WO2022261251A1 - Immunogenic compositions comprising tumour-associated antigen - Google Patents

Immunogenic compositions comprising tumour-associated antigen Download PDF

Info

Publication number
WO2022261251A1
WO2022261251A1 PCT/US2022/032730 US2022032730W WO2022261251A1 WO 2022261251 A1 WO2022261251 A1 WO 2022261251A1 US 2022032730 W US2022032730 W US 2022032730W WO 2022261251 A1 WO2022261251 A1 WO 2022261251A1
Authority
WO
WIPO (PCT)
Prior art keywords
immunogenic composition
taca
stn
cancer
carrier protein
Prior art date
Application number
PCT/US2022/032730
Other languages
French (fr)
Inventor
John Robert HASERICK
Khanita KARAVEG
Shiteshu SHRIMAL
Nicholas M. VALIANTE
Original Assignee
Glyde Bio Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glyde Bio Inc. filed Critical Glyde Bio Inc.
Publication of WO2022261251A1 publication Critical patent/WO2022261251A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/646Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the entire peptide or protein drug conjugate elicits an immune response, e.g. conjugate vaccines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/001172Sialyl-Thomson-nouvelle antigen [sTn]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55577Saponins; Quil A; QS21; ISCOMS
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6037Bacterial toxins, e.g. diphteria toxoid [DT], tetanus toxoid [TT]

Definitions

  • This invention is in the field of immunogenic compositions comprising conjugates of tumour-associated carbohydrate antigens (TACAs) and carrier proteins.
  • TACAs tumour-associated carbohydrate antigens
  • carrier proteins carrier proteins
  • TACAs tumour-associated carbohydrate antigens
  • carbohydrates are T-independent antigens, they are poorly immunogenic and often only give rise to short-lived, IgM-based responses. Conjugation to a carrier protein can convert T-independent antigens into T- dependent antigens, thereby enhancing memory responses and allowing immunity to develop.
  • Antibodies specific for TACAs should, in theory, slow growth and/or induce apoptosis of tumour cells.
  • TACAs There are two major classes of TACAs, namely glycoprotein antigens and glycolipid antigens.
  • Glycoprotein antigens include the O-linked carbohydrates Thomsen-nouveau (Tn), sialyl-Tn (STn), and Thomsen-Friedenreich (TF).
  • Tn is often overexpressed in breast, prostate, and stomach cancers
  • STn is overexpressed in breast, colon, lung, ovary, prostate, and stomach cancers
  • TF is overexpressed in breast, colon, ovary, prostate, and stomach cancers [3].
  • Tn, STn, and TF are also associated with various infectious diseases [133], [132], [133].
  • the invention provides conjugates comprising Tn, STn, or TF conjugated to a carrier protein.
  • the invention provides immunogenic compositions comprising Tn, STn, and TF, each conjugated to a carrier protein.
  • the conjugates and immunogenic compositions may be used as vaccines for treating and/or preventing cancer.
  • the invention provides a method for immunising a patient against cancer, the method comprising a step of administering to the patient an immunogenic composition of the first aspect of the invention.
  • Immunogenic compositions In one embodiment, the invention provides an immunogenic composition comprising Tn conjugated to a carrier protein, STn conjugated to a carrier protein, and TF conjugated to a carrier protein. The inventors have found that these compositions may induce higher IgG responses than monovalent compositions (i.e. compositions comprising one of Tn, STn, or TF) and may not exhibit significant cross-reactivity. In some embodiments, the compositions comprise only one or two of these conjugates. For example, the compositions may be monovalent or bivalent.
  • the compositions comprise other conjugates, in particular conjugates having other TACAs.
  • the compositions may be tetravalent, pentavalent, hexavalent, etc.
  • the immunogenic compositions of the invention do not comprise any TACAs other than Tn, STn, and TF. In other words, the immunogenic compositions are typically trivalent.
  • Tumour-associated carbohydrate antigens TACAs
  • the present invention makes use of the TACAs Tn (GalNAc ⁇ 1 –), STn (Neu5Ac ⁇ 2,6 GalNAc ⁇ 1 –), and TF (Gal ⁇ 3GalNAc ⁇ 1 –) (see FIG. 1A).
  • TACAs are closely related, chemically and biosynthetically. They occur naturally as O-linked glycans, meaning they link to the hydroxyl group of a serine or threonine residue of a protein. The inventors have selected these TACAs for use in the invention based on a variety of reasons.
  • TACAs tumour-associated antigens
  • TAAs tumour-associated antigens
  • the invention also make use of other TACAs, for example glycolipid TACAs (e.g. Globo series, gangliosides, and/or Lewis structure series).
  • the immunogenic compositions of the invention do not comprise any glycolipid TACAs.
  • the immunogenic compositions of the invention do not comprise Globo-H.
  • the immunogenic compositions of the invention do not comprise any TACAs other than Tn, STn, and/or TF.
  • TACAs used in the invention are typically in their native form as described above.
  • TACAs are modified.
  • N- acyl modified Tn, STn, and/or TF are used, for example as described in [11], [12], or [13].
  • /V-fluoroacetyl, /V-phenylacetyl and/or /V-chlorophenylacetyl modified Tn, STn, and/or TF are used [14].
  • fluorine-substituted TACAs are used to improve ligand binding and/or enhance the immune response [12], [15].
  • TF is modified to have a hydrophobic terminal residue such as benzyl, p-nitrophenyl, butyl, propyl, ethyl, or methyl, as described in [16].
  • S-glycosides and C-glycosides are used to lengthen in vivo lifetime [15].
  • further optional modifications include sulfation and acetylation.
  • TACA mimetics are used.
  • the conformationally constrained Tn mimetic described in [17] is used. Modified TACAs and mimetics may reduce immune tolerance, reduce TACA degradation, and/or increase immunogenicity. However, generally, the TACAs used in the invention are not modified.
  • the ratio of the TACAs may vary.
  • the ratio (w/w) of each TACA to each other TACA is from about 5:1 to about 1:5, from about 4:1 to about 1:4, from about 3:1 to about 1:3; from about 2:1 to about 1:2, from about 1.5:1 to about 1:1.5, from about 1.4:1 to about 1:1.4, from about 1.3:1 to about 1:1.3, from about 1.2:1 to about 1:1.2, from about 1.1:1 to about 1:1.1.
  • the ratio (w/w) of each TACA to each other TACA is about 1:1.
  • trivalent compositions e.g.
  • each TACA typically constitutes from about 15% to about 50%, from about 20% to about 45%, or from about 25% to about 40% (w/w) of the total TACAs. In a particular embodiment, each TACA constitutes from about 30% to about 35% (w/w) of the total number of TACAs. In a particular embodiment, in trivalent compositions, the three TACAs are present in a molecular ratio of about 1:1:1. Such compositions allow for easier manufacture. Tn, STn, and TF can be synthesised by techniques known in the art. TACAs and PEG3-COOH functionalised TACAs are available fromshire Research Laboratories Inc. (Ottawa, ON, Canada). Carrier proteins
  • the invention provides conjugates that comprise TACAs (typically Tn, STn, and/or TF) conjugated to a carrier protein.
  • TACAs typically Tn, STn, and/or TF
  • covalent conjugation of TACAs to carrier proteins enhances the immunogenicity of the TACAs as it converts them from T-independent antigens to T-dependent antigens, thus allowing priming for immunological memory.
  • Conjugation to carrier proteins is a well-known technique in vaccinology.
  • the invention involves the use of carrier proteins, typically immunogenic carrier proteins.
  • carrier proteins include bacterial toxins or toxoids, such as diphtheria toxoid or tetanus toxoid. Fragments of toxins or toxoids can also be used e.g. fragment C of tetanus toxoid [18].
  • Cross-Reactive-Material-197 is a diphtheria toxin which has lost the ADP- ribosyltransferase activity of the native toxin as a result of genetic mutation [19-21].
  • CRM197 is the carrier protein, because CRM197 is not itself glycosylated, the amines of CRM197 are amenable to modifications, and CRM197 has been used in approved prophylactic vaccines (e.g. Menveo ® and Prevnar13 ® vaccines). Also, the inventors found that conjugates using CRM197 as the carrier protein for Tn, STn, and TF resulted in high levels of IgG compared to other carrier proteins.
  • CRM197 is a recombinant CRM197 because it is easy and efficient to synthesise.
  • recombinant CRM197 for use in the invention is produced in E.
  • recombinant CRM197 for use in the invention is produced in a Pseudomonas host cell (including, for example, those described and produced in [25] and [26]). Recombinant CRM197 for use in the invention can also be produced using methods described in [27].
  • Other carrier proteins for use in the invention include Keyhole limpet haemocyanin (KLH) [28], the N.
  • influenzae [39,40], pneumococcal surface protein PspA [41], pneumolysin [42], iron-uptake proteins [43], toxin A or B from C.difficile [44], recombinant Pseudomonas aeruginosa exoprotein A (rEPA) [45], a GBS protein, etc.
  • more than one type of carrier protein is used in the invention e.g. to reduce the risk of carrier suppression.
  • different types of carrier proteins are used for different types of TACAs, e.g. Tn is conjugated to CRM197 while STn is conjugated to tetanus toxoid.
  • more than one type of carrier protein is used for a particular type of TACA, e.g. some Tn molecules are conjugated to CRM197 and other Tn molecules are conjugated to tetanus toxoid. In general, however, it is preferred to use the same carrier protein for all TACAs (typically CRM197).
  • a single carrier protein molecule is conjugated to more than one type of TACA.
  • a single carrier protein is conjugated to two or three of Tn, STn, and TF.
  • different TACAs are mixed prior to the conjugation reaction.
  • each carrier protein molecule is conjugated to a single type of TACA (e.g. Tn, STn, or TF).
  • the immunogenic compositions of the invention comprises a first conjugate comprising Tn conjugated to a first carrier protein, a second conjugate comprising STn conjugated to a second carrier protein, and a third conjugate comprising TF conjugated to a third carrier protein.
  • the first, second, and third carrier proteins are all CRM197.
  • the precise number of TACAs per carrier molecule can vary. Generally, the average number of TACA molecules per carrier molecule is at least about 20% of the total number of accessible conjugation sites on the carrier molecule, in particular from about 20% to about 100% of the total number of accessible conjugation sites. Typically, the average number of TACA molecules per carrier is at least about 30% of the total number of accessible conjugation sites, in particular from about 30% to about 100% of the total number of accessible conjugation sites.
  • CRM197 contains approximately 40 sites that could be conjugated to a TACA, if the protein was fully denatured. However, in the native protein only approximately 20 of these sites are accessible to modification. Previous publications have described which lysines in CRM197 are most reactive [46].
  • the carrier protein is a diphtheria toxin or toxoid, such as CRM197
  • the average number of TACA molecules per carrier molecule is typically at least about 4, in particular from about 4 to about 15.
  • the average number of TACA molecules per carrier molecule is at least about 6, in particular from about 6 to about 15. The inventors found that conjugates having these TACA:carrier ratios were highly immunogenic.
  • the average number of TACA molecules per carrier molecule is at least about 7. In some embodiments, for CRM197-STn conjugates, the average number of TACA molecules per carrier molecule is at least about 5. In some embodiments, for CRM197-TF conjugates, the average number of TACA molecules per carrier molecule is at least about 6, in particular from about 7 to about 10.
  • TACAs may be conjugated to carrier proteins covalently or non-covalently.
  • the TACAs are conjugated to the carrier proteins covalently, e.g. via a linker.
  • the TACAs are non-covalently conjugated to the carrier proteins.
  • the reducing end of the TACA is functionalised with a reactive moiety, for example p-nitrophenyl, maleimide, or an aldehyde-containing group, which is then conjugated to the carrier protein through amide bond formation, Michael addition or reductive amination [14].
  • a reactive moiety for example p-nitrophenyl, maleimide, or an aldehyde-containing group
  • the carrier protein through amide bond formation, Michael addition or reductive amination [14].
  • some conjugation methods used in the art are inefficient, destroy/denature the TACA, or result in a linker between the TACA and the carrier protein which itself is immunogenic and elicits high levels of anti-linker antibodies.
  • each TACA is conjugated to its carrier covalently via a linker which is non-immunogenic and/or elicits low levels of anti-linker antibodies (typically 1 or 2 orders of magnitude lower than anti-TACA antibodies).
  • linkers comprise serine residues, threonine residues, polyethylene glycol (PEG) groups, alkane groups, thiol groups, maleimide groups, amide groups, thio-ether groups, thioester groups, ether groups, aminoxy groups, oxime groups, hydrazone groups, or combinations thereof.
  • PEG polyethylene glycol
  • linkers comprise serine and/or threonine residues, e.g. when the TACA is O-linked to a serine or threonine residue.
  • the linker comprises a single serine residue.
  • the linker comprises a single threonine residue.
  • the linker comprises both serine and threonine residues.
  • the serine or threonine may be covalently bound to a lysine residue in the carrier protein via a maleimide-containing moiety. Linkers comprising serine and/or threonine are advantageous because TACAs are naturally found on serine and threonine residues, and they exhibit low immunogenicity.
  • linkers comprise an alkane group, i.e. -(CH ⁇ 2) n -, wherein n is typically from 2 to 8, from 2 to 6, or from 2 to 4. In some embodiments, n is 2, 3, 4, 5, 6, 7, or 8. In some embodiments, n is 4.
  • the TACA is O-linked to an alkane-carboxyl which is reacted with the amine group in the side chain of a lysine residue in the carrier protein. Alkane linkers are advantageous because they exhibit low immunogenicity.
  • linkers comprise polyethylene glycol (PEG), i.e. -(O-CH 2 -CH 2 ) n -, wherein n is typically from 2 to 12, from 3 to 10, from 3 to 8, or from 3 to 6, or preferably is 3 or 4. In some embodiments, n is 3, 4, 5, 6, 7, or 8. In some embodiments n is from 3 to 8. In some embodiments, n is from 3 to 6. In some embodiments, n is 3 or 4. In a particular embodiment, the linker comprises PEG-3 (i.e. -(O -CH 2 -CH 2 ) n - wherein n is 3). PEG linkers are particularly advantageous because they exhibit low immunogenicity, low cross-reactivity, few side-effects, and allow for easy manufacture at the preferred TACA:carrier ratios.
  • PEG linkers are particularly advantageous because they exhibit low immunogenicity, low cross-reactivity, few side-effects, and allow for easy manufacture at the preferred TACA:carrier ratios.
  • the TACA is O-linked to PEG(n)-carboxyl, which is reacted with the amine group in the side chain of a lysine residue in the carrier protein .
  • An exemplary resulting conjugate is CRM197-PEG3-Tn 1 shown in FIG. IB.
  • the TACA is O-linked to PEG(n)-azide, PEG(n)-thiol, or PEG(n)-amine, which is reacted with a modified lysine residue in the carrier protein.
  • the amine group in the side chain of the lysine may be modified with a bifunctional linker.
  • a homobifunctional linker of the formula X-L-X may be used, wherein the two X groups are the same as each other and can react with the azide, thiol, or amine in the O-linked TACA and the amine group in the side chain of the lysine, and wherein L is a linking moiety in the linker.
  • a heterobifunctional linker of the formula X-L-X is used, wherein the two X groups are different and one can react with the azide, thiol, or amine in the O-linked TACA and the other can react with the amine group in the side chain of the lysine, and wherein L is a linking moiety in the linker.
  • An exemplary X group is N- hydroxysuccinimide.
  • Exemplary heterobifunctional crosslinkers are GMBS (N-y- maleimidobutyryloxysuccinimide ester) and Alkyne, Succinimidyl Ester (3- propargyloxypropanoic acid, succinimidyl ester).
  • An exemplary resulting conjugate is CRM197- PEG3-Tn 2 shown in FIG. IB.
  • PEG(n)-azide e.g. in CRM197-PEG3-Tn 2 shown in FIG. IB
  • PEG(n)-carboxyl e.g. in CRM197-PEG3-Tn 1 shown in FIG. IB
  • strongly immunogenic constructs without eliciting high levels of anti-PEG antibodies.
  • Exemplary linkers are shown in the conjugates in FIG. IB (only Tn conjugates are shown but any of Tn, STn, of TF could be used).
  • the invention also provides methods of producing conjugates (and conjugates obtainable by these methods), comprising (a) providing a TACA and a carrier molecule; (b) conjugating the TACA to a carrier molecule.
  • the method comprises: (a) providing a PEG3-COOH functionalised TACA selected from Tn-PEG3-COOH, STn-PEG3-COOH, and TF-PEG3-COOH, and a CRM197; (b) conjugating the TACA to the CRM197 by (i) activating the PEG3-COOH functionalised TACA with 1-ethyl-3-(-3-dimethylaminopropyl) carbodiimide hydrochloride (EDC) and N-Hydroxysuccinimide (NHS), optionally at a 1:2:2 ratio of TACA:EDC:NHS in 0.1M 2- ethanesulfonic acid (MES) buffer pH 5.0 supplemented with 10-14% v/v dimethyl sulfoxide (DMSO)
  • the method further comprises c) filtering out reactants, biproducts, residual solvent and/or concentrate, optionally by performing buffer exchange on the reaction mixture, further optionally by dialysis or tangential flow filtration.
  • the TACAs used in the invention are conjugated to carrier proteins via peptide backbones, for example as described in [47], [48], [49], [50], or [51].
  • the TACAs may be in a clustered formation.
  • the TACAs used in the invention are not present on a peptide backbone.
  • the TACAs used in the invention are not in a clustered formation.
  • the conjugates used in the invention comprise a polypeptide derived from a mucin, for example as described in [49], [52], [53], or [54].
  • a polypeptide derived from a mucin means a polypeptide of 8 or more amino acids in length, having at least about 80% identity to a region of any of MUC1-22.
  • the conjugates used in the invention do not comprise a polypeptide derived from a mucin.
  • the conjugates used in the invention comprise a T cell epitope, for example a PV (KLFAVWKITYKDT), a pan-DR epitope ("PADRE", aAKXVAAWTLKAa) as described in [55], and/or OVA 323-339 (ISQAVHAAHAEINEAGR) as described in [14].
  • T cell epitope for example a PV (KLFAVWKITYKDT), a pan-DR epitope ("PADRE”, aAKXVAAWTLKAa) as described in [55], and/or OVA 323-339 (ISQAVHAAHAEINEAGR) as described in [14].
  • PADRE pan-DR epitope
  • OVA 323-339 ISQAVHAAHAEINEAGR
  • each of the conjugates used in the invention comprises or consists of a TACA linked to a carrier protein via a linker.
  • each of the conjugates used in the invention comprises or consists of a TACA selected from Tn, STn, and TF, linked to CRM197 via a linker comprising a PEG, more preferably PEG-3.
  • the immunogenic composition of the invention comprises precisely two of the TACAs described herein, i.e. the composition is bivalent.
  • the composition comprises Tn conjugated to a carrier protein and STn conjugated to a carrier protein, since Tn and STn are very closely related and are usually present on the same tumour.
  • the composition comprises STn conjugated to a carrier protein and TF conjugated to a carrier protein, since STn can elicit antibodies against Tn.
  • the composition comprises Tn conjugated to a carrier protein and TF conjugated to a carrier protein.
  • the immunogenic composition of the invention comprises only one of the TACAs described herein, i.e. the composition is monovalent.
  • the immunogenic composition comprises STn conjugated to a carrier protein, since the inventors have observed that STn alone can elicit a Tn and STn response (these TACAs are very closely related, chemically and biosynthetically).
  • the immunogenic compositions of the invention may further comprise a pharmaceutically acceptable carrier.
  • Typical pharmaceutically acceptable carriers include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition. Suitable carriers are typically large, slowly metabolised macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, sucrose [56], trehalose [57], lactose, and lipid aggregates (such as oil droplets or liposomes). Such carriers are well known in the art.
  • the immunogenic compositions may also contain diluents, such as water, saline, glycerol, etc.
  • compositions of the invention may be in aqueous form (i.e. solutions or suspensions) or in a dried form (e.g. lyophilised). If a dried immunogenic composition is used then it will be reconstituted into a liquid medium prior to injection. Lyophilisation of conjugate vaccines are known in the art.
  • the immunogenic compositions of the invention include conjugates comprising more than one type of TACA (e.g.
  • Tn, STn, and TF it is typical for the conjugates to be prepared separately, mixed, and then lyophilised. In this way, lyophilised compositions comprising multiple (preferably three) conjugates as described herein may be prepared.
  • a sugar alcohol e.g. mannitol
  • a disaccharide e.g. sucrose or trehalose
  • compositions may be presented in vials, or they may be presented in ready-filled syringes.
  • the syringes may be supplied with or without needles.
  • a syringe will include a single dose of the composition, whereas a vial may include a single dose or multiple doses.
  • compositions of the invention are also suitable for reconstituting other vaccines from a lyophilised form.
  • the invention provides a kit, which may comprise two vials, or may comprise one ready-filled syringe and one vial, with the contents of the syringe being used to reactivate the contents of the vial prior to injection.
  • compositions of the invention may be packaged in unit dose form or in multiple dose form.
  • vials are preferred to pre-filled syringes.
  • Effective dosage volumes can be routinely established, but a typical human dose of the composition has a volume of 0.5ml e.g. for intramuscular injection.
  • the pH of the composition is preferably between 6 and 8, preferably about 7. Stable pH may be maintained by the use of a buffer.
  • the immunogenic compositions of the invention typically comprise disodium monohydrogen phosphate and potassium dihydrogen phosphate buffer. If a composition comprises an aluminium hydroxide salt, it is preferred to use a histidine buffer [59].
  • the composition may be sterile and/or pyrogen-free. Compositions of the invention may be isotonic with respect to humans.
  • compositions of the invention are immunogenic, and are more preferably vaccine compositions.
  • Vaccines according to the invention may either be prophylactic (i.e. to prevent cancer) or therapeutic (i.e. to treat cancer), but will typically be therapeutic.
  • Immunogenic compositions used as vaccines comprise an immunologically effective amount of each antigen, as well as any other components, as needed.
  • immunologically effective amount it is meant that the administration of that amount to a subject, either in a single dose or as part of a series, is effective for treatment and/or prevention. This amount varies depending upon the taxonomic group of subject to be treated (e.g.
  • non-human primate, primate, etc. the health and physical condition of the subject to be treated, age, the capacity of the subject immune system to synthesise antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
  • compositions of the invention may be prepared in various forms.
  • the compositions may be prepared as injectables, either as liquid solutions or suspensions.
  • the composition may be prepared for pulmonary administration e.g. as an inhaler, using a fine powder or a spray.
  • the composition may be prepared as a suppository or pessary.
  • the composition may be prepared for nasal, aural or ocular administration e.g. as spray, drops, gel or powder [e.g. refs 60 & 61].
  • Compositions of the invention may include an antimicrobial, particularly when packaged in multiple dose format.
  • compositions of the invention may comprise detergent e.g. a Tween (polysorbate), such as Tween 80.
  • Detergents are generally present at low levels e.g. ⁇ 0.01%.
  • compositions of the invention may include sodium salts (typically sodium chloride and potassium chloride) to give tonicity.
  • sodium salts typically sodium chloride and potassium chloride
  • a concentration of 10 ⁇ 2mg/ml NaCI is typical.
  • Compositions of the invention will generally include one or more buffers. Potassium phosphate monobasic buffers and sodium phosphate dibasic buffers are typical.
  • the invention also provides methods of producing pharmaceutical compositions comprising combining two or more conjugates of the invention.
  • the method comprises combining: a) a first conjugate comprising Tn conjugated to a first carrier protein; b) a second conjugate comprising STn conjugated to a second carrier protein; and c) a third conjugate comprising TF conjugated to a third carrier protein, optionally with a pharmaceutically acceptable carrier.
  • conjugates a)-c) are combined in a ratio (w/w) of about 1:1:1.
  • the first, second, and third carrier proteins are each CRM197.
  • Adjuvants are each CRM197.
  • compositions of the invention may be administered in conjunction with other immunoregulatory agents.
  • compositions may include one or more adjuvants described in detail below.
  • Mineral containing compositions suitable for use as adjuvants in the invention include mineral salts, such as aluminium salts and calcium salts (or mixtures thereof).
  • Calcium salts include calcium phosphate (e.g. the "CAP" particles disclosed in [62]).
  • Aluminium salts include hydroxides, phosphates, sulfates, etc., with the salts taking any suitable form (e.g. gel, crystalline, amorphous, etc.). Adsorption to these salts is preferred.
  • the mineral containing compositions may also be formulated as a particle of metal salt [63].
  • the adjuvants known as aluminium hydroxide and aluminium phosphate may be used. These names are conventional, but are used for convenience only, as neither is a precise description of the actual chemical compound which is present (e.g. see chapter 9 of [64]).
  • the invention can use any of the "hydroxide” or "phosphate” adjuvants that are in general use as adjuvants.
  • the adjuvants known as "aluminium hydroxide” are typically aluminium oxyhydroxide salts, which are usually at least partially crystalline.
  • the adjuvants known as "aluminium phosphate” are typically aluminium hydroxyphosphates, often also containing a small amount of sulfate ( i.e . aluminium hydroxyphosphate sulfate). They may be obtained by precipitation, and the reaction conditions and concentrations during precipitation influence the degree of substitution of phosphate for hydroxyl in the salt.
  • Alhydrogel ® may be used as an adjuvant.
  • Oil emulsion compositions suitable for use as adjuvants in the invention include oil-in-water emulsions, such as AS03 or the nanoemulsion AddaS03TM (AS03), and squalene-water emulsions, such as MF59 (5% Squalene, 0.5% Tween 80, and 0.5% Span 85, formulated into submicron particles using a microfluidizer) (Chapter 10 of [64]; see also [65]-[67]).
  • oil-in-water emulsions such as AS03 or the nanoemulsion AddaS03TM (AS03)
  • squalene-water emulsions such as MF59 (5% Squalene, 0.5% Tween 80, and 0.5% Span 85, formulated into submicron particles using a microfluidizer) (Chapter 10 of [64]; see also [65]-[67]).
  • Particularly preferred adjuvants for use in the compositions are submicron oil-in-water emulsions.
  • Submicron oil-in-water emulsions, methods of making the same and immunostimulating agents, such as muramyl peptides, for use in the compositions, are described in detail in [65] & [68]-[69].
  • CFA Complete Freund's adjuvant
  • IFA incomplete Freund's adjuvant
  • Saponin formulations such as those described in (chapter 22 of[126]) may also be used as adjuvants in the invention.
  • Saponins are a heterologous group of sterol glycosides and triterpenoid glycosides that are found in the bark, leaves, stems, roots and even flowers of a wide range of plant species. Saponins isolated from the bark of the Quillaia soponorio Molina tree have been widely studied as adjuvants. Saponin can also be commercially obtained from Smilax ornata (sarsaparilla), Gypsophilla paniculata (brides veil), and Saponaria officianalis (soap root).
  • Saponin adjuvant formulations include purified formulations, such as QS21, as well as lipid formulations, such as ISCOMs.
  • Saponin compositions have been purified using HPLC and RP-HPLC. Specific purified fractions using these techniques have been identified, including QS7, QS17, QS18, QS21, QH-A, QH-B and QH-C.
  • the saponin is QS21.
  • a method of production of QS21 is disclosed in[70].
  • Saponin formulations may also comprise a sterol, such as cholesterol [71].
  • ISCOMs immunostimulating complexes
  • phospholipid such as phosphatidylethanolamine or phosphatidylcholine.
  • Any known saponin can be used in ISCOMs.
  • the ISCOM includes one or more of QuilA, QHA and QHC. ISCOMs are further described in [71]-[73].
  • the ISCOMS may be devoid of additional detergent(s) [74].
  • Virosomes and virus-like particles can also be used as adjuvants in the invention.
  • These structures generally contain one or more proteins from a virus optionally combined or formulated with a phospholipid. They are generally non-pathogenic, non-replicating and generally do not contain any of the native viral genome.
  • the viral proteins may be recombinantly produced or isolated from whole viruses.
  • viral proteins suitable for use in virosomes or VLPs include proteins derived from influenza virus (such as HA or NA), Hepatitis B virus (such as core or capsid proteins), Hepatitis E virus, measles virus, Sindbis virus, Rotavirus, Foot-and-Mouth Disease virus, Retrovirus, Norwalk virus, human Papilloma virus, HIV, RNA- phages, Qß-phage (such as coat proteins), GA-phage, fr-phage, AP205 phage, and Ty (such as retrotransposon Ty protein pi).
  • VLPs are discussed further in[77]-[82].
  • Virosomes are discussed further in, for example [83].
  • Bacterial or microbial derivatives are discussed further in, for example [83].
  • Adjuvants suitable for use in the invention include bacterial or microbial derivatives such as non-toxic derivatives of enterobacterial lipopolysaccharide (LPS), Lipid A derivatives, immunostimulatory oligonucleotides and ADP-ribosylating toxins and detoxified derivatives thereof.
  • LPS enterobacterial lipopolysaccharide
  • Lipid A derivatives Lipid A derivatives
  • immunostimulatory oligonucleotides and ADP-ribosylating toxins and detoxified derivatives thereof.
  • Non-toxic derivatives of LPS include monophosphoryl lipid A (MPLA) and 3-O-deacylated MPL (3dMPL).
  • 3dMPL is a mixture of 3 de-O-acylated MPLA with 4, 5 or 6 acylated chains.
  • a preferred "small particle" form of 3 De-O-acylated MPLA is disclosed in [84]. Such "small particles" of 3dMPL are small enough to be sterile filtered through a 0.22 ⁇ m membrane [84].
  • Other non-toxic LPS derivatives include MPLA mimics, such as aminoalkyl glucosaminide phosphate derivatives e.g. RC-529 [85,86].
  • the MPLA analog/derivative is selected from Monophosphoryl 3-Deacyl Lipid A and Monophosphoryl Hexa-acyl Lipid A, 3- Deacyl (e.g. PHAD ® , 3D-PHAD ® , and 3D-6A-PHAD ® - Avanti Polar Lipids). .
  • Lipid A derivatives include derivatives of lipid A from Escherichia coli such as OM-174.
  • OM-174 is described for example in [87] & [88].
  • Immunostimulatory oligonucleotides suitable for use as adjuvants in the invention include nucleotide sequences containing a CpG motif (a dinucleotide sequence containing an unmethylated cytosine linked by a phosphate bond to a guanosine). Double-stranded RNAs and oligonucleotides containing palindromic or poly(dG) sequences have also been shown to be immunostimulatory.
  • the CpG's can include nucleotide modifications/analogs such as phosphorothioate modifications and can be double-stranded or single-stranded. [89], [90] and [91] disclose possible analog substitutions e.g. replacement of guanosine with 2'-deoxy-7-deazaguanosine. The adjuvant effect of CpG oligonucleotides is further discussed in [92]-[97].
  • the CpG sequence may be directed to TLR9, such as the motif GTCGTT or TTCGTT [98].
  • the CpG sequence may be specific for inducing a Th1 immune response, such as a CpG-A ODN, or it may be more specific for inducing a B cell response, such a CpG-B ODN.
  • CpG-A and CpG-B ODNs are discussed in refs. 99-101.
  • the CpG is a CpG-B ODN, for example ODN1826 may be used in mice and ODN7909 (AKA ODN2006) may be used in humans.
  • the CpG oligonucleotide is constructed so that the 5' end is accessible for receptor recognition.
  • two CpG oligonucleotide sequences may be attached at their 3' ends to form "immunomers". See, for example, [98] & [102]-[104].
  • Bacterial ADP-ribosylating toxins and detoxified derivatives thereof may be used as adjuvants in the invention.
  • the protein is derived from E.coli (E.coli heat labile enterotoxin "LT"), cholera ("CT”), or pertussis ("PT”).
  • LT E.coli heat labile enterotoxin
  • CT cholera
  • PT pertussis
  • the toxin or toxoid is preferably in the form of a holotoxin, comprising both A and B subunits.
  • the A subunit contains a detoxifying mutation; preferably the B subunit is not mutated.
  • the adjuvant is a detoxified LT mutant such as LT-K63, LT-R72, and LT-G192.
  • the use of ADP- ribosylating toxins and detoxified derivatives thereof, particularly LT-K63 and LT-R72, as adjuvants can be found in refs. 107-114. Numerical reference for amino acid substitutions is preferably based on the alignments of the A and B subunits of ADP-ribosylating toxins set forth in [115].
  • Pam3CSK4 (Pam3CysSerLys4) may also be used as an adjuvant. It is a synthetic lipopeptide and a TLR2/TLR2 ligand and mimics the acylated amino terminus of bacterial lipopeptides.
  • Trehalose-6, 6-dibehenate may also be used as an adjuvant. It is a synthetic analog of trehalose-6, 6-dimycolate (TDM, also known as cord factor), which is a well-known immunostimulatory component of M. tuberculosis. TDB binds the C-Type lectin, Mincle.
  • Human immunomodulators suitable for use as adjuvants in the invention include cytokines, such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12 [116], etc.) [117], interferons (e.g. interferon- ⁇ ), macrophage colony stimulating factor, and tumour necrosis factor.
  • cytokines such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12 [116], etc.) [117], interferons (e.g. interferon- ⁇ ), macrophage colony stimulating factor, and tumour necrosis factor.
  • Bioadhesives and mucoadhesives may also be used as adjuvants in the invention.
  • Suitable bioadhesives include esterified hyaluronic acid microspheres [118] or mucoadhesives such as cross-linked derivatives of poly(acrylic acid), polyvinyl alcohol, polyvinyl pyrollidone, polysaccharides and carboxymethylcellulose. Chitosan and derivatives thereof may also be used as adjuvants in the invention [119].
  • Microparticles may also be used as adjuvants in the invention.
  • Microparticles i.e. a particle of ⁇ 100nm to ⁇ 150 ⁇ m in diameter, more preferably ⁇ 200nm to ⁇ 30 ⁇ m in diameter, and most preferably ⁇ 500nm to ⁇ 10 ⁇ m in diameter
  • materials that are biodegradable and non-toxic e.g. a poly(a-hydroxy acid), a polyhydroxybutyric acid, a polyorthoester, a polyanhydride, a polycaprolactone, etc.
  • a negatively-charged surface e.g. with SDS
  • a positively-charged surface e.g. with a cationic detergent, such as CTAB
  • liposome formulations suitable for use as adjuvants are described in [120]-[121] and Chapters 13 & 14 of [64].
  • liposomes are used.
  • liposomes may be composed of the phospholipid 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and cholesterol.
  • DOPC 1,2-dioleoyl-sn-glycero-3-phosphocholine
  • Liposomes provide a stable solution and consistent doses of adjuvants.
  • liposomes can impart better injection site and patient tolerance.
  • Polyoxyethylene ether and polyoxyethylene ester formulations Adjuvants suitable for use in the invention include polyoxyethylene ethers and polyoxyethylene esters [122]. Such formulations further include polyoxyethylene sorbitan ester surfactants in combination with an octoxynol [123] as well as polyoxyethylene alkyl ethers or ester surfactants in combination with at least one additional non-ionic surfactant such as an octoxynol [124].
  • Preferred polyoxyethylene ethers are selected from the following group: polyoxyethylene-9-lauryl ether (laureth 9), polyoxyethylene-9-steoryl ether, polyoxytheylene-8- steoryl ether, polyoxyethylene-4-lauryl ether, polyoxyethylene-35-lauryl ether, and polyoxyethylene-23-lauryl ether.
  • PCPP Polyphosphazene
  • PCPP formulations are described, for example, in [125] and [126].
  • muramyl peptides suitable for use as adjuvants in the invention include N-acetyl- muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), and N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1'-2'-dipalmitoyl-sn- glycero-3-hydroxyphosphoryloxy)-ethylamine MTP-PE).
  • thr-MDP N-acetyl- muramyl-L-threonyl-D-isoglutamine
  • nor-MDP N-acetyl-normuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1'-2'-dipalmitoyl
  • imidazoquinolone compounds suitable for use adjuvants in the invention include Imiquamod and its homologues ( e,g . "Resiquimod 3M”), described further in [127] and [128].
  • thiosemicarbazone compounds as well as methods of formulating, manufacturing, and screening for compounds all suitable for use as adjuvants in the invention include those described in [129].
  • the thiosemicarbazones are particularly effective in the stimulation of human peripheral blood mononuclear cells for the production of cytokines, such as TNF-a.
  • tryptanthrin compounds as well as methods of formulating, manufacturing, and screening for compounds all suitable for use as adjuvants in the invention include those described in [130].
  • the tryptanthrin compounds are particularly effective in the stimulation of human peripheral blood mononuclear cells for the production of cytokines, such as TNF-a.
  • Polyinosinic:polycytidylic acid may be used as an adjuvant.
  • Poly l:C is a synthetic analog of dsRNA and interacts with TLR3.
  • the invention may also comprise combinations of aspects of one or more of the adjuvants identified above.
  • two or more adjuvants may be used in compositions of the invention.
  • the invention also provides a method for raising an immune response in a subject, comprising administering a conjugate or composition of the invention to the subject.
  • the immune response is preferably preventative (prophylactic) and/or therapeutic, and preferably involves antibodies.
  • the method may raise a booster response.
  • the invention also provides a conjugate or composition of the invention for use as a medicament or for use in a method of prevention/treatment.
  • the medicament or method is preferably able to raise an immune response in a subject (i.e. it is an immunogenic composition) and is more preferably a vaccine.
  • the invention also provides the use of a composition of the invention in the manufacture of a medicament for raising an immune response in a subject.
  • the method may comprise further administering a checkpoint inhibitor to the subject.
  • the invention also provides a checkpoint inhibitor for use in a method of treatment, wherein the method comprises administering a conjugate or composition of the invention.
  • Suitable checkpoint inhibitors include an anti-PD-1 or anti-PD-L1 antibody, for example nivolumab, pembrolizumab, and/or atezolizumab.
  • Broadly active checkpoint inhibitors allow for a more antigen-specific response induced by a vaccine of the invention to target tumours more effectively, and may exhibit synergy with the vaccine.
  • Checkpoint inhibitors may be administered simultaneously or at different time points from the conjugate or composition of the invention.
  • Checkpoint inhibitors may be administered as part of the same composition as the conjugate or composition of the invention or separately.
  • these methods and uses are for the prevention and/or treatment of cancer (i.e. a malignant neoplasm).
  • the cancer is a solid tumour.
  • the cancer is an epithelial cancer or carcinoma, for example an adenocarcinoma or a squamous cell carcinoma.
  • the cancer is pancreatic cancer, breast cancer, colon cancer, gastro-intestinal cancer, prostate cancer, lung cancer, ovarian cancer, or stomach cancer, because these types of cancer have been associated with high expression of Tn, STn, and TF.
  • the cancer may be stage 0, I, II, III, or IV.
  • the cancer may be metastatic cancer.
  • the cancer may be relapsed or refractory to other treatments.
  • the cancer may comprise cancer cells expressing one or more of Tn, STn, and TF, for example two of Tn, STn, and TF, on the cell surface.
  • the cancer cells may overexpress one or more of Tn, STn, and TF, for example two of Tn, STn, and TF, compared to a reference expression level or control sample.
  • a control sample may be healthy (i.e. non-cancer) cells, which may be tissue- matched and/or patient-matched.
  • the method may comprise determining the level of one or more of Tn, STn, and TF expression on cancer cells obtained from the subject.
  • the method may comprise identifying the subject as being suitable for treatment if one or more of Tn, STn, and TF is expressed on the cancer cells or is overexpressed on the cancer cells compared to a reference expression level or control sample.
  • compositions comprising Tn, STn, or TF conjugates are used to prevent and/or treat infections with parasites expressing Tn, STn, or TF.
  • compositions comprising Tn conjugates are used to prevent and/or treat infections with parasites expressing Tn, for example Fasciola hepatica [131], Schistostoma mansoni, Echinococcus granulosus, or Cryptosporidium parvum [133].
  • compositions comprising STn conjugates are used to prevent and/or treat infections with parasites expressing STn, for example Fasciola hepatica [131] or Trypanosoma cruzi [132].
  • compositions comprising Tn, STn, or TF conjugates may be used to prevent and/or treat viral infections resulting in host cell expression of Tn, STn, or TF.
  • compositions comprising Tn conjugates may be used to prevent and/or treat HIV-1, since HIV-1 infection of T cells has been reported to induce Tn-antigen expression in lymphocytes and anti-Tn antibodies have been shown to block the infection of lymphocytes by HIV-1.
  • compositions comprising STn conjugates may be used to prevent and/or treat H1N1, since infection with H1N1 influenza virus has been shown to cause upregulated expression of STn [IBS].
  • the subject is typically a mammal.
  • the mammal is preferably a human.
  • a preferred class of humans is humans at risk of developing a cancer or infectious disease as described above.
  • a preferred class of humans is humans having been diagnosed with a cancer or infectious disease as described above.
  • compositions of the invention will generally be administered directly to a patient.
  • Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue), or by rectal, oral, vaginal, topical, transdermal, intranasal, ocular, aural, pulmonary or other mucosal administration.
  • parenteral injection e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue
  • rectal oral, vaginal, topical, transdermal, intranasal, ocular, aural, pulmonary or other mucosal administration.
  • intramuscular administration is used, for example to the deltoid.
  • Administration may be in two parts, one in each deltoid.
  • Injection may be via a needle (e.g. a hypodermic needle), but needle-free injection may alternatively be used
  • Dosage treatment can be a single dose schedule or a multiple dose schedule. Typically, a multiple dose schedule is used. In examples, up to or about 7 doses are administered. As an example, doses may be administered on days 1, 4, 8, 22, 36, 50 and 64. Multiple doses may be used in a primary immunisation schedule and/or in a booster immunisation schedule. A primary dose schedule may be followed by a booster dose schedule. Suitable timing between priming doses (e.g. between 4-16 weeks), and between priming and boosting, can be routinely determined.
  • composition comprising X may consist exclusively of X or may include something additional e.g. X + Y.
  • x means, for example, x ⁇ 10%.
  • the word “substantially” does not exclude “completely” e.g. a composition which is “substantially free” from Y may be completely free from Y. Where necessary, the word “substantially” may be omitted from the definition of the invention.
  • a process comprising a step of mixing two or more components does not require any specific order of mixing. Thus components can be mixed in any order. Where there are three components then two components can be combined with each other, and then the combination may be combined with the third component, etc.
  • FIG. 1A shows the structures of Tn (GalNAc ⁇ 1 -), STn (Neu5Ac ⁇ 2,6 GalNAc ⁇ 1 ), and TF (Gal ⁇ 3 GalNAc ⁇ 1 -).
  • FIG. 1B shows the structures of exemplary glycoconjugates for use in the invention. Only Tn is shown, but STn or TF can also be used.
  • FIG. 1C shows the three glycoconjugates used in the exemplified GVI.
  • FIG.1B and FIG.1C only a single representative lysine of CRM197 is shown, for brevity.
  • FIG. 2 shows the chemical conjugation scheme for the GVI. The bioconjugation of Tn/STn/TF- PEG3-COOH to CRM197 is performed in a standard two step activation and conjugation procedure.
  • TACAs are activated with 1-ethyl-3-(-3-dimethylaminopropyl) carbodiimide hydrochloride (EDC) and N-Hydroxysuccinimide (NHS) a 1:2:2 ratio of TACA:EDC:NHS, in 0.1M 2-ethanesulfonic acid (MES) buffer pH 5.0 supplemented with 10-14% v/v dimethyl sulfoxide (DMSO), to make each respective NHS-ester.
  • MES 2-ethanesulfonic acid
  • DMSO dimethyl sulfoxide
  • FIGS. 3A and 3B show conjugation level testing by MALDI-TOF-MS.
  • FIG. 3A is a table showing the calculated number of modifications per CRM197 molecule for five lots of each of the three monovalent glycoconjugates.
  • FIG.3B shows results from in vivo potency testing of monovalent glycoconjugates.
  • C57BL/6 mice were immunised per group with monovalent formulations containing CRM197-PEG3-Tn/STn/TF 1 along with QS21 and ODN1826 adjuvants on day 0, 3, 7 and 21.
  • ELISA was performed with day 28 mouse sera on plates coated with TACAs (Tn-PAA, STn-PAA, or TF-PAA) and GMT's were calculated from IgG dilution curve.
  • FIGS. 4A and 4B show conjugation level testing by MALDI-TOF-MS, at different TACA:CRM97 ratios.
  • FIG. 4A is a table showing the calculated number of modifications for each monovalent glycoconjugate reacted with different mole:mole ratios of TACA:CRM197. As the ratio of glycoconjugates is reduced, there are less modifications observed on the final product.
  • FIG. 4B shows that lower TACA conjugation leads to decreased in vivo immune response. Groups of mice were immunised with monovalent glycoconjugates having different TACA densities along with QS21 and ODN1826. Day 28 sera was analysed by ELISA as above.
  • FIGS. 5A-5D show that GVI compositions induce potent and specific antibody responses in mouse models. IgG responses against Tn, STn and TF are illustrated by serial dilution curves of sera after immunisation with a GVI composition, lot 1, (FIGS. 5A-5C) and GMT for multiple lots (FIG. 5D). Groups of mice were immunised intramuscularly on days 0, 3, 7, 21, and 35 with a GVI composition containing CRM197-PEG3-Tn/STn/TF 1 glycoconjugates along with QS21 and ODN1826.
  • mice were bled 7 days after the fourth (day 28) and fifth (day 42) vaccinations and serological responses were analyzed by ELISA to determine the antibody titers against Tn, STn and TF as above.
  • IgG titration curves (FIGS. 5A-5C) and GMT's - day 28 (FIG. 5D) were calculated and plotted.
  • Anti-Tn (5F4 clone - SBH), anti-STn (3F1 clone - SBH), and biotinylated PNA lectin (Vector labs) served as positive controls (starting dilution 1 mg/ml) for Tn, STn and TF antigen respectively while Pre-bleed served as a negative control.
  • FIGS. 6A-6E show that GVI compositions induce tumour binding and tumouricidal IgG.
  • FIGS. 6A- 6C show surface binding of serially diluted sera. Mice immunised with 10, 5, 3.3 or 1 mg of the GVI along with QS21 and ODN1826 with Jurkat (FIG. 6A), Ov90 (FIG. 6B) and Colo201 (FIG. 6C) cells were evaluated by flow cytometry and mean fluorescence intensities (MFI) were plotted. "Pre” means pre-bleed, which served as a negative control.
  • Anti-B mAb refers to an anti-STn mAb, which served as positive control.
  • FIGS. 6D and 6E show that antibodies raised against the GVI mediate complement-dependent cytotoxicity (CDC) to kill Tn and STn containing tumour cells. Cytotoxicity was determined using the calcein AM based CDC assay. Anti-Tn and anti-STn monoclonal antibodies served as positive controls. FIGS. 6D and 6E show an average of 5 independent experiments.
  • FIG. 7 shows results from a glycan microarray study. Polyclonal sera from mice inoculated with trivalent GVI were screened against an O-linked glycan microarray and probed with a fluorescently labeled anti-mouse-lgG. X-axis represents glycan ID numbers while their abundance as detected by sera is plotted as relative fluorescence intensity on Y-axis. For controls and list of O-glycans printed on the array, see [134].
  • FIGS. 8A-8F show results from a long term stability study.
  • Lot 1 of each of the CRM197-PEG3-Tn/STn/TF 1 glycoconjugates was stored at 4°C as drug substances for 300+ days.
  • Each material was periodically removed, and a mock GVI produced mixing equal volumes of the three glycoconjugate.
  • the mock GVI was analyzed by SEC-UHPLC to determine the percentages of monomer, dimer, and HMW species (trimer, tetramer, and higher order).
  • a stable SEC profile was observed over time, suggesting that the drug substances are stable at 4°C over time.
  • FIG. 9 shows results of an adjuvant screening.
  • Mice were immunised with the trivalent GVI using a variety of different adjuvant mixed and formats, including liquid immunogen + liquid adjuvant mixes (QS21 alone, QS21 + ODN1826, QS21 + poly l:C, QS21 + poly l:C +ODN1826), liquid immunogen + liposome mixes (QS21+PHAD ® , QS21 + 3D-PHAD ® , QS21 + 3D-6A-PHAD ® , QS21 + MPLA, QS21 + MPLA + ODN1826), and immunogen/adjuvant absorbed to aluminum hydroxide "Alum" (Alum, Alum + QS21, Alum + QS21 + MPLA). Sera were collected at day 28 and ELISA performed, as above.
  • liquid immunogen + liquid adjuvant mixes QS21 alone, QS21 + ODN1826, QS21 + poly l:C, QS21 + poly l:
  • FIGS. 10A and 10B show the results from linker screening.
  • FIG. 10A shows IgG responses to different doses of serine or PEG3 linked conjugates. Animals were dosed with different amounts of trivalent GVIs with linkers comprising serine (see “CRM197-Ser-Tn 1" in FIG. IB; in CRM197-Ser-STn/TF 1, Tn is simply replaced with STn/TF) or PEG3 (see “CRM197-PEG3-Tn 1" in FIG. IB; in CRM197-PEG3-STn/TF 1, Tn is simply replaced with STn/TF).
  • FIG. 10B shows the detection of anti-linker antibodies.
  • This graph compares the level of IgG anti-PEG (i.e. anti- linker) antibodies induced by two CRM197-PEG3-Tn glycoconjugates having different conjugation chemistries (see “CRM197-PEG3-Tn 1” and “CRM197-PEG3-Tn 2” in FIG. 1B). The levels of antibodies were probed by ELISA.
  • the antigens Tn-PAA and mPEG4- modified bovine serum albumin (mPEG4-BSA) were used for coating.
  • the anti-sera from animals inoculated with CRM197-PEG3-Tn 2 have appreciable levels of anti-PEG antibodies, while the anti-sera from animals immunized CRM197-PEG3-Tn 1 do not have detectable anti-PEG antibodies.
  • An exemplary trivalent glycoconjugate vaccine intermediate (“GVI”) is a trivalent mixture of three different TACAs – the Tn antigen (GalNAc ⁇ 1 -), the STn antigen (Neu5Ac ⁇ 2,6 -GalNAc ⁇ 1 -) and the TF antigen (Gal ⁇ 3 GalNAc ⁇ 1 ) – each conjugated to a CRM197 carrier protein [135], [136].
  • Structural representation of the three glycoconjugates of the GVI is shown in FIG. 1C (only a single, representative TACA is shown on each CRM197).
  • each CRM197 molecule contains about 6 to about 15 TACAs per molecule.
  • Each glycoconjugate is manufactured separately and combined in a final formulation.
  • the three PEG3-COOH functionalised TACAs namely the Tn-PEG3-COOH (GalNAc ⁇ -O-PEG3- COOH), TF-PEG3-OOH (Gal ⁇ (1-3)GalNAc ⁇ -O-PEG3-COOH), and STn-PEG3-COOH (NeuAc ⁇ (2,6)GalNAc ⁇ -O-PEG3-COOH) compounds
  • Tn-PEG3-COOH GalNAc ⁇ -O-PEG3- COOH
  • TF-PEG3-OOH Ga ⁇ (1-3)GalNAc ⁇ -O-PEG3-COOH
  • STn-PEG3-COOH NeAc ⁇ (2,6)GalNAc ⁇ -O-PEG3-COOH
  • the amino acid sequence of the CRM197 used in these experiments was identical to the CRM197 used in approved prophylactic conjugates [137]. All non-custom materials used in the productions of each GVI, such as: the activating chemicals for conjugation (EDC (1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC)) and NHS (N- Hydroxysuccinimide)); buffering agents (MES (2-ethanesulfonic acid), sodium phosphate dibasic, and potassium phosphate monobasic); salts (potassium chloride, sodium chloride); and pH adjusting acids and bases (sodium hydroxide, hydrochloric acid), are all readily commercially available.
  • EDC activating chemicals for conjugation
  • NHS N- Hydroxysuccinimide
  • buffering agents MES (2-ethanesulfonic acid
  • sodium phosphate dibasic sodium phosphate dibasic
  • potassium phosphate monobasic sodium phosphate dibasic
  • salts potassium chloride
  • the PEG3 functionalised TACAs are rendered active and conjugated to CRM197 (a detailed chemical conjugation scheme is shown in FIG.2).
  • the conjugation chemistry is well established in the vaccine industry [138], [139], [140].
  • the residual chemicals of the reaction, DMSO, MES, NHS, and EDC-isourea and their by-products, are eliminated after buffer exchange.
  • each CRM197 molecule is conjugated to about 6 to about 15 TACAs per molecule.
  • FIG. 3A shows the calculated average number of TACAs per CRM197 molecule for five lots of each of the three glycoconjugates.
  • Results from a routine experiment are shown in FIGS. 5A-5C.
  • the first step was to determine the antibody titers against the three TACA targets using ELISA.
  • the TACAs are scaffolded on polyacrylamide (PAA) in this screening format so that the titers measured are solely against the TACA and not the CRM197 carrier protein and/or any other protein.
  • PAA polyacrylamide
  • a weak IgM response (average from 20 mice Tn - 11,840; STn - 4,230 and TF - 4,622) and a very strong and reproducible IgG response (average from 20 mice Tn - 623,582; STn - 1,534,852 and TF - 441,586) was observed against all three TACAs (Tn, STn and TF) at day 28.
  • Another booster dose increased IgG titers of TF to 760,672 as observed in IgG dilution curve against TF measured at day 42.
  • GVI compositions induced 100-500X higher IgG titers against Tn, STn and TF in mice than previous vaccine candidates described, e.g. [141], [142], [143], [144], [145], [146]. Also, the IgG responses observed are superior to pre-clinical and clinical candidates advanced previously. This is best shown by the differential IgG to IgM responses induced by GVI compositions. Induction of B cell class switching (from IgM to IgG) is a hallmark of vaccination and a demonstration of the anamnestic nature of secondary exposure to an antigen [147].
  • IgM production to IgG is associated with key immunological events including the generation of B cell memory, affinity maturation and production of exponentially higher titers of IgG over the initial IgM dominated response.
  • the literature clearly shows IgG titers are equivalent to IgM titers in experimental animals and patients [4], [5], [6], [51], [50], [141], [142], [143]. Both IgG and IgM were detected at ⁇ 1:10,000 GMT for the three antigens, even after repeated immunisations. These data indicate that the first- generation vaccines were sub-optimal, in that they cannot induce a classical vaccine response.
  • the IgG titers induced by GVI compositions in mouse models are 100-500X more than the specific IgM titers generated after 4 immunisations over 28 days (IgG > 1:100,000 GMT vs. IgM ⁇ 1:10,000 for all three glycan antigens).
  • GVI compositions are capable of inducing rapid, durable, boostable and high levels of circulating IgG against the aberrant tumour glycans, Tn, STn and TF.
  • the proposed mechanisms of GVI compositions induced IgG efficacy are the same as those for anti-tumour antibody therapeutics and vaccines, such as anti- Her2 or anti-CD20 Tx mAbs [148], [149]. Binding differentially to tumour over healthy tissue, the vaccine-induced anti-glycan antibodies target the tumour leading to cell death through diverse mechanisms. These include engagement of the patient's immune system through CDC, ADCC and ADCP and potentially direct signalling. The antibodies elicited by GVI compositions bind Tn, STn and TF in the absence of protein/peptide context. That is, they are targeting the glycan alone and not a single glycoprotein, per se. Thus, in immunological terms, the antibodies are hapten-specific [150], [151]. ii. Tumour cell binding and tumouricidal IgG
  • TLR3 agonists e.g. poly l:C
  • TLR9 agonists e.g. ODN1826
  • TLR4 agonists e.g. MPLA
  • Th2 adjuvants e.g. Alum
  • Th1/Th2 adjuvants e.g. QS21
  • Multiple different formulations were screened, including including liquid immunogen + liquid adjuvant mixes, liquid immunogen + liposome mixes, and immunogen/adjuvant absorbed to aluminum hydroxide "alum".
  • use of adjuvants can help maximise IgG titers.
  • linkers comprising serine are suitable alternatives to linkers comprising PEG3 (see FIG. 10A).
  • CRM197-PEG3-Tn/STn/TF conjugates may have different conjugations chemistries, for example see “CRM197-PEG3-Tn 1" and “CRM197-PEG3-Tn 2" in FIG. IB. Whilst CRM197-PEG3- Tn 1 and CRM197-PEG3-Tn 2 resulted in similar anti-Tn IgG levels, CRM197-PEG3-Tn 1 elicited significantly lower levels of anti-PEG (i.e., anti-linker) antibodies (FIG. 10B).
  • anti-PEG i.e., anti-linker
  • Conjugates comprising other linkers, e.g., other PEG groups (e.g., PEG4-PEG8), threonine, or alkane groups (e.g., C2-C8 alkanes), or other conjugation chemistries, are expected to achieve similar results.
  • linkers e.g., other PEG groups (e.g., PEG4-PEG8), threonine, or alkane groups (e.g., C2-C8 alkanes), or other conjugation chemistries

Landscapes

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

Abstract

The invention relates to immunogenic compositions comprising: a) a conjugate comprising the tumour-associated antigen (TACA) Tn covalently conjugated to a carrier protein; b) a conjugate comprising the TACA STn covalently conjugated to a carrier protein; and c) a conjugate comprising the TACA TF covalently conjugated to a carrier protein.

Description

IMMUNOGENIC COMPOSITIONS COMPRISING TUMOUR-ASSOCIATED ANTIGEN
RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application No. 63/208,207, filed June 8, 2021, the entire contents of which are incorporated herein by reference.
TECHNICAL FIELD
This invention is in the field of immunogenic compositions comprising conjugates of tumour- associated carbohydrate antigens (TACAs) and carrier proteins. The compositions are useful for treating cancer.
BACKGROUND ART
Cancer cells often express aberrant cell surface carbohydrates, which differentiate them from healthy cells. There is increasing evidence that these carbohydrates provide an immuno- suppressive shield for the tumour [1]. Thus, these tumour-associated carbohydrate antigens (TACAs) are promising therapeutic targets. However, as carbohydrates are T-independent antigens, they are poorly immunogenic and often only give rise to short-lived, IgM-based responses. Conjugation to a carrier protein can convert T-independent antigens into T- dependent antigens, thereby enhancing memory responses and allowing immunity to develop. Antibodies specific for TACAs should, in theory, slow growth and/or induce apoptosis of tumour cells.
There are two major classes of TACAs, namely glycoprotein antigens and glycolipid antigens. Glycoprotein antigens include the O-linked carbohydrates Thomsen-nouveau (Tn), sialyl-Tn (STn), and Thomsen-Friedenreich (TF). One or more of Tn, STn, and TF are expressed on the surface of most solid tumour types and are shared between diverse histologies and patient populations [2]. In detail, Tn is often overexpressed in breast, prostate, and stomach cancers, STn is overexpressed in breast, colon, lung, ovary, prostate, and stomach cancers, and TF is overexpressed in breast, colon, ovary, prostate, and stomach cancers [3].
Tn, STn, and TF are also associated with various infectious diseases [133], [132], [133].
Attempts to target TACAs with vaccines have shown pre-clinical promise. They have generally been well tolerated in clinical trials and induce anti-glycan antibodies in cancer patients. So far, however, there have been no clear clinical successes [4], [5], [6], [51], [50]. Thus, there remains a need for further and improved TACA conjugate vaccines. DISCLOSURE OF THE INVENTION In a first aspect, the invention provides conjugates comprising Tn, STn, or TF conjugated to a carrier protein. In a second aspect, the invention provides immunogenic compositions comprising Tn, STn, and TF, each conjugated to a carrier protein. The conjugates and immunogenic compositions may be used as vaccines for treating and/or preventing cancer. In another aspect, the invention provides a method for immunising a patient against cancer, the method comprising a step of administering to the patient an immunogenic composition of the first aspect of the invention. Immunogenic compositions In one embodiment, the invention provides an immunogenic composition comprising Tn conjugated to a carrier protein, STn conjugated to a carrier protein, and TF conjugated to a carrier protein. The inventors have found that these compositions may induce higher IgG responses than monovalent compositions (i.e. compositions comprising one of Tn, STn, or TF) and may not exhibit significant cross-reactivity. In some embodiments, the compositions comprise only one or two of these conjugates. For example, the compositions may be monovalent or bivalent. In some embodiments, the compositions comprise other conjugates, in particular conjugates having other TACAs. For example, the compositions may be tetravalent, pentavalent, hexavalent, etc. However, typically, the immunogenic compositions of the invention do not comprise any TACAs other than Tn, STn, and TF. In other words, the immunogenic compositions are typically trivalent. Tumour-associated carbohydrate antigens (TACAs) The present invention makes use of the TACAs Tn (GalNAcα1–), STn (Neu5Acα2,6GalNAcα1–), and TF (Galβ3GalNAcα1–) (see FIG. 1A). These three TACAs are closely related, chemically and biosynthetically. They occur naturally as O-linked glycans, meaning they link to the hydroxyl group of a serine or threonine residue of a protein. The inventors have selected these TACAs for use in the invention based on a variety of reasons. For example: they exhibit broad and high expression on a variety of solid tumours; they have strong tumour/normal differential expression compared to other tumour-associated antigens (TAAs) [7], [8], [9], [10]; Tn, STn, and TF expression in cancer is controlled by similar genetic lesions in the glycosylation machinery; the genes controlling their expression are under selection in many solid tumours; they are often co-expressed by a single tumour [10]; expression of these TACAs strongly correlates with poor outcomes in cancer patients. In some embodiments, the invention also make use of other TACAs, for example glycolipid TACAs (e.g. Globo series, gangliosides, and/or Lewis structure series). However, generally, the immunogenic compositions of the invention do not comprise any glycolipid TACAs. Typically, the immunogenic compositions of the invention do not comprise Globo-H. Preferably, the immunogenic compositions of the invention do not comprise any TACAs other than Tn, STn, and/or TF.
TACAs used in the invention are typically in their native form as described above. In some embodiments, TACAs are modified. In some embodiments, N- acyl modified Tn, STn, and/or TF are used, for example as described in [11], [12], or [13]. In some embodiments, /V-fluoroacetyl, /V-phenylacetyl and/or /V-chlorophenylacetyl modified Tn, STn, and/or TF are used [14]. In some embodiments, fluorine-substituted TACAs are used to improve ligand binding and/or enhance the immune response [12], [15]. In some embodiments, TF is modified to have a hydrophobic terminal residue such as benzyl, p-nitrophenyl, butyl, propyl, ethyl, or methyl, as described in [16]. In some embodiments, S-glycosides and C-glycosides are used to lengthen in vivo lifetime [15]. In some embodiments, further optional modifications include sulfation and acetylation. In some embodiments, TACA mimetics are used. In some embodiments, the conformationally constrained Tn mimetic described in [17] is used. Modified TACAs and mimetics may reduce immune tolerance, reduce TACA degradation, and/or increase immunogenicity. However, generally, the TACAs used in the invention are not modified.
In the invention, the ratio of the TACAs may vary. In some embodiments, the ratio (w/w) of each TACA to each other TACA is from about 5:1 to about 1:5, from about 4:1 to about 1:4, from about 3:1 to about 1:3; from about 2:1 to about 1:2, from about 1.5:1 to about 1:1.5, from about 1.4:1 to about 1:1.4, from about 1.3:1 to about 1:1.3, from about 1.2:1 to about 1:1.2, from about 1.1:1 to about 1:1.1. In particular embodiments, the ratio (w/w) of each TACA to each other TACA is about 1:1. In trivalent compositions (e.g. wherein the TACAs are Tn, STn, and TF) each TACA typically constitutes from about 15% to about 50%, from about 20% to about 45%, or from about 25% to about 40% (w/w) of the total TACAs. In a particular embodiment, each TACA constitutes from about 30% to about 35% (w/w) of the total number of TACAs. In a particular embodiment, in trivalent compositions, the three TACAs are present in a molecular ratio of about 1:1:1. Such compositions allow for easier manufacture. Tn, STn, and TF can be synthesised by techniques known in the art. TACAs and PEG3-COOH functionalised TACAs are available from Sussex Research Laboratories Inc. (Ottawa, ON, Canada). Carrier proteins
The invention provides conjugates that comprise TACAs (typically Tn, STn, and/or TF) conjugated to a carrier protein. In general, covalent conjugation of TACAs to carrier proteins enhances the immunogenicity of the TACAs as it converts them from T-independent antigens to T-dependent antigens, thus allowing priming for immunological memory. Conjugation to carrier proteins is a well-known technique in vaccinology.
The invention involves the use of carrier proteins, typically immunogenic carrier proteins. Useful carrier proteins include bacterial toxins or toxoids, such as diphtheria toxoid or tetanus toxoid. Fragments of toxins or toxoids can also be used e.g. fragment C of tetanus toxoid [18]. Cross-Reactive-Material-197 (CRM197) is a diphtheria toxin which has lost the ADP- ribosyltransferase activity of the native toxin as a result of genetic mutation [19-21]. In a particular embodiment of the invention, CRM197 is the carrier protein, because CRM197 is not itself glycosylated, the amines of CRM197 are amenable to modifications, and CRM197 has been used in approved prophylactic vaccines (e.g. Menveo® and Prevnar13® vaccines). Also, the inventors found that conjugates using CRM197 as the carrier protein for Tn, STn, and TF resulted in high levels of IgG compared to other carrier proteins. In some embodiments, CRM197 is a recombinant CRM197 because it is easy and efficient to synthesise. In some embodiments, recombinant CRM197 for use in the invention is produced in E. coli (including, for example, those described and produced in [22], [23], and [24]). In some embodiments, recombinant CRM197 for use in the invention is produced in a Pseudomonas host cell (including, for example, those described and produced in [25] and [26]). Recombinant CRM197 for use in the invention can also be produced using methods described in [27]. Other carrier proteins for use in the invention include Keyhole limpet haemocyanin (KLH) [28], the N. meningitidis outer membrane protein [29], synthetic peptides [30,31], heat shock proteins [32,33], pertussis proteins [34,35], cytokines [36], lymphokines [36], hormones [36], growth factors [36], human serum albumin (preferably recombinant), ovalbumin, artificial proteins comprising multiple human CD4+ T cell epitopes from various pathogen-derived antigens [37] such as N19 [38], protein D from H. influenzae [39,40], pneumococcal surface protein PspA [41], pneumolysin [42], iron-uptake proteins [43], toxin A or B from C.difficile [44], recombinant Pseudomonas aeruginosa exoprotein A (rEPA) [45], a GBS protein, etc.
In some embodiments, more than one type of carrier protein is used in the invention e.g. to reduce the risk of carrier suppression. For example, in some embodiments, different types of carrier proteins are used for different types of TACAs, e.g. Tn is conjugated to CRM197 while STn is conjugated to tetanus toxoid. In some embodiments, more than one type of carrier protein is used for a particular type of TACA, e.g. some Tn molecules are conjugated to CRM197 and other Tn molecules are conjugated to tetanus toxoid. In general, however, it is preferred to use the same carrier protein for all TACAs (typically CRM197).
Conjugates
In some embodiments, a single carrier protein molecule is conjugated to more than one type of TACA. For example, a single carrier protein is conjugated to two or three of Tn, STn, and TF. To achieve this goal, different TACAs are mixed prior to the conjugation reaction. In general, however, it is preferred that each carrier protein molecule is conjugated to a single type of TACA (e.g. Tn, STn, or TF). Such embodiments are preferred because it is easier to perform three separate conjugation reactions (each using a different type of TACA) and subsequently mix the conjugates, it enables the manufacturer to easily control the ratio of Tn:STn:TF in the final composition, and it enables the manufacturer to control the number of molecules of a given TACA present on each carrier (the TACA:carrier ratio). Thus, in some embodiments, the immunogenic compositions of the invention comprises a first conjugate comprising Tn conjugated to a first carrier protein, a second conjugate comprising STn conjugated to a second carrier protein, and a third conjugate comprising TF conjugated to a third carrier protein. Typically, the first, second, and third carrier proteins are all CRM197.
The precise number of TACAs per carrier molecule can vary. Generally, the average number of TACA molecules per carrier molecule is at least about 20% of the total number of accessible conjugation sites on the carrier molecule, in particular from about 20% to about 100% of the total number of accessible conjugation sites. Typically, the average number of TACA molecules per carrier is at least about 30% of the total number of accessible conjugation sites, in particular from about 30% to about 100% of the total number of accessible conjugation sites.
CRM197 contains approximately 40 sites that could be conjugated to a TACA, if the protein was fully denatured. However, in the native protein only approximately 20 of these sites are accessible to modification. Previous publications have described which lysines in CRM197 are most reactive [46]. In some embodiments wherein the carrier protein is a diphtheria toxin or toxoid, such as CRM197, the average number of TACA molecules per carrier molecule is typically at least about 4, in particular from about 4 to about 15. In some embodiments, the average number of TACA molecules per carrier molecule is at least about 6, in particular from about 6 to about 15. The inventors found that conjugates having these TACA:carrier ratios were highly immunogenic. In some embodiments, for CRM197-Tn conjugates, the average number of TACA molecules per carrier molecule is at least about 7. In some embodiments, for CRM197- STn conjugates, the average number of TACA molecules per carrier molecule is at least about 5. In some embodiments, for CRM197-TF conjugates, the average number of TACA molecules per carrier molecule is at least about 6, in particular from about 7 to about 10.
Linkers
TACAs may be conjugated to carrier proteins covalently or non-covalently. In some embodiments of the invention, the TACAs are conjugated to the carrier proteins covalently, e.g. via a linker. In other embodiments of the invention, the TACAs are non-covalently conjugated to the carrier proteins.
Generally in the art, to covalently conjugate a TACA to a carrier protein, the reducing end of the TACA is functionalised with a reactive moiety, for example p-nitrophenyl, maleimide, or an aldehyde-containing group, which is then conjugated to the carrier protein through amide bond formation, Michael addition or reductive amination [14]. However, some conjugation methods used in the art are inefficient, destroy/denature the TACA, or result in a linker between the TACA and the carrier protein which itself is immunogenic and elicits high levels of anti-linker antibodies.
In some embodiments, each TACA is conjugated to its carrier covalently via a linker which is non-immunogenic and/or elicits low levels of anti-linker antibodies (typically 1 or 2 orders of magnitude lower than anti-TACA antibodies).
In some embodiments, linkers comprise serine residues, threonine residues, polyethylene glycol (PEG) groups, alkane groups, thiol groups, maleimide groups, amide groups, thio-ether groups, thioester groups, ether groups, aminoxy groups, oxime groups, hydrazone groups, or combinations thereof.
In some embodiments, linkers comprise serine and/or threonine residues, e.g. when the TACA is O-linked to a serine or threonine residue. In some embodiments, the linker comprises a single serine residue. In some embodiments, the linker comprises a single threonine residue. In some embodiments, the linker comprises both serine and threonine residues. In some embodiments, the serine or threonine may be covalently bound to a lysine residue in the carrier protein via a maleimide-containing moiety. Linkers comprising serine and/or threonine are advantageous because TACAs are naturally found on serine and threonine residues, and they exhibit low immunogenicity. In some embodiments, linkers comprise an alkane group, i.e. -(CH¬2)n-, wherein n is typically from 2 to 8, from 2 to 6, or from 2 to 4. In some embodiments, n is 2, 3, 4, 5, 6, 7, or 8. In some embodiments, n is 4. In some embodiments, the TACA is O-linked to an alkane-carboxyl which is reacted with the amine group in the side chain of a lysine residue in the carrier protein. Alkane linkers are advantageous because they exhibit low immunogenicity.
In some embodiments, linkers comprise polyethylene glycol (PEG), i.e. -(O-CH2-CH2)n-, wherein n is typically from 2 to 12, from 3 to 10, from 3 to 8, or from 3 to 6, or preferably is 3 or 4. In some embodiments, n is 3, 4, 5, 6, 7, or 8. In some embodiments n is from 3 to 8. In some embodiments, n is from 3 to 6. In some embodiments, n is 3 or 4. In a particular embodiment, the linker comprises PEG-3 (i.e. -(O -CH2-CH2)n- wherein n is 3). PEG linkers are particularly advantageous because they exhibit low immunogenicity, low cross-reactivity, few side-effects, and allow for easy manufacture at the preferred TACA:carrier ratios.
In some embodiments, the TACA is O-linked to PEG(n)-carboxyl, which is reacted with the amine group in the side chain of a lysine residue in the carrier protein . An exemplary resulting conjugate is CRM197-PEG3-Tn 1 shown in FIG. IB. In some embodiments, the TACA is O-linked to PEG(n)-azide, PEG(n)-thiol, or PEG(n)-amine, which is reacted with a modified lysine residue in the carrier protein.
In these embodiments, the amine group in the side chain of the lysine may be modified with a bifunctional linker. For example, a homobifunctional linker of the formula X-L-X may be used, wherein the two X groups are the same as each other and can react with the azide, thiol, or amine in the O-linked TACA and the amine group in the side chain of the lysine, and wherein L is a linking moiety in the linker. In another example, a heterobifunctional linker of the formula X-L-X is used, wherein the two X groups are different and one can react with the azide, thiol, or amine in the O-linked TACA and the other can react with the amine group in the side chain of the lysine, and wherein L is a linking moiety in the linker. An exemplary X group is N- hydroxysuccinimide. Exemplary heterobifunctional crosslinkers are GMBS (N-y- maleimidobutyryloxysuccinimide ester) and Alkyne, Succinimidyl Ester (3- propargyloxypropanoic acid, succinimidyl ester). An exemplary resulting conjugate is CRM197- PEG3-Tn 2 shown in FIG. IB.
The use of PEG(n)-azide (e.g. in CRM197-PEG3-Tn 2 shown in FIG. IB) may result in a more immunogenic construct, but may also elicit higher levels of anti-PEG antibodies. In contrast, the use of PEG(n)-carboxyl (e.g. in CRM197-PEG3-Tn 1 shown in FIG. IB) may result in strongly immunogenic constructs without eliciting high levels of anti-PEG antibodies.
Exemplary linkers are shown in the conjugates in FIG. IB (only Tn conjugates are shown but any of Tn, STn, of TF could be used).
The invention also provides methods of producing conjugates (and conjugates obtainable by these methods), comprising (a) providing a TACA and a carrier molecule; (b) conjugating the TACA to a carrier molecule. Typically, the method comprises: (a) providing a PEG3-COOH functionalised TACA selected from Tn-PEG3-COOH, STn-PEG3-COOH, and TF-PEG3-COOH, and a CRM197; (b) conjugating the TACA to the CRM197 by (i) activating the PEG3-COOH functionalised TACA with 1-ethyl-3-(-3-dimethylaminopropyl) carbodiimide hydrochloride (EDC) and N-Hydroxysuccinimide (NHS), optionally at a 1:2:2 ratio of TACA:EDC:NHS in 0.1M 2- ethanesulfonic acid (MES) buffer pH 5.0 supplemented with 10-14% v/v dimethyl sulfoxide (DMSO) (alternatively, sulfo-NHS can be used so that DMSO is not required), to make each respective NHS-ester, and (ii) adding the reaction mixture to the CRM197, optionally in phosphate buffered saline in a 50:1 ratio of activated TACA:protein. A typical chemical conjugation scheme is depicted in FIG. 2. In some embodiments, the method further comprises c) filtering out reactants, biproducts, residual solvent and/or concentrate, optionally by performing buffer exchange on the reaction mixture, further optionally by dialysis or tangential flow filtration.
Other
In some embodiments, the TACAs used in the invention are conjugated to carrier proteins via peptide backbones, for example as described in [47], [48], [49], [50], or [51]. Thus, the TACAs may be in a clustered formation. However, generally, the TACAs used in the invention are not present on a peptide backbone. Thus, generally, the TACAs used in the invention are not in a clustered formation.
In some embodiments, the conjugates used in the invention comprise a polypeptide derived from a mucin, for example as described in [49], [52], [53], or [54]. As used herein, "a polypeptide derived from a mucin" means a polypeptide of 8 or more amino acids in length, having at least about 80% identity to a region of any of MUC1-22. However, generally, the conjugates used in the invention do not comprise a polypeptide derived from a mucin.
In some embodiments, the conjugates used in the invention comprise a T cell epitope, for example a PV (KLFAVWKITYKDT), a pan-DR epitope ("PADRE", aAKXVAAWTLKAa) as described in [55], and/or OVA323-339 (ISQAVHAAHAEINEAGR) as described in [14]. However, generally, the conjugates used in the invention do not comprise T cell epitopes.
In some embodiments, each of the conjugates used in the invention comprises or consists of a TACA linked to a carrier protein via a linker. In some embodiments, each of the conjugates used in the invention comprises or consists of a TACA selected from Tn, STn, and TF, linked to CRM197 via a linker comprising a PEG, more preferably PEG-3.
In some embodiments, the immunogenic composition of the invention comprises precisely two of the TACAs described herein, i.e. the composition is bivalent. In some embodiments, the composition comprises Tn conjugated to a carrier protein and STn conjugated to a carrier protein, since Tn and STn are very closely related and are usually present on the same tumour. In some embodiments, the composition comprises STn conjugated to a carrier protein and TF conjugated to a carrier protein, since STn can elicit antibodies against Tn. In some embodiments, the composition comprises Tn conjugated to a carrier protein and TF conjugated to a carrier protein.
In some embodiments, the immunogenic composition of the invention comprises only one of the TACAs described herein, i.e. the composition is monovalent. In some embodiments, the immunogenic composition comprises STn conjugated to a carrier protein, since the inventors have observed that STn alone can elicit a Tn and STn response (these TACAs are very closely related, chemically and biosynthetically).
Pharmaceutical compositions and uses
The immunogenic compositions of the invention may further comprise a pharmaceutically acceptable carrier. Typical pharmaceutically acceptable carriers include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition. Suitable carriers are typically large, slowly metabolised macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, sucrose [56], trehalose [57], lactose, and lipid aggregates (such as oil droplets or liposomes). Such carriers are well known in the art. The immunogenic compositions may also contain diluents, such as water, saline, glycerol, etc. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present. Sterile pyrogen-free, phosphate-buffered physiologic saline is a typical carrier. A thorough discussion of suitable pharmaceutically acceptable excipients is available in reference [58]. Compositions of the invention may be in aqueous form (i.e. solutions or suspensions) or in a dried form (e.g. lyophilised). If a dried immunogenic composition is used then it will be reconstituted into a liquid medium prior to injection. Lyophilisation of conjugate vaccines are known in the art. When the immunogenic compositions of the invention include conjugates comprising more than one type of TACA (e.g. Tn, STn, and TF), it is typical for the conjugates to be prepared separately, mixed, and then lyophilised. In this way, lyophilised compositions comprising multiple (preferably three) conjugates as described herein may be prepared. To stabilise conjugates during lyophilisation, it may be preferred to include a sugar alcohol (e.g. mannitol) and/or a disaccharide (e.g. sucrose or trehalose).
Compositions may be presented in vials, or they may be presented in ready-filled syringes. The syringes may be supplied with or without needles. A syringe will include a single dose of the composition, whereas a vial may include a single dose or multiple doses.
Aqueous compositions of the invention are also suitable for reconstituting other vaccines from a lyophilised form. Where a composition of the invention is to be used for such extemporaneous reconstitution, the invention provides a kit, which may comprise two vials, or may comprise one ready-filled syringe and one vial, with the contents of the syringe being used to reactivate the contents of the vial prior to injection.
Compositions of the invention may be packaged in unit dose form or in multiple dose form. For multiple dose forms, vials are preferred to pre-filled syringes. Effective dosage volumes can be routinely established, but a typical human dose of the composition has a volume of 0.5ml e.g. for intramuscular injection.
The pH of the composition is preferably between 6 and 8, preferably about 7. Stable pH may be maintained by the use of a buffer. The immunogenic compositions of the invention typically comprise disodium monohydrogen phosphate and potassium dihydrogen phosphate buffer. If a composition comprises an aluminium hydroxide salt, it is preferred to use a histidine buffer [59]. The composition may be sterile and/or pyrogen-free. Compositions of the invention may be isotonic with respect to humans.
Compositions of the invention are immunogenic, and are more preferably vaccine compositions. Vaccines according to the invention may either be prophylactic (i.e. to prevent cancer) or therapeutic (i.e. to treat cancer), but will typically be therapeutic. Immunogenic compositions used as vaccines comprise an immunologically effective amount of each antigen, as well as any other components, as needed. By "immunologically effective amount", it is meant that the administration of that amount to a subject, either in a single dose or as part of a series, is effective for treatment and/or prevention. This amount varies depending upon the taxonomic group of subject to be treated (e.g. non-human primate, primate, etc.), the health and physical condition of the subject to be treated, age, the capacity of the subject immune system to synthesise antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
Cancer affects various tissues of the body and so the compositions of the invention may be prepared in various forms. For example, the compositions may be prepared as injectables, either as liquid solutions or suspensions. The composition may be prepared for pulmonary administration e.g. as an inhaler, using a fine powder or a spray. The composition may be prepared as a suppository or pessary. The composition may be prepared for nasal, aural or ocular administration e.g. as spray, drops, gel or powder [e.g. refs 60 & 61]. Compositions of the invention may include an antimicrobial, particularly when packaged in multiple dose format.
Compositions of the invention may comprise detergent e.g. a Tween (polysorbate), such as Tween 80. Detergents are generally present at low levels e.g. <0.01%.
Compositions of the invention may include sodium salts (typically sodium chloride and potassium chloride) to give tonicity. A concentration of 10±2mg/ml NaCI is typical.
Compositions of the invention will generally include one or more buffers. Potassium phosphate monobasic buffers and sodium phosphate dibasic buffers are typical.
The invention also provides methods of producing pharmaceutical compositions comprising combining two or more conjugates of the invention. Typically, the method comprises combining: a) a first conjugate comprising Tn conjugated to a first carrier protein; b) a second conjugate comprising STn conjugated to a second carrier protein; and c) a third conjugate comprising TF conjugated to a third carrier protein, optionally with a pharmaceutically acceptable carrier. Typically, conjugates a)-c) are combined in a ratio (w/w) of about 1:1:1. Preferably, the first, second, and third carrier proteins are each CRM197. Adjuvants
Compositions of the invention may be administered in conjunction with other immunoregulatory agents. In particular, compositions may include one or more adjuvants described in detail below.
Mineral-containing compositions
Mineral containing compositions suitable for use as adjuvants in the invention include mineral salts, such as aluminium salts and calcium salts (or mixtures thereof). Calcium salts include calcium phosphate (e.g. the "CAP" particles disclosed in [62]). Aluminium salts include hydroxides, phosphates, sulfates, etc., with the salts taking any suitable form (e.g. gel, crystalline, amorphous, etc.). Adsorption to these salts is preferred. The mineral containing compositions may also be formulated as a particle of metal salt [63].
The adjuvants known as aluminium hydroxide and aluminium phosphate may be used. These names are conventional, but are used for convenience only, as neither is a precise description of the actual chemical compound which is present (e.g. see chapter 9 of [64]). The invention can use any of the "hydroxide" or "phosphate" adjuvants that are in general use as adjuvants. The adjuvants known as "aluminium hydroxide" are typically aluminium oxyhydroxide salts, which are usually at least partially crystalline. The adjuvants known as "aluminium phosphate" are typically aluminium hydroxyphosphates, often also containing a small amount of sulfate ( i.e . aluminium hydroxyphosphate sulfate). They may be obtained by precipitation, and the reaction conditions and concentrations during precipitation influence the degree of substitution of phosphate for hydroxyl in the salt. In examples, Alhydrogel® may be used as an adjuvant.
Oil Emulsions
Oil emulsion compositions suitable for use as adjuvants in the invention include oil-in-water emulsions, such as AS03 or the nanoemulsion AddaS03™ (AS03), and squalene-water emulsions, such as MF59 (5% Squalene, 0.5% Tween 80, and 0.5% Span 85, formulated into submicron particles using a microfluidizer) (Chapter 10 of [64]; see also [65]-[67]).
Particularly preferred adjuvants for use in the compositions are submicron oil-in-water emulsions. Submicron oil-in-water emulsions, methods of making the same and immunostimulating agents, such as muramyl peptides, for use in the compositions, are described in detail in [65] & [68]-[69].
Complete Freund's adjuvant (CFA) and incomplete Freund's adjuvant (IFA) may also be used as adjuvants in the invention. Saponin formulations
Saponin formulations such as those described in (chapter 22 of[126]) may also be used as adjuvants in the invention. Saponins are a heterologous group of sterol glycosides and triterpenoid glycosides that are found in the bark, leaves, stems, roots and even flowers of a wide range of plant species. Saponins isolated from the bark of the Quillaia soponorio Molina tree have been widely studied as adjuvants. Saponin can also be commercially obtained from Smilax ornata (sarsaparilla), Gypsophilla paniculata (brides veil), and Saponaria officianalis (soap root). Saponin adjuvant formulations include purified formulations, such as QS21, as well as lipid formulations, such as ISCOMs.
Saponin compositions have been purified using HPLC and RP-HPLC. Specific purified fractions using these techniques have been identified, including QS7, QS17, QS18, QS21, QH-A, QH-B and QH-C. Preferably, the saponin is QS21. A method of production of QS21 is disclosed in[70]. Saponin formulations may also comprise a sterol, such as cholesterol [71].
Combinations of saponins and cholesterols can be used to form unique particles called immunostimulating complexes (ISCOMs) (chapter 23 of [64]). ISCOMs typically also include a phospholipid such as phosphatidylethanolamine or phosphatidylcholine. Any known saponin can be used in ISCOMs. Preferably, the ISCOM includes one or more of QuilA, QHA and QHC. ISCOMs are further described in [71]-[73]. Optionally, the ISCOMS may be devoid of additional detergent(s) [74].
A review of the development of saponin based adjuvants can be found in [75] & [76].
Virosomes and virus-like particles Virosomes and virus-like particles (VLPs) can also be used as adjuvants in the invention. These structures generally contain one or more proteins from a virus optionally combined or formulated with a phospholipid. They are generally non-pathogenic, non-replicating and generally do not contain any of the native viral genome. The viral proteins may be recombinantly produced or isolated from whole viruses. These viral proteins suitable for use in virosomes or VLPs include proteins derived from influenza virus (such as HA or NA), Hepatitis B virus (such as core or capsid proteins), Hepatitis E virus, measles virus, Sindbis virus, Rotavirus, Foot-and-Mouth Disease virus, Retrovirus, Norwalk virus, human Papilloma virus, HIV, RNA- phages, Qß-phage (such as coat proteins), GA-phage, fr-phage, AP205 phage, and Ty (such as retrotransposon Ty protein pi). VLPs are discussed further in[77]-[82]. Virosomes are discussed further in, for example [83]. Bacterial or microbial derivatives
Adjuvants suitable for use in the invention include bacterial or microbial derivatives such as non-toxic derivatives of enterobacterial lipopolysaccharide (LPS), Lipid A derivatives, immunostimulatory oligonucleotides and ADP-ribosylating toxins and detoxified derivatives thereof.
Non-toxic derivatives of LPS include monophosphoryl lipid A (MPLA) and 3-O-deacylated MPL (3dMPL). 3dMPL is a mixture of 3 de-O-acylated MPLA with 4, 5 or 6 acylated chains. A preferred "small particle" form of 3 De-O-acylated MPLA is disclosed in [84]. Such "small particles" of 3dMPL are small enough to be sterile filtered through a 0.22μm membrane [84]. Other non-toxic LPS derivatives include MPLA mimics, such as aminoalkyl glucosaminide phosphate derivatives e.g. RC-529 [85,86]. In some embodiments, the MPLA analog/derivative is selected from Monophosphoryl 3-Deacyl Lipid A and Monophosphoryl Hexa-acyl Lipid A, 3- Deacyl (e.g. PHAD®, 3D-PHAD®, and 3D-6A-PHAD® - Avanti Polar Lipids). .
Lipid A derivatives include derivatives of lipid A from Escherichia coli such as OM-174. OM-174 is described for example in [87] & [88].
Immunostimulatory oligonucleotides suitable for use as adjuvants in the invention include nucleotide sequences containing a CpG motif (a dinucleotide sequence containing an unmethylated cytosine linked by a phosphate bond to a guanosine). Double-stranded RNAs and oligonucleotides containing palindromic or poly(dG) sequences have also been shown to be immunostimulatory.
The CpG's can include nucleotide modifications/analogs such as phosphorothioate modifications and can be double-stranded or single-stranded. [89], [90] and [91] disclose possible analog substitutions e.g. replacement of guanosine with 2'-deoxy-7-deazaguanosine. The adjuvant effect of CpG oligonucleotides is further discussed in [92]-[97].
The CpG sequence may be directed to TLR9, such as the motif GTCGTT or TTCGTT [98]. The CpG sequence may be specific for inducing a Th1 immune response, such as a CpG-A ODN, or it may be more specific for inducing a B cell response, such a CpG-B ODN. CpG-A and CpG-B ODNs are discussed in refs. 99-101. Preferably, the CpG is a CpG-B ODN, for example ODN1826 may be used in mice and ODN7909 (AKA ODN2006) may be used in humans.
Preferably, the CpG oligonucleotide is constructed so that the 5' end is accessible for receptor recognition. Optionally, two CpG oligonucleotide sequences may be attached at their 3' ends to form "immunomers". See, for example, [98] & [102]-[104]. Bacterial ADP-ribosylating toxins and detoxified derivatives thereof may be used as adjuvants in the invention. Preferably, the protein is derived from E.coli (E.coli heat labile enterotoxin "LT"), cholera ("CT"), or pertussis ("PT"). The use of detoxified ADP-ribosylating toxins as mucosal adjuvants is described in ref. 105 and as parenteral adjuvants in ref. 106. The toxin or toxoid is preferably in the form of a holotoxin, comprising both A and B subunits. Preferably, the A subunit contains a detoxifying mutation; preferably the B subunit is not mutated. Preferably, the adjuvant is a detoxified LT mutant such as LT-K63, LT-R72, and LT-G192. The use of ADP- ribosylating toxins and detoxified derivatives thereof, particularly LT-K63 and LT-R72, as adjuvants can be found in refs. 107-114. Numerical reference for amino acid substitutions is preferably based on the alignments of the A and B subunits of ADP-ribosylating toxins set forth in [115].
Pam3CSK4 (Pam3CysSerLys4) may also be used as an adjuvant. It is a synthetic lipopeptide and a TLR2/TLR2 ligand and mimics the acylated amino terminus of bacterial lipopeptides.
Trehalose-6, 6-dibehenate (TDB) may also be used as an adjuvant. It is a synthetic analog of trehalose-6, 6-dimycolate (TDM, also known as cord factor), which is a well-known immunostimulatory component of M. tuberculosis. TDB binds the C-Type lectin, Mincle.
Human immunomodulators
Human immunomodulators suitable for use as adjuvants in the invention include cytokines, such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12 [116], etc.) [117], interferons (e.g. interferon-γ), macrophage colony stimulating factor, and tumour necrosis factor.
Bioadhesives and Mucoadhesives
Bioadhesives and mucoadhesives may also be used as adjuvants in the invention. Suitable bioadhesives include esterified hyaluronic acid microspheres [118] or mucoadhesives such as cross-linked derivatives of poly(acrylic acid), polyvinyl alcohol, polyvinyl pyrollidone, polysaccharides and carboxymethylcellulose. Chitosan and derivatives thereof may also be used as adjuvants in the invention [119].
Microparticles
Microparticles may also be used as adjuvants in the invention. Microparticles (i.e. a particle of ~100nm to ~150μm in diameter, more preferably ~200nm to ~30μm in diameter, and most preferably ~500nm to ~10μm in diameter) formed from materials that are biodegradable and non-toxic (e.g. a poly(a-hydroxy acid), a polyhydroxybutyric acid, a polyorthoester, a polyanhydride, a polycaprolactone, etc.), with poly(lactide-co-glycolide) are preferred, optionally treated to have a negatively-charged surface (e.g. with SDS) or a positively-charged surface (e.g. with a cationic detergent, such as CTAB).
Liposomes
Examples of liposome formulations suitable for use as adjuvants are described in [120]-[121] and Chapters 13 & 14 of [64].
Typically, cholesterol liposomes are used. For example, liposomes may be composed of the phospholipid 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and cholesterol. Liposomes provide a stable solution and consistent doses of adjuvants. In addition, liposomes can impart better injection site and patient tolerance.
Polyoxyethylene ether and polyoxyethylene ester formulations Adjuvants suitable for use in the invention include polyoxyethylene ethers and polyoxyethylene esters [122]. Such formulations further include polyoxyethylene sorbitan ester surfactants in combination with an octoxynol [123] as well as polyoxyethylene alkyl ethers or ester surfactants in combination with at least one additional non-ionic surfactant such as an octoxynol [124]. Preferred polyoxyethylene ethers are selected from the following group: polyoxyethylene-9-lauryl ether (laureth 9), polyoxyethylene-9-steoryl ether, polyoxytheylene-8- steoryl ether, polyoxyethylene-4-lauryl ether, polyoxyethylene-35-lauryl ether, and polyoxyethylene-23-lauryl ether.
Polyphosphazene (PCPP)
PCPP formulations are described, for example, in [125] and [126].
Muramyl peptides
Examples of muramyl peptides suitable for use as adjuvants in the invention include N-acetyl- muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), and N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1'-2'-dipalmitoyl-sn- glycero-3-hydroxyphosphoryloxy)-ethylamine MTP-PE).
Imidazoquinolone Compounds.
Examples of imidazoquinolone compounds suitable for use adjuvants in the invention include Imiquamod and its homologues ( e,g . "Resiquimod 3M"), described further in [127] and [128].
Thiosemicarbazone Compounds.
Examples of thiosemicarbazone compounds, as well as methods of formulating, manufacturing, and screening for compounds all suitable for use as adjuvants in the invention include those described in [129]. The thiosemicarbazones are particularly effective in the stimulation of human peripheral blood mononuclear cells for the production of cytokines, such as TNF-a.
Tryptanthrin Compounds.
Examples of tryptanthrin compounds, as well as methods of formulating, manufacturing, and screening for compounds all suitable for use as adjuvants in the invention include those described in [130]. The tryptanthrin compounds are particularly effective in the stimulation of human peripheral blood mononuclear cells for the production of cytokines, such as TNF-a.
Poly l:C
Polyinosinic:polycytidylic acid (poly l:C) may be used as an adjuvant. Poly l:C is a synthetic analog of dsRNA and interacts with TLR3.
Combinations
The invention may also comprise combinations of aspects of one or more of the adjuvants identified above. For example, two or more adjuvants may be used in compositions of the invention.
Methods of treatment
The invention also provides a method for raising an immune response in a subject, comprising administering a conjugate or composition of the invention to the subject. The immune response is preferably preventative (prophylactic) and/or therapeutic, and preferably involves antibodies. The method may raise a booster response.
The invention also provides a conjugate or composition of the invention for use as a medicament or for use in a method of prevention/treatment. The medicament or method is preferably able to raise an immune response in a subject (i.e. it is an immunogenic composition) and is more preferably a vaccine.
The invention also provides the use of a composition of the invention in the manufacture of a medicament for raising an immune response in a subject.
The method may comprise further administering a checkpoint inhibitor to the subject. Thus, the invention also provides a checkpoint inhibitor for use in a method of treatment, wherein the method comprises administering a conjugate or composition of the invention. Suitable checkpoint inhibitors include an anti-PD-1 or anti-PD-L1 antibody, for example nivolumab, pembrolizumab, and/or atezolizumab. Broadly active checkpoint inhibitors allow for a more antigen-specific response induced by a vaccine of the invention to target tumours more effectively, and may exhibit synergy with the vaccine. Checkpoint inhibitors may be administered simultaneously or at different time points from the conjugate or composition of the invention. Checkpoint inhibitors may be administered as part of the same composition as the conjugate or composition of the invention or separately.
Typically, these methods and uses are for the prevention and/or treatment of cancer (i.e. a malignant neoplasm). Typically, the cancer is a solid tumour. Typically, the cancer is an epithelial cancer or carcinoma, for example an adenocarcinoma or a squamous cell carcinoma. Preferably, the cancer is pancreatic cancer, breast cancer, colon cancer, gastro-intestinal cancer, prostate cancer, lung cancer, ovarian cancer, or stomach cancer, because these types of cancer have been associated with high expression of Tn, STn, and TF. The cancer may be stage 0, I, II, III, or IV. The cancer may be metastatic cancer. The cancer may be relapsed or refractory to other treatments.
The cancer may comprise cancer cells expressing one or more of Tn, STn, and TF, for example two of Tn, STn, and TF, on the cell surface. The cancer cells may overexpress one or more of Tn, STn, and TF, for example two of Tn, STn, and TF, compared to a reference expression level or control sample. A control sample may be healthy (i.e. non-cancer) cells, which may be tissue- matched and/or patient-matched.
Thus, the method may comprise determining the level of one or more of Tn, STn, and TF expression on cancer cells obtained from the subject. The method may comprise identifying the subject as being suitable for treatment if one or more of Tn, STn, and TF is expressed on the cancer cells or is overexpressed on the cancer cells compared to a reference expression level or control sample.
Alternatively, these methods and uses are for the prevention and/or treatment of an infectious diseases. In some embodiments, compositions comprising Tn, STn, or TF conjugates are used to prevent and/or treat infections with parasites expressing Tn, STn, or TF. In some embodiments, compositions comprising Tn conjugates are used to prevent and/or treat infections with parasites expressing Tn, for example Fasciola hepatica [131], Schistostoma mansoni, Echinococcus granulosus, or Cryptosporidium parvum [133]. In some embodiments, compositions comprising STn conjugates are used to prevent and/or treat infections with parasites expressing STn, for example Fasciola hepatica [131] or Trypanosoma cruzi [132]. In some embodiments, compositions comprising Tn, STn, or TF conjugates may be used to prevent and/or treat viral infections resulting in host cell expression of Tn, STn, or TF. For example, compositions comprising Tn conjugates may be used to prevent and/or treat HIV-1, since HIV-1 infection of T cells has been reported to induce Tn-antigen expression in lymphocytes and anti-Tn antibodies have been shown to block the infection of lymphocytes by HIV-1. As another example, compositions comprising STn conjugates may be used to prevent and/or treat H1N1, since infection with H1N1 influenza virus has been shown to cause upregulated expression of STn [IBS].
The subject is typically a mammal. The mammal is preferably a human. Where the immunogenic composition is for prophylactic use, a preferred class of humans is humans at risk of developing a cancer or infectious disease as described above. Where the immunogenic composition is for therapeutic use, a preferred class of humans is humans having been diagnosed with a cancer or infectious disease as described above.
Compositions of the invention will generally be administered directly to a patient. Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue), or by rectal, oral, vaginal, topical, transdermal, intranasal, ocular, aural, pulmonary or other mucosal administration. Typically, intramuscular administration is used, for example to the deltoid. Administration may be in two parts, one in each deltoid. Injection may be via a needle (e.g. a hypodermic needle), but needle-free injection may alternatively be used.
Dosage treatment can be a single dose schedule or a multiple dose schedule. Typically, a multiple dose schedule is used. In examples, up to or about 7 doses are administered. As an example, doses may be administered on days 1, 4, 8, 22, 36, 50 and 64. Multiple doses may be used in a primary immunisation schedule and/or in a booster immunisation schedule. A primary dose schedule may be followed by a booster dose schedule. Suitable timing between priming doses (e.g. between 4-16 weeks), and between priming and boosting, can be routinely determined.
General
The term "comprising" encompasses "including" as well as "consisting" e.g. a composition "comprising" X may consist exclusively of X or may include something additional e.g. X + Y.
The term "about" in relation to a numerical value x means, for example, x±10%. The word “substantially” does not exclude “completely” e.g. a composition which is “substantially free” from Y may be completely free from Y. Where necessary, the word “substantially” may be omitted from the definition of the invention. Unless specifically stated, a process comprising a step of mixing two or more components does not require any specific order of mixing. Thus components can be mixed in any order. Where there are three components then two components can be combined with each other, and then the combination may be combined with the third component, etc. Unless otherwise stated, identity between polypeptide sequences is preferably determined by the Smith-Waterman homology search algorithm as implemented in the MPSRCH program (Oxford Molecular), using an affine gap search with parameters gap open penalty=12 and gap extension penalty=1. BRIEF DESCRIPTION OF DRAWINGS FIG. 1A shows the structures of Tn (GalNAcα1-), STn (Neu5Acα2,6GalNAcα1), and TF (Galβ3GalNAcα1-). FIG. 1B shows the structures of exemplary glycoconjugates for use in the invention. Only Tn is shown, but STn or TF can also be used. FIG. 1C shows the three glycoconjugates used in the exemplified GVI. In FIG.1B and FIG.1C only a single representative lysine of CRM197 is shown, for brevity. FIG. 2 shows the chemical conjugation scheme for the GVI. The bioconjugation of Tn/STn/TF- PEG3-COOH to CRM197 is performed in a standard two step activation and conjugation procedure. PEG3-COOH TACAs are activated with 1-ethyl-3-(-3-dimethylaminopropyl) carbodiimide hydrochloride (EDC) and N-Hydroxysuccinimide (NHS) a 1:2:2 ratio of TACA:EDC:NHS, in 0.1M 2-ethanesulfonic acid (MES) buffer pH 5.0 supplemented with 10-14% v/v dimethyl sulfoxide (DMSO), to make each respective NHS-ester. The reaction mixture is added to the CRM197 in phosphate buffered saline in a 50:1 ratio of activated TACA:protein, in a stirred cell. The reaction mixture is buffer exchanged via dialysis or tangential flow filtration to buffer exchange, filter out reactants, biproducts, residual solvent and concentrate. FIGS. 3A and 3B show conjugation level testing by MALDI-TOF-MS. FIG. 3A is a table showing the calculated number of modifications per CRM197 molecule for five lots of each of the three monovalent glycoconjugates. FIG.3B shows results from in vivo potency testing of monovalent glycoconjugates. C57BL/6 mice were immunised per group with monovalent formulations containing CRM197-PEG3-Tn/STn/TF 1 along with QS21 and ODN1826 adjuvants on day 0, 3, 7 and 21. ELISA was performed with day 28 mouse sera on plates coated with TACAs (Tn-PAA, STn-PAA, or TF-PAA) and GMT's were calculated from IgG dilution curve.
FIGS. 4A and 4B show conjugation level testing by MALDI-TOF-MS, at different TACA:CRM97 ratios. FIG. 4A is a table showing the calculated number of modifications for each monovalent glycoconjugate reacted with different mole:mole ratios of TACA:CRM197. As the ratio of glycoconjugates is reduced, there are less modifications observed on the final product. FIG. 4B shows that lower TACA conjugation leads to decreased in vivo immune response. Groups of mice were immunised with monovalent glycoconjugates having different TACA densities along with QS21 and ODN1826. Day 28 sera was analysed by ELISA as above.
FIGS. 5A-5D show that GVI compositions induce potent and specific antibody responses in mouse models. IgG responses against Tn, STn and TF are illustrated by serial dilution curves of sera after immunisation with a GVI composition, lot 1, (FIGS. 5A-5C) and GMT for multiple lots (FIG. 5D). Groups of mice were immunised intramuscularly on days 0, 3, 7, 21, and 35 with a GVI composition containing CRM197-PEG3-Tn/STn/TF 1 glycoconjugates along with QS21 and ODN1826. Mice were bled 7 days after the fourth (day 28) and fifth (day 42) vaccinations and serological responses were analyzed by ELISA to determine the antibody titers against Tn, STn and TF as above. IgG titration curves (FIGS. 5A-5C) and GMT's - day 28 (FIG. 5D) were calculated and plotted. Anti-Tn (5F4 clone - SBH), anti-STn (3F1 clone - SBH), and biotinylated PNA lectin (Vector labs) served as positive controls (starting dilution 1 mg/ml) for Tn, STn and TF antigen respectively while Pre-bleed served as a negative control.
FIGS. 6A-6E show that GVI compositions induce tumour binding and tumouricidal IgG. FIGS. 6A- 6C show surface binding of serially diluted sera. Mice immunised with 10, 5, 3.3 or 1 mg of the GVI along with QS21 and ODN1826 with Jurkat (FIG. 6A), Ov90 (FIG. 6B) and Colo201 (FIG. 6C) cells were evaluated by flow cytometry and mean fluorescence intensities (MFI) were plotted. "Pre" means pre-bleed, which served as a negative control. "Anti-B mAb" refers to an anti-STn mAb, which served as positive control. FIGS. 6D and 6E show that antibodies raised against the GVI mediate complement-dependent cytotoxicity (CDC) to kill Tn and STn containing tumour cells. Cytotoxicity was determined using the calcein AM based CDC assay. Anti-Tn and anti-STn monoclonal antibodies served as positive controls. FIGS. 6D and 6E show an average of 5 independent experiments. FIG. 7 shows results from a glycan microarray study. Polyclonal sera from mice inoculated with trivalent GVI were screened against an O-linked glycan microarray and probed with a fluorescently labeled anti-mouse-lgG. X-axis represents glycan ID numbers while their abundance as detected by sera is plotted as relative fluorescence intensity on Y-axis. For controls and list of O-glycans printed on the array, see [134].
FIGS. 8A-8F show results from a long term stability study. For FIGS. 8A-8C, Lot 1 of each of the CRM197-PEG3-Tn/STn/TF 1 glycoconjugates was stored at 4°C as drug substances for 300+ days. Each material was periodically removed, and a mock GVI produced mixing equal volumes of the three glycoconjugate. The mock GVI was analyzed by SEC-UHPLC to determine the percentages of monomer, dimer, and HMW species (trimer, tetramer, and higher order). A stable SEC profile was observed over time, suggesting that the drug substances are stable at 4°C over time. For FIGS. 8D-8F, these materials were also incorporated into in vivo studies using four 6 weeks old female C57BL/6 mice per group, with animals inoculated with a trivalent GVI ("ABC") or monovalent formulations ("A", "B", and "C" represent Tn, STn, and TF, respectively), all with QS21 and ODN1826, inoculated at days = 0, 3, 7, 21, 35, serum collected on day 28 and 42 and subsequently analyzed by ELISA for IgG titer against each antigen.
FIG. 9 shows results of an adjuvant screening. Mice were immunised with the trivalent GVI using a variety of different adjuvant mixed and formats, including liquid immunogen + liquid adjuvant mixes (QS21 alone, QS21 + ODN1826, QS21 + poly l:C, QS21 + poly l:C +ODN1826), liquid immunogen + liposome mixes (QS21+PHAD®, QS21 + 3D-PHAD®, QS21 + 3D-6A-PHAD®, QS21 + MPLA, QS21 + MPLA + ODN1826), and immunogen/adjuvant absorbed to aluminum hydroxide "Alum" (Alum, Alum + QS21, Alum + QS21 + MPLA). Sera were collected at day 28 and ELISA performed, as above.
FIGS. 10A and 10B show the results from linker screening. FIG. 10A shows IgG responses to different doses of serine or PEG3 linked conjugates. Animals were dosed with different amounts of trivalent GVIs with linkers comprising serine (see "CRM197-Ser-Tn 1" in FIG. IB; in CRM197-Ser-STn/TF 1, Tn is simply replaced with STn/TF) or PEG3 (see "CRM197-PEG3-Tn 1" in FIG. IB; in CRM197-PEG3-STn/TF 1, Tn is simply replaced with STn/TF). The anti-sera was probed via ELISA for total IgG on plates coated with Tn-PAA, STn-PAA, or TF-PAA. The GMTs were measured by serial dilution of test samples compared to a standard. FIG. 10B shows the detection of anti-linker antibodies. This graph compares the level of IgG anti-PEG (i.e. anti- linker) antibodies induced by two CRM197-PEG3-Tn glycoconjugates having different conjugation chemistries (see “CRM197-PEG3-Tn 1” and “CRM197-PEG3-Tn 2” in FIG. 1B). The levels of antibodies were probed by ELISA. The antigens Tn-PAA and mPEG4- modified bovine serum albumin (mPEG4-BSA) were used for coating. The anti-sera from animals inoculated with CRM197-PEG3-Tn 2 have appreciable levels of anti-PEG antibodies, while the anti-sera from animals immunized CRM197-PEG3-Tn 1 do not have detectable anti-PEG antibodies. MODES FOR CARRYING OUT THE INVENTION EXAMPLE 1 - Production of exemplary glycoconjugates An exemplary trivalent glycoconjugate vaccine intermediate (“GVI”) is a trivalent mixture of three different TACAs – the Tn antigen (GalNAcα1-), the STn antigen (Neu5Acα2,6-GalNAcα1-) and the TF antigen (Galβ3GalNAcα1) – each conjugated to a CRM197 carrier protein [135], [136]. Structural representation of the three glycoconjugates of the GVI is shown in FIG. 1C (only a single, representative TACA is shown on each CRM197). Typically, each CRM197 molecule contains about 6 to about 15 TACAs per molecule. Each glycoconjugate is manufactured separately and combined in a final formulation. The three PEG3-COOH functionalised TACAs (namely the Tn-PEG3-COOH (GalNAcα-O-PEG3- COOH), TF-PEG3-OOH (Galβ(1-3)GalNAcα-O-PEG3-COOH), and STn-PEG3-COOH (NeuAcα(2,6)GalNAcα-O-PEG3-COOH) compounds) were supplied by Sussex Research Laboratories, Inc. (Ottawa, ON, Canada). The amino acid sequence of the CRM197 used in these experiments was identical to the CRM197 used in approved prophylactic conjugates [137]. All non-custom materials used in the productions of each GVI, such as: the activating chemicals for conjugation (EDC (1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC)) and NHS (N- Hydroxysuccinimide)); buffering agents (MES (2-ethanesulfonic acid), sodium phosphate dibasic, and potassium phosphate monobasic); salts (potassium chloride, sodium chloride); and pH adjusting acids and bases (sodium hydroxide, hydrochloric acid), are all readily commercially available. The PEG3 functionalised TACAs are rendered active and conjugated to CRM197 (a detailed chemical conjugation scheme is shown in FIG.2). The conjugation chemistry is well established in the vaccine industry [138], [139], [140]. The residual chemicals of the reaction, DMSO, MES, NHS, and EDC-isourea and their by-products, are eliminated after buffer exchange. When an input ratio of activated TACA:protein of 50:1 is used, each CRM197 molecule is conjugated to about 6 to about 15 TACAs per molecule. EXAMPLE 2 - Conjugation efficiency
Conjugation efficiency was measured by MALDI-TOF MS. Conjugation levels for each of the CRM197-PEG3-Tn/STn/TF glycoconjugates were reproducible across five lots. FIG. 3A shows the calculated average number of TACAs per CRM197 molecule for five lots of each of the three glycoconjugates.
EXAMPLE 3 - Monovalent glycoconjugates i. In vivo potency
The in vivo testing of each of the five lots of the individual CRM197-PEG3-Tn/STn/TF glycoconjugates resulted in very reproducible IgG titers at day 28 in C57BL/6 mice, as assessed by TACA ELISA (see FIG. 3B). Data analysis clearly demonstrates monovalent glycoconjugates are capable of inducing a robust and reproducible immune response (IgG GMT > 100000) each time against each tested TACA. ii. Effect of conjugation levels on in vivo potency
Studies were performed on a series of glycoconjugates with decreasing TACA:CRM197 ratios. As described above, a 50:1 TACA:CRM197 ratio achieved up to 15 TACA molecules per CRM197 molecule (see FIG. 3A).
We reduced the TACA:CRM197 ratio to 25:1, 10:1, and 5:1. As expected, the decreased ratios led to a concomitant decrease in conjugation efficiency for all 3 TACAs to CRM197 as determined by MALDI-TOF-MS (see FIG. 4A).
There is a strong concordance between the TACA:CRM197 ratios and the level of antigen- specific IgG induced in mice (see FIG. 4B); a decrease in TACA density on carrier protein leads to decrease in immune response against the targeted antigen. At lower ratios (particularly 5:1) there are an insufficient number of modifications to elicit a favourable immune response to the TACAs. Higher ratios (particularly 50:1 and 25:1) elicit stronger immune responses against the TACAs.
EXAMPLE 4 - Trivalent compositions
Iterative in vivo testing was performed with a GVI composition comprising the adjuvants QS21 and ODN1826 as an experimental vaccine in mice. The resulting antisera were screened in a series of assays that measure GMT for each TACA alone, differential binding to antigen expressing and antigen negative cells, and differential tumouricidal activity against the same cells. Throughout all studies the best commercially available mAb controls for all antigen targets were used as comparators (the mAbs for the TF antigen tested thus far detect TF antigen on live tumour cells and TF conjugated to CRM197, but do not detect TF in the ELISA used herein). i. In vivo potency of trivalent compositions
Results from a routine experiment are shown in FIGS. 5A-5C. The first step was to determine the antibody titers against the three TACA targets using ELISA. The TACAs are scaffolded on polyacrylamide (PAA) in this screening format so that the titers measured are solely against the TACA and not the CRM197 carrier protein and/or any other protein.
A weak IgM response (average from 20 mice Tn - 11,840; STn - 4,230 and TF - 4,622) and a very strong and reproducible IgG response (average from 20 mice Tn - 623,582; STn - 1,534,852 and TF - 441,586) was observed against all three TACAs (Tn, STn and TF) at day 28. Another booster dose increased IgG titers of TF to 760,672 as observed in IgG dilution curve against TF measured at day 42.
Analysis of GVI sera against TACAs for multiple lots clearly demonstrates that the generated immune response against TACAs by the GVI composition is quite consistent and reproducible (see FIG. 5D). The titers are consistently at maximum seen for mouse GMT's against recombinant proteins (» 1:100,000).
Surprisingly, GVI compositions induced 100-500X higher IgG titers against Tn, STn and TF in mice than previous vaccine candidates described, e.g. [141], [142], [143], [144], [145], [146]. Also, the IgG responses observed are superior to pre-clinical and clinical candidates advanced previously. This is best shown by the differential IgG to IgM responses induced by GVI compositions. Induction of B cell class switching (from IgM to IgG) is a hallmark of vaccination and a demonstration of the anamnestic nature of secondary exposure to an antigen [147]. The switch from IgM production to IgG is associated with key immunological events including the generation of B cell memory, affinity maturation and production of exponentially higher titers of IgG over the initial IgM dominated response. For all the historical vaccines, the literature clearly shows IgG titers are equivalent to IgM titers in experimental animals and patients [4], [5], [6], [51], [50], [141], [142], [143]. Both IgG and IgM were detected at < 1:10,000 GMT for the three antigens, even after repeated immunisations. These data indicate that the first- generation vaccines were sub-optimal, in that they cannot induce a classical vaccine response. By contrast, the IgG titers induced by GVI compositions in mouse models are 100-500X more than the specific IgM titers generated after 4 immunisations over 28 days (IgG > 1:100,000 GMT vs. IgM ≤ 1:10,000 for all three glycan antigens). Thus, GVI compositions are capable of inducing rapid, durable, boostable and high levels of circulating IgG against the aberrant tumour glycans, Tn, STn and TF.
Without being bound by theory, the proposed mechanisms of GVI compositions induced IgG efficacy are the same as those for anti-tumour antibody therapeutics and vaccines, such as anti- Her2 or anti-CD20 Tx mAbs [148], [149]. Binding differentially to tumour over healthy tissue, the vaccine-induced anti-glycan antibodies target the tumour leading to cell death through diverse mechanisms. These include engagement of the patient's immune system through CDC, ADCC and ADCP and potentially direct signalling. The antibodies elicited by GVI compositions bind Tn, STn and TF in the absence of protein/peptide context. That is, they are targeting the glycan alone and not a single glycoprotein, per se. Thus, in immunological terms, the antibodies are hapten-specific [150], [151]. ii. Tumour cell binding and tumouricidal IgG
Whilst Ig binding to TACAs in an ELISA format is informative, Ig binding to live tumour cell lines expressing TACAs and inducing their destruction is the key pharmacodynamic (PD) effect desired from GVI compositions. Binding to antigen bearing tumour cell lines was assessed by flow cytometry (see FIGS. 6A-6C). The cell binding data clearly show that a GVI composition comprising QS21 and ODN1826 induced IgG binding cells bearing the target TACAs (Jurkat cells express Tn and STn, while Ov90 cells express STn only), but not those negative for Tn, STn, and TF (Colo201 cells). This selective binding leads to cell lysis if complement is added to the assay and cell viability is determined (see FIGS. 6D and 6E). Notably, it had previously been suggested in the literature that Tn, STn and TF antigens were incapable of inducing tumouricidal antibodies [152]. The results clearly challenge this misconception.
EXAMPLE 5 - IgG specificity
A glycan microarray platform was exploited to understand further the specificity of the antisera generated across >90 related mammalian O-linked glycan structures scaffolded in different ways (FIG. 7). The results show that GVI IgG responses are not just potent but also highly specific for the three target glycans, namely Tn, STn, and TF. No cross-reactivity towards other glycans on the array was observed, indicating that the IgG responses induced by GVI are specific for the targeted glycans with minimal cross-reactivity to other closely related structures, including those naturally expressed in humans. EXAMPLE 6 - Long term stability
The long term stability of the three monovalent glycoconjugates was studied. A consistent size exclusion profile was observed over a 300-day period, suggesting that each monovalent glycoconjugate is stable at 4-8°C condition (see FIGS. 8A-8C). Furthermore, in vivo potency as measured by geometric mean titers of IgG against C57BL/6 mice was consistent over time in 4 or 5 different in vivo studies (FIGS. 8D-8F).
EXAMPLE 7 - Adjuvant screening
Multiple different adjuvant classes were screened, including TLR3 agonists (e.g. poly l:C), TLR9 agonists (e.g. ODN1826), TLR4 agonists (e.g. MPLA), Th2 adjuvants (e.g. Alum), and Th1/Th2 adjuvants (e.g. QS21). Multiple different formulations were screened, including including liquid immunogen + liquid adjuvant mixes, liquid immunogen + liposome mixes, and immunogen/adjuvant absorbed to aluminum hydroxide "alum". Clearly, use of adjuvants can help maximise IgG titers.
EXAMPLE 8 - Alternative linkers In the above examples, CRM197-PEG3-Tn/STn/TF 1 conjugates (see FIG. IB) were exemplified. However, other linkers and conjugations chemistries may also be used in the invention.
For example, linkers comprising serine are suitable alternatives to linkers comprising PEG3 (see FIG. 10A).
Also, CRM197-PEG3-Tn/STn/TF conjugates may have different conjugations chemistries, for example see "CRM197-PEG3-Tn 1" and "CRM197-PEG3-Tn 2" in FIG. IB. Whilst CRM197-PEG3- Tn 1 and CRM197-PEG3-Tn 2 resulted in similar anti-Tn IgG levels, CRM197-PEG3-Tn 1 elicited significantly lower levels of anti-PEG (i.e., anti-linker) antibodies (FIG. 10B).
Conjugates comprising other linkers, e.g., other PEG groups (e.g., PEG4-PEG8), threonine, or alkane groups (e.g., C2-C8 alkanes), or other conjugation chemistries, are expected to achieve similar results.
REFERENCES
[1] Laubli and Borsig (2019) Front Immunol 10:2120.
[2] Munkley and Elliot (2016) Oncotarget 7:35478-35489.
[3] Astronomo (2010) Not Rev Drug Discov 9:308-324.
[4] MacLean (1993) Cancer Immunol Immunother 36, 215-222.
[5] Adluri (1995) Cancer Immunol Immunother 41, 185-192. [6] MacLean (1992) J Immunother 11, 292-305.
[7] Matsumoto (2020) Glycobiology 30, 282-300
[8] Julien (2012) Biomolecules 2, 435-466.
[9] Karacosta (2018) Transl Oncol 11, 450-466.
[10] Sun (2018) BMC Cancer 18, 827.
[11] Sun (2016) Chem Med Chem 11(10):1090-1096
[12] Song (2017) Octotarget 8(29):47330-47343.
[13] Song (2019) Glycoconjugate J 36:399-408.
[14] Mettu (2020) J Biol Sci 27(9):1-22.
[15] Tyrikos-Ergas (2020) Biomacromol 21(1):18-29
[16] WO09/066245.
[17] Amedei (2020) iScience 101250.
[18] W02005/000346.
[19] Anonymous (Jan 2002) Research Disclosure, 453077.
[20] Anderson (1983) Infect Immun 39(1):233-238.
[21] Anderson (1985) J Clin Invest 76(1):52-59.
[22] WO2014/102265
[23] US10,093,704
[24] W02015/117093
[25] US8,530,171
[26] WO2011/126811
[27] WO2018/143648
[28] Swaminathan (2014) BrJ Clin Pharmacol 78(5):1135-1142
[29] EP-A-0372501.
[30] EP-A-0378881.
[31] EP-A-0427347.
[32] W093/17712
[33] W094/03208.
[34] W098/58668.
[35] EP-A-0471177.
[36] WO91/01146
[37] Falugi (2001) EurJ Immunol 31:3816-24.
[38] Baraldo (2004) Infect Immun 72:4884-87.
[39] EP-A-0594610.
[40] WO00/56360.
[41] W002/091998.
[42] Kuo (1995) Infect Immun 63:2706-13.
[43] WO01/72337
[44] WOOO/61761.
[45] WO00/33882
[46] Moginger (2016) Sci Rep 6:20488 [47] Ragupathi (2006) J Am Chem Soc 128(8):2715-2725
[48] Gilewski (2007) Clin Cancer Res 15(13):2977-85
[49] Sabbatini (2007) Clin Cancer Res 13(14):4170-4177
[50] Slovin (2005) Cancer Immunol Immunother 54(7):694-702
[51] Slovin (2003) J Clin Oncol 21(23):4294-4298
[52] Slovin (2007) Cancer Immunol Immother 56(12):1921-1930
[53] Palitzsch (2016) Angew Chem Int Ed Engl 55(8):2894-2898
[54] Malekan (2013) Bioorg Med Chem 21(16):4778-4785
[55] W017/48823.
[56] Paoletti (2001) Vaccine 19:2118-2126.
[57] WO00/56365.
[58] Gennaro (2000) Remington: The Science and Practice of Pharmacy. 20th edition, ISBN: 0683306472.
[59] W003/009869.
[60] Almeida & Alpar (1996) J. Drug Targeting 3:455-467.
[61] Agarwal & Mishra (1999) Indian J Exp Biol 37:6-16.
[62] US patent 6355271.
[63] WO00/23105.
[64] Vaccine Design... (1995) eds. Powell & Newman. ISBN: 030644867X. Plenum.
[65] WO90/14837.
[66] Podda (2001) Vaccine 19:2673-80.
[67] Frey (2003) Vaccine 21:4234-7.
[68] US Patent 6,299,884.
[69] US Patent 6,451,325.
[70] US patent 5,057,540.
[71] W096/33739.
[72] EP-A-0109942.
[73] W096/11711.
[74] WO00/07621.
[75] Barr (1998) Advanced Drug Delivery Reviews 32:247-271.
[76] Sjolanderet (1998) Advanced Drug Delivery Reviews 32:321-338.
[77] Niikura (2002) Virology 293:273-280.
[78] Lenz (2001) J Immunol 166:5346-5355.
[79] Pinto (2003) J Infect Dis 188:327-338.
[80] Gerber (2001) Virol 75:4752-4760.
[81] W003/024480
[82] W003/024481
[83] Gluck (2002) Vaccine 20:B10-B16.
[84] EP-A-0689454.
[85] Johnson (1999) Bioorg Med Chem Lett 9:2273-2278.
[86] Evans (2003) Expert Rev Vaccines 2:219-229.
[87] Meraldi (2003) Vaccine 21:2485-2491.
[88] Pajak (2003) Vaccine 21:836-842. [89] Kandimalla (2003) Nucleic Acids Research 31:2393-2400.
[90] WO02/26757.
[91] W099/62923.
[92] Krieg (2003) Nature Medicine 9:831-835.
[93] McCluskie (2002) FEMS Immunology and Medical Microbiology 32:179-185.
[94] W098/40100.
[95] US patent 6,207,646.
[96] US patent 6,239,116.
[97] US patent 6,429,199.
[98] Kandimalla (2003) Biochemical Society Transactions 31 (part 3):654-658.
[99] Blackwell (2003) J Immunol 170:4061-4068.
[100] Krieg (2002) Trends Immunol 23:64-65.
[101] WOO1/95935.
[102] Kandimalla (2003) BBRC 306:948-953.
[103] Bhagat (2003) BBRC 300:853-861.
[104] WO03/035836.
[105] W095/17211.
[106] W098/42375.
[107] Beignon (2002) Infect imrnun 70:3012-3019.
[108] Pizza (2001) Vaccine 19:2534-2541.
[109] Pizza (2000) Int J Med Microbiol 290:455-461.
[110] Scharton-Kersten (2000) Infect Imrnun 68:5306-5313.
[111] Ryan (1999) Infect Imrnun 67:6270-6280.
[112] Partidos (1999) Immunol Lett 67:209-216.
[113] Peppoloni (2003) Expert Rev Vaccines 2:285-293.
[114] Pine (2002) J Control Release 85:263-270.
[115] Domenighini (1995) Mol Microbiol 15:1165-1167.
[116] WO99/40936.
[117] W099/44636.
[118] Singh (2001) J Cont Release 70:267-276.
[119] WO99/27960.
[120] US patent 6,090,406
[121] EP-A-0626169.
[122] W099/52549.
[123] W001/21207.
[124] WO01/21152.
[125] Andrianov (1998) Biomaterials 19:109-115.
[126] Payne (1998) Adv Drug Delivery Review 31:185-196.
[127] Stanley (2002) Clin Exp Dermatol 27:571-577.
[128] Jones (2003) Curr Opin Investig Drugs 4:214-218.
[129] W004/60308
[130] WO04/64759. [131] Freire (2003) IntJ Parasitol 33(1):47-56
[132] Freire (2003) Biochem Biophys Res Commun 312(4):1309-16
[133] Ju (2011) Angew Chem Int Ed Engl 50(8):1770-91
[134] https://www.zbiotech.com/o-elvcan-array.html
[135] Malito (2012) PNAS 109:5229-5234
[136] WO 2015/117093 A1
[137] Hickey (2018) J Pharm Sci 107, 1806-1819.
[138] Lockyer (2020) Vaccine 38, 2859-2869.
[139] Micoli (2011) Vaccine 29, 712-720.
[140] Cuello (2007) Vaccine 25, 1798-1805.
[141] Kuduk (1998) J Am Chem Soc 120, 12474-12485.
[142] Zhang (1995) Cancer Res 55, 3364-3368.
[143] Ragupathi (2003) Cancer Immunol Immunother 52, 608-616.
[144] Julien (2009) BrJ Cancer 100, 1746-1754.
[145] Fung (1990) Cancer Res 50, 4308.
[146] Lo-Man (200) J Immunol 166, 2849.
[147] Snapper and Mond (1993) Immunol Today 14, 15-17.
[148] Scott (2012) Nat Rev Cancer 12, 278-287.
[149] Maximiano (2016) BioDrugs 30, 75-86.
[150] Polonskaya (2019) Curr Opin Immunol 59: 65-71.
[151] Cummings (2009) Mol Biosyst 5(10):1087-104.
[152] Ragupathi (2005) J Immunol 174:5706-5712

Claims

1. An immunogenic composition comprising: a) a conjugate comprising the tumour-associated antigen (TACA) Tn covalently conjugated to a carrier protein; b) a conjugate comprising the TACA STn covalently conjugated to a carrier protein; and c) a conjugate comprising the TACA TF covalently conjugated to a carrier protein, wherein the immunogenic composition does not comprise any other TACAs.
2. The immunogenic composition of claim 1, wherein the carrier protein is an immunogenic carrier protein.
3. The immunogenic composition of claim 2, wherein the immunogenic carrier protein in a), b), and/or c) is a diphtheria toxin or toxoid, or a tetanus toxin or toxoid.
4. The immunogenic composition of any preceding claim, wherein the immunogenic carrier protein in a), b), and/or c) is CRM197.
5. The immunogenic composition of any preceding claim, wherein in a), b), and/or c) the TACA is covalently conjugated to the carrier protein via a linker.
6. The immunogenic composition of claim 5, wherein the linker comprises a polyethylene glycol (PEG).
7. The immunogenic composition of claim 6, wherein the PEG is from PEG-2 to PEG-12.
8. The immunogenic composition of claim 7, wherein the PEG is PEG-3.
9. The immunogenic composition of any preceding claim, wherein each TACA is covalently linked to a CRM197 carrier protein via a PEG-3 linker and an amide bond.
10. The immunogenic composition of any preceding claims, wherein the average number of TACA molecules covalently conjugated to each carrier molecule is at least about 4.
11. The immunogenic composition of claim 9, wherein the average number of TACA molecules covalently conjugated to each carrier molecule is at least about 6.
12. The immunogenic composition of any preceding claim, wherein the ratio (w/w) of each TACA to each other TACA is from about 2:1 to about 1:2.
13. The immunogenic composition of claim 12, wherein the ratio (w/w) of Tn:STn:TF in the composition is about 1:1:1.
14. The immunogenic composition of any preceding claim, wherein the immunogenic composition comprises: a) a first conjugate comprising Tn covalently conjugated to a first carrier protein; b) a second conjugate comprising STn covalently conjugated to a second carrier protein; and c) a third conjugate comprising TF covalently conjugated to a third carrier protein.
15. The immunogenic composition of claim 14, wherein the first, second, and third carrier proteins are each CRM197.
16. The immunogenic composition of any preceding claim, wherein one or more of Tn, STn, and TF are modified compared to native Tn, STn, or TF or are analogs of native Tn, STn, and TF.
17. A pharmaceutical composition comprising the immunogenic composition of any of the preceding claims and a pharmaceutically acceptable excipient or carrier.
18. The pharmaceutical composition of claim 17, further comprising one or more adjuvants.
19. A trivalent vaccine comprising the immunogenic composition of any of claims 1-16 or the pharmaceutical composition of claim 17 or 18.
20. The immunogenic composition of any of claims 1-16, for use in a method of treatment.
21. A method of treating or preventing cancer in a subject, the method comprising administering the immunogenic composition of any of claims 1-16 to the subject.
22. The immunogenic composition of any of claims 1-16, for use in a method of treating or preventing cancer.
23. The method of claim 21 or the immunogenic composition for the use of claim 22, wherein the cancer is a solid tumour.
24. The method or the immunogenic composition for the use of claim 23, wherein the cancer is selected from the group consisting of: pancreatic cancer, breast cancer, colon cancer, gastro-intestinal cancer, prostate cancer, lung cancer, ovarian cancer, and stomach cancer.
25. The method of claim 21 or the immunogenic composition for the use of claim 22, wherein the subject is human.
PCT/US2022/032730 2021-06-08 2022-06-08 Immunogenic compositions comprising tumour-associated antigen WO2022261251A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163208207P 2021-06-08 2021-06-08
US63/208,207 2021-06-08

Publications (1)

Publication Number Publication Date
WO2022261251A1 true WO2022261251A1 (en) 2022-12-15

Family

ID=82595039

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/032730 WO2022261251A1 (en) 2021-06-08 2022-06-08 Immunogenic compositions comprising tumour-associated antigen

Country Status (1)

Country Link
WO (1) WO2022261251A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US6090406A (en) 1984-04-12 2000-07-18 The Liposome Company, Inc. Potentiation of immune responses with liposomal adjuvants
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6299884B1 (en) 1989-05-25 2001-10-09 Chiron Corporation Adjuvant formulation comprising a submicron oil droplet emulsion
US6355271B1 (en) 1999-02-03 2002-03-12 Biosante Pharmaceuticals, Inc. Therapeutic calcium phosphate particles and methods of manufacture and use
US6429199B1 (en) 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6090406A (en) 1984-04-12 2000-07-18 The Liposome Company, Inc. Potentiation of immune responses with liposomal adjuvants
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US6299884B1 (en) 1989-05-25 2001-10-09 Chiron Corporation Adjuvant formulation comprising a submicron oil droplet emulsion
US6451325B1 (en) 1989-05-25 2002-09-17 Chiron Corporation Adjuvant formulation comprising a submicron oil droplet emulsion
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6429199B1 (en) 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US6355271B1 (en) 1999-02-03 2002-03-12 Biosante Pharmaceuticals, Inc. Therapeutic calcium phosphate particles and methods of manufacture and use

Non-Patent Citations (87)

* Cited by examiner, † Cited by third party
Title
ADLURI, CANCER IMMUNOL IMMUNOTHER, vol. 41, 1995, pages 185 - 192
AGARWALMISHRA, INDIAN J EXP BIOL, vol. 37, 1999, pages 6 - 16
ALMEIDAALPAR, J. DRUG TARGETING, vol. 3, 1996, pages 455 - 467
AMEDEI, ISCIENCE, 2020, pages 101250
ANDERSON, INFECT IMMUN, vol. 39, no. 1, 1983, pages 233 - 238
ANDERSON, J CLIN INVEST, vol. 76, no. 1, 1985, pages 52 - 59
ANDRIANOV, BIOMATERIALS, vol. 19, 1998, pages 109 - 115
ASTRONOMO, NAT REV DRUG DISCOV, vol. 9, 2010, pages 308 - 324
BARALDO, INFECT IMMUN, vol. 72, 2004, pages 4884 - 87
BEIGNON, INFECT IMMUN, vol. 70, 2002, pages 3012 - 3019
BLACKWELL, J IMMUNOL, vol. 170, 2003, pages 4061 - 4068
CUELLO, VACCINE, vol. 25, 2007, pages 1798 - 1805
CUMMINGS, MOL BIOSYST, vol. 5, no. 10, 2009, pages 1087 - 104
DOMENIGHINI, MOL MICROBIOL, vol. 15, 1995, pages 1165 - 1167
FALUGI, EUR J IMMUNOL, vol. 31, 2001, pages 3816 - 24
FREIRE, BIOCHEM BIOPHYS RES COMMUN, vol. 312, no. 4, 2003, pages 1309 - 16
FREIRE, INT J PARASITOL, vol. 33, no. l, 2003, pages 47 - 56
FUNG, CANCER RES, vol. 50, 1990, pages 4308
GERBER, VIROL, vol. 75, 2001, pages 4752 - 4760
GLUCK, VACCINE, vol. 20, 2002, pages B10 - B16
HICKEY, J PHARM SCI, vol. 107, 2018, pages 1806 - 1819
JOHNSON, BIOORG MED CHEM LETT, vol. 9, 1999, pages 2273 - 2278
JONES, CURR OPIN INVESTIG DRUGS, vol. 4, 2003, pages 214 - 218
JU, ANGEW CHEM INT ED ENGL, vol. 50, no. 8, 2011, pages 1770 - 91
JULIEN, BIOMOLECULES, vol. 2, 2012, pages 435 - 466
JULIEN, BR J CANCER, vol. 100, 2009, pages 1746 - 1754
KANDIMALLA, BBRC, vol. 300, 2003, pages 853 - 861
KANDIMALLA, BIOCHEMICAL SOCIETY TRANSACTIONS, vol. 31, 2003, pages 654 - 658
KANDIMALLA, NUCLEIC ACIDS RESEARCH, vol. 31, 2003, pages 2393 - 2400
KARACOSTA, TRANSL ONCOL, vol. 11, 2018, pages 450 - 466
KRIEG, NATURE MEDICINE, vol. 9, 2003, pages 831 - 835
KRIEG, TRENDS IMMUNOL, vol. 23, 2002, pages 64 - 65
KUDUK, J AM CHEM SOC, vol. 120, 1998, pages 12474 - 12485
KUO, INFECT IMMUN, vol. 63, 1995, pages 2706 - 13
LAUBLIBORSIG, FRONT IMMUNOL, vol. 10, 2019, pages 2120
LENZ, J IMMUNOL, vol. 166, 2001, pages 5346 - 5355
LOCKYER, VACCINE, vol. 38, 2020, pages 2859 - 2869
MACLEAN, CANCER IMMUNOL IMMUNOTHER, vol. 36, 1993, pages 215 - 222
MACLEAN, J IMMUNOTHER, vol. 11, 1992, pages 292 - 305
MALEKAN, BIOORG MED CHEM, vol. 21, no. 16, 2013, pages 4778 - 4785
MALITO, PNAS, vol. 109, 2012, pages 5229 - 5234
MATSUMOTO, GLYCOBIOLOGY, vol. 30, 2020, pages 282 - 300
MAXIMIANO, BIODRUGS, vol. 30, 2016, pages 75 - 86
MCCLUSKIE, FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY, vol. 32, 2002, pages 179 - 185
MERALDI, VACCINE, vol. 21, 2003, pages 2485 - 2491
METTU, J BIOL SCI, vol. 27, no. 9, 2020, pages 1 - 22
MICOLI, VACCINE, vol. 29, 2011, pages 712 - 720
MOGINGER, SCI REP, vol. 6, 2016, pages 20488
MUNKLEYELLIOT, ONCOTARGET, vol. 7, 2016, pages 35478 - 35489
NIIKURA, VIROLOGY, vol. 293, 2002, pages 273 - 280
PALITZSCH, ANGEW CHEM INT ED ENGL, vol. 55, no. 8, 2016, pages 2894 - 2898
PAOLETTI, VACCINE, vol. 19, 2001, pages 2534 - 2541
PARTIDOS, IMMUNOL LETT, vol. 67, 1999, pages 209 - 216
PAYNE, ADV DRUG DELIVERY REVIEW, vol. 31, 1998, pages 185 - 196
PEPPOLONI, EXPERT REV VACCINES, vol. 2, 2003, pages 285 - 293
PINE, J CONTROL RELEASE, vol. 85, 2002, pages 263 - 270
PINTO, J INFECT DIS, vol. 188, 2003, pages 327 - 338
PIZZA, INT J MED MICROBIOL, vol. 290, 2000, pages 455 - 461
POLONSKAYA, CURR OPIN IMMUNOL, vol. 59, 2019, pages 65 - 71
RAGUPATHI G ET AL: "A PRECLINICAL STUDY COMPARING APPROACHES FOR AUGMENTING THE IMMUNOGENICITY OF A HEPTAVALENT KLH-CONJUGATE VACCINE AGAINST EPITHELIAL CANCERS", CANCER IMMUNOLOGY IMMUNOTHERAPY, SPRINGER, BERLIN/HEIDELBERG, vol. 52, no. 10, 1 October 2003 (2003-10-01), pages 608 - 616, XP008060191, ISSN: 0340-7004, DOI: 10.1007/S00262-003-0399-2 *
RAGUPATHI, CANCER IMMUNOL IMMUNOTHER, vol. 52, 2003, pages 608 - 616
RAGUPATHI, J AM CHEM SOC, vol. 128, no. 8, 2006, pages 2715 - 2725
RAGUPATHI, J IMMUNOL, vol. 174, 2005, pages 5706 - 5712
RESEARCH DISCLOSURE, January 2002 (2002-01-01), pages 453077
RYAN, INFECT IMMUN, vol. 67, 1999, pages 6270 - 6280
SABBATINI PJ ET AL: "Pilot Study of a Heptavalent Vaccine-Keyhole Limpet Hemocyanin Conjugate plus QS21 in Patients with Epithelial Ovarian, Fallopian Tube, or Peritoneal Cancer", CLINICAL CANCER RESEARCH, vol. 13, no. 14, 15 July 2007 (2007-07-15), pages 4170 - 4177, XP055012365, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-06-2949 *
SABBATINI, CLIN CANCER RES, vol. 13, no. 14, 2007, pages 4170 - 4177
SCHARTONKERSTEN, INFECT IMMUN, vol. 68, 2000, pages 5306 - 5313
SCOTT, NAT REV CANCER, vol. 12, 2012, pages 278 - 287
SINGH, J CONT RELEASE, vol. 70, 2001, pages 267 - 276
SJOLANDERET, ADVANCED DRUG DELIVERY REVIEWS, vol. 32, 1998, pages 321 - 338
SLOVIN, CANCER IMMUNOL IMMOTHER, vol. 56, no. 12, 2007, pages 1921 - 1930
SLOVIN, CANCER IMMUNOL IMMUNOTHER, vol. 54, no. 7, 2005, pages 694 - 702
SLOVIN, J CLIN ONCOL, vol. 21, no. 23, 2003, pages 4294 - 4298
SNAPPERMOND, IMMUNOL TODAY, vol. 14, 1993, pages 15 - 17
SONG C ET AL: "Fluorine-modified sialyl-Tn-CRM197 vaccine elicits a robust immune response", GLYCOCONJUGATE JOURNAL, CHAPMAN & HALL, BOSTON, vol. 36, no. 5, 2 July 2019 (2019-07-02), pages 399 - 408, XP036885573, ISSN: 0282-0080, [retrieved on 20190702], DOI: 10.1007/S10719-019-09884-0 *
SONG C ET AL: "Synthesis and immunological evaluation ofN-acyl modified Tn analogues as anticancer vaccine candidates", BIOORGANIC & MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 24, no. 4, 8 January 2016 (2016-01-08), pages 915 - 920, XP029399971, ISSN: 0968-0896, DOI: 10.1016/J.BMC.2016.01.015 *
SONG, GLYCOCONJUGATE J, vol. 36, 2019, pages 399 - 408
SONG, OCTOTARGET, vol. 8, no. 29, 2017, pages 47330 - 47343
STANLEY, CLIN EXP DERMATOL, vol. 27, 2002, pages 571 - 577
STEFANETTI G ET AL: "Click chemistry compared to thiol chemistry for the synthesis of site-selective glycoconjugate vaccines using CRMas carrier protein", GLYCOCONJUGATE JOURNAL, CHAPMAN & HALL, BOSTON, vol. 37, no. 5, 13 June 2020 (2020-06-13), pages 611 - 622, XP037249264, ISSN: 0282-0080, [retrieved on 20200613], DOI: 10.1007/S10719-020-09930-2 *
SUN S ET AL: "Synthesis and Evaluation of Glycoconjugates Comprising N -Acyl-Modified Thomsen-Friedenreich Antigens as Anticancer Vaccines", CHEMMEDCHEM COMMUNICATIONS, vol. 11, no. 10, 14 April 2016 (2016-04-14), DE, pages 1090 - 1096, XP055960096, ISSN: 1860-7179, DOI: 10.1002/cmdc.201600094 *
SUN, BMC CANCER, vol. 18, 2018, pages 827
SUN, CHEM MED CHEM, vol. 11, no. 10, 2016, pages 1090 - 1096
SWAMINATHAN, BR J CLIN PHARMACOL, vol. 78, no. 5, 2014, pages 1135 - 1142
TYRIKOSERGAS, BIOMACROMOL, vol. 21, no. 1, 2020, pages 18 - 29
ZHANG, CANCER RES, vol. 55, 1995, pages 3364 - 3368

Similar Documents

Publication Publication Date Title
ES2329059T3 (en) MENINGOCOCIC CONJUGATES COMBINED WITH A COMMON TRANSPORTING PROTEIN.
US10286055B2 (en) Immunogenic composition
US20150283232A1 (en) Immunogenic compositions
NZ560432A (en) Conjugation of streptococcal capusular saccharides
JP2008538183A (en) Type B influenzae
KR102112663B1 (en) Salmonella conjugate vaccines
ES2774729T3 (en) Cycloalkino derivative saccharides
CN104411328A (en) Composition
WO2016142880A1 (en) Compositions and uses
WO2022261251A1 (en) Immunogenic compositions comprising tumour-associated antigen
WO2015153883A1 (en) Vaccination using plant virus particles linked to her2 antigens
WO2006109186A2 (en) Early endosome neutralising compouds as vaccine adjuvants

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22743636

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE