WO2005061523A1 - Glycosaminoglycan (gag) mimetics - Google Patents

Glycosaminoglycan (gag) mimetics Download PDF

Info

Publication number
WO2005061523A1
WO2005061523A1 PCT/AU2004/001800 AU2004001800W WO2005061523A1 WO 2005061523 A1 WO2005061523 A1 WO 2005061523A1 AU 2004001800 W AU2004001800 W AU 2004001800W WO 2005061523 A1 WO2005061523 A1 WO 2005061523A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
ome
mhz
compounds
deoxy
Prior art date
Application number
PCT/AU2004/001800
Other languages
French (fr)
Inventor
Robert Hugh Don
Vito Ferro
Ian Bytheway
Siska Cochran
Jon Krueger Fairweather
Edward Timothy Hammond
Tomislav Karoli
Cai Ping Li
Ligong Liu
Original Assignee
Progen Industries Limited
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
Priority claimed from AU2003907107A external-priority patent/AU2003907107A0/en
Application filed by Progen Industries Limited filed Critical Progen Industries Limited
Priority to CA002551181A priority Critical patent/CA2551181A1/en
Priority to JP2006545842A priority patent/JP2007515434A/en
Priority to AU2004303434A priority patent/AU2004303434A1/en
Priority to EP04802102A priority patent/EP1699806A1/en
Priority to BRPI0417750-9A priority patent/BRPI0417750A/en
Priority to MXPA06007194A priority patent/MXPA06007194A/en
Publication of WO2005061523A1 publication Critical patent/WO2005061523A1/en
Priority to IL176474A priority patent/IL176474A0/en
Priority to NO20063366A priority patent/NO20063366L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H5/00Compounds containing saccharide radicals in which the hetero bonds to oxygen have been replaced by the same number of hetero bonds to halogen, nitrogen, sulfur, selenium, or tellurium
    • C07H5/08Compounds containing saccharide radicals in which the hetero bonds to oxygen have been replaced by the same number of hetero bonds to halogen, nitrogen, sulfur, selenium, or tellurium to sulfur, selenium or tellurium
    • C07H5/10Compounds containing saccharide radicals in which the hetero bonds to oxygen have been replaced by the same number of hetero bonds to halogen, nitrogen, sulfur, selenium, or tellurium to sulfur, selenium or tellurium to sulfur
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H5/00Compounds containing saccharide radicals in which the hetero bonds to oxygen have been replaced by the same number of hetero bonds to halogen, nitrogen, sulfur, selenium, or tellurium
    • C07H5/04Compounds containing saccharide radicals in which the hetero bonds to oxygen have been replaced by the same number of hetero bonds to halogen, nitrogen, sulfur, selenium, or tellurium to nitrogen
    • C07H5/06Aminosugars
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H11/00Compounds containing saccharide radicals esterified by inorganic acids; Metal salts thereof
    • C07H11/04Phosphates; Phosphites; Polyphosphates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H13/00Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids
    • C07H13/12Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids by acids having the group -X-C(=X)-X-, or halides thereof, in which each X means nitrogen, oxygen, sulfur, selenium or tellurium, e.g. carbonic acid, carbamic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/02Acyclic radicals, not substituted by cyclic structures
    • C07H15/04Acyclic radicals, not substituted by cyclic structures attached to an oxygen atom of the saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/18Acyclic radicals, substituted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H3/00Compounds containing only hydrogen atoms and saccharide radicals having only carbon, hydrogen, and oxygen atoms

Definitions

  • the invention that is the subject of this application lies in the area of compounds that mimic the structure of certain carbohydrates. More particularly, the invention lies in the area of glycosaminoglycan (GAG) mimetics. Specifically, the invention relates to compounds comprising at least one charged group that are designed to mimic the structure of GAGs. The invention also relates to methods for the preparation of the compounds, compositions comprising the compounds, and use of the compounds and compositions thereof for the antiangiogenic, antimetastatic, anti-inflammatory, anticoagulant, antithiOmbotic, and/or antimicrobial treatment of a mammalian subject.
  • GAG glycosaminoglycan
  • the invention further relates to the use ofthe compounds and compositions thereof in the treatment of a mammalian subject having a condition amenable to treatment with such agents.
  • GAGs Glycosaminoglycans
  • ECM extracellular matrix
  • HS-GAGs play key roles in cell growth and development, angiogenesis, coagulation, tumour metastasis, cell adhesion, activation of growth factors, binding of cytokines and chemokines, and infection by bacteria and viruses [4-6].
  • angiogenesis coagulation
  • tumour metastasis cell adhesion
  • activation of growth factors binding of cytokines and chemokines
  • infection by bacteria and viruses [4-6].
  • GAG mimetics molecules that mimic the structure of certain GAGs — which molecules are referred to as “GAG mimetics” — can bind to GAG-binding proteins and modulate their biological activity: e.g., the activation of AT-III by various pentasaccharides [7,8], or the activation of fibroblast growth factors (FGFs) by sucrose octasulfate [9].
  • FGFs fibroblast growth factors
  • anticancer agents that have been developed to target HS-binding angiogenic growth factors include polysulfonated compounds [10], suramin and the related suradistas [11], and sulfated oligosaccharides [12,13].
  • the present invention relates to novel, small molecule GAG mimetics that bind to GAG-binding proteins and modulate their functions.
  • the compounds incorporate at least one negatively charged group (preferably a sulfo group) to interact with the positively charged residues in the GAG-binding site of the target proteins, and also contain one or more substituents to form interactions with other protein residues in and around the above-mentioned binding site.
  • Important and distinguishing features ofthe compounds described herein are that they have fewer sulfo groups and are of lower molecular weight than previously described polysulfated GAG mimetics such as the sulfated oligosaccharides [12,13]. Another important feature is that their structures are based on cyclic scaffolds (e.g., a monosaccharide) with sulfo groups and other substituents placed in specific, pre-defined orientations about the ring, thus differing significantly from the simple, randomly charged GAG mimetics described by Kisilevsky [14].
  • SPR surface plasmon resonance
  • a selection of compounds are shown to inhibit the HS-mediated infection of cells and cell-to-cell spread by herpes simplex virus.
  • One aspect of the present invention is the utilisation of the Ugi reaction [15,16] to provide a diverse array of GAG mimetics. The capacity for variation in the manner in which the individual charged structures are connected to one another or to other functional groups as well as the scope of application to mimic the diverse structural variation of GAGs is demonstrated.
  • the functionalisation of the cyclic scaffolds is not limited to the Ugi reaction. For example, the use of many other reactions such as alkylation, acylation and cycloaddition is demonstrated.
  • n is an integer of from 0 to 2;
  • Z is N, N(O), O. S, S(O), S(O) 2 , P, P(O), P(O) 2 , Si, Si(O), or Si(O) 2 ;
  • each X is independently C, C(O), N, N(O), O, S, S(O), S(O) 2 , P.
  • each of R ⁇ to R 6 is independently a bond or is selected from the group consisting of: hydrogen; halogen; straight chain, cyclic, branched, substituted, heterocyclic, heteroatom substituted or unsubstituted alkyl, alkenyl, alkynyl, aryl, or heteroaryl; phosphoryl groups such as phosphate, thiophosphate -O-P(S)(OH) 2 ; phosphate esters -O-P(O)(OR) 2 ; thiophosphate esters -O-P(OR) 2 ; phosphonate -O-P(O)OHR; thiophosphonate -O-P(S)OHR; substituted phosphonate -O-P(O)OR 1 R 2 ; substituted thiophosphonate -O-P(S)OR 1 R 2 ; -O-
  • a pharmaceutical or veterinary composition for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis, and/or microbial infection, which composition comprises at least one compound according to the first embodiment together with a pharmaceutically or veterinarially acceptable carrier or diluent for said at least one compound.
  • a pharmaceutically or veterinarially acceptable carrier or diluent for said at least one compound.
  • a compound according to the first embodiment in the manufacture of a medicament for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis, and/or microbial infection.
  • a method for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis, and/or microbial infection comprises administering to the subject an effective amount of at least one compound according to the first embodiment, or a composition comprising said at least one compound.
  • processes for synthesising the compounds according to the first embodimrent as defined above are provided.
  • alkyl, aryl and other substituent groups are used in accordance with their usual meaning in the art. For example, alkyl and aryl groups would normally have from 1 to 10 carbon atoms.
  • two ofthe groups Ri to R 5 may be connected to each other to form a bicyclic strucure; or the cyclic structure of formula I may contain a double bond, i.e., two contiguous XRi to XR 5 groups may be bonds.
  • Preferred compounds ofthe invention have the general structures of formulae III— VI, as defined in Tables 1-4 below. In order that the invention may be more readily understood and put into practice, one or more preferred embodiments thereof will now be described, by way of example only.
  • GAG glycosaminoglycan HS heparan sulfate FGF fibroblast growth factor aFGF acidic fibroblast growth factor (or FGF-1) bFGF basic fibroblast growth factor (or FGF-2) NEGF vascular endothelial growth factor SPR surface plasmon resonance HSN herpes simplex virus
  • FGF-1 FGF acidic fibroblast growth factor
  • FGF-2 FGF basic fibroblast growth factor
  • NEGF vascular endothelial growth factor SPR surface plasmon resonance HSN herpes simplex virus
  • GAG mimetics of the invention can be synthesised using a number of different routes, including the Ugi reaction, and generally incorporating sulfonation in the process.
  • Preferred compounds according to the first embodiment of the invention as defined above include those embraced by generic structures I and II and those included in Tables 1-4 below.
  • the compounds according to the invention have utility in the prevention or treatment in mammalian subjects of a disorder resulting from angiogenesis, metastasis, inflammation, microbial infection, coagulation or thrombosis.
  • the compounds have particular utility in the treatment of the foregoing disorders in humans.
  • the compounds are typically administered as a component of a pharmaceutical composition as described in the following paragraphs.
  • Pharmaceutical compositions for oral administration can be in tablet, capsule, powder or liquid form.
  • a tablet can include a solid carrier such as gelatine or an adjuvant or an inert diluent.
  • Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, a mineral oil or a synthetic oil.
  • Physiological saline solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. Such compositions and preparations will generally contain at least 0.1 wt% of the compound.
  • Parenteral administration includes administration by the following routes: intravenously, cutaneously or subcutaneously, nasally, intramuscularly, intraocularly, transepithelially, intraperitoneally and topically. Topical administration includes dermal, ocular, rectal, nasal, as well as administration by inhalation or by aerosol means.
  • compositions according to the invention can further include a pharmaceutically or veterinarially acceptable excipient, buffer, stabiliser, isotonicising agent, preservative or antioxidant or any other material known to those of skill in the art.
  • compositions typically include such substances so as to maintain the composition at a close to physiological pH or at least within a range of about pH 5.0 to about pH 8.0.
  • compositions according to the invention can also include active ingredients in addition to the at least one compound. Such ingredients will be principally chosen for their efficacy as antiangiogenic, antimetastatic, anti-inflammatory, anticoagulant, antithrombotic, antimicrobial agents but can be chosen for their efficacy against any associated condition.
  • compositions for administration to a human subject will include between about 0.01 and 100 mg of the compound per kg of body weight and more preferably between about 0.1 and 10 mg/kg of body weight.
  • the compounds can be included in compositions as pharmaceutically or veterinarially acceptable derivatives thereof.
  • derivatives of the compounds includes salts, coordination complexes with metal irons such as Mn 2+ and Zn 2+ , esters such as in vivo hydrolysable esters, free acids or bases, hydrates, or prodrugs.
  • Compounds having acidic groups such as phosphates or sulfates can form salts with alkaline or alkaline earth metals such as Na, K, Mg and Ca, and with organic amines such as triethylamine and Tris (2-hydroxyethyl) amine.
  • Salts can also be formed between compounds with basic groups, such as amines, with inorganic acids such as hydrochloric acid, phosphoric acid or sulfuric acid, or organic acids such as acetic acid, citric acid, benzoic acid, fumaric acid, or tartaric acid.
  • inorganic acids such as hydrochloric acid, phosphoric acid or sulfuric acid
  • organic acids such as acetic acid, citric acid, benzoic acid, fumaric acid, or tartaric acid.
  • Compounds having both acidic and basic groups can form internal salts.
  • Esters can be formed between hydroxyl or carboxylic acid groups present in the compound and an appropriate carboxylic acid or alcohol reaction partner, using techniques that will be well known to those of skill in the art.
  • Prodrug derivatives ofthe compounds ofthe invention can be transformed in vivo or in vitro into the parent compounds.
  • prodrugs are glycolipid derivatives in which one or more lipid moieties are provided as substituents on the moieties, leading to the release of the free form of the compound by cleavage with an enzyme having phospholipase activity.
  • Prodrugs of compounds of the invention include the use of protecting groups which may be removed in vivo to release the active compound or serve to inhibit clearance of the drug. Suitable protecting groups will be known to those of skill in the art and include an acetate group.
  • compounds according to the invention have utility in the manufacture of a medicament for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis and/or microbial infection.
  • Processes for the manufacture of such medicaments will be known to those of skill in the art and include the processes used to manufacture the pharmaceutical compositions described above.
  • the compounds falling within the scope of the invention have been found to have bind growth factors. In particular, it has been established that the compounds have affinity for aFGF, bFGF and NEGF.
  • the compounds thus have utility as antiangiogenic, antimetastatic and/or anti-inflammatory agents in the treatment of mammalian subjects including humans.
  • the uses of the compounds include the treatment of angiogenesis-dependent diseases such as angiogenesis associated with the growth of solid tumours, and proliferative retinopathies, as well as the treatment of inflammatory diseases and conditions such as rheumatoid arthritis.
  • the compounds may also activate the growth factors and could thus be used in cardiovascular treatments.
  • the compounds of the invention additionally have utility as anticoagulant or antithrombotic agents.
  • the compounds can therefore be used for both the prophylaxis and treatment of many thrombotic and cardiovascular diseases, the most notable of these being deep venous thrombosis, pulmonary embolism, thrombotic stroke, peripheral arterial thrombosis, unstable angina and myocardial infarction.
  • compositions of the charged aminoacid compounds can be delivered orally, the compounds are an attractive alternative to warfarin, a widely used oral anticoagulant with severe side effects.
  • the compounds of the invention additonally have been found to inhibit viral infection and thus have utility as antiviral agents in the treatment or prevention of many viral infections.
  • the compounds of the invention are particularly suited for the treatment or prevention of infection resulting from pathogens which utilise HS as an attachment/entry receptor [6], for example, HSN, HIN, Dengue virus, Yellow fever virus, Cytomegalovirus and Hepatitis C virus.
  • the compounds ofthe invention are also suited for the treatment or prevention of infection resulting from non-viral microbial pathogens which utilise HS as an attachment/entry, for example, Plasmodium (malaria). Most notable is the inhibition by the compounds ofthe invention ofthe cell-to-cell spread of HSN-1 and HSN-2. Having broadly described the invention, non-limiting examples ofthe compounds, their synthesis, and their biological activities, will now be given with reference to the accompanying Tables which will be briefly described in the following section of this specification.
  • the pure fractions were evaporated and co-evaporated (H 2 O) and then lyophilised (H 2 O) to yield the sulfated product.
  • H 2 O lyophilised
  • the product was passed through an ion- exchange resin column (AG ® -50W-X8, Na + form, 1x4 cm, deionized H O, 15 mL) in order to transfer the product uniformly into the sodium salt form.
  • the solution collected was evaporated and lyophilised to give the final product as a colourless glass or white power.
  • Size exclusion chromatography Size exclusion chromatography (SEC) was performed over Bio-Gel P-2 in a 5 x 100 cm column with a flow rate of 2.8 mL/min of 0.1 M NH 4 HCO 3 , collecting 2.8 min (7.8 mL) fractions. Fractions were analysed for carbohydrate content by TLC (charring) and/or for poly- charged species by the dimethyl methylene blue test, and then for purity by capillary electrophoresis (CE) and those deemed to be free of salt were pooled and lyophilised.
  • TLC charring
  • CE capillary electrophoresis
  • Dimethyl methylene blue Test Dimethyl methylene blue (DMB) reagent was prepared by dissolving 16 mg of DMB in 1 L of deionized water containing 3.04 g of glycine, 2.37 g of NaCI. 0.1 M HC1 (95 ml) was added to adjust the pH to 3.0. The stock solution was stored in a brown coloured bottle at r.t. (the solution was stable for at least 3 months under such conditions).
  • a 96-well microtitre plate was loaded with 10 ⁇ L of fraction solution per well. 55 ⁇ L of DMB stock solution was added into each used well. An instant colour change from blue to pink indicated the presence of polycharged species, i.e., sulfated product fractions.
  • General procedure for NIS glycosylations Glycosyl acceptor (1 eq), thioglycoside donor (1.1 eq), 500 mg of freshly activated powdered 3 A molecular sieves and 10 mL of dry DCM were stirred at -20° for 20 min before 1.3 eq of NIS and 1 drop of TfOH were added.
  • the mixture was loaded in a miniclave (B ⁇ chi AG, Uster/Switzerland) and stirred under hydrogen atmosphere (50 psi) for 2-10 h. Alternatively, the mixture was bubbled with hydrogen gas for 1 h then stirred at r.t. under 1 atmosphere of hydrogen for 1-5 days. The reaction was monitored by TLC (EtOAc or MeCN-water 10:1). The mixture was filtered and rinsed with MeOH, or EtOH. The filtrate was evaporated and dried under high vacuum, checked by 1H NMR, freeze- dried and used directly for sulfonation.
  • Step a Methyl 3,4,6-tri- -acetyl-2-0-benzyl- -O-galactopyranosyl-(l- ⁇ 4)-2,3,6- tri-O-benzyl- ⁇ -O-glucopyranoside.
  • Step b Methyl 3,4,6-tri-0-acetyl-a-O-galactopyranosyl-(l ⁇ 4)-fi-O-glucopyranoside.
  • Pearlman's catalyst (20 mg) and 20 ⁇ L of acetic acid were added to a solution of 90 mg (106 ⁇ mol) of methyl 3,4,6-tri-0-acetyl-2-0-benzyl-a-O-galactopyranosyl-(l ⁇ 4)-2,3,6- tri-O-benzyl- ⁇ -O-glucopyranoside in 10 mL of MeOH.
  • An atmosphere of hydrogen was applied with 3 vacuum purges and the suspension was stirred for 3 days.
  • Step c Methyl 2-0-sulfo- -O-galactopyranosyl-(l ⁇ 4)-2,3,6-tri-0-sulfo ⁇ -O-glucopyranoside, tetrasodium salt (PG2038)
  • the above disaccharide (32.2 mg, 0.667 mmol), was subjected to the standard sulfonation and deacetylation procedures to give the title compound as a white foam (4.0 mg, 7.8%, 96% purity, CE: 7.18 min).
  • Example 2 PG2046 and PG2047 Step a: 2-Azido-3, 4, 6-tri-0-benzoyl-2-deoxy-a-D-glucopyranosyl-(l ⁇ 4)-l, 6-anhydro-2-azido- 2-deoxy-3-0-benzyl- -D-glucopyranose
  • Step b 2-Deoxy-2-sulfamido-a-O-glucopyranosyl-(l ⁇ 4)-l, 6-anhydro-2-deoxy-2-sulfamido-3- 0-benzyl- -O-glucopyranose, disodium salt (PG2046)
  • Pearlman's catalyst 11 mg
  • ammonium formate 300 mg
  • the mixture was cooled to r.t., filtered (0.2 ⁇ m) and evaporated.
  • the crude amine was purified by SPE (300 mg C18 Waters cartridge, equilibrated with 5:95 MeOH:H 2 O, gradient eluted 5:95 to 100:0 MeOH:H 2 O) to yield 53 mg of the diamine (58%). Without further purification, to the diamine was added DMF (5 mL), SO 3 « Me 3 N (41 mg, 295 ⁇ mol) and NaHCO 3 (40 mg, 475 ⁇ mol). The mixture was heated to 60° for 1 h then cooled to rt and quenched with ice and Na 2 CO 3 (sat. aqueous).
  • Step c 2-Deoxy-2-sulfamido-a-O-glucopyranosyl-(l ⁇ 4)-l, 6-anhydro-2-deoxy-2-sulfamido- - O-glucopyranoside, disodium salt
  • PG2047 A mixture of 2-deoxy-2-sulfamido-a-O-glucopyranosyl-(l ⁇ 4)-l, 6-anhydro-2-deoxy-2- sulfamido-3-O-benzyl- ⁇ -O-glucopyranoside, disodium salt (12.9 mg, 20.8 ⁇ mol) and Pearlman's catalyst (5 mg) in purified water (2 mL) was subjected to 50 psi H 2 overnight.
  • Example 3 PG2039 and PG2037 Step a: Methyl 3,4-di-0-acetyl-2,6-di-0-benzyl-a-O-galactopyranosyl-(l ⁇ 4)-2,3,6- tri-0-benzyl- -O-glucopyranoside Methyl 2,3,6-tri-O-benzyl- ⁇ -O-glucopyranoside (287 mg; 618 ⁇ mol), 302 mg (618 ⁇ mol) of ethyl 3,4-di-0-acetyl-2,6-0-dibenzyl-l-thio-fi-D-galactopyranoside [21] and 700 mg of 3 A molecular sieves were subjected to the general NIS glycosylation procedure using 181 mg (803 ⁇ mol
  • Step b Methyl 3,4-di-0-acetyl-a-O-galactopyranosyl-(l ⁇ 4)- -D-glucopyranoside.
  • methyl 3,4-di-0-acetyl-2,6-di- 0-benzyl-a-O-galactopyranosyl-(l ⁇ 4)-2,3,6-tri-0-benzyl-fi-O-glucopyranoside 88 mg, 98.8 ⁇ mol
  • Step c Methyl 2,6-di-0-sulfo-a-O-galactopyranosyl-(l ⁇ 4)-2,3,6-tri-0-sulfo- -O- glucopyranoside, pentasodium salt (PG2039)
  • PG2039 pentasodium salt
  • 42 mg (95.4 ⁇ mol) of methyl 3,4-di-0-acetyl-a-O-galactopyranosyl-(l ⁇ 4)- -O-glucopyranoside was converted to the title compound as a white powder (14.8 mg, 18%, CE: 6.12 min).
  • Step d Methyl 2,6-di-0-benzyl-3,4-di-0-methyl ⁇ -O-galactopyranosyl-(l ⁇ 4)-2,3,6-tri-0- benzyl- -O-glucopyranoside
  • methyl 3,4-di-O- acetyl-2, 6-di-0-benzyl-a-O-galactopyranosyl-(l ⁇ 4)-2, 3, 6-tri-O-benzyl- ⁇ -O-glucopyranoside 72 mg, 80.8 ⁇ mol
  • Step e Methyl 3,4-di-0-methyl-a-O-galactopyranosyl-(l ⁇ 4)-fi-O-glucopyranoside
  • methyl 2,6-di-0-benzyl-3,4-di-0- methyl-a-O-galactopyranosyl-(l ⁇ 4)-2,3,6-tri-0-benzyl- -O-glucopyranoside (62.1 mg, 75.1 ⁇ mol) was deprotected to give the title compound as colourless gum (28 mg, 97%).
  • Stepf Methyl 3, 4-di-0-methyl-2, 6-di-0-sulfo-a-O-galactopyranosyl-(l ⁇ 4)-2, 3, 6-tri-O- sulfo-fi-O-glucopyranoside, pentasodium salt (PG2037) Following the standard sulfonation procedure, methyl 3,4-di-O-methyl-a-O- galactopyranosyl-(l ⁇ 4)- -O-glucopyranoside (28 mg, 72.8 ⁇ mol) gave the title compound (3.2 mg, 4.9%).
  • Example 4 PG2053 and PG2042 Methyl 4-0-Allyl-2,3-di-0-sulfo- -L-rhamnoside, disodium salt (PG2053).
  • the title compound was obtained from methyl 2,3-O-isopropylidene- -L- rhamnopyranoside [22] via the general alkylation (with allyl bromide) and deprotection procedure followed by the general sulfonation procedure, as a colourless powder.
  • CE t m 10.48 min.
  • Example 5 PG2024 Methyl 4-0-Benzyl-2,3-di-0-sulfo- -L-rhamnoside, disodium salt (PG2024)
  • the title compound was obtained from methyl 2,3-O-isopropylidene- -L- rhamnopyranoside via the general alkylation (with benzyl bromide) and deprotection procedure followed by the general sulfonation procedure, as a colourless powder.
  • CE t m 10.82 min.
  • Example 6 PG2054 Step a: Methyl 4-O-benzoyl-a- -rhamnoside A solution of methyl 2,3- -isopropylidene- - -rhamnopyranoside (200 mg, 920 ⁇ mol), benzoyl chloride (193 mg, 1.38 mmol) and Et 3 N (364 ⁇ L, 2.76 mmol) in DCM (10 mL) was stirred overnight. The resulting suspension (Et 3 N » HCl precipitates) was diluted with DCM (50 mL) and washed with NaHCO 3 (sat. aqueous), water then brine, dried (MgSO 4 ) and evaporated.
  • Step b 3-0-Benzyl-4,6-di-0-sulfo-l,2-dihydro-O-glucal, Disodium salt (PG2041). 4,6-0-Benzylidene-l,2-dihydro-D-glucal was subjected to the alkylation (benzyl bromide), de-protection and sulfonation general procedures to yield the title compound as a colourless powder.
  • CE t m 15.40 min.
  • Step a 1, 6-Anhydro-3-0-methyl- -O-glucopyranose.
  • -Toluenesulfonyl chloride (790 mg, 4.14 mmol) was added to a cooled (0°) suspension of 3-O-methyl-D-glucopyranose (804 mg, 4.14 mmol) in pyridine (10 mL) and the reaction mixture stirred (0° ⁇ r.t, 1.5 h).
  • Ac 2 O (1.5 mL, 15 mmol) and N,N-dimethylaminopyrdine (50 mg) were then introduced and stirring continued (r.t., 4 h).
  • the residual oil was subjected to flash chromatography (20-50% EtOAc/hexanes) to yield an inseparable mixture of 2,4-di-0-acetyl-l,6-anhydro-3-0-methyl- -D-glucopyranose (a) and l,2,4-tri-0-acetyl-3-0-methyl-6-0-tosyl- -O-glucopyranose (b) (in a ratio of 3:1) as a pale yellow oil (466 mg). The ratio was determined by integration of the H-l and 3-OMe signals observed in the 1H ⁇ MR spectrum.
  • Example 9 PG2012 and PG2013 Step a: ' N-benzyl- ⁇ N-(cyclohexylacetamido)-l, 2, 3, 4-tetra-O-acetyl- -glucuronamide
  • D-glucuronic acid (0.950 g, 4.89 mmol)
  • solutions of each of the following three reagents benzylamine (2 M in MeO ⁇ , 2.45 mL, 4.89 mmol), formaldehyde (2 M in MeO ⁇ , 2.45 mL, 4.89 mmol) and cyclohexylisocyanide (1 M in MeO ⁇ , 4.89 mL, 4.89 mmol) were loaded into a reaction vessel and the mixture stirred at r.t.
  • Step b Y ⁇ -benzyl- ⁇ -(cyclohexylacetamido)-l, 2, 3, 4-teti"a-0-sulfo-a ⁇ D-glucuronamide, tetrasodium salt (PG2012) and ⁇ -benzyl- ⁇ N-(cyclohexylacetamido)-l,2,3-tri-0-sulfo-a-D- glucuronamide, trisodium salt (PG2013) Following the general procedure for deacetylation, the above tetraacete (0.441 g, 0.747 mmol) was deacetylated to give ⁇ N-benzyl- ⁇ N-(cyclohexylacetamido)-D-glucuronamide as pale- yellow glass (0.316 g, 100%).
  • the polar part was purified by LH20 column (x2) and ion exchange column to give tetrasulfate PG2012 as an off-white powder after lyophilisation (7.6 mg, 1.5%).
  • Example 10 PG2064 Step a: 2-(N-acetyl- ' N-cyclohexyl)amino- ' N-(methyl 2,3,4-tri-0-benzyl-6-deoxy-a-D- mannopyranos-6-yl)acetamide
  • acetic acid (2 M in MeOH, 60 ⁇ L, 119 ⁇ mol)
  • cyclohexylamine (2 M in MeOH, 60 ⁇ L, 119 ⁇ mol
  • formaldehyde 2 M in MeOH, 60 ⁇ L, 119 ⁇ mol
  • methyl 2,3,4-tri-0-benzyl-6-deoxy-6-isocyano- -D-mannopyranoside 0.721 M in CHC1 3 , 150 ⁇ L, 108 ⁇ mol
  • Step b 2-(N-acetyl- ⁇ N-cyclohexyl)amino- ' N-( methyl 6-deoxy -2,3,4-tri-O-sulfo-a-D- mannopyranos ⁇ 6-yl)acetamide, trisodium salt (PG2064)
  • PG2064 2-(N-acetyl- ⁇ N-cyclohexyl)amino- ' N-( methyl 6-deoxy -2,3,4-tri-O-sulfo-a-D- mannopyranos ⁇ 6-yl)acetamide, trisodium salt
  • Step a Following the general procedure for the Ugi reaction, monomethyl succinate (15.7 mg, 0.119 mmol) and a solution of each ofthe following three reagents: ethylamine (2 M in MeOH, 60 ⁇ L, 119 ⁇ mol), formaldehyde (2 M in MeOH, 60 ⁇ L, 119 ⁇ mol) and methyl 2,3,4-tri-O- benzyl-6-deoxy-6-isocyano-a-D-mannopyranoside (0.721 M in CHCI 3 , 150 ⁇ L, 108 ⁇ mol) was loaded into a 2 mL sample vial and the mixture stirred at r.t. for 19 h.
  • ethylamine (2 M in MeOH, 60 ⁇ L, 119 ⁇ mol
  • formaldehyde 2 M in MeOH, 60 ⁇ L, 119 ⁇ mol
  • 13 C (100 MHz, CDCI 3 , ⁇ 77.0): major rotamer, 173.32, 171.73, 168.94, 138.20, 138.17, 138.09, 128.22, 128.18, 128.10, 127.76, 127.63, 127.49, 127.46, 98.94, 79.97, 75.38, 75.01, 74.84, 73.01, 72.01, 70.10, 54.63, 51.63, 49.84, 43.59, 39.63, 28.99, 27.24, 13.45.
  • Step b (PG2068) Following the general procedure for deprotection of benzyl ethers, a mixture of the above tribenzyl ether (46.8 mg, 0.0706 mmol), 20% palladium on activated charcoal (30 mg) in MeOH (3 mL) was stirred under hydrogen atmosphere at 50 psi for 2 h.
  • Step a 3-Chlorophenylacetic acid (223 mg, 1.307 mmol) was dissolved in MeCN (3 mL). Ammonia solution (28%, 0.26 mL, 3.8 mmol) was added. The mixture was swirled for a while and evaporated in vacuo. The residue was suspended in MeCN (3 mL), filtered and the white solid was washed with MeCN and freeze-dried to afford ammonium 3-chlorophenylacetate (0.195 g, 80%).
  • 13 C (100 MHz, CDC1 3 , ⁇ 77.0): 169.67, 166.81, 138.27, 138.11, 137.70, 135.16, 134.31, 129.82, 129.40, 128.34, 128.31, 128.01, 127.85, 127.78, 127.70, 127.68, 127.60, 127.53, 127.46, 98.90, 79.96, 75.08, 75.04, 74.71, 73.58, 72.73, 72.19, 72.13, 69.78, 68.49, 63.17, 40.25, 39.27.
  • Step b (PG2075) Following the general procedure for deprotection of benzyl ethers, a mixture of the above tetrabenzyl ether (34.8 mg, 0.0439 mmol), 20% palladium on activated charcoal (26 mg) in MeOH (2 mL) was stirred under hydrogen atmosphere at 50 psi for 2 h. General work-up gave the tetrol intermediate as a colourless gum. Following the general procedure for sulfonation, the above tetrol was sulfonated . The residue was purified via SEC (Bio-Gel P-2).
  • Example 13 PG2014 Step a Following the general procedure for the Ugi reaction, 2-(benzyl3,4,6-tri-0-benzyl- -D- mannopyranoside-2-yl) acetic acid (50 mg, 0.0835 mmol) and a solution of each following three reagents: benzylamine (2 M in MeOH, 41.8 ⁇ L, 0.0835 mmol), formaldehyde (2 M in MeOH, 41.8 ⁇ L, 0.0835 mmol) and 2-isocyanoethyl 2,3,4,6-tetra-O-benzyl- -D- mannopyranoside (0.415 M in MeOH, 201.4 ⁇ L, 0.0835 mmol) was loaded into a 2 mL sample vial and the mixture stirred at r.t.
  • Step b (PG2014). Following the general procedure for the deprotection of benzyl ethers, a mixture of the above octabenzyl ether (35 mg, 0.0267 mmol) and 20% palladium on activated charcoal (10 mg) in EtOH (4 mL) was stirred under hydrogen atmosphere at 50 psi for 2 h. General work- up gave the octol intermediate as a colourless gum. Following the general procedure for sulfonation, the above octol was sulfonated (sulfur trioxide trimethylamine complex, 60 °C, 19 h). The residue was purified via SEC (Bio-Gel P-2).
  • Example 14 PG2016 Step a Following the general procedure for the Ugi reaction, tr r ⁇ -l,4-diaminocyclohexane (6.3 mg, 0.055 mmol) and a solution of each following three reagents: 2 -(methyl 2,3,4-tri-O- benzyl -O-mannopyranoside-6-yl)acetic acid (0.91 M in MeOH, 121 ⁇ L, 0.11 mmol), formaldehyde (2 M in MeOH, 55 ⁇ L, 0.11 mmol) and cyclohexylisocyanide (1 M in MeOH, 110 ⁇ L, 0.11 mmol) was loaded into a 2 mL sample vial and the mixture stirred at r.t.
  • 2 -(methyl 2,3,4-tri-O- benzyl -O-mannopyranoside-6-yl)acetic acid (0.91 M in MeOH, 121 ⁇ L, 0.11 mmol)
  • PG2016 Following the general procedure for the deprotection of benzyl ethers, a mixture of the above hexabenzyl ether (33 mg, 0.0235 mmol) and 20% palladium on activated charcoal (65 mg) in MeOH (2.8 mL) was stirred under hydrogen atmosphere at 1 atm for 5 days. General work-up gave the hexol intermediate as a colourless gum. Following the general procedure for sulfonation, the above hexol was sulfonated. The residue was purified via sequential column chromatography (SEC on Bio-Gel P-2 followed by ion exchange column) to give PG2016 as a white powder (12.2 mg, 35%).
  • Example 15 PG2015 Step a Following the general procedure for the Ugi reaction, 3,3-dimethylglutaric acid (7.1 mg, 0.0443 mmol) and a solution of each following three reagents: 3-aminopropyl 2,3,4,6- tetra-O-benzyl- -D-mannopyranoside (0.642 M in MeOH, 138 ⁇ L, 0.0886 mmol), formaldehyde (2 M in MeOH, 44.3 ⁇ L, 0.0886 mmol) and cyclohexylisocyanide (1 M in MeOH, 88.6 ⁇ L, 0..0886 mmol) were loaded into a 2 mL sample vial and the mixture stirred at r.t.
  • 3-aminopropyl 2,3,4,6- tetra-O-benzyl- -D-mannopyranoside 0.642 M in MeOH, 138 ⁇ L, 0.0886 mmol
  • formaldehyde (2 M in Me
  • Step b (PG2015). Following the general procedure for the deprotection of benzyl ethers, a mixture of the above hexabenzyl ether (32.3 mg, 0.0202 mmol), 20% palladium on activated charcoal (41 mg) in MeOH (2.8 mL) was stirred under hydrogen atmosphere at 1 atm for 5 days. General work- up gave the octol intermediate as a colourless gum. Following the general procedure for sulfonation, the above octol was sulfonated. The residue was purified via SEC (Bio-Gel P-2) to give PG2015 as a white powder (12.6 mg, 37%).
  • Ethyl 2,6-Di-0-benzyl-3,4-di-0-sulfo- ⁇ -D-galactopyranoside, disodium salt (PG2155) The title compound was obtained from ethyl 2,6-di-0-benzyl-3,4-di-0-sulfo- ⁇ -D- galactopyranoside [23] via the general sulfonation procedure as a colourless powder.
  • Step a Methyl 4-0-allyl-6-azido-6-deoxy-2,3-di-0-isopropylidene- -D-mannopyranoside
  • methyl 6-azido-6-deoxy-a-D-mannopyranoside 311 mg, 1.419 mmol
  • 2,2-dimethoxypropane 4.7 mL, 0.3 M
  • ( ⁇ )-camphor-lO-sulfonic acid (16 mg, 0.0709 mmol, 5 mol%).
  • Step b Methyl 4-0-allyl-6-azido-6-deoxy- -D-mannopyranoside
  • Methyl 4-0-allyl-6-azido-6-deoxy-2, 3-di-O-isopropylidene-a-D-mannopyranoside (56 mg, 0.187 mmol) was dissolved in MeCN-MeOH-H 2 O (3 mL, 3 mL and 0.2 mL respectively) and treated with p-toluenesulfonic acid monohydrate (7 mg, 0.0374 mmol, 20 mol%). The mixture was stirred at r.t. for 5 h and triethylamine (0.4 mL) added.
  • the mixtures were combined and evaporated.
  • the residue was re-dissolved in dichloromethane (50 mL) and stirred with IM ammonium chloride solution (50 mL).
  • IM ammonium chloride solution 50 mL
  • the less polar spots were slowly disappeared and converted into the polar product.
  • the conversion was not further improved after 40 min:
  • the dichloromethane phase was separated and stirred with 0.5 M HC1 solution (50 mL) for another 20 min. TLC indicated no further change.
  • the DCM phase was separated and washed with brine- IM NaOH, dried (MgSO 4 ).
  • Step b Methyl 6-azido-3-0-benzyl-6-deoxy-a-D-mannopyranoside and methyl 6-azido-2-0- benzyl-6-deoxy- a-D-mannopyranoside
  • DMF 7.8 mL, 0.1 M
  • sodium cyanoborohydride 589 mg, 9.37 mmol, 12 eq
  • molecular sieve 3A 1 g
  • Step c Methyl 6-azido-2-0-benzyl-6-deoxy-2,3-di-0-sulfonato-a-D-mannopyranoside disodium salt (PG2160) Methyl 6-azido-2-0-benzyl-6-deoxy-a-D-mannopyranoside was sulfonated according to the standard procedure to yield the title compound as a white powder, 64 mg, 59%.
  • Step d Methyl 6-azido-3-0-benzyl-6-deoxy-2,4-di-0-sulfonato-a-D-mannopyranoside disodium salt (PG2161) Methyl 6-azido-3-0-benzyl-6-deoxy- a-D-mannopyranoside (64 mg) was sulfonated according to the standard procedure to yield the title compound as a white powder, 58 mg, 66%.
  • Example 19 PG2170 Allyl 6-azido-2, 3-0-disulfonato-6-deoxy-4-0-(l-naphthylmethyl)- a-D-mannopyranoside disodium salt (containing 10% of 2-naphthylmethyl isomer) (PG2170)
  • the source ofthe minor isomer is the commercial 9:1 mixture of 1- and 2-bromomethylnapthalene used in step a.
  • Example 20 Biological Testing of Compounds Methods 1.
  • Growth Factor Binding Binding affinities of ligands for the growth factors were measured using a surface plasmon resonance (SPR) based solution affinity assay. The principle of the assay is that heparin immobilised on a sensorchip surface distinguishes between free and bound growth factor in an equilibrated solution of the growth factor and a ligand. Upon injection of the solution, the free growth factor binds to the immobilised heparin, is detected as an increase in the SPR response and its concentration thus determined.
  • SPR surface plasmon resonance
  • a decrease in the free growth factor concentration as a function of the ligand concentration allows for the calculation of the dissociation constant, K d . It is important to note that ligand binding to the growth factors can only be detected when the interaction involves the heparin binding site, thus eliminating the chance of evaluating non-specific binding to other sites on the protein.
  • a 1 : 1 stoichiometry has been assumed for all protein: ligand interactions. The preparation of heparin-coated sensorchips, via immobilisation of biotinylated BSA- heparin on a streptavidin-coated sensorchip, has been described [24].
  • Heparin has also been immobilised via aldehyde coupling using either adipic acid dihydrazide or 1,4-diaminobutane.
  • solutions were prepared containing a fixed concentration of protein and varying concentrations ofthe ligand in buffer.
  • Ligands binding to FGF-1 and VEGF were measured in HBS-EP buffer (10 mM HEPES, pH 7.4, 150 mM NaCI, 3.0 mM EDTA and 0.005% (v/v) polysorbate 20), while binding to FGF-2 was measured in HBS-EP buffer containing 0.3 M NaCI [24].
  • HBS-EP buffer 10 mM HEPES, pH 7.4, 150 mM NaCI, 3.0 mM EDTA and 0.005% (v/v) polysorbate 20
  • binding to FGF-2 was measured in HBS-EP buffer containing 0.3 M NaCI [24].
  • Prior to injection samples were maintained at 4 °C to maximise protein stability.
  • K d values are the values fit, using the binding equation, to a plot of [P] versus [L] tota i- Where K d values were measured in duplicate, the values represent the average ofthe duplicate measurements. It has been shown that GAG mimetics that bind tightly to these growth factors elicit a biological response in vivo [24].
  • HSV herpes simplex virus
  • HSV-1 and HSV-2 two types of herpes simplex virus
  • GMK AHl African green monkey kidney cells
  • the viral strains used were herpes simplex virus type 1 (HSV-1) KOS321 strain [27] and HSV-2 strain 333 [28]. In both assays the compounds were tested at 200 ⁇ M.
  • HSV-1 herpes simplex virus type 1
  • HSV-2 strain 333 [28] HSV-2 strain 333 [28]. In both assays the compounds were tested at 200 ⁇ M.
  • the compounds were mixed with the virus, incubated for 10 min at room temperature and then the mixture was added to cells, and kept on cells for lh only to allow (or not) the virus attachment to/entry into the cells. Thus this assay reflects whether or not the compound in question has the ability to bind to the virus particles and block its attachment to/entry into the cells. An inhibition is manifested as a decreased number of viral plaques.
  • HSV spread The next assay, termed HSV spread, relies on the addition of compound to the cells after the virus attachment/entry steps have already occurred. This assay reflects whether the examined compound has the ability to inhibit virus transmission from an infected to an uninfected cell (cell-to-cell spread) and in addition whether the compound has the ability to enter the cells and inhibit viral replication. Lack of compound activity in the assay of virus infectivity but some activity in the virus spread assay suggests that the compound acts by entering the cells and inhibition of viral replication step(s). An inhibition is manifested as a reduction in the size of viral plaques.
  • results are expressed as % of control, ie., as the number (infectivity assay) or the size (spread assay) of viral plaques developed in the presence of compound relative to the mock-treated controls (no compound). Results The results ofthe tests as described in the preceding section are presented in Tables 1 to
  • R D CH 2 OS0 3 Na
  • R G H 2038 77.9 ⁇ M 2.10 mM 368 ⁇ M
  • R A OMe
  • R F ,R H OH
  • R B ,Rc,R E ,R OS0 3 Na 2039 21.8 ⁇ M 3.50 mM 1.27 mM
  • Rj/R K H/OMe (anomeric mixture);
  • R S -R N -CH 2 0-; 2045 392 ⁇ M 3.40 mM 1.07 mM
  • R M OS0 3 Na;
  • R Q OBn;
  • R L ,R 0 ,R P ,R R H
  • R ⁇ NHCOCH 2 ⁇ Ph(2,4-di-Cl);
  • R Mj R N ,R Q OS ⁇ 3 Na; 2096 35.7 ⁇ M 141 ⁇ M 20.4 ⁇ M
  • R s * CH 2 OS0 3 Na;
  • R P ,RR H
  • Rj/R K H/OMe (anomeric mixture);
  • RS-RN -CH 2 0-; 2165 1.13 mM ⁇ 25.5 mM 1.70 mM
  • R M OBn;
  • R Q OS0 3 Na;
  • RJ,R K ,RL,RO,RP,RR H 2166 3.60 mM 1.90 mM 2.70 mM
  • R ⁇ l,2,3,4-tetra-0-sodi ⁇ un sulfonato-D-glucuronoyl
  • R x COCH 2 C(CH 3 ) 2 CH 2 CO; 2015 2.94 ⁇ M 7.56 ⁇ M 267 nM sulfo- ⁇ -D-mannopyranos- 1 -0-yl)-propyl

Abstract

The invention relates to compounds that are designed to mimic the structure of GAGs; methods for the preparation of the compounds; compositions comprising the compounds; and use of the compounds and compositions thereof for the anti-angiogenic, anti-metastatic, anti-inflammatory, anticoagulant, antithrombotic, and/or antimicrobial treatment of a mammalian subject.

Description

GLYCOSAMINOGLYCAN (GAG) MIMETICS
TECHNICAL FIELD The invention that is the subject of this application lies in the area of compounds that mimic the structure of certain carbohydrates. More particularly, the invention lies in the area of glycosaminoglycan (GAG) mimetics. Specifically, the invention relates to compounds comprising at least one charged group that are designed to mimic the structure of GAGs. The invention also relates to methods for the preparation of the compounds, compositions comprising the compounds, and use of the compounds and compositions thereof for the antiangiogenic, antimetastatic, anti-inflammatory, anticoagulant, antithiOmbotic, and/or antimicrobial treatment of a mammalian subject. The invention further relates to the use ofthe compounds and compositions thereof in the treatment of a mammalian subject having a condition amenable to treatment with such agents. BACKGROUND ART Glycosaminoglycans (GAGs) are linear, polyanionic polysaccharides that are produced by most animal cells and are usually found attached to a protein core [1,2]. GAGs occur abundantly (as proteoglycans) and are extruded by cells to the cell surface and into the extracellular matrix (ECM) [3], It has been recognised that GAGs, especially those belonging to the heparan sulfate (HS) family (HS-GAGs), mediate numerous physiological processes. For example, HS-GAGs play key roles in cell growth and development, angiogenesis, coagulation, tumour metastasis, cell adhesion, activation of growth factors, binding of cytokines and chemokines, and infection by bacteria and viruses [4-6]. In recent years there has been a dramatic increase in the list of proteins that interact with GAGs and the list continues to grow. The emerging view is that unique sequences of extracellular GAGs bind specifically to important proteins and by doing so influence fundamental biological processes. It has been shown that molecules that mimic the structure of certain GAGs — which molecules are referred to as "GAG mimetics" — can bind to GAG-binding proteins and modulate their biological activity: e.g., the activation of AT-III by various pentasaccharides [7,8], or the activation of fibroblast growth factors (FGFs) by sucrose octasulfate [9]. Similarly, it has been shown that GAG mimetics can antagonise the binding of a GAG to its target protein and in so doing inhibit that protein's biological or disease function. For example, anticancer agents that have been developed to target HS-binding angiogenic growth factors include polysulfonated compounds [10], suramin and the related suradistas [11], and sulfated oligosaccharides [12,13]. The present invention relates to novel, small molecule GAG mimetics that bind to GAG-binding proteins and modulate their functions. The compounds incorporate at least one negatively charged group (preferably a sulfo group) to interact with the positively charged residues in the GAG-binding site of the target proteins, and also contain one or more substituents to form interactions with other protein residues in and around the above-mentioned binding site. Important and distinguishing features ofthe compounds described herein are that they have fewer sulfo groups and are of lower molecular weight than previously described polysulfated GAG mimetics such as the sulfated oligosaccharides [12,13]. Another important feature is that their structures are based on cyclic scaffolds (e.g., a monosaccharide) with sulfo groups and other substituents placed in specific, pre-defined orientations about the ring, thus differing significantly from the simple, randomly charged GAG mimetics described by Kisilevsky [14]. The binding of the compounds described herein to a selection of HS-binding, angiogenic growth factors is demonstrated via a surface plasmon resonance (SPR) solution affinity assay. Additionally, a selection of compounds are shown to inhibit the HS-mediated infection of cells and cell-to-cell spread by herpes simplex virus. One aspect of the present invention is the utilisation of the Ugi reaction [15,16] to provide a diverse array of GAG mimetics. The capacity for variation in the manner in which the individual charged structures are connected to one another or to other functional groups as well as the scope of application to mimic the diverse structural variation of GAGs is demonstrated. As will be apparent to those skilled in the art, the functionalisation of the cyclic scaffolds is not limited to the Ugi reaction. For example, the use of many other reactions such as alkylation, acylation and cycloaddition is demonstrated. SUMMARY OF THE INVENTION It is an object of the invention to provide novel charged compounds that have utility as GAG mimetics. It is a further object of the invention to provide effective synthetic routes for the preparation ofthe subject compounds. According to a first embodiment of the invention, there is provided a compound of the formula
Figure imgf000004_0001
wherein: n is an integer of from 0 to 2; Z is N, N(O), O. S, S(O), S(O)2, P, P(O), P(O)2, Si, Si(O), or Si(O)2; each X is independently C, C(O), N, N(O), O, S, S(O), S(O)2, P. P(O), P(O)2, Si, Si(O),)2 or is a bond; and each of R\ to R6 is independently a bond or is selected from the group consisting of: hydrogen; halogen; straight chain, cyclic, branched, substituted, heterocyclic, heteroatom substituted or unsubstituted alkyl, alkenyl, alkynyl, aryl, or heteroaryl; phosphoryl groups such as phosphate, thiophosphate -O-P(S)(OH)2; phosphate esters -O-P(O)(OR)2; thiophosphate esters -O-P(S)(OR)2; phosphonate -O-P(O)OHR; thiophosphonate -O-P(S)OHR; substituted phosphonate -O-P(O)OR1R2; substituted thiophosphonate -O-P(S)OR1R2; -O-P(S)(OH)(SH); and cyclic phosphate; other phosphorus containing compounds such.as phosphoramidite -O-P(OR)-NR1R2; and phosphoramidate -O-P(O)(OR)-NR!R2; sulfur groups such as -O-S(O)(OH), -SH, -SR, -S(→O)-R, S(O)2R, RO-S(O)2\ -O-SO2NH2, -O-SO2RiR2 or sulfamide -NHSO2NH2; amino groups such as -NHR, -N ιR2, -NHAc, -NHCOR, -NH-O-COR, - NHSO3, -NHSO2R, -N(SO2R)2, and/or amidino groups such as -NH- C(=NH)NH2 and/or ureido groups such as -NH-CO-NRiR2 or thiouriedo groups such as -H-C(S)-NH2; another unit ofthe structure I, attached through any position, where Z, X and Rt to R6 are as defined above; or a substructure based upon a group ofthe following formula:
Figure imgf000005_0001
wherein: Y is a bond or is selected from the group consisting of: straight chain, cyclic, branched, substituted, heterocyclic, heteroatom substituted or unsubstituted alkyl; straight chain, cyclic, branched, substituted, heterocyclic, heteroatom substituted or unsubstituted acyl; and aryl, substituted aryl, heteroaryl; and each of R7 to Rπ is independently at least one structure according to formula I, or a structure according to formula II; with the provisos that: when Z is O, and X is O or a bond, then all of Ri to R5 are not H or CH OH; or when Z is N and X is O or a bond, then all of Ri to R6 are not H. According to a second embodiment ofthe invention, there is provided a pharmaceutical or veterinary composition for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis, and/or microbial infection, which composition comprises at least one compound according to the first embodiment together with a pharmaceutically or veterinarially acceptable carrier or diluent for said at least one compound. According to a third embodiment of the invention, there is provided the use of a compound according to the first embodiment in the manufacture of a medicament for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis, and/or microbial infection. According to a fourth embodiment of the invention there is provided a method for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis, and/or microbial infection, which method comprises administering to the subject an effective amount of at least one compound according to the first embodiment, or a composition comprising said at least one compound. In other embodiments of the invention, there are provided processes for synthesising the compounds according to the first embodimrent as defined above. With further regard to the compounds of the first embodiment, if not otherwise specified, alkyl, aryl and other substituent groups are used in accordance with their usual meaning in the art. For example, alkyl and aryl groups would normally have from 1 to 10 carbon atoms. Additionally, two ofthe groups Ri to R5 may be connected to each other to form a bicyclic strucure; or the cyclic structure of formula I may contain a double bond, i.e., two contiguous XRi to XR5 groups may be bonds. Preferred compounds ofthe invention have the general structures of formulae III— VI, as defined in Tables 1-4 below. In order that the invention may be more readily understood and put into practice, one or more preferred embodiments thereof will now be described, by way of example only. DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS The following abbreviations are used herein: GAG glycosaminoglycan HS heparan sulfate FGF fibroblast growth factor aFGF acidic fibroblast growth factor (or FGF-1) bFGF basic fibroblast growth factor (or FGF-2) NEGF vascular endothelial growth factor SPR surface plasmon resonance HSN herpes simplex virus The present inventors have found that a broad range of compounds with GAG mimetic properties can be synthesised using a number of different strategies as illustrated below in the examples. These compounds have utility in the prevention or treatment in mammalian subjects of a disorder resulting from angiogenesis, metastasis, inflammation, microbial infections, coagulation or thrombosis. This utility results from the ability of the compounds to modulate the activity of GAG-binding proteins responsible for disease processes. The GAG mimetics of the invention, as indicated above, can be synthesised using a number of different routes, including the Ugi reaction, and generally incorporating sulfonation in the process. Preferred compounds according to the first embodiment of the invention as defined above include those embraced by generic structures I and II and those included in Tables 1-4 below. As indicated above, the compounds according to the invention have utility in the prevention or treatment in mammalian subjects of a disorder resulting from angiogenesis, metastasis, inflammation, microbial infection, coagulation or thrombosis. The compounds have particular utility in the treatment of the foregoing disorders in humans. The compounds are typically administered as a component of a pharmaceutical composition as described in the following paragraphs. Pharmaceutical compositions for oral administration can be in tablet, capsule, powder or liquid form. A tablet can include a solid carrier such as gelatine or an adjuvant or an inert diluent. Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, a mineral oil or a synthetic oil. Physiological saline solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. Such compositions and preparations will generally contain at least 0.1 wt% of the compound. Parenteral administration includes administration by the following routes: intravenously, cutaneously or subcutaneously, nasally, intramuscularly, intraocularly, transepithelially, intraperitoneally and topically. Topical administration includes dermal, ocular, rectal, nasal, as well as administration by inhalation or by aerosol means. For intravenous, cutaneous or subcutaneous injection, or injection at a site where treatment is desired, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-firee and has suitable pH, isotonicity and stability. Those of skill in the art will be well able to prepare suitable solutions using, for example, solutions of the subject compounds or derivatives thereof. In addition to the at least one compound and a carrier or diluent, compositions according to the invention can further include a pharmaceutically or veterinarially acceptable excipient, buffer, stabiliser, isotonicising agent, preservative or antioxidant or any other material known to those of skill in the art. It will be appreciated by the person of skill that such materials should be non-toxic and should not interfere with the efficacy of the compound(s). The precise nature of any additive may depend on the route of administration of the composition: that is, whether the composition is to be administered orally or parenterally. With regard to buffers, aqueous compositions typically include such substances so as to maintain the composition at a close to physiological pH or at least within a range of about pH 5.0 to about pH 8.0. Compositions according to the invention can also include active ingredients in addition to the at least one compound. Such ingredients will be principally chosen for their efficacy as antiangiogenic, antimetastatic, anti-inflammatory, anticoagulant, antithrombotic, antimicrobial agents but can be chosen for their efficacy against any associated condition. A pharmaceutical or veterinary composition according to the invention will be administered to a subject in either a prophylactically effective or a therapeutically effective amount as necessary for the particular situation under consideration. The actual amount of at least one compound administered by way of a composition, and rate and time-course of administration, will depend on the nature and severity of the condition being treated or the prophylaxis required. Prescription of treatment such as decisions on dosage and the like will be within the skill of the medical practitioner or veterinarian responsible for the care of the subject. Typically however, compositions for administration to a human subject will include between about 0.01 and 100 mg of the compound per kg of body weight and more preferably between about 0.1 and 10 mg/kg of body weight. The compounds can be included in compositions as pharmaceutically or veterinarially acceptable derivatives thereof. As used herein "derivatives" of the compounds includes salts, coordination complexes with metal irons such as Mn2+ and Zn2+, esters such as in vivo hydrolysable esters, free acids or bases, hydrates, or prodrugs. Compounds having acidic groups such as phosphates or sulfates can form salts with alkaline or alkaline earth metals such as Na, K, Mg and Ca, and with organic amines such as triethylamine and Tris (2-hydroxyethyl) amine. Salts can also be formed between compounds with basic groups, such as amines, with inorganic acids such as hydrochloric acid, phosphoric acid or sulfuric acid, or organic acids such as acetic acid, citric acid, benzoic acid, fumaric acid, or tartaric acid. Compounds having both acidic and basic groups can form internal salts. Esters can be formed between hydroxyl or carboxylic acid groups present in the compound and an appropriate carboxylic acid or alcohol reaction partner, using techniques that will be well known to those of skill in the art. Prodrug derivatives ofthe compounds ofthe invention can be transformed in vivo or in vitro into the parent compounds. Typically, at least one of the biological activities of a parent compound may be suppressed in the prodrug form of the compound, and can be activated by conversion of the prodrug to the parent compound or a metabolite thereof. Examples of prodrugs are glycolipid derivatives in which one or more lipid moieties are provided as substituents on the moieties, leading to the release of the free form of the compound by cleavage with an enzyme having phospholipase activity. Prodrugs of compounds of the invention include the use of protecting groups which may be removed in vivo to release the active compound or serve to inhibit clearance of the drug. Suitable protecting groups will be known to those of skill in the art and include an acetate group. As also indicated above, compounds according to the invention have utility in the manufacture of a medicament for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis and/or microbial infection. Processes for the manufacture of such medicaments will be known to those of skill in the art and include the processes used to manufacture the pharmaceutical compositions described above. The compounds falling within the scope of the invention have been found to have bind growth factors. In particular, it has been established that the compounds have affinity for aFGF, bFGF and NEGF. The compounds thus have utility as antiangiogenic, antimetastatic and/or anti-inflammatory agents in the treatment of mammalian subjects including humans. The uses of the compounds include the treatment of angiogenesis-dependent diseases such as angiogenesis associated with the growth of solid tumours, and proliferative retinopathies, as well as the treatment of inflammatory diseases and conditions such as rheumatoid arthritis. The compounds may also activate the growth factors and could thus be used in cardiovascular treatments. As further indicated above, the compounds of the invention additionally have utility as anticoagulant or antithrombotic agents. The compounds can therefore be used for both the prophylaxis and treatment of many thrombotic and cardiovascular diseases, the most notable of these being deep venous thrombosis, pulmonary embolism, thrombotic stroke, peripheral arterial thrombosis, unstable angina and myocardial infarction. Since compositions of the charged aminoacid compounds can be delivered orally, the compounds are an attractive alternative to warfarin, a widely used oral anticoagulant with severe side effects. The compounds of the invention additonally have been found to inhibit viral infection and thus have utility as antiviral agents in the treatment or prevention of many viral infections. The compounds of the invention are particularly suited for the treatment or prevention of infection resulting from pathogens which utilise HS as an attachment/entry receptor [6], for example, HSN, HIN, Dengue virus, Yellow fever virus, Cytomegalovirus and Hepatitis C virus. Similarly, the compounds ofthe invention are also suited for the treatment or prevention of infection resulting from non-viral microbial pathogens which utilise HS as an attachment/entry, for example, Plasmodium (malaria). Most notable is the inhibition by the compounds ofthe invention ofthe cell-to-cell spread of HSN-1 and HSN-2. Having broadly described the invention, non-limiting examples ofthe compounds, their synthesis, and their biological activities, will now be given with reference to the accompanying Tables which will be briefly described in the following section of this specification. General Procedures General procedure for al ylation and deprotection ofdiols The diol (1 eq.) in DMF was added dropwise to a cooled (0°), stirred suspension of pre- washed (hexane) ΝaH (5 eq.) in DMF. Once the addition was complete, stirring was maintained (0°→r.t., 20 min). The mixture was cooled (0°, 5 min) and the alkyl halide (2 eq.) was introduced dropwise with continued stirring (0°— »-r.t., o/n). The mixture was cooled once again (0°) and MeOH (5 mL) was introduced with continued stirring (5 min). The solvent was evaporated and the residue subjected to workup (EtOAc) and flash chromatography to homogeneity (TLC). This residue was co-evaporated (2 x 10 mL MeCΝ). The crude mixture andjXTsOH-HjO (50 mg) in MeOH/MeCΝ (1:1) was heated under reflux (1 h). The mixture was cooled (r.t.) and Et3Ν (100 μL) was added prior to evaporation ofthe solvent. The residue was subjected to flash chromatography (EtOAc/hexane) to yield the diol. General procedure for sulfonation of alcohols A mixture of the alcohol and SOytrimethylamine (2 eq per hydroxyl group) in DMF was heated (60°, o/n). The cooled (r.t.) reaction mixture was treated with MeOH and then made basic (to pH>10) by the addition of Na2CO3 (10% w/w). The mixture was filtered and the filtrate evaporated and co-evaporated (H2O). Where deacylation of the sulfated product was required, the crude product was taken up in water and IM NaOH was added (2 eq per acyl group). When deprotection was complete the product was carried through to the next stage. The crude sulfated material in H2O was subjected to size exclusion chromatography. The pure fractions were evaporated and co-evaporated (H2O) and then lyophilised (H2O) to yield the sulfated product. When required, after lyophilisation the product was passed through an ion- exchange resin column (AG®-50W-X8, Na+ form, 1x4 cm, deionized H O, 15 mL) in order to transfer the product uniformly into the sodium salt form. The solution collected was evaporated and lyophilised to give the final product as a colourless glass or white power. Size exclusion chromatography Size exclusion chromatography (SEC) was performed over Bio-Gel P-2 in a 5 x 100 cm column with a flow rate of 2.8 mL/min of 0.1 M NH4HCO3, collecting 2.8 min (7.8 mL) fractions. Fractions were analysed for carbohydrate content by TLC (charring) and/or for poly- charged species by the dimethyl methylene blue test, and then for purity by capillary electrophoresis (CE) and those deemed to be free of salt were pooled and lyophilised. In the cases of the presence of undersulfated by-products or other salt contaminants (normally only small amounts, but often detected), an LH20 SEC step (2 x 95 cm, deionized water, 1.2 mL/min, 3.5 min per vial) was applied to remove them completely. Dimethyl methylene blue Test Dimethyl methylene blue (DMB) reagent was prepared by dissolving 16 mg of DMB in 1 L of deionized water containing 3.04 g of glycine, 2.37 g of NaCI. 0.1 M HC1 (95 ml) was added to adjust the pH to 3.0. The stock solution was stored in a brown coloured bottle at r.t. (the solution was stable for at least 3 months under such conditions). A 96-well microtitre plate was loaded with 10 μL of fraction solution per well. 55 μL of DMB stock solution was added into each used well. An instant colour change from blue to pink indicated the presence of polycharged species, i.e., sulfated product fractions. General procedure for NIS glycosylations Glycosyl acceptor (1 eq), thioglycoside donor (1.1 eq), 500 mg of freshly activated powdered 3 A molecular sieves and 10 mL of dry DCM were stirred at -20° for 20 min before 1.3 eq of NIS and 1 drop of TfOH were added. Stirring was continued at -20° until the reaction was complete by TLC (~1 h) before 400 μL of Et3N was added. Evaporation (in vacuo) onto silica gel and flash chromatography yielded the glycosylated product. General procedure for Ugi four-component reaction Solutions ofthe acid (1 eq), amine (1 eq), carbonyl compound (1 eq) and isocyanide (1 eq) in MeOH, MeOH-THF (varied ratios) or CHC13 were transferred into a reaction vial (final concentration: 0.1-0.5 M). When D-glucuronic acid was the acid component, it was added as a solid. In the case of bis-acid, bis-amine, bis-aldehyde or bis-isocyanide, the amount was 0.5 eq. The mixture was stirred or shaken at r.t. or 60 °C for 1 h to 5 days. The progress ofthe reaction was monitored by TLC. The mixture was evaporated and the residue was purified by flash chromatography or dried completely under high vacuum followed by direct peracetylation and purification by flash chromatography. General procedure for acetylation of hydroxyl groups: The corresponding alcohol was dissolved in DCM-pyridine (15:1 v/v, 0.15 M) containing DMAP (0.42 mol%). Acetic anhydride (2 eq per hydroxyl) was added and the mixture was stirred at r.t. o/n. The mixture was poured into ice-chilled 0.5 M HC1 and extracted with CHC13. The organic phase was separated and washed with cold 0.5 M HC1 (x2), brine, and dried (MgSO4). The solution was filtered and evaporated. The residue was purified by flash chromatography (gradient elution with hexane-EtOAc) to give pure product. General procedure for Zemplen deacetylation/debenzoylation: A solution of the acetate/benzoate in anhydrous MeOH (0.1 M) was treated with a solution of sodium methoxide in MeOH (1.35 M, 0.2-0.6 eq). The mixture was stirred at r.t. for 1-3 h (monitored by TLC). Acidic resin AG®-50W-X8 (H1" form) was added to adjust the pH to 6-7, the mixture was filtered and the resin was rinsed with MeOH. The combined filtrate and washings were evaporated in vacuo and thoroughly dried to give the poly-ol product. General procedure for deprotection of benzyl ethers via hydrogenolysis To a solution of the benzyl ether-protected compound (0.03 mmol) in MeOH or EtOH (2 mL) was added 5% Pd/C or 20% Pd(OH)2 on charcoal (30 mg or excess). The mixture was loaded in a miniclave (Bϋchi AG, Uster/Switzerland) and stirred under hydrogen atmosphere (50 psi) for 2-10 h. Alternatively, the mixture was bubbled with hydrogen gas for 1 h then stirred at r.t. under 1 atmosphere of hydrogen for 1-5 days. The reaction was monitored by TLC (EtOAc or MeCN-water 10:1). The mixture was filtered and rinsed with MeOH, or EtOH. The filtrate was evaporated and dried under high vacuum, checked by 1H NMR, freeze- dried and used directly for sulfonation. Methylation of hydroxyl groups The dried poly-ol was dissolved in anhydrous DMF (0.04 M) under argon and stirred with NaH (60% suspension in mineral oil, 1.2 eq per hydroxyl) at r.t. for 1 h. Iodomethane (1.2 eq per hydroxyl) was added and stirring continued o/n. MeOH was added and the mixture was evaporated onto silica and purified by flash chromatography. General procedure for Huisgen cycloaddition reactions. The sulfated sugar azide was dissolved in water (0.75 M) and a solution of acetylene in t-butanol (0.9 M, 1 eq) was added. To this mixture was added a solution of copper (II) sulfate (0.3 M in water, 5 mol%) and a solution of sodium ascorbate (1 M, in water, 20 mol%). The mixture was shaken on a minishaker at r.t. o/n, and purified by column chromatography (silica 1x18 cm, gradient elution with EtOAc-MeOH-H2O 50:2:1, 20:2:1 to 10:2:1) to give the corresponding triazole product. Example 1: PG2038
Step a: Methyl 3,4,6-tri- -acetyl-2-0-benzyl- -O-galactopyranosyl-(l-→4)-2,3,6- tri-O-benzyl-β-O-glucopyranoside. Methyl 2,3,6-tri-O-benzyl- -O-glucopyranoside [17] (150 mg, 322 μmol), methyl 3,4,6- tri-O-acetyl-2-O-benzyl-l-thio-fi-O-galactopyranoside [18] (151 mg, 354 μmol) and 200 mg of 3A molecular sieves were subjected to the general NIS glycosylation procedure using 95 mg (422 μmol) of NIS. Flash chromatography (gradient elution 20:80 to 25:75 EtOAc:hexanes) yielded 274 mg of partially deacetylated material. To this mixture was added 10 mL of DCM, 200 μL of acetic anhydride, 200 of μL Et3N and 2 mg of DMAP, and the solution was stirred for 1 h before evaporation and flash chromatography (gradient elution 25:75 to 30:70 EtOAc:hexanes) to give 180 mg (66%) of the title compound as a colourless glass. 1H n.m.r. (400 MHz, CDC13) δ: 7.05-7.35 (m, 20H, 4χPh), 5.74 (d, 1H, J1;2= 4.0, HI11), 5.29 (dd, 1H, J3,4 **** 3.2, J4,5 = 1.2, H411), 5.23 (dd, 1H, J2,3 = 10.8, H3π), 4.92 (d, 1H, Jgem= 12.0, PhCH2), 4.85 (d, 1H, Jgem -=- 10.8, PhCH2), 4.55-4.69 (m, 4H, PhCH2), 4.42 (AB, 1H, Jgem = 12.0, PhCH2), 4.40 (AB, 1H, PhCH2), 4.32 (d, 1H, J1;2 = 7.6, HI1), 4.10 (dt, 1H, J5,6 = 6.8, H5π), 3.88-3.96 (m, H, HS^Hδ1), 3.82 (dd, 1H, Jgem = 11.1, H6π), 3.70-3.76 (m. 4H, H2χH6I+H2II+H3I), 3.56 (s, 3H, OMe), 3.5-3.6 (m, 1H, H41), 3.45 (dd, 1H, J2,3 = 9.0, H21), 2.02 (s, 3H, Ac), 1.93 (s, 3H, Ac), 1.88 (s, 3H, Ac). 13C n.m.r. (100 MHz, CDC13) δ: 170.17, 170.06, 169.87, 138.82, 138.20, 138.13, 137.68, 128.33, 128.25, 128.19, 128.04, 127.61, 127.57, 127.49, 127.46, 127.01, 126.39, 104.42, 97.12, 84.49, 82.26, 74.46, 74.27, 73.69, 73.31, 73.28, 73.02, 69.47, 69.17, 68.35, 66.60, 61.62, 56.96, 20.69, 20.63, 20.58. Step b: Methyl 3,4,6-tri-0-acetyl-a-O-galactopyranosyl-(l→4)-fi-O-glucopyranoside. Pearlman's catalyst (20 mg) and 20 μL of acetic acid were added to a solution of 90 mg (106 μmol) of methyl 3,4,6-tri-0-acetyl-2-0-benzyl-a-O-galactopyranosyl-(l→4)-2,3,6- tri-O-benzyl-β-O-glucopyranoside in 10 mL of MeOH. An atmosphere of hydrogen was applied with 3 vacuum purges and the suspension was stirred for 3 days. After filtration, evaporation and co-evaporation with PhMe the residue was subjected to flash chromatography (gradient elution 100:0 to 100:3 EtOAc:MeH) to yield 47 mg (91%) ofthe title compound. 1H n.m.r. (400 MHz, CD3OD) δ: 5.39 (br d, 1H, J3,4 = 3.0, H4π), 5.32 (d, 1H, J1;2= 3.8, HI11), 5.10 (dd, 1H5 J2!3 = 10.6, H3π), 4.38 (br t, 1H, J5,6 = 6.8, H5π), 4.20 (d, 1H, J1)2 *= 7.8, HI1), 4.09 (app d (ABX), 2H, J5,6 = 6.5,Η6Π), 4.00 (dd, 1H, H2π), 3.92 (dd, 1H, J5;6A= 1.7, Jgem= 12.2, H6A1), 3.80 (dd, 1H, J5j6B= 4.8, H6B1), 3.62 (dis t, 1H, J2,3~3;4= 9.1, H31), 3.56 (part obs t, 1H, J3j4~4j5 = 9.3, H41), 3.53 (s, 3H, OMe), 3.42 (ddd, IH, J4j5 = 9.4, H51), 3.22 (dd, IH, J2;3 = 9.1, H21), 2.10 (s, 3H, AcO), 2.05 (s, 3H, AcO), 2.00 (s, 3H, AcO).
Step c: Methyl 2-0-sulfo- -O-galactopyranosyl-(l→4)-2,3,6-tri-0-sulfo~ -O-glucopyranoside, tetrasodium salt (PG2038) The above disaccharide (32.2 mg, 0.667 mmol), was subjected to the standard sulfonation and deacetylation procedures to give the title compound as a white foam (4.0 mg, 7.8%, 96% purity, CE: 7.18 min). 1H NMR (D2O, 400 MHz): 5.473 (d, IH, Jπi-aiX 3.6, HI11), 4.833 (d, IH, Jπ-2i = 2.8, HI1), 4.60 (overlapped with water, IH, H31), 4.551 (m, IH, H21), 4.306 (dd, IH, π-3π = 10.2, H2π), 4.17-4.06 (m, 4H, H41, H51 and H61), 3.902 (d, IH, J3iι-4iι =* 3.6, H4π), 3.867 (dd, IH, H3π), 3.616 (dd, IH, J6axii-6eqii = 12.0, J5ιι-6axπ = 7.2, Hoax11), 3.564 (dd, IH, J5ii-6eqii= 5.2, H6eqπ), 3.363 (dd, IH, H5π), 3.343 (s, 3H, CH3O). Example 2: PG2046 and PG2047 Step a: 2-Azido-3, 4, 6-tri-0-benzoyl-2-deoxy-a-D-glucopyranosyl-(l→4)-l, 6-anhydro-2-azido- 2-deoxy-3-0-benzyl- -D-glucopyranose A solution of 3,4,6-tri-0-acetyl-2-azido-2-deoxy-D-glucopyranosyl trichloro- acetimidate [19] (201 mg, 0.453 mmol) and l,6-anhydro-2-azido-3-0-benzyl-2-deoxy-fi-D- glucopyranose [20] (84 mg, 0.302 mmol) in 1,2-DCE (5 mL) was stirred in the presence of activated mol. sieves (300 mg of 3A powder) under an atmosphere of argon (r.t., 30 min). The mixture was cooled (-20°) with continued stirring (10 min) and TBDMSOTf (21 μL, 0.091 mmol) was introduced drop-wise and stirring maintained (-20°, 10 min). Et3N (100 μL) was introduced and the mixture filtered and evaporated. The residue was subjected to aqueous workup (EtOAc) and flash chromatography (10-40% EtOAc/hexanes) to yield a pale yellow coloured oil (130 mg). This residue was co-evaporated (2 x 10 mL MeCN) then subjected to the Zemplen deacetylation general procedure. The product was subjected to aqueous workup (EtOAc) to yield a colourless oil (98 mg). This residue was co-evaporated (2 x 10 mL MeCN). BzCl (210 μL, 1.81 mmol) was added to a solution of the crude product (0.302 mmol, max.) and pyridine (2 mL) in 1,2-DCE (3 mL) and the combined mixture stirred (r.t., o/n). The mixture was cooled (0°) and MeOH (2 mL) was introduced with continued stirring (0°→r.t., 2 min) before evaporation and co-evaporation (toluene) of the solvent. The residue was subjected to aqueous workup (EtOAc) and flash chromatography (10-30% EtOAc/hexanes) to yield two compounds. Firstly, the title compound as a colourless foam (101 mg, 46%, 3 steps). 1H NMR (400 MHz, CDC13) 3.l l (s, 1 H; H-21), 3.41 (dd, 1 H, J1;2 3.7, J2,3 10.7 Hz; H-61), 3.61 (s, 1 H; H- 31), 3.39 (s, 1 H; H-41), 4.05 (d, 1 H, J6>6 7.3 Hz; H-61), 4.41-4.49 (m, 2 H; H-611), 4.55, 4.68 (AB quartet, JA,B 11.9 Hz; CH2Ph), 4.79 (ddd, 1 H, J4;5 10.3, J5;6 2.9, 5.9 Hz; H-5π), 4.91 (br d, 1 H, J5,6 5.5 Hz; H-51), 5.08 (d, 1 H, Jlj2 3.6 Hz; H-lπ), 5.51 (dd, 1 H, J3>4 9.5, J4;5 10.2 Hz, H- 4π), 5.60 (s, 1 H,; H-l1), 6.10 (dd, 1 H, J2j3 10.7, J3,4 9.3 Hz; H-3π), 7.29-7.55, 7.89-8.03 (2 m, 20 H; ArH). 13C NMR (100 MHz, CDC13) δ 58.74, 61.47, 63.30, 64.84, 69.19, 69.49, 70.52, 73.17, 74.62, 78.12, 79.57 (11 C; C-2l-6l, C2π-6π, CH2Ph), 100.71, 101.16 (2 C; C-l1, C-lπ), 128.10, 128.42, 128.57, 128.61, 128.64, 128.81, 128.88, 129.15, 129.86, 129.90, 130.06, 130.17, 133.43, 133.53, 133.74, 137.48 (Ar), 165.61, 165.62, 166.26 (3 C; C=O). Next, 2-azido-3, 4, 6-tri-0-benzoyl~2-deoxy-β-O-glucopyranosyl-(l→4)-l, 6-anhydro-2- azido-2-deoxy-3-0-benzyl-fi-O-glucopyranose as a colourless oil (27 mg, 12%, 3 steps). 1H NMR (400 MHz, CDC13) 3.19 (s, 1 H; H-21), 3.74 (dd, 1 H, J5;6 6.2, J6;6 7.1 Hz; H-61), 3.79-3.88 (m, 2 H; H-2π, H-31), 3.88 (ddd, J4;5 9.2, J5;6 3.1, 4.7 Hz; H-5π), 3.95 (br s, 1 H; H- 41), 4.10 (d, 1 H, J6;6 7.3 Hz; H-61), 4.34 (dd, 1 H; J5,6 4.9, J6,6 12.2 Hz; H-6π), 4.50 (dd, 1 H, J5,6 3.1, J6,6 12.3 Hz, H-6π), 4.52, 4.59 (AB quartet, JA,B 12.0 Hz; CH2Ph), 4.65 (d, 1 H, Jι,2 7.9 Hz; H-lπ), 4.69 (br d, 1 H, J5,6 5.5 Hz; H-51), 5.44 (t, 1 H, J2,3=3,4 9.7 Hz; H-3π), 5.49 (br s, 1 H; H-l1), 5.51 (t, 1 H, J3A^ι5 9.6 Hz; H-4π), 7.23-7.50, 7.84-7.96 (2 m, 20 H; ArH). Step b: 2-Deoxy-2-sulfamido-a-O-glucopyranosyl-(l→4)-l, 6-anhydro-2-deoxy-2-sulfamido-3- 0-benzyl- -O-glucopyranose, disodium salt (PG2046) A mixture of 2-azido-3,4,6-tri-0-benzoyl-2-deoxy-a-O-glucopyranosyl-(l→4)-l,6- anhydro-2-azido-2-deoxy-3-0-benzyl- -D-glucopyranoside (127 μmol), Pearlman's catalyst (11 mg), and ammonium formate (300 mg) in 2:1 MeOH:EtOAc (7 mL) was heated to 65° under argon until complete by TLC. The mixture was cooled to r.t., filtered (0.2 μm) and evaporated. The crude amine was purified by SPE (300 mg C18 Waters cartridge, equilibrated with 5:95 MeOH:H2O, gradient eluted 5:95 to 100:0 MeOH:H2O) to yield 53 mg of the diamine (58%). Without further purification, to the diamine was added DMF (5 mL), SO3 «Me3N (41 mg, 295 μmol) and NaHCO3 (40 mg, 475 μmol). The mixture was heated to 60° for 1 h then cooled to rt and quenched with ice and Na2CO3 (sat. aqueous). This suspension was stored at -18° o/n and the sample was filtered. The filtrate was evaporated. Water (1 mL) and NaOH (250 μL, IM) were added and the solution was stirred overnight then loaded directly onto the SEC column (general procedures) to yield 22 mg (28 % over three steps) of the title compound. 1H NMR (400 MHz, D2O, solvent suppressed) δ: 7.35-7.21 (m, 5H, ArH), 5.43 (br s, IH, HI1), 5.18 (d, IH, J1-2 *= 3.6, HI11), 4.72-4.691 (m, IH, H51), 4.54-4.521 (m, 2H, ArCH2), 4.05 (d, IH, Jgem = 7.9, H6A1), 3.85 (br s, IH, H31), 3.76-3.58 (m, 5H), 3.51 (dd, IH, J2-3 = 10.4, J3-4 = 9.1, H3π), 3.34 (t, IH, J3-4~4-5 = 9.2, H4π), 3.23 (br s, IH, H21), 3.12 (dd, IH, H2π). 13C NMR (100 MHz, CDC13) δ: 133.3, 124.8, 124.6, 124.4, 96.9, 95.1, 72.7, 71.5, 70.8, 68.3, 68.2, 67.2, 66.0, 61.0, 56.6, 54.0, 49.8.
Step c: 2-Deoxy-2-sulfamido-a-O-glucopyranosyl-(l→4)-l, 6-anhydro-2-deoxy-2-sulfamido- - O-glucopyranoside, disodium salt (PG2047) A mixture of 2-deoxy-2-sulfamido-a-O-glucopyranosyl-(l→4)-l, 6-anhydro-2-deoxy-2- sulfamido-3-O-benzyl-β-O-glucopyranoside, disodium salt (12.9 mg, 20.8 μmol) and Pearlman's catalyst (5 mg) in purified water (2 mL) was subjected to 50 psi H2 overnight. The mixture was filtered and lyophilised to yield 10.7 mg (98 %) of the title compound. 1H NMR (400 MHz, D2O) δ: 5.47 (br s, IH, HI1), 5.20 (d, IH, J1-2 = 3.5, HI11), 4.68 (br d, IH, J5-4 = 5.5, H5), 4.07 (d, IH, Jgem = 1.6, H6A1), 3.98 (br s, IH, H31), 3.75-3.64 (m, 4H), 3.52 (t, IH, J2.3.-3-4 = 9.3, H3π), 3.34 (t, IH, J3-4^.5 = 9.3, H4π), 3.13 (obs. dd2, IH, H2π), 3.11 (br s, IH, H21). Example 3: PG2039 and PG2037 Step a: Methyl 3,4-di-0-acetyl-2,6-di-0-benzyl-a-O-galactopyranosyl-(l→4)-2,3,6- tri-0-benzyl- -O-glucopyranoside Methyl 2,3,6-tri-O-benzyl-β-O-glucopyranoside (287 mg; 618 μmol), 302 mg (618 μmol) of ethyl 3,4-di-0-acetyl-2,6-0-dibenzyl-l-thio-fi-D-galactopyranoside [21] and 700 mg of 3 A molecular sieves were subjected to the general NIS glycosylation procedure using 181 mg (803 μmol, 1.3eq) of NIS. Flash chromatography (2.5 x 20 cm, gradient elution 1:5 to 1:3 EtOAc:Hexanes) yielded the title compound as a colourless gum (176 mg, 32%). 1H NMR (400 MHz, CDC13, 400 MHz): 7.40-7.12 (m, 25H, Ph), 5.818 (d, IH, Jm._ιι = 3.6, HI11), 5.481 (d, IH, J41I-3iι = 3.2, H4π), 5.309 (dd, IH, J3ii-2n = 10.8, J31I-4π = 3.2, H3π), 4.980 (d, IH, Jgem = 11.6, a-PhCH2), 4.904 (d, IH, Jgem = 11.2, b-PhCH2), 4.748 (d, IH, Jgem = 11.6, a-PhCH2), 4.67-4.57 (m, 3H, b-PhCH2 and c-PhCH2), 4.479 (d, IH, Jgem = 12.8, d-PhCH2), 4.443 (d, IH, Jgem *= 12.8, d-PhCH2), 4.415 (d, IH, Jgem = 11.6, e-PhCH2), 4.360 (d, IH, Jιua = 8.0, HI1), 4.201 (d, IH, Jgem = 12.6, e-PhCH2), 4.153 (t, IH, J5II-6aχiι = 7.2, J5ii-6eqii -= 6.0, H511), 4.072 (t, IH, J4I-3i = 9.0, J41-5i = 9.0, H41), 3.858 (dd, IH, J2π-3π = 10.8, J2π-1π = 3.6, H2π), 3.82-3.76 (m, 3H, H31, Hoax1 and H6 eq1), 3.597 (s, 3H, OMe), 3.62-3.57 (m, IH, H51), 3.507 (t, IH, J2I.3I =
Affected by the solvent suppression signal. Partially obscured by H21. 8.4, J2HI = 8.0, H21), 3.347 (dd, IH, J6eqii-6axii = 9.2, J6eqπ-5iι = 6.0, H6eqπ), 3.291 (dd, IH, J6axii-6ec.ii *--- 9.2, J6axII-5„ = 7.2, Hoax11), 1.958 (s, 3H, OAc), 1.930 (s, 3H, OAc). 13C NMR (100 MHz, CDCI3, 100 MHz): 169.94 (CO), 169.78 (CO), 138.88, 138.35, 138.24, 137.73 and 137.64 (5x ipso-V ), 128.25, 128.22, 128.21, 128.15, 128.12, 128.00, 127.80, 127.57, 127.52, 127.46, 127.41, 127.37, 126.93, 126.37, 104.40, 97.16, 84.63, 82.31, 74.42, 74.30, 73.70, 73.25, 73.16, 73.14, 72.98, 69.73, 69.07, 68.89, 67.64, 67.50, 56.86, 20.71, 20.55. Step b: Methyl 3,4-di-0-acetyl-a-O-galactopyranosyl-(l→4)- -D-glucopyranoside. Following the standard debenzylation procedure, methyl 3,4-di-0-acetyl-2,6-di- 0-benzyl-a-O-galactopyranosyl-(l→4)-2,3,6-tri-0-benzyl-fi-O-glucopyranoside (88 mg, 98.8 μmol) was deprotected to give the title compound as a colourless powder (42 mg, 97%). 1H NMR (D2O, 400 MHz): 5.394 (d, IH, Jin-2n= 3.6, HI11), 5.294 (d, IH, J4π-3πX 3.2, H4π), 4.953 (dd, IH, J3II.2II *--- 10.4, J3II-4H- 3.2, H3n), 4.229 (d, IH, Jn-2I-= 8.4, HI1), 4.080 (t, IH, J5π-6axiι = 6.4, J5n-6eqii *= 6.0, H5π), 3.965 (dd, IH, J2n-3ii= 10.4, J__.m = 3.6, H2π), 3.803 (dd, IH, J6eqι-6eqι = 12.0, J6eqι-5ι= 1.6, Hόeq1), 3.67-3.59 (m, 2H, Hδax1 and H31), 3.54-3.40 (m, 4H, H41, H51 and H6π), 3.407 (s, 3H, OMe), 3.134 (dd, IH, ι-31 = 9.2, J_IΛ\ **** 8.4, H21), 2.012 (s, 3H, OAc), 1.909 (s, 3H, OAc). 13C NMR (D2O, 100 MHz): 173.57, 173.47, 103.20, 99.71, 77.12, 76.30, 74.57, 73.09, 70.89, 69.94, 69.04, 66.45, 60.82, 60.18, 57.31, 20.34, 20.09. Step c: Methyl 2,6-di-0-sulfo-a-O-galactopyranosyl-(l→4)-2,3,6-tri-0-sulfo- -O- glucopyranoside, pentasodium salt (PG2039) Following the standard sulfonation/deacetylation procedures, 42 mg (95.4 μmol) of methyl 3,4-di-0-acetyl-a-O-galactopyranosyl-(l→4)- -O-glucopyranoside was converted to the title compound as a white powder (14.8 mg, 18%, CE: 6.12 min). 1H NMR (D2O, 400 MHz): 5.404 (d, IH, Jin-2n = 3.6, HI11), 4.756 (d, IH, Jn-2ι= 3.6, HI1), 4.60 (overlappped with water, IH, H31), 4.448 (dd, IH, J2ι-3ι = 3.2, H21), 4.296 (dd, IH, n-311 = 10.0, H2π), 4.23-4.00 (m, 7H, H61, H51, H6π, H41 and H5π), 3.958 (dd, IH, J3ii-4n = 3.6, J4n-5*= 0.8, H4π), 3.930 (dd, lH, H3π), 3.367 (s, 3H, CH3O).
Step d: Methyl 2,6-di-0-benzyl-3,4-di-0-methyl~ -O-galactopyranosyl-(l→4)-2,3,6-tri-0- benzyl- -O-glucopyranoside Following the standard deacetylation and methylation procedures, methyl 3,4-di-O- acetyl-2, 6-di-0-benzyl-a-O-galactopyranosyl-(l→4)-2, 3, 6-tri-O-benzyl-β-O-glucopyranoside (72 mg, 80.8 μmol) was converted into the title compound as colourless gum (62.7 mg, 93%). 1H NMR (CDCI3, 400 MHz): 7.35-7.08 (m, 25H, Ph), 5.717 (d, IH, Jιπ-2π = 3.6, HI11), 4.856 (d, IH, Jgem *= 11.2, a-PhCH2), 4.843 (d, IH, Jgem *= 10.8, b-PhCH2), 4.695 (d, 2H, Jgem = 12.0, a-PhCH2 and c-PhCH2), 4.631 (d, IH, Jgem = 12.4, d-PhCH2), 4.571 (d, IH, Jgem = 10.8, b-PhCH2), 4.500 (d, IH, J^rrX 12.4, d-PhCH2), 4.450 (d, IH, Jgem = 11.6, c-PhCH2), 4.433 (d, IH, Jgem = 11.2, e-PhCH2), 4.359 (d, IH, Jeaa = 11.2, e-PhCH2), 4.303 (d, IH, J1I-2ι= 7.6, HI1), 3.949 (t, IH, J4I-3I = 9.0, J41-51 = 9.0, H41), 3.871 (dd, IH, J5π-6axII = 7.2, J5Ii-6eqii = 6.4, H5π), 3.791 (dd, IH, J__-m = 10.4, J2π-m = 3.6, H211), 3.77-3.68 (m, 4H, H4π, H31, Hδax1 and Hόeq1)), 3.59-3.53 (m, 2H, H51 and H6π), 3.551 (s, 3H, OMe), 3.51-3.40 (m, 3H, H3π, H6π and H21), 3.492 (s, 3H, OMe), 3.433 (s, 3H, OMe). Step e: Methyl 3,4-di-0-methyl-a-O-galactopyranosyl-(l→4)-fi-O-glucopyranoside Following the standard debenzylation procedure, methyl 2,6-di-0-benzyl-3,4-di-0- methyl-a-O-galactopyranosyl-(l→4)-2,3,6-tri-0-benzyl- -O-glucopyranoside (62.1 mg, 75.1 μmol) was deprotected to give the title compound as colourless gum (28 mg, 97%). 1H NMR (D20, 400 MHz): 5.232 (d, IH, Jm-w = 4.4, HI11), 4.217 (d, IH, J1I-2ι- 8.0, HI1), 3.83-3.75 (m, 3H, H4π, H5π and H61), 3.682 (dd, IH, J2n-3πX 10.4, J2Iι-ιπ = 4.4, H2π), 3.64-3.52 (m, 4H, H61, H3π and H6π), 3.47-3.38 (m, 3H, H41, H51 and H3π), 3.400 (s, 3H, OMe), 3.340 (s, 6H, 2xOMe), 3.117 (dd, IH, J21-31 = 9.6, J_ι-u = 8.0, H21). 13C NMR (D2O, 100 MHz): 103.20, 99.64, 79.35, 76.92, 76.34, 75.35, 74.65, 73.06, 72.03, 68.00, 61.02, 60.89, 60.86, 57.30, 56.94. Stepf: Methyl 3, 4-di-0-methyl-2, 6-di-0-sulfo-a-O-galactopyranosyl-(l→4)-2, 3, 6-tri-O- sulfo-fi-O-glucopyranoside, pentasodium salt (PG2037) Following the standard sulfonation procedure, methyl 3,4-di-O-methyl-a-O- galactopyranosyl-(l→4)- -O-glucopyranoside (28 mg, 72.8 μmol) gave the title compound (3.2 mg, 4.9%). 1H NMR (400 MHz, D2O): 5.357 (d, IH, Jln-2ii= 3.2, HI11), 4.766 (d, IH, Jπ-2ι = 3.6, HI1), 4.60 (overlappped with water, IH, H31), 4.455 (dd, IH, J2ι-3ι= 2.8, H21), 4.304 (dd, IH, J2-3π = 10.0, H2π), 4.22-3.99 (m, 5H, H51, H61, H41 and H5π), 4.002 (d, 2H, J511-6I1 = 6.8, H6π), 3.886 (d, IH, J3π-4iι= 3.2, H4π), 3.667 (dd, IH, H3π), 3.398 (s, 3H, CH3O), 3.367 (s, 3H, CH3O), 3.356 (s, 3H, CH3O). Example 4: PG2053 and PG2042 Methyl 4-0-Allyl-2,3-di-0-sulfo- -L-rhamnoside, disodium salt (PG2053). The title compound was obtained from methyl 2,3-O-isopropylidene- -L- rhamnopyranoside [22] via the general alkylation (with allyl bromide) and deprotection procedure followed by the general sulfonation procedure, as a colourless powder. CE tm = 10.48 min. 1H NMR (400 MHz, D2O) £ 1.19 (d, 3 H, J5,6 6.4 Hz; H-6), 3.26 (s, 3 H; OMe); 3.29-3.40 (m, 1 H; H-4), 3.59-3.67 (m, 1 H; H-5), 4.00-4.05, 4.18-4.22 (2 m, 2 H; OCH2), 4.41-4.42 (m, 1 H; H-3), 4.63-4.64 (m, 2 H; H-2), 4.83 (s, 1 H; H-l), 5.07-5.21 (m, 2 H; *=CH2), 5.76-5.88 (m, 1 H; =CH). When a reduced quantity (1 eq.) of SO3»trimethylamine was employed, methyl 4-0- allyl-2-O-sulfo-a- -rhamnoside, sodium salt (PG2042) was exclusively obtained. CE tm > 25.00 min. 1H NMR (400 MHz, D2O) £1.19 (d, 3 H, J5,6 6.4 Hz; H-6), 3.16 (t, 1 H, J3,4 3.1, J4;5 9.7 Hz; H-4), 3.25 (s, 3 H; OMe), 3.54-3.58 (m, 1 H; H-5), 3.76 (dd, 1 H, J2;3 9.7 Hz; H-3), 4.03-4.18 (m, 2 H; OCH2), 4.34-4.35 (m, 1 H; H-2), 4.80 (s, 1 H; H-l), 5.08-5.22 (m, 2 H; =CH2), 5.78-5.88 (m, 1 H; -CH). Example 5: PG2024 Methyl 4-0-Benzyl-2,3-di-0-sulfo- -L-rhamnoside, disodium salt (PG2024) The title compound was obtained from methyl 2,3-O-isopropylidene- -L- rhamnopyranoside via the general alkylation (with benzyl bromide) and deprotection procedure followed by the general sulfonation procedure, as a colourless powder. CE tm = 10.82 min. 1H NMR (400 MHz, D2O) £1.00 (d, 3 H, J5,6 6.8 Hz; H-6), 3.23 (s, 3 H; OMe); 3.78-3.80 (m, 1 H; H-4), 3.88-3.94 (m, 1 H; H-5), 4.41-4.43 (m, 1 H; H-2), 4.52-4.56 (m, 2 H; H-3), 4.54, 4.78 (AB quartet, JA,B 12.0 Hz; CH2Ph), 4.90 (dd, 1 H, J1;2 1.2 Hz; H-l), 7.20-7.36 (m, 5 H; ArH). Example 6: PG2054 Step a: Methyl 4-O-benzoyl-a- -rhamnoside A solution of methyl 2,3- -isopropylidene- - -rhamnopyranoside (200 mg, 920 μmol), benzoyl chloride (193 mg, 1.38 mmol) and Et3N (364 μL, 2.76 mmol) in DCM (10 mL) was stirred overnight. The resulting suspension (Et3N»HCl precipitates) was diluted with DCM (50 mL) and washed with NaHCO3 (sat. aqueous), water then brine, dried (MgSO4) and evaporated. The residue was taken up in 50 mL of 1:1 MeCN:H2O and p-TsOH (10 mg) was added. The resulting solution was stirred until the reaction was complete (TLC, ~4 h), evaporated and subjected to flash chromatography (1:1 EtOAc:hexanes) to give 165 mg (64 % over two steps) of the title compound as a colourless solid. 1H NMR (400 MHz, CDC13) δ: 8.03-8.00 (m, 2H, Hortho), 7.55 (tt, IH, JHp-Hm = 7.5, JHp-Ho = 1.3, Hpara), 7.43-7.39 (m, 2H, Hmeta), 5.09 (dis t, IH, J4-3~ -5 = 9.3, H4), 4.73 (br s, IH, HI), 4.01-3.97 (m, 2H, H2+H3), 3.91 (dq, IH, J4-5 = 9.7, J5-6 = 6.4, H5), 3.53-3.46 (br s, 2H, OH), 3.38 (s, 3H, OMe), 1.25 (d, 3H, H6). 13C NMR (100 MHz, CDC13) δ: 167.1, 133.3, 129.8, 129.5, 128.3, 100.6, 75.7, 70.8, 70.1, 65.7, 55.0, 17.4. Step b: Methyl 4-0-Benzoyl-2,3-di-0-sulfo-a-L-rhamnoside, Disodium salt (PG2054) The title compound was obtained from methyl 4-O-benzoyl- - -rhamnoside via the general sulfonation procedure as a colourless powder. CE tm = 11.14 min. 1H NMR (400
MHz, D2O) £ 1.14 (d, 3 H, J5,6 6.3 Hz; H-6), 3.33 (s, 3 H; OMe), 3.99-4.07 (m, 1 H; H-5), 4.66-4.73 (m, 2 H; H-2, -3), 4.95 (d, 1 H, J1;2 1.4 Hz; H-l), 5.04 (t, 1 H, J3,4=4>5 9.6 Hz; H-4),
7.35-7.41, 7.53-7.55, 7.92-7.93 (3 m, 5 H; Ph). Example 7: PG2041 Step a: 4,6-0-Benzylidene-l,2-dihydro-O-glucal. A mixture of tri-O-acetyl-O-glucal (1.7 g, 6.25 mmol), AcOH (50 μL) and Pd(OH)2/C (100 mg) in MeOH (15 mL) was vigorously stirred under H2 (1 atm.) overnight. The mixture was filtered, the solvent evaporated and the residue subjected to flash chromatography (10- 50% EtOAc/hexanes) to yield tri-0-acetyl-l,2-dihydro-O-glucal as a colourless oil. This residue was co-evaporated (2 x 10 mL MeCN) then subjected to the Zemplen deacetylation general procedure to yield 1,2-dihydro-D-glucal as a colourless oil (825 mg, 89%). This residue was co-evaporated (2 x 10 mL MeCN). -TsOH.H2O (50 mg) was added to a solution of the 1,2-dihydro-O-glucal (495 mg, 3.34 mmol) and α,α-dimethoxytoluene (753 μL, 5.01 mmol) in DMF (5 mL) and the combined mixture stirred (60°, 1 h). Et3N (100 μL) was introduced and the solvent was evaporated. The residue was subjected to flash chromatography (0-5% MeOH/CHCl3) to yield the title compound as colourless needles (503 mg, 64%). 1H NMR (400 MHz, CDC13) £ 1.72-2.01 (m, 2 H; H-2), 3.27-3.33 (m, 1 H; H-5), 3.41 (dd, 1 H, J3,4 8.8, J5;6 9.1 Hz; H-4), 3.49-3.56 (m, 1 H; H-3), 3.67 (t, 1 H, J5,6=6,6 10.3 Hz; H-6), 3.81-3.87, 3.93-3.98 (2 m, 2 H; H-l), 4.25 (dd, 1 H; J5;6 4.9 Hz; H-6), 5.53 (s, 1 H; CHPh), 7.23-7.52 (m, 5 H, CHPh). 13C NMR (100 MHz, CDCI3) £ 33.47 (C-2); 66.46, 69.07, 69.64, 71.32 (4 C; C- 1,-4,-5,-6), 84.14, (C-3), 102.16 (CHPh), 126.43, 128.55, 129.34, 137.50 (4 C; Ph).
Step b: 3-0-Benzyl-4,6-di-0-sulfo-l,2-dihydro-O-glucal, Disodium salt (PG2041). 4,6-0-Benzylidene-l,2-dihydro-D-glucal was subjected to the alkylation (benzyl bromide), de-protection and sulfonation general procedures to yield the title compound as a colourless powder. CE tm = 15.40 min. 1H NMR (400 MHz, CDC13) £ 1.48-1.53, 1.97-2.03 (2 m, 2 H; H-2), 3.30-3.71 (m, 1 H; H-l), 3.52-3.57 (m, 1 H; H-5), 3.60-3.66 (m, 1 H; H-3), 3.78- 3.83 (m, 1 H; H-l), 3.97 (dd, 1 H, J5;6 8.0, J6,6 11.4 Hz; H-6), 3.98 (t, 1 H, J3,4=4,s 8.9 Hz; H-4), 4.34 (dd, 1 H, J5>6 2.3 Hz; H-6), 4.52-4.67 (m, 2 H; CH2Ph), 7.21-7.36 (m, 5 H; Ph). Example 8: PG2030
Step a: 1, 6-Anhydro-3-0-methyl- -O-glucopyranose. -Toluenesulfonyl chloride (790 mg, 4.14 mmol) was added to a cooled (0°) suspension of 3-O-methyl-D-glucopyranose (804 mg, 4.14 mmol) in pyridine (10 mL) and the reaction mixture stirred (0°→r.t, 1.5 h). Ac2O (1.5 mL, 15 mmol) and N,N-dimethylaminopyrdine (50 mg) were then introduced and stirring continued (r.t., 4 h). The mixture was then cooled (0°) and MeOH (3 mL) was added and stirring maintained (10 min) prior to evaporation ofthe solvent. The residual oil was dissolved (EtOAc) and subjected to workup yielding the tosylate as a pale yellow coloured oil (1.93 g). A mixture of the crude tosylate (1.93 g) and ΝaOH (20 mL of 1.0 M, 20 mmol) in EtOH (20 mL) was heated (80°, 1 h). The mixture was neutralised with acetic acid and the solvent evaporated and co-evaporated (toluene). The crude residue was treated with pyridine (10 mL), Ac2O (5 mL) and NN-dimethylaminopyridine (50 mg) and the combined mixture stirred (r.t., o/n). The mixture was treated with ice- water (10 mL) and stirring continued (r.t., 3 h) before being subjected to workup (EtOAc). The residual oil was subjected to flash chromatography (20-50% EtOAc/hexanes) to yield an inseparable mixture of 2,4-di-0-acetyl-l,6-anhydro-3-0-methyl- -D-glucopyranose (a) and l,2,4-tri-0-acetyl-3-0-methyl-6-0-tosyl- -O-glucopyranose (b) (in a ratio of 3:1) as a pale yellow oil (466 mg). The ratio was determined by integration of the H-l and 3-OMe signals observed in the 1HΝMR spectrum. Partial 1H NMR (400 MHz, CDC13) £ 3.32 (s, 3 H; OMe b), 3.45 (s, 3 H; OMe a); 5.22 (br s, 1 H; H-l b), 5.42 (br s, 1 H; H-l a). The mixture of the two compounds (456 mg) was subjected to the Zemplen deacetylation general method and the residue subjected to flash chromatography (0-5% MeOH/EtOAc) to yield the title compound as a colourless oil (162 mg, 33%, 3 steps). 1HNMR (400 MHz, CDC13): £ 3.27- 3.30 (m, 1 H; H-3), 3.38 (s, 3 H; OMe), 3.57-3.59 (m, 1 H; H-2), 3.63-3.65 (m, 1 H; H-4), 3.70 (dd, 1 H, J5,6 = 5.6, J6,6 = 7.2 Hz; H-6), 4.06 (d, 1 H, J6>6 = 7.2 Hz; H-6), 4.48-4.51 (m, 1 H; H- 5), 4.39 (br s, 1 H; H-l). Step b: 1, 6-Anhydro-4-0-benzyl-3-0-methyl- -O-glucopyranose. A mixture of l,6-anhydro-3-0-methyl-β-O-glucopyranose (155 mg, 0.88 mmol) and Bu2SnO (241 mg, 0.97 mmol) in toluene (18 mL) was heated under reflux (with azeotropic removal of water) until the solution was one-half the original volume. The mixture was cooled (80°), BnBr (104 μL, 0.88 mmol) and BvuNBr (567 mg, 1.76 mmol) were introduced and stirring continued (o/n). The mixture was treated with MeOH (2 mL) and H2O (1 mL) with continued stirring (10 min) prior to evaporation of the solvent. The residue was subjected to workup (EtOAc) and flash chromatography (20-60% EtOAc/ hexanes) to yield two compounds. Firstly, the title compound was produced as a colourless oil (94 mg, 40%). 1H NMR (400 MHz, CDC13): £ 2.58 (d, 1 H, J2;OH 6.4 Hz; OH), 3.32-3.43 (m, 5 H; H-3, H-4, OMe), 3.52-3.57 (m, 1 H; H-2), 3.68-3.72, 4.01-4.04 (2 m, 2 H; H-6), 4.55-4.58 (m, 1 H; H-5), 4.64 (s, 2 H; CH2Ph), 5.39-5.40 (m, 1 Η; Η-l), 7.28-7.36 (m, 5 H; ArH). Secondly, l,6-anhydro-2-0-benzyl-3-0-methyl-$-D-glucopyranose was afforded as a colourless oil (91 mg, 39%). 1H NMR (400 MHz, CDC13): £ 2.90 (br s, 1 H; OH), 3.31-3.34 (m, 4 H; H-2, OMe), 3.36-3.38 (m, 1 H; H-3), 3.58 (br s, 1 H; H-4), 3.68 (dd, 1 H, J5,6 6.0 Hz, J6,6 7.2 Hz; H-6), 4.08 (dd, 1 H, J5,6 0.8 Hz, J6,6 7.2 Hz; H-6), 4.47-4.49 (m, 1 H; H-5), 4.56, 4.62 (AB quartet, JAjB 12.0 Hz; CH2Ph), 5.40-5.41 (m, 1 Η; Η-l), 7.26-7.36 (m, 5 Η; ArΗ). Step c: 1, 6-Anhydro-4-0-benzyl-3-0-methyl-2-0-sulfo-β-D-glucopyranose, sodium salt (PG2030) l,6-Anhydro-4-0-benzyl-3-0-methyl-^>-D-glucopyranose (84 mg, 0.32 mmol) was sulfonated according to the general procedure and subjected to flash chromatography (50/2/l→10/2/l EtOAc/MeOΗ/Η2O) prior to SEC to yield the title compound as a pale yellow coloured powder (70 mg, 60%); CE tm = 5.62 min; 1H NMR (400 MHz, D2O) £3.19 (s, 3 H; OCH3); 3.43-3.45 (m, 1 H; H-4), 3.52-3.53 (m, 1 H; H-3), 3.57 (dd, 1 H, J5;6 = 5.9 Hz, J6,6 = 7.8 Hz; H-6), 3.82 (dd, 1 H, J5,6 = 1.1 Hz, J6,6 = 7.8 Hz; H-6), 3.97-3.99 (m, 1 H; H-5), 4.59-4.61 (m, 3 H; H-2, CH2Ph), 5.41 (br s, 1 Η; Η-l), 7.22-7.34 (m, 5 Η; ArΗ). Example 9: PG2012 and PG2013 Step a: 'N-benzyl-~N-(cyclohexylacetamido)-l, 2, 3, 4-tetra-O-acetyl- -glucuronamide Following the general procedure for the Ugi reaction, D-glucuronic acid (0.950 g, 4.89 mmol), and solutions of each of the following three reagents: benzylamine (2 M in MeOΗ, 2.45 mL, 4.89 mmol), formaldehyde (2 M in MeOΗ, 2.45 mL, 4.89 mmol) and cyclohexylisocyanide (1 M in MeOΗ, 4.89 mL, 4.89 mmol) were loaded into a reaction vessel and the mixture stirred at r.t. for 19 h. The volatiles were removed under reduced pressure and dried under high vacuum to afford ~N-benzyl-~N-(cyclohexylacetamido)-O-glucuronamide as a yellow foam. Following the general procedure for acetylation, the above crude Ugi product was peracetylated to give the title compound as pale-yellow foam 1.929 g, 66% (two steps, Rf = 0.37, hexane-EtOAc 1 :1) after flash chromatography (gradient elution with hexanes-EtOAc 2:1 to 1 : 1). 1H NMR (CDCI3, 400 MHz) was very complicated due to the presence of anomers and rotamers. The spectrum was not simplified after the temperature was raised to 55 °C. However, in pyridine-^ at 100 °C, each set of rotamers was coalesced in some degree into much more simplified structure, thus two anomers were clearly observed (α:β ratio = 69:31). 1H NMR (CDC13, 400 MHz, 25 °C): 7.41-7.14 (m, 5H, Ph), 6.337 (d, 0.39H, J= 3.6), 6.300 (d, 0.29H, J- 3.6), 5.969 (br d, 0.52H, J- 8), 5.823 (br d, 0.09H, J= 8.4), 5.66-5.41 (m, 2.25H), 5.28-5.09 (m, 1.3H), 4.92-4.58 (m, 2.25H), 4.411 (d, J= 10) and 4.395 (d, J= 14, 0.55H), 4.271 (d, 0.1 IH, J= 9.6), 4.219 (d, 0.09H, J= 17.2), 4.125 (d, 0.17H, J= 14), 4.098 (d, 0.17H, J= 14.4), 3.994 (d, J= 15.2) and 3.963 (d, J= 14.8, 0.89H), 3.82-3.59 (m, 2.04H), 2.190, 2.111, 2.038, 2.033, 2.025, 2.023, 2.016, 2.014, 2.008, 1.998, 1.983, 1.944 and 1.927 (all singlet, 12H, Ac), 1.89-1.55 (m, 5H, cyclohexyl-CH2), 1.41-0.83 (m, 5H, cyclohexyl-CH2). 1H NMR (CDCI3, 400 MHz, 55 °C): 7.38-7.16 (m, 5H, Ph), 6.336 (d, J= 3.2, ) and 6.153 (d, J*= 3.2, 0.7H ), 5.939 (br d, 0.6H, J = 6.8), 5.721 (br d, 0.2H, J = 7.2), 5.65-5.55 (m, 1.3H), 5.52-5.41 (m, IH,), 5.28-5.10 (m, 1.4H), 4.85-4.58 (m, 2.4H), 4.48-4.40 (m, 0.6H), 4.318 (d, 0.1H, J = 9.2), 4.205 (d, 0.1H, J= 17.6), 4.02-3.93 (m, 0.9H), 3.84-3.62 (m, 2.2H), 2.179, 2.090, 2.021, 2.012, 2.001, 1.989, 1.983, 1.975, 1.968, 1.959 and 1.935 (all singlet, 12H, Ac), 1.88-1.56 (m, 5H, cyclohexyl-CH2), 1.42-0.88 (m, 5H, cyclohexyl-CH2). 1H NMR (pyridine- d , 400 MHz, δ7.22, 100 °C): only typical sugar protons are given; the remaining signals (except acetate singlets) were complicated and appeared as broad lumps, α-anomer, 6.672 (d, J = 3.6, glu-Hl), 5.456 (dd, J= 9.6, 3.6, glu-H2); β-anomer, 6.206 (d, J= 8.0, glu-Hl), 5.741 (t, J= 9.2, glu-H4 or H5), 5.515 (dd, J**** 8.8, 8.0, glu-H2).
Step b: Y\-benzyl-~ -(cyclohexylacetamido)-l, 2, 3, 4-teti"a-0-sulfo-a~D-glucuronamide, tetrasodium salt (PG2012) and~ -benzyl-~N-(cyclohexylacetamido)-l,2,3-tri-0-sulfo-a-D- glucuronamide, trisodium salt (PG2013) Following the general procedure for deacetylation, the above tetraacete (0.441 g, 0.747 mmol) was deacetylated to give ~N-benzyl-~N-(cyclohexylacetamido)-D-glucuronamide as pale- yellow glass (0.316 g, 100%). Following the general procedure for sulfonation, the above tetrol (0.257 g, 0.608 mmol) was sulfonated (using sulfur trioxide pyridine complex, 60 °C, 19 h). The residue was co- evaporated with toluene and purified by flash chromatography [2.5 x 20cm, eluted with EtOAc, MeCN, MeCN-Et3N (10:1), MeCN-Et3N-H2O (110:2:11)]. The fractions were divided into two parts according to TLC and CE. The less polar part was purified again by flash chromatography, LH20 (x2) and ion exchange chromatography to give trisulfate PG2013 as white fluffy powder after lyophilisation (19.3 mg, 4.4%). 1H NMR (D2O, 400 MHz): two rotamers in a ratio of 56:44. major rotamer, δ 7.36-7.11 (m, 5H, Ph), 5.946 (d, IH, J- 3.2, HI), 4.894 (d, IH, J*= 9.6, H5), 4.748 (d, IH, J= 16, a-CH2), 4.685 (d, IH, J*= 16, a-CH2), 4.502 (t, IH, J= 10.4, 9.6, H3), 4.306 (dd, IH, J= 9.6, 3.6, H2), 4.005 (t, IH, J= 9.6, 8.8, H4), 3.869 (s, 2H, b-CH2), 3.42-3.32 (m, IH, cyclohexyl-CHN), 1.64-1.36 (m, 5H, cyclohexyl-CH2), 1.20-0.92 (m, 5H, cyclohexyl-CH2); minor rotamer, 7.36-7.11 (m, 5H, Ph), 5.905 (d, IH, J= 3.2, HI), 4.578 (d, IH, J= 10, H5), 4.523 (s, 2H, c-CH2), 4.478 (t, IH, J= 10.4, 9.6, H3), 4.321 (d, IH, J= 17.6, d-CH2), 4.281 (dd, IH, J*= 9.6, 3.2, H2), 4.039 (t, IH, J= 9.6, 9.2, H4), 3.900 (d, IH, J = 17.6, d-CH2), 3.42-3.32 (m, IH, cyclohexyl-CHN), 1.64-1.36 (m, 5H, cyclohexyl-CH2), 1.20-0.92 (m, 5H, cyclohexyl-CH2). 13C NMR (D2O, 100 MHz, no reference): double-up of each signals due to two rotamers, 169.96 (amide-CON), 169.75 (amide-CON), 168.91 (amide-CON), 168.85 (amide-CON), 135.45 (ipso-Fh), 135.20 (ipso- Ph), 129.16, 129.07, 128.47, 128.32, 128.20 and 128.13 (meta-, ortho- and para-V ), 95.67 and 95.65 (glu-Cl), 77.84 and 77.75 (glu-C3), 73.76 and 73.71 (glu-C2), 70.66 and 70.18 (glu- C4), 69.23 and 68.70 (glu-C5), 52.87 (a-CH2), 51.12 (c-CH2), 50.32 (d-CH2), 50.02 (b-CH2), 49.25 and 49.00 (cyclohexyl-CHN), 31.89 and 31.86, 25.08 and 25.05, 24.47 and 24.38 (cyclohexyl-CH2). ES-LRMS (+ve, m/z): C21H27N2Na3O16S3 required 728.02, found 751 (M+Na+), 729 (M+H+); ES-HRMS (+ve, m/z): M+Na+, C21H27N2Na4O16S3 required 751.0113, found 750.1087; M+H+, C21H28N2Na3O16S3 required 729.0294, found 729.0242. The polar part was purified by LH20 column (x2) and ion exchange column to give tetrasulfate PG2012 as an off-white powder after lyophilisation (7.6 mg, 1.5%). 1H NMR (D2O, 400 MHz): two rotamers in a molar ratio of 70:30. Major rotamer, δ 7.34-7.16 (m, 5H, Ph), 5.954 (d, IH, J= 3.6, HI), 5.235 (d, IH, J= 9.6, H5), 4.904 (d, IH, J= 15.6, a-CH2), 4.67-4.57 (overlapped with water, IH, H3), 4.536 (t, IH, J= 9.6, 8.8, H4), 4.466 (d, IH, J= 15.6, a-CH2), 4.394 (dd, IH, J= 9.8, 3.4, H2), 3.927 (d, IH, J= 16.8, b-CH2), 3.749 (d, IH, J= 16.8, b-CH2), 3.30-3.20 (m, IH, cyclohexyl-CHN), 1.65-1.35 (m, 5H, cyclohexyl-CH2), 1.18-0.92 (m, 5H, cyclohexyl-CH2); minor rotamer, 7.34-7.16 (m, 5H, Ph), 5.912 (d, IH, J= 3.4, HI), 4.77-4.72 (m, 2H, H5 and H3 or H4), 4.689 (d, IH, J = 15.2, c-CH2), 4.67-4.56 (overlapped with water, IH, H4 or H3), 4.373 (dd, IH, J= 9.8, 3.4, H2), 4.256 (d, IH, J= 18.4, d-CH2), 4.215 (d, IH, J *= 15.2, c-CH2), 3.936 (d, IH, J*= 18.4, d-CH2), 3.36-3.26 (m, IH, cyclohexyl-CHN), 1.65-1.35 (m, 5H, cyclohexyl-CH2), 1.18-0.92 (m, 5H, cyclohexyl-CH2). 13C NMR (D20, 100 MHz, no reference): major rotamer, 172.35 (amide-CON), 171.69 (amide- CON), 137.22 (ipso-? ), 131.71 and 131.58 (meta- and ortho-?h), 131.11 (pαrα-Ph), 97.68 (glu-Cl), 78.08 (glu-C4), 77.60 (glu-C3), 76.43 (glu-C2), 70.10 (glu-C5), 55.81 (a-CH2), 53.17 (b-CH2), 51.77 (cyclohexyl-CHN), 34.17 (cylcohexyl-CH2), 27.56 (cylcohexyl-CH2), 27.13 (cylcohexyl-CH2); minor rotamer (only typical peaks shown), 97.65 (glu-Cl), 78.22 (glu-C4), 77.77 (glu-C3), 76.33 (glu-C2), 71.01 (glu-C5), 53.26 (d-CH2), 53.79 (c-CH2), 52.10 (cyclohexyl-CHN), 34.28 (cylcohexyl-CH2), 27.52 (cylcohexyl-CH2), 27.08 (cylcohexyl-CH2). ES-MS (+ve, m/z): C21H26N2Na4O19S4 required 829.96, found 853 (M+Na+), 831 (M+l ). ES- HRMS (+ve, m/z): M+Na+, C21H26N2Na5O19S4 required 852.9501, found 852.9334; M+H1", C21H27N2Na4O19S4 required 830.9682, found 830.9635. Example 10: PG2064 Step a: 2-(N-acetyl-'N-cyclohexyl)amino-'N-(methyl 2,3,4-tri-0-benzyl-6-deoxy-a-D- mannopyranos-6-yl)acetamide Following the general procedure for the Ugi reaction, a solution of each of the following four reagents: acetic acid (2 M in MeOH, 60 μL, 119 μmol), cyclohexylamine (2 M in MeOH, 60 μL, 119 μmol), formaldehyde (2 M in MeOH, 60 μL, 119 μmol) and methyl 2,3,4-tri-0-benzyl-6-deoxy-6-isocyano- -D-mannopyranoside (0.721 M in CHC13, 150 μL, 108 μmol) was loaded into a 4 mL sample vial and the mixture stirred at 60 °C for 19 h. The volatiles were removed under reduced pressure and purified by flash chromatography (gradient elution with hexane-EtOAc 4: 1 to 1 :4) to afford the title compound as a colourless gum, 42 mg, 60% (Rf = 0.49, EtOAc). 1H NMR (CDC13, 400 MHz): two rotamers in a ratio of 72:28. δ 7.38-7.26 (m, 15H, 3 x C6H5), 6.933 (t, 72% x IH, J = 4.4, NH in major rotamer), 6.357 (t, . 28% x IH, J= 5.8, NH in minor rotamer), 4.91-4.41 (m, 7H, sugar-Hl and 3 x PhCH2), 4.04- 3.42 (m, 9H, sugar-H2-6, NCH2CO and cyclohexyl-CH), 3.305 (s, 72% x 3H, CH3O in major rotamer), 3.266 (s, 28% x 3H, CH3O in minor rotamer), 2.067 (s, 72% x 3H, CH3CO in major rotamer), 2.012 (s, 28% x 3H, CH3CO in major rotamer), 1.85-1.00 (m, 10H, cyclohexyl-CH2). Step b: 2-(N-acetyl-~N-cyclohexyl)amino-'N-( methyl 6-deoxy -2,3,4-tri-O-sulfo-a-D- mannopyranos~6-yl)acetamide, trisodium salt (PG2064) Following the general procedure for deprotection of benzyl ethers, a mixture of the above tribenzyl ether (42 mg, 0.065 mmol), 20% palladium on activated charcoal (22 mg) in MeOH (2 mL) was stirred under hydrogen atmosphere at 50 psi for 10 h. General work-up gave the triol intermediate as a colourless gum. Following the general procedure for sulfonation, the triol was sulfonated (sulfur trioxide trimethylamine complex, 60 °C, 19 h) and the crude was evaporated. The residue was purified via sequential SEC (Bio Gel P-2 followed by LH20). The pure product was converted to the sodium salt by passing through an ion exchange column to give the title compound as a white fluffy powder after lyophilisation (3.1 mg, 7.0%, two steps). 1H NMR (D2O, int. ref. acetone at 2.05, 400 MHz): two rotamers in a ratio of 63:37. δ 4.844 (s, IH, sugar-Hl), 4.678 (s, IH, sugar-H2), 4.51-4.45 (m, IH, sugar-H3), 4.240 (t, 37% x IH, J= 9.6, sugar-H4 in minor rotamer), 4.214 (t, 63% x IH, J= 9.6, sugar-H4 in major rotamer), 3.980 (s, 37% x 2H, COCH2N in minor rotamer), 3.825 (s, 63%) x 2H, COCH2N in major rotamer), 3.78-3.59 (m, 3H, sugar-H5, one sugar-H6 and cyclohexyl-CH), 3.31-3.17 (m, 4H, one sugar-H6 and CH3O [3.253, s, 3H]), 2.060 (s, 67% x 3H, CH3CO in major rotamer), 1.900 (s, 33% x 3H, CH3CO in minor rotamer), 1.70-0.88 (m, 10H, cyclohexyl-CH2). Example 11: PG2068
Step a Following the general procedure for the Ugi reaction, monomethyl succinate (15.7 mg, 0.119 mmol) and a solution of each ofthe following three reagents: ethylamine (2 M in MeOH, 60 μL, 119 μmol), formaldehyde (2 M in MeOH, 60 μL, 119 μmol) and methyl 2,3,4-tri-O- benzyl-6-deoxy-6-isocyano-a-D-mannopyranoside (0.721 M in CHCI3, 150 μL, 108 μmol) was loaded into a 2 mL sample vial and the mixture stirred at r.t. for 19 h. The volatiles were removed under reduced pressure and purified by flash chromatography (gradient elution with hexane-EtOAc 1:1 to 1:4 then EtOAc) to afford pure product as a colourless gum (46.8 mg, 65%). 1H NMR (CDCI3, 400 MHz): two rotamers in a ratio of 73:27. δ 7.38-7.25 (m, 15H, Ph), 6.598 (t, 73% x IH, J= 6, NH), 6.529 (t, 27% x IH, J= 6, NH), 4.93-4.61 (m, 7H, sugar-Hl and 3 x PhCH2), 4.14-3.24 (m, 16H, sugar 6 x H, NCH2CO, 2 x CH3O [singlets at 3.660 and 3.301, 73%; 3.648 and 3.276, 27%] and ethyl-CH2), 2.70-2.42 (m, 4H, COCH2CH2CO), 1.138 (t, 73% x 3H, J= 7, ethyl-CH3), 1.014 (t, 27% x 3H, J= 7, ethyl-CH3). 13C (100 MHz, CDCI3, δ 77.0): major rotamer, 173.32, 171.73, 168.94, 138.20, 138.17, 138.09, 128.22, 128.18, 128.10, 127.76, 127.63, 127.49, 127.46, 98.94, 79.97, 75.38, 75.01, 74.84, 73.01, 72.01, 70.10, 54.63, 51.63, 49.84, 43.59, 39.63, 28.99, 27.24, 13.45. minor rotamer (only non-overlapped peaks), 173.26, 171.54, 168.01, 128.27, 127.69, 127.55, 98.98, 79.85, 75.14, 74.40, 72.86, 71.92, 69.97, 54.74, 50.91, 49.72, 42.10, 28.90, 27.83, 12.29. Step b (PG2068) Following the general procedure for deprotection of benzyl ethers, a mixture of the above tribenzyl ether (46.8 mg, 0.0706 mmol), 20% palladium on activated charcoal (30 mg) in MeOH (3 mL) was stirred under hydrogen atmosphere at 50 psi for 2 h. General work-up gave the triol intermediate as a colourless gum. Following the general procedure for sulfonation, the triol was sulfonated (sulfur trioxide trimethylamine complex, 60 °C, 19 h). The residue was dissolved in IM NaOH (3 mL, 0.16 M). The mixture was stirred at room temperature overnight and concentrated under reduced pressure. The residue was purified via sequential SEC (Bio-Gel P-2 followed by LH20). The pure product was converted into the sodium salt by passing through an ion exchange column to give the PG2068 as a white powder (4.9 mg, 9.8%, two steps). !H NMR (D2O, int. ref. acetone at 2.05, 400 MHz): two rotamers in a ratio of 70:30. δ 4.86-4.84 (m, IH, sugar-Hl), 4.69-4.67 (m, IH, sugar-H2), 4.50-4.46 (m, IH, sugar-H3), 4.28-4.19 (m, IH, sugar-H4), 4.072 (s, 30% x 2H, 2H of COCH2N in minor rotamer), 3.987 (d, 35% x 2H, J= 16.8, IH of COCH2N in major rotamer), 3.815 (d, 35% x 2H, J= 16.8, IH of COCH2N in major rotamer), 3.79-3.69 (m, 2H, sugar-H5 and one sugar- H6), 3.44-3.18 (m, 6H, one sugar-H6, ethyl-CH2 and CH3O [3.245, s, 3H]), 2.633 (t, J= 6.8) and 2.45-2.37 (m, total 4H, COCH2CH2CO2), 1.054 (t, 70% x 3H, J**** 7.2, CH3O), 0.906 (t, 30% χ 3H, J= 7.2, CH3O). Example 12: PG2075
Step a 3-Chlorophenylacetic acid (223 mg, 1.307 mmol) was dissolved in MeCN (3 mL). Ammonia solution (28%, 0.26 mL, 3.8 mmol) was added. The mixture was swirled for a while and evaporated in vacuo. The residue was suspended in MeCN (3 mL), filtered and the white solid was washed with MeCN and freeze-dried to afford ammonium 3-chlorophenylacetate (0.195 g, 80%). Following the general procedure for the Ugi reaction, the above ammonium salt (22.5 mg, 0.120 mmol) and a solution of each following two reagents: formaldehyde (2 M in MeOH, 60 μL, 119 μmol) and 2-isocyanoethyl 2,3,4,6-tetra-O-benzyl-a-D-mannopyranoside (0.762 M in CHCI3, 157 μL, 120 μmol) was loaded into a 2 mL sample vial and the mixture stirred at r.t. for 19 h. The volatiles were removed under reduced pressure and the residue purified by flash chromatography to give the product as a colourless gum (34.8 mg, 37%). 1H NMR (CDCI3, 400 MHz): δ 7.39-7.06 (m, 24H, 4 x C6H5 and 1 x H ), 6.731 (t, IH, J= 6.0, NH), 4.878 (d, IH, J= 10.8, a-CH2), 4.844 (d, IH, J= 2.0, sugar-Hl), 4.770 (d, IH, J= 12.4, b-CH2), 4.723 (d, IH, J= 12.4, b-CH2), 4.640 (s, 2H, c-CH2), 4.589 (d, IH, J= 11.6, d-CH2), 4.535 (d, IH, J= 11.6, d-CH2), 4.505 (d, IH, J= 10.8, a-CH2), 4.446 (d, IH, J= 14.8, e-CH2), 4.362 (d, IH, J= 14.8, e-CH2), 3.90-3.51 (m, 12H), 3.38-3.29 (m, IH). 13C (100 MHz, CDC13, δ 77.0): 169.67, 166.81, 138.27, 138.11, 137.70, 135.16, 134.31, 129.82, 129.40, 128.34, 128.31, 128.01, 127.85, 127.78, 127.70, 127.68, 127.60, 127.53, 127.46, 98.90, 79.96, 75.08, 75.04, 74.71, 73.58, 72.73, 72.19, 72.13, 69.78, 68.49, 63.17, 40.25, 39.27. Step b (PG2075) Following the general procedure for deprotection of benzyl ethers, a mixture of the above tetrabenzyl ether (34.8 mg, 0.0439 mmol), 20% palladium on activated charcoal (26 mg) in MeOH (2 mL) was stirred under hydrogen atmosphere at 50 psi for 2 h. General work-up gave the tetrol intermediate as a colourless gum. Following the general procedure for sulfonation, the above tetrol was sulfonated . The residue was purified via SEC (Bio-Gel P-2). The pure product was converted into the sodium salt by passing through an ion exchange column to give PG2075 as a white fluffy powder after lyophilisation (10.6 mg, 28%, two steps). 1H NMR (D2O, int. ref. acetone at 2.05, 400 MHz): δ 7.30-7.11 (m, 4H, Ar), 5.00-4.97 (m, IH, sugar-Hl), 4.72-4.28 (m, 3H, sugar-H2, H3 and H4), 4.21-3.30 (m, 11H, sugar-H5, H6 and 4 x CH2). Example 13: PG2014 Step a Following the general procedure for the Ugi reaction, 2-(benzyl3,4,6-tri-0-benzyl- -D- mannopyranoside-2-yl) acetic acid (50 mg, 0.0835 mmol) and a solution of each following three reagents: benzylamine (2 M in MeOH, 41.8 μL, 0.0835 mmol), formaldehyde (2 M in MeOH, 41.8 μL, 0.0835 mmol) and 2-isocyanoethyl 2,3,4,6-tetra-O-benzyl- -D- mannopyranoside (0.415 M in MeOH, 201.4 μL, 0.0835 mmol) was loaded into a 2 mL sample vial and the mixture stirred at r.t. for 19 h. General work-up gave the product as a colourless gum (38.9 mg, 36%). 1H NMR (400 MHz): two rotamers around amide CO-NH single bond in a ratio of 66:34. δ 7.40-7.05 (m, 45H, 9 x C6H5), 6.66 (t, 0.66H, J= 5.6, CONH-major rotamer) and 6.40 (t, 0.34H, J = 5.4, CONH-minor rotamer), 5.104 (d, 0.66H, J = 1.2, Hl'-major rotamer) and 5.046 (s, 0.34H, Hl'-minor rotamer), 4.86-4.20 (m, 21H), 3.97-3.48 (m, 16H), 3.36 (q, 0.66H, J= 5.6, major rotamer) and 3.26 (q, 0.34H, J= 5.6, minor rotamer). Step b (PG2014). Following the general procedure for the deprotection of benzyl ethers, a mixture of the above octabenzyl ether (35 mg, 0.0267 mmol) and 20% palladium on activated charcoal (10 mg) in EtOH (4 mL) was stirred under hydrogen atmosphere at 50 psi for 2 h. General work- up gave the octol intermediate as a colourless gum. Following the general procedure for sulfonation, the above octol was sulfonated (sulfur trioxide trimethylamine complex, 60 °C, 19 h). The residue was purified via SEC (Bio-Gel P-2). The pure product was converted into the sodium salt by passing through an ion exchange column to give PG2014 as a white powder (16.6 mg, 44%, two steps). 1H NMR (D2O, 400 MHz, complicated due to two rotamers): δ 7.32-7.14 (m, 5H, Ph), 5.80-5.66 (m, 0.5H), 5.44-5.39 (m, 0.5H), 5.04-4.96 (m, 1.5H), 4.80- 4.20 (m, 9H, overlapped with water), 4.18-3.78 (m, 7.5H), 3.76-3.46 (m, 1.5H), 3.42-2.98 (m, 3.5H). Example 14: PG2016 Step a Following the general procedure for the Ugi reaction, tr rø-l,4-diaminocyclohexane (6.3 mg, 0.055 mmol) and a solution of each following three reagents: 2 -(methyl 2,3,4-tri-O- benzyl -O-mannopyranoside-6-yl)acetic acid (0.91 M in MeOH, 121 μL, 0.11 mmol), formaldehyde (2 M in MeOH, 55 μL, 0.11 mmol) and cyclohexylisocyanide (1 M in MeOH, 110 μL, 0.11 mmol) was loaded into a 2 mL sample vial and the mixture stirred at r.t. for 5 days. The volatiles were removed under reduced pressure and purified by flash chromatography (gradient elution with hexanes-EtOAc 2:1 to 1:4) to give the product as a colourless gum, 33.0 mg, 43% (Rf = 0.24, DCM-MeOH 95:5 or Rf *=* 0.48, MeCN-EtOAc 1:1). 1H NMR (CDC13, 400 MHz, very complicated due to rotamers): δ 7.50-7.20 (m, 30H, Ph), 6.62 (br s, 1.1H), 6.48 (br s, 0.38H), 5.96 (br d, 0.26H, J= 8), 5.79 (br d, 0.26H, J= 10), 4.92-4.85 (m, 2H), 4.78-4.56 (m, 12H), 4.28-4.22 (m, 2.8H), 4.35-4.06 (m, 1.6H), 3.94-3.56 (m, 19.6H), 3.28 (s, 6H, OMe), 1.88-1.42 (m, 16H), 1.36-1.00 (m, 12H). Step b. (PG2016). Following the general procedure for the deprotection of benzyl ethers, a mixture of the above hexabenzyl ether (33 mg, 0.0235 mmol) and 20% palladium on activated charcoal (65 mg) in MeOH (2.8 mL) was stirred under hydrogen atmosphere at 1 atm for 5 days. General work-up gave the hexol intermediate as a colourless gum. Following the general procedure for sulfonation, the above hexol was sulfonated. The residue was purified via sequential column chromatography (SEC on Bio-Gel P-2 followed by ion exchange column) to give PG2016 as a white powder (12.2 mg, 35%). 1H NMR (D2O, 400 MHz): δ 4.97-4.92 (m, 2H, man-Hl), 4.77- 4.75 (m, 2H, man-H2), 4.58-4.52 (m, 2H, man-H3), 4.46-4.08 (m, 6H, containing man-H4 at 4.46-4.36, and OCH2CO), 3.98-3.80 (m, 8H, man-H5, man-H6 and NCH2CO), 3.80-3.32 (m, 6H, containing man-H6 at 3.80-3.64, and cyclohexyl-CH), 3.318 (s, 6H, OMe), 1.82-1.34 (m, 18H, cyclohexyl-CH2), 1.26-1.00 (m, 10H, cyclohexyl-CH2). ES-MS (+ve, m/z): C40H62N4Na6O34S6 required 1472.10, found 1495 (M+Na+), 1473 (M+H+). ES-HRMS (+ve, m/z): M+Na+, C40H62N4Na7O34S6 required 1495.0853, found 1495.0957; M+H , C40H63N4Na6O34S6 required 1473.1034, found 1473.1082. Example 15: PG2015 Step a Following the general procedure for the Ugi reaction, 3,3-dimethylglutaric acid (7.1 mg, 0.0443 mmol) and a solution of each following three reagents: 3-aminopropyl 2,3,4,6- tetra-O-benzyl- -D-mannopyranoside (0.642 M in MeOH, 138 μL, 0.0886 mmol), formaldehyde (2 M in MeOH, 44.3 μL, 0.0886 mmol) and cyclohexylisocyanide (1 M in MeOH, 88.6 μL, 0..0886 mmol) were loaded into a 2 mL sample vial and the mixture stirred at r.t. for 5 days. The volatiles were removed under reduced pressure and purified by flash chromatography (gradient elution with hexanes-EtOAc 2:1 to 1 :4) to give the product as a colourless gum, 32.3 mg, 46% (Rf = 0.45, hexane-EtOAc 1 :3). 1H NMR (CDC13, 400 MHz, very complicated due to rotamers): δ 7.38-7.21 (m, 40H, Ph), 7.003 (d, 0.41H, J= 7.7), 6.930 (br s, 0,21H), 6.726 (d, 0.4H, J= 8.4), 6.597 (d, 0.73H, J= 8.8), 6.487 (br s, 0.25H), 4.85-4.78 (m, 4H), 4.76-4.59 (m, 10H), 4.54-4.45 (m, 4H), 4.00-3.62 (m, 20H), 3.46-3.14 (m, 6H), 2.52-2.22 (m, 4H), 1.90-1.52 (m, 15H), 1.34-1.00 (m, 15H). Step b (PG2015). Following the general procedure for the deprotection of benzyl ethers, a mixture of the above hexabenzyl ether (32.3 mg, 0.0202 mmol), 20% palladium on activated charcoal (41 mg) in MeOH (2.8 mL) was stirred under hydrogen atmosphere at 1 atm for 5 days. General work- up gave the octol intermediate as a colourless gum. Following the general procedure for sulfonation, the above octol was sulfonated. The residue was purified via SEC (Bio-Gel P-2) to give PG2015 as a white powder (12.6 mg, 37%). 1H NMR (D2O, 400 MHz): δ 5.020 (d, 2H, J = 1.6, man-Hl), 4.759 (br s, 2H, man-H2), 4.66-4.56 (m, 2H, man-H3, overlapped with water), 4.46-4.41 ,(m, 2H, man-H6), 4.265 (t, 2H, J= 9.6, 9.2, man-H4), 4.10-3.96 (m, 4H, man-H5 and man-H6), 4.05-3.14 (m, 14H, NCH2CO, cyclohexyl-CH and NCH2CH2CH2O), 2.50-2.11 (m, 4Η, CCH2CO), 1.84-1.42 (m, 14H, cyclohexyl-CH2 and NCH2CH2CH2O), 1.24-0.93 (m, 16H, cyclohexyl-CH2 and Me). ES-MS (+ve, m/z): C41H93NnO37S7 (7 x SO3NH4) required 1556, found 1578 (M+Na"1"), 1556 (M+H+). ES-HRMS (+ve, m/z): M+H+ C41H93NπO37S7 required 1556.3857, found 1556.3783. Example 16: PG2155.
Ethyl 2,6-Di-0-benzyl-3,4-di-0-sulfo-β-D-galactopyranoside, disodium salt (PG2155) The title compound was obtained from ethyl 2,6-di-0-benzyl-3,4-di-0-sulfo-β-D- galactopyranoside [23] via the general sulfonation procedure as a colourless powder. 1H NMR (400 MHz, D2O) £ 1.10 (dd, 3 H; CH2CH3); 2.49-2.66 (m, 2H, CH2CH3); 3.59 (dd, 1 H, Jw 9.7 Hz; H-2); 3.65 (dd, 1 H, J5,6a 3.3, J6a,6b 11.0 Hz; H-6a); 3.68 (dd, IH, J5,6b 4.0 Hz; H-6b); 3.83 (m, 1 H; H-5); 4.36 (dd, 1 H, J3,4 3.0 Hz; H-3); 4.46 (s, 2 H; CH2Ph); 4.48 (d, 1 H; H-l); 4.60, 4.75 (AB quartet, J 10.3 Hz; CH2Ph); 4.85 (dd, 1 H; J4,5 0.0 Hz; H-4); 7.22-7.29, 7.37-7.39 (2 m, 10 H; ArH). Example 17: PG2163.
Step a: Methyl 4-0-allyl-6-azido-6-deoxy-2,3-di-0-isopropylidene- -D-mannopyranoside A solution of methyl 6-azido-6-deoxy-a-D-mannopyranoside (311 mg, 1.419 mmol) in 2,2-dimethoxypropane (4.7 mL, 0.3 M) was treated with (±)-camphor-lO-sulfonic acid (16 mg, 0.0709 mmol, 5 mol%). The mixture was stirred at r.t. for 1 h. TLC indicated the complete conversion to the product (Rf = 0.40, EtOAc-hexane = 17:83). The mixture was basified by addition of sat. Na2CO3 (aq. sol.) and evaporated under vacuum. The residue was extracted with EtOAc (30 mL) and the EtOAc solution washed with brine, dried (MgSO4). Filtration and evaporation gave a gum, which was co-evaporated with toluene once. The final colourless gum was dissolved in anhydrous DMF (3.5 mL, 0.4 M) and stirred with NaH (60% dispersion in mineral oil, 163 mg, 4.257 mmol, 3 eq) for lh. Allyl bromide (360 μL, 4.257 mmol, 3 eq) was added and the mixture stirred at r.t. for another 6 h, treated with methanol (1 mL) and evaporated to dryness. The residue was purified by silica column chromatography (2.5x18 cm, eluted with EtOAc-hexane 1:10 to 1:6) to give the title compound as a colourless gum (0.281 mg, 66% over 2 steps). 1H NMR (CDC13, 400 MHz): 5.85 (m, IH, allyl-2'), 5.23 (ddd, IH, J2.3.trans = 17.2, J3,-gem= 3.6, Jv.? = 1.6, H3'tøris), 5.17-5.13 (m, IH, H3'cis), 4.89 (s, IH, HI), 4.35 (dddd, IH, Jrgem = 12.4, Jv.? = 5.2, J= 1.6, HI'), 4.16 (dd, IH, J2-3 = 5.6, J3-4 = 7.2, H3), 4.09 (d, IH, H2), 4.05 (dddd, IH, HI'), 3.67 (ddd, IH, J4-5 = 10.4, J5-6ax= 6.8, J5-6eq *= 2.4, H5), 3.48 (dd, IH, J6ax-6eq *= 13.2, H6eq), 3.40 (dd, IH, J6aχ-6eq = 13.2, J5-6ax = 6.8, H6ax), 3.33 (dd, IH, H5), 1.50 (s, 3H, Me), 1.30 (s, 3H, Me). 13C NMR (CDC13, 100 MHz): 134.4, 117.1, 109.2, 98.0, 78.2, 76.2, 75.6, 71.5, 68.0, 54.9, 51.6, 27.8, 26.1.
Step b: Methyl 4-0-allyl-6-azido-6-deoxy- -D-mannopyranoside Methyl 4-0-allyl-6-azido-6-deoxy-2, 3-di-O-isopropylidene-a-D-mannopyranoside (56 mg, 0.187 mmol) was dissolved in MeCN-MeOH-H2O (3 mL, 3 mL and 0.2 mL respectively) and treated with p-toluenesulfonic acid monohydrate (7 mg, 0.0374 mmol, 20 mol%). The mixture was stirred at r.t. for 5 h and triethylamine (0.4 mL) added. The mixture was evaporated and the residue purified by column chromatography (silica 1x18 cm, eluted with EtOAc-hexane 1:6 to 2:1) to give the product as a colourless waxy solid (34.8 mg, 72%). 1H NMR (CDC13, 400 MHz): 5.91 (m, IH, allyl-H2'), 5.28 (ddd, IH, J^ s = 16.8, J3.trans.3Ois = 3.0, Jr-3. = 1.6, allyl-H3'trans), 5.20 (ddd, IH, J2.-3.cIs = 10.0, J^ans-sOis = 3.0, Jv.y = 1.6, allyl-ffi'cV), 4.72 (ddd, IH, J1-2 = 2.0, HI), 4.28 (dddd, IH, Jgem = 12.4, J -2. = 5.6, J= 1.6, allyl-1*), 4.13 (dddd, IH, allyl-1'), 3.92 (dd, IH, J2-3 = 3.6, H2), 3.88 (dd, IH, J3-4 = 9.6, H3), 3.70 (ddd, IH, J -5 *= 9.6, J5-6ax= 5.6, J5-6eq= 2.8, H5), 3.54-3.44 (m, 3H, H4, H6ax and H6eq), 3.39 (s, 3H, MeO), 2.62 (br s, 2H, OH).
Step c: Methyl 4-0-allyl-6-azido-2-0-benzyl-6-deoxy-2,3-di-0-sulfonato-a-D- mannopyranoside disodium salt (PG2163) Methyl 4-0-allyl-6-azido-6-deoxy-a-D-mannopyranoside was sulfonated according to the standard procedure to yield the title compound as a white powder, 55 mg (89%). Rf = 0.20 (EtOAc-MeOH-H2O=10:2:l). 1H NMR (D2O, 400 MHz): 5.88-5.76 (m, IH, allyl-2'), 5.20 (d, IH, J2'-3tπ-ns = 17.2, allyl-3 'trans), 5.12 (d, IH, J2.-3'cis = 10.0, allyl-3'cis), 4.91 (s, IH, HI), 4.65 (br s, IH, J2-3 = 3.2, H2), 4.48 (dd, IH, J2 = 3.2, J3-4= 9.2, H3), 4.21 (dd, IH, Jgern= 11.6, Jv.x = 5.6, allyl-1'), 4.00 (dd, IH, Jv._> = 6.4, allyl-1'), 3.71-3.67 (m, IH, H5), 3.58 (dd, IH, J6eq-6aχ = 13.6, J5-6eq = 2.0, H6eq), 3.58 (dd, IH, J4-5 *= 9.6, H4), 3.46 (dd, IH, J6eq-6ax = 13.6, J5.6sκ = 5.6, H6ax), 3.30 (s, 3H, MeO). 13C NMR (D2O, 100 MHz, internal MeOH at 49.05 ppm): 133.9, 119.2, 98.4, 76.1, 75.3, 74.2, 73.1, 70.7, 55.4, 50.8. Example 18: PG2160, PG2161 and PG2173. Step a: Methyl 6-azido-6-deoxy-2,3-di-0-benzylidene-a-D-mannopyranoside Methyl 6-azido-6-deoxy-α-D-mannopyranoside (1.011 g, 4.61 mmol) was dissolved in anhydrous DMF (9 mL) and acetonitrile (9 mL). Benzaldehyde dimethyl acetal (1.38 mL, 9.22 mmol, 2 eq) and (±)-camphor-lO-sulfonic acid (214 mg, 0.922 mmol, 20 mol%) were added in that order. The mixture was stirred under house vacuum at 60 °C (external) overnight, and the volatile materials were removed on rotavap. The residue was loaded on silica gel and purified by column (silica 2.5x20 cm, gradient elution with hexane-ethyl aceate 10:1, 9:1, 7:1, 5:1, 4:1, 3:1 to 2:1). The fractions were pooled into two parts. The less polar part (Rf = 0.55 and 0.51, hexane-EtOAc 3:1) was a mixture of 4-acetals and the polar part was mainly the 4-OH product. The mixtures were combined and evaporated. The residue was re-dissolved in dichloromethane (50 mL) and stirred with IM ammonium chloride solution (50 mL). The less polar spots were slowly disappeared and converted into the polar product. However, the conversion was not further improved after 40 min: Thus the dichloromethane phase was separated and stirred with 0.5 M HC1 solution (50 mL) for another 20 min. TLC indicated no further change. The DCM phase was separated and washed with brine- IM NaOH, dried (MgSO4). The dried DCM solution was filtered, evaporated and the residue was purified by silica column as above to give the title compound as a colourless gummy solid (0.543 g, 38%, Rf = 0.29, EtOAc-hexane = 1:3). 1H NMR (CDC13, 400 MHz): two benzylidene epimers in a ratio of 1:1. 7.49-7.36 (m, 5H, C6H5), 6.126 and 5.902 (2xs, IH, benzylidene-CH), 5.066 and 4.987 (2xs, IH, sugar-Hl), 4.396 (dd, 0.5H, J= 6.4, 5.6, sugar-H3), 4.248 (dd, 0.5H, J= 6.4, 6.0, sugar-H3), 4.214 (dd, 0.5H, J = 6.0, sugar-H2), 4.102 (dd, 0.5H, J = 4.8, sugar-H2), 3.86-3.38 (m, 4H), 3.466 and 3.424 (2xs, 3H, CH3O), 2.984 (br s, IH, OH). 13C NMR (CDCI3, 100 MHz): 138.01, 136.41, 129.38, 128.99, 128.24, 128.12, 126.41, 125.86, 103.85, 102.60, 97.74, 97.53, 79.36, 77.75, 77.47, 74.81, 70.00, 68.91, 68.42, 66.97, 54.78, 51.18 and 51.10. Step b: Methyl 6-azido-3-0-benzyl-6-deoxy-a-D-mannopyranoside and methyl 6-azido-2-0- benzyl-6-deoxy- a-D-mannopyranoside A solution of methyl 6-azido-6-deoxy-2,3-di-O-benzylidene-α-D-mannopyranoside (240 mg, 0.781 mmol) in DMF (7.8 mL, 0.1 M) was treated with sodium cyanoborohydride (589 mg, 9.37 mmol, 12 eq) and molecular sieve 3A (1 g). The mixture was stirred at r.t. for 20 min, then at 70 °C while TFA (0.361 mL, 4.686 mmol, 6 eq) was added slowly. After addition, the mixture was stirred at 70 °C for 6 h, cooled to 0 °C and basified by addition of solid Na2CO3. The cold mixture was filtered and the cake washed with EtOAc. The filtrate and washings was extracted once with sat. Na2CO3. Evaporation gave a gum, which was purified by column chromatography (silica 2.5x18 cm, eluted with EtOAc-hexane 1 :4 to 1 : 1) to give methyl 6-azido-3-0-benzyl-6-deoxy- a-D-mannopyranoside (colourless gum, 64 mg, 26%, Rf - 0.45, EtOAc-hexane=l:l); 1H NMR (CDCI3, 400 MHz): 7.41-7.32 (m, 5H, Ph), 4.78 (d, IH, J1-2 = 1.6, HI), 4.70 (d, IH, Jgem = 12.0, CH2), 4.56 (d, IH, CH2), 4.02 (dd, IH, J2-3 = 3.2, H2), 3.78 (dd, IH, J3-4 = 8.4, J4-5 = 10.0, H4), 3.73 (ddd, IH, J5-6ax= 6.0, J5-6eq *= 3.2, H5), 3.64 (dd, IH, H3), 3.52 (dd, IH, J6aχ-6eq = 13.2, H6eq), 3.47 (dd, IH, J6ax.6eq = 13.2, J5-6ax = 6.0, H6ax), 3.40 (s, 3H, MeO), 2.21 (br s, 2H, 2xOH) and methyl 6-azido-2-0-benzyl-6-deoxy-a-D- mannopyranoside (colourless waxy solid, 62 mg, 26%, Rf = 0.27, EtOAc-hexane-=l:l). 1H NMR (CDCI3, 400 MHz): 7.38-7.28 (m, 5H, Ph), 4.78 (s, IH, HI), 4.71 (d, IH, J= 11.6, CH2), 4.53 (d, IH, = 11.6, CH2), 3.75-3.61 (m, 4H, H2, H3, H4 and H5), 3.53 (dd, 1H, J= 13.2, 1.5, H6eq), 3.46-3.40 (dm, IH, J= 13.2, H6ax), 3.38 (s, 3H, MeO), 2.83 (br s, 2H, 2xOH). Step c: Methyl 6-azido-2-0-benzyl-6-deoxy-2,3-di-0-sulfonato-a-D-mannopyranoside disodium salt (PG2160) Methyl 6-azido-2-0-benzyl-6-deoxy-a-D-mannopyranoside was sulfonated according to the standard procedure to yield the title compound as a white powder, 64 mg, 59%. 1H NMR (D2O, 400 MHz): 7.38-7.26 (m, 5H, Ph), 4.72 (d, IH, Jgem = 12.0, CH2Ph), 4.59 (d, IH, J1-2 = 2.0, HI), 4.58 (d, IH, CH2Ph), 4.49 (d, IH, J2-3 = 2.8, J3-4 = 9.6, H3), 4.44 (dd, IH, J4-5 = 9.6, H4), 4.07 (dd, IH, H2), 3.78 (ddd, IH, J5-6ax = 5.6, J5-6eq = 2.4, H5), 3.62 (dd, IH, J6ax-6eq = 13.2, H6eq), 3.51 (dd, IH, J6ax-6eq= 13.2, J5-6ax= 5.6, H6ax), 3.22 (s, 3H, MeO). Step d: Methyl 6-azido-3-0-benzyl-6-deoxy-2,4-di-0-sulfonato-a-D-mannopyranoside disodium salt (PG2161) Methyl 6-azido-3-0-benzyl-6-deoxy- a-D-mannopyranoside (64 mg) was sulfonated according to the standard procedure to yield the title compound as a white powder, 58 mg, 66%. 1H NMR (D2O, 400 MHz): 7.42-7.21 (m, 5H, Ph), 4.92 (d, IH, J1-2 = 2.4, HI), 4.67 (d, IH, Jgem- 12.4, CH2Ph), 4.60 (d, IH, CH2Ph), 4.56 (dd, IH, J2-3 = 3.2, H2), 4.35 (dd, IH, J3-4 = 9.6, J4-5 = 9.6, H4), 3.80 (dd, IH, H3), 3.75 (ddd, IH, J5-6ax - 6.0, J5-6eq = 2.8, H5), 3.66 (dd, IH, J6ax-6eq-= 13.4, H6eq), 3.54 (dd, IH, J6ax-6eq= 13.4, J5-6ax= 6.0, H6ax), 3.29 (s, 3H, MeO). Step e: Methyl 6-[l '-(4-phenyl)triazolyl]-2-0-benzyl-6-deoxy-2,3-di-0-sulfonato-a-D- mannopyranoside disodium salt (PG2173) Methyl 6-azido-2-0-benzyl-6-deoxy-2, 3 -di-O-sulfonato- -D-mannopyranoside disod- ium salt was subjected to the Huisgen reaction general procedure using phenyl acetylene to yield the title compound as a white powder 6.8 mg, 67%, Rf = 0.34, EtOAc-MeOH-H2O = 10:2:1. 1H NMR (D2O, 400 MHz): 8.26 (s, IH, triazole), 7.64-7.60 (m, 2H), 7.37-7.15 (m, 8H), 4.89 (dd, IH, J*= 14.4, 2.8, H3), 4.67 (d, IH, J= 12.0, PhCH2), 4.57-4.39 (m, 5H, PhCH2, HI, H4 and H6eq and H6ax), 4.04 (dd, IH, J= 2.8, 2.0, H2), 3.99 (ddd, IH, J= 9.2, 8.8, 2.4, H5), 2.83 (s, 3H, MeO). Example 19: PG2170 Allyl 6-azido-2, 3-0-disulfonato-6-deoxy-4-0-(l-naphthylmethyl)- a-D-mannopyranoside disodium salt (containing 10% of 2-naphthylmethyl isomer) (PG2170) The title compound, prepared analogously to PG2163 beginning with allyl 6-azido-6- deoxy- a-D-mannopyranoside, was obtained as a white powder 87.5 mg, 87%, Rf = 0.28
(major), 0.22 (minor), EtOAc-MeOH-H2O = 10:2:1. 1H NMR (D20, 400 MHz): 7.87 (d, IH, J = 8.4, naphthyl), 7.65-7.54 (m, 2H, naphthyl), 7.33-7.19 (m, 4H, naphthyl), 5.68 (ddt, IH, Jaiiyi2'-3'trans = 22.0, nyi2'-3'cis = 10.8, Jaiiyii'-2' = 6.0, allyl-2'), 5.23 (AB quartet, 2H, Jgem = 12.0, naphthyl-CH2), 5.17-5.02 (m, 3H, allyl-3' and HI), 4.74 (dd, IH, J1-2-= 2.0, J2-3 = 3.2, H2), 4.64 (d, IH, J3-4= 9.6, H3), 3.90 (dd, IH, ιιyιrgem= 13.2, J- 6.0, allyl-1'), 3.81 (dd, IH, allyl-1'), 3.64 (t, IH, J4-5 = 9.6, H4), 3.39 (ddd, IH, J5-6eq= 2.0, J5-6ax= 5.2, H5), 2.78 (dd, IH, J6eq-6ax = 13.6, H6eq), 2.68 (dd, IH,
Figure imgf000035_0001
13.6, H6ax). Typical signals for minor isomer 2-naphthylmethyl derivative: 4.84 (AB quartet, 2H, Jgem = 11.2, naphthyl-CH2), 3.52 (ddd, IH, J4.5 = 10.0, J5-6eq= 2.4, J5-6ax= 6.0, H5), 3.07 (dd, IH, J6eq-6ax= 13.6, H6eq), 2.96 (dd, IH, eci-δax*--- 13.6,
Figure imgf000035_0002
6.0, H6ax). The source ofthe minor isomer is the commercial 9:1 mixture of 1- and 2-bromomethylnapthalene used in step a. Additional compounds were synthesized using appropriate modifications of the syntheses detailed above in Examples 1 to 19. These additional compounds are included in the tables giving the results of biological testing of compounds according to the invention. Example 20 Biological Testing of Compounds Methods 1. Growth Factor Binding Binding affinities of ligands for the growth factors were measured using a surface plasmon resonance (SPR) based solution affinity assay. The principle of the assay is that heparin immobilised on a sensorchip surface distinguishes between free and bound growth factor in an equilibrated solution of the growth factor and a ligand. Upon injection of the solution, the free growth factor binds to the immobilised heparin, is detected as an increase in the SPR response and its concentration thus determined. A decrease in the free growth factor concentration as a function of the ligand concentration allows for the calculation of the dissociation constant, Kd. It is important to note that ligand binding to the growth factors can only be detected when the interaction involves the heparin binding site, thus eliminating the chance of evaluating non-specific binding to other sites on the protein. A 1 : 1 stoichiometry has been assumed for all protein: ligand interactions. The preparation of heparin-coated sensorchips, via immobilisation of biotinylated BSA- heparin on a streptavidin-coated sensorchip, has been described [24]. Heparin has also been immobilised via aldehyde coupling using either adipic acid dihydrazide or 1,4-diaminobutane. For each Kd measurement, solutions were prepared containing a fixed concentration of protein and varying concentrations ofthe ligand in buffer. Ligands binding to FGF-1 and VEGF were measured in HBS-EP buffer (10 mM HEPES, pH 7.4, 150 mM NaCI, 3.0 mM EDTA and 0.005% (v/v) polysorbate 20), while binding to FGF-2 was measured in HBS-EP buffer containing 0.3 M NaCI [24]. Prior to injection, samples were maintained at 4 °C to maximise protein stability. For each assay mixture, 50-200 μL of solution was injected at 5-40 μL/min and the relative binding response measured. All surface binding experiments were performed at 25 °C. The surface was regenerated by injection of 40 μL of 4M NaCI at 40 μL/min, followed by injection of 40 μL of buffer at 40 μL/min. Sensorgram data were analysed using the BIAevaluation software (BIAcore).
Background sensorgrams were subtracted from experimental sensorgrams to produce curves of specific binding, and baselines were subsequently adjusted to zero for all curves. The relative binding response for each injection was converted to free protein concentration using the equation p]=- p\ \total rn. where r is the relative binding response and rm is the maximal binding response. Binding equilibria established in solution prior to injection were assumed to be of 1:1 stoichiometry. Therefore, for the equilibrium,
P + L === P-L
where P corresponds to the growth factor protein, L is the ligand, and P-L is the protein: ligand complex, the equilibrium equation is
Figure imgf000036_0001
and the binding equation [24] can be expressed as
Figure imgf000036_0002
The Kd values given are the values fit, using the binding equation, to a plot of [P] versus [L]totai- Where Kd values were measured in duplicate, the values represent the average ofthe duplicate measurements. It has been shown that GAG mimetics that bind tightly to these growth factors elicit a biological response in vivo [24]. 2. Antiviral Assays. Selected compounds were tested against two types of herpes simplex virus (HSV), i.e., HSV-1 and HSV-2, in two assays for inhibition of viral infectivity and cell-to-cell spread, as described by Nyberg et al. [25]. Monolayer cultures of African green monkey kidney cells (GMK AHl) [26] cultivated in 6-well cluster plates, were used. The viral strains used were herpes simplex virus type 1 (HSV-1) KOS321 strain [27] and HSV-2 strain 333 [28]. In both assays the compounds were tested at 200 μM. (i) In the assay of HSV infectivity, the compounds were mixed with the virus, incubated for 10 min at room temperature and then the mixture was added to cells, and kept on cells for lh only to allow (or not) the virus attachment to/entry into the cells. Thus this assay reflects whether or not the compound in question has the ability to bind to the virus particles and block its attachment to/entry into the cells. An inhibition is manifested as a decreased number of viral plaques. (ii) The next assay, termed HSV spread, relies on the addition of compound to the cells after the virus attachment/entry steps have already occurred. This assay reflects whether the examined compound has the ability to inhibit virus transmission from an infected to an uninfected cell (cell-to-cell spread) and in addition whether the compound has the ability to enter the cells and inhibit viral replication. Lack of compound activity in the assay of virus infectivity but some activity in the virus spread assay suggests that the compound acts by entering the cells and inhibition of viral replication step(s). An inhibition is manifested as a reduction in the size of viral plaques. The results (see Table 5) are expressed as % of control, ie., as the number (infectivity assay) or the size (spread assay) of viral plaques developed in the presence of compound relative to the mock-treated controls (no compound). Results The results ofthe tests as described in the preceding section are presented in Tables 1 to
5. Table 1
Figure imgf000037_0001
PG# KdaFGF Kd bFGF Kd VEGF KdFGF-4
Figure imgf000038_0001
RA,RF,RH=OMe; RB,Rc,RE,Rι=OS03Na; 2037 47.7 μM 507 μM 645 μM
RD=CH2OS03Na; RG=H 2038 77.9 μM 2.10 mM 368 μM
Figure imgf000038_0002
RA=OMe; RF,RH=OH; RB,Rc,RE,R OS03Na; 2039 21.8 μM 3.50 mM 1.27 mM
Figure imgf000038_0003
RF,RG,RI=OH; RD-RA=-CH20-; RB,RE=NHS03Na; 2046 6.35 mM 3.70 mM 1.50 mM
Rc=OBn; RH=H
RF,RG,RRc=OH; RD-RA=-CH20-; RB,RE=NHS03Na; 2047 388 μM 1.95 mM 2.55 mM RH=H
RF,RH,RI=OH; RA=OMe; RB.RcRE^OSOsNa; 2063 1.39 mM 2.35 mM 2.59 mM D>RG =H
Table 2
Figure imgf000038_0004
PG# KdaFGF Kd bFGF Kd VEGF
RK=OMe; RM,Ro=OS03Na; RQ=OBn; RR=CH3; 2023 1.76 mM 4.90 mM 2.27 mM RJ>RJ )RP)RS =H
RK=OMe; RM,Ro*=0SO3Na; RP=OBn; RR=CH3; 2024 4.73 mM 3.65 mM 6.40 mM
Figure imgf000038_0005
Figure imgf000038_0006
RK=OMe,RM,Ro=OS03Na;RQ=Oallyl;RR=CH3; ^ ^^ >1000mM 236 M
Kjjl ji ^K-PjK-S-'-!
^JJ1R^L>R1 D")CΛΛPJK0R"-_;τ1?-IM=OSθ3Na:RN=OMe;RQ=°Bn; 2030 317 μM 4.61 mM
Figure imgf000038_0007
? ;^ ;5=CH2θS°3Na; 2°4 9.38 mM 5.10 mM 1.04 mM
^ ΛJ,K ,KN,; R RQϊ, S-11 3Na;Rθ=OH;Rp=°allyl;RR=CH3; 2042 3.05 mM 10.7 mM 2.59 mM PG # Kd aFGF Kd bFGF Kd VEGF 2043 6.43 mM 17.4 mM 1.73 mM
Figure imgf000039_0001
RK =OMe; RM,Ro=OS03Na; RP=OOCCH2CH2Ph; RR=CH3; 2044 366 μM 1.55 mM 1.65 mM RJ,RL,RN,RQ>RS =H
Rj/RK=H/OMe (anomeric mixture); RS-RN = -CH20-; 2045 392 μM 3.40 mM 1.07 mM RM=OS03Na; RQ=OBn; RL,R0,RP,RR=H
Figure imgf000039_0002
RK=OMe; RN,RM=OBn; Rg-=OS03Na; Rs=CH2OS03Na; 2049 1.51 mM » 60.0 μM 2.72 mM RJ,RL,RO,RP!RR =H
RK=OMe; RM =OS03Na; RN,Ro=OBn; 2050 3.31 mM 8.25 mM 10.00 mM RJ,RL,RO,RP,RR,RS =H
2051 2.46 mM > 20.4 mM 4.63 mM
Figure imgf000039_0003
RK=OMe; RM,Ro=OS03Na; RP=OOCCH2OPh; RR=CH3; 2052 5.92 mM 4.50 mM 686 μM RJJ L,RN)RQ>RS =H
RK=OMe; RM,Ro=OS03Na; RP=Oallyl; RR=CH3; 2053 1.30 mM 5.17 mM 343 μM RJ)R >RNJ Q)RS =H
RK=OMe; RM,R0-=OS03Na; RP=OBz; RR=CH3; 2054 454 μM 2.73 mM 403 μM RJ)RL)RN;RQ>RS=H
RK=OMe; RM,Ro=*OS03Na; RP=OOCPh(p-OMe); RR=CH3; 2056 797 μM 2.45 mM 485 μM RJ)RL,RN) QJRS =H
2079 1.92 mM ~ 3.45 mM 1.73 mM
Figure imgf000039_0004
RS-RN= -CH20-; RM=OS03Na; RQ=OBn; 2080 1.62 mM ~ 11.7 mM 1.36 mM RJ;RK)R > O)RP)RR=H 2085 9.60 mM ~ 17.4 mM 8.90 mM
Figure imgf000039_0005
RK=OMe; RM)Ro=OS03Na; RP=0(CH2)3OPh; RR=CH3; 2086 3.05 mM 1.50 mM RJ>R )RN)RQ)RS =:H
RK=OMe; RM;Ro-=OS03Na; RP=OH; RR=CH3; 2087 566 μM 2.35 mM 899 μM RJ;RL, NJ Q)RS =H
RK=OMe; RM,Ro=OS03Na; RP=0(CH2)3Ph; RR=CH3; 2088 676 μM 3.00 mM 761 μM J)R ;RN»RQ)RS =H
RK=OMe; RM,Ro=OS03Na; RP=OCH2(2-Napthyl); 2089 1.20 mM 2.15 mM 2.33 mM RR=CH3; RJ,RL,RN;RQ,RS =H
RK=OMe; RM,Ro=OS03Na; RP=OCH2(£)CH=CHPh; 2090 3.85 mM 2.50 mM 3.02 mM RR=CH3; RJ,RL,RN,RQ,RS=H
RK=OMe; RM,R0=OS03Na; RP=OCH2(l-Napthyl); 2091 1.37 mM 1.50 mM 1.98 mM RR=CH3; RJ,RL,RN,RQ,RS-=H PG # Kd aFGF Kd bFGF Kd VEGF
RK=OMe; RM,Ro=OS03Na; RP=OCH2Ph(p-Me); RR=CH3; 2092 2.70 mM 2.85 mM 2.86 mM RJ,RL,RN,RQ,RS=H
Figure imgf000040_0001
Rκ=NHCOCH2θPh(2,4-di-Cl); RMjRN,RQ=OSθ3Na; 2096 35.7 μM 141 μM 20.4 μM Rs *=CH2OS03Na; RJ,RL,RO;RP,RR=H
Rj=OMe; RL,RN=OBz; R0=OSO3Na; Rs=CH2OS03Na; 2097 127 μM 2.05 mM 267 μM K>R J OJRP;RR =H
RK=OMe; RM,Ro=0SO3Na; RP-=OCH2Ph(p-CF3); RR=CH3; 2098 X85 mM 2.65 mM 2.60 mM R.,RL>RN>RQ!RS =H RP=OCH2Ph(».-CF3); RR=CH3; 2099 1.30 mM 2.85 mM 5.70 mM
Figure imgf000040_0002
Rj=OMe; RL,RQ=OS03Na; RN=Oallyl; Rs=CH2Oallyl; 2100 1.25 mM 18.1 mM 146 μM RKJRMJ OJRPJ R^H
Rj=OMe; RL,RN=OH; RQ=OS03Na; Rs=CH2OS03Na; 2101 77.3 μM 188 μM RK> 5RO; P3RR =H
RK=OMe; RM,R0=OS03Na; RP=OMe; RR=CH3; 2102 116 μM 1.30 mM 206 μM
RJ;RL>RNJRQJRS=H
RK=OMe; RM,Ro=OS03Na; RP=OCH2Cyclopropyl; 2103 5.50 mM 4.20 mM 3.00 mM RR=CH3; RJ,R ,RN,RQ,RS=H
RK=N3; RM,Ro*=OS03Na; RP=OBn; RR=CH3; 2104 1.80 mM 2.45 mM 3.30 mM RJ,R ;RN> Q>RS =H
RJ=N3; RM=OS03Na;R0=OH; RP=OBn; RR=CH3; 2105 1.85 mM 2.10 mM 8.30 mM RK, L)RN;RQ, S =H
RK=OBn; RM,Ro=OS03Na; RP=OMe; RR=CH3; 2106 1.31 mM 3.43 mM 1.45 mM RJ;R JRNJRQ;RS =H
RK=OMe; RM,RN=OOCPh( -OMe); RQ=OS03Na; 2107 645 μM Rs=CH2OS03Na; RJ,RL,RO,RP,RR=H
RK=OMe; RM,RN=OCH2(£)CH=CHP1.; RQ=OS03Na; 2108 563 μM Rs=CH2OS03Na; RJ,RL,RO,RP,RR=H
Figure imgf000040_0003
RK=OMe; RM,R0=OS03Na; RP=OOCP (3,4-di-Cl); 2111 1.30 mM RR=CH; RJ,RL,RN,RQ,RS =H 1.60 mM
Figure imgf000040_0004
RK=OMe; RM,R0=OS03Na; RP=OOCCH2Ph(3,4-di-Cl); 2113 RR=CH3; RJ,RL,RN,RQ,RS =H 1.45 mM 1.60 mM 1.60 mM PG# KdaFGF Kd bFGF Kd VEGF
2114 3.85 mM 2.30 mM 888 μM
2129 5.40 mM 9.10 mM 3.00 mM
Figure imgf000041_0001
Rs=CH2Oallyl; 2130 1.05 mM 7.85 mM 361 μM
Figure imgf000041_0002
RK=OMe; RM,Ro=OS03Na; RP=OEt; RR=CH3; 2131 8.30 mM 7.20 mM 21.8 mM JJRLJRNJRQJRS13"!!
Figure imgf000041_0003
2139 884 μM ~2.70mM 383 μM
Figure imgf000041_0004
Figure imgf000041_0005
Rj=OMe; RL,RQ=OS03Na; RN=Oallyl; 2142 3.00 mM 481 μM Rs*=CH2OCH2(CH3)C=CH2; RK,RM;RO>RP) R=H 1.10 mM 398 μM
Figure imgf000041_0006
Figure imgf000041_0007
RN=Oallyl; Rs=CH2OH; 2145 ~5.95mM ~23.0mM 1.60 mM RN=Oallyl; Rs=CH2OBn; 2147 1.50 mM 9.10 mM 2.10 mM
Figure imgf000041_0008
Rs=CH2OCH2(3- 2148 2.40 mM ~13.0mM 981 μM
Figure imgf000041_0009
2149 2.50 mM ~6.70mM 709 μM
2150 »6.00mM ~24.2mM 7.80 mM
Figure imgf000041_0010
Rs=CH2Oallyl; 2151 3.50 mM ~9.90mM 935 μM
Figure imgf000041_0011
Figure imgf000041_0012
2153 ~11.5mM »42.1mM -ll.lmM
2154 ~3.70mM ~26.7mM 2.15 mM
Figure imgf000041_0013
PG # Kd aFGF Kd bFGF Kd VEGF
Figure imgf000042_0001
RN=Oallyl; RS=CH2N3; 2156 360 μM 1.65 mM 1.04 mM
Figure imgf000042_0002
Figure imgf000042_0003
Rj=OMe; RL=OH;RN=OS03Na; RQ=OCH2(2-napthyl); 2158 1.55 mM ~ 10.3 mM 985 μM
Figure imgf000042_0004
Rj=OMe; RL,RQ=OH; RN=OS03Na; RS=CH2NH2; 2159 1.90 mM 62.3 mM 6.40 mM
Figure imgf000042_0005
Rj=OMe; RL=OBn; RN,RQ=OS03Na; RS=CH2N3; 2160 4.40 mM 2.50 mM 12.9 mM
Figure imgf000042_0006
Figure imgf000042_0007
Rj=OMe; RL,RN=OS03Na; RQ=Oallyl; RS=CH2N3; 2163 396 μM 4.80 mM 61.2 μM
Figure imgf000042_0008
Rj=OMe; RL,RN=OS03Na; Ro=OCH2(2-napthyl); 2164 1.35 mM 1.70 mM 1.30 mM
Figure imgf000042_0009
Rj/RK=H/OMe (anomeric mixture); RS-RN= -CH20-; 2165 1.13 mM ~ 25.5 mM 1.70 mM RM=OBn; RQ=OS03Na; RJ,RK,RL,RO,RP,RR=H 2166 3.60 mM 1.90 mM 2.70 mM
Figure imgf000042_0010
Figure imgf000042_0011
2171 845 μM ~ 18.9 mM 2.00 mM
Figure imgf000042_0012
Rj=OMe; RL=OBn; RN,RQ=OS03Na; RS=CH2NH2; 2172 3.80 mM ~ 5.90 mM 2.30 mM RKJ M. OJRPJRR31"!!
Rj=OMe; RL=OBn; H2(4-phenyl- 2173 18.9 μM 918 μM 89.3 μM [l,2,3]triazol-l-yl);
Figure imgf000042_0013
Rj=Oallyl; RL=OH; RN=OS03Na; RQ=OBz; RS=CH2N3; 2174 278 μM ~ 19.3 mM 846 μM
Figure imgf000042_0014
Figure imgf000042_0015
PG # Kd aFGF Kd bFGF Kd VEGF RS=CH2(4- l-yl); 2178 130 μM 1.20 mM 60.9 μM
Figure imgf000043_0001
2179 52.9 μM 143 μM 10.3 μM 2180 91.9 μM 1.90 mM
Figure imgf000043_0002
Rι=OMe; RL,RN=OS03Na; RQ=Oallyl; RS=CH2(4- (CH2NHCOCH2OPh)-[l,2,3]triazol-l-yl); 2181 365 μM 203 μM
RK>RM;ROJRP;RR =H
Rj=OMe; RL,RN=OS03Na; RQ=Oallyl; RS=CH2(4- 2182 107 μM (CH2NHCOPh)-[l,2,3]triazol-l-yl); RK,RM,Ro,RP,RR=H 847 μM
Rj=OMe; RL,RN=OS03Na; Rς Oallyl; RS=CH2(4-(CH2-.V- 2183 324 μM phthalimido)-[l,2,3]triazol-l-yl); RK,RM,Ro,RP,RR=H 82.7 μM
Rj=OMe; RL,RN=OS03Na; Ro=Oallyl; RS=CH2(4- (CH2 HS02Ph(p-Me))-[l,2,3]triazol-l-yl); 2184 388 μM 1.6 mM
RKJ M) 0JRP;RR =H
Figure imgf000043_0003
Rj=OMe; RL,RN=O [l,2,3]triazol-l-yl);
Figure imgf000043_0004
Rj=OMe; RL=OS03Na; RN=OH; Ro=OCH2(2-napthyl); Rs=CH2N3; RK,R ,ROJRPJ R=H 2187 320 μM
Figure imgf000043_0005
Rj=OMe; RL)RN=OS03Na; Rς Oallyl; Rs=CH2NHS02Me; 2189
RK> >R0;RP;RR=H 191 μM 35.0 μM
Figure imgf000043_0006
Rj=OMe; RL,RN=OS03Na; RQ=Oallyl; RS=CH2(4- (CH2NHCO(o-C02Na)phenyl)-[ 1 ,2,3]triazol- 1-yl); 2193 1.20 mM 270 μM RKJRMJROJRP)RR=H
Figure imgf000043_0007
; 2194 1.25 mM >2.8 mM
R >RM>ROIRP;RR=H
Figure imgf000043_0008
PG # Kd aFGF Kd bFGF Kd VEGF
RK=OBn; RM,R0 *=OS03Na; RP=OBn; RR=CH3; 2196 170 μM 2.50 mM RJ,RL,RN,RQ,RS=H
Figure imgf000044_0001
Table 3
Figure imgf000044_0002
PG # Kd aFGF Kd bFGF Kd VEGF
Rτ=l,2,3,4-tetra-0-sodiιun sulfonato-D-glucuronoyl;
Figure imgf000044_0003
Rτ= 1 -O-Me-2,3 ,4-tri-O-sodium sulfonato-α-D- mannopyranos-6-yl-acetyl; Ru=CH2CH2OS03Na; Rγ=H ; 2008 296 μM 551 μM 335 μM Rw=cyclohexyl
RT=Ac; Ru=2-(2,3,4,6-tetra-0-sodium sulfonato-α-D- mannopyranos-l-C*-yl-)-ethyl; Rv=H; Rw=2-(2,3,4,6-tetra- 2009 428 μM (9-sodium sulfonato-α-D-mam opyranos- 1 -0-yl-)-ethyl
Rτ=3-(2,3,4,6-tetra-C-sodium sulfonato-α-D- mannopyranos- 1 -0-yl-)-propyl; Ro="COCH2CH2Ph; 2010 556 μM Rw=cyclohexyl; Rv-H
Rτ=l,2,3,4-tetra-0-sodium sulfonato-D-glucuronoyl; 2011 62.4 μM Ru=Bn; R ^cyclohexyl; Rv=P
Rτ=l,2,3,4-tetra-0-sodium sulfonato-α-D-glucuronoyl; 2012 122 μM 505 μM Ru=Bn; R =cyclohexyl; RV=H
Figure imgf000044_0004
Rτ=l,3,4,6-tetra-0-sodium sulfonato-α-D-mannopyranos-2- yl-acetyl; Ru=Bn; Rγ=H; Rw=2-(2,3,4,6-tetra-0-sodium 2014 5.09 μM 85.1 μM 8.82 μM sulfonato-α-D-mannopyranos- 1 -0-yl-)-ethyl
Rτ=3-(2,3,4,6-tetra-( -sodium sulfonato-α-D- mannopyranos-l-0-yl-)-propyl; Rυ= CO(CH2)3Ph; Rv=H; 2018 R =cyclohexyl
Rτ=l,2,3,4-tetra-0-sodium sulfonato-α-D-glucuronoyl; 2020 Ru,Rw=Bn; RV=H 104 μM 206 μM 437 μM
Rτ=l-0-Me-2,3,4-tri-C*-sodium sulfonato-α-D- 2032 mannopyranos-6-yl-acetyl; Ru=Bn; Rw=cyclohexyl; Rγ=H 260 μM 201 μM 705 μM PG# Kd aFGF Kd bFGF Kd VEGF
Rτ=l-0-Me-2,3,4-tri-0-sodium sulfonato-α-D- mannopyranos-6-yl-acetyl; Ru=Bn; Rv^H; Rw=2-(2,3,4,6- 2034 37.6 μM 16.5 μM 115 μM tetra-O-sodium sulfonato-α-D-mannopyranos- 1 -0-yl-)-ethyl
RT=Ac; Ru=Bn; RV=H; Rw=2-(2,3,4,6-tetra-0-sodium 2035 sulfonato-α-D-mannopyranos- 1 -<9-yl-)-ethyl 24.8 μM 287 μM 76.6 μM
118 μM 2.50 mM 1.10 mM
Figure imgf000045_0001
RT=Ac; Ru=CH2CH2Ph; RV=H; Rw=6-deoxy-l~0-Me-2,3,4- 2058 224 μM 682 μM tri-O-sodium sulfonato-α-D-mannopyranos-6-yl 109 μM
RT=Ac; Ru=CH2CH2Ph; RV=H; R^ό-deoxy-l-O-Me-2,3,4- 2058 224 μM 682 μM tri-O-sodium sulfonato-α-D-mannopyranos-6-yl 109 μM
RT=Ac; Ru=Bn; RV=H; Rw=6-deoxy-l-0-Me-2,3,4-tri-C>- 2059 140 μM sodium sulfonato-α-D-mannopyranos-6-yl 192 μM 77.0 μM
RT=Ac; Ru=Bn; RV=H; Rw=6-deoxy-l-C>-Me-2,3,4-tri-0- 2059 140 μM sodium sulfonato-α-D-mannopyranos-6-yl 192 μM 77.0 μM
RT=Ac; Ru=Ph; RV=H; Rw=6-deoxy-l-0-Me-2,3,4-tri-0- 2060 196 μM 481 μM 76.3 μM sodium sulfonato-α-D-mannopyranos-6-yl
RT=Ac; Ru=cyclohexyl; RV=H; Rw=6-deoxy-l-0-Me-2,3,4- 2064 314 μM 413 μM 1.70 mM tri-O-sodium sulfonato-α-D-mannopyranos-6-yl
Rτ=Ac; Ru=CH2CH2OS03Na; RV=H; Rw=6-deoxy-l-0- 2065 94.8 μM 241 μM 283 μM Me-2,3 ,4-tri-0-sodium sulfonato-α-D-mannopyranos-6-yl
RT=m-ClPhCH2CO; Ru=H;
Figure imgf000045_0002
2066 37.4 μM 433 μM 45.3 μM 2,3,4-tri-(9-sodium sulfonato-α-D-marmopyranos-6-yl
RT=Et; Ru=CO(CH2)2COONa; Rγ=H; Rw=6-deoxy-l-0- 2068 338 μM 291 μM 207 μM Me-2,3 ,4-tri-O-sodium sulfonato-α-D-mamιopyranos-6-yl
RT=Et; Ru=CO(CH2)wCOONa; Rv==H; Rw=6-deoxy-l-0- 2069 160 μM 477 μM 104 μM Me-2,3 ,4-tri-O-sodium sulfonato-α-D-mannopyranos-6-yl
RT=Et; Ru=CO(CH2)9OS03Na; RV=H; Rw=6-deoxy-l-0- 2070 l l l μM 243 μM 545 μM Me-2,3,4-tri-0-sodium sulfonato-α-D-mannopyranos-6-yl
RT=Et; Ru=CO(CH2)wCOONa; RV=H; Rw=6-deoxy-l-0- Me-2,3 ,4-tri-O-sodium sulfonato-α-D-mannopyranos-6-yl, 2071 119 μM 2.24 mM 161 μM mixture of mono- and di- sulfates
RT=Ac; Ru=cyclohexyl; RV=H; R =6-deoxy-l-0-Me-2,3,4- tri-O-sodium sulfonato-α-D-mannopyranos-6-yl, mixture of 2072 264 μM 2.98 mM 277 μM mono- and di-sulfates
Figure imgf000045_0003
sulfonato-α-D-mannopyranos- 1 -< -yl-)-ethyl 2073 97.4 μM 236 μM 402 μM
RT=Ac; Ru=(CH2)2Ph; RV=H; Rw=2-(2,3,4,6-tetra-( - sodium sulfonato-α-D-mannopyranos- 1 -0-yl-)-ethyl 2074 11.8 μM 113 μM 28.8 μM
Rτ= m-ClPhCH2CO; RU;RV=H; Rw=2-(2,3,4,6-tetra-0- sodium sulfonato-α-D-mannopyranos-l-C>-yl-)-ethyl 2075 171 μM 837 μM 90.8 μM PG # Kd aFGF Kd bFGF Kd VEGF
RT=Et; Ru=CO(CH2)2C02Na; Rγ=H; Rw=2-(2,3,4,6-tetra- 2076 43.4 μM 118 μM 40.3 μM O-sodium sulfonato-α-D-mannopyranos- 1 -0-yl-)-ethyl
RT=Et; Ru=CO(CH2)4C02Na; Rγ=H; Rw=2-(2,3,4,6-tetra- 2077 43.6 μM 188 μM 81.1 μM O-sodium sulfonato-α-D-mannopyranos- 1 -<9-yl-)-ethyl
RT=Et; Ru=CO(CH2)9OS03Na; RV=H; Rw=2-(2,3,4,6-tetra- 2078 20.0 μM O-sodium sulfonato-α-D-mannopyranos- 1 -Oyl-)-ethyl 157 μM 49.6 μM
Rr=Bz; Ru=Et; RV=H; Rw=6-deoxy-l-0-Me-2,3,4-tri-0- 2081 sodium sulfonato-α-D-mannopyranos-6-yl 366 μM 480 μM 1.10 mM
RT=CO(CH2)2Ph; R0=Et; Rγ=H; Rw=6-deoxy-l-0-Me- 2082 2,3 ,4-tri-O-sodium sulfonato-α-D-mannopyranos-6-yl 596 μM
403 μM 80.7 μM
Figure imgf000046_0001
RT=COCH2OS03Na; Ru=Et; Rγ=H; Rw=6-deoxy-l-0-Me- 2084 161 μM 2,3 ,4-tri-O-sodium sulfonato-α-D-mannopyranos-6-yl 192 μM 277 μM
Rw=cyclohexyl; Ru=Ac; RV=H; Rτ=6-deoxy-l-0-Me-2,3,4- 2094 246 μM tri-O-sodium sulfonato-α-D-mannopyranos-6-yl 565 μM l.lO mM
Rw=cyclohexyl; Ru=Ac; RV=H; Rτ=6-deoxy-l-0-Me-2,3- 0-benzylidene,4-0-sodium sulfonato-α-D-mannopyranos-6- 2115 5.10 mM 3.90 mM yi
Figure imgf000046_0002
2,3 ,4-tri-O-sodium sulfonato-α-D-mannopyranos-6-yl 369 μM 411 μM 3.00 mM
RT=Ac; Ru=cycloheptyl; Rγ=H; R^ό-deoxy-l-O-Me^^- 2117 1.50 mM di-O-sodium sulfonato-α-D-mannopyranos-6-yl 2.90 mM
RT=Ac; Ru=cyclooctyl; RV=H; R^ό-deoxy-l-O-Me^^-di- 2120 1.60 mM O-sodium sulfonato-α-D-mamιopyranos-6-yl 11.0 mM
RT=COCH2Ph(p-CF3); Ru=Et; RV=H; Rw=6-deoxy-l-0- 2122 Me-2,4-di-0-sodium sulfonato-α-D-mannopyranos-6-yl 3.70 mM 1.20 mM
RT=COCH2Ph(p-CF3); Ru=Et; Rγ=H; Rw=6-deoxy-l-0- 2124 242 μM Me-2,3-di-0-sodium sulfonato-α-D-mannopyranos-6-yl 570 μM
R =cyclohexyl; Ru=Ac; Rv=H; Rτ=6-deoxy-l-C-Me-4-0- 2125 26.6 mM sodium sulfonato-α-D-mannopyranos-6-yl ~ 166 mM
RT=Ac; Ru=cyclododecyl; RV=H; R^ό-deoxy-l-OMe- 2126 2,3 ,4-tri-O-sodium sulfonato-α-D-mannopyranos-6-yl 265 μM 483 μM 1.20 mM
RT=Ac; Ru=4-t-butylcyclohexyl; Rv=H; Rw=6-deoxy-l-< - 2132 Me-2,3,4-tri-0-sodium sulfonato-α-D-mannopyranos-6-yl 243 μM 544 μM 1.00 mM
RT=Ac; Ru=l-(l-adamantyl)-ethyl; Rγ=H; R^ό-deoxy-l- 2133 0-Me-2,4-di-0-sodium sulfonato-α-D-mannopyranos-6-yl 398 μM
Figure imgf000046_0003
di-O-sodium sulfonato-α-D-mannopyranos-6-yl PG# Kd aFGF Kd bFGF Kd VEGF
Rτ=6-deoxy- 1 -<9-Me-2,3 ,4-tri-0-sodium sulfonato-α-D- mannopyranos-6-yl; Ru=CH2CO]SfHcyclo exyl; 2138 418 μM 449 μM 1.34 mM Rw=cyclohexyl; RV=H
Rw=cyclohexyl; Ru=CHO; RV=H; Rτ=6-deoxy-l-0-Me- 2162 1.20 mM 2,3 ,4-tri-O-sodium sulfonato-α-D-mannopyranos-6-yl
Table 4
Figure imgf000047_0001
PG # Kd aFGF Kd bFGF Kd VEGF
Rx=COCH2C(CH3)2CH2CO;
Figure imgf000047_0002
2015 2.94 μM 7.56 μM 267 nM sulfo-α-D-mannopyranos- 1 -0-yl)-propyl
Rx= 1 ,4-tτ-αrø-cyclohexyl; Rγ= 1 -O-Me-2,3 ,4-tri-O-sodium 2016 32.6 μM 81.8 μM 931 nM sulfo-α-D-mannopyranos-6-yl-acetyl
Figure imgf000047_0003
2057 18.8 μM 61.6 μM 55.6 μM sulfo-α-D-mannopyranos- 1 -0-yl)-ethyl, undersulfated
Figure imgf000047_0004
2062 10.3 μM 29.7 μM 17.3 μM sulfo-α-D-mannopyranos- 1 -0-yl)-ethyl
Table 5: Antiviral Testing Results PG# HSV-1 Infectivity HSV-2 Infectivity HSV-1 Spread HSV-2 Spread
2000 100.8 NT (= = not tested) 82.1 NT
2001 85.6 NT 88.3 NT 2002 89.2 NT 113.8 NT
2003 97 NT 95.2 NT
2005 102.8 NT 131.7 NT 2040 106.6 NT 86.2 NT 2041 108.3 NT 60 NT 2042 92 NT 107.6 NT PG# HSV-1 Infectivity HSV-2 Infectivity HSV-1 Spread HSV-2 Spread
2000 100.8 NT (=nottested) 82.1 NT
2001 85.6 NT 88.3
2002 89.2 NT 113.8 NT 2003 97 NT 95.2 NT 2005 102.8 NT 131.7 NT
2040 106.6 NT 86.2 NT
2041 108.3 NT 60 NT
2044 73.5 NT 77.9 NT
2085 99.2 NT 64.1 52.8
2091 81.8 135.6 74.9 58.9
2092 90.3 101.4 75.4 50
2093 90.3 121.6 74.9 55.5
2097 101.8 111.5 45 42.2
2098 81.8 116.3 68.4 54.4
2099 89.5 115.9 76 43.3
2103 80.8 112 94.2 70
2111 100.3 105.3 57.9 55.5
2112 95.6 90.9 74.9 76.7
2113 95.1 91.3 46.2 31.1
2114 86.4 99 71.3 68.9
2139 86.8 81.8 47.8 20
2146 108.5 92 56.2 52.8
2145 92.1 76 73.7 77.8
2151 100 75.5 84.5 86.1 The results presented in Tables 1 to 4 demonstrate that the broad range of compounds embraced by the invention have strong affinity for GAG-binding growth factors and may thus serve as modulators of their activity. The results in presented in Table 5 demonstrate that the compounds do indeed possess in vivo activity. The foregoing embodiments are illustrative only ofthe principles ofthe invention, and various modifications and changes will readily occur to those skilled in the art. The invention is capable of being practiced and carried out in various ways and in other embodiments. It is also to be understood that the terminology employed herein is for the purpose of description and should not be regarded as limiting. The term "comprise" and variants ofthe term such as "comprises" or "comprising" are used herein to denote the inclusion of a stated integer or stated integers but not to exclude any other integer or any other integers, unless in the context or usage an exclusive interpretation of the term is required. Any reference to publications cited in this specification is not an admission that the disclosures constitute common general knowledge in Australia.
REFERENCES
[I] Kjellen, L.; Lindahl, U. Annu. Rev. Biochem. 1991, 60, 443.
[2] Sugahara, K.; Kitagawa, H. Curr. Opin. Struct. Biol. 2000, 10, 518.
[3] 20179369 Tumova, S.; Woods, A.; Couchman, J.R. Int. J. Biochem. Cell Biol. 2000, 32, 269.
[4] Capila, I.; Linhardt, R.J. Angew. Chem. Int. Ed. 2002, 41, 391.
[5] Casu, B.; Lindahl, U. Adv. Carbohydr. Chem. Biochem. 2001, 57, 159.
[6] Liu, J.; Thorp, S.C. Med. Res. Rev. 2002, 22, 1.
[7] van Boeckel, C.A.A.; Petitou, M. Angew. Chem. Int. Ed. Engl. 1993, 32, 1671. [8] Petitou, M.; Herault, J.P.; Bernat, A.; Driguez, P.A.; Duchaussoy, P.; Lormeau, J.C.; Herbert, J.M. Nature 1999, 398, 417. [9] Yeh, B.K.; Eliseenkova, AN.; Plotnikov, A.Ν.; Green, D.; Pinnell, J.; Polat, T.; Gritli-Linde, A.; Linhardt, R.J.; Mohammadi, M. Mol. Cell. Biol. 2002, 22, 7184. [10] Liekens, S.; Leali, D.; Neyts, J.; Esnouf, R.; Rusnati, M.; Dell Era, P.; Maudgal, P.C; De Clercq, E.; Presta, M. Mol. Pharmacol. 1999, 56, 204.
[I I] Sola, F.; Farao, M.; Pesenti, E.; Marsiglio, A.; Mongelli, N.; Grandi, M. Cancer Chemother. Pharmacol. 1995, 36, 211.
[12] Foxall, C; Wei, Z.; Schaefer, M.E.; Casabonne, M.; Fugedi, P.; Peto, C; Castellot, J.J., Jr; Brandley, B.K. J. Cell. Physiol. 1996, 168, 651. [13] Parish, C.R.; Freeman, C; Brown, K.J.; Francis, D.J.; Cowden, W.B. Cancer Res. 1999, 59, 3433. [14] Kisilevsky, R.; Green, A.M.; Gervais, F. 2001, US Patent No. 6,310,073. [15] Dόmling, A.; Ugi, I. Angew. Chem. Int. Ed. 2000, 39, 3168 and references cited therein. [16] Lockhoff, O.; Frappa, I. Combinatorial Chemistry & High Throughput Screening 2002, 5, 361 and references cited therein. [17] Hanessian, S. Preparative Carbohydrate Chemistry; Chapter 3; Marcel Dekker Inc.: New York, 1996. [18] Pozsgay, N.; Trinh, L.; Shiloach, J.; Robbins, J.B.; Donohue-Rolfe, A.; Calderwood, S.B. Bioconjugate Chem. 1996, 7, 45.
[19] Dasgupta, F.; Masada, R.I. Carbohydr. Res. 2002, 337, 1055.
[20] Hori. H.; Νishida, Y.; Ohrui, H.; Meguro, H. J. Org. Chem. 1989, 54, 1346.
[21 ] Prepared analogously to ref. 18. [22] Liptak, A.; Imre, J.; Nanasi, P. Carbohydr. Res. 1981, 92, 154. [23] Agoston, K.; Kerekgyartό, J.; Hajkό, J.; Batta, G.; Lefeber, D J.; Kamerling, J.P.; Nliegenthart, J.F. Chem. Eur. J. 2002, 8, 151. [24] Cochran, S.; Li, C; Fairweather, J.K.; Kett, W.C; Coombe, D.R.; Ferro, N. J. Med. Chem. 2003, 46, 4601.
[25] Nyberg, K.; Ekblad, M.; Bergstrδm, T.; Freeman, C; Parish, C.R.; Ferro, N.; Trybala, E. Antiviral Res. 2004, 63, 15. [26] Gunalp, A. Proc. Soc. Exp. Biol. Med. 1965, 118, 185.
[27] Holland, T.C.; Homa, F.L.; Marlin, S.D.; Levine, M.; Glorioso, J. J. Virol. 1984, 52, 566.
[28] Duff, R.; Rapp, F. Nat. New Biol. 1971, 233, 48.

Claims

A compound of the formula
Figure imgf000052_0001
wherein: n is an integer of from 0 to 2;
Z is N, N(O), O, S, S(O), S(O)2, P, P(O), P(O)2, Si, Si(O), or Si(O)2; each X is independently C, C(O), N, N(O), O, S, S(O), S(O)2, P, P(O), P(O)2, Si, Si(O),)2 or is a bond; and each of Ri to R6 is independently a bond or is selected from the group consisting of: hydrogen; halogen; straight chain, cyclic, branched, substituted, heterocyclic, heteroatom substituted or unsubstituted alkyl, alkenyl, alkynyl, aryl, or heteroaryl; phosphoryl groups such as phosphate, thiophosphate -O-P(S)(OH) ; phosphate esters -O-P(O)(OR)2; thiophosphate esters -O-P(S)(OR)2; phosphonate -O-P(O)OHR; thiophosphonate -O-P(S)OHR; substituted phosphonate -O-P(O)OR1R2; substituted thiophosphonate -O-P(S)OR1R2; -O-P(S)(OH)(SH); and cyclic phosphate; other phosphorus containing compounds such as phosphoramidite -O-P(OR)-NR1R2; and phosphoramidate -O-P(O)(OR)- RιRa; sulfur groups such as -O-S(O)(OH), -SH, -SR, -S(→O)-R, S(O)2R, RO-S(O)2\ -O-SO2NH2, -O-SO2R1R2 or sulfamide -NHSO2NH2; amino groups such as -NHR, -NRιR2, -NHAc, -NHCOR, -NH-O-COR, - NHSO3, -NHSO2R, -N(SO2R)2, and/or amidino groups such as -NH- C(=NH)NH2 and/or ureido groups such as -NH-CO-NRiR2 or thiouriedo groups such as -H-C(S)-NH2; another unit ofthe structure I, attached through any position, where Z, X and Ri to R6 are as defined above; or a substructure based upon a group ofthe following formula:
Figure imgf000053_0001
wherein: Y is a bond or is selected from the group consisting of: straight chain, cyclic, branched, substituted, heterocyclic, heteroatom substituted or unsubstituted alkyl; straight chain, cyclic, branched, substituted, heterocyclic, heteroatom substituted or unsubstituted acyl; and aryl, substituted aryl, heteroaryl; and each of R to Ri i is independently at least one structure according to formula I, or a structure according to formula II; with the provisos that: when Z is O, and X is O or a bond, then all of Ri to R5 are not H or CH2OH; or when Z is N and X is O or a bond, then all of Ri to R6 are not H.
2. A compound according to claim 1, wherein said compound is PG2024, PG2037, PG2046, PG2155, as hereinbefore described.
3. A compound according to claim 1, wherein said compound is any one ofthe compounds of Tables 1-4 ofthe description.
4. A pharmaceutical or veterinary composition for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis, and/or microbial infection, which composition comprises at least one compound according to claim 1 together with a pharmaceutically or veterinarially acceptable carrier or diluent for said at least one compound.
5. The composition according to claim 4 which further includes a pharmaceutically or veterinarially acceptable excipient, buffer, stabiliser, isotonicising agent, preservative or antioxidant.
6. The composition according to claim 4, wherein said compound is present therein as an ester, a free acid or base, a hydrate, or a prodrug.
7. Use of a compound according to claim 1 in the manufacture of a medicament for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis, and/or microbial infection.
8. The use according to claim 7, wherein said mammalian subject is a human subject.
9. A method for the prevention or treatment in a mammalian subject of a disorder resulting from angiogenesis, metastasis, inflammation, coagulation, thrombosis, and/or microbial infection, which method comprises administering to the subject an effective amount of at least one compound according to claim 1 , or a composition comprising said at least one compound.
10. The method according to claim 9 wherein said mammalian subject is a human subject.
11. The method according to claim 9, wherein said disorder resulting from angiogenesis is a proliferative retinopathy or angiogenesis resulting from the growth of a solid tumour.
12. The method according to claim 9, wherein said disorder resulting from inflammation is rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, allograft rejection or chronic asthma.
13. The method according to claim 9, wherein said disorder resulting from coagulation and/or thrombosis is deep venous thrombosis, pulmonary embolism, thrombotic stroke, peripheral arterial thrombosis, unstable angina or myocardial infarction.
14. The method according to claim 9, wherein said disorder resulting from viral infection is Herpes Simplex.
PCT/AU2004/001800 2003-12-23 2004-12-21 Glycosaminoglycan (gag) mimetics WO2005061523A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
CA002551181A CA2551181A1 (en) 2003-12-23 2004-12-21 Glycosaminoglycan (gag) mimetics
JP2006545842A JP2007515434A (en) 2003-12-23 2004-12-21 Glycosaminoglycan (GAG) mimics
AU2004303434A AU2004303434A1 (en) 2003-12-23 2004-12-21 Glycosaminoglycan (GAG) mimetics
EP04802102A EP1699806A1 (en) 2003-12-23 2004-12-21 Glycosaminoglycan (gag) mimetics
BRPI0417750-9A BRPI0417750A (en) 2003-12-23 2004-12-21 compound of the formula; pharmaceutical or veterinary composition; compound use, and method for prevention and treatment in the area of mimetic glycosaminoglycan (gag)
MXPA06007194A MXPA06007194A (en) 2003-12-23 2004-12-21 Glycosaminoglycan (gag) mimetics.
IL176474A IL176474A0 (en) 2003-12-23 2006-06-21 Glycosaminoglycan (gag) mimetics
NO20063366A NO20063366L (en) 2003-12-23 2006-07-20 Glycosamylcan (GAG) mimetics

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2003907107 2003-12-23
AU2003907107A AU2003907107A0 (en) 2003-12-23 Glycosaminoglycan (GAG) Mimetics

Publications (1)

Publication Number Publication Date
WO2005061523A1 true WO2005061523A1 (en) 2005-07-07

Family

ID=34705560

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2004/001800 WO2005061523A1 (en) 2003-12-23 2004-12-21 Glycosaminoglycan (gag) mimetics

Country Status (11)

Country Link
EP (1) EP1699806A1 (en)
JP (1) JP2007515434A (en)
KR (1) KR20060129296A (en)
CN (1) CN1906203A (en)
BR (1) BRPI0417750A (en)
CA (1) CA2551181A1 (en)
IL (1) IL176474A0 (en)
MX (1) MXPA06007194A (en)
NO (1) NO20063366L (en)
RU (1) RU2006126718A (en)
WO (1) WO2005061523A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010037179A1 (en) * 2008-10-03 2010-04-08 Glycan Biosciences Pty Ltd Anionic conjugates of glycosylated bacterial metabolite
US8143289B2 (en) 2008-11-13 2012-03-27 Merck Sharp & Dohme Corp. Aminotetrahydropyrans as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US8716482B2 (en) 2009-09-25 2014-05-06 Merck Sharp & Dohme Corp. Substituted aminopiperidines as dipeptidyl peptidase-IV inhibitors for the treatment of diabetes
US9045523B2 (en) 2006-11-14 2015-06-02 Pharos Pharmaceuticals Inc. Compounds
US9504702B2 (en) 2010-08-05 2016-11-29 Seattle Genetics, Inc. Methods of inhibition of protein fucosylation in vivo using fucose analogs
US9816069B2 (en) 2008-05-02 2017-11-14 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
WO2018068090A1 (en) * 2016-10-12 2018-04-19 Glycan Biosciences Llc Therapeutic agents and methods of producing same
WO2019113645A1 (en) * 2017-12-15 2019-06-20 The Australian National University Compounds for treating and preventing extracellular histone mediated pathologies
US10350228B2 (en) 2012-08-23 2019-07-16 Seattle Genetics, Inc. Treatment of sickle cell disease and inflammatory conditions
WO2020172698A1 (en) * 2019-02-25 2020-09-03 The Australian National University Compounds for treating and preventing net associated complications
US11401339B2 (en) 2018-08-23 2022-08-02 Seagen Inc. Anti-TIGIT antibodies
US11548908B2 (en) 2017-12-29 2023-01-10 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3
US11845771B2 (en) 2018-12-27 2023-12-19 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3
US11873317B2 (en) 2018-12-27 2024-01-16 Glycomimetics, Inc. Galectin-3 inhibiting c-glycosides

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100790007B1 (en) * 2006-12-28 2008-01-02 경상대학교산학협력단 A method for purification glycosaminoglycans from ascidian tunic
JP5777200B2 (en) * 2010-11-05 2015-09-09 学校法人常翔学園 Anti-dengue virus agent
CN102775450B (en) * 2012-07-03 2015-04-08 华东师范大学 Method for preparing 1,6-Anhydro-2-azido-2-deoxy-beta-D-glucopyranose
WO2015067594A1 (en) * 2013-11-05 2015-05-14 Nestec S.A. Use of glucosamine-enriched plant compositions
CN104914057B (en) * 2015-05-25 2018-06-19 上海中医药大学附属曙光医院 Simple and fast detects the kit and its detection method of glycosaminoglycan
CN115368332B (en) * 2022-08-22 2024-03-12 三峡大学 Synthesis method and application of 6-aryl/alkylthio-2H-pyran-3-ketone compound

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4459293A (en) * 1981-12-28 1984-07-10 American Cyanamid Company Method of modulating the complement system by administering bis-[β-D-glucopyranosyl-1-thio (or sulfinyl or sulfonyl)]-aryline sulfate derivatives and the cation salts thereof
WO1985000973A1 (en) * 1983-08-29 1985-03-14 American Cyanamid Company SUBSITUTED PHENYL-1-THIO(POLY-O-SULFO)-alpha(OR beta)-D-GLUCOPYRANOSIDES
WO1993024506A1 (en) * 1992-05-26 1993-12-09 Alberta Research Council IMMUNOSUPPRESSIVE AND TOLEROGENIC MODIFIED LEWISC AND LacNAc COMPOUNDS
WO1996003413A2 (en) * 1994-07-27 1996-02-08 Genzyme Limited Regioselective sulfation
WO1997018222A2 (en) * 1995-11-13 1997-05-22 Glycomed, Inc. Novel oligosaccharide glycosides having mammalian immunosuppressive and tolerogenic properties
US5700918A (en) * 1994-04-01 1997-12-23 Nippon Shinyaku Co., Ltd. Moranoline derivative
JPH11315092A (en) * 1998-03-05 1999-11-16 Agency Of Ind Science & Technol Sulfated galactose compound, its intermediate and production of sulfated galactose compound
WO1999065480A1 (en) * 1998-06-16 1999-12-23 Ono Pharmaceutical Co., Ltd. Glucopyranose derivatives and preventives and/or remedies for hiv infection containing the same as the active ingredient
JP2001048857A (en) * 1999-08-11 2001-02-20 Ono Pharmaceut Co Ltd Glucopyranose derivative
WO2003038054A2 (en) * 2001-10-31 2003-05-08 New York University Structure-based design and synthesis of fgf inhibitors and fgf modulator compounds

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4459293A (en) * 1981-12-28 1984-07-10 American Cyanamid Company Method of modulating the complement system by administering bis-[β-D-glucopyranosyl-1-thio (or sulfinyl or sulfonyl)]-aryline sulfate derivatives and the cation salts thereof
WO1985000973A1 (en) * 1983-08-29 1985-03-14 American Cyanamid Company SUBSITUTED PHENYL-1-THIO(POLY-O-SULFO)-alpha(OR beta)-D-GLUCOPYRANOSIDES
WO1993024506A1 (en) * 1992-05-26 1993-12-09 Alberta Research Council IMMUNOSUPPRESSIVE AND TOLEROGENIC MODIFIED LEWISC AND LacNAc COMPOUNDS
US5700918A (en) * 1994-04-01 1997-12-23 Nippon Shinyaku Co., Ltd. Moranoline derivative
WO1996003413A2 (en) * 1994-07-27 1996-02-08 Genzyme Limited Regioselective sulfation
WO1997018222A2 (en) * 1995-11-13 1997-05-22 Glycomed, Inc. Novel oligosaccharide glycosides having mammalian immunosuppressive and tolerogenic properties
JPH11315092A (en) * 1998-03-05 1999-11-16 Agency Of Ind Science & Technol Sulfated galactose compound, its intermediate and production of sulfated galactose compound
WO1999065480A1 (en) * 1998-06-16 1999-12-23 Ono Pharmaceutical Co., Ltd. Glucopyranose derivatives and preventives and/or remedies for hiv infection containing the same as the active ingredient
JP2001048857A (en) * 1999-08-11 2001-02-20 Ono Pharmaceut Co Ltd Glucopyranose derivative
WO2003038054A2 (en) * 2001-10-31 2003-05-08 New York University Structure-based design and synthesis of fgf inhibitors and fgf modulator compounds

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 14, no. 9, 2004, pages 2221 - 2226 *
BIOORGANIC & MEDICINAL CHEMISTRY, vol. 10, no. 8, 2002, pages 2445 - 2460 *
CARBOHYDRATE RESEARCH, vol. 327, no. 3, 2000, pages 223 - 260 *
CHEMICAL & PHARMACEUTICAL BULLETIN, vol. 47, no. 9, 1999, pages 1237 - 1245 *
DATABASE CA [online] BYTHEWAY I. ET AL: "Validation of Molecular Docking Calculation Involving FGF-1 and FGF-2", Database accession no. 140:314439 *
DATABASE CA [online] FAROUX-CORLAY B. ET AL: "Synthesis of single- and double-chain fluorocarbon and hydrocarbon galactosyl amphiphiles and their anti-HIV-1 activity", Database accession no. 133:267051 *
DATABASE CA [online] LIU L. ET AL: "Application of the four-component Ugi condensation for the preparation of sulfated glycoconjugate libraries", Database accession no. 141:54554 *
DATABASE CA [online] MULLIGAN M.S. ET AL: "In vitro and in vivo selectin-blocking activities of sulfated lipids and sulfated sialyl compounds", Database accession no. 129:107414 *
DATABASE CA [online] SAKAGAMI M. ET AL: "Synthesis and evaluation of biantennary oligosaccharide ligands mimicking Sialyl Lewis X", Database accession no. 131:322848 *
DATABASE CA [online] YOSHIIZUMI K. ET AL: "Studies on scavenger receptor inhibitors. Part 1: synthesis and structure-activity relationships of novel derivatives of sulfatides", Database accession no. 138:82903 *
DATABASE WPI Week 199612, Derwent World Patents Index; Class B03, AN 1996-116981 *
DATABASE WPI Week 200009, Derwent World Patents Index; Class B03, AN 2000-100762 *
DATABASE WPI Week 200010, Derwent World Patents Index; Class B03, AN 2000-116716 *
DATABASE WPI Week 200136, Derwent World Patents Index; Class B03, AN 2001-337999 *
INTERNATIONAL IMMUNOLOGY, vol. 10, no. 5, 1998, pages 569 - 575 *
JOURNAL OF MEDICINAL CHEMISTRY, vol. 47, no. 7, 2004, pages 1683 - 1693 *

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9045523B2 (en) 2006-11-14 2015-06-02 Pharos Pharmaceuticals Inc. Compounds
US9816069B2 (en) 2008-05-02 2017-11-14 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
US11319526B2 (en) 2008-05-02 2022-05-03 Seagen Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
US10443035B2 (en) 2008-05-02 2019-10-15 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
US9346845B2 (en) 2008-10-03 2016-05-24 Glycan Biosciences Llc Anionic conjugates of glycosylated bacterial metabolite
US8791245B2 (en) 2008-10-03 2014-07-29 Glycan Bioscience LLC Anionic conjugates of glycosylated bacterial metabolite
AU2009299118B2 (en) * 2008-10-03 2015-12-17 Glycan Biosciences Llc Anionic conjugates of glycosylated bacterial metabolite
WO2010037179A1 (en) * 2008-10-03 2010-04-08 Glycan Biosciences Pty Ltd Anionic conjugates of glycosylated bacterial metabolite
US8772328B2 (en) 2008-11-13 2014-07-08 Merck Sharp & Dohme Corp. Aminotetrahydropyrans as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US8951965B2 (en) 2008-11-13 2015-02-10 Merck Sharp & Dohme Corp. Aminotetrahydropyrans as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US9138426B2 (en) 2008-11-13 2015-09-22 Merck Sharp & Dohme Corp. Aminotetrahydropyrans as dipeptidyl peptidase-IV inhibitors for the treatment of diabetes
US9278976B2 (en) 2008-11-13 2016-03-08 Merck Sharp & Dohme Corp. Aminotetrahydropyrans as dipeptidyl peptidase-IV inhibitors for the treatment of diabetes
US8592371B2 (en) 2008-11-13 2013-11-26 Merck Sharpe & Dohme Corp. Aminotetrahydropyrans as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US9403790B2 (en) 2008-11-13 2016-08-02 Merck Sharp & Dohme Corp. Aminotetrahydropyrans as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US8415297B2 (en) 2008-11-13 2013-04-09 Merck Sharp & Dohme Corp. Aminotetrahydropyrans as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US8143289B2 (en) 2008-11-13 2012-03-27 Merck Sharp & Dohme Corp. Aminotetrahydropyrans as dipeptidyl peptidase-IV inhibitors for the treatment or prevention of diabetes
US8716482B2 (en) 2009-09-25 2014-05-06 Merck Sharp & Dohme Corp. Substituted aminopiperidines as dipeptidyl peptidase-IV inhibitors for the treatment of diabetes
US11033561B2 (en) 2010-08-05 2021-06-15 Seagen Inc. Methods of inhibition of protein fucosylation in vivo using fucose analogs
US10342811B2 (en) 2010-08-05 2019-07-09 Seattle Genetics, Inc. Methods of inhibition of protein fucosylation in vivo using fucose analogs
US9504702B2 (en) 2010-08-05 2016-11-29 Seattle Genetics, Inc. Methods of inhibition of protein fucosylation in vivo using fucose analogs
US10350228B2 (en) 2012-08-23 2019-07-16 Seattle Genetics, Inc. Treatment of sickle cell disease and inflammatory conditions
CN110573520B (en) * 2016-10-12 2024-02-09 格莱科新诺威逊私人有限公司 Therapeutic agent and method for producing the same
IL265971A (en) * 2016-10-12 2019-06-30 Glycan Biosciences Llc Therapeutic agents and methods of producing sa
CN110573520A (en) * 2016-10-12 2019-12-13 格莱科新诺威逊私人有限公司 Therapeutic agents and methods of making the same
US11903958B2 (en) 2016-10-12 2024-02-20 Glycosynnovations Pty Ltd Therapeutic agents and methods of producing same
WO2018068090A1 (en) * 2016-10-12 2018-04-19 Glycan Biosciences Llc Therapeutic agents and methods of producing same
WO2019113645A1 (en) * 2017-12-15 2019-06-20 The Australian National University Compounds for treating and preventing extracellular histone mediated pathologies
US11628179B2 (en) 2017-12-15 2023-04-18 The Australian National University Compounds for treating and preventing extracellular histone mediated pathologies
US11548908B2 (en) 2017-12-29 2023-01-10 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3
US11401339B2 (en) 2018-08-23 2022-08-02 Seagen Inc. Anti-TIGIT antibodies
US11845771B2 (en) 2018-12-27 2023-12-19 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3
US11873317B2 (en) 2018-12-27 2024-01-16 Glycomimetics, Inc. Galectin-3 inhibiting c-glycosides
WO2020172698A1 (en) * 2019-02-25 2020-09-03 The Australian National University Compounds for treating and preventing net associated complications

Also Published As

Publication number Publication date
CN1906203A (en) 2007-01-31
EP1699806A1 (en) 2006-09-13
CA2551181A1 (en) 2005-07-07
KR20060129296A (en) 2006-12-15
RU2006126718A (en) 2008-01-27
BRPI0417750A (en) 2007-04-10
MXPA06007194A (en) 2007-01-19
JP2007515434A (en) 2007-06-14
NO20063366L (en) 2006-07-20
IL176474A0 (en) 2006-10-05

Similar Documents

Publication Publication Date Title
WO2005061523A1 (en) Glycosaminoglycan (gag) mimetics
RU2392281C2 (en) Sulphated derivatives of oligosaccharides
EP2382226B1 (en) Process for the synthesis of l-fucosyl di- or oligosaccharides and novel 2,3,4 tribenzyl-fucosyl derivatives intermediates thereof
SK284881B6 (en) Synthetic polysaccharides, preparation method therefor and pharmaceutical compositions containing same
US10174132B2 (en) Heparan sulfate synthesis
Eklind et al. Large-Scale Synthesis of a Lewis b Tetrasaccharide Derivative, its Acrylamide Copolymer, and Related DI-and Trisaccharides for Use in Adhesion Inhibition Studies with Helicobacter Pylori.
US5220008A (en) Oligosaccharide inhibitors for influenza virus
TW202334175A (en) Novel oligosaccharide, manufacturing intermediate for novel oligosaccharide, and method for manufacturing these
US5254676A (en) Oligosaccharide inhibitors for influenza virus
AU2004303434A1 (en) Glycosaminoglycan (GAG) mimetics
Bindschädler et al. Synthesis of a potential 10E4 tetrasaccharide antigen involved in scrapie pathogenesis
WO2012121617A1 (en) Oligosaccharide compounds
US20010041677A1 (en) Compounds and their uses
Buijsman et al. Synthesis of a Pentasaccharide–Oligonucleotide Conjugate: A Novel Antithrombotic Agent
EP1409498B1 (en) Inositol phosphoglycan derivatives and their medical uses
US20110212907A1 (en) Hexadecasaccharides with antithrombotic activity, including a covalent bond and an amino chain
EP1152007A1 (en) Ether type lipid a1-carboxylic acid analogues
Wessel et al. The Synthesis of the 2′′-and 2′′′-Monodeoxygenated Analogues of β-Maltosyl-(1→ 4)-Trehalose
MXPA01008080A (en) Ether type lipid a1-carboxylic acid analogues
MXPA06013555A (en) Azasugar derivatives, heparanase inhibitors, method for preparing same, compositions containing same, use thereof
JP2004075670A (en) Carboxymethyl analog at glucosyl lipid a1 position
TW200413400A (en) Lipid a derivatives which have glucose back-bone as the reducing sugar part

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2004303434

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 3514/DELNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 176474

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 200605169

Country of ref document: ZA

Ref document number: PA/a/2006/007194

Country of ref document: MX

Ref document number: 2006545842

Country of ref document: JP

Ref document number: 2551181

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

ENP Entry into the national phase

Ref document number: 2004303434

Country of ref document: AU

Date of ref document: 20041221

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004303434

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004802102

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200480040751.0

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 1020067014850

Country of ref document: KR

Ref document number: 1020067014837

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1020067014837

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2006126718

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 2004802102

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020067014850

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 2004802102

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0417750

Country of ref document: BR