WO2005018632A1 - Treatment of neurodegenerative conditions - Google Patents

Treatment of neurodegenerative conditions Download PDF

Info

Publication number
WO2005018632A1
WO2005018632A1 PCT/GB2004/003524 GB2004003524W WO2005018632A1 WO 2005018632 A1 WO2005018632 A1 WO 2005018632A1 GB 2004003524 W GB2004003524 W GB 2004003524W WO 2005018632 A1 WO2005018632 A1 WO 2005018632A1
Authority
WO
WIPO (PCT)
Prior art keywords
acid
linolenic acid
lipid
dihomo
disease
Prior art date
Application number
PCT/GB2004/003524
Other languages
French (fr)
Inventor
Laurence S. Harbige
Michael J. Leach
Mohammed Sharief
Paul Barraclough
Original Assignee
Btg International 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 GB0319358A external-priority patent/GB0319358D0/en
Priority claimed from GB0410846A external-priority patent/GB0410846D0/en
Priority to CN200480030644XA priority Critical patent/CN1867328B/en
Priority to CH00249/06A priority patent/CH698539A8/en
Priority to EP04768085A priority patent/EP1660071A1/en
Priority to DE112004001520T priority patent/DE112004001520B4/en
Priority to CA2534202A priority patent/CA2534202C/en
Application filed by Btg International Limited filed Critical Btg International Limited
Priority to US10/567,778 priority patent/US7964641B2/en
Priority to AU2004266480A priority patent/AU2004266480B2/en
Priority to GB0603646A priority patent/GB2422373B/en
Priority to JP2006523678A priority patent/JP2007502805A/en
Publication of WO2005018632A1 publication Critical patent/WO2005018632A1/en
Priority to IS8246A priority patent/IS8246A/en
Priority to FI20060154A priority patent/FI20060154A/en
Priority to NO20061232A priority patent/NO20061232L/en
Priority to US12/654,779 priority patent/US20100113595A1/en
Priority to US12/654,780 priority patent/US20100113810A1/en
Priority to US13/363,705 priority patent/US20120142775A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/62Halogen-containing esters
    • C07C69/65Halogen-containing esters of unsaturated acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/23Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms
    • A61K31/232Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms having three or more double bonds, e.g. etretinate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C67/00Preparation of carboxylic acid esters
    • C07C67/14Preparation of carboxylic acid esters from carboxylic acid halides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C67/00Preparation of carboxylic acid esters
    • C07C67/28Preparation of carboxylic acid esters by modifying the hydroxylic moiety of the ester, such modification not being an introduction of an ester group
    • C07C67/29Preparation of carboxylic acid esters by modifying the hydroxylic moiety of the ester, such modification not being an introduction of an ester group by introduction of oxygen-containing functional groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/52Esters of acyclic unsaturated carboxylic acids having the esterified carboxyl group bound to an acyclic carbon atom
    • C07C69/587Monocarboxylic acid esters having at least two carbon-to-carbon double bonds

Definitions

  • the present invention relates to a method for treating neurodegenerative conditions, particularly those in which increase in transforming growth factor ⁇ (TGF- ⁇ ) is beneficial, particularly TGF- ⁇ l. More particularly the present invention provides treatment for neurodegenerative conditions, particularly those such as demyelinating diseases, such as multiple sclerosis, Alzheimer's and Parkinsons diseases and the degenerative sequelae associated with head trauma, stroke and intracranial bleeds, whereby neuronal function may be improved or restored from an impaired condition, eg. by remyeleination.
  • TGF- ⁇ transforming growth factor ⁇
  • the present invention provides treatment for neurodegenerative conditions, particularly those such as demyelinating diseases, such as multiple sclerosis, Alzheimer's and Parkinsons diseases and the degenerative sequelae associated with head trauma, stroke and intracranial bleeds, whereby neuronal function may be improved or restored from an impaired condition, eg. by remyeleination.
  • TGF- ⁇ l and PGE 2 production has been shown to be increased non-specifically in ⁇ -linolenic acid fed mice ex vivo.
  • TGF- ⁇ 1 has been reported to protect in acute and relapsing EAE ((Racke et al (1993); Santambrogio et al (1993)), and PG inhibitors such as indomethacin augment, and thus worsen, the disease (Ovadia & Paterson (1982)).
  • Cytokines are implicated in the pathogenesis of MS, with many studies showing an increase in myelinotoxic inflammatory cytokines (TNF- ⁇ , IL-1 ⁇ and LFN- ⁇ ) coinciding with the relapse phase of the disease.
  • TGF- ⁇ l cytokine transforming growth factor-betal
  • TGF- ⁇ l is expressed in the CNS and, in oral-tolerance-induced protection in EAE, TGF- ⁇ and PGE are expressed in the brain (Karpus & Swanborg (1991); Khoury et al (1992)). Harbige ((1998) concluded that dietary ⁇ -linolenic acid effects on EAE are mediated through Th 3 -like mechanisms involving TGF- ⁇ l and possibly through superoxide dismutase antioxidant activity.
  • T cell depleters and modulators such as cyclophosphamide
  • T-cells indeed produce beneficial cytokines, such as TGF- ⁇ l, as well as deleterious ones in man.
  • David Baker of Institute of Neurology UK summed up the disparity between what is effective in the EAE and in MS with a paper entitled 'Everything stops EAE, nothing stops MS' at the 10 th May 2004 UK MS Frontiers meeting of the UK MS Society. It is clear that immunosuppression alone cannot cure MS.
  • MS patients in addition to the autoimmune disease, that leads to membrane abnormality, cytokine dysregulation and subsequent immune attack and lesioning.
  • the 'gold standard' treatment for MS remains interferon, such as with ⁇ - Avonex ®, Rebif ® and other interferon preparations.
  • This gold standard treatment only addresses needs of some, eg 30%, of the patients and even in these symptom improvement is restricted to reduced severity of relapses. Whilst symptoms may be reduced in a proportion of patients, the disease tends to progress to further disability and death due to underlying degeneration.
  • PBMC peripheral blood mononuclear cell production
  • pro-inflammatory cytokines TNF- ⁇ and IL-l ⁇ were significantly and markedly ( ⁇ 70%) reduced and they either maintained or increased the PBMC membrane long chain omega-6 fatty acids dihomo- ⁇ -linolenic acid (DHLA) and arachidonic acid (AA) in contrast to patients taking placebo who demonstrated loss of these fatty acids over the course of the trial period.
  • DHLA dihomo- ⁇ -linolenic acid
  • AA arachidonic acid
  • MOG myelin oligodendrocyte glycoprotein
  • blackcurrant seed oil 72 % w/w 18:3n-6, GLA
  • blackcurrant seed oil has a low sn-2 GLA with most of the GLA in the sn-1 and sn-3 positions (Lawson and Hughes 1988).
  • TG-GLA structured triacylgcerol containing three GLA moieties
  • the present inventors now set out, in view of their results for high sn-2- ⁇ - linolenic acid Borage Oil, to demonstrate that it is indeed the presence of an sn-2- ⁇ - linolenic acid, dihomo- ⁇ -linolenic acid or arachidonic acid residue in a glyceride, particularly a triglyceride, that gives it efficacy in treating EAE, CREAE and the human disease MS.
  • the present invention provides a method of treating a patient in need of therapy for a neurodegenerative disease comprising administering to that patient a therapeutically effective dose of a defined structure lipid glyceride comprising a glycerol moiety esterifed with one or more fatty acid moieties, characterised in that the lipid has a fatty acid moiety at the sn-2 position selected from the group of residues consisting of residues of ⁇ -linolenic acid, dihomo- ⁇ -linolenic acid and arachidonic acid.
  • Particularly advantageously treated neurodegenerative diseases are those involving demyelination.
  • the present method specifically arrests underlying neurodegeneration and restores neuronal function.
  • the method normalises neuronal membrane composition, and restores healthy PBMC spontaneuosly released TGF- ⁇ l/TNF ⁇ ratios and the ratios of TGF- ⁇ l with other PBMC released cytokines.
  • the method arrests neurodegeneration in multiple sclerosis of all types but particularly relapsing remitting, primary progressive and chronic progressive MS and the restoration, in part or completely, of neuronal function such as measured, eg. By MRI or CAT scan or by EDSS score.
  • Such method may also be used in treatment of cerebral impairment after stroke, head trauma and intracranial bleeding where there is demyelination or neuronal damage.
  • the lipid is administered for a duration and at a dose sufficient to maintain or elevate TGF- ⁇ levels in the patient to therapeutic levels.
  • therapeutic levels is meant levels at least consistent with healthy subjects.
  • the dose is such as to produce a TGF- ⁇ 1/TNF- ⁇ ratio spontaneously released from peripheral blood mononuclear cells (PBMCs) isolated from blood of a patient, after 18 months of daily dosing, of 0.4 to 3.0, at least 0.5, more preferably at least 0.75 and most preferably at least 1.
  • PBMCs peripheral blood mononuclear cells
  • the dose is such as to produce a TGF- ⁇ 1/IL-l ⁇ ratio in blood of a patient, after 18 months of daily dosing, of at least 0.5, more preferably at least 0.75 and most preferably at least 1.
  • said levels are produced after 12 months and more preferably after 6 months.
  • the amount of lipid administered daily will be between 0.5 and 30 grams, orally dosed, still more preferably between 1 and 20 grams and most preferably between 1 and 18 grams, typically 3 to 5 grams.
  • the sn-2 moiety is that of a ⁇ -linolenic acid residue
  • the dose may be toward the higher end of these ranges, particuarly where the sn-1 and sn-3 moieties are relatively inert, eg.
  • the lipid is a monoglyceride, diglyceride or triglyceride containing the at least one sn-2 ⁇ -linolenic acid, dihomo- ⁇ -linolenic acid or arachidonic acid moiety of general Formula I below:
  • R and R are independently selected from hydrogen and acyl groups, and R 2 is selected from the group consisting of ⁇ -linolenic acid, dihomo- ⁇ - linolenic acid and arachidonic acid residues.
  • acyl groups are defined as comprising at least one carbonyl group on the end of an optionally substituted hydrocarbyl chain selected from alkyl and alkenyl chains, the carbonyl group being directly attached by its carbon to the oxygen of the glycerol residue shown in Formula 1.
  • Preferred acyl groups R 1 and R 3 are saturated fatty acid moities of formula - CO-(CH 2 ) n -CH 3 , wherein n is an integer selected from 1 to 22, more preferably being 4 to 16, still more preferably being from 5 to 12, most preferably being from 6 to 10.
  • Particularly preferred acyl groups are those of caprylic and capric acids, particularly being 1,3-dicaprylic or 1,3-dicapric glycerols having the ⁇ -linolenic acid, dihomo- ⁇ - linolenic acid or arachidonic acid moiety at the sn-2 position.
  • Preferred glycerides for use in the invention are triglycerides.
  • US 4701469 describes some potential triglycerides for nutraceutical use that the present inventors have determined may be used in the method of the invention, although it only specifically describes 1,3-dioctanyl triglycerides wherein the sn-2 acid is of an EFA, only 1,3-dioctanoyl eicosapenta glycerol is described as having been prepared. These are said to useful in inter alia immunomodulation, but although a number of diseases are specified, use in immunosuppresion in neurodegeneration and MS are not listed.
  • R 1 to R 3 for inclusion in the compound of formula I are simple saturated fatty acids or naturally occurring fatty acids with structural or metabolic function, such as medium chain or long chain fatty acids, there are other possibilities. Particularly preferred fatty acids are those that are utilised primarily by the metabolism for producing energy. Where fatty acids are structural, that is utilised in membranes, they are conveniently such as ⁇ -linolenic acid, linoleic acid, dihomo- ⁇ -linolenic acid and arachidonic acid residues. By residue is meant the moiety that remains after the fatty acid carboxyl group esterifies to one of the hydroxy groups of the glycerol molecule.
  • sn-1 and sn-3 are selected from fatty acids that are metabolised in the human to yield energy as opposed to a fatty acid that is primarily directed to the structural membrane pool: such preferred acids include oleic acid and palmitic acid.
  • sn-1 and sn-3 fatty acid chain (R and R ) is unsaturated it may also be that of other essential fatty acids, such as the n-3 acids such as stearidonic acid, eicosapentanoic acid and docosahyexanoic acid.
  • the fatty acid is optionally substituted these will preferably be by hydroxy, oxo, carboxyl, alkyl, alkenyl and alkoxy groups.
  • the hydrocarbyl chain is preferably one of from 1 to 30 carbon atoms in length, more preferably from 4 to 28 carbon atoms in length, still more preferably 4 to 24 carbon atoms in length. Most preferably the hydrocarbyl chain is that of a fatty acid, more particularly a mono or polyunsaturated fatty acid.
  • a fatty acid more particularly a mono or polyunsaturated fatty acid.
  • Many of the preferred lipids for use in the method of the invention are known and may be prepared by chemical process known in the art. For example, many are commercially available, such as trigamma-linolenin, known as TLG, but herein referred to as GGG, reflecting the identity of groups R R R where G represents ⁇ - linolenic acid residues. GGG is commercially available from Nu-Check-Prep Inc.
  • EP 0300844 describes its synthesis using a base-catalysed trans-esterification of triacetin with methyl gamma linolenate to give a mixture containing 80% GGG, unreacted methyl ⁇ - linolenate and 10% mono- and di-glycerides.
  • Triarachidin is known and small quantities can be obtained commercially eg. from Sigma AAA has been synthesised from arachidonic acid by using immobilised lipase patented for angiogenisis-enhancing activity US 4888324.
  • the present inventors prefer the use of the mono- ⁇ -linolenic acid, dihomo- ⁇ -linolenic acid or arachidonic acid sn-2 ester triglycerides because they administer less of the immunomodulatory and proinflammatory fatty acids ⁇ -linolenic acid, dihomo- ⁇ -linolenic acid or arachidonic acid whilst retaining the enhanced activity that the sn-2 ⁇ -linolenic acid, dihomo- ⁇ - linolenic acid or arachidomc acid moiety provides with regard to the desired membrane normalising and disease modifying effect.
  • Novel lipids which are preferred are accessible by processes and methods set out in the Examples herein. Most preferred lipids are those where there is just a single ⁇ -linolenic acid, dihomo- ⁇ -linolenic acid or arachidonic acid moiety esterified to the glycerol at sn-2, with the flanking sn-1 and sn-3 acids being unsaturated medium chain or long chain acids.
  • novel lipids disclosed herein including compounds of formula II
  • R 1 and R 3 are the same and are -C(O)(CH 2 ) n CH 3 wherein n is selected from 4 to 14, more preferably 6 to 10 and most preferably 7, 8 or 9 and R 2 is selected from ⁇ -linolenyl, dihomo- ⁇ -linolenyl and arachidonyl.
  • a further aspect of the present invention provides a method for synthesis of a compound of general formula III
  • R 1 and R 3 are the same and are -C(O)(CH 2 ) n CH 3 wherein n is selected from 4 to 14, more preferably 6 to 10 and most preferably 7, 8 or 9 and R 2 is ⁇ - linolenyl residue, dihomo- ⁇ -linolenyl residue or arachidonyl residue comprising reacting 1,3-dihydroxyacetone with a compound of formula X-C(O)(CH 2 ) n CH 3 wherein X is selected from Cl, Br and I, to give the corresponding l,3-di-( C(O)(CH 2 ) n CH 3 ) 2-keto compound reducing the keto group to the corresponding l,3-di-( C(O)(CH 2 ) n CH 3 ) 2-ol and reacting that with ⁇ -linolenyl chloride or dihomo- ⁇ -linolenyl chloride or arachidonyl chloride.
  • n is selected from 4 to
  • R 1 to R 3 are the same and selected from ⁇ -linolenyl residue, dihomo- ⁇ -linolenyl residue or arachidonyl residue comprising reacting the corresponding ⁇ -linolenyl chloride, dihomo- ⁇ - linolenyl chloride or arachidonyl chloride with glycerol.
  • Synthesis of some of these compounds is described below and schemes shown in the figures below. For example, a single-step esterification of glycerol using GLA and a coupling agent, such as DCCI/DMAP (1.1-Dicylcohexylcarbodiimide/ 4- dimethylaminopyridine) coupling reagents may be carried out.
  • DCCI/DMAP 1.1-Dicylcohexylcarbodiimide/ 4- dimethylaminopyridine
  • GLA-C1 prepared from ⁇ -linolenic acid and oxalyl chloride
  • glycerol in dichloromethane/pyridine with good yields at scale-up to 250 g purified by column chromatography.
  • Yukiki Watanebe, Yoichi, Takagi, Yoshiaki (Nisshin Oil Mills Ltd, Japan) describes a related but different technique for purification of tri- ⁇ -linolenin
  • the applicant further provides an improved process which allows glycerol to react with more than 3 equivalents of decanoyl chloride and purified the tricaprin product by recrystallisation.
  • Further aspects of the present invention provide use of triglyceride oils as described above for the manufacture of a medicament for the treatment of neurodegenerative diseases as set out for the method of the invention.
  • Particularly preferred medicaments are for the arresting and reversing of neurodegeneration in multiple sclerosis of all types but particularly relapsing remitting, primary progressive and chronic progressive and the restoration, in part or completely, of neuronal integrity function such as measured, eg. By MRI or CAT scan or by EDSS score.
  • TGF- ⁇ l responsive diseases may be treated as set out previously.
  • the lipids for use in the present invention may be administered by any of the conventional vehicles known in pharmacy. Most conveniently they are administered as neat oils or in admixture with foodstuffs, in the form of capsules containing such oils, or in enterically coated forms. Other forms will occur to those skilled in the art but Remington Pharmaceutical Sciences 19 Edition. It will be realised by those skilled in the art that other beneficial agents may be combined with the lipids for use in the present invention or otherwise form part of a treatment regime with the lipids. These might be ion channel blockers, eg. sodium chamiel blockers, interferons ( ⁇ , ⁇ , or ⁇ ), T-cell depleters, steroids or other palliative agents.
  • ion channel blockers eg. sodium chamiel blockers, interferons ( ⁇ , ⁇ , or ⁇ )
  • T-cell depleters steroids or other palliative agents.
  • Table 1 Shows the compositional % Total fatty acid content of various triglyceride oils and protective effect in EAE.
  • Table 2 Shows the parameters of the three treatment groups in high sn-2 GLA Borage Oil trial described in PCT/GB04/002089.
  • Table 3 Shows the effect of various forms of GLA on EAE incidence and clinical score in SJL mice: lower score indicating improved therapeutic effect.
  • Table 4 Shows the failure of enriched Blackcurrent oil, a high GLA, but low sn-2- GLA, plant oil, to match fungal and Borage oils in EAE.
  • Figure 1 Shows spontaneous peripheral blood mononuclear cell cytokine production in placebo and high sn-2 ⁇ -linolenic acid, PCT/GB04/002089 trial oil treated human MS patients at 18 months.
  • Figure 2 Shows the effect of placebo and low dose (5g/day) high sn-2 GLA Borage oil on human MS patient EDSS score as compared to high dose (15g/day) displayed as a histogram with months treatment on the x axis.
  • Figure 3 Shows the effect of placebo, low dose and high dose high sn-2 GLA Borage oil on human MS patient Mean Relapse rate (%) as histogram with months on x axis.
  • Figure 4 Shows the reaction scheme for synthesis of a single fatty acid triacylglyceride for use in the method and use of this invention.
  • Figure 5 Shows the reaction scheme for synthesis of control compound tricaprin.
  • Figure 6 Shows the reaction scheme for synthesis of CGC, a mixed fatty acid triacylglyceride of the invention.
  • Figure 7 Shows the reaction scheme for synthesis of C-DHGLA-C, a mixed fatty acid triacylglyceride of the invention.
  • Figure 8 Shows the reaction scheme for synthesis of control compound GCG, 1,3- dicapryl, 2- ⁇ -linolenic acid.
  • Figure 9 Shows the reaction scheme for synthesis of C-AA-C, a mixed fatty acid traiacylglyceride of the invention.
  • High sn-2 Borage oil (PCT/GB04/002089) trial. Twenty-eight active relapsing-remitting (two relapses in the preceding 18 months) multiple sclerosis patients (ages ranging from 18 to 65 yrs) were entered into a double-blind placebo controlled trial to investigate the effects of encapsulated borage oil on clinical activity and laboratory parameters over 18 months.
  • This oil was of high sn-2 ⁇ -linolenic (GLA) content (>40% of sn-2 residues being ⁇ -linolenic acid ) with low monene (eg. erusic acid) content and had no added Vitamin E, a known immunomodulator.
  • GLA high sn-2 ⁇ -linolenic
  • Exclusion criteria include any form of steroid or immunosuppressive drug treatment, pregnancy, hyperlipidemia, regular use of aspirin or related drugs and vitamin or fatty acid supplementation within the previous three months.
  • Results are shown in Tables 1 and 2 and Figures 1 to 5.
  • the primary outcome parameter was the number of clinical relapses between baseline (Month 0) and the end of treatment (Month 18).
  • Secondary outcome parameters included: the time to first clinical relapse; severity of relapses, as assessed by EDSS score and the use of steroid treatment; and changes in EDSS at Month 3, 6, 9, 12, and 18 compared to baseline and defined as at least 1.0 point increase in the EDSS that is sustained for 3 months or at least 1.5 point increase on the EDSS from the baseline EDSS that is sustained for 3 months.
  • Eleven patients were in the placebo group seven patients had been taking low- dose Borage oil, and ten patients had been taking high-dose Borage oil.
  • the study drug was well-tolerated, and there were no serious adverse events during the 18- month trial.
  • TNF- ⁇ , IL-l ⁇ and IFN- ⁇ in cell culture supernatants and plasma were detected using commercially available paired antibodies enabling cytokine detection in an ELISA format (R&D systems Ltd, Abingdon, UK).
  • the sensitivities for the TNF- ⁇ and IFN- ⁇ ELISAs were 15.6-lOOOpg/ml and 3.9-250 ⁇ g/ml for IL-l ⁇ .
  • Patient 1 The first patient was a 48 year old woman who had had a clinically active, relapsing remitting MS for 9 years. She had originally worked as a full-time administrator at the local Health Authority, but she was unable to perform her duties because of her severe MS. Therefore, she later worked as a part-time secretary, but still had difficulties in mobilization because of muscles stiffness and sensory disturbances. She was also experiencing severe clinical relapses at an average of one relapse every nine months. Most of these relapses had resulted in hospital admissions for steroid therapy. In view of her active MS, she was recruited into the Borage oil trial.
  • Patient 2 The second case was a 46-year old woman who also had a clinically active relapsing remitting MS for 8 years. She had originally worked as a shop assistant, but became unemployed after MS was diagnosed. Her symptoms included difficulty with mobilisation and painful sensory symptoms in both legs. She had experienced three clinical relapses in the two years preceding the clinical trial, and had been admitted to hospital twice for steroid therapy. Consequently, she was recruited into the Borage oil trial, but her walking continued to deteriorate. Six months into the trial, she need to use a walking stick and also received treatment with Baclofen to reduce low limb spasticity. Approximately ten months after starting the Borage oil trial, she was admitted to hospital because of severe clinical relapse, which was treated with steroids.
  • the DCM was dried over magnesium sulphate, filtered and concentrated in vacuo to a brown oil ( ⁇ 21 g).
  • the oil was purified on a silica column using 5% ether in hexane at first and then 10%. 15.6g (77% yield) of a clear oil was obtained. By tic this material contained a small amount of free GLA. (This material was repurified at a later date)
  • glycerol tri-6,9,12-linolenate (gamma linolenic acid triglyceride, trigammalinolenin, GGG) was prepared from 96.2 % GLA by a two-step acid chloride route. It is a clear, pale yellow oil and was stored under nitrogen in the freezer. The GLA content was 97.1 % and no C20:l, C22:l, or C24:l acids were detected). The HPLC purity was 93.6 %.
  • the precipitated pyridine hydrochloride was removed by filtration and washed with dichloromethane. The combined washing and filtrate was then washed with aqueous solutions (20 ml) of 5% sodium chloride, 5% sodium bicarbonate, 0.1N hydrochloric acid, and 5% sodium chloride. The dichloromethane layer was then dried over MgSO 4 and the solvent removed in vacuo. The residual oil crystallised on standing. This material was recrystallised from isopropanol (40 ml) to give 15.6 g (86% yield) of a waxy white solid.
  • the precipitated pyridine hydrochloride was removed by filtration and washed with dichloromethane. The combined washing and filtrate was then washed with aqueous solutions (300 ml) of 5% sodium chloride, 5% sodium bicarbonate, 0. IN hydrochloric acid, and 5% sodium chloride. The dichloromethane layer was then dried over MgSO 4 and the solvent removed in vacuo. The residual oil crystallised on standing. This material was recrystalhsed from isopropanol (400 ml) to give 228 g (86% yield) of a waxy white solid.
  • glycerol tridecanoate (tricaprin, CCC) was been prepared from decanoyl chloride (98 %) by a one-step process (scheme given below). It is a white, low-melting solid and was stored under nitrogen in the freezer. The C content was 99.9 % of fatty acid content and the HPLC purity was 97.9 %.
  • CGC was prepared by reaction of 1,3-Dicaprin with GLA-chloride in dichloromethane-pyridine.
  • 1,3-Dicaprin was prepared by sodium borohydride reduction of l,3-didecanoyloxypropan-2-one, which was in turn prepared by reaction of decanoyl chloride with 1,3-dihydroxyacetone.
  • the intermediate 1,3-dicaprin must be handled with care since it can undergo acyl migration on exposure to acids , bases and heat.
  • the residual oily acid chloride (GLA-Cl) was then added dropwise over 15 min (ice /water cooling) to a stirred solution of 1,3-dicaprin (11.2g, 0.028 mol), DCM (50 ml), pyridine (2.42 ml, 2.37 g, 0.03 mol) and 4- dimethylaminopyridine (0.10 g, 0.0008 mol, 0.03 equiv) at 10-15°C .
  • the temperature was maintained by ice-water cooling.
  • the reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM.
  • 1,3-Dicaprin The above ketone (158 g, 0.40 mol) was dissolved in tetrahydrofuran (THF, 2.25 L). Water (50 ml) was then added, the solution cooled to 5°C, and sodium borohydride (5.66 g, 1.5eq) added portionwise below 10°C. The reaction mixture was monitored by HPLC (C18, eluted with ACN at lml min ⁇ 210nm) (Note: only about 4.5g of the borohydride was in fact added, as all SM had reacted). The reaction mixture was stirred at RT for lh and then concentrated in vacuo to remove THF.
  • THF tetrahydrofuran
  • CGC 1,3-Dicaprin 2-gammalinolenoate
  • GLA95 1,3-Dicaprin 2-gammalinolenoate
  • DCM dichloromethane
  • oxalyl chloride 55.7 ml, 82.3 g, 0.65 mol, 1.5eq
  • the mixture was stirred at RT overnight and then concentrated in vacuo to remove DCM and excess oxalyl chloride.
  • the residual oily acid chloride (GLA-Cl) was then added dropwise over 30-40 min at 10-15 °C (ice /water cooling) to a stirred solution of 1,3-dicaprin (164.7g, 0.41 mol), DCM (650 ml), pyridine (33.3 ml, 32.5 g, 0.41 mol) and 4-dimethylaminopyridine (1.50 g, 0.012 mol, 0.03 equiv) at 10-15°C .
  • the reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM.
  • DHLA (3.93g, 12.8 mmol, 1 eq) was dissolved in dichloromethane (DCM, 20 ml) and stirred at room temperature under a nitrogen atmosphere.
  • Oxalyl chloride (1.69 ml, 2.46 g, 19.4 mmol, 1.5 eq) was added dropwise over 1-2 min, and left stirring at room temperature overnight. The resulting solution was concentrated in vacuo to remove DCM and excess oxalyl chloride.
  • DHLA-C1 The residual oily acid chloride (DHLA-C1) was then added dropwise over 5 min at 25°C to a stirred mixture of 1,3-dicaprin (4.91 g, 12.2 mmol, 0.95 eq), pyridine (0.98 ml, 0.96 g 12.1 mmol, 0.95 eq) and 4-dimethylam inopyridine (DMAP, 8 mg, 0.07 mmol, 0.03 eq).
  • DMAP 4-dimethylam inopyridine
  • the residual oily acid chloride (A-Cl) was then added dropwise over 15 min to a pre- warmed (25 °C) stirred mixture of glycerol (5.11 g, 0.055 mol, 1 eq), pyridine (13.5 ml, 13.2 g, 0.17 mol, 3 eq) and 4-dimethylamino pyridine (DMAP, 0.20 g, 0.002 mol, 0.03 eq).
  • DMAP 4-dimethylamino pyridine
  • the temperature of the reaction mixture rose to 42 °C during the addition and a gentle reflux was observed.
  • the mixture was stirred at 30-40 °C and monitored by HPLC. After 2 h, no further product formation was observed.
  • the precipitated pyridine hydrochloride was filtered off and washed with DCM.
  • a second batch (17.8 g) was produced from 39.8 g of arachidonic acid, The two batches were combined and residual solvents removed under vacuo to give 40.5 g (43%) of a mobile pale yellow oil. HPLC purity 84.8% GLC analysis 94.3% AA (arachidonic acid).
  • the residual oily acid chloride (GLA-Cl) was then added dropwise over 20 min at 25°C to a stirred mixture of 1,3-dihydroxyacetone dimer (28.99 g, 0.32 mol, 1.0 equiv), pyridine (52 ml, 50.9 g 0.64 mol, 2.0 equiv), 4-dimethylaminopyridine (2.36 g, 0.02 mol, 0.06 equiv) and dichloromethane (DCM, 600 ml) at room temperature under nitrogen. The temperature of the reaction mixture was allowed to rise to 40°C and the mixture was stirred for a further 2 h under nitrogen (monitored by HPLC).
  • 1,3-dihydroxyacetone dimer 28.99 g, 0.32 mol, 1.0 equiv
  • pyridine 52 ml, 50.9 g 0.64 mol, 2.0 equiv
  • 4-dimethylaminopyridine (2.36 g, 0.02 mol, 0.06 equiv
  • 1,3-Di- ⁇ -linolenin 2-decanoate (Glycerol l,3-dioctadeca-(6Z,9ZJ2Z -trienoate 2- decanoate or GCG)
  • Decanoyl chloride (13.5 ml, 12.4 g, 0.065 mol, 1.1 eq) was added to a stirred solution of 1,3-di- ⁇ -linolenin (36.1 g, 0.059 mol, leq), dry pyridine (5.7 ml, 5.6 g, 0.07 mol, l.leq), 4-dimethylaminopyridine (0.2 g, 0.002 mol, 0.03 eq) and dichloromethane (DCM, 150 ml) over ca.
  • DCM dichloromethane
  • 1,3-Dicaprin 2-arachidonate Glycerol 1,3-didecanoate 2-eicosatetra-(5-Z,8-Z,ll- ZJ4-Z)enoate or CAP
  • This triglyceride is known.
  • CAC has been identified as a constituent of lymph lipids following administration of safflower oil to rats.
  • WO 03 013,497 describing an arachidonic acid containing triglyceride (produced by culturing Mortierella alpina) useful for diseases caused by brain hypofunction, but specifically for cognition enhancement.
  • the two intermediates used in the synthesis of CAC are known.
  • the synthesis of CAC from 1,3-dicaprin, and the purification of this are all novel.
  • CAC was prepared by reaction of 1 ,3-Dicaprin with arachidonyl chloride in dichloromethane-pyridine.
  • 1,3-Dicaprin was prepared by sodium borohydride reduction of l,3-didecanoyloxypropan-2-one, which was in turn prepared by reaction of decanoyl chloride with 1,3-dihydroxyacetone.
  • the intermediate 1,3-dicaprin must be handled with care since it can undergo acyl migration on exposure to acids , bases and heat.
  • An older method 6 of making 1,3-dicaprin, by catalysed addition of decanoic acid to a glycidol ester (from epichlorohydrin) was deemed less attractive because of more severe reaction conditions and acyl migration problems.
  • the final product, CAC was purified by careful column chromatography on silica which removed byproducts.
  • the residual oily acid chloride (GLA-Cl) was then added dropwise over 15 min (ice /water cooling) to a stirred solution of 1,3-dicaprin (11.2g, 0.028 mol), DCM (50 ml), pyridine (2.42 ml, 2.37 g, 0.03 mol) and 4- dimethylammopyridine (0.10 g, 0.0008 mol, 0.03 equiv) at 10-15°C .
  • the temperature was maintained by ice-water cooling.
  • the reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM.
  • the pyridine hydrochloride formed was removed by filtration and washed with DCM. The combined filtrate and washings were then washed with 1 x 150ml portions of 5%NaCl, 5%NaHCO 3 , 0.1N HCl, 5%NaCl. The solution was then dried over MgSO 4 and concentrated in vacuo to a yellowish semi-solid. This was then crystallised from methanol (500ml) to give a white solid. The yield was 158 g (60%). 1,3-Dicaprin The above ketone (158 g, 0.40 mol) was dissolved in tetrahydrofuran (THF, 2.25 L).
  • the residual oily acid chloride (GLA-Cl) was then added dropwise over 30-40 min at 10-15°C (ice /water cooling) to a stirred solution of 1,3-dicaprin (94.2 g, 0.24 mol), DCM (450 ml), pyridine (19.1 ml, 18.6 g, 0.24 mol) and 4-dimethylaminopyridine (1.72 1.50 g, 0.014 mol, 0.06 equiv) at 10-15°C .
  • the reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM.
  • glycerol l,3-didecanoate-2-arachidonate (l,3-dicaprin-2-AA, CAC) has been prepared from decanoyl chloride (98 %) and Arachidonic acid (95%) by a three-step process (scheme given below). It is a very pale yellow oil and was stored under nitrogen in the freezer. The HPLC purity is 95.8 %.
  • OGO was here prepared by reaction of 1,3-Doleoin with GLA-chloride in dichloromethane-pyridine.
  • 1,3-Diolein was prepared by sodium borohydride reduction of l,3-dioleoylpropan-2-one, which was in turn prepared by reaction of oleoyl chloride with 1,3-dihydroxyacetone.
  • the intermediate 1,3-dioleolin must be handled with care since it can undergo acyl migration on exposure to acids , bases and heat.
  • Older methods 7 ' 8 of making 1,3-dioleoin, via mono-tritylglycerols or glycidyl esters was deemed less attractive because of more steps and acyl migration problems.
  • the final product, OGO was purified by careful column chromatography on silica which removed by-products.
  • 1,3-Diolein 2-gammalinolenoate (O-G-O) ⁇ -Linolenic acid (GLA95, 41.2 g, 0.15 mol, 1.1 equiv) was dissolved in dichloromethane (DCM, 250 ml). The resulting solution was stirred at RT under nitrogen and oxalyl chloride (19.1 ml, 28.2 g, 0.22 mol, 1.65 equiv) added dropwise over 5 mins. The mixture was stirred at RT overnight and then concentrated in vacuo to remove DCM and excess oxalyl chloride.
  • DCM dichloromethane
  • the residual oily acid chloride (GLA-Cl) was then added dropwise over 15 min (ice /water cooling) to a stirred solution of 1,3- diolein (83.5g, 0.13 mol), DCM (250 ml), pyridine (10.9 ml, 10.6 g, 0.14 mol) and 4- dimethylaminopyridine (0.49 g, 0.004 mol, 0.15 equiv) at 10-15°C .
  • the temperature was maintained by ice-water cooling.
  • the reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM.
  • glycerol l,3-oleoate-2-gammalinolenoate (l,3-dioleate-2-GLA, OGO) was prepared from oleoyl chloride (98 %) by a three-step process (scheme given below). It was an almost colourless oil (slight yellow tinge) and is being stored under nitrogen in the freezer. The HPLC purity was 89.4 %.
  • the proton-decoupled 13 C NMR spectra with suppressed NOE were collected at 21 °C in a 5-mm broadband probe on a Joel 500 MHz spectrometer operating at 125.728 MHz.
  • Waltz decoupling was the chosen mode of decoupling and was gated on only during the 14.89s acquisition time.
  • the relaxation delay was set at 30 sees and the pulse angle was 90°.
  • the spectral window used was ca.35 ppm (from 173.5 to 172.6 ppm) with a 170 ppm offset.
  • the spectra were internally referenced to CDC1 at 77.0 ppm.
  • the approximate number of scans collected for adequate signal-to- noise ranged from 300 to 1200 scans depending on the concentration and purity of the sample.
  • the total acquisition time for the experiments ranged between 2-8h e.g
  • CREAE Chronic Relapsing Experimental Autoimmune Encephalomyelitis
  • CREAE was induced in C57B1/6 and SJL mice.
  • Animals were injected subcutaneously with 100 ⁇ g of the neuroantigen peptide MOG 35-55 (amino acid sequence MEVGWYRSPFSRVVHLYRNGK Genemed Synthesis, hie) or 1 mg of mouse spinal cord homogenate (SCH), in phosphate buffered saline (PBS), emulsified by sonication for 10 min at room temperature, in incomplete Freund's adjuvant (DIFCO, Detroit, USA) supplemented with 480 ⁇ g of mycobacteria tuberculosis and 60 ⁇ g of Mycobacteria butyricium (DIFCO, Detroit, USA) on days 0 and 7 as described previously (Morris-Downes, MM., et al 2002).
  • PBS phosphate buffered saline
  • DIFCO incomplete Freund's adjuvant
  • mycobacteria tuberculosis 60 ⁇ g of Myco
  • MOG EAE in C57BL mice Treatment was from Days 5 to 21 inclusive except C-DHLA-C group where treatment was from days 5 to 15 inclusive. Animals were culled on PSD 25. [Five animals from an untreated group, 3 animals from control CCC treatment group, 5 animals from GGG 150ul treatment group and 2 animals from GGG 350ul treatment group were sampled for histological analysis on PSD 20].
  • Treatment was from Days 6 to 20 inclusive.
  • C-A-C are all capable of reducing severity of CREAE whereas compounds G-C-G and C-C-C failed to treat the condition.
  • Compound O-G-O is believed to work if the dose is adjusted.
  • the arachidnoic acid compounds are effective, but lead to death of some animals. Surviving animals had much reduced disease. It is believed that the dose of these compounds may be reduced still further to provide survival with satisfactory treatment.
  • Some of the studies show a bell shaped response curve for compounds C-G-C and G-G-G, suggesting that very high doses are not optimal, as set out above. Such dosing can be conveniently determined by those skilled art, eg.
  • Genain CP. Cannella B., hauser SL and Raine CS. Identification of autoantibodies associated with myelin damage in multiple sclerosis. Nature Med 1999:5,170-175.
  • Transforming growth factor beta I reduces infarct size after experimental cerebral ischemia in a rabbit model .Stroke 24,558-562.
  • TGF-beta I protects hippocampal neurons against degeneration caused by transient global ischaemia. Dose-response relationship and potential neuroprotective mechanisms. Stroke, 27, 1609-1614.
  • Hollifield RD Harbige LS, PhM-Dinh D, Sharief M.
  • Evidence for cytokine Dysregulation in Multiple Sclerosis Peripheral Blood Mononuclear cell production of pro-inflammmatory and anti-inflammatory cytokines during relapse and remission. Autoimmunity, 2003 36(3):133-141.
  • Schiefer HB Hancock DS, Loew FM. Long-term effects of partially hydrogenated herring oil on the rat myocardium. Drug Nutr Interact. 1982;1(2):89-102.
  • Schluesener HJ Lider O. Transforming growth factors beta 1 and beta 2: cytokines with identical immunosuppressive effects and a potential role in the regulation of autoimmune T cell function. J Neuroimmunol 1989; 24:249-58.
  • FGO Fungal Oil
  • BOO Borage Oil
  • EPO Evening Primrose Oil
  • SAF Safflower Oil

Abstract

A method is provided for treating a patient in need of therapy for a neurodegenerative disease comprising administering to that patient a therapeutically effective dose of a lipid glyceride comprising a glycerol moiety and a fatty acid moiety, the fatty acid moiety being selected from the group consisting of Ϝ-linolenic acid, dihomo-Ϝ-linolenic acid and arachidonic acid characterised in that the selected fatty acid moiety is attached to the glycerol moiety at its sn-2 position. Preferably the method is that wherein the lipid is administered for a duration and at a dose sufficient to maintain or elevate TGF-β1 levels in the patient to therapeutic levels.

Description

TREATMENT OF NEURODEGENERATIVE CONDITIONS. The present invention relates to a method for treating neurodegenerative conditions, particularly those in which increase in transforming growth factor β (TGF- β) is beneficial, particularly TGF-βl. More particularly the present invention provides treatment for neurodegenerative conditions, particularly those such as demyelinating diseases, such as multiple sclerosis, Alzheimer's and Parkinsons diseases and the degenerative sequelae associated with head trauma, stroke and intracranial bleeds, whereby neuronal function may be improved or restored from an impaired condition, eg. by remyeleination. Further provided are novel use of known and novel compounds comprising unsaturated fatty acid moieties for the manufacture of medicaments capable of effectively treating such conditions, more particularly being capable of achieving previously unattained levels of success with regard to recovery of neurological function. The inventor's copending unpublished patent application PCT/GB04/002089, incorporated herein by reference, relates to the use of plant and fungal oils for the treatment of neurodegenerative diseases. These oils have high percentages of the essential fatty acid γ-linolenic acid (GLA) at the sn-2 position of their lipids, typically being over 40% of the sn-2 fatty acid total of the oil. It is well reported in the literature that essential fatty acids (EFAs) of the n-3 and n-6 unsaturation pattern have beneficial effect in a wide variety of human physiological disorders, including autoimmune diasese (WO 02/02105). Harbige (1998) Proc. Nut. Soc. 57, 555-562 reviewed the supplementation of diet with n-3 and n-6 acids in autoimmune disease states, and particularly noted evidence of benefit of γ-linolenic (GLA) and/or linoleic acid (LA) rich oils. Bates et al noted that lipid oils comprising a mixture of linoleic acid and γ- linolenic acid residues had been suggested back in 1957 to be possibly more efficacious in treating inflammation and autoimmune diseases, but found that at 3g oil per day (Naudicelle Evening Primrose oil 7:1 LA:GLA) patients who had relapses became more ill on the trial oil than on the control. Although the aetiology of MS remains unknown studies have shown that MS patients have higher than normal neuro-antigen autoreactive T-cells levels. These T- cells react inter alia to myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein (MOG) and are in an increased state of activation compared with healthy controls. The actual processes of axonal damage e.g. chronic inflammation, demyelination and astrogliosis in MS is complex but white matter inflammation and demyelination are considered to deteπnine disease severity, whilst recent studies suggested that axonal damage in MS begins in the early stages of the disease and contributes to disability (De Stefano et al, 2001). Experimental autoimmune encephalomyelitis (EAE) is the most frequently used animal model for immune mediated effects of MS. Studies in the guinea-pig have shown that linoleic acid partially suppresses the incidence and severity of EAE (Meade et al (1978)). (Harbige et al (1995), 1997b) demonstrated disease modifying effects of linoleic acid and γ-linolenic acid on clinical and histopathological manifestations of EAE. Depending on dose, γ-linolenic acid was fully protective in acute rat EAE whereas linoleic acid had dose-dependent action on the clinical severity but did not abolish it. Despite these experimental findings, it is recognised that the human disease, multiple sclerosis, is highly complex and can be conversely exacerbated and ameliorated by the activity of T-cells and other immune response factors. It is thought that the n-6 fatty acids promote autoimmune and inflammatory disease based upon results obtained with linoleic acid only. TGF-βl and PGE2 production has been shown to be increased non-specifically in γ-linolenic acid fed mice ex vivo. TGF-β 1 has been reported to protect in acute and relapsing EAE ((Racke et al (1993); Santambrogio et al (1993)), and PG inhibitors such as indomethacin augment, and thus worsen, the disease (Ovadia & Paterson (1982)). Cytokines are implicated in the pathogenesis of MS, with many studies showing an increase in myelinotoxic inflammatory cytokines (TNF-α, IL-1 β and LFN- γ) coinciding with the relapse phase of the disease. Conversely, levels of the anti- inflammatory and immunosuppressive cytokine transforming growth factor-betal (TGF-βl) appear to be reduced during a phase of relapse and increase as the patient enters remission. Thus the balance between biologically active TGF-βl and the pro- inflammatory TNF-α, IL-lβ and IFN-γ appears to be dysregulated during MS relapse- remission. During natural recovery phase from EAE, TGF-βl -secreting T-cells inhibit
EAE effector cells, TGF-βl is expressed in the CNS and, in oral-tolerance-induced protection in EAE, TGF-β and PGE are expressed in the brain (Karpus & Swanborg (1991); Khoury et al (1992)). Harbige ((1998) concluded that dietary γ-linolenic acid effects on EAE are mediated through Th3-like mechanisms involving TGF-βl and possibly through superoxide dismutase antioxidant activity. Borage oil (typically 20% to 23% γ-linolenic acid and 34 to 40% linoleic acid per 100% fatty acid content) and Mucor javanicus fungal oil (see Figure 1) have been shown to be effective in the EAE animal model used to identify MS candidates, whilst never having been shown to be significantly effective in the human disease. High levels of linoleic rich oil containing low levels of γ-linolenic acid (EPO: linoleic acid:γ-linolenic acid 7:1) partially suppressed the incidence and severity of EAE in rat (Mertin & Stackpoole, 1978) whereas the Bates' Naudicelle study referred to above led to worsening of patients. In spite of the use of Borage oil and other GLA/LA containing oils such as Evening Primrose oil by multiple sclerosis sufferers over the past 30 years or so, the vast majority of patients fail to recover from the disease, showing no significant improvement, with the underlying disease continuing to progress to death. It has been suggested to use, inter alia, γ-linolenic acid and linoleic acid rich Borage oil as a means to provide immuno-suppression in multiple sclerosis (US 4,058,594). Critially, the dose suggested is 2.4 grams of oil per day and no actual evidence of efficacy is provided. This is much lower than the low 5g/day dose found to be ineffective in vivo in man in the PCT/GB04/002089 study. Other more dramatic immunosuppressant treatments, including T cell depleters and modulators such as cyclophosphamide, are also shown to be effective in the EAE model, but where these are employed in the human multiple sclerosis disease symptoms improve, but the underlying disease continues to progress. T-cells indeed produce beneficial cytokines, such as TGF-βl, as well as deleterious ones in man. David Baker of Institute of Neurology, UK summed up the disparity between what is effective in the EAE and in MS with a paper entitled 'Everything stops EAE, nothing stops MS' at the 10th May 2004 UK MS Frontiers meeting of the UK MS Society. It is clear that immunosuppression alone cannot cure MS. This is almost certainly due to a fundamental underlying metabolic disorder in MS patients, in addition to the autoimmune disease, that leads to membrane abnormality, cytokine dysregulation and subsequent immune attack and lesioning. Although patients go into remission in relapse-remitting disease, the underlying demyelination proceeds. The 'gold standard' treatment for MS remains interferon, such as with β- Avonex ®, Rebif ® and other interferon preparations. This gold standard treatment only addresses needs of some, eg 30%, of the patients and even in these symptom improvement is restricted to reduced severity of relapses. Whilst symptoms may be reduced in a proportion of patients, the disease tends to progress to further disability and death due to underlying degeneration. In their as yet unpublished PCT/GB04/002089 study the present inventors have surprisingly determined that with compliance to a 'high dose' treatment with triglyceride oil containing high levels of sn-2 γ-linolenic acid (>40% of residues at the sn-2 being of γ-linolenic acid) with suitable accompanying fatty acid content, remarkable levels of improvement in almost all symptoms of MS can be achieved, way surpassing that provided by the current gold standard treatment. Such success is particularly surprising in the light of the prior use of other γ-linolenic acid containing preparations without success, such as the Naudicelle study. The PCT/GB04/002089 study shows that over an 18-month period, patients taking high dose (15g/day) selected high sn-2 γ-linolenic acid borage oil showed significant (ρ<0.001) and marked improvements in EDSS score, a reduced rate of relapse, symptomatic relief of muscle spasticity and painful sensory symptoms, and improved objective measures of cognitive functions. Low doses of 5g/day of this borage oil were without effect. Patients taking the highest dose of this borage oil maintained their level of peripheral blood mononuclear cell production (PBMC) of TGF-βl during the trial period, their pro-inflammatory cytokines TNF-α and IL-lβ were significantly and markedly (<70%) reduced and they either maintained or increased the PBMC membrane long chain omega-6 fatty acids dihomo-γ-linolenic acid (DHLA) and arachidonic acid (AA) in contrast to patients taking placebo who demonstrated loss of these fatty acids over the course of the trial period. This whilst immuno-suppression would be expected to reduce increase of active lesioning and neurodegeneration, the high sn-2 GLA oil treatment apparently targeted maintenance and/or increase of key membrane lipid components that are otherwise specifically lost in MS, being consistent with a correction of a metabolic defect not otherwise effectively treated by current therapies. The fact that the low dose (5 grams/day) had no effect on this supports such determination. γ-Linolenic acid (18:3n-6, or GLA) is known to be rapidly converted to longer-chain omega-6 polyunsaturated fatty acids dihomo-γ-linolenic acid and arachidonic acid in vivo (Phylactos et al 1994, Harbige et al 1995, 2000). Therefore to determine how to increase the level of membrane long chain omega-6 fatty acids in MS the inventors have reviewed their results obtained with several GLA-containing oils:- both fungal (from Mucor javanicus) and plant (BoraRQ officianalis), Evening primrose Oenothera spp. or Blackcurrant Ribes spp) as well as a synthetic tri-GLA oil as GLA delivery systems in an in vivo experimental animal model of MS known as chronic relapsing experimental autoimmune encephalomyelitis (CREAE). Induction of EAE in rats does not produce histological features of demyelination (Brosnan et al 1988) but induces an acute mono-phasic disease pattern, unlike MS which is characterised by CNS demyelination and is in the majority of cases clinically relapsing-remitting. Chronic relapsing and demyelinating EAE models (CREAE) however are characterised by demyelination and relapse phases. With the demonstration that myelin oligodendrocyte glycoprotein (MOG) is an important neuroantigenic target in MS (Genain et al 1999) and the demonstration of far greater responses of peripheral blood auto-reactive lymphocytes to this neuroantigen, compared with MBP, in MS (Kerlero de Rosbo et al 1993, 1997) MOG induced CREAE has become the animal model of choice with features closely resembling those observed in MS ( Fazakerely et al 1997, Genain et al 1999, Amor et al 1994). Evidence from the inventor's CREAE and rat EAE feeding studies indicates that an enriched blackcurrant seed oil (72 % w/w 18:3n-6, GLA) did not protect against EAE (see Table 3). Importantly blackcurrant seed oil has a low sn-2 GLA with most of the GLA in the sn-1 and sn-3 positions (Lawson and Hughes 1988). Furthermore a structured triacylgcerol containing three GLA moieties (TG-GLA) provided protective effects similar to that of the borage oil used in CREAE (Table 2). This would also be consistent with the sn-2 GLA being important i.e. the outer pair sn-1 and sn-3 GLA being enzymatically removed in vivo and probably undergoing oxidation leaving the sn-2 GLA only. This selective hydrolysis arises from the known ability of specific lipases to remove the sn-1 and sn-3 fatty acids from triacylgycerol molecules but an apparent protection of the sn-2 position in vivo (Lawson and Hughes 1988, Kyle 1990). This review has led the inventors to postulate that glycerides having sn-2-γ- linolenic acid, dihomo-γ-linolenic acid or arachidonic acid residues will be superior in correcting MS metabolism even to the high sn-2-γ-linolenic acid Borage oil of their earlier trial. This would allow lower doses of lipid to be taken and/or possibly decrease the time of treatment which would result in beneficial effect. Table 3 of EP 0520624 (Efamol Holdings) compares the triglyceride content of Evening Primrose and Borage Oils, the former being taught to be more therapeutically effective than the latter for a variety of GLA responsive disorders. This document indicates Borage oil to have twenty seven different trigyceride components, only 20% of which have sn-2 GLA. Page 3, lines 40-42 notes that biological testing has shown that equal amounts of GLA may indeed have very different effects when that GLA is supplied as different oil sources. Crucially, it then directs the reader to one particular fraction present in Evening Primrose Oil (EPO), but not Borage Oil, as being responsible for the former's superior effect in raising PGE1 (see EP 0520624 Chart page 4 and Table 2) and thus anti-inflammatory effect: that fraction being identified as di-linoeoyl-mono-ga ma-linolenyl-glycerol (DLMG) which it states to be 18 to 19% of the total triglyceride in EPO. Crictically, page 6 clearly teaches that the position of the GLA, in sn-1, 2 or 3, is not important to this effect. Dines et al (1994) Proceedings of the Physiological Society, Aberdeen Meeting 14-16 September 1994 report on studies of treatment of diabetic neuropathy neuronal damage with γ-linolenic acid containing oils of the type advocated by EP 0520624 and again note that Borage Oil was not very effective in treating this neurodegeneration whereas Evening primrose oil was. The paper concludes that Borage Oil contains other constituents that interfere with GLA activity. The present inventors now set out, in view of their results for high sn-2-γ- linolenic acid Borage Oil, to demonstrate that it is indeed the presence of an sn-2-γ- linolenic acid, dihomo-γ-linolenic acid or arachidonic acid residue in a glyceride, particularly a triglyceride, that gives it efficacy in treating EAE, CREAE and the human disease MS. In a first aspect the present invention provides a method of treating a patient in need of therapy for a neurodegenerative disease comprising administering to that patient a therapeutically effective dose of a defined structure lipid glyceride comprising a glycerol moiety esterifed with one or more fatty acid moieties, characterised in that the lipid has a fatty acid moiety at the sn-2 position selected from the group of residues consisting of residues of γ-linolenic acid, dihomo-γ-linolenic acid and arachidonic acid. Particularly advantageously treated neurodegenerative diseases are those involving demyelination. The present method specifically arrests underlying neurodegeneration and restores neuronal function. Particularly the method normalises neuronal membrane composition, and restores healthy PBMC spontaneuosly released TGF-βl/TNFα ratios and the ratios of TGF-βl with other PBMC released cytokines. Most advantageously the method arrests neurodegeneration in multiple sclerosis of all types but particularly relapsing remitting, primary progressive and chronic progressive MS and the restoration, in part or completely, of neuronal function such as measured, eg. By MRI or CAT scan or by EDSS score. Such method may also be used in treatment of cerebral impairment after stroke, head trauma and intracranial bleeding where there is demyelination or neuronal damage. Further application is provided in treating other chronic demyelination such as in Alzheimer's and Parkinson's disease. Preferably the the lipid is administered for a duration and at a dose sufficient to maintain or elevate TGF-β levels in the patient to therapeutic levels. By therapeutic levels is meant levels at least consistent with healthy subjects. Preferably the dose is such as to produce a TGF-β 1/TNF-α ratio spontaneously released from peripheral blood mononuclear cells (PBMCs) isolated from blood of a patient, after 18 months of daily dosing, of 0.4 to 3.0, at least 0.5, more preferably at least 0.75 and most preferably at least 1. Preferably the dose is such as to produce a TGF-β 1/IL-lβ ratio in blood of a patient, after 18 months of daily dosing, of at least 0.5, more preferably at least 0.75 and most preferably at least 1. Preferably said levels are produced after 12 months and more preferably after 6 months. Typically the amount of lipid administered daily will be between 0.5 and 30 grams, orally dosed, still more preferably between 1 and 20 grams and most preferably between 1 and 18 grams, typically 3 to 5 grams. Where the sn-2 moiety is that of a γ-linolenic acid residue, the dose may be toward the higher end of these ranges, particuarly where the sn-1 and sn-3 moieties are relatively inert, eg. being metabolically utilised acids such as saturated fatty acids. Where the sn-2 moiety is that of a dihomo-γ-linolenic acid residue, the dose may be less, whilst where the sn-2 moiety is that of an aracidonic acid residue, efficacy is higher, but dosing should be more cautious, due to possibilities of unwanted side effects at higher levels. More preferably the method is characterised in that the lipid is a monoglyceride, diglyceride or triglyceride containing the at least one sn-2 γ-linolenic acid, dihomo-γ-linolenic acid or arachidonic acid moiety of general Formula I below:
Figure imgf000011_0001
Formula I wherein R and R are independently selected from hydrogen and acyl groups, and R2 is selected from the group consisting of γ-linolenic acid, dihomo-γ- linolenic acid and arachidonic acid residues. For the purpose of the present invention acyl groups are defined as comprising at least one carbonyl group on the end of an optionally substituted hydrocarbyl chain selected from alkyl and alkenyl chains, the carbonyl group being directly attached by its carbon to the oxygen of the glycerol residue shown in Formula 1. Preferred acyl groups R1 and R3 are saturated fatty acid moities of formula - CO-(CH2)n-CH3, wherein n is an integer selected from 1 to 22, more preferably being 4 to 16, still more preferably being from 5 to 12, most preferably being from 6 to 10. Particularly preferred acyl groups are those of caprylic and capric acids, particularly being 1,3-dicaprylic or 1,3-dicapric glycerols having the γ-linolenic acid, dihomo-γ- linolenic acid or arachidonic acid moiety at the sn-2 position.. Preferred glycerides for use in the invention are triglycerides. US 4701469 describes some potential triglycerides for nutraceutical use that the present inventors have determined may be used in the method of the invention, although it only specifically describes 1,3-dioctanyl triglycerides wherein the sn-2 acid is of an EFA, only 1,3-dioctanoyl eicosapenta glycerol is described as having been prepared. These are said to useful in inter alia immunomodulation, but although a number of diseases are specified, use in immunosuppresion in neurodegeneration and MS are not listed. Whilst most preferred groups R1 to R3 for inclusion in the compound of formula I are simple saturated fatty acids or naturally occurring fatty acids with structural or metabolic function, such as medium chain or long chain fatty acids, there are other possibilities. Particularly preferred fatty acids are those that are utilised primarily by the metabolism for producing energy. Where fatty acids are structural, that is utilised in membranes, they are conveniently such as γ-linolenic acid, linoleic acid, dihomo-γ-linolenic acid and arachidonic acid residues. By residue is meant the moiety that remains after the fatty acid carboxyl group esterifies to one of the hydroxy groups of the glycerol molecule. Other preferred acids for sn-1 and sn-3 are selected from fatty acids that are metabolised in the human to yield energy as opposed to a fatty acid that is primarily directed to the structural membrane pool: such preferred acids include oleic acid and palmitic acid. Where the sn-1 and sn-3 fatty acid chain (R and R ) is unsaturated it may also be that of other essential fatty acids, such as the n-3 acids such as stearidonic acid, eicosapentanoic acid and docosahyexanoic acid. Where the fatty acid is optionally substituted these will preferably be by hydroxy, oxo, carboxyl, alkyl, alkenyl and alkoxy groups. The hydrocarbyl chain is preferably one of from 1 to 30 carbon atoms in length, more preferably from 4 to 28 carbon atoms in length, still more preferably 4 to 24 carbon atoms in length. Most preferably the hydrocarbyl chain is that of a fatty acid, more particularly a mono or polyunsaturated fatty acid. Many of the preferred lipids for use in the method of the invention are known and may be prepared by chemical process known in the art. For example, many are commercially available, such as trigamma-linolenin, known as TLG, but herein referred to as GGG, reflecting the identity of groups R R R where G represents γ- linolenic acid residues. GGG is commercially available from Nu-Check-Prep Inc. EP 0300844 describes its synthesis using a base-catalysed trans-esterification of triacetin with methyl gamma linolenate to give a mixture containing 80% GGG, unreacted methyl γ- linolenate and 10% mono- and di-glycerides. Triarachidin is known and small quantities can be obtained commercially eg. from Sigma AAA has been synthesised from arachidonic acid by using immobilised lipase patented for angiogenisis-enhancing activity US 4888324. However, whilst the tri and di-γ-linolenic acid, dihomo-γ-linolenic acid or arachidonic acid di or triglycerides may be used, the present inventors prefer the use of the mono-γ-linolenic acid, dihomo-γ-linolenic acid or arachidonic acid sn-2 ester triglycerides because they administer less of the immunomodulatory and proinflammatory fatty acids γ-linolenic acid, dihomo-γ-linolenic acid or arachidonic acid whilst retaining the enhanced activity that the sn-2 γ-linolenic acid, dihomo-γ- linolenic acid or arachidomc acid moiety provides with regard to the desired membrane normalising and disease modifying effect. Novel lipids which are preferred are accessible by processes and methods set out in the Examples herein. Most preferred lipids are those where there is just a single γ-linolenic acid, dihomo-γ-linolenic acid or arachidonic acid moiety esterified to the glycerol at sn-2, with the flanking sn-1 and sn-3 acids being unsaturated medium chain or long chain acids. Thus a further aspect of the present invention provides novel lipids disclosed herein including compounds of formula II
Figure imgf000013_0001
wherein R1 and R3 are the same and are -C(O)(CH2)nCH3 wherein n is selected from 4 to 14, more preferably 6 to 10 and most preferably 7, 8 or 9 and R2 is selected from γ-linolenyl, dihomo-γ-linolenyl and arachidonyl. A further aspect of the present invention provides a method for synthesis of a compound of general formula III
Figure imgf000014_0001
wherein R1 and R3 are the same and are -C(O)(CH2)nCH3 wherein n is selected from 4 to 14, more preferably 6 to 10 and most preferably 7, 8 or 9 and R2 is γ- linolenyl residue, dihomo-γ-linolenyl residue or arachidonyl residue comprising reacting 1,3-dihydroxyacetone with a compound of formula X-C(O)(CH2)nCH3 wherein X is selected from Cl, Br and I, to give the corresponding l,3-di-( C(O)(CH2)nCH3) 2-keto compound reducing the keto group to the corresponding l,3-di-( C(O)(CH2)nCH3) 2-ol and reacting that with γ-linolenyl chloride or dihomo-γ-linolenyl chloride or arachidonyl chloride. A still further aspect of the present invention provides a method for synthesis of a compound of general formula IV
Figure imgf000014_0002
wherein R1 to R3 are the same and selected from γ-linolenyl residue, dihomo- γ-linolenyl residue or arachidonyl residue comprising reacting the corresponding γ-linolenyl chloride, dihomo-γ- linolenyl chloride or arachidonyl chloride with glycerol. Synthesis of some of these compounds is described below and schemes shown in the figures below. For example, a single-step esterification of glycerol using GLA and a coupling agent, such as DCCI/DMAP (1.1-Dicylcohexylcarbodiimide/ 4- dimethylaminopyridine) coupling reagents may be carried out. This method gives a good yield but generates impurities that, unless removed, make the final oil cloudy. This may be circumvented by using a coupling agent such as EDCI (l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride) which gives rise to water- soluble by-products that are easier to remove. Jpn. Kokai Tokkyo Koho JP 05310638 A2 22Nov 1993 Heisei, 6pp. describes the preparation of tri-α-linolenin (LnLnLn where Ln is linoleic acid) using DCCI, and analogous but different reaction. A alternative approach provides a two-step sequence that utilises reaction of
GLA-C1 (prepared from γ-linolenic acid and oxalyl chloride) and glycerol in dichloromethane/pyridine with good yields at scale-up to 250 g purified by column chromatography. Jpn. Kokai Tokkyo Koho JP 04328199 A2 17 Nov 1992 Heisei,
5pp. (Japan) Concentration of a-linolenic acid triglyceride by flash chromatography.
Ando, Yukiki, Watanebe, Yoichi, Takagi, Yoshiaki (Nisshin Oil Mills Ltd, Japan) describes a related but different technique for purification of tri-α-linolenin
(LnLnLn). Comparative example tricaprin (glycerol tridecanate) is a known compound commercially available from Sigma. It has been prepared by reaction of methyl decanoate and sodium glyceroxide with subsequent purification of the crude product by column chromatography (see E. S. Lutton and A. J. Fehl, Lipids, 5, 90-99 (1970)) An alternative method involves the acid-catalysed reaction of glycerol with decanoic acid followed by four crystallisations (see L. H. Jenson and A. J. Mabis,
Ada Ctγst., 21, 770 (1966)). The applicant further provides an improved process which allows glycerol to react with more than 3 equivalents of decanoyl chloride and purified the tricaprin product by recrystallisation. Further aspects of the present invention provide use of triglyceride oils as described above for the manufacture of a medicament for the treatment of neurodegenerative diseases as set out for the method of the invention. Particularly preferred medicaments are for the arresting and reversing of neurodegeneration in multiple sclerosis of all types but particularly relapsing remitting, primary progressive and chronic progressive and the restoration, in part or completely, of neuronal integrity function such as measured, eg. By MRI or CAT scan or by EDSS score. Other TGF-βl responsive diseases may be treated as set out previously. The lipids for use in the present invention may be administered by any of the conventional vehicles known in pharmacy. Most conveniently they are administered as neat oils or in admixture with foodstuffs, in the form of capsules containing such oils, or in enterically coated forms. Other forms will occur to those skilled in the art but Remington Pharmaceutical Sciences 19 Edition. It will be realised by those skilled in the art that other beneficial agents may be combined with the lipids for use in the present invention or otherwise form part of a treatment regime with the lipids. These might be ion channel blockers, eg. sodium chamiel blockers, interferons (α, β, or γ), T-cell depleters, steroids or other palliative agents. It will further be realsied that where the immune and inflammatory responses are being modulated, such combinations will need to be made carefully, given the complex nature of these systems. However, given the delayed response to the present oils, shorter acting agents might be beneficial in the first months of treatment before the TGF-βl levels are normalised, as long as the additional treatment does not impede this normalization process. The synthesis of structured lipids for use in the present invention is described below together with synthesis of comparative examples. Some of these lipids are novel while others are known but have not been used for the treatment of the invention. The present invention will now be described by way of Example only by reference to the following non-limiting Tables, Examples and Figures. Further embodiments falling within the scope of the invention will occur to those skilled in the art in the light of these.
TABLES Table 1 : Shows the compositional % Total fatty acid content of various triglyceride oils and protective effect in EAE.
Table 2: Shows the parameters of the three treatment groups in high sn-2 GLA Borage Oil trial described in PCT/GB04/002089.
Table 3: Shows the effect of various forms of GLA on EAE incidence and clinical score in SJL mice: lower score indicating improved therapeutic effect.
Table 4: Shows the failure of enriched Blackcurrent oil, a high GLA, but low sn-2- GLA, plant oil, to match fungal and Borage oils in EAE.
FIGURES
Figure 1 : Shows spontaneous peripheral blood mononuclear cell cytokine production in placebo and high sn-2 γ-linolenic acid, PCT/GB04/002089 trial oil treated human MS patients at 18 months.
Figure 2: Shows the effect of placebo and low dose (5g/day) high sn-2 GLA Borage oil on human MS patient EDSS score as compared to high dose (15g/day) displayed as a histogram with months treatment on the x axis.
Figure 3: Shows the effect of placebo, low dose and high dose high sn-2 GLA Borage oil on human MS patient Mean Relapse rate (%) as histogram with months on x axis.
Figure 4: Shows the reaction scheme for synthesis of a single fatty acid triacylglyceride for use in the method and use of this invention. Figure 5: Shows the reaction scheme for synthesis of control compound tricaprin.
Figure 6: Shows the reaction scheme for synthesis of CGC, a mixed fatty acid triacylglyceride of the invention.
Figure 7: Shows the reaction scheme for synthesis of C-DHGLA-C, a mixed fatty acid triacylglyceride of the invention.
Figure 8: Shows the reaction scheme for synthesis of control compound GCG, 1,3- dicapryl, 2-γ-linolenic acid.
Figure 9: Shows the reaction scheme for synthesis of C-AA-C, a mixed fatty acid traiacylglyceride of the invention.
Figure 10 to 19 show the results of EAE studies in SJL and C57BL mice as set out in the examples below. (DHLA=DHGLA: A=AA)
EXAMPLES
High sn-2 Borage oil (PCT/GB04/002089) trial. Twenty-eight active relapsing-remitting (two relapses in the preceding 18 months) multiple sclerosis patients (ages ranging from 18 to 65 yrs) were entered into a double-blind placebo controlled trial to investigate the effects of encapsulated borage oil on clinical activity and laboratory parameters over 18 months. This oil was of high sn-2 γ-linolenic (GLA) content (>40% of sn-2 residues being γ-linolenic acid ) with low monene (eg. erusic acid) content and had no added Vitamin E, a known immunomodulator. Patients were recruited from neurology out-patient clinics at two inner city hospitals; hospital informed consent was obtained on first (baseline) visit. Exclusion criteria include any form of steroid or immunosuppressive drug treatment, pregnancy, hyperlipidemia, regular use of aspirin or related drugs and vitamin or fatty acid supplementation within the previous three months. Only patients meeting all the following criteria were included in the trial: fa) able to provide informed consent prior to treatment, with the full understanding that consent may be withdrawn at any time without prejudice; (b) male or female outpatients aged 18 to 60 years inclusive; (c) have confirmed diagnosis of clinically definite relapsing MS; (d) have had at least three documented clinical relapses in the past two years; fe) have a baseline Expanded Disability Scoring Scale (EDSS) score of 0.0-5.5 inclusive, provided they have well documented exacerbations; and (f) healthy, apart from the MS-related symptoms, as confirmed by the medical history, physical examination and clinical chemistry, urine and haematological tests. Patients were randomly allocated by the Pharmacy Department to one of three groups each containing 12 patients:
• One clinical group (n=12) to receive placebo (5 g of Polyethylene Glycol 400)
• Second clinical group (n=12) to receive low-dose (5 g) refined Borage officinalis
• Third clinical group (n=12) to receive high-dose (15 g) refined Borage officinalis Supplementation was in the form of one gram oil capsules daily (5/day for low dose, 15/day high dose) for 18 months duration. Borage officinalis oil and omega-6 polyunsaturated fatty acids are food ingredients that are generally recognised as safe for human consumption (GRAS). There are no classification or labelling requirements under EC regulations. Clinical assessment included: Extended Disability Scale Scores (EDSS) and clinical relapse record. Venous blood (50 mis) was obtained for laboratory studies on the 1st, 3rd, 6th, 12th, 15th, and 18th month of supplementation. The following biochemical and immunological parameters were investigated on each visit for comparison with pre-treatment data and between group data: • Stimulated and unstimulated ex vivo peripheral blood mononuclear cell cytokine production: changes in TGF-βl, IFN-γ, TNF-α, IL-lβ, LL-6 and IFN-β, which are implicated in the pathogenesis of MS. Cytokine and related gene expression. • Soluble adhesion molecules in serum particularly ICAM-1 and VCAM-1 • Peripheral blood mononuclear cell membrane fatty acids and plasma phospholipid fatty acid composition.
Results are shown in Tables 1 and 2 and Figures 1 to 5. The primary outcome parameter was the number of clinical relapses between baseline (Month 0) and the end of treatment (Month 18). Secondary outcome parameters included: the time to first clinical relapse; severity of relapses, as assessed by EDSS score and the use of steroid treatment; and changes in EDSS at Month 3, 6, 9, 12, and 18 compared to baseline and defined as at least 1.0 point increase in the EDSS that is sustained for 3 months or at least 1.5 point increase on the EDSS from the baseline EDSS that is sustained for 3 months. Eleven patients were in the placebo group, seven patients had been taking low- dose Borage oil, and ten patients had been taking high-dose Borage oil. The study drug was well-tolerated, and there were no serious adverse events during the 18- month trial.
Isolation and Culture of PBMC Heparinised whole blood was diluted with an equal volume of Hanks' balanced salt solution (Sigma, UK) and the resulting diluted blood layered onto Lymphoprep (Nycomed, Oslo, Norway). Following density centrifugation at 800g for 30 minutes the PBMC were removed from the interface and diluted in Hanks' solution. The cells were then washed twice by centrifugation for 10 minutes at 250g. The resulting final pellet was then resuspended in culture medium consisting of RPMI-1640 medium (Sigma, UK) supplemented with 2mM L-glutamine, 100U penicillin and lOOμg streptomycin (Sigma, UK) and 10% autologous plasma. 2x106 per ml PBMC, >95% viable as judged by trypan blue exclusion, were added to tissue culture tubes (Bibby Sterilin Ltd, Stone, UK) and incubated for 24h at 37°C with 5% CO2. The concentration of antigen, cell density and time of culture were all determined in previous kinetic experiments to determine maximum cytokine production (data not shown). Routine cytospin preparations were also prepared for subsequent differential counts. Following incubation the cells were removed from culture by centrifugation at 250g for 10 minutes, the resulting supernatants were then removed, aliquoted and stored at -70°C.
Preparation of Plasma Samples
10ml of heparinised blood was spun at 250g for 10 minutes. The resulting plasma layer was then removed, aliquoted and stored at -70°C.
Detection of Pro-inflammatory Cytokines
TNF-α, IL-lβ and IFN-γ in cell culture supernatants and plasma were detected using commercially available paired antibodies enabling cytokine detection in an ELISA format (R&D systems Ltd, Abingdon, UK). The sensitivities for the TNF-α and IFN-γ ELISAs were 15.6-lOOOpg/ml and 3.9-250ρg/ml for IL-lβ.
Detection of Biologically Active TGF-βl
Biologically active TGF-βl in cell culture supernatants and plasma were detected using the commercially available Emax ELISA system with a sensitivity of 15.6- lOOOpg/ml (Promega, Southampton, UK).
Statistical Analysis Differences in cytokine production were compared using Student's t-test and Mann- Whitney [/-test and were considered significant when p values were less than 0.05. RESULTS Two patients had developed diarrhoea, both of whom were later confirmed to have been taking high-dose Borage oil. The diarrhoea was mild in one patient, but was moderately severe in the second patient, who later discontinued the study drug. The code was not broken and the diarrhoea had stopped after the discontinuation of the drug, but reappeared upon re-challenge. Therefore, this patient was withdrawn from the trial. The remaining patients who were treated with high-dose Borage oil showed excellent clinical improvement on all primary and secondary outcome criteria. For example, their mean EDSS score after 6 months of treatment had improved from baseline EDSS (Figure 1). More importantly, the mean number of clinical relapses had significantly reduced after 6 months of treatment when compared to the number of relapses in the placebo group (Figure 2). In contrast, patients who had been receiving low-dose Borage oil did not show any clinical improvement when compared to the placebo group. In addition to its beneficial effect on MS disease activity, high dose Borage oil provided some symptomatic relief of muscle spasticity (stiffness) and painful sensory symptoms, and also improved cognitive functions. As can be seen for the figures below, relapse rate after 9, 12 and 18 months was down to zero in the high dose group. The increase seen at 15 months was due to the patient dropping out of this group. The following are tliree briefcase histories to illustrate the therapeutic benefits of high dose high sn-2 GLA Borage oil. The first two are from the trial while the third is a post trial patient for whom MRI studies were obtained.
Patient 1 (Treatment); The first patient was a 48 year old woman who had had a clinically active, relapsing remitting MS for 9 years. She had originally worked as a full-time administrator at the local Health Authority, but she was unable to perform her duties because of her severe MS. Therefore, she later worked as a part-time secretary, but still had difficulties in mobilization because of muscles stiffness and sensory disturbances. She was also experiencing severe clinical relapses at an average of one relapse every nine months. Most of these relapses had resulted in hospital admissions for steroid therapy. In view of her active MS, she was recruited into the Borage oil trial. There were no adverse events relating to the study, and after taking the medication for four months, she experienced good improvement in her walking and sensory symptoms. About nine months after therapy, she was well enough to start full-time employment. In addition, she remained relapse- free for the 18-month duration of the clinical trial. Following the conclusion of the trial, the treatment code revealed that she was taking high-dose Borage oil.
Patient 2 (Control): The second case was a 46-year old woman who also had a clinically active relapsing remitting MS for 8 years. She had originally worked as a shop assistant, but became unemployed after MS was diagnosed. Her symptoms included difficulty with mobilisation and painful sensory symptoms in both legs. She had experienced three clinical relapses in the two years preceding the clinical trial, and had been admitted to hospital twice for steroid therapy. Consequently, she was recruited into the Borage oil trial, but her walking continued to deteriorate. Six months into the trial, she need to use a walking stick and also received treatment with Baclofen to reduce low limb spasticity. Approximately ten months after starting the Borage oil trial, she was admitted to hospital because of severe clinical relapse, which was treated with steroids. She later developed bladder disturbances and began to use a wheelchair for long journeys. The treatment code was broken after the conclusion of the 18-month trial, and she was found to have been taking placebo. Since then, she started using a walking frame for journeys exceeding 50 yards. Patient 3: Treatment (additional to trial) The third case was a 26 year-old man who was diagnosed with definite MS in April 2001. His symptoms had started in 1999 when he complained of diffuse, intractable pain affecting various parts of his body, particularly the left side of the chest and abdomen. This was followed by intermittent numbness in the hands and feet, associated with fluctuating weakness. There were also distressing bladder symptoms in the form of urinary frequency and urgency. The diagnosis of MS in 2001 was based on his relapsing remitting symptoms, and was confirmed by positive cerebrospinal fluid analysis and magnetic resonance imaging (MRI) of the brain, which showed multiple white matter abnormalities in both cerebral hemispheres. Symptoms did not respond to various pharmaceutical therapies. In April 2003, oral supplementation with the present high dose Borage oil was commenced. The patient reported dramatic improvement in his symptoms within three months of starting this oral supplementation. His painful sensory symptoms disappeared completely. He reported no numbness or weakness since May 2003, and noticed significant improvement in his bladder control. The oral supplementation caused no adverse events. A repeat brain MRI was undertaken to verify the reported improvement in Mr N's symptoms. The repeat MRI showed a reduction in the size and distribution of the white matter abnormalities.
EXAMPLES; Structured sn-2 lipids
In all the examples below higher purity is obtained by use of higher purity starting material γ-linolenic, dihomo-γ-linolenic or arachidonic acid, such as is available eg from Sigma Aldrich. GLA 95 indicates 95% pure γ-linolenic acid.
Synthesis Example 1: synthesis of Trigammalinolenin
1) Acid chloride method 2.0 g (7.2 mmol, 3.1 equiv) GLA95 (95% pure γ-linolenic acid) was dissolved in 10 ml DCM. l.Olg (0.71 ml, 8.0 mmol, 3.4 equiv) oxalyl chloride in 5 ml DCM added dropwise over 2-3 min under nitrogen. Stirred at RT overnight. Reaction mixture concentrated in vacuo to remove DCM and excess oxalyl chloride. This acid chloride was then added dropwise over 2-3 min to a stirred mixture of 215 mg (2.3 mmol, 1 equiv) of glycerol, 0.58 ml (3.1 equiv) pyridine and 10 ml DCM under nitrogen. The mixture was stirred at RT overnight. The pyridine hydrochloride formed was then filtered off and washed with DCM. The solution was washed 1 x 4 ml water, 0.1N HC1, 5% sodium bicarbonate and 5% NaCl. Dried over magnesium sulphate, filtered and concentrated in vacuo to a yellow oil. This oil was purified on a silica column using 10% ether in hexane as eluting solvent. A clear colourless oil was obtained, a sample of which was trans- esterified and subsequently analysed by GC. The product contained 96.3% GLA
2) DCCI method 2.19 g GLA95 (3.15 equiv), 230 mg (1 equiv) glycerol, 153 mg DMAP (0.5 equiv) were stirred in 10 ml DCM under nitrogen. 1.85 g DCCI (3.6 equiv) in 5 ml DCM was added. The reaction mixture was stirred at RT under nitrogen overnight. The DCU formed was filtered and washed with DCM. DCM washed 1 x 5mls N HC1, water, 5% sodium bicarbonate and water. Dried over magnesium sulphate, filtered and concentrated in vacuo to an oil. This oil was then purified on a silica column using 10% ether in hexane as eluting solvent. 1.47 g (67%) of a slightly cloudy oil was obtained. A sample of this product was trans-esterified and subjected to GC analysis. The product contained 95.8% GLA.
Scale-up 20 g (0.072 mol, 3.1 equiv) of GLA95 (gamma linolenic acid, 95%) was dissolved in 100 ml DCM. 13.7g (9.3 ml, 0.11 mol, 4.78 equiv) oxalyl chloride was added over 3-4 min under nitrogen. The reaction mixture was stirred under nitrogen overnight. It was then concentrated in vacuo to remove DCM and excess oxalyl chloride. This oil was then added dropwise over ca 5 min to a stirred mixture of 2.14g (0.023 mol, 1 equiv) of glycerol, 100 ml DCM and 5.8 ml (5.68 g, 0.072 mol, 3.1 equiv) of pyridine under nitrogen. 85 mg (0.7 mmol, 0.03 equiv) of DMAP (4- dimethylaminopyridine) catalyst was added.. The mixture was stirred at RT overnight. Pyridine hydrochlori.de was filtered off and washed with DCM. The DCM solution was washed lx 25 ml: water, 10% sodium bicarbonate, 0.1N HC1, 5% NaCl. (Emulsions formed during this process, especially at first). The DCM was dried over magnesium sulphate, filtered and concentrated in vacuo to a brown oil (~21 g). The oil was purified on a silica column using 5% ether in hexane at first and then 10%. 15.6g (77% yield) of a clear oil was obtained. By tic this material contained a small amount of free GLA. (This material was repurified at a later date)
Large Scale-up The above reaction was repeated on 10 times scale. Thus, 200 g of GLA95, 1L DCM, 137 g of oxalyl chloride, and 21.4 g of glycerol were used. On the addition of the acid chloride the reaction mixture was cooled in a cold water bath and the temperature kept below 35°C. 250g of a brown oil were produced. This was initially
purified on a 500 gram silica column. The oil was dissolved in 200 ml hexane and applied to the column. The column was eluted at first with hexane, then 5% ether in hexane and then 10%. Fractions were collected and analysed by tic eventually yielding two batches of oils. The first A (66 g) contained a small amount of front running impurity and a little GLA (slower running than TGL), the second fraction B (99g) was clear of front running impurity and contained a little GLA. The large scale reaction was repeated using 169 g of GLA and gave two fractions as above. This time there was 85g of 'A' fraction and 54g of 'B' fraction. Both batches of 'A' were combined and re-purified on a 500g silica column. The 'B' fractions were treated in a similar manner (15g of material from the small-scale reaction were also added to this batch). Some fractions from the above were again re-purified to eventually give 259 grams of oil. The oil was pumped down on a rotary evaporator under high vacuum to constant weight - 256g. This represents an overall yield of 65%. Analysis of product.
GC A small sample was trans-esterified and subjected to GC analysis: The GLA content was 97.1%. The main impurity was linoleic acid - 1.91%. Note: The original GLA95 that was used for the synthesis contained 96.2%
GLA and 2.42% linoleic acid.
HPLC An HPLC method was developed using a reversed phase column (Hypersil C18 4.6 x 100 mm), eluting with 80/20 acetonitrile/THF. Detection was by UV at 210 nm. This showed the product to be a mixture of three components. The main peak (93.6%) was the required product. A slower running impurity (representing 5.0% of the product) was probably a GGL1 triglyceride (LI = linoleic acid). A second impurity was slightly faster running and represented 1.4% of the product. Note: Absorption at 210 nm varies considerably between triglycerides of differing fatty acid content. For example trigammalinolenin has a UV absorbtion 5-6 times greater than that of trilinolenin
Summary 254 g of glycerol tri-6,9,12-linolenate (gamma linolenic acid triglyceride, trigammalinolenin, GGG) was prepared from 96.2 % GLA by a two-step acid chloride route. It is a clear, pale yellow oil and was stored under nitrogen in the freezer. The GLA content was 97.1 % and no C20:l, C22:l, or C24:l acids were detected). The HPLC purity was 93.6 %.
Synthesis of higher purity GGG would is readily achievable using GLA 98 (98% γ- linolenic acid: Scotia ) or higher starting material. Comparative lipid 1: synthesis Tricaprin (Glycerol tridecanoate)
Small Scale Glycerol (3.0 g, 0.0325 mol, 1 eq) pyridine (8.1 ml, 0.10 mol, 3.1 eq) and dichloromethane (100 ml) were stirred at room temperature under nitrogen. Decanoyl chloride (21 ml, 19.25 g, 0.10 mol, 3.1 equiv) was then added dropwise over 5 min, with external cooling in a water bath to keep the temperature at 30-35 °C. When the addition was complete 4-dimethylaminopyridine (DMAP (0.12 g, 1 mmol, 0.03 eq) was added and the mixture stirred under nitrogen at room temperature overnight. The precipitated pyridine hydrochloride was removed by filtration and washed with dichloromethane. The combined washing and filtrate was then washed with aqueous solutions (20 ml) of 5% sodium chloride, 5% sodium bicarbonate, 0.1N hydrochloric acid, and 5% sodium chloride. The dichloromethane layer was then dried over MgSO4 and the solvent removed in vacuo. The residual oil crystallised on standing. This material was recrystallised from isopropanol (40 ml) to give 15.6 g (86% yield) of a waxy white solid.
Analysis GC - 99.8% pure HPLC (C18 4.6 x 100 mm, ACN/THF 85/15 1 ml/min, λ 210 nm) - 94.9% pure
Large Scale The above was repeated on 15 times the scale. Glycerol (45.0 g, 0.49 mol, 1 eq), pyridine (121.5 ml, 1.50 mol, 3.1 eq) and dichloromethane (1.5 L) were stirred at room temperature under nitrogen. Decanoyl chloride (315 ml, 288.8 g, 1.50 mol, 3.1 equiv) was then added dropwise over 15 min, with external cooling in a water bath to keep the temperature at 30-35 °C. When the addition was complete 4-dimethylammopyridine (DMAP (1.8 g, 15 mmol, 0.03 eq) was added and the mixture stirred under nitrogen at room temperature overnight. The precipitated pyridine hydrochloride was removed by filtration and washed with dichloromethane. The combined washing and filtrate was then washed with aqueous solutions (300 ml) of 5% sodium chloride, 5% sodium bicarbonate, 0. IN hydrochloric acid, and 5% sodium chloride. The dichloromethane layer was then dried over MgSO4 and the solvent removed in vacuo. The residual oil crystallised on standing. This material was recrystalhsed from isopropanol (400 ml) to give 228 g (86% yield) of a waxy white solid.
Analysis GC - 99.8% pure HPLC (C18 4.6 x 100 mm, ACN/THF 85/15 1 ml/min, λ 210 nm) - 94.9% pure A further batch was made and combined with the small-scale batch above and recrystalhsed from isopropanol to give 44 g of product. The above batches were combined (268 g) and reanalysed: GC 99.9% pure HPLC 97.9%
Summary
263 g of glycerol tridecanoate (tricaprin, CCC) was been prepared from decanoyl chloride (98 %) by a one-step process (scheme given below). It is a white, low-melting solid and was stored under nitrogen in the freezer. The C content was 99.9 % of fatty acid content and the HPLC purity was 97.9 %. Synthesis Example 2; 1,3-Dicaprin 2-gammalinolenoate (Glycerol 1,3- didecanoate 2-octadecatri(6-Z,9-Z,12-Z)enoate or CGC) This triglyceride is novel. Unlike CGC, its isomer CLnC (Ln = α-linolenic acid), has been identified (see K. Long et al Biotechnol. Lett. ,20, 369-372 (1998). and H. Mu, P. Kalo et al, Eur. J. Lipid Sci. Technol, 102, 202-211(2000). as a component of coconut oil. In addition, CLxC (Lx = a linolenic acid of unspecified double bond position) has been described (see J. Gresti et al. J. Dairy Sci., 76, 1850-1869 (1993)), The two intermediates used in the synthesis of CGC are known (see L. El
Kihel et al Arzneim -Forsch./Drug Res., 46, 1040-1044 (1996) and US 4178299. The last step described below is novel and the first two stages are also inventive since they are more suitable for large scale production than those previously reported. CGC was prepared by reaction of 1,3-Dicaprin with GLA-chloride in dichloromethane-pyridine. 1,3-Dicaprin was prepared by sodium borohydride reduction of l,3-didecanoyloxypropan-2-one, which was in turn prepared by reaction of decanoyl chloride with 1,3-dihydroxyacetone. The intermediate 1,3-dicaprin must be handled with care since it can undergo acyl migration on exposure to acids , bases and heat. An older method of making 1,3-dicaprin has been described (see A. P. J. Mank et al Chem. Physics Lipids, 16, 107-114 (1976). A versatile, flexible synthesis of 1,3-diglycerides and triglycerides. by catalysed addition of decanoic acid to a glycidol ester (from epichlorohydrin) is less attractive because of more severe reaction conditions and acyl migration problems. The final product, CGC, was purified by careful column chromatography on silica which removed by-products.
Small Scale 1, S-didecanoyloxypropan-2-one Decanoyl chloride (40.0 ml, 36.8 g, 0.19 mol, 1.98 equiv) was added dropwise over 10-15 min to a stirred suspension of 1,3-dihydroxyacetone dimer (8.68 g, 0.048 mol, 1.0 equiv), pyridine (15.6 ml, 0.19 mol), 4-dimethylaminopyridine (0.18 g, 0.0014 mol, 0.03 equiv) and dichloromethane (DCM, 150 ml) at room temperature under nitrogen. The temperature of the reaction mixture was kept below 30°C by cooling in a cold water bath. The reaction mixture was stirred at RT under nitrogen overnight. The pyridine hydrochloride formed was removed by filtration and washed with DCM. The combined filtrate and washings were then washed with 1 x 25ml portions of 5%NaCl, 5%NaHCO3, 0.1N HC1, 5%NaCl. The solution was then dried over MgSO and concentrated in vacuo to a yellowish semi-solid. This was then crystallised from methanol (150 ml) to give a white solid. The yield was 28.2 g (73%). 1,3-Dicaprin The above ketone (28.2 g, 0.071 mol) was dissolved in tetrahydrofuran (THF, 200 ml). Water (10 ml) was then added, the solution cooled to 5°C, and sodium borohydride (5.38 g, 0.14 mol) added portionwise below 10°C. The reaction mixture was stirred at RT for lh and then concentrated in vacuo to remove THF. The residue was partitioned between ethyl acetate and 5% sodium chloride solution. The aqueous phase was re-extracted with ethyl acetate and the combined extracts dried over MgSO4 and concentrated in vacuo to a waxy solid. This was crystallised twice from hexane to give 11.2g (40%) of a white solid. (99%+ pure by HPLC) 1 ,3-Dicaprin 2-gammalinolenoate (CGC) Gamma-linolenic acid (GLA95, 8.34 g, 0.03 mol) was dissolved in dichloromethane (DCM, 60 ml). The resulting solution was stirred at RT under nitrogen and oxalyl chloride (3.9 ml, 5.67 g, 0.044 mol) added dropwise over 5 mins. The mixture was stirred at RT overnight and then concentrated in vacuo to remove DCM and excess oxalyl chloride. The residual oily acid chloride (GLA-Cl) was then added dropwise over 15 min (ice /water cooling) to a stirred solution of 1,3-dicaprin (11.2g, 0.028 mol), DCM (50 ml), pyridine (2.42 ml, 2.37 g, 0.03 mol) and 4- dimethylaminopyridine (0.10 g, 0.0008 mol, 0.03 equiv) at 10-15°C . The temperature was maintained by ice-water cooling. The reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM. The combined washing and filtrate was washed with 1 x 20ml portions of 5%NaCl, 5%NaHCO3, 0.1N HC1, 5%NaCl. The solution was then dried over MgSO4 and the solvent removed in vacuo.The residual brown oil was purified by column chromatography on silica. Elution with hexane and then with 5% ether/hexane gave 10.3g (56%) of a colourless oil. The structure was confirmed by 13C NMR and GLC. Purity determined by HPLC.
Large Scale 1, 3-didecanoyloxypropan-2-one Decanoyl chloride (272 ml, 250 g, 1.3 mol, 2 equiv) was added dropwise over
10-15 min to a stirred suspension of 1,3-dihydroxyacetone dimer (59.1 g, 0.65 mol, 1.0 equiv), pyridine (106 ml, 103.7g 1.3 mol), 4-dimethylaminopyridine (2.38 g, 0.02 mol, 0.03 equiv) and dichloromethane (DCM, 750ml) at room temperature under nitrogen. The temperature of the reaction mixture was kept below 30°C by cooling in a cold water bath. The reaction mixture was stirred at RT under nitrogen overnight. The pyridine hydrochloride formed was removed by filtration and washed with DCM. The combined filtrate and washings were then washed with 1 x 150ml portions of 5%NaCl, 5%NaHCO3, 0.1N HC1, 5%NaCl. The solution was then dried over MgSO4 and concentrated in vacuo to a yellowish semi-solid. This was then crystallised from methanol (500ml) to give a white solid. The yield was 158 g (60%).
1,3-Dicaprin The above ketone (158 g, 0.40 mol) was dissolved in tetrahydrofuran (THF, 2.25 L). Water (50 ml) was then added, the solution cooled to 5°C, and sodium borohydride (5.66 g, 1.5eq) added portionwise below 10°C. The reaction mixture was monitored by HPLC (C18, eluted with ACN at lml min λ210nm) (Note: only about 4.5g of the borohydride was in fact added, as all SM had reacted).The reaction mixture was stirred at RT for lh and then concentrated in vacuo to remove THF. The residue was partitioned between ethyl acetate and 5% sodium chloride solution. The aqueous phase was re-extracted with ethyl acetate and the combined extracts dried over MgSO and concentrated in vacuo to a waxy solid. This was crystallised twice from hexane to give 96g (60%) of a white solid. (98% pure by HPLC)
1,3-Dicaprin 2-gammalinolenoate (CGC) Gamma-linolenic acid (GLA95, 120.2g, 0.43mol) was dissolved in dichloromethane (DCM, 750 ml). The resulting solution was stirred at RT under nitrogen and oxalyl chloride (55.7 ml, 82.3 g, 0.65 mol, 1.5eq) added dropwise at 15- 20°C over 15 mins. The mixture was stirred at RT overnight and then concentrated in vacuo to remove DCM and excess oxalyl chloride. The residual oily acid chloride (GLA-Cl) was then added dropwise over 30-40 min at 10-15 °C (ice /water cooling) to a stirred solution of 1,3-dicaprin (164.7g, 0.41 mol), DCM (650 ml), pyridine (33.3 ml, 32.5 g, 0.41 mol) and 4-dimethylaminopyridine (1.50 g, 0.012 mol, 0.03 equiv) at 10-15°C .The reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM. The combined washing and filtrate was washed with 1 x 150 ml portions of 5%NaCl, 5%NaHCO3, 0.1N HC1, 5%NaCl. The solution was then dried over MgSO4 and the solvent removed in vacuo to a brown oil (275g). The scale of the above three reactions was the largest on which each was carried out. The borohydride reduction produced, in addition to 1,3-dicaprin, a by- product in variable yield. The presence of this by-product greatly affected the yield of the isolated pure 1,3-dicaprin; the by-product could only be removed by two crystallisations of the crude product. Since the final product, CGC, is purified by column chromatography, it is imperative that the 1,3-dicaprin used for the final step is as pure as possible! From the above reactions about 440g of crude CGC was produced as a brown oil. This was purified on a series of silica columns using hexane followed by 2-3% ether/hexane. The purification required 7 or 8 columns, using 3-4 kilos of silica, 25-30 litres of solvent (recycling solvent kept this figure low - in practice over 100 litres were used) The resulting product, a clear almost colourless oil, (264grams) was 96.4% pure by HPLC (C18 4.6 x 100mm, eluted with 85/15 ACN/THF at lml/min. UV detection λ210nm). GC indicated a ratio of 66.1/33.9 C/G. NMR analysis indicated the product to have the correct CGC structure and be of at least 95% purity: δc (500 MHz, CDC13) 172.65 (2-GLA carbonyl), 173.25 (1,3- capric carbonyl). Ratio of signals 2.04:1. No signal at 173.0 indicating absence of 1.3-GLA. Trace signal at 172.79 could be oleic acid impurity in GLA or 2-capric acid.
Summary 264 g of glycerol l,3-didecanoate-2-gammalinolenoate (l,3-dicaprin-2-GLA, CGC) has been prepared from decanoyl chloride (98 %) by a three-step process (scheme given below). It is an almost colourless oil (slight yellow tinge) and was stored under nitrogen in the freezer. The HPLC purity was 96.4 %.
Synthesis Example 3 l,3-Didecanoate-2-dihomo-γ-linolenoate (Glycerol l,3-didecanoate2- eicosa- (8Z,llZ,14Z)-trienoate or C(DHLA)C
This triglyceride appears to be novel - no reference to it has been found.
DHLA (3.93g, 12.8 mmol, 1 eq) was dissolved in dichloromethane (DCM, 20 ml) and stirred at room temperature under a nitrogen atmosphere. Oxalyl chloride (1.69 ml, 2.46 g, 19.4 mmol, 1.5 eq) was added dropwise over 1-2 min, and left stirring at room temperature overnight. The resulting solution was concentrated in vacuo to remove DCM and excess oxalyl chloride. The residual oily acid chloride (DHLA-C1) was then added dropwise over 5 min at 25°C to a stirred mixture of 1,3-dicaprin (4.91 g, 12.2 mmol, 0.95 eq), pyridine (0.98 ml, 0.96 g 12.1 mmol, 0.95 eq) and 4-dimethylam inopyridine (DMAP, 8 mg, 0.07 mmol, 0.03 eq). The reaction temperature rose to 32 °C during the addition. The reaction was stirred at 30-35°C and monitored by HPLC. The reaction was stopped after 1.5h. The precipitated pyridine hydrochloride was filtered off and washed with DCM. The combined filtrate and washings were then washed with 1 x 10 ml portions of 5% NaCl, 5% NaHCO3, 0.1N HC1, 5% NaCl. The solution was then dried over MgSO4 and concentrated in vacuo to give the crude product as a yellow-orange oil (8.9 g, 86% purity by HPLC). This oil was chromatographed on silica gel (250 g). Elution with hexane and diethyl ether-hexane (2-6%) gave a purified product as a pale yellow oil. Treatment of a hexane solution with decolourising charcoal and removal of the solvent in vacuo gave C(DHLA)C as a clear colourless oil (6.48g, 98.9% purity by HPLC).
Synthesis Example 4 Triarachidin (Glycerol trieicosotetra5-Z,8-Z,ll-Z,14Z-eneoate) or AAA
Arachidonic acid (50.9 g, 0.17 mol, 3 eq) was dissolved in dichloromethane (DCM, 175 ml) and stirred at room temperature under a nitrogen atmosphere. Oxalyl Chloride (21.9 ml, 31.9 g, 0.25 mol, 4.4 eq) was then added to the stirred solution over 5 min and the temperature increased by 4 °C. The resulting yellow-green mixture was stirred at RT overnight and then concentrated in vacuo to remove DCM and excess oxalyl chloride. The residual oily acid chloride (A-Cl) was then added dropwise over 15 min to a pre- warmed (25 °C) stirred mixture of glycerol (5.11 g, 0.055 mol, 1 eq), pyridine (13.5 ml, 13.2 g, 0.17 mol, 3 eq) and 4-dimethylamino pyridine (DMAP, 0.20 g, 0.002 mol, 0.03 eq). The temperature of the reaction mixture rose to 42 °C during the addition and a gentle reflux was observed. The mixture was stirred at 30-40 °C and monitored by HPLC. After 2 h, no further product formation was observed. The precipitated pyridine hydrochloride was filtered off and washed with DCM. The combined filtrate and washings were then washed with 1 x 50 ml portions of 5% NaCl, 5% NaHCO3, 0.1N HC1, 5% NaCl. The solution was then dried over MgSO4 and concentrated in vacuo to give the crude product as a yellow-orange oil (57 g). This oil was purified by column chromatography on silica gel (ca. 600 g). Elution with hexane and diethyl ether(2-4%)-hexane gave 22.8 g of the product as an oil. A second batch (17.8 g) was produced from 39.8 g of arachidonic acid, The two batches were combined and residual solvents removed under vacuo to give 40.5 g (43%) of a mobile pale yellow oil. HPLC purity 84.8% GLC analysis 94.3% AA (arachidonic acid).
Comparative Lipid 2 1 ,3-Di(octadeca-6Z,9Z, 12Z-enoyloxy)proρan-2-one
(l,3-Di(γ-linolenoyloxy)propan-2one ,GonG) Stage 1 intermediate for GCG Gamma-linolenic acid (GLA95, 197g, 0.71 mol, 2.2 equiv) was dissolved in dichloromethane (DCM, 600 ml) contained in a 2L 3 necked flask. The resulting solution was stirred at RT under nitrogen. Oxalyl chloride (93 ml, 136 g, 1.07 mol, 3.3eq) was added dropwise at 15-20°C over 15 min. The brown mixture was stirred at RT overnight and then concentrated in vacuo to remove DCM and excess oxalyl chloride. The residual oily acid chloride (GLA-Cl) was then added dropwise over 20 min at 25°C to a stirred mixture of 1,3-dihydroxyacetone dimer (28.99 g, 0.32 mol, 1.0 equiv), pyridine (52 ml, 50.9 g 0.64 mol, 2.0 equiv), 4-dimethylaminopyridine (2.36 g, 0.02 mol, 0.06 equiv) and dichloromethane (DCM, 600 ml) at room temperature under nitrogen. The temperature of the reaction mixture was allowed to rise to 40°C and the mixture was stirred for a further 2 h under nitrogen (monitored by HPLC). The pyridine hydrochloride that formed was removed by filtration and washed with DCM. The combined filtrate and washings were then washed with 1 x 150 ml portions of 5% NaCl, 5% NaHCO3, 0.1N HCl, 5% NaCl. The solution was then dried over MgSO and concentrated in vacuo to give ca. 200 g of a yellow oil. This material was partially purified by column chromatography on silica (600 g). Elution with hexane and then ether-hexane mixtures (2-15%) gave 42 g of a pale yellow oil. This oil was chromatographed again on silica (600 g) and eluted with hexane and then 1-10% ether-hexane to give the product (95.9% purity) as a pale yellow oil. The yield was 42 g (17%).
1 ,3-Di(octadeca-6Z.9Z, 12Z-enoyloxy)propan-2-ol
(l,3-Di(γ-linolenoyloxy)propan-2-ol or 3-Di-gamma-linolenin GolG) Stage 2 intermediate for GCG 13-Di(γ-linolenoyloxy)propan-2-one (GonG, 25.5 g, 0.04 mol, 1 eq) was dissolved in tetrahydrofuran (THF, 375 ml) and water (12.7 ml). The solution was vigorously stirred at -20°C, care was taken to keep the reaction temperature below - 15°C. Sodium borohydride (790 mg, 0.02 mol, 1.25 eq) was added portionwise to the stirred solution over 3 mins. The reaction mixture was stirred for a further 10 mins at -20°C and hexane (380 ml) then added. The still cold mixture was then washed with water (2 x 200 ml), dried over MgSO and concentrated in vacuo to give the title compound as a brown oil (27.8g) (82.6 % purity by HPLC, less than 1% migrated material). Another batch was prepared and combined with the first to give 50 g of crude product. This material was purified by column chromatography on silica gel (400 g). Elution with hexane and diethyl ether-hexane mixture (5-20%) gave 36.1 g of the product as a pale oil (91.5 % purity). (N.B. Care should be taken not to leave the compound on the silica overnight as it appears to undergo a migration reaction, giving GGol)
1,3-Di-γ-linolenin 2-decanoate (Glycerol l,3-dioctadeca-(6Z,9ZJ2Z -trienoate 2- decanoate or GCG) Decanoyl chloride (13.5 ml, 12.4 g, 0.065 mol, 1.1 eq) was added to a stirred solution of 1,3-di-γ-linolenin (36.1 g, 0.059 mol, leq), dry pyridine (5.7 ml, 5.6 g, 0.07 mol, l.leq), 4-dimethylaminopyridine (0.2 g, 0.002 mol, 0.03 eq) and dichloromethane (DCM, 150 ml) over ca. 10 mins. The temperature was maintained at 17 °C -23 °C during addition. The reaction was then stirred at 30-35 °C and monitored by HPLC. A further 1-2 ml of decanoyl chloride was added after 1 h, 1.5 h and 2 h. Further addition appeared to increase the conversion to product as determined by HPLC. After 3 h the reaction mixture was filtered and the filtrate washed with DCM. The combined filtrate and washings were then washed with 1 x 50 ml portions of 5% NaCl, 5% NaHCO3, 0.1N HCl, 5% NaCl. The DCM extract was then dried over MgSO and concentrated in vacuo to give the crude product as a pale yellow oil; (purity 90% by HPLC). The oil was purified by column chromatography on silica gel (600 g). Elution with hexane and diethyl ether-hexane (1.5-2.5 then 3.5%) gave the product (GCG.) as a clear oil; (35.5 g 96.1% purity by HPLC). Another 7.5 g of pure lipid was obtained by further chromatography on some of the fractions containing only a small amount of impurity.
Synthesis Example 5
1,3-Dicaprin 2-arachidonate (Glycerol 1,3-didecanoate 2-eicosatetra-(5-Z,8-Z,ll- ZJ4-Z)enoate or CAP This triglyceride is known. CAC has been identified as a constituent of lymph lipids following administration of safflower oil to rats. WO 03 013,497 describing an arachidonic acid containing triglyceride (produced by culturing Mortierella alpina) useful for diseases caused by brain hypofunction, but specifically for cognition enhancement. The two intermediates used in the synthesis of CAC are known. The synthesis of CAC from 1,3-dicaprin, and the purification of this are all novel. Here CAC was prepared by reaction of 1 ,3-Dicaprin with arachidonyl chloride in dichloromethane-pyridine. 1,3-Dicaprin was prepared by sodium borohydride reduction of l,3-didecanoyloxypropan-2-one, which was in turn prepared by reaction of decanoyl chloride with 1,3-dihydroxyacetone. The intermediate 1,3-dicaprin must be handled with care since it can undergo acyl migration on exposure to acids , bases and heat. An older method 6 of making 1,3-dicaprin, by catalysed addition of decanoic acid to a glycidol ester (from epichlorohydrin) was deemed less attractive because of more severe reaction conditions and acyl migration problems. The final product, CAC, was purified by careful column chromatography on silica which removed byproducts.
1,3-Dicaprin 2-arachidonate (CAC) Arachidonic acid (AA96, 8.34 g, 0.03 mol) was dissolved in dichloromethane (DCM, 60 ml). The resulting solution was stirred at RT under nitrogen and oxalyl chloride (3.9 ml, 5.67 g, 0.044 mol) added dropwise over 5 mins. The mixture was stirred at RT overnight and then concentrated in vacuo to remove DCM and excess oxalyl chloride. The residual oily acid chloride (GLA-Cl) was then added dropwise over 15 min (ice /water cooling) to a stirred solution of 1,3-dicaprin (11.2g, 0.028 mol), DCM (50 ml), pyridine (2.42 ml, 2.37 g, 0.03 mol) and 4- dimethylammopyridine (0.10 g, 0.0008 mol, 0.03 equiv) at 10-15°C . The temperature was maintained by ice-water cooling. The reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM. The combined washing and filtrate was washed with 1 x 20ml portions of 5%NaCl, 5%NaHCO3, 0.1N HCl, 5%NaCl. The solution was then dried over MgSO4 and the solvent removed in vacuo. The residual brown oil was purified by column chromatography on silica. Elution with hexane and then with 5% ether/hexane gave 10.3g (56%) of a colourless oil. The structure was confirmed by 13C NMR and GLC. Purity determined by HPLC.
Large Scale
1 ,3 -didecano yloxyprop an-2-one Decanoyl chloride (272 ml, 250 g, 1.3 mol, 2 equiv) was added dropwise over 10-15 min to a stirred suspension of 1,3-dihydroxyacetone dimer (59.1 g, 0.65 mol, 1.0 equiv), pyridine (106 ml, 103.7g 1.3 mol), 4-dimethylaminopyridine (2.38 g, 0.02 mol, 0.03 equiv) and dichloromethane (DCM, 750ml) at room temperature under nitrogen. The temperature of the reaction mixture was kept below 30°C by cooling in a cold water bath. The reaction mixture was stirred at RT under nitrogen overnight. The pyridine hydrochloride formed was removed by filtration and washed with DCM. The combined filtrate and washings were then washed with 1 x 150ml portions of 5%NaCl, 5%NaHCO3, 0.1N HCl, 5%NaCl. The solution was then dried over MgSO4 and concentrated in vacuo to a yellowish semi-solid. This was then crystallised from methanol (500ml) to give a white solid. The yield was 158 g (60%). 1,3-Dicaprin The above ketone (158 g, 0.40 mol) was dissolved in tetrahydrofuran (THF, 2.25 L). Water (50 ml) was then added, the solution cooled to 5°C, and sodium borohydride (5.66 g, 1.5eq) added portionwise below 10°C. The reaction mixture was monitored by HPLC (C18, eluted with ACN at lml/min λ210nm) (Note: only about 4.5g of the borohydride was in fact added, as all SM had reacted).The reaction mixture was stirred at RT for lh and then concentrated in vacuo to remove THF. The residue was partitioned between ethyl acetate and 5% sodium chloride solution. The aqueous phase was re-extracted with ethyl acetate and the combined extracts dried over MgSO4 and concentrated in vacuo to a waxy solid. This was crystallised twice from hexane to give 96g (60%) of a white solid. (98% pure by HPLC)
1,3-Dicaprin 2-arachidonate (CAC) Arachidonic acid (AA96, 78.8 g, 0.26 mol) was dissolved in dichloromethane (DCM, 425 ml). The resulting solution was stirred at RT under nitrogen and oxalyl chloride (33.9 ml, 49.4 g, 0.39 mol, 1.5eq) added dropwise at 15-20°C over 15 mins. The mixture was stirred at RT overnight and then concentrated in vacuo to remove DCM and excess oxalyl chloride. The residual oily acid chloride (GLA-Cl) was then added dropwise over 30-40 min at 10-15°C (ice /water cooling) to a stirred solution of 1,3-dicaprin (94.2 g, 0.24 mol), DCM (450 ml), pyridine (19.1 ml, 18.6 g, 0.24 mol) and 4-dimethylaminopyridine (1.72 1.50 g, 0.014 mol, 0.06 equiv) at 10-15°C .The reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM. The combined washing and filtrate was washed with 1 x 150 ml portions of 5%NaCl, 5%NaHCO3, 0.1N HCl, 5%NaCl. The solution was then dried over MgSO4 and the solvent removed in vacuo to a brown oil (171 g). The scale of the above three reactions was the largest on which each was carried out. The borohydride reduction produced, in addition to 1,3-dicaprin, a byproduct in variable yield. The presence of this by-product greatly affected the yield of the isolated pure 1,3-dicaprin; the by-product could only be removed by two crystallisations of the crude product. Since the final product, CAC, is purified by column chromatography, it is imperative that the 1,3-dicaprin used for the final step is as pure as possible! 412 g of crude CAC was produced as a brown oil from the above reactions. This material was purified on a series of silica columns using hexane followed by 1- 3% ether/hexane. The purification required 7 or 8 columns, using 3-4 kilos of silica, and 100 litres of solvent. The resulting product, a clear very pale yellow oil, (295grams) was 95.8% pure by HPLC (C18 4.6 x 100mm, eluted with 85/15 ACN/THF at lml/min. UV detection λ210nm). GC indicated a ratio of 66.3/32.1 C/A (1.6% impurity carried through from the 5% impurity in A).
Summary
295 g of glycerol l,3-didecanoate-2-arachidonate (l,3-dicaprin-2-AA, CAC) has been prepared from decanoyl chloride (98 %) and Arachidonic acid (95%) by a three-step process (scheme given below). It is a very pale yellow oil and was stored under nitrogen in the freezer. The HPLC purity is 95.8 %.
Synthesis Example 7 1,3-Dioleoin 2-gammalinolenoate (Glycerol 1.3-dioctadeca-9Z-enoate 2- octadecatri(6-Z.9-Z,12-Z)enoate or OGO) This triglyceride is known: a carbon-14 labelled version has been prepared by normal chemical synthesis and the normal unlabelled form by biochemical synthesis using lipases. OGO is not a major component of borage oil but its isomer OOG is (9%). The two intermediates used in the synthesis of CGC are known. The last step is novel. The use of, the synthesis of from 1,3-dioleoin, and the purification of CGC are all believed novel. In general triglycerides CXC are prefened over OXO on patent and cost of goods grounds. OGO was here prepared by reaction of 1,3-Doleoin with GLA-chloride in dichloromethane-pyridine. 1,3-Diolein was prepared by sodium borohydride reduction of l,3-dioleoylpropan-2-one, which was in turn prepared by reaction of oleoyl chloride with 1,3-dihydroxyacetone. The intermediate 1,3-dioleolin must be handled with care since it can undergo acyl migration on exposure to acids , bases and heat. Older methods 7'8 of making 1,3-dioleoin, via mono-tritylglycerols or glycidyl esters was deemed less attractive because of more steps and acyl migration problems. The final product, OGO, was purified by careful column chromatography on silica which removed by-products.
Small Scale
1.3-dioleoylpropan-2-one 155.1g Oleic acid (155.1 g, 0.55 mol, 1.0 equiv, Croda 094 RV05192) was dissolved in dichloromethane (DCM, 500 ml). The solution was stirred at room temperature (RT) under nitrogen and 104.4g (1.5eq 71mls) oxalyl chloride (104.4 g, 71.8 ml, 0.82 mol, 1.5 equiv) was added dropwise at 15-20°C over about 20 mins. The reaction mixture was stirred overnight at RT. The excess oxalyl chloride and DCM were removed in vacuo and the residual oily acid chloride was added dropwise over 15-20 min to a stirred suspension of 1,3-dihydroxyacetone dimer (22.5g, 0.24 mol of monomer), pyridine (40.4 ml), 4-dimethylaminopyridine (1.83g) and dichloromethane (DCM, 500ml) at room temperature under nitrogen. The temperature of the reaction mixture was kept below 20°C by cooling in an ice/water bath. The reaction mixture was stirred at RT under nitrogen overnight. The pyridine hydrochloride formed was removed by filtration and washed with DCM. The combined filtrate and washings were then washed with 1 x 150ml portions of 5%NaCl, 5%NaHCO3, 0.1N HCl, 5%NaCl. The solution was then dried over MgSO4 and concentrated in vacuo to an orange/brown semi-solid. This was triturated in methanol and stored in the 'fridge overnight. The solid deposited (90% pure by HPLC) was then crystallised from diisopropyl ether (DLPE) and methanol to give 51.3g of an off white solid which was 95% pure by HPLC. Further crystallisation from DIPE/methanol yielded 41g (27%) of a 98% pure product.
1,3-Diolein The above ketone (32.8 g, 0.053 mol) was dissolved in tetrahydrofuran (THF,
250 ml). Water (10 ml) was then added, the solution cooled to 5°C, and sodium borohydride added portionwise below 10°C. The reaction was followed by HPLC (C18, ACN/THF 90/10 at 2mls/min ,λ210nm) and after all the starting ketone had reacted the addition of the borohydride was stopped (830mg ,0.022 mol added). The mixture was then concentrated in vacuo to remove THF. The residue was partitioned between ethyl acetate and water. The aqueous phase was re-extracted with ethyl acetate and the combined extracts dried over MgSO and concentrated in vacuo to an oil (~33g) which solidified on cooling.The product (68% pure by HPLC) was crystallised from 100ml hexane at -20°C (in the freezer) overnight This product (92% pure 21.1g) was recrystalhsed from hexane (50ml) to give 18.28g (56%yield) of a product 97.5% pure by HPLC.
1,3-Diolein 2-gammalinolenoate (O-G-O) γ-Linolenic acid (GLA95, 41.2 g, 0.15 mol, 1.1 equiv) was dissolved in dichloromethane (DCM, 250 ml). The resulting solution was stirred at RT under nitrogen and oxalyl chloride (19.1 ml, 28.2 g, 0.22 mol, 1.65 equiv) added dropwise over 5 mins. The mixture was stirred at RT overnight and then concentrated in vacuo to remove DCM and excess oxalyl chloride. The residual oily acid chloride (GLA-Cl) was then added dropwise over 15 min (ice /water cooling) to a stirred solution of 1,3- diolein (83.5g, 0.13 mol), DCM (250 ml), pyridine (10.9 ml, 10.6 g, 0.14 mol) and 4- dimethylaminopyridine (0.49 g, 0.004 mol, 0.15 equiv) at 10-15°C . The temperature was maintained by ice-water cooling. The reaction mixture was stirred at RT under nitrogen overnight. Pyridine hydrochloride was removed by filtration and washed with DCM. The combined washing and filtrate was washed with 1 x 80ml portions of 5%NaCl, 5%NaHCO3, 0. IN HCl, 5%NaCl. The solution was then dried over MgSO4 and the solvent removed in vacuo.The residual brown oil was purified by column chromatography on silica. Elution with hexane and then with 5% ether/hexane gave 63.6g (54%) of a colourless oil. Purity detennined by HPLC.
Summary
64 g of glycerol l,3-oleoate-2-gammalinolenoate (l,3-dioleate-2-GLA, OGO) was prepared from oleoyl chloride (98 %) by a three-step process (scheme given below). It was an almost colourless oil (slight yellow tinge) and is being stored under nitrogen in the freezer. The HPLC purity was 89.4 %.
13C NMR Data for Structured lipids
GGG δc (125.7 MHz, CDC13) 172.69 (IC, C-2 carbonyl), 173.09 (2C, C-1, C-3 carbonyls)
CGC δc (125.7 MHz, CDC13) 172.76 (IC, C-2 carbonyl), 173.17 (2C, C-1, C-3 carbonyls)
CAC δc (125.7 MHz, CDC13) 172.65 (IC, C-2 carbonyl), 173.28 (2C, C-1, C-3 carbonyls)
C(DHLA)C δc (125.7 MHz, CDC13) 172.83 (IC, C-2 carbonyl), 173.30 (2C, C-1, C- 3 carbonyls)
GCG δc (125.7 MHz, CDC13) 172.91 (IC, C-2 carbonyl), 173.11 (2C, C-1, C-3 carbonyls)
OGO δc (125.7 MHz, CDC13) 172.69 (IC, C-2 carbonyl), 173.25 (2C, C-1, C-3 carbonyls)
AAA δc (125.7 MHz, CDC13) 172.66 (IC, C-2 carbonyl), 173.04 (2C, C-1, C-3 carbonyls)
CCC δc (125.7 MHz, CDC13) 172.81 (IC, C-2 carbonyl), 173.21 (2C, C-1, C-3 carbonyls)
Experimental Procedure
The proton-decoupled 13C NMR spectra with suppressed NOE were collected at 21 °C in a 5-mm broadband probe on a Joel 500 MHz spectrometer operating at 125.728 MHz. Waltz decoupling was the chosen mode of decoupling and was gated on only during the 14.89s acquisition time. The relaxation delay was set at 30 sees and the pulse angle was 90°. The spectral window used was ca.35 ppm (from 173.5 to 172.6 ppm) with a 170 ppm offset. The spectra were internally referenced to CDC1 at 77.0 ppm. Typically, the approximate number of scans collected for adequate signal-to- noise ranged from 300 to 1200 scans depending on the concentration and purity of the sample. The total acquisition time for the experiments ranged between 2-8h e.g
1272 scans; data points 65,536. Concentrated solutions up to 20% w/v were employed when possible to reduce the acquisition time The chemical shifts quoted vary with the concentration of the solution.
BIOLOGICAL STUDIES.
Chronic Relapsing Experimental Autoimmune Encephalomyelitis (CREAE)
Studies .
Induction and Clinical Assessment of EAE
CREAE was induced in C57B1/6 and SJL mice. Animals were injected subcutaneously with 100 μg of the neuroantigen peptide MOG 35-55 (amino acid sequence MEVGWYRSPFSRVVHLYRNGK Genemed Synthesis, hie) or 1 mg of mouse spinal cord homogenate (SCH), in phosphate buffered saline (PBS), emulsified by sonication for 10 min at room temperature, in incomplete Freund's adjuvant (DIFCO, Detroit, USA) supplemented with 480 μg of mycobacteria tuberculosis and 60μg of Mycobacteria butyricium (DIFCO, Detroit, USA) on days 0 and 7 as described previously (Morris-Downes, MM., et al 2002). In addition to optimise the disease mice also received 200 ng (intraperitoneally) of Bordetella pertussis toxin dissolved in PBS administered lhr and 24 hrs after immunization with the MOG neuroantigen and for SCH days 0, 1, 7 and 8. Animals were weighed from day 5 onwards and examined daily for clinical neurological signs by two experienced investigators and graded according to a previously validated grading scheme (Morris-Downes, MM. et al 2002 and others): 0 = normal; 1 = limp tail and feet; 2 = impaired righting reflex; 3 = partial hind limb paralysis; 4 = complete hindlimb paralysis; 5 = moribund; 6 = death. Animals exhibiting clinical signs of a lesser severity grade than typically observed were scored as 0.5 less than the indicated grade.
Reference
Morris-Downes, MM., et al (2002). Pathological and regulatory effects of anti-myelin antibodies in experimental allergic encephalomyehtis in mice. /. Neuroimmunol. 125. 114-124. The mean group EAE score was compared for each test group compared to a respective control group by non-parametric statistical analysis (Mann Whitney U Test). All MOG-CREAE studies comprised a treatment control group (C-C-C or saline as selected from the above study). Each structured lipid was tested at 3 dose levels, all treaments being orally administered for 2 weeks from day 7 after inoculation. All treatment groups will contained 10 animals. On completion of studies (day 21), brain and spinal cord were be removed and half of the samples were processed for signs of CNS perivascular mononuclear leucocyte-infiltrated sites and demyelination.
Studies were as follows:
Study 2: Spinal cord homogenate(SCH) EAE in SJL mice. EAE Induction: lmg SCH day 0 +day 7 sc. 200ng Pertussis toxin day 0,1, 7 & 8 ip.lOmice/group. Mice were treated from day 7 to 21 with CCC or CGC.
Study 3: SCH EAE in SJL mice: Treatment was from PSD 7 to 21, both days inclusive.
Study 4: MOG EAE in C57BL mice: Treatment was from PSD 7 to 21, both days inclusive.
Study 5: SCH EAE in SJL mice: Treatment was from PSD 5 to 18, both days inclusive.
Study 6: MOG EAE in C57BL mice: Treatment was from Days 5 to 21 inclusive except C-DHLA-C group where treatment was from days 5 to 15 inclusive. Animals were culled on PSD 25. [Five animals from an untreated group, 3 animals from control CCC treatment group, 5 animals from GGG 150ul treatment group and 2 animals from GGG 350ul treatment group were sampled for histological analysis on PSD 20].
Study 7: SCH EAE in SJL mice
Treatment was from Days 6 to 20 inclusive.
Study 2 - Spinal cord homogenate (SCH) in SJL mice :-tested CGC (50/150/350ul); CCC (350ul). GGG.(50/350ul) [Severe disease observed]
Study 3 - SCH/SJL mice:- tested CCC (50/150/ '50ul) CGC (25/50/150/350ul) GGG (50/150/350ul) OGO. (25/50/150/350ul) [Severe disease observed]
Study 4 - MOG/ C57BL mice:- tested CCC (50/150/350ul) CGC (25/50/150/350ul) GGG (50/150/35 Oul) OGO. (25/50/150/350ul)
Study 6 - MOG/C57BL mice:- tested CCC (150ul) C-DHLA-C (50ul) CAC (50/350ul) AAA (50/150ul) GCG (50ul) CGC (5 Oul) GGG.(150/350ul) [Pathology: CCC; GGG]
Histological examination of the submitted samples of brain and spinal cord showed lesions typical of experimental allergic encephalomyehtis. Localised and diffuse lesions were characterised by gliosis, myelin vacuolation, axonal degeneration and perivascular cuffing with lymphocytes, macrophages and neutrophils. Spinal cord lesions were mostly located in subpial white matter and brain lesions mostly occurred in the cerebellar white matter. Lesions were more severe in the spinal cords than in the brains and whereas all animals with brain lesions had lesions in the spinal cord, not all animals with cord lesions had lesions in the brain. Variation in the severity of changes between individual mice is summarised using a semi-quantitative five point grading system. Untreated mice had histological scores of 3-4 which correlated with EAE scores of 1.5-3. One mouse showed little pathological change with a zero score. In the GGG treated mice, the majority showed no abnormalities. Two mice from this group had histological scores of 2 and 3 respectively which correlated with EAE severity. scores of 1 and 1.5 The results of the four studies are shown in Figures 11 to 20 below These show that the compounds G-G-G, A- A- A, C-G-C, C-DHGLA-C, and
C-A-C are all capable of reducing severity of CREAE whereas compounds G-C-G and C-C-C failed to treat the condition. Compound O-G-O is believed to work if the dose is adjusted. As cautioned in the description, the arachidnoic acid compounds are effective, but lead to death of some animals. Surviving animals had much reduced disease. It is believed that the dose of these compounds may be reduced still further to provide survival with satisfactory treatment. Some of the studies show a bell shaped response curve for compounds C-G-C and G-G-G, suggesting that very high doses are not optimal, as set out above. Such dosing can be conveniently determined by those skilled art, eg. By dose escalatio and monitoring TGF-β 1/TNF-α spontaneously release ratio changes from PBMCs. Given the PCT/GB04/002089 high sn-2 γ-linolenic acid results, the lack of efficacy of low sn-2 black-current oil and G-C-G in CREAE and the low dose efficacy of C-G-C and C-DHGLA-C in Figure 20, it can be seen that sn-2 -γ-linolenic acid, dihomo-γ-linolenic acid and arachidonic acid lipids provide a novel treatment for MS that far exceeds any current therapy outcome in that lesions are repaired and difficult symptoms are resolved: decreasing EDSS over a period of years being so far unachieved in other treatments. References Amor S, Groome N, Linington C, Morris MM, Dornmair K, Gardinier MV, Matthieu JM, Baker D. Identification of epitopes of myelin oligodendrocyte glycoprotein for the induction of experimental allergic encephalomyehtis in SJL and Biozzi AB H mice. J Immunol. 1994 Nov 15;153(10):4349-56.
Beck J, Rondot P, Catinot L et al. Increased production of interferon gamma and tumor necrosis factor precedes clinical manifestation in multiple sclerosis: do cytokines trigger off exacerbations? ActaNeurol Scand 1988; 78:318-23.
' Bertolotto A, Capobianco M, Malucchi S et al. Transforming growth factor betal (TGFbetal) mRNA level correlates with magnetic resonance imaging disease activity in multiple sclerosis patients. Neurosci Lett 1999; 263:21-4. Bertolotto A, Malucchi S, Capobianco M et al. Quantitative PCR reveals increased levels of tumor necrosis factor-alpha mRNA in peripheral blood mononuclear cells of multiple sclerosis patients during relapses. J Interferon Cytokine Res 1999; 19:575- 81. Brosnan CF., Selmaj K and Raine CS. Hypothesis: a role for tumor necrosis factor in immune-mediated demyelination and its relevance to multiple sclerosis. J Neuroimmunol 1988:18,87-94.
Brosnan CF and Raine CS. Mechanisms of immune injury in multiple sclerosis. Brain Pathol. 1996:6,243-257.
Burns J, Bartholomew B, Lobo S. Isolation of myelin basic protein-specific T cells predominantly from the memory T-cell compartment in multiple sclerosis. Ann Neurol 1999; 45:33-9. Cannella B, Raine CS. The adhesion molecule and cytokine profile of multiple sclerosis lesions. Ann Neurol 1995; 37:424-35.
Chou YK, Bourdette DN, Offner H et al. Frequency of T cells specific for myelin basic protein and myelin proteolipid protein in blood and cerebrospinal fluid in multiple sclerosis. J Neuroimmunol 1992; 38:105-14.
De Stefano N., Narayanan S., Francis GS., Arnaoutelis R., Tartaglia MC, Antel JP., matthews PM and Arnold DL. Evidence of axonal damage in the early stages of multiple sclerosis and its relevance to disability. Arch Neurol. 2001: 58(1), 65-70.
Ewing C, Bernard CC. Insights into the aetiology and pathogenesis of multiple sclerosis. Immunol Cell Biol 1998; 76:47-54.
Fazakerly JK. Molecular biology of multiple sclerosis. Wiley and Sons Ltd. 1997,255-273.
Fredrikson S, Soderstrom M, Hillert J et al. Multiple sclerosis: occurrence of myelin basic protein peptide-reactive T cells in healthy family members. Acta Neurol Scand 1994; 89:184-9.
Genain CP., Cannella B., hauser SL and Raine CS. Identification of autoantibodies associated with myelin damage in multiple sclerosis. Nature Med 1999:5,170-175.
Gross CE, Bednar MM, Howard DB and Spom MB (1993) Transforming growth factor beta I reduces infarct size after experimental cerebral ischemia in a rabbit model .Stroke 24,558-562.
Harbige, LS, Crawford MA, Jones J, Preece AW and Forti A. Dietary intervention studies on the phosphoglyceride fatty acids and electrophoreitic mobility of erythrocytes in multiple sclerosis. Prog. Lipid Res 1986:25,243-248.
Harbige LS. Nutrition and immunity with emphasis on infection and autoimmune disease. (1996) Nutr Health, 10(4):285-312.
Harbige LS (1998) Dietary n-6 and n-3 fatty acids in immunity and autoimmue disea.se.Proceedings of the Nutrition Society 57, 555-562.
Harbige LS, Yeatman N, Amor S & Crawford MA (1995) Prevention of experimental autoimmune encephalomyehtis in Lewis rats by a novel source of y-linolenic acid. British Journal of Nutrition 74,701-715.
Harbige LS., Layward L., Morris-Downes MM., Dumonde DC and Amor S. The protective effects of omega-6 fatty acids in experimental autoimmune encephalomyehtis (EAE) in relation to transforming growth factor-beta 1 (TGF- betal) up-regulation and increased prostaglandin E2 (PGE2) production. Clin Exp Immunol 2000:122,445-452.
Henrich Noack P, Prehn JH, and Kriegistein J. (1996) TGF-beta I protects hippocampal neurons against degeneration caused by transient global ischaemia. Dose-response relationship and potential neuroprotective mechanisms. Stroke, 27, 1609-1614.
Hirsch RL, Panitch HS, Johnson KP. Lymphocytes from multiple sclerosis patients produce elevated levels of gamma interferon in vitro. J Clin Immunol 1985; 5:386-9.
Hollifield RD, Harbige LS, PhM-Dinh D, Sharief M. Evidence for cytokine Dysregulation in Multiple Sclerosis: Peripheral Blood Mononuclear cell production of pro-inflammmatory and anti-inflammatory cytokines during relapse and remission. Autoimmunity, 2003 36(3):133-141.
Imamura K, Suzumura A, Hayashi F et al. Cytokine production by peripheral blood monocytes/macrophages in multiple sclerosis patients. Acta Neurol Scand 1993; 87:281-5.
Issazadeh S, Lorentzen JC, Mustafa MI et al. Cytokines in relapsing experimental autoimmune encephalomyehtis in DA rats: persistent mRNA expression of promflammatory cytokines and absent expression of interleukin-10 and transforming growth factor-beta. J Neuroimmunol 1996; 69:103-15.
Johns LD, Sriram S. Experimental allergic encephalomyehtis: neutralizing antibody to TGF beta 1 enhances the clinical severity of the disease. J Neuroimmunol 1993; 47:1-7.
Kerlero de Rosbo N, Hoffman M, Mendel I et al. Predominance of the autoimmune response to myelin oligodendrocyte glycoprotein (MOG) in multiple sclerosis: reactivity to the extracellular domain of MOG is directed against three main regions. Eur J Immunol 1997; 27:3059-69.
Kerlero de Rosbo N, Milo R, Lees MB et al. Reactivity to myelin antigens in multiple sclerosis. Peripheral blood lymphocytes respond predominantly to myelin oligodendrocyte glycoprotein. J Clin Invest 1993; 92:2602-8.
Khalil N. TGF-beta: from latent to active. Microbes Infect 1999; 1:1255-63.
Krupinski J, Kumar P, Kumar S, and Kaluza J. (1996) Increased expression of TGF- beta I in brain tissue after ischemic stroke in humans. Stroke, 27, 852-857. Kuroda Y, Shimamoto Y. Human tumor necrosis factor-alpha augments experimental allergic encephalomyehtis in rats. J Neuroimmunol 1991; 34:159-64.
Lu CZ, Jensen MA, Arnason BG. Interferon gamma- and interleukin-4-secreting cells in multiple sclerosis. J Neuroimmunol 1993; 46:123-8.
Maimone D, Reder AT, Gregory S. T cell lymphokine-induced secretion of cytokines by monocytes from patients with multiple sclerosis. Cell Immunol 1993; 146:96-106.
Martino G, Hartung H-P. hnmunopathogenesis of multiple sclerosis: the role of T cells. Curr Opin Neurol 1999; 12:309-21.
McCarron RM, Wang L, Racke MK et al. Cytokine-regulated adhesion between encephalitogenic T lymphocytes and cerebro vascular endothelial cells. J Neuroimmunol 1993; 43:23-30.
McDonald WI, Compston A, Edan G, Goodkin D, Hartung HP, Lublin FD, McFariand HF, Paty DW, Pohnan CH, Reingold SC, Sandberg-Wollheim M, Sibley W, Thompson A, van den Noort S, Weinshenker BY, Wolinsky JS.
Recommended diagnostic criteria for multiple sclerosis: guidelines from the International Panel on the diagnosis of multiple sclerosis. Ann Neurol. 2001 Jul;50(l):121-7.
Merrill JE, Strom SR, Ellison GW et al. In vitro study of mediators of inflammation in multiple sclerosis. J Clin Immunol 1989; 9:84-96.
Merrill JE, Zimmerman RP. Natural and induced cytotoxicity of oligodendrocytes by microglia is inhibitable by TGF beta. Glia 1991; 4:327-31. Miyazono K, Hellman U, Wernstedt C et al. Latent high molecular weight complex of transforming growth factor beta 1. Purification from human platelets and structural characterization. J Biol Chem 1988; 263:6407-15.
Mokhtarian F, Shi Y, Shirazian D et al. Defective production of anti-inflammatory cytokine, TGF-beta by T cell lines of patients with active multiple sclerosis. J Immunol 1994; 152:6003-10.
Navikas V, Link H. Review: cytokines and the pathogenesis of multiple sclerosis. J Neurosci Res 1996; 45:322-33.
Noseworthy JH. Progress in determining the causes and treatment of multiple sclerosis. Nature 1999:399(6738 Suppl), A40-47.
Ota K, Matsui M, Milford EL et al. T-cell recognition of an immunodominant myelin basic protein epitope in multiple sclerosis. Nature 1990; 346:183-7.
Perkin GD, Wolinsky JS. Fast facts-Multiple Sclerosis, 1st Edn. Oxford, UK: Health Press, 2000.
Philippe J, Debruyne J, Leroux-Roels G et al. In vitro TNF-alpha, IL-2 and IFN- gamma production as markers of relapses in multiple sclerosis. Clin Neurol Neurosurg 1996; 98:286-90.
Phylactos AC, Ghebremeskel K, Costeloe K, Leaf AA, Harbige LS, Crawford MA. (1994) Polyunsaturated fatty acids and antioxidants in early development. Possible prevention of oxygen-induced disorders.Eur J Clin Nutr. 48Suppl 2:S17-23.
Prehn JH, Peruche B, Unsicker K and Kriegistein J. (1993) Isoform-specific effects of transforming growth factor-beta on degeneration of primary neuronal cultures induced by cytotoxic hypoxia or glutamate. J.Neurochem. 60,1665-1672.
Rack MK, Sriram S, Calrimi J, Cannella B, Raine CS & McFarim DE (1993) Long- term treatment of chronic relapsing experimental allergic encephalomyehtis by transforming growth factor-p2. Journal ofNeuroimmunology, 46, 175-183.
Racke MK, Cannella B, Albert P et al. Evidence of endogenous regulatory function of transforming growth factor-beta 1 in experimental allergic encephalomyehtis. h t Immunol 1992; 4:615-20.
Rieckmann P, Albrecht M, Kitze B et al. Cytokine mRNA levels in mononuclear blood cells from patients with multiple sclerosis. Neurology 1994; 44:1523-6.
Rieckmann P, Albrecht M, Kitze B et al. Tumor necrosis factor-alpha messenger RNA expression in patients with relapsing-remitting multiple sclerosis is associated with disease activity. Ann Neurol 1995; 37:82-8.
Ruddle NH, Bergman CM, McGrath KM et al. An antibody to lymphotoxin and tumor necrosis factor prevents transfer of experimental allergic encephalomyehtis. J Exp Med 1990; 172:1193-200.
Santambrogio L, Hochwald GM, Saxena B, Leu CH, Martz JE, Carlino JA, Ruddle NH, Palladino MA, Gold LI & Thorbecke GJ (1993) Studies on the mechamsms by which Transforming Growth Factor-p protects against allergic encephalomyehtis. Journal of Immunology 151, 1116-1127.
Schiefer HB, Hancock DS, Loew FM. Long-term effects of partially hydrogenated herring oil on the rat myocardium. Drug Nutr Interact. 1982;1(2):89-102. Schluesener HJ, Lider O. Transforming growth factors beta 1 and beta 2: cytokines with identical immunosuppressive effects and a potential role in the regulation of autoimmune T cell function. J Neuroimmunol 1989; 24:249-58.
Selmaj K, Raine CS, Cannella B et al. Identification of lymphotoxin and tumor necrosis factor in multiple sclerosis lesions. J Clin Invest 1991; 87:949-54.
Selmaj K, Raine CS, Farooq M et al. Cytokine cytotoxicity against oligodendrocytes. Apoptosis induced by lymphotoxin. J Immunol 1991; 147:1522-9.
Sharief MK, Thompson EJ. In vivo relationship of tumor necrosis factor-alpha to blood-brain barrier damage in patients with active multiple sclerosis. J Neuroimmunol 1992; 38:27-33.
Tej ada-Simon MV, Hong J, Rivera VM et al. Reactivity pattern and cytokine profile of T cells primed by myelin peptides in multiple sclerosis and healthy individuals. Eur J Immunol 2001; 31:907-17.
Vartanian T, Li Y, Zhao M et al. Interferon-gamma-induced oligodendrocyte cell death: implications for the pathogenesis of multiple sclerosis. Mol Med 1995; 1:732- 43.
Vivien D, Bemaudin M, Buisson A, Divoux D, MacKenzie ET and Nouvelot A.(1998) Evidence of type I and type II transforming growth factor-beta receptors in central nervous tissues: changes induced by focal cerebral ischemia. J. Neurochem. 70,2296-2304.
Zhang J, Markovic-Plese S, Lacet B et al. Increased frequency of interleukin 2- responsive T cells specific for myelin basic protein and proteolipid protein in peripheral blood and cerebrospinal fluid of patients with multiple sclerosis. J Exp Med 1994; 179:973-84.
Japanese Patent 6172263 (1994) Y. Kosugi et al, Agency of Industrial Science & Technology High-purity arachidonic acid triglyceride and its production.
US Patent 4,888,324 (1989) N. Catsimpoolas et al, Angio-Medical Corporation Method for enhancing angiogenesis with lipid containing molecules.
Y. Kosugi and N. Azuma, JAmer. Oil Chem. Soc, 71, 1397-1403 (1994).
Synthesis of Triacylglycerol from polyunsaturated fatty acid by immobilized ipase.
J. W. Hageman et al, J. Amer, Oil Chem.Soc, 49, 118-xxx (1972) Preparation of glycerin and their uses.
E. S. Lutton and A. J. Fehl, Lipids, 5, 90-99 (1970).
The polymorphism of odd and even saturated single acid triglycerides, C8-C22.
D. Horrobin, A. McMordie, M. S. Manku (Scotia Holdings PLC UK) Eur. Pat. Appl EP 609078 3 Aug 1994. Phospholipids containing two different unsaturated fatty acids for use in therapy, nutrition, and cosmetics.
Y.-S. Huang, X. Lin, P. R. Redden and D. F. Horrobin, J. Am. Oil Chem. Soc,
72,625-631,(1995). In vitro Hydrolysis of Natural and Synthetic γ-Linolenic Acid- Containing Triacylglycerols by Pancreatic Lipase
K. Osada, K. Takahashi, M. Hatano and M. Hosokawa, Nippon Suisan Gakkaishi., 57, 119-125 ( 1991). Chem. Abstr., 115:278299 Molecular Species of Enzymically-synthesized Polyunsaturated Fatty acid-rich Triglycerides. J.-W. Liu, S. DeMichele, M. Bergana, E.. Bobik, Jr., C. Hastilow, Lu-Te
Chuang, P. Mukerji and J.-S. Huang., J. Am. Oil Chem. Soc, 78, 489-
493 (2001) Characterization of Oil Exhibiting High γ-Linolenic Acid from a Genetically transformed Canola Strain.
D. R. Kodali, D. Atkinson, T. G. Redgrave and D. Small, J. Lipid Res., 28, 403-413 (1987).Structure and polymorphism of 18-Carbon Fatty Acid Triacylglycerols: Effect of Unsaturation and Substitution in the 2-Position
P. H. Bentley and W. McCrae, J. Org. Chem. 35, 2082-2083 (1970) An Efficient Synthesis of Symmetrical 1,3-Diglycerides.
M. Berger, K. Laumen and M. P. Schneider, J. Am. Oil. Chem. Soc, 69, 955- 959,(1992). Enzymatic Esterification of Glycerol 1. Lipase-Catalyzed Synthesis of
Regioisomerically Pure 1,3-sn-Diacylglycerols.
A. P. J. Mank, J. P. Ward and D. A. van Dorp, Chem. Physics Lipids, 16, 107-114 (1976). A versatile, flexible synthesis of 1,3-diglycerides and triglycerides.
L. Hartman, Chem. Rev., 58, 845-867 (1958) and references therein. Advances in the Synthesis of Glycerides of Fatty Acids
TABLE 1 Compositional (% Total FAs) Characteristics of Various Oils and their Protective Effects in EAE
Treatment 18:2n-6 18:3n-6 18:2n-6/18:3n-6 18:1 n-9 INCIDENCE OF EAE
C CD FGO 17 20 0.6 35 0/10 CΛ
BOO 37 24 1.5 15 3/10 m
CΛ ∞
I m EPO 71 9.4 7.5 7/10 m
73 c SAF 66 17 9/10 m r σ> Controls 9/10
FGO, Fungal Oil; BOO, Borage Oil; EPO, Evening Primrose Oil, SAF, Safflower Oil.
TABLE 2 Treatment Groups- PCT/GB04/002089 Borage oil-MS trial
C
ΓJΓJ CΛ
I m m
73 c m r σ>
Figure imgf000061_0001
Table 3. Molecular Species Comparison of Triacylglveerol-GLA (TG-GLA), Ethyl-Ester-GLA f EE-GLA) and PCT/GB04/002089 Boraao Officinalis O»-GLA (BOR-GLA) in MOG-induced CREAE in SJL Mice
Treatment No. with EAE Mean Clinical Score
Control 10/11 33±13
EE-GLAa 5/6 3.Q±0.8
TG-GLA3 3/6 l.G±13c
BOR-GLAb 3/6 1.0±1.2C
1 Animals given 100 μl of test lipid; b25Qμl BOR-GLA given. Significance of difference compared with controls, c p <0.05
F:\500\l 34\WO01 Spec\040916b.doc
Table 4. Effect of enriched black-currant seed oil (73% GLA) on the incidence of EAE
C % Incidence of EAE (Days after immunisation)
ΓJΓJ CΛ 13 17 21
C
H Controls (n=10) 60 90 10 m *
CΛ ^
I m Blackcurrant (n=10) 10 80 70 m
H 10 c m Note: Blackcurrant oil delays the incidence but does not provide full protection. Animals were fed 7 days after sensitization (immunisation). r σ>
15

Claims

CLAIMS.
1. A method of treating a patient in need of therapy for a neurodegenerative disease comprising administering to that patient a therapeutically effective dose of a defined structure lipid glyceride comprising a glycerol moiety esterifed with one or more fatty acid moieties, characterised in that the lipid has a fatty acid moiety at the sn-2 position selected from the group consisting of γ-linolenic acid, dihomo-γ-linolenic acid and arachidonic acid
2. A method as claimed in Claim 1 wherein the neurodegenerative disease involves demyelination.
3. A method as claimed in Claim 1 wherein the treatment specifically arrests underlying neurodegeneration and restores neuronal function.
4. A method as claimed in Claim 1 which normalises neuronal membrane composition with respect to γ-linolenic acid, dihomo-γ-linolenic acid and arachidonic acid lipid content.
5. A method as claimed in Claim 1 which restores healthy TGF-β 1/TNF ratios as measured from spontaneous release from peripheral blood mononuclear cell release.
6. A method as claimed in Claim 1 wherein the disease is multiple sclerosis.
7. A method as claimed in Claim 1 wherein the disease is relapsing remitting multiple sclerosis, primary progressive multiple sclerosis or chronic progressive multiple sclerosis.
8. A method as claimed in Claim 1 wherein the disease is multiple sclerosis and the treatment restores, in part or completely, neuronal function or neuronal integrity as measured by one or more of MRI scan, CAT scan or by EDSS score.
9. A method as claimed in Claim 1 wherein the treatment is of cerebral impairment after stroke, head trauma and intracranial bleeding, Alzheimer's disease or Parkinson's disease where there is demyelination or neuronal damage.
10. A method as claimed in Claiml wherein the lipid is administered for a duration and at a dose sufficient to maintain or elevate TGF-βl levels in the patient to therapeutic levels.
11. A method as claimed in Claim 1 wherein the lipid is administered for a duration and at a dose sufficient to maintain or elevate TGF-βl levels in the patient to a TGF-β 1/TNF-α ratio released spontaneously from peripheral blood mononuclear cells isolated from the blood of a patient, after 18 months of daily dosing, of 0.4 to 3.0, at least 0.5, more preferably at least 0.75 and most preferably at least 1.
12. A method as claimed in Claiml 1 wherein the dose is such as to produce a TGF-β 1/IL-lβ ratio in PBMCs isolated from blood of a patient, after 18 months of daily dosing, of at least of at least 0.75.
13. A method as claimed in Claim 1 wherein the amount of lipid administered is between 0.5 and 30 grams, typically 3 to 5 grams, per day.
14. A method as claimed in Claim 1 wherein the lipid is a mono glyceride, diglyceride or triglyceride containing the at least one sn-2 γ-linolenic acid, dihomo-γ- linolenic acid or arachidonic acid moiety, the lipid being of of general Formula I
Figure imgf000066_0001
Formula I
wherein R1 and R3 are independently selected from hydrogen and acyl groups, and R2 is selected from the group consisting of γ-linolenic acid, dihomo-γ-linolemc acid and arachidonic acid residues having their carbonyl carbon attached to the oxygen of the glycerol moiety.
15. A method as claimed in Claim 1 wherein R1 and R3 are saturated fatty acid moieties of formula -CO-(CH2)n-CH3, wherein n is an integer selected from 1 to 22,
16. A method as claimed in Claim 15 wherein R1 and R3 are the same and n is an integer of from 5 to 12.
17. A method as claimed in Claim 16 wherein n is an integer of from 6 to 10.
18. A method as claimed in Claim 14 wherein R1 and R3 are selected from the group consisting of essential fatty acids or physiologically acceptable fatty acids metabolisable by the human body.
19. A method as claimed in Claim 14 wherein R1, R2 and R3 are all the same and are selected from the group consisting of of γ-linolenic acid, dihomo-γ-linolenic acid and arachidonic acid residues.
20. A pharmaceutial composition characterised in that it comprises a defined structure lipid glyceride comprising a glycerol moiety esterifed with one or more fatty acid moieties, characterised in that the lipid has a fatty acid moiety at the sn-2 position selected from the group consisting of γ-linolenic acid, dihomo-γ-linolenic acid.
20. A pharmaceutial composition for treating neurodegeneration characterised in that it comprises a defined structure lipid glyceride comprising a glycerol moiety esterifed with one or more fatty acid moieties, characterised in that the lipid has a fatty acid moiety at the sn-2 position selected from the group consisting of γ-linolenic acid, dihomo-γ-linolenic acid and arachidonic acid.
21. A pharmaceutical composition for treating demyelinating disease comprising a glycerol moiety esterifed with one or more fatty acid moieties, characterised in that the lipid has a fatty acid moiety at the sn-2 position selected from the group consisting of γ-linolenic acid, dihomo-γ-linolenic acid and arachidonic acid.
22. A lipid of formula II
Figure imgf000067_0001
wherein R1 and R3 are the same and are -C(O)(CH2)nCH wherein n is selected from 4 to 14, more preferably 6 to 10 and most preferably 7, 8 or 9 and R2 is selected from γ-linolenyl, dihomo-γ-linolenyl and arachidonyl residues.
23. Use of a defined structure lipid glyceride comprising a glycerol moiety esterifed with one or more fatty acid moieties, characterised in that the lipid has a fatty acid moiety at the sn-2 position selected from the group consisting of γ-linolenic acid, dihomo-γ-linolenic acid and arachidonic acid, for the manufacture of a medicament for the treatment of neurodegenerative disease
24. Use as claimed in Claim 23. wherein the degenerative disease is a demyelinating disease.
25. Use as claimed in Claim 23 wherein the disease is multiple sclerosis.
26. Use as claimed in Claim 23 wherein the medicament normalises neuronal membrane composition with respect to lipid γ-linolenic acid, dihomo-γ-linolenic acid and arachidonic acid levels.
27. Use as claimed in Claim 23 wherein the medicament restores TGF-β 1/TNFα ratios spontaneously released from peripheral blood mononuclear cells of a patient to healthy levels.
28. Use as claimed in Claim 23 wherein treatment is for multiple sclerosis or the degenerative sequelae associated with head trauma, stroke and intracranial bleeds or neuronal damage caused by Alzheimer's or Parkinson's disease.
29. Use as claimed in Claim 23 wherein the medicament repairs CNS lesions.
30. Use as claimed in Claim 23 wherein the medicament relieves muscle spasticity and/or pain.
31. Use as claimed in Claim 23 wherein the medicament eliminates relapses.
32. Use as claimed in Claim 23 wherein the medicament improves EDSS score by at least 1 unit over a period of 1 years treatment.
33. Use as claimed in Claim 23 wherein the medicament is sufficient to restore EDSS of a patient with EDSS above 2.5 to below 2 over a period of 1 years treatment.
34. Use as claimed in Claim 23 wherein there is improvement in bladder control.
35. A method for synthesis of a compound of general formula III
Figure imgf000069_0001
wherein R1 and R3 are the same and are -C(O)(CH2)nCH3 wherein n is selected from 4 to 14, more preferably 6 to 10 and most preferably 1, 8 or 9 and R is γ- linolenyl residue, dihomo-γ-linolenyl residue or arachidonyl residue comprising reacting 1 ,3-dihydroxyacetone with a compound of formula X-C(O)(CH2)nCH3 wherein X is selected from Cl, Br and I, to give the corresponding l,3-di-( C(O)(CH2)nCH3) 2-keto compound reducing the keto group to the corresponding l,3-di-( C(O)(CH )nCH3) 2-ol and reacting that with γ-linolenyl halide or dihomo-γ-linolenyl halide or arachidonyl halide, wherein halide is chloride, bromide or iodide.
36. A method for synthesis of a compound of general formula IV
Figure imgf000070_0001
wherein R1 to R3 are the same and selected from γ-linolenyl residue, dihomo-γ- linolenyl residue or arachidonyl residue comprising reacting the corresponding γ-linolenyl halide, dihomo-γ-linolenyl halide or arachidonyl halide, wherem halide is chloride, bromide or iodide, with glycerol
37. A lipid selected from the group consisting of Glycerol 1,3-didecanoate- 2-octadecatri(6-Z,9-Z,12-Z)enoate Glycerol l,3-didecanoate-2- eicosa-(8Z,llZ,14Z)-trienoate Glycerol trieicosotetra5-Z,8-Z,ll-Z,14Z-eneoate
38. A lipid as claimed in Claim 37 for use in therapy.
PCT/GB2004/003524 2003-08-18 2004-08-13 Treatment of neurodegenerative conditions WO2005018632A1 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
GB0603646A GB2422373B (en) 2003-08-18 2004-08-13 Treatment of neurodegenerative conditions
JP2006523678A JP2007502805A (en) 2003-08-18 2004-08-13 Treatment of neurodegenerative conditions
AU2004266480A AU2004266480B2 (en) 2003-08-18 2004-08-13 Treatment of neurodegenerative conditions
CH00249/06A CH698539A8 (en) 2003-08-18 2004-08-13 Compounds with unsaturated fatty acid residues for the preparation of medicaments for the treatment of multiple sclerosis.
EP04768085A EP1660071A1 (en) 2003-08-18 2004-08-13 Treatment of neurodegenerative conditions
DE112004001520T DE112004001520B4 (en) 2003-08-18 2004-08-13 Use of a lipid glyceride of specified structure for the manufacture of a medicament for the treatment of multiple sclerosis
CA2534202A CA2534202C (en) 2003-08-18 2004-08-13 Lipid glycerides for the treatment of neurodegenerative conditions involving demyelination
CN200480030644XA CN1867328B (en) 2003-08-18 2004-08-13 Use of lipid glyceride in preparing medicine for treatment of neurodegenerative conditions
US10/567,778 US7964641B2 (en) 2003-08-18 2004-08-13 Treatment of neurodegenerative conditions
IS8246A IS8246A (en) 2003-08-18 2006-01-19 Treatment of neurodegenerative conditions
FI20060154A FI20060154A (en) 2003-08-18 2006-02-17 Treatment of neurogenerative conditions
NO20061232A NO20061232L (en) 2003-08-18 2006-03-17 Treatment of neurodegenerative conditions
US12/654,780 US20100113810A1 (en) 2003-08-18 2009-12-31 Treatment of neurodegenerative conditions
US12/654,779 US20100113595A1 (en) 2003-08-18 2009-12-31 Treatment of neurodegenerative conditions
US13/363,705 US20120142775A1 (en) 2003-08-18 2012-02-01 Treatment of neurodegenerative conditions

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB0319358.8 2003-08-18
GB0319358A GB0319358D0 (en) 2003-08-18 2003-08-18 Treatment of neurodegenerative conditions
GB0410846A GB0410846D0 (en) 2004-05-14 2004-05-14 Treatment of neurodegenerative conditions
GB0410846.0 2004-05-14

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/654,780 Division US20100113810A1 (en) 2003-08-18 2009-12-31 Treatment of neurodegenerative conditions
US12/654,779 Continuation US20100113595A1 (en) 2003-08-18 2009-12-31 Treatment of neurodegenerative conditions

Publications (1)

Publication Number Publication Date
WO2005018632A1 true WO2005018632A1 (en) 2005-03-03

Family

ID=34219616

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2004/003524 WO2005018632A1 (en) 2003-08-18 2004-08-13 Treatment of neurodegenerative conditions

Country Status (13)

Country Link
US (4) US7964641B2 (en)
EP (2) EP2324828A1 (en)
JP (1) JP2007502805A (en)
AU (1) AU2004266480B2 (en)
CA (1) CA2534202C (en)
CH (1) CH698539A8 (en)
DE (1) DE112004001520B4 (en)
ES (1) ES2304094B2 (en)
FI (1) FI20060154A (en)
GB (1) GB2422373B (en)
IS (1) IS8246A (en)
NO (1) NO20061232L (en)
WO (1) WO2005018632A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006092623A1 (en) * 2005-03-02 2006-09-08 Btg International Limited Cytokine modulators using cyclic glycerides of essential polyunsaturated fatty acids
WO2006092622A1 (en) * 2005-03-02 2006-09-08 Btg International Limited Treatment of cytokine dysregulation by using sn-2 gamma-linolenoyl, gamma-dihomolinolenoyl or arachidonoyl patty acid glycerol monoesters
US20080004909A1 (en) * 2005-11-30 2008-01-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Computational systems related to nutraceuticals
WO2008081989A1 (en) * 2006-12-28 2008-07-10 Suntory Holdings Limited Nerve regeneration agent
US7935729B2 (en) 2003-05-14 2011-05-03 Btg International Limited Use of triglyceride oils containing γ-linolenic acid residues and linoleic acid residues for the treatment of neurodegenerative disease
US7964641B2 (en) 2003-08-18 2011-06-21 Btg International Limited Treatment of neurodegenerative conditions
US8202907B2 (en) 2004-09-17 2012-06-19 Suntory Holdings Limited Composition with preventive or improvement effect on symptoms or diseases associated with stress-induced behavior disorders
US8372812B2 (en) 2009-02-26 2013-02-12 Aker Biomarine Asa Phospholipid and protein tablets
US8697138B2 (en) 2007-03-28 2014-04-15 Aker Biomarine As Methods of using krill oil to treat risk factors for cardiovascular, metabolic, and inflammatory disorders
US8793141B2 (en) 2005-11-17 2014-07-29 The Invention Science Fund I, Llc Assistance related to health
US9028877B2 (en) 2007-03-28 2015-05-12 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US9168241B2 (en) 2005-06-30 2015-10-27 Suntory Holdings Limited Compositions ameliorating a reduced diurnal activity and/or depressive symptoms
US9867856B2 (en) 2014-01-10 2018-01-16 Aker Biomarine Antarctic As Phospholipid compositions and their preparation
US10042980B2 (en) 2005-11-17 2018-08-07 Gearbox Llc Providing assistance related to health
US10296720B2 (en) 2005-11-30 2019-05-21 Gearbox Llc Computational systems and methods related to nutraceuticals
US10456412B2 (en) 2015-02-11 2019-10-29 Aker Biomarine Antarctic As Lipid extraction processes
US10704011B2 (en) 2013-06-14 2020-07-07 Aker Biomarine Antarctic As Lipid extraction processes
US10744146B2 (en) 2001-08-02 2020-08-18 Suntory Holdings Limited Composition having effects of preventing or ameliorating conditions or diseases caused by brain hypofunction
US10864223B2 (en) 2015-02-11 2020-12-15 Aker Biomarine Antarctic As Lipid compositions

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2535966C2 (en) * 2008-09-12 2014-12-20 Криопрашис Криобиоложия Лтда. Cell-based therapy of ischemic tissue
TR201808471T4 (en) 2010-07-05 2018-07-23 Beth Israel Deaconess Medical Ct Inc Sn-2-monoacylglycerols and lipid malabsorption.
JP2016023163A (en) * 2014-07-22 2016-02-08 出光興産株式会社 AGENT FOR REDUCING OXIDATIVE STRESS ON NERVE CELLS COMPRISING γ-LINOLENIC ACID
CN105468156A (en) * 2015-11-25 2016-04-06 新乡医学院 Novel asynchronous brain-computer interface system based on alpha wave control
KR102054401B1 (en) * 2018-03-26 2019-12-10 주식회사 엔지켐생명과학 1,2-diacylglycerol compound, method for preparing the same and immunomodulating agent including the same as active ingredient
CN113164377A (en) * 2018-10-18 2021-07-23 Ds生物制药有限公司 DGLA and/or 15-HETRE for the treatment of inflammatory, fibrotic and proliferative conditions

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4701469A (en) * 1983-04-15 1987-10-20 Roussel Uclaf Triglycerides, process for therapeutical applications and compositions containing them
WO2001097793A2 (en) * 2000-06-23 2001-12-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem 2-arachidonylglycerol (2-ag) - an inhibitor of tumor necrosis factor -alfa and neuroprotector of brain in closed head injury
WO2003013497A1 (en) * 2001-08-02 2003-02-20 Suntory Limited Compositions having effects of preventing or ameliorating conditions or diseases caused by brain hypofunction

Family Cites Families (132)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2077371A (en) 1937-04-13 Synthetic drx
US655579A (en) * 1900-02-27 1900-08-07 Draper Co Lease-forming mechanism.
US2617791A (en) 1949-09-15 1952-11-11 Trojan Powder Co Recovery of valuable products from pentaerythritol mother liquor
US3158541A (en) 1959-12-18 1964-11-24 Escambia Chem Corp Product for reduction of blood cholesterol concentration
US3082228A (en) 1959-12-18 1963-03-19 Escambia Chem Corp Method for producing monoesters of polyunsaturated fatty acids
FR2003425A1 (en) 1968-03-07 1969-11-07 Unilever Nv EDIBLE PLASTIC FAT COMPOSITIONS AND PROCESS FOR PREPARING THEM
US3558656A (en) 1968-04-19 1971-01-26 Smith Kline French Lab Glycerol trichloroethyl carbonate and derivatives
US3671563A (en) 1968-04-19 1972-06-20 Smith Kline French Lab Glycerol 3-(2,2,2-trichloroethyl) carbonate
US3676472A (en) 1969-07-28 1972-07-11 American Home Prod Certain linoleic and linolenic acid ester fractions of vegetable oils and derivatives thereof
US3748348A (en) 1970-07-27 1973-07-24 Lever Brothers Ltd Directed-interesterified glyceridic oils having a high linoleic acid content and process for their production
US3671557A (en) 1970-09-17 1972-06-20 Smith Kline French Lab 1,2-diacylglycerol 3-(2,2,2-trichloroethyl) carbonates
GB1370021A (en) 1971-03-25 1974-10-09 Unilever Ltd Process for preparing usaturated carboxylic acids
US3855254A (en) 1972-03-31 1974-12-17 Lever Brothers Ltd Interesterification process
GB1506563A (en) 1974-04-25 1978-04-05 Williams J Immunosuppressive agents
US4058594A (en) * 1974-04-25 1977-11-15 John Williams Immuno-suppressive agents
US3988446A (en) 1974-11-07 1976-10-26 Abbott Laboratories Glycerides with anti-inflammatory properties
US3972907A (en) 1975-03-24 1976-08-03 G. D. Searle & Co. Anti-hyperlipidemic fatty acids and esters
US4048202A (en) 1975-04-11 1977-09-13 G. D. Searle & Co. 3-O-Alkanoylglyceric acids
US4178299A (en) 1978-03-27 1979-12-11 Abbott Laboratories Process for preparing 1,3-diacyl glycerols
DE2967049D1 (en) * 1978-04-11 1984-07-19 Efamol Ltd Pharmaceutical and dietary composition comprising gamma-linolenic acids
US4181670A (en) 1978-12-11 1980-01-01 G. D. Searle & Co. Phenyl 5Z,8Z,11Z,14Z-eicosatetraenoate and congeners
EP0092076B1 (en) 1982-04-16 1986-12-17 Societe Des Produits Nestle S.A. Lipid composition for oral, enteral or parenteral feeding
US4607052A (en) 1983-04-15 1986-08-19 Roussel-Uclaf Triglycerides, dietetic and therapeutical applications and compositions containing them
US4701468A (en) * 1983-04-15 1987-10-20 Roussel-Uclaf Oxidized triglycerides having therapeutic utility
GB8404463D0 (en) 1984-02-21 1984-03-28 Efamol Ltd Microbiological production of essential fatty acids
AT383130B (en) 1984-05-15 1987-05-25 Chemie Linz Ag METHOD FOR THE PRODUCTION OF PHOSPHATIDYLCHOLINES AND PHOSPHATIDYLETHANOLAMINES SUBSTITUTED DIFFERENTLY AT C1 AND C2 OVER THE NEW COMPOUNDS 1-0-TRITYLGLYCEROPHOSPHOCHOLIN OR RELATED (1-0, N-DITYHOHOLINE)
US5077312A (en) 1984-07-08 1991-12-31 Oncogen Biologically active lipids binding membrane receptors
GB2178752B (en) 1985-07-12 1989-10-11 Unilever Plc Substitute milk fat
US4888324A (en) 1985-10-01 1989-12-19 Angio-Medical Corporation Method for enhancing angiogenesis with lipid containing molecules
GB8524275D0 (en) 1985-10-02 1985-11-06 Efamol Ltd Pharmaceutical & dietary compositions
US5151291A (en) 1985-12-27 1992-09-29 Nisshin Flour Milling Co., Ltd. Glycerides of eicosapentaenoic acid, processes for preparing the same and oil and fat products containing the same
US5306730A (en) 1986-02-03 1994-04-26 Kabushiki Kaisha Yakult Honsha Botulinum toxin neutralizer
US5227403A (en) 1986-10-01 1993-07-13 The Nisshin Oil Mills, Ltd. Fats and oils having superior digestibility and absorptivity
US4867965A (en) 1986-10-02 1989-09-19 Revlon, Inc. Fatty acid diesters
FR2617161B1 (en) 1987-06-29 1989-10-27 Azar Robert NOVEL UNSATURATED FATTY ACID GLYCERIDES AND PROCESS FOR OBTAINING SAME
US4832975A (en) 1987-09-29 1989-05-23 The Procter & Gamble Company Tailored triglycerides having improved autoignition characteristics
DK565288D0 (en) * 1988-10-11 1988-10-11 Novo Industri As PROCEDURE FOR THE PREPARATION OF TRIGLYCERIDES, APPLICATION OF SUCH TRIGLYCERIDES, AND AN EMULSION CONTAINING SUCH TRIGLYCERIDES
US5008126A (en) 1989-06-27 1991-04-16 Nabisco Brands, Inc. Long chain diol diesters as low calorie fat mimetics
GB9026648D0 (en) 1990-12-07 1991-01-23 Efamol Holdings Nutrition
US5922345A (en) 1990-12-07 1999-07-13 Scotia Holdings Plc Nutrition
US5658767A (en) 1991-01-24 1997-08-19 Martek Corporation Arachidonic acid and methods for the production and use thereof
PH11992043811B1 (en) 1991-01-24 2002-08-22 Martek Corp Arachidonic acid and methods for the production and use thereof
IT1247165B (en) 1991-03-15 1994-12-12 Fidia Spa THERAPEUTIC USE OF PHOSPHATIDYLSERINE AND DERIVATIVES IN DEGENERATIVE PATHOLOGIES, ALSO ASSOCIATED WITH IMMUNITARY DYSFUNCTIONS.
JPH04328199A (en) 1991-04-26 1992-11-17 Nisshin Oil Mills Ltd:The Concentration of alpha-linolenic acid-containing triglyceride
GB9111900D0 (en) 1991-06-03 1991-07-24 Efamol Holdings Fatty acid compositions
US5674901A (en) 1995-06-01 1997-10-07 Wisconsin Alumni Research Foundation Methods of treating animals to maintain or increase CD-4 and CD-8 cell populations
JPH05310638A (en) 1992-05-01 1993-11-22 Idemitsu Petrochem Co Ltd Production of unsaturated fatty acid triglyceride
JPH06172263A (en) 1992-08-14 1994-06-21 Agency Of Ind Science & Technol High-purity arachidonic acid triglyceride and its production
US5618955A (en) 1992-11-30 1997-04-08 Yissum Research Development Company Of The Hebrew University Of Jerusalem Fatty acid derivatives and pharmaceutical compositions containing same
WO1994015464A1 (en) 1993-01-15 1994-07-21 Abbott Laboratories Structured lipids
GB9301446D0 (en) 1993-01-26 1993-03-17 Scotia Holdings Plc Internal radiation damage
GB9301629D0 (en) 1993-01-27 1993-03-17 Scotia Holdings Plc Formulations containing unsaturated fatty acids
JPH06279311A (en) 1993-03-26 1994-10-04 Sagami Chem Res Center Activation agent for protein kinase c isozyme
JPH0741421A (en) 1993-05-28 1995-02-10 Suntory Ltd Preventive agent or improver for medical symptom using leukotriene b4 (ltb4)
US20050027004A1 (en) 1993-06-09 2005-02-03 Martek Biosciences Corporation Methods of treating senile dementia and Alzheimer's diseases using docosahexaenoic acid and arachidonic acid compositions
US5663450A (en) 1993-08-17 1997-09-02 Cv Therapeutics Macrophage lipid chemoattractant
EP0734723A4 (en) 1993-12-29 2001-04-11 Kowa Tekuno Sachi Co Ltd Therapeutic composition for hyperparathyroidism of patient subjected to artificial dialysis
JPH07309773A (en) 1994-05-16 1995-11-28 Sagami Chem Res Center Acetylcholine release promoter
FR2722410B1 (en) 1994-07-15 1996-10-04 Grinda Jean Robert PROCESS FOR THE STABILIZATION OF POLYUNSATURATED FATTY ACIDS AND THE USE OF THESE STABILIZED ENTHERAPEUTIC PRODUCTS
JP3770628B2 (en) 1994-08-09 2006-04-26 サントリー株式会社 Prevention and amelioration of medical symptoms through delayed allergic reaction
EP0707850A1 (en) 1994-09-21 1996-04-24 Scotia Holdings Plc Use of polyunsaturated fatty acids for the manufacture of a medicament for the treatment of brest pain
EP0711503A3 (en) 1994-11-14 1997-11-26 Scotia Holdings Plc Milk fortified with GLA and/or DGLA
US5990163A (en) 1995-01-13 1999-11-23 The Salk Institute For Biological Studies Selective modulation of processes mediated by retinoid X receptors, and compounds useful therefor
US6410078B1 (en) 1995-04-28 2002-06-25 Loders-Croklaan B.V. Triglycerides, rich in polyunsaturated fatty acids
US6765020B2 (en) 1995-08-07 2004-07-20 Suntory Limited Drugs for prevention and treatment of diseases caused by abnormalities in cartilage tissues
GB9519661D0 (en) 1995-09-27 1995-11-29 Scotia Holdings Plc Fatty acid treatment
US6015798A (en) 1995-10-10 2000-01-18 Colgate Palmolive Company Method for reducing the damaging effects of radiation therapy on animal skin and mucosa
US5776913A (en) 1995-10-10 1998-07-07 Colgate Palmolive Company Therapeutic diet for metabolic abnormalities found in animals with lymphoma
WO1997038688A1 (en) 1996-04-12 1997-10-23 Peptide Technology Pty. Limited Methods of treating immunopathologies using polyunsaturated fattyacids
US5753702A (en) 1996-05-22 1998-05-19 University Of Vermont Arachidonic acid metabolite, 16-hete
CA2271166C (en) 1996-06-03 2006-08-29 Croda International Plc Compositions and uses thereof
US6340485B1 (en) 1996-06-03 2002-01-22 Croda International Plc Compositions and uses thereof
CA2259330C (en) 1996-06-29 2003-03-18 The Scottish Agricultural College Improvement of male fertility with antioxidants and/or polyunsaturated fatty acids
GB9617847D0 (en) 1996-08-27 1996-10-09 Scotia Holdings Plc Fatty acid treatment
CZ293704B6 (en) 1996-10-11 2004-07-14 Scarista Limited Pharmaceutical preparation for the treatment of schizophrenia and/or tardive dyskinesia comprising eicosapentaenoic acid and/or stearidonic acid as well as process for preparing a medicament for treating schizophrenia and/or tardive dyskinesia
US6080787A (en) 1997-02-21 2000-06-27 Abbott Laboratories Methods for reducing the incidence of necrotizing enterocolitis
DK1039892T3 (en) 1997-02-21 2004-08-30 Univ Tennessee Res Foundation Use of polyunsaturated fatty acids to reduce the incidence of necrotizing enterocolitis
US6201022B1 (en) 1997-03-27 2001-03-13 Myorx, Inc. Methods for treating neurotransmitter-mediated pain syndromes by topically administering an omega fatty acid
US6051754A (en) 1997-04-11 2000-04-18 Abbott Laboratories Methods and compositions for synthesis of long chain poly-unsaturated fatty acids in plants
US6432684B1 (en) 1997-04-11 2002-08-13 Abbott Laboratories Human desaturase gene and uses thereof
US5968809A (en) 1997-04-11 1999-10-19 Abbot Laboratories Methods and compositions for synthesis of long chain poly-unsaturated fatty acids
US20020037876A1 (en) 1998-06-25 2002-03-28 Yissum Research Development Company Of Hebrew University Of Jerusalem Carboxylic acids and derivatives thereof and pharmaceutical compositions containing them
GB9715444D0 (en) 1997-07-22 1997-09-24 Scotia Holdings Plc Therapeutic and dietary compositions
US6369252B1 (en) 1998-02-26 2002-04-09 The University Of Georgia Research Foundation, Inc. Structured lipids
EP1066235B1 (en) 1998-04-03 2003-10-01 Yissum Research And Development Company Of The Hebrew University Of Jerusalem Synthetic endogenous cannabinoid analogues and uses thereof
US7101914B2 (en) 1998-05-04 2006-09-05 Natural Asa Isomer enriched conjugated linoleic acid compositions
US6696584B2 (en) 1998-05-04 2004-02-24 Natural Asa Isomer enriched conjugated linoleic acid compositions
US6214372B1 (en) 1998-05-04 2001-04-10 Con Lin Co., Inc. Method of using isomer enriched conjugated linoleic acid compositions
ATE375712T1 (en) 1998-08-04 2007-11-15 Cargill Inc PROMOTERS OF FATTY ACID DESATURASE FROM PLANTS
KR100325581B1 (en) 1998-08-07 2002-08-24 오우택 Analgesic compositions containing substances that result from lipoxygenase metabolism of arachidonic acid
US6555579B2 (en) 1998-08-13 2003-04-29 The Wistar Institute Methods for reducing atherosclerotic plaques
GB9901809D0 (en) 1999-01-27 1999-03-17 Scarista Limited Highly purified ethgyl epa and other epa derivatives for psychiatric and neurological disorderes
JP3779505B2 (en) 1999-08-24 2006-05-31 花王株式会社 Oil composition
US6426367B1 (en) 1999-09-09 2002-07-30 Efa Sciences Llc Methods for selectively occluding blood supplies to neoplasias
US6340705B1 (en) 1999-09-10 2002-01-22 Monsanto Technology, Llc Use of α-linolenic acid metabolites for treatment or prevention of cancer
PT1088552E (en) 1999-09-30 2006-06-30 Loders Croklaan Bv COMPOSITIONS CONTAINING PINOLENIC ACID AND ITS UTILIZATION AS A COMPONENT IN HEALTH
GB9923738D0 (en) 1999-10-07 1999-12-08 Nestle Sa Nutritional composition
US20010047036A1 (en) 1999-12-17 2001-11-29 Vanderhoof Jon A. Composition for improving the proliferative response during adaptation of the gastrointestinal tract and use in short bowel syndrome
EG22407A (en) 2000-02-17 2003-01-29 Iams Company Method for improving bone modeling and chondrocyte functioning in growing canines
US6361806B1 (en) 2000-02-23 2002-03-26 Michael P. Allen Composition for and method of topical administration to effect changes in subcutaneous adipose tissue
EP1129711B1 (en) 2000-02-24 2004-01-28 Unilever N.V. Pinolenic acid against diabetes
GB0016045D0 (en) 2000-06-29 2000-08-23 Laxdale Limited Therapeutic combinations of fatty acids
JP2002047176A (en) 2000-08-04 2002-02-12 Idemitsu Technofine Co Ltd Ige production suppressing agent
US6664230B1 (en) 2000-08-24 2003-12-16 The Regents Of The University Of California Orally administered peptides to ameliorate atherosclerosis
FR2815227B1 (en) 2000-10-17 2003-04-11 Schwartz Laboratoires Robert ANTI-STRESS COMPOSITION FOR PRIMARY INCORPORATION IN NUTRITIONAL VEHICLES
NO20005718A (en) 2000-11-13 2001-06-05 Ethics Cosmeceuticals Ab Composition for skin containing chitosan-conjugated CLA and chitosan-conjugated vitamin A or a <beta> -cyclodextrin-conjugated vitamin A and methods for its preparation and use
US6528040B1 (en) 2001-01-18 2003-03-04 Maurine Pearson EMU oil-based formulations for use as an analgesic, anesthetic and antipruritic
WO2002069964A1 (en) 2001-03-05 2002-09-12 Ernest Stephen P Enteral formulation
JP2004531568A (en) 2001-05-30 2004-10-14 ラクスデイル リミテッド Coenzyme Q and eicosapentaenoic acid (EPA)
CA2452401C (en) * 2001-07-02 2013-02-26 Suntory Limited Process for producing fat comprising triglyceride containing highly unsaturated fatty acid
EP1285590A1 (en) 2001-08-08 2003-02-26 Société des Produits Nestlé S.A. Lipid blends
AU784852B2 (en) 2001-08-10 2006-07-06 Mars, Incorporated Canine support diet
US20030032674A1 (en) * 2001-08-13 2003-02-13 Hwang Daniel H. Use of unsaturated fatty acids to treat severe inflammatory diseases
NL1019368C2 (en) 2001-11-14 2003-05-20 Nutricia Nv Preparation for improving receptor performance.
US6677470B2 (en) 2001-11-20 2004-01-13 Natural Asa Functional acylglycerides
ITMI20012732A1 (en) 2001-12-20 2003-06-20 Health Pharma S R L FOOD SUPPLEMENT FOR NEUROPATHICS
AUPS082102A0 (en) 2002-03-01 2002-03-21 Women's And Children's Hospital Therapeutic properties of oils
CN101072509A (en) 2002-03-08 2007-11-14 孟山都技术有限公司 Treatment and prevention of inflammatory disorders
US20040048926A1 (en) 2002-03-15 2004-03-11 Hoffman Dennis Robert Use of docosahexaenoic acid and arachidonic acid to enhance the visual development of term infants breast-fed up to the age of six months
WO2003097034A1 (en) 2002-05-17 2003-11-27 Nikken Chemicals Co., Ltd. TGF-α EXPRESSION INHIBITORS
US7335481B2 (en) 2002-07-24 2008-02-26 Christer Owman Methods of identifying compounds that affect a fatty acid cell-surface receptor
AU2003304108B2 (en) 2002-10-30 2007-03-22 Spherics, Inc. Nanoparticulate bioactive agents
US7074418B2 (en) 2002-11-18 2006-07-11 Changaris David G Conjugated fatty acid based emulsion and methods for preparing and using same
US20040229950A1 (en) 2002-11-26 2004-11-18 Vanderhoek Jack Y. Method and composition with conjugated linoleic acid esters
US6841573B2 (en) 2002-11-27 2005-01-11 Molecular Nutrition Use of arachidonic acid as a method of increasing skeletal muscle mass
US20040209953A1 (en) 2002-12-06 2004-10-21 Wai Lee Theresa Siu-Ling Glyceride compositions and methods of making and using same
US20040208939A1 (en) 2003-04-18 2004-10-21 Barry Sears Novel dietary compositions to reduce inflammation
GB0311081D0 (en) * 2003-05-14 2003-06-18 Btg Internat Limted Treatment of neurodegenerative conditions
EP1651169A4 (en) * 2003-08-01 2008-02-27 Medarex Inc Combination therapies for multiple sclerosis
CA2534202C (en) 2003-08-18 2012-01-17 Btg International Limited Lipid glycerides for the treatment of neurodegenerative conditions involving demyelination
EP1711173A2 (en) 2003-12-31 2006-10-18 Igennus Limited Formulation containing an eicosapentaenoic acid or an ester thereof and a triterpene or ester thereof
GB0504362D0 (en) * 2005-03-02 2005-04-06 Btg Int Ltd Cytokine modulators

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4701469A (en) * 1983-04-15 1987-10-20 Roussel Uclaf Triglycerides, process for therapeutical applications and compositions containing them
WO2001097793A2 (en) * 2000-06-23 2001-12-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem 2-arachidonylglycerol (2-ag) - an inhibitor of tumor necrosis factor -alfa and neuroprotector of brain in closed head injury
WO2003013497A1 (en) * 2001-08-02 2003-02-20 Suntory Limited Compositions having effects of preventing or ameliorating conditions or diseases caused by brain hypofunction

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10744146B2 (en) 2001-08-02 2020-08-18 Suntory Holdings Limited Composition having effects of preventing or ameliorating conditions or diseases caused by brain hypofunction
US7935729B2 (en) 2003-05-14 2011-05-03 Btg International Limited Use of triglyceride oils containing γ-linolenic acid residues and linoleic acid residues for the treatment of neurodegenerative disease
US7964641B2 (en) 2003-08-18 2011-06-21 Btg International Limited Treatment of neurodegenerative conditions
US8367729B2 (en) 2004-09-17 2013-02-05 Suntory Holdings Limited Composition with preventive or improvement effect on symptoms or diseases associated with stress-induced behavior disorders
US8202907B2 (en) 2004-09-17 2012-06-19 Suntory Holdings Limited Composition with preventive or improvement effect on symptoms or diseases associated with stress-induced behavior disorders
US8114903B2 (en) 2005-03-02 2012-02-14 Btg International Limited Cytokine modulators using cyclic glycerides of essential polyunsaturated fatty acids
WO2006092622A1 (en) * 2005-03-02 2006-09-08 Btg International Limited Treatment of cytokine dysregulation by using sn-2 gamma-linolenoyl, gamma-dihomolinolenoyl or arachidonoyl patty acid glycerol monoesters
WO2006092623A1 (en) * 2005-03-02 2006-09-08 Btg International Limited Cytokine modulators using cyclic glycerides of essential polyunsaturated fatty acids
US9168241B2 (en) 2005-06-30 2015-10-27 Suntory Holdings Limited Compositions ameliorating a reduced diurnal activity and/or depressive symptoms
US10042980B2 (en) 2005-11-17 2018-08-07 Gearbox Llc Providing assistance related to health
US8793141B2 (en) 2005-11-17 2014-07-29 The Invention Science Fund I, Llc Assistance related to health
US20080004909A1 (en) * 2005-11-30 2008-01-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Computational systems related to nutraceuticals
US10296720B2 (en) 2005-11-30 2019-05-21 Gearbox Llc Computational systems and methods related to nutraceuticals
KR101578498B1 (en) * 2006-12-28 2015-12-18 산토리 홀딩스 가부시키가이샤 Nerve regeneration agent
KR20140117705A (en) * 2006-12-28 2014-10-07 산토리 홀딩스 가부시키가이샤 Nerve regeneration agent
WO2008081989A1 (en) * 2006-12-28 2008-07-10 Suntory Holdings Limited Nerve regeneration agent
EP3593797A1 (en) * 2006-12-28 2020-01-15 Suntory Holdings Limited Nerve-regenerating agent
US8383677B2 (en) 2006-12-28 2013-02-26 Suntory Holdings Limited Nerve-regenerating agent
US10543237B2 (en) 2007-03-28 2020-01-28 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US9889163B2 (en) 2007-03-28 2018-02-13 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US8697138B2 (en) 2007-03-28 2014-04-15 Aker Biomarine As Methods of using krill oil to treat risk factors for cardiovascular, metabolic, and inflammatory disorders
US9220735B2 (en) 2007-03-28 2015-12-29 Aker Biomarine Antarctic As Methods of using krill oil to treat risk factors for cardiovascular, metabolic, and inflammatory disorders
US9320765B2 (en) 2007-03-28 2016-04-26 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US9375453B2 (en) 2007-03-28 2016-06-28 Aker Biomarine Antarctic As Methods for producing bioeffective krill oil compositions
US9644169B2 (en) 2007-03-28 2017-05-09 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US9644170B2 (en) 2007-03-28 2017-05-09 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US9730966B2 (en) 2007-03-28 2017-08-15 Aker Biomarine Antartic As Method of reducing appetite in a human subject comprising administering krill oil composition
US9816046B2 (en) 2007-03-28 2017-11-14 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US11865143B2 (en) 2007-03-28 2024-01-09 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US9119864B2 (en) 2007-03-28 2015-09-01 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US10010567B2 (en) 2007-03-28 2018-07-03 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US9078905B2 (en) 2007-03-28 2015-07-14 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US9034388B2 (en) 2007-03-28 2015-05-19 Aker Biomarine Antartic As Bioeffective krill oil compositions
US9028877B2 (en) 2007-03-28 2015-05-12 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US9072752B1 (en) 2007-03-28 2015-07-07 Aker Biomarine Antarctic As Bioeffective krill oil compositions
US8372812B2 (en) 2009-02-26 2013-02-12 Aker Biomarine Asa Phospholipid and protein tablets
US10704011B2 (en) 2013-06-14 2020-07-07 Aker Biomarine Antarctic As Lipid extraction processes
US11578289B2 (en) 2013-06-14 2023-02-14 Aker Biomarine Antarctic As Lipid extraction processes
US9867856B2 (en) 2014-01-10 2018-01-16 Aker Biomarine Antarctic As Phospholipid compositions and their preparation
US10456412B2 (en) 2015-02-11 2019-10-29 Aker Biomarine Antarctic As Lipid extraction processes
US10864223B2 (en) 2015-02-11 2020-12-15 Aker Biomarine Antarctic As Lipid compositions
US11819509B2 (en) 2015-02-11 2023-11-21 Aker Biomarine Antarctic As Lipid compositions

Also Published As

Publication number Publication date
GB2422373B (en) 2008-05-21
AU2004266480A1 (en) 2005-03-03
GB0603646D0 (en) 2006-04-05
US20100113595A1 (en) 2010-05-06
AU2004266480B2 (en) 2011-03-03
US20080194684A1 (en) 2008-08-14
IS8246A (en) 2006-01-19
US7964641B2 (en) 2011-06-21
US20120142775A1 (en) 2012-06-07
EP2324828A1 (en) 2011-05-25
ES2304094A1 (en) 2008-09-01
CH698539A8 (en) 2009-10-15
JP2007502805A (en) 2007-02-15
EP1660071A1 (en) 2006-05-31
CA2534202A1 (en) 2005-03-03
US20100113810A1 (en) 2010-05-06
CA2534202C (en) 2012-01-17
DE112004001520T5 (en) 2006-07-06
NO20061232L (en) 2006-05-15
GB2422373A (en) 2006-07-26
FI20060154A (en) 2006-02-17
ES2304094B2 (en) 2010-10-14
CH698539B1 (en) 2009-08-31
DE112004001520B4 (en) 2008-04-10

Similar Documents

Publication Publication Date Title
US20100113810A1 (en) Treatment of neurodegenerative conditions
CA2524036C (en) Treatment of neurodegenerative conditions
US9796658B2 (en) Production and purification of esters of polyunsaturated fatty acids
US20100130608A1 (en) Compositions and methods for reducing triglyceride levels
JP2010509204A (en) Fatty acid alcohol
US20090023807A1 (en) Treatment of Cytokine Dysregulation by Using Sn-2 Gamma-Linolenoyl, Gamma-Diho-Molinolenoyl or Arachidonoyl Patty Acid Glycerol Monoesters
US8114903B2 (en) Cytokine modulators using cyclic glycerides of essential polyunsaturated fatty acids
US20090036410A1 (en) Structured Phospholipids
GB2442164A (en) Treatment of neurodegenerative conditions
GB2442161A (en) Treatment of neurodegenerative conditions
CN1867328B (en) Use of lipid glyceride in preparing medicine for treatment of neurodegenerative conditions

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200480030644.X

Country of ref document: CN

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): 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 IT 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

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2534202

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2004266480

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1120040015200

Country of ref document: DE

Ref document number: 20060154

Country of ref document: FI

Ref document number: 2006523678

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 0603646.1

Country of ref document: GB

Ref document number: 0603646

Country of ref document: GB

WWE Wipo information: entry into national phase

Ref document number: 2004768085

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2004266480

Country of ref document: AU

Date of ref document: 20040813

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004266480

Country of ref document: AU

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
WWP Wipo information: published in national office

Ref document number: 2004768085

Country of ref document: EP

RET De translation (de og part 6b)

Ref document number: 112004001520

Country of ref document: DE

Date of ref document: 20060706

Kind code of ref document: P

WWE Wipo information: entry into national phase

Ref document number: 112004001520

Country of ref document: DE

REG Reference to national code

Ref country code: DE

Ref legal event code: 8607

WWE Wipo information: entry into national phase

Ref document number: 10567778

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 200650014

Country of ref document: ES

Kind code of ref document: A

WWG Wipo information: grant in national office

Ref document number: 200650014

Country of ref document: ES

Kind code of ref document: A