WO1999047101A2 - Compositions containing lysophosphatidic acids which inhibit apoptosis and uses thereof - Google Patents

Compositions containing lysophosphatidic acids which inhibit apoptosis and uses thereof Download PDF

Info

Publication number
WO1999047101A2
WO1999047101A2 PCT/US1999/005943 US9905943W WO9947101A2 WO 1999047101 A2 WO1999047101 A2 WO 1999047101A2 US 9905943 W US9905943 W US 9905943W WO 9947101 A2 WO9947101 A2 WO 9947101A2
Authority
WO
WIPO (PCT)
Prior art keywords
lpa
compound
composition according
composition
mmol
Prior art date
Application number
PCT/US1999/005943
Other languages
French (fr)
Other versions
WO1999047101A3 (en
Inventor
John G. Goddard
Donald H. Picker
Samuil R. Umansky
Steven Price
Jacobus C. H. M. Wijkmans
Edward Andrew Boyd
Anthony David Baxter
Original Assignee
Lxr Biotechnology, Inc.
Oxford Asymmetry 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
Application filed by Lxr Biotechnology, Inc., Oxford Asymmetry Limited filed Critical Lxr Biotechnology, Inc.
Priority to EP99913956A priority Critical patent/EP1069895A4/en
Priority to US09/646,599 priority patent/US6949528B1/en
Priority to AU31915/99A priority patent/AU3191599A/en
Publication of WO1999047101A2 publication Critical patent/WO1999047101A2/en
Publication of WO1999047101A3 publication Critical patent/WO1999047101A3/en
Priority to US11/236,429 priority patent/US7259273B1/en
Priority to US11/834,505 priority patent/US20080076736A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds

Definitions

  • compositions containing lysophosphatidic acid or analogs and derivatives thereof, which have been found to exhibit anti-apoptotic activity and or to preserve or restore cell, tissue or organ function.
  • LPA lysophosphatidic acid or analogs and derivatives thereof
  • This invention specifically relates to therapeutically effective reverse ester, reverse thioester, and bis-LPA analogs encompassed by the term LPA. Additionally, this invention relates to methods of use of these therapeutically effective compositions.
  • LPI lysophosphatidylcholine
  • LPC lysophosphatidylcholine
  • apoptosis studies of apoptosis suggest that a common metabolic pathway leading to apoptosis can be initiated by a wide variety of signals, including hormones, serum growth factor deprivation, chemotherapeutic agents, ionizing radiation, and infection by human immunodeficiency virus
  • HTV hypertension
  • Apoptosis can also be induced by mild, non-catastrophic cell injury and can be concomitant with adjacent necrosis. Agents that affect the biological control of apoptosis thus have therapeutic utility in numerous clinical indications.
  • Apoptosis is an active cellular response to a physiologic or external signal and can be modulated by interfering with the apoptotic pathway. Conversely, by definition, necrosis can be prevented only by decreasing cell injury. Prevention of apoptosis by upregulation of bcl-2 and bcl-x expression, or by inhibitors of ICE-like proteases are typical examples of modulation of cell death. Vvnaxv-ty Molekylyarnaya Biologiya 30:285-295 (1996); VauxandStrasser N S93:2239-2244 (1996); Nunez etal. (1994) Immunol. Today 15:582-588; and Whyte (1996) Trends in Cell Biol. 6:245-148.
  • heart transplantation the donor heart is flushed with approximately 1 liter of cold organ preservation solution (OPS) to arrest the heart. Cooling is supplemented by surrounding the heart with iced saline. The chilled, arrested heart is then surgically excised, immersed in cold OPS, packed in ice and rushed to the recipient center.
  • OPS cold organ preservation solution
  • the diseased heart is excised.
  • the preserved donor heart is then removed from the OPS, trimmed appropriately and transplanted into the recipient. Blood is allowed to flow to the transplanted heart.
  • the transplanted heart will then either resume beating spontaneously or will require chemical and/or electrical treatment to restore normal rhythm.
  • the cardiopulmonary bypass is discontinued and the recipient's chest closed.
  • Most non-transplant surgical procedures on the heart require that the heart be arrested for a period ranging from 1 to 4_hours. During this time, the heart is kept cool by external cooling as well as by periodically reflushing an OPS through the coronary arteries.
  • the OPS composition is designed to rapidly arrest the heart and to keep it in good condition during the period of standstill so that it will resume normal function when the procedure is finished.
  • cardioplegic procedures the heart is exposed in the chest and, at a minimum, the aortic root is isolated. A vascular clamp is applied across the aorta and approximately 1 liter of cold OPS is flushed into the aortic root through a needle.
  • Venting is provided through the left ventricle, pulmonary artery or the right atrium and the effluent, which can contain high levels of potassium, is suctioned out of the chest. This, together with external cooling, produces rapid cessation of contractions. During the period of arrest, the patient's circulation is maintained artificially using cardiopulmonary bypass.
  • the donor organ is cooled rapidly by flushing cold solutions through the organ's vascular system and maintaining the organ at temperatures near 0°C for the purpose of greatly slowing the metabolic rate.
  • the flush solution composition is designed to cause the heart to rapidly stop beating as well as to preserve it.
  • Wicomb et al. reported the beneficial effects of a solution of PEG 8000 and horseradish peroxidase on rabbit hearts preserved by oxygenated low pressure perfusion for 24 hours. Wicombetal. (1989) Transplantation Proceedings 21:1366-1368. The substitution of PEG20M as the colloid for hydroxyethyl starch (HES) of the UW solution also yielded excellent cardiac function.
  • HES hydroxyethyl starch
  • PEG20M consists of two or more molecules of PEG 6000-8000 joined by a bisphenol epoxide linker (CAS # 37225-26-6; CAS name Oxirane, 2, 2' [(l-methyl-ethylidene)bis(4, l-phenyleneoxy methylene)]bis-, polymer with (-hydro-(-hydroxypoly(oxy-l,2-ethanediyl).
  • the substitution of PEG20M for HES also allowed baboon heart storage up to 48 hours and increased cardiac output (CO) under conditions of microperfusion. Wicomb et al. (1986) J. Surg. Res. 40:276; and Wicomb et al. (1989) Transplantation 48:6-9.
  • Microperfusion is a hypoxic, very-low-flow perfusion with flowrates such as 3 ml/g heart wt/24 hour, which is 1/500 of that typical of conventional continuous perfusion. Wicomb et al. (1989) Transplantation 48:6-9.
  • An improved OPS, CardiosolTM heart preservation solution contained the substitution of PEG20M for HES and eliminated five components of UW solution (penicillin, dexamethasone, insulin, allopurinol, and adenosine). Wicomb et al. (1990) Transplantation 49:261-264; and United States Patent No.4,938,961.
  • CardiosolTMheart preservation solution contains 5% or 10% by weight PEG 20M (Union Carbide Chemicals and Plastics Co., Inc., Washington, WV), 40 mM sodium, 125 mM potassium, 5 mM magnesium, 25 mM phosphate, 5 mM sulfate, 100 mM lactobionate, 30 mM raffinose, and 3 mM glutathione.
  • PEG 20M Union Carbide Chemicals and Plastics Co., Inc., Charleston, WV
  • a variety of food supplements containing, in part, partially processed plant extracts have been used to ameliorate the gastrointestinal disorders that often accompany chemotherapy, radiation and ADDS.
  • the supplements generally contain carbohydrates, fat and plant protein hydrolysates. See, e.g., Tomei and Cope et al. in Apoptosis: The Molecular Basis of Cell Death
  • This invention relates to therapeutically effective compositions of matter. More specifically, it encompasses compositions containing lysophosphatidic acid or analogs and derivatives thereof, (collectively "LPA") which have been found to exhibit anti-apoptotic activity or to preserve or restore cell, tissue or organ function. Additionally, this invention relates to methods of use of these therapeutically effective compositions.
  • LPA lysophosphatidic acid or analogs and derivatives thereof
  • compositions containing a compound of the following formula:
  • each W is independently selected from one of the following structures II- V
  • R is an alkyl having between about 10 and about 24 carbon atoms, wherein between 0 and 11, inclusive, of the carbon-carbon bonds are unsaturated. More specifically, R is an alkyl having 18 carbon atoms, wherein 1 or 2 of the carbon-carbon bonds are unsaturated, and mixtures thereof. Even more specifically, the composition comprises
  • composition contains a compound of the following formula:
  • each W is independently SH, OH, OCH 2 CH(NH 2 )C0 2 H, OCHCH 3 CH(NH 2 )C0 2 H, OPO 3 H 2 , OP0 2 HOP0 3 H 2 or Q, wherein when one W is Q, the other W is OH, and wherein Q is one of the following structures:
  • Y is O or S
  • the composition comprises a compound selected from the group consisting of reverse ester-LPA, reverse thioester-LPA or a salt of either.
  • the composition comprises: 3 Oleyl l-thiophosphoryl-2-0- methyl-r ⁇ c-glycerate, or a salt thereof.
  • the present invention also provides methods of treating apoptosis, preserving or restoring function in a cell, tissue or organ comprising administering in vivo or in vitro a therapeutically effective amount of a pharmaceutically acceptable composition of the present invention.
  • the present composition can also contain a potentiating component including, for example, a polyethylene glycol, a protein, or a lipid membrane structure.
  • compositions of the present invention also comprise pharmaceutically acceptable excipients, which can be, without limitation, selected from the group encompassing topical pharmaceutically acceptable carrier, cosmetic carrier, sterile solutions, sterile isotonic solutions, ingestable liquids, pharmaceutically acceptable aerosols and solutions for organ/tissue/cell preservation and/or transplantation.
  • pharmaceutically effective agents such as, without limitation, drugs, antibiotics, wound healing agents and antioxidants.
  • compositions of the present invention comprising the steps of: forming a lipid dispersion comprising LPA; providing at least one of said components; and combining the products of steps a) and b), and more specifically, wherein the lipid dispersion is formed by the steps of: a) dissolving LPA and any other lipids in organic solvent; b) removing the solvent to form dried lipid; and c) dispersing the dried lipid into aqueous media by the steps of: i) forming an even lipid dispersion; and ii) forming an even dispersion of lipid membrane structures.
  • compositions of the compounds of the present invention comprising administering a therapeutically effective amount of a pharmaceutically acceptable composition of the compounds of the present invention.
  • Such composition can further comprise a potentiating component.
  • Treatment can comprise administering to the patient at least one intravenous bolus of an effective amount of the composition.
  • methods of the present invention decrease apoptosis-related problems associated with immunosuppressing viruses, chemotherapeutic agents, or radiation and immunosuppressive drugs.
  • Fig. 1 is a bar graph depicting the effect of various concentrations of 18: 1-LPA analog on protection of C3H/10T1/2 cells from serum deprivation.
  • LPA was prepared with or without a potentiating component (bovine serum albumin or phosphatidyl glycerol vesicles).
  • the open section of the bars represents adherent cells and the solid section of the bars represents non-adherent cells.
  • "LPA" refers to 18: 1-LPA in all figures.
  • the white sections represent adherent cells and the dark sections represent non-adherent cells.
  • Fig. 1-8 and 11 the white sections represent adherent cells and the dark sections represent non-adherent cells.
  • FIG. 2 is a bar graph depicting the percentage of adherent cells (open sections) and non-adherent cells (solid sections) in the C3H/10T1/2 assay after 24 hours exposure to serum-free medium to which 18: 1-LPA was added as a 10% (by weight) mixture in various phospholipid membrane structures. For all treatments, 18: 1-LPA was delivered at 0.25, 0.75, 2.25 or 6.75 (g/mL. The following list provides the compositions represented by each column in Fig.
  • Fig. 3 is a bar graph depicting protection of serum-deprived C3H/10T1/2 cells by Five Phospholipid Mixture (referred to as "Elirex” in the figure) and 18: 1-LPA formulations in phosphotidyl glycerol (PG) and phosphotidyl glycerol/phosphotidylcholine (PG/PC) membrane structures.
  • Elirex Five Phospholipid Mixture
  • PG phosphotidyl glycerol
  • PG/PC phosphotidyl glycerol/phosphotidylcholine
  • Fig. 4 is a bar graph depicting protection of serum-deprived C3H/10T1/2 cells by 18: 1-LPA and lysophosphotidylserine (LPS).
  • Fig.5 is a bar graph depicting protection of C3H/10T1/2 cells from serum-deprivation by soy-derived, 18:1 and 16:0 LPA.
  • Fig. 6 is a bar graph depicting C3H/10T1/2 cell protection from serum-deprivation by 18:1 LPA and 18:0 LPA alone or in PG membrane structures.
  • Fig. 8 is a bar graph depicting protection of serum-deprived C3H/10T1/2 cells by 18: 1-LPA in Five Phospholipid Mixture (labeled "Elirex") and PC or chemically modified PC membrane structures.
  • Fig. 9 depicts an autoradiograph of a non-denaturing polyacrylamide gel of proteins separated following incubation with 3 H-labeled 18: 1-LPA.
  • Fig. 13 is a graph depicting the prevention of cardiomyocyte death induced by serum/glucose deprivation by PEG, Five Phospholipid Mixture (referred to in the figure as
  • Fig. 14 is a graph depicting the treatment of rat neonatal cardiomyocytes with PEG plus
  • Fig. 16 is a bar graph depicting the actual infarct size compared to the area at risk in the rat heart model treated with placebo (soUd bar) or a dose of the combination of Five PhosphoUpid Mixture and PEG (gray bar, Usted in key as "APM/PEG").
  • 'Treating apoptosis is herein defined as adrninistering to a cell, tissue, organ or organism exhibiting apoptosis, or at risk of apoptosis, a treatment to effect beneficial or desired clinical results, including, but not limited to preventing or diminishing apoptosis.
  • phosphoUpid structures are weU defined in the Uterature, they can vary with respect to Upid chain length and saturation.
  • “Lysophosphatidic acids” or “LPAs” as used herein shall encompass the following structures and descriptions and will also include related structures known in the art provided they are effective in producing therapeutic response.
  • LPA has the following general structure:
  • UB is used in reference to the various structures herein to describe the number of unsaturated carbon atoms in R. For example, if R is 18 and UB is 1, R contains 18 carbon atoms, with 1 unsaturated bond.
  • Some LPA analogs are also referred to herein as R:UB-LPA (i.e.
  • LPA includes LPA having any one of a variety of fatty acids esterified at the C 1 position.
  • Examples include LPA wherein the fatty acid ester is lauryl, myristyl, palmityl, stearyl, palmitoleyl, oleyl or linoleyl.
  • the composition where R is 18 and UB is 1, shall herein be referred to as "18: 1-LPA").
  • suitable phospholipids see any chemical catalog of a phospholipid suppUer, for instance, the (1994) Avanti Polar Lipids Inc. catalog, particularly pages 14 and 21.
  • R can be an unsubstituted or substituted, saturated or unsaturated, straight or branched chain alkyl having from about 10 to about 24 carbon atoms.
  • R can have between 0 and (n-2)/2 unsaturated bonds, wherein n is the number of carbon atoms in R.
  • Substitutions include, but are not limited to, halogen, hydroxy, phenyl, amino or acylamino.
  • LPA also encompasses LPA analogs. Given the examples provided herein, it can be determined readily if an LPA analog exerts sufficient anti-apoptotic activity to be suitable for the uses contemplated herein. A wide variety of LPA analogs are known in the art and many of these can be purchased from commercial sources such as Avanti Polar Lipids Inc.
  • W is OH;
  • Z is OH, SH, NH 2 , halogen, OPOjH H,
  • R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably 24-30, more preferably 24-28 and most preferably about 24 carbon atoms, or ((CH 2 ) ⁇ n O) p (CH 2 ) I -W where p is an integer from 1 to about 900 and where each m is independently an integer from 2 to about 10 and W is OH, or 0(CH 2 ) q CH 3 where q is an integer from 0 to about 10; Y is O or S; and n is an integer from 0 to about 10.
  • R is between about 10 and 24, UB is 0-11, and mixtures thereof. More preferably, R is between about 14 and 20, UB is 0-6, and mixtures thereof. Even more preferably, R is between about 16 and 18, UB is 0-3, and mixtures thereof. Most preferably, R is 18, UB is 1 or 2, and mixtures thereof.
  • ethers and thioethers at the Cl position having the structure: o
  • W is OH
  • Z is OH, CH 2 OH, SH, NH 2 , halogen, OPOsHj, H or SOjH
  • R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably where p is an integer from 1 to about 900 and where each m is independently an integer from 2 to about 10 and W is OH, or 0(CH 2 ) q CH 3 where q is an integer from 0 to about 10; Y is O or S; and n is an integer from 0 to about 10. Simon et al. (1982) Biochem. Biophys. Res. Comm. 108:1743-1750.
  • R is between about 10 and 24, UB is 0-11, and mixtures thereof. More preferably, R is between about 14 and 20, UB is 0-6, and mixtures thereof. Even more preferably,
  • R is between about 16 and 18, UB is 0-3, and mixtures thereof. Most preferably, R is 18, UB is 1 or 2, and mixtures thereof.
  • glycero LPA analogs having the structure:
  • R is between aboutlO and 24, UB is 0-11, and mixtures thereof. More preferably, R is between about 14 and 20, UB is 0-6, and mixtures thereof. Even more preferably, R is between about 16 and 18, UB is 0-3, and mixtures thereof. Most preferably, R is 18, UB is 1 or 2, and mixtures thereof.
  • R is an amino acid side chain or a branched amino acid side chain, or an alkylated amino acid side chain, unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably 24-30, more preferably 24-28 and most preferably about 24 carbon atoms, or ((CH 2 ) m O) p (CH 2 ) I -W where p is an integer from 1 to about 900 and where each m is independently an integer from 2 to about 10 and W is OH, or 0(CH 2 ) q CH 3 where q is an integer from 0 to about 10; Y is O or S; and n is an integer from 0 to about 10.
  • R is between about 10 and 24, UB is 0-11, and mixtures thereof. More preferably, R is between about 14 and 20, UB is 0-6, and mixtures thereof. Even more preferably, R is between about 16 and 18, UB is 0-3, and mixtures thereof. Most preferably,
  • R is 18, UB is 1 or 2, and mixtures thereof.
  • each W is independently selected from one of the following structures (XI, XH, Xffl, XTV): X Z
  • LPA Naturally occurring derivatives are also encompassed in the term "LPA.” Such derivatives include, but are not limited to, PHYPLA or cLPA. Murakami-Murofushi et al. (1992) J. Biol. Chem.267:21512-21517. CycUc derivatives can also be synthesized by methods known in the art.
  • the phospholipids can be obtained from any source including, but not limited to, commercial, isolated from a variety of different plants (including plant organs) and animals or created synthetically. Preferably the plants are in the soybean family, but the phospholipids can be isolated from other plants including, but not limited to, those in the leguminosae (beans and peas etc.).
  • the phospholipids can also be isolated from partially purified plant extracts including, but not limited to, soy molasses, lecithin (fluid, deoiled or other forms), partially purified protein concentrates, partially purified protein hydrolysates, defatted soy flakes, refined soy oils, soy grits, soy flours and other soy fractions from which lipids can be extracted.
  • phospholipids of the present invention can be isolated from a particular species of plant, plant extract or organ within a plant.
  • U.S. Patent No. 3,365,440 describes extraction oflipids from soybeans.
  • U.S. Patent Nos. 5,567,425; 5,602,885; 5,624,675; 5,635,186; 5,635,187 have further general descriptions of a variety of techniques useful for the present invention.
  • the phospholipids can be obtained from plant sources by any method known in the art provided it results in purification of at least one of the phospholipids of the invention. A variety of methods are known in the art for purifying and analyzing phosphoUpids from plant sources.
  • PhosphoUpids can also be derived from animal sources.
  • the animal is a mammal.
  • the phosphoUpids are derived from Uver cells.
  • Such phosphoUpids are commercially available or can be purified from animal tissue by methods known in the art, for instance from animal and egg lecithin or from the compositions described in WO 95/15173.
  • PhosphoUpids in general, and LPAs in particular, can also be derived from blood.
  • the phosphoUpids of the invention can also be synthesized by methods known in the art. Suitable semi-synthetic phospholipids and their synthesis are described in Kates, Techniques of Lipidology ( 1972). A synthesis of lysophosphatidic acid is described in W. Stoffel and G.D. Wolf, Chemische Synthese von l-0-[3H]Palmitoyl-L-glycerin-3-phosphate (L-3-Lysophosphatidsaure), ChemJBer., 347 (1966) 94-101. The synthesis of various cyclic phosphate LPAs is described in
  • the phospholipids can be used. Purity can be assayed by any method known in the art such as two dimensional TLC or HPLC and assayed for total Upids, phospholipids or phosphate. Various suitable methods are outlined in Kates (1972).
  • the phosphoUpids must be of sufficient purity such that, when mixed at a total concentration of about 10 mg/rnL, the mixture can be sonicated as described below to provide a relatively translucent solution.
  • the phospholipids are at least 90% pure, more preferably, they are at least 95% pure and, most preferably, they are at least 99% pure.
  • the references cited herein are incorporated by reference in their entirety.
  • potentiating component is defined as a molecule which potentiates the therapeutic activity of LPA. Potentiating components include, but are not limited to, proteins, other phospholipids, polyethylene glycols (PEG), Upid membrane structure forming compounds, polypeptides, modified polypeptides and polymers.
  • LPA is presented in combination with a potentiating component such as BSA and/or PEG.
  • Alternative potentiating components appropriate to protect the activity of LPA can be identified by performing a screen as described in Example 2, in the presence and absence of the potentiating component to be tested.
  • An appropriate potentiating component for use in the present invention can easily be selected by combining the composition to be evaluated with a therapeutic amount of LPA in a therapeutically acceptable solution and evaluating the combination for its mitogenic activity, storage stability, in vivo half life, and for anti-apoptotic activity by any method known in the art, including those described herein. If the mixture has acceptable stability, is not therapeutically unacceptable due to mitogenic activity, and displays anti-apoptotic activity, or preserves or restores cell, tissue or organ function, the potentiating component is appropriate for use in the present invention.
  • the phosphoUpids PA; PI; lysophosphatidic acid; LPI; and LPC are present in the composition in a range of ratios from 0-20:5-20:2-16:0-4:0-8, respectively.
  • these phosphoUpids are in a ratio of approximately 2-15:8-15:6-10:2-4:2-8, respectively.
  • these phospholipids are in a ratio of approximately 10:10:8:2:4, respectively.
  • LPA is combined with PEG prior to therapeutic use.
  • PEGs constitute a diverse group of molecules. Only those that potentiate the therapeutic efficacy of LPA are encompassed herein. It is within the skill of one in the art to determine whether a particular PEG is suitable for use in the claimed compositions. Such a determination can be made, for instance, by the methods described herein.
  • Polyethylene glycol (“PEG”) ((-Hydro-(-hydroxypoly (oxy-l,2-ethanediyl)), is known by numerous designations including macrogel; PEG; Carbowax; Jeffox; Nycoline; Pluracol E; Poly-G; Polyglycol E; and
  • PEG refers to the Uquid and solid polymers of the general formula H(OCH 2 -CH 2 ) folkOH, where n is greater than or equal to 4. In general, each PEG is foUowed by a number which corresponds to its average MW. PEG syntheses are described for instance in Hibbert (1939) J. Am. Chem. Soc. 61:1905-1910. For review, see also, PoweU, III in Handbook of Water-Soluble Gums &Resins, R.L. Davidson ed. (McGraw-HiU, New York, 1980)pp. 18/1-18/31.
  • PEGs have found use as water-soluble lubricants for rubber molds, textile fibers, and metal-forming operations, in food and food packaging, in hair preparations and in cosmetics in general and as ointment and suppository bases in pharmaceutical compositions.
  • PEGs are clear, viscous Uquids or white soUds that dissolve in water to form transparent solutions. They are soluble in many organic solvents and readily soluble in aromatic hydrocarbons. They are only slightly soluble in aliphatic hydrocarbons. Typically, they do not hydrolyze on storage. PEGs have low toxicity. The molecular weights of PEG compositions Usted herein are given in number averages rather than weight averages.
  • PEG20M consists of two or more molecules of PEG having approximate molecular weights of 6000-10,000 joined by a bisphenol epoxide linker (CAS # 37225-26-6; CAS name Oxirane, 2, 2' [(l-methyl-ethylidene)bis(4,l-phenyleneoxy methylene)]bis-, polymer with (-hydro-(-hydroxypoly(oxy-l,2-ethanediyl).
  • PEG20L is a substantially linear PEG having an average molecular weight of about 20,000 Daltons (available from several commercial sources including, but not limited to, ClariantHoechst Celanese, Fluka and Nippon Oils and Fats). The molecular weights of PEG compositions listed herein are given in number averages rather than weight averages. Various other molecular weights of linear PEG are also available from several commercial sources.
  • PEG has been used in a number of pharmacologic applications.
  • Conjugation to PEG also increases the solubility and biological half-Ufe of cytokines.
  • Conjugates of PEG and glucocerebroside have been formulated for treating Gaucher's disease.
  • PEG has also been conjugated to such enzymes as adenosine deaminase, amidase bovine and asparaginase, for therapeutic use. See Delgado et al. (1992) Crit. Rev. Ther. Drug. Carrier Syst. 9:249-304; and Burnham (1994) Am. J. Hosp. Pharm. 51:210-218, for review.
  • compositions comprising therapeutically effective amounts of LPA and PEG.
  • the PEG to LPA weight ratio is such that the LPA therapeutic activity is potentiated by the PEG.
  • the PEG to LPA weight ratio is such that the LPA therapeutic activity is potentiated by the PEG.
  • LPA weight ratio of the composition is preferably 1-100,000: 1 and most preferably 10-10,000: 1.
  • PEG can be in the molecular weight range from about 6,000 to about 500,000.
  • PEG of 35,000 molecular weight is also preferred. PEG of higher molecular weight may have clearance problems when administered in vivo. Thus, PEG of molecular weight greater than 35,000 is preferably used in compositions for topical deUvery.
  • linear PEG or “linear polymer” means that each PEG molecule comprises a single subunit without molecular linkages. By “having an average molecular weight of about 20,000 Daltons” is meant that individual linear polymers can vary in length, but the average molecular weight is about 20,000 Daltons.
  • molecular weight refers to the average molecular weight of a number of molecules in any given sample, as commonly used in the art.
  • a sample of PEG 2,000 might contain a statistical mixture of polymer molecules ranging in weight from, for example, 1,500 to 2,500 Daltons with one molecule differing slightly from the next over a range. Specification of a range of molecular weight indicates that the average molecular weight can be any value between the limits specified, and can include molecules outside those limits.
  • PEG PEG is present in an effective concentration and is essentially free of impurities.
  • impurities is meant the products produced when PEG is oxidized.
  • smaU molecular fragments are formed such as formate, methyl formate, formaldehyde, acetaldehyde, etc., all of which are defined here as impurities.
  • impurities can be removed by any method known in the art, including, but not limited to, dialysis, which removes not only small molecular impurities such as formaldehyde, but also removes hydroperoxides, as evidenced by spectrophotometry.
  • dialysis which removes not only small molecular impurities such as formaldehyde, but also removes hydroperoxides, as evidenced by spectrophotometry.
  • conductivity is measured. When dialysis is effective, conductivity drops markedly to 10-20 microsiemens.
  • Removal of impurities by dialysis can be performed through a CeUulose Acetate HoUow Fiber Dialyzer (Baxter Model CA 110) but the same procedure can also be performed by
  • a therapeutically effective amount is meant an amount sufficient to effect beneficial or desired clinical results.
  • a therapeutically effective amount can be administered in one or more administrations.
  • PEG is present in a therapeutically effective amount.
  • an effective amount is the amount required to improve the ability of an OPS to preserve organs.
  • the preferred composition is where phosphoUpids are combined in a ratio of about 10:10:8:2:4 by weight.
  • a ratio of "about” means that the ratios of the phospholipids can range approximately up to 15% but preferably not more than 5%. More preferably, the ratios are within ⁇ 0.5%.
  • PEG has been used as a potentiating component for therapeutic agents, so the capacity of other macromolecules and macromolecular structures to serve as potentiating components has been explored. Included among these are various lipid membrane structures and proteins.
  • Lipid Membrane Structures Lipid membrane structures (LMSs), including Uposomes, micelles, multilamellar vesicles and cellular membrane isolates, have been used as vehicles for delivering therapeutic agents.
  • LMSs Lipid membrane structures
  • LMSs are lamellar lipid particles wherein polar head groups of a polar Upid are arranged to face an aqueous phase of an interface to form membrane structures.
  • a "Uposome” or “Upid vesicle” is a small vesicle bounded by at least one and possibly more than one bilayer Upid membrane. Liposomes are completely closed Upid bilayer membranes which contain entrapped aqueous volume. Liposomes are vesicles which can be unilameUar (single membrane) or multilamellar (onion-like structures characterized by multiple membrane bUayers, each separated from the next by an aqueous layer).
  • the bilayer is composed of two Upid monolayers having a hydrophobic "tail” region and a hydrophilic "head” region.
  • liposomes In the membrane bilayer, the hydrophobic (nonpolar) "tails” of the Upid monolayers orient toward the center of the bilayer, whereas the hydrophilic (polar) "heads” orient toward the aqueous phase.
  • the basic structure of liposomes can be made by a variety of techniques known in the art. These Upid vesicles are made artificially from phospholipids, glycolipids, lipids, steroids such as cholesterol, bolaamphiles related molecules, or a combination thereof by any technique known in the art, including, but not limited to sonication, extrusion, or removal of detergent from lipiddetergent complexes.
  • a Uposome can also optionally comprise additional components associated with the outer surface, such as a tissue targeting component.
  • a "Upid membrane” or “lipid bilayer” need not consist exclusively of lipids, but can additionally contain any percentage of other components, including, but not limited to, cholesterol and other steroids, proteins of any length, and other amphipathic molecules, providing the general structure of the membrane is a sheet of two hydrophilic surfaces sandwiching a hydrophobic core.
  • Suitable lipids include, but are not limited to, those discussed in Lasic (1993)
  • the lipid bilayer making up the Uposome can comprise phospholipids, glycolipids, steroids, and their equivalents .
  • the lipid bilayer can also be made of amphipathic proteins and lipidsoluble chemicals.
  • a composition is chosen that allows the membrane to be formed with reproducible quaUties, such as diameter, and is stable in the presence of elements expected to occur where the Uposome is to be used, such as physiological buffers and circulating molecules.
  • the Uposome is resilient to effects of manipulation by storage, freezing, and mixing with pharmaceutical excipients.
  • Lipids suitable for incorporation into lipid membrane structures include, but are not Umited to, natural, semi-synthetic or synthetic mono- or di-glycerophospholipids including phosphatidylcholines, phosphatidylethanolamines, phosphatidylglycerols, phosphatidylinositols, phosphatidic acids, phosphatidylserines, glycero- and cardioUpins. Sphingolipids such as sphingomyelin and cerebrosides can also be incorporated.
  • lipids can have alternative stereochemistry with, e.g., the phospho group at the sn-l or sn-2 positions.
  • the hydrophobic chains can be attached to the glycerol backbone by acyl, ether, alkyl or other linkages.
  • Derivatives of these Upids are also suitable for incorporation into Uposomes.
  • Derivatives suitable for use include, but are not limited to, haloalkyl derivatives, including those in which all or some of the hydrogen atoms of the alkyl chains are substituted with, for example, fluorine.
  • cholesterol and other amphipathic steroids, bolaamphiphiles include, but are not limited to, haloalkyl derivatives, including those in which all or some of the hydrogen atoms of the alkyl chains are substituted with, for example, fluorine.
  • cholesterol and other amphipathic steroids, bolaamphiphiles include, but are not limited to, haloalkyl derivatives, including those
  • Liposomes can be composed of a single Upid or mixtures of two or more different Upids.
  • the Upid bilayer of the Uposome is formed primarily from phospholipids. More preferably, the phospholipid composition is a complex mixture, comprising a combination of phosphatidylcholine (PC), phosphatidic acid (PA), phosphatidylethanolamine (PE), phosphatidylglycerol (PG) and sphingomyeUn (SM).
  • the LMS can further comprise additional Upids such as phosphatidylinositol (PI), phosphatidylserine (PS), or cardioUpin (diphosphatidylglycerol).
  • PI phosphatidylinositol
  • PS phosphatidylserine
  • cardioUpin diphosphatidylglycerol
  • SM can be replaced with a greater proportion of PC, PE, or a combination thereof.
  • PS can optionally be replaced with phosphatidylglycerol (PG).
  • PG phosphatidylglycerol
  • at least PC and PE are included; more preferably, at least three of the group PC, PS, PE, and SM are included.
  • the composition is chosen so as to confer upon the LMS stability during both storage and administration.Each phosphoUpid described above can vary in its structure depending on the fatty acid moieties that are esterified to the glycerol moiety of the phospholipid. Generally, most commercially available forms of a particular phosphoUpid can be used. However, phospholipids containing particular fatty acid moieties may be preferred for certain applications.
  • the LMS also includes cholesterol or a related steroid to improve the rigidity of the membrane.
  • Any amount of cholesterol can be used.
  • a preferred ratio of total cholesterol to Upid is between about 0.5 and about 1.2 moles of cholesterol per mole of Upid. More preferred is a molar ratio of about 0.8 to about 1.2: 1; even more preferred is a molar ratio of about 0.9 to about 1.1:1; still more preferred is a molar ratio of about 1.0:1.0.
  • Other molecules that can be used to increase the rigidity of the membrane include crossUnked phospholipids.
  • Liposomes for use in vivo are those with an enhanced ability to evade the reticuloendotheUal system, thereby giving them a longer period in which to reach the target cell.
  • Effective Upid compositions in this regard are those with a large proportion of SM and cholesterol, or SM and PI.
  • Liposomes with prolonged circulation time also include those that comprise the monosialogangUoside GMl, glucuronide, or PEG.
  • cholesterol can be added at the ratios indicated above to a lipid mixture consisting of any combination of SM, PI, glucuronide, PEG, and other suitable components.
  • Liposomes can also be provided with molecules at the surface that target them to the ceU of interest.
  • Such smaU molecules can be attached by incorporating into the Upid bilayer a functionaUzed phosphoUpid (US . Patents 5,052,421 and 5,540,935) or a functionalized cholesterol
  • Polypeptides can be attached covalently to the Upid bilayer (EP Patent 0036277), to a glycophosphohpid (U.S. Patent 5,374,548), to a carboxylated phosphoUpid (U.S. Patent 4,762,915), to a derivatized sterol (U.S. Patent 5,000,960), or to a peptide anchor (U.S. Patent 5,109,113).
  • the polypeptide comprises a hydrophobic domain, it can be incorporated directly into the lipid bilayer, either by forming the liposome in its presence, or by performing the liposome and inserting the polypeptide subsequently using a suitable detergent. Tranum-Jensenetal. (1994) J. Membrane Biol. 140: 215-23; EP Patent 0047480; and U.S. Patent 5,252,348.
  • Liposomes have been prepared with mammalian-derived peptides such as cytokines (U.S. Patent 5,258,499), transferrin (Stavridis et al.), antibody (Laukkanen et al. (1994) Biochem.
  • mammalian-derived peptides such as cytokines (U.S. Patent 5,258,499), transferrin (Stavridis et al.), antibody (Laukkanen et al. (1994) Biochem.
  • Liposomes have been prepared with artificial peptides, such as a 14-residue amphipathic sequence which is a fusogenic GALA-type peptide. Puyal et al. (1994) Biochim. Biophys. Acta 1195:259-266. Liposomes have also been prepared with viral components: for example, the F and G glycoprotein of respiratory syncytial virus (RSV) (U.S. Patent 5,252,348), reovirus M cell attachment protein (Rubas etal. (1990) ⁇ ⁇ /cro ⁇ «c ⁇ pj «/ ⁇ 'o j 7:385-395), influenza virus surface protein (WO 92/19267; EP 0047480; and Nussbaum etal.
  • RSV respiratory syncytial virus
  • the present invention also encompasses compositions comprising micelles and methods of using such compositions.
  • MiceUes in aqueous solution both non-ionic, cationic and anionic, have been described in the literature in numerous pubUcations. Mittal (1977) Micellization, Solubilization andMicroemulsions Plenum Press, New York; Mittal (1979) Solution Chemistry of Surfactants Plenum Press, New York; Menger ( 1977) In Biorganic Chemistry III. Macro- and Multicomponent Systems Van Tanelen, Ed. Academic Press, New York; and Menger ( 1979) Ace.
  • Micelles is a term appUed to aggregates which form from tenside molecules in aqueous solutions above a specific temperature or a characteristic concentration. This concentration is called the critical micellization concentration, or cmc. When the cmc is exceeded, the monomer concentration remains practically constant and the excess tenside molecules form micelles. They can occur in various shapes (spheres, rods, discs) depending on the chemical constitution of the tenside and on the temperature, concentration or ionic strength of the solution. The micelles have characteristic aggregation numbers with usually only a small distribution spread. Reaching the cmc is manifest by abrupt changes in the surface tension, the osmotic pressure, the electrical conductivity and the viscosity.
  • Micelles are thermodynamically stable association colloids of surfactant substances in which the hydrophobic radicals of the monomers lie in the interior of the aggregates and are held together by hydrophobic interaction; the hydrophilic groups face the water and by solvation provide the solubility of the colloid.
  • a process for preparing liposomes containing LPA is as foUows.
  • An aqueous dispersion of Uposomes is prepared from membrane components, such as phosphoUpids (e.g. phosphatidylcholine, phosphatidylglycerol, sphingomyelin and phosphatidylethanolamine) and glycoUpids according to known methods as disclosed. Ann. Rev. Biophys. Bioeng. 9:467 (1980).
  • the Uposomes can further contain sterols (e.g., cholesterol and cholestanol), dialkylphosphates, diacylphosphatidic acids, stearylamine, (-tocopherol, etc., in the liposomal membrane.
  • aqueous solution of LPA is added to the liposomal dispersion at a concentration sufficient to produce a therapeutically effective final product.
  • the mixture is allowed to stand for a given period of time, preferably under warming at a temperature more than the phase transition temperature of the membrane or above 40°C, followed by cooling to produce liposomes containing LPA in the liposomal membrane.
  • the desired liposomes can also be prepared by previously mixing the above-described membrane components and LPA and treating the mixture in accordance with known methods for preparing liposomes.
  • the Upid vesicles can be prepared by any suitable technique known in the art. Methods include, but are not Umited to, microencapsulation, microfluidization, LLC method, ethanol injection, freon injection, the "bubble” method, detergent dialysis, hydration, sonication, and reverse-phase evaporation (reviewed in Watwe et al.). For example, ultrasonication and dialysis methods generally produce small unilameUar vesicles, while extrusion and reverse-phase evaporation generally produce larger sized vesicles. Techniques can be combined in order to provide vesicles with the most desirable attributes. One particularly useful method is microfluidization.
  • the invention encompasses LMSs containing tissue or cellular targeting components.
  • tissue or cellular targeting components are components of a LMS that enhance its accumulation at certain tissue or cellular sites in preference to other tissue or ceUular sites when administered to an intact animal, organ, or ceU culture.
  • a targeting component is generally accessible from outside the liposome and is, therefore, generally either bound to the outer surface or inserted into the outer lipid bilayer.
  • a targeting component can be, for example, a peptide, a region of a larger peptide, an antibody or antigen binding fragment thereof, a nucleic acid, a carbohydrate, a region of a complex carbohydrate, a special lipid, or a small molecule such as a drug, hormone, or hapten, attached to any of the aforementioned molecules.
  • the LMSs can be targeted to any cell type that is undergoing an inappropriate level of apoptosis.
  • Such cells include, but are not limited to, cardiomyocytes, endothelial cells, neuronal cells, hepatocytes, glomerulocytes, lung cells, mucosal cells, skin cells, heart ceUs and cancer cells.
  • LMSs can be targeted to such cell types in various ways.
  • a LMS can be modified to contain an antibody, or a fragment of an antibody, specific for a cell surface molecule, or marker, found solely or primarily on a given cell type. Antibodies with specificity toward cell type-specific cell surface markers are known in the art.
  • compositions of the present invention can additionally comprise surfactants.
  • Surfactants can be cationic, anionic, amphiphiUc, or nonionic.
  • a preferred class of surfactants are nonionic surfactants; particularly preferred are those that are water soluble.
  • Nonionic, water soluble surfactants include polyoxyethylene derivatives of fatty alcohols, fatty acid ester of fatty alcohols and glyceryl esters, wherein the polyoxyethylene group is coupled via an ether linkage to an alcohol group.
  • Examples include, but are not Umited to, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene castor oil derivatives, polyoxyethylene hardened castor oU derivatives, fatty acid sodium salts, sodium cholates, polyexyethylene fatty acid ester and polyoxyethylene alkyl ethers.
  • Proteins other than serum albumin have been characterized or impUcated as having fatty acid/Upid binding capabiUties. These include the fatty acid binding proteins (FABPs), the Upid binding proteins, long-chain fatty acyl-CoA (LCFA-CoA) binding proteins, phosphoUpid transfer proteins, and Ca + /lipid binding proteins.
  • FABPs fatty acid binding proteins
  • LCFA-CoA long-chain fatty acyl-CoA binding proteins
  • phosphoUpid transfer proteins and Ca + /lipid binding proteins.
  • Lipid-binding proteins are a family of fatty acid and retinoid transport proteins. Some are intraceUular, for example, while others are secreted from the cell. IntraceUular lipid binding proteins include cellular retinoic acid binding proteins, CRABP
  • the lipid-binding protein family includes many FABP.
  • the FABP are relatively smaU proteins (13-15 kDa) capable of binding long-chain fatty acids (LCFA) and their coenzyme A and L-camitine esters .
  • LCFA-CoA play an important physiological role in intermediary metaboUsm of fatty acids, but recent data indicate that they also can be potent regulators of ceU functions .
  • LCFA-CoA typically exist within a cell bound to membrane lipids and/or proteins. Proteins which bind LCFA-CoA include LCFA CoA binding protein, FABP and sterol carrier protein-2. Gossett et al. (1996) Lipids 31:895-918.
  • a protein that enhances a therapeutic effect of LPA can be a naturally-occurring or a synthetic protein, a protein fragment, or can contain lipid or sugar moieties.
  • the protein is a lipid-binding or carrier protein.
  • the protein can contain other modifications including, but not Umited to glycosylation, phosphorylation, myristylation, sulfation and hydroxylation.
  • the protein can be a hybrid protein, part of which confers the property of enhancing a therapeutic effect of LPA, and another part of which confers some other desirable property such as targeting to a particular cell type, enhanced in vivo stability, and the like.
  • Preferred proteins include, but are not Umited to, albumin, soy and plant proteins, cytochrome C, low density Upoprotein, acyl carrier protein and alphafeto-protein.
  • Suitable polypeptides include any known in the art. Modified polypeptides contain any of a variety of modifications, including, but not Umited to glycosylation, phosphorylation, myristylation, sulfationandhydroxylation. As used herein, a suitable polypeptide is one that will protect LPA to preserve its activity. Examples of binding proteins include, but are not limited to, fatty acid binding proteins, albumins such as bovine serum albumin (BSA) and pea albumin.
  • BSA bovine serum albumin
  • Binding as used herein means that the LPA and the protein form non-covalent complexes with one another. Binding of a protein to LPA can be determined by a variety of methods known in the art, including a non-denaturing gel electrophoresis binding assay as described in Example_3 or a size shift assay as described in Example_3.
  • the protein/LPA compositions optionally exhibit a reduction in the mitogenic activity of LPA.
  • Mitogenic activity can be measured by any method known in the art, for instance, measurement of uptake of 3 H-thymidine by cells treated with LPA with or without the addition of protein.
  • LP Aprotein compositions of the present invention can be prepared in a variety of ways.
  • LPA and a protein can be suspended in any biocompatible buffer, for example, bicarbonate buffered saline, at a ratio of about 0.00001 % to 10%, more preferably about 0.001 % to 1 %.
  • the mixtures can then be sonicated for about 5_minutes or until the mixture is clear or can be sterile filtered.
  • the protein concentration of the LP A/protein compositions can be from about 0.001 to about 50 mg/mL, more preferably from about 0.01 to about lO mg/mL, even more preferably from about 0.05 to about l.Ojmg/mL.
  • the protein component of the LP A/protein compositions of the present invention are substantially pure, i.e., the protein is substantially free of lipids, other proteins, or any other material that might affect the activity of the LP A/protein complex.
  • the protein is at least about 75% pure, more preferably at least about 85% pure and still more preferably at least about 95% pure. 5.
  • Other Optional Components are also be from about 0.001 to about 50 mg/mL, more preferably from about 0.01 to about lO mg/mL, even more preferably from about 0.05 to about l.Ojmg/mL.
  • the protein component of the LP A/protein compositions of the present invention are substantially pure, i.e., the protein is substantially free of
  • Suitable polymers can be any known in the art of pharmaceuticals and include, but are not Umited to, naturaUy-occurring polymers such as dextrans, hydroxyethyl starch, and polysaccharides; and synthetic polymers. Examples of naturally occurring polymers include proteins, glycopeptides, polysaccharides, dextran and lipids.
  • the additional polymer can be a synthetic polymer.
  • Examples of synthetic polymers which are suitable for use in the present invention include, but are not Umited to, polyalkyl glycols (PAG) such as PEG, polyoxyethylated polyols (POP) such as polyoxyethylated glycerol (POG), polytrimethylene glycol (PTG) polypropylene glycol (PPG), polyhydroxyethyl methacrylate, polyvinyl alcohol (PVA), polyacryhc acid, polyethyloxazoUne, polyacrylamide, polyvinylpyrroUdone (PVP), polyamino acids, polyurethane and polyphosphazene.
  • the synthetic polymers can also be linear or branched, substituted or unsubstituted, homopolymeric, copolymers, or block copolymers of two or more different synthetic monomers.
  • the synthetic polymers can have the following generic structure:
  • R, and R 3 are the same or different and are H, H 3 C, OH, R 2 or a reactive group (as described below); where R 2 is a linear or branched substituted or unsubstituted alkyl group; where
  • Biodegradable polymers can also be included in the compositions. These include, but are not Umited to poly(lactide), poly(glycoUde) poly(vinyl alcohol), crosslinked collagen.
  • the polymers can also include polyglycolic acid, polyethylene terephthalate, polybutyl lactose, polycaprolactone, D-polylactic acid, L-polylactic acid and poly-L-lysine and polymeric mixtures thereof.
  • Suitable polymers also include polysaccharides.
  • Suitable polysaccharides include, but are not limited to, trehalose, glucose, maltose, lactose, maltulose, iso-maltulose, lactulose, mono-reducing glycosides of polyhydroxy compounds selected from sugar alcohols, other straight chain polyalcohols, raffinose, stachyose, melezitose, dextran, sucrose and sugar alcohols thereof, maltitol, lactitol, iso-maltulose, palatinit, 2-D-glucopyranosyl- 1 -(-6-mannitol and their individual sugar alcohols.
  • the compositions can further include pharmaceutically acceptable excipients.
  • compositions include, but are not limited to, topical pharmaceutically acceptable carrier, cosmetic carrier, sterile solutions, sterile isotonic solutions, ingestable liquids, pharmaceutically acceptable aerosols and other constituents used in solutions for organ/tissue/cell preservation and/or transplantation.
  • the compositions can further include additional pharmaceutically effective agents.
  • Suitable classes of pharmaceuticaUy effective agents include, but are not Umited to, drugs, antibiotics, wound healing agents and antioxidants.
  • Suitable drugs include, but are not Umited to, those listed in Table 1 and from the foUowing classes: antipyretic and anti-inflammatory drugs, analgesics, antiarthritics, antispasmodics, antidepressants, antipsychotics, tranquiUzers, antianxiety drugs, narcotic antagonists, antiparkinsonism agents, cholinergic antagonists, chemotherapeutic agents, immuno-suppressive agents, antiviral agents, parasiticides, appetite suppressants, antiemetics, antihistamines, antimigraine agents, coronary vasodilators, cerebral vasodUators, peripheral vasodilators, hormonal agents, contraceptives, antithrombotic agents, diuretics, antihypertensive agents, cardiovascular drugs, opioids, and vitamins are all included in the compositions described herein.
  • Suitable antibiotics include, but are not limited to, ampicillin, tetracycline, chloramphenicol, erythromycin, amphotericin B and penicillin.
  • Suitable wound healing agents include, but are not Umited to, transforming growth factors, platelet-derived growth factors, epidermal growth factors and fibroblast growth factors .
  • Suitable antioxidants include, but are not limited to, naturaUy-occurring antioxidants such as tocopherols, (e.g., (-tocopherol, vitamin E), ascorbic acid (Vitamin_C), (-carotene (vitamin A), dihydroUpoamide and flavenoids; and synthetic antioxidants such as butylated hydroxytoluene, butylated hydroxyanisole, Trolox®, propyl gallate, other phenoUc antioxidants and phenothiazines; and chelators such as desferoxamide, HBED and
  • compositions for Use in the Present Invention The invention further includes methods of formulating compositions for use in the present invention, some of which appear below and some of which appear throughout the text of the specification.
  • compositions of the present invention can be in either a Uquid or soUd form.
  • the LPA can be concentrated for dilution prior to use.
  • the components are in a concentration suitable for immediate use.
  • a soUd addition of a predetermined amount of an aqueous solution will result in the appropriate concentration of the components.
  • the soUd can also be in powder form, for use in therapies including, but not Umited to inhalation therapies.
  • the LPA is preferably present in an amount of from about 0.00001% to about 10% (weight/volume). More preferably, the LPA is present in an amount of from about 0.0001% to about 1% (weight/volume). Most preferably, the LPA is present in an amount of from about 0.005% to about 1% (weight/volume).
  • the LPA is present in an amount of from about 0.00001% to 50% (weight/weight). More preferably, the LPA is present in an amount of from about 0.001% to 1% (weight/weight).
  • LPA can be suspended in any buffered solution that is preferably free of divalent cations having a pH range of 2-10, more preferably, about 4-8 and most preferably about 6-8.
  • Suitable buffers include, but are not Umited to, D-PBS (phosphate buffered saline, free of calcium and magnesium salts; Gibco BRL) or 50 mM ammomum bicarbonate containing isotonic sodium chloride.
  • the buffered solution is preferably physiologically acceptable.
  • a wide range ofpH values are effective.
  • the pH is between about 5.5 to about 8.
  • any pH at which the composition is at least minimally therapeutically effective is suitable for use.
  • the mixture has been found to be most active at pH 8.
  • the phosphoUpids are suspended in 50 mM ammomum bicarbonate / 0.154 M sodium chloride, 250_(M EDTA with a pH of 7.7-8.0.
  • the mixture is dispersed in order to achieve maximal activity.
  • Any method of dispersion that forms particles of about 5-450 microns is acceptable and about 30-100 microns is preferred. These methods include, but are not Umited to, microfluidization, extrusion and sonication, provided that the method does not denature or otherwise chemicaUy modify the phosphoUpids in such a manner as to render them toxic or of substantially diminished therapeutic activity.
  • the mixture is sonicated until optical clarity is attained although sonication can be continued beyond this point provided the mixture is not overheated.
  • the preferred sonication parameters are those provided in the examples herein.
  • optical clarity indicates that the mixture changes from opaque to translucent. This change is readily monitored visually; no further measurements are necessary.
  • translucent can be defined as when the mixture has an O.D. 600 of less than about 0.2 AU.Concentrations of up to approximately 50 mg/mL phosphoUpids can be prepared. Preferably, 10 mg/mL solutions are used.
  • sonication if used, is in 5 minute alternating cycles, with 5 minutes of sonication foUowed by 5 minutes of equihbration. However, this can be varied, depending on the volume of mixture being sonicated and the heat generated by sonication.
  • the total length of sonication depends on the concentration and volume of the mixture being sonicated and the power output of the sonicator. Sonication should proceed until the mixture has become translucent. Typically, mixtures are sonicated for 3 to 90 minutes. Preferably, sonication proceeds by several periods of 5 minutes each, 6 to 12 total periods, with 1 to 5 minutes between each period to allow equihbration and dissipation of heat.
  • the temperature of the water bath should not exceed about 60°C. Preferably, the temperature of the water bath is not allowed to exceed 37°C.
  • the sonicated mixture is passed through a sterile filter before use.
  • the sterile filter has a 0.2 micron cut off.
  • compositions can be sterilized at any point. Sterilization can be by any method known in the art, and encompasses, but is not limited to, heat sterilization, steam sterilization, ultrafiltration, sterile filtration and ultraviolet light sterilization. Sterilization is essential for most of the methods of treatment, although for certain applications, it may not be necessary, or the level of sterilization required can be reduced.
  • the compositions can also be prepared and dried to form a solid. The soUd is suitable for use as a powder or pill, or in solution upon reconstitution. Any method of drying is suitable for use herein, including, but not limited to, freeze-drying, air drying, spray drying and fluidized bed evaporation, vacuum drying and rotary evaporation
  • compositions are stored under anoxic conditions.
  • Any method of such storage known in the art is suitable for use herein, including, but not Umited to, storage under an inert gas such as argon.
  • the anti-apoptotic activity of the compositions of the present invention can be measured in many anti-apoptosis assays known in the art. These include, but are not Umited to, the serum deprivation of the C3H/10T1/2 ceU assay described in detaU in Example 2. Furthermore, in vivo apoptosis inhibition can be measured by any method known in the art. Methods for determining therapeutic efficacy in treating an ischemic event are known in the art and described herein. Methods for determining efficacy in organ storage and transplantation are known in the art and described herein.
  • compositions described herein can be measured or determined by any method known in the art. For instance, there are a variety of wound healing assays described in the art and cited herein.
  • the invention further comprises any of the above-described compositions in combination with a pharmaceutically acceptable vehicle.
  • the level of purity of the components necessary for the composition can be determined empirically and is within the skiU of one in the art.
  • the compositions are suitable for use in a variety of disorders, as described below, and in both human and veterinary appUcations.
  • compositions are pharmaceutically acceptable due to their low toxicity in the therapeutic dosage range, stability and ability to be inco ⁇ orated into a wide variety of vehicles for numerous routes of administration.
  • the compositions can be administered alone or in combination with other pharmaceutically effective agents including, but not limited to, antibiotics, wound healing agents, antioxidants and other therapeutic agents as previously described.
  • compositions can contain at least a therapeutically effective amount of at least one of the above-described compositions and at least one physiologically acceptable carrier.
  • a physiologically acceptable carrier is one that does not cause an adverse physical reaction upon administration and one in which the compositions are sufficiently soluble to deliver a therapeutically effective amount of the compound.
  • the therapeutically effective amount of the compositions depends in part upon the manner of introduction and the indication to be treated and other criteria evident to one of ordinary skill in the art. Typically, a therapeutically effective amount is one sufficient to ameliorate or cure the condition being treated as evidenced by diminishment of the symptoms compared to a control.
  • a therapeutically effective amount is from about 0.0001% or 1 ⁇ g/mL by weight ofthe phospholipid mixture although a wide range of effective amounts can be used for different indications and can be determined empiricaUy.
  • the route(s) of administration useful in a particular indication are discussed below and are weU known to one of skiU in the art.
  • Suitable cell types for treatment and/or preservation include, but are not limited to, eukaryotic and prokaryotic cells, such as bacterial cells, plant cells, yeast cells, fungi cells, insect ceUs, mammaUan cells, and human cells in particular.
  • Mammalian cell types encompass cardiomyocytes, endotheUal cells, neuronal cells, hepatocytes, renal ceUs, lung cells, mucosal cells, pancreatic cells, gastrointestinal cells, corneal cells and skin cells. These cell types, and the tissues and organs they form, are suitable for treatment and/or preservation by the methods of the present invention. These cell types can be treated either in vivo or in vitro using methods of the present invention.
  • Routes of administration include, but are not limited to, topical, transdermal, parenteral, gastrointestinal, transbronchial, transalveolar, and in vitro treatment of cells, tissues or organs foUowed by in vitro administration of treated ceUs, tissues or organs.
  • Internal routes of administration encompass any method of in vivo administration other than solely by topical application to the skin.
  • Surface administration is accomplished via application of a cream, gel, rinse, etc. containing a therapeutically effective amount of the compositions.
  • Transdermal administration is accomplished by application of a cream, rinse, gel, etc. capable of allowing the active components to penetrate the skin and enter the blood stream.
  • Parenteral routes of administration include, but are not Umited to, direct injection such as intravenous, intramuscular, intraperitoneal or subcutaneous injection.
  • Gastrointestinal routes of administration include, but are not limited to, ingestion and rectal.
  • Transbronchial and transalveolar routes of administration include, but are not Umited to, inhalation, either via the mouth or intranasally (for example, of a mist or a dry powder) and direct injection into an airway, such as through a tracheotomy. While the compositions can be topicaUy administered alone, it may be desirable to administer them in a mixture with a topical physiologically or cosmetically acceptable carrier.
  • Topical pharmaceutically acceptable carrier is any substantiaUy non-toxic carrier conventionally useable for topical administration of pharmaceutical agents in which the compositions wiU remain stable and bioavailable when appUed directly to skin or mucosal surfaces .
  • the compositions can be dissolved in a liquid, dispersed or emulsified in a medium in a conventional manner to form a Uquid preparation or mixed with a semi-solid (gel) or soUd carrier to form a paste, powder, ointment, cream, lotion or the like.
  • Suitable topical pharmaceutically acceptable carriers include water, petroleum jeUy (Vaseline), petrolatum, mineral oil, vegetable oil, animal oil, organic and inorganic waxes, such as microcrystaUine, paraffin and ozocerite wax, natural polymers, such as xanthanes, gelatin, ceUulose, collagen, starch, or gum arabic, synthetic polymers, such as discussed below, alcohols, polyols, and the like.
  • the carrier can be a water miscible carrier composition that is substantiaUy miscible in water.
  • Such water miscible topical pbarmaceuticaUy acceptable carrier composition can include those made with one or more appropriate ingredients set forth above but can also include sustained or delayed release carriers, including water containing, water dispersible or water soluble compositions, such as liposomes, microsponges, microspheres or microcapsules, aqueous base ointments, water-in-oil or oil-in-water emulsions, gels or the like.
  • sustained or delayed release carriers including water containing, water dispersible or water soluble compositions, such as liposomes, microsponges, microspheres or microcapsules, aqueous base ointments, water-in-oil or oil-in-water emulsions, gels or the like.
  • the topical pharmaceutically acceptable carrier comprises a sustained release or delayed release carrier.
  • the carrier is any material capable of sustained or delayed release of the compositions to provide a more efficient administration resulting in one or more of less frequent and/or decreased dosage of the compositions, ease of handling, and extended or delayed effects on dermatologic conditions.
  • the carrier is capable of releasing the compositions when exposed to any oily, fatty, waxy, or moist environment on the area being treated or by diffusing or by release dependent on the degree of loading of the compositions to the carrier in order to obtain release thereof.
  • Non-limiting examples of such carriers include liposomes, microsponges, microspheres, or microcapsules of natural and synthetic polymers and the like.
  • suitable carriers for sustained or delayed release in a moist environment include gelatin, gum arabic, xanthane polymers; by degree of loading include lignin polymers and the like; by oily, fatty or waxy environment include thermoplastic or flexible thermoset resin or elastomer including thermoplastic resins such as polyvinyl halides, polyvinyl esters, polyvinyUdene halides and halogenated polyolefins, elastomers such as brasiUensis, polydienes, and halogenated natural and synthetic rubbers, and flexible thermoset resins such as polyurethanes, epoxy resins and the like.
  • the sustained or delayed release carrier is a liposome, microsponge, microsphere or gel.
  • compositions used in the method of treating dermatologic conditions of the invention are applied directly to the areas to be treated. While not required, it is desirable that the topical composition maintain the active components at the desired location for about 24 to 48 hours, or a length of time sufficient to exert therapeutic efficacy.
  • one or more additional ingredients conventionally found in topical pharmaceutical or cosmetic compositions can be included with the carrier, such as a moisturizers, humectants, odor modifiers, buffers, pigments, preservatives, Vitamins such as A, C and E, emulsifiers, dispersing agents, wetting agents, odor-modifying agents, gelling agents, stabilizers, propeUants, antimicrobial agents, sunscreens, enzymes and the like.
  • additional ingredients include superoxide dismutase, stearyl alcohol, isopropyl myristate, sorbitan monooleate, polyoxyethylene stearate, propylene glycol, water, alkali or alkaline earth lauryl sulfate, methylparaben, octyl dimethyl-p-amino benzoic acid (Padimate O), uric acid, reticuUn, polymucosaccharides, hydroxyethyl starch (such as, DuPont Pentafraction), hyaluronic acids, aloe vera, lecithin, polyoxyethylene sorbitan monooleate, Vitamin A or C, tocopherol (Vitamin E), alpha-hydroxy of alpha-keto acids such as pyruvic, lactic or glycolic acids, or any of the topical ingredients disclosed in U.S
  • topical carrier can also be a topical cosmeticaUy acceptable carrier.
  • topical cosmeticaUy acceptable carrier as used herein is meant any substantiaUy non-toxic carrier conventionaUy Useable for topical administration of cosmetics in which the compositions will remain stable and bioavailable when appUed directly to the skin surface.
  • Suitable cosmeticaUy acceptable carriers are known to those of skill in the art and include, but are not limited to, cosmeticaUy acceptable Uquids, creams, oUs, lotions, ointments, gels, or soUds, such as conventional cosmetic night creams, foundation creams, suntan lotions, sunscreens, hand lotions, make-up and make-up bases, masks and the like.
  • cosmeticaUy acceptable carriers and pharmaceuticaUy acceptable carriers are similar, if not often identical, in nature so that most of the earlier discussion on pharmaceuticaUy acceptable carriers also appUes to cosmeticaUy acceptable carriers.
  • compositions can contain other ingredients conventional in cosmetics including perfumes, estrogen, Vitamins A, C or E, alpha-hydroxy or alpha-keto acids such as pyruvic, lactic or glycolic acids, lanolin, vaseline, aloe vera, methyl or propyl paraben, pigments and the like.
  • compositions used to treat dermatologic conditions or diseases can vary depending on such factors as condition of the skin, age of the skin, the particular ratio of phospholipids or degree of the purity of phosphoUpids employed, the type of formulation and carrier ingredients used, frequency of administration, overall health of the individual being treated and the like.
  • the precise amount for any particular patient use can be determined by those of skill in the dermatologic art taking into consideration these factors and the present disclosure.
  • the composition is administered in at least two doses and no more than about six doses per day, or less when a sustained or delayed release form is used.
  • compositions for topical, oral and parenteral administration usually contain from about 0.001% to about 10% by weight of the LPA compared to the total weight of the composition, preferably from about 0.01% to about 2% by weight of the mixture to the pharmaceutical composition, and especially from about 0.1% to about 1.5% by weight of the mixture to the pharmaceutical composition.
  • the topical composition is administered by applying a coating or layer to the skin or mucosal area desired to be treated.
  • a coating or layer is applied to the skin or mucosal area desired to be treated.
  • the applied material is rubbed into the area. Applications need not be rubbed into the skin and the layer or coating can be left on the skin overnight.
  • compositions suitable for transdermal administration including, but not Umited to, pharmaceuticaUy acceptable lotions, suspensions, oils, creams, ointments, rinses, gels and liposomal carriers suspended in a suitable vehicle in which a therapeutically effective amount of the compositions has been admixed.
  • Such compositions are appUed directly to the skin or inco ⁇ orated into a protective carrier such as a transdermal device (so-called "patch").
  • a transdermal device so-called "patch”
  • suitable creams, ointments etc. can be found, for instance, in the Physician's Desk Reference.
  • suitable transdermal devices are described, for instance, in U.S. Patent No. 4,818,540 (
  • the present invention includes compositions suitable for parenteral administration including, but not Umited to, pharmaceuticaUy acceptable sterile isotonic solutions. Such solutions include, but are not Umited to, saline and phosphate buffered saline for intravenous, intramuscular, intraperitoneal or subcutaneous injection.
  • compositions suitable for gastrointestinal administration including, but not limited to, pharmaceuticaUy acceptable powders, pills or Uquids for ingestion and suppositories for rectal administration.
  • the present invention includes compositions suitable for transbronchial and transalveolar administration including, but not Umited to, various types of pharmaceutically acceptable aerosols for inhalation, both liquid and powder forms.
  • An example of a drug administered in the form of an aerosol is pentamidine which is administered to AIDS patients by inhalation to prevent pneumonia caused by Pneumocystis carnii.
  • the present invention encompasses methods of delivery including, but not Umited to, delivery by catheter inserted into a vessel. Where deUvery of the LPA containing compositions is desired to prevent or minimize damage resulting from cardiac ischemia the present invention encompasses intracoronary delivery by guide catheter.
  • the present invention further encompasses devices suitable for transbronchial and transalveolar administration of the compositions.
  • Such devices include, but are not Umited to, atomizers and vaporizers.
  • the present invention also includes devices for electrical or direct injection. Electrical injection, or iontophoresis, is the process of using a small electrical current to drive charged elements, compounds and drugs through the skin to deliver the therapeutic compound to the local tissues or to the whole body without breaking the skin.
  • the present invention encompasses solutions suitable for flushing, perfusion, and storage of organs and tissues prior to or during transplantation. Such solutions are described in Chien et_al. (1993) "Hibernation Induction Trigger for Organ Preservation" in Medical InteUigence Unit,
  • compositions described herein can be used, for instance, to replace and improve on much more impure soy preparations currently in use.
  • organ preservation solution an aqueous solution specifically designed to preserve organs.
  • the organ is the heart.
  • the solutions are used in organ transplantation, but are also useful for use in cardioplegia during open heart surgery.
  • the OPS can also be used to flush the organ to be transplanted either prior to or after harvesting, or both.
  • this solution contains between about 0.00001% to about 10%, preferably about
  • this solution additionally contains between about 0.1% and 20% by weight PEG. More preferably, this solution contains between about 2% and 15% by weight PEG and most preferably, this solution contains about 8% by weight PEG.
  • the PEG is PEG20L.
  • this solution contains between about 2% and 15 % by weight PEG and can also contain effective amounts of: (a) a buffer such as NaOH, preferably about 30-40 mM, (or sufficient to result in pH of 7.2 to 7.9); (b) an impermeant anion such as Lactobionic acid, preferably about 100 mM; (c) a component providing phosphate such as KH 2 P0 4 , preferably about 25 mM; (d) a component providing potassium such as KOH, preferably about 100 mM; and (e) a component controlling cell swelling such as Raffinose, preferably about 30 mM.
  • a buffer such as NaOH, preferably about 30-40 mM, (or sufficient to result in pH of 7.2 to 7.9)
  • an impermeant anion such as Lactobionic acid, preferably about 100 mM
  • a component providing phosphate such as KH 2 P0 4 , preferably about 25 mM
  • a component providing potassium such as KOH, preferably
  • the OPS also contains effective amounts of any component known in the art of organ preservation. These include, but are not Umited to glutathione, parahydroxyanisole (PHA), desferoxamine, and nitroglycerin.
  • PHA parahydroxyanisole
  • desferoxamine desferoxamine
  • nitroglycerin any component known in the art of organ preservation.
  • compositions are meant to describe, but not limit, the compositions suitable for use in the invention.
  • the methods of producing the various compositions are within the ability of one skilled in the art and are not described in detail here.
  • suitable devices for injection, topical application, atomizers and vaporizers are known in the art and will not be described in detail.
  • the invention further provides methods of treatment comprising administering an amount of the compositions effective to inhibit apoptosis or to preserve or restore cell, tissue or organ function. These methods entail administration of a therapeuticaUy effective amount of the above-described compositions.
  • Various indications which can be treated include, but are not Umited to, those related to apoptosis, ischemia, traumatic injury and reperfusion damage.
  • Those conditions related to apoptosis include, but are not Umited to, dermatological effects of aging, the effects of reperfusion after an ischemic event, immunosuppression, gastrointestinal perturbations, cardiovascular disorders, rejection of tissue transplantation, wound healing and Alzheimer's disease.
  • the treatment can also diminish the apoptosis-related problems associated with immunosuppressing viruses, chemotherapeutic agents, or radiation and immunosuppressive drugs.
  • compositions are also suitable for use in organ transplantation during aU phases of transplantation.
  • the compositions can be used to prepare the organ by administering an amount of the compositions to the donor effective to stabilize or preserve the organ.
  • the organ can be perfused and/or preserved in OPS containing the compositions.
  • the organ recipient can then be administered an amount of the compositions effective to enhance organ stabiUty and function.
  • the compositions are also particularly suitable for use in treating cardioplegia whether related to transplantation or other surgical intervention.
  • Apoptosis related problems are caused by a variety of stimuli which include, but are not Umited to, viruses including, but not Umited to, HTV, chemotherapeutic agents, and radiation. These stimuli trigger apoptosis in a variety of disorders, including, but not Umited to, those of the digestive tract tissues and associated gastrointestinal perturbations.
  • Gastrointestinal perturbations include, but are not Umited to, damage to the lining of the gut, severe chronic ulcers, colitis, radiation induced damage, chemotherapy induced damage, and the perturbation of the gastrointestinal tract caused by parasites, and diarrhea from any other cause.
  • Various viral and bacterial infections are known to result in gastrointestinal perturbations; the compositions are also suitable for use in treatment of the side effects associated with these infections.
  • the compositions are particularly suited for use in ameliorating the gastrointestinal disturbances associated with chemotherapy.
  • the compositions are suitable for use not only in preventing the diarrhea associated with chemotherapy but also the nausea.
  • the compositions are particularly suited to treatment of various gastrointestinal conditions in animals, including, but not
  • gastrointestinal conditions are preferably by gastrointestinal administration.
  • an effective amount of the compositions can be conveniently mixed in with the feed.
  • administration can be by any method known in the art of gastrointestinal administration.
  • administration is oral.
  • compositions can be administered to immunodeficient patients, particularly HIV-positive patients, to prevent or at least mitigate apoptotic death of T ceUs associated with the condition, which results in the exacerbation of immunodeficiencies as seen in patients with AIDS.
  • administration to such patients is parenterally, but can also be transdermaUy or gastrointestinaUy.
  • compositions can also be administered to treat apoptosis associated with reperfusion damage involved in a variety of conditions, including, but not Umited to, coronary artery obstruction; cerebral infarction; spinal/head trauma and concomitant severe paralysis; reperfusion damage due to other insults such as frostbite; coronary angioplasty, blood vessel attachment, limb attachment, organ attachment and kidney reperfusion.
  • Myocardial and cerebral infarctions are caused generally by a sudden insufficiency of arterial or venous blood supply due to emboli, thrombi, or pressure that produces a macroscopic area of necrosis; the heart, brain, spleen, kidney, intestine, lung and testes are likely to be affected.
  • CeU death occurs in tissue surrounding the infarct upon reperfusion of blood to the area; thus, the compositions are effective if administered at the onset of the infarct, during reperfusion, or shortly thereafter.
  • the invention includes methods of treating reperfusion damage by administering a therapeutically effective amount of the compositions to a patient in need of such therapy.
  • the invention further encompasses a method of reducing the damage associated with myocardial and cerebral infarctions for patients with a high risk of heart attack and stroke by administering a therapeutically effective amount of the compositions to a patient in need of such therapy.
  • treatment of such damage is by parenteral administration of the compositions of the invention.
  • Any other suitable method can be used, however, for instance, direct cardiac injection in the case of myocardial infarct. Devices for such injection are known in the art, for instance the Aboject cardiac syringe.
  • the invention further provides methods of limiting and preventing apoptosis in cells, or otherwise preserving cells, during the culture or maintenance of mammaUan organs, tissues, and ceUs, by the addition of an effective amount of the compositions to any media or solutions used in the art of culturing or maintaining mammaUan organs, tissues, and cells.
  • the invention further encompasses media and solutions known in the art of culturing and maintaining mammaUan organs, tissues and cells, which comprise an amount of the compositions effective to preserve or restore cell, tissue or organ function, or limit or prevent apoptosis of the ceUs in culture.
  • mammaUan cell culture media comprising an effective amount of at least one composition and the use of such media to preserve or restore cell, tissue or organ function, or to limit or prevent apoptosis in mammalian cell culture.
  • An effective amount is one which decreases the rate of apoptosis and/or preserves the cells, tissue or organ.
  • the compositions have been found to limit or prevent apoptosis under circumstances in which cells are subjected to mild traumas which would normally stimulate apoptosis.
  • Such traumas can include, but are not Umited to, low level irradiation, thawing of frozen cell stocks, rapid changes in the temperature, pH, osmolarity, or ion concentration of culture media, prolonged exposure to non-optimal temperature, pH, osmolarity, or ion concentration of the culture media, exposure to cytotoxins, disassociation of cells from an intact tissue in the preparation of primary cell cultures, serum deprivation (or growth in serum-free media).
  • the invention encompasses compositions comprising tissue culture medium and an effective amount of the compositions.
  • Serum-free media to which the compositions can be added as anti-apoptotic media supplements include, but are not limited to, AIM V® Media, Neuman and Tytell's Serumless Media, TroweU's T8 Media,
  • suitable mammalian cell culture media to which the compositions can be added as anti-apoptotic media supplements include, but are not Umited to, Basal Media Eagle's, Fischer's Media, McCoy's Media, Media 199,RPMI Media 1630and 1640, Media based on F-10&F-12 Nutrient Mixtures, Leibovitz's L-15 Media, Glasgow Minimum Essential Media, and Dulbecco's Modified Eagle
  • MammaUan cell culture media to which the compositions can be added further comprise any media supplement known in the art, including, but not Umited to, sugars, Vitamins, hormones, metaUoproteins, antibiotics, antimycotics, growth factors, lipoproteins and sera.
  • the invention further encompasses solutions for maintaining mammaUan organs prior to transplantation, which comprise an effective amount of the compositions, and the use of such solutions to preserve or restore organ function or to limit or prevent apoptosis in such mammaUan organs during their surgical removal and handling prior to transplantation.
  • the solutions can be used to flush, perfuse and/or store the organs.
  • concentrations of the compositions required to limit or prevent damage to the organs can be determined empirically by one skiUed in the art by methods such as those found in the example provided below, as well as other methods known in the art.
  • compositions can be topically appUed to the skin to treat a variety of dermatologic conditions. These conditions include, but are not limited to, hair loss and wrinkling due to age and/or photo damage.
  • the present invention thus encompasses methods of treating dermatological conditions.
  • hair loss can be caused by apoptosis of the ceUs of the hair follicles. Therefore, the compositions are suitable for use in topical treatment of the skin to prevent continued hair loss. Stenn et al. (1994) J.Jnvest. Dermatol. 103:107-111. As discussed above, these conditions are preferably treated by topical application of a composition comprising an effective amount of the compositions.
  • An effective amount of the compositions is one which ameliorates or dirninishes the symptoms of the dermatologic conditions.
  • the treatment results in resolution of the dermatologic condition or restoration of normal skin function; however, any amelioration or lessening of symptoms is encompassed by the invention.
  • EXAMPLE 1 Structure and Synthesis of Various Analogs of Lysophosphatidic Acid
  • the following example sets forth the synthetic methodology and analytical data used in the construction and characterization of several lysophosphatidic acid analogs and derivatives.
  • experimental procedures and analytical data has been provided for intermediates used in the construction of these LPAs.
  • reaction schemes which show the synthetic routes used in compound construction.
  • the following nomenclature and abbreviations are used in the naming of the compounds:
  • the obtained oil was treated with 1/1 2M HCl THF (150 mL, v/v) for 16 hours.
  • the resulting mixture was concentrated and redissolved in ethyl acetate (150 mL), washed with H 2 O (3 x 50 mL), dried (MgS0 4 ) and evaporated to dryness.
  • the crude product was purified by siUca-gel column chromatography [eluent: CH 2 Cl 2 (ethyl acetate/CH 2 Cl 2 , 50/50, v/v] to yield diol Compound 2 (8.4 g, 73%) as a white soUd.
  • the residual oU was purified by silica-gel column chromatography [eluent: ethyl acetate hexane, 91/9, v/v] to furnish epoxide Compound 10 (1.65 g, 72%) as a colourless oil.
  • Alcohol Compound 40 (48.8 g, 0.16 mmol) was added to a mixture of NaH (4.0 g, 0.17 mol) in dry THF (500 mL). After 5 min, Mel (10 mL, 0.17 mol) was added and, after another 3 hours, the mixture was quenched with H 2 0 (150 mL), extracted into diethyl ether (500 mL), dried (MgS0 4 ) and concentrated.
  • ESI-MS (m/z, +ve): 581 (MSa + , 100%); ESI-MS (m/z, -ve): 504 ( ⁇ f-NCCtf 2 CH 2 , 100%).
  • Example 2 Anti-apoptotic Activity Assay In order to determine the apoptotic activity of the claimed invention, the following method of analysis was used. The cell assay is described in detail in U.S. Patent Nos. 5,637,486, 5,620,888, and 5,681,703, and Tomei et al. (1993) Proc Natl. Acad. Sci. 90:853-857. Briefly, mouse fibroblast C3B/10T1/2 cells (clone 8) were obtained from ATCC (RockviUe, MD) and were maintained in exponential growth phase in which the cell cycle is randomly distributed and no cells are arrested in Go, and in quiescence.
  • AU non-adherent or loosely adherent cells were removed from the culture dish and counted by appropriate techniques, typically counting by electronic particle counting instrument. These are the apoptotic cells, the serum deprived released cells (SDR), released by the action of cultivation in serum-free medium. Approximately 95% of these released cells are apoptotic as shown by both ultrastructure analysis and DNA fragmentation analysis.
  • SDR serum deprived released cells
  • the remaining adherent cells were exposed to a buffered, typicaUy pB_7.3, balanced salt solution such as Hanks Balanced Salt Solution without calcium and magnesium salts containing 0.05% trypsin and 0.53 mM ethylene cUaminetetraacetic acid (EDTA).
  • Each culture was incubated at either room temperature or 37(C on a rocking platform to ensure uniform distribution of the trypsin reagent over the culture surface.
  • typicaUy 10 minutes the released cells were removed from each culture dish and measured by the same means as described above, typically electronic particle counting.
  • This ADH cell count is comprised of both trypsin resistant and trypsin sensitive ceUs as described in U.S. Patent Nos.
  • Anti-apoptotic activity is expressed in the following examples as the calculated concentration of material ((g/ml of media) required to save 50% of the ceUs released on serum free treatment.
  • Example 3 Preparation of Five PhosphoUpid Mixture
  • PA, PI, LPI, LPC available, for example, from Avanti® Polar Lipids, Inc.
  • PA, PI, LPI, LPC available, for example, from Avanti® Polar Lipids, Inc.
  • PA, PI, LPI, LPC available, for example, from Avanti® Polar Lipids, Inc.
  • PA, PI, LPI, LPC available, for example, from Avanti® Polar Lipids, Inc.
  • Total concentrations of phospholipids of greater than 10 mg/mL can be used provided that clarity is obtainable upon sonication.
  • Total concentrations of up to about 50 mg/mL have been utilized.
  • the phosphoUpid mixtures are suspended in a buffer and the mixture is placed in a disposable borosilicate glass, preferably 1-2 mL in a 16 x 100 mm tube or 0.5-2 mL in a 13 x 100 mm tube, or up to 1 mL in a 12 x 75 mm tube.
  • the combination of phosphoUpids is then sonicated.
  • a small bath sonicator is used, such as a that sold by Laboratory SuppUes, HicksviUe, NY.
  • the temperature of the water bath is between about 21 and 50°C, preferably between about 21 °C and about 40°C. The optimal temperature depends on the phospholipids used and can be determined empirically.
  • the water level is adjusted so that it is approximately the same height as the phosphoUpid mixture in the glass tube(s).
  • a probe sonicator can be used (Fisher Scientific Sonic Dismembrator model 550), as long as care is taken to prevent overheating of the mixture.
  • the mixture was sonicated for between 3 and 90 minutes, with alternating 5 minute intervals of sonication foUowed by 5 minutes of thermal equilibration, in a 1-2 ml volume until the mixture became translucent and passed readily through a filter attached to a 5 ml syringe with a pore size of 0.22 (m.
  • sonication is for 5-10 minutes.
  • compositions were stored for one week at 4°C, room temperature, and 65 °C .
  • results show loss of activity after storage at 65°C, while the compositions stored at 4°C or at room temperature do not have a significant loss of activity.
  • the concentration of LPA was varied as was the chain length to determine the effects of these parameters on activity.
  • LPA Anti-apoptotic activity of LPA
  • Log phase cells were seeded in 60 mm Perri dishes at 175-350 ceUs per cm2 and maintained in Basal Medium Eagle (BME) supplemented with 10% Beat Inactivated Fetal Bovine Serum (HIFBS). On day 3 the cells were given fresh media. Treatment began on day 5 when the media containing serum was removed and replaced with the LPA mixture to be tested.
  • BME Basal Medium Eagle
  • HIFBS Beat Inactivated Fetal Bovine Serum
  • SDR serum deprived released
  • ADH adherent
  • Adherent cells were remove by treatment with 5 mL of Hanks Balanced Salt Solution (HBSS) without ions containing 0.05%trypsin and 0.53 mMEDTAand were counted with the lower threshold setting at 11.01 (m. AU samples were tested in tripUcate and serum-deprived controls (BME only) were assayed at both the beginning and end of each experiment.
  • HBSS Hanks Balanced Salt Solution
  • the dose shown is the lowest concentration at which cytotoxic effects were observed.
  • LPA LPA was incorporated into Upid membrane structures of various Upid compositions . Except in the Five PhosphoUpid Mixture treatments, LPA was presented to the cells as 18: 1-LPA, and was tested in all preparations at four concentrations: 0.25, 0.75, 2.25 and 6.75
  • Positively charged particles containing 18: 1-LPA combined with the neutral phosphoUpid phosphatidyl choline (PC) and 20 mol% of the positively charged lipid l,2-dioleoyl-3-trimethylammonium-propane (TAP) were efficacious in protecting 70 to 80 % of ceUs, compared with PC/TAP alone, offered no protection over control.
  • 18:1 -LPA into the negatively charged phosphatidyl glycerol (PG) gave even better protection, with up to 90% of ceUs protected as compared to alone which offered no protection over control.
  • inclusion in the 18:1-LPA/PG particles of 5 to 10 mol% of the neutral phosphoUpid phosphatidylethanolamine (PE) with a long polyethylene glycol (2000 MW) covalently linked to the polar head group resulted in reduced efficacy of 18 : 1 -LPA.
  • PE neutral phosphoUpid phosphatidylethanolamine
  • LPA was then tested in two different weight ratios with PG, using 18: 1-LPA in concentrations representing LPA equivalents to Five PhosphoUpid Mixture at 1, 3, or 10 (g/ml. As shown in Fig. 3, LPA equivalent to 10 (g/ml Five PhosphoUpid Mixture incorporated into particles at 10% by weight with PG protected approximately 65% of ceUs from apoptosis. This degree of protection was roughly the same as that afforded by Five Phospholipid Mixture alone.
  • LPS lysophosphatidyl serine
  • Soy derived LPA (soy-LPA) (Avanti® Polar Lipids, Inc., Alabaster, AL) was stored in an organic solution and was dried immediately prior to assay using a Speed Vac (Savant Instruments, HicksvUle, NY). Tested proteins were resuspended at lOjmg/mL in a bicarbonate buffered saline EDTA solution (50_mM NH,BC0 3 , 104 mM NaCl, 250_(M EDTA) prior to being added to dried soy-LPA at a 1 : 10_soy-LPA to protein (weightweight) ratio.
  • the mixture was then sonicated for 5 minutes using a high power 80 Watt sonication bath (Laboratory SuppUes Co., Hicksville, NY). Following sonication, concentrated LP A/protein mixtures were left standing for 15 minutes at room temperature prior to dilution in cell culture assay media.
  • SDR serum deprived release
  • ADH adherent
  • Adherent cells were removed by treatment with a Hanks' balanced salt solution containing 0.05% trypsin and 0.53_mM EDTA and were counted with the lower threshold setting at 11.0 l_(m. AU samples were tested in tripUcate and serum-deprived controls (BME only) were assayed at both the beginning and end of each experiment.
  • soy-LPA When soy-LPA was added together with protein, the degree of protection from apoptosis was, for certain proteins, more than additive.
  • FAD fatty acid depleted bovine serum albumin
  • BSA bovine serum albumin
  • soy-LPA alone protected 8.0% of cells, wh ⁇ e FAD BSA plus soy-LPA saved 58.7% of the cells.
  • Similar effects were seen for mixtures of soy-LPA with FAD soy protein, cytochrome c and low density Upoprotein (LDL) and, to a lesser degree, acyl carrier protein, casein and myoglobin. Soybean trypsin inhibitor, ovalbumin, retinol binding protein and myelin basic protein did not enhance the anti-apoptotic activity of LPA, while bromolain was toxic to the ceUs.
  • Binding of LPA to protein was examined using an electrophoresis radiobinding detection assay and a size shift analysis assay.
  • Electrophoresis radiobinding assay Proteins were analyzed on native polyacrylamide gels foUowing incubation with 3 B-labeled 18: 1-LPA. 3 B-labeled lysophosphatidic acid (1 -oleyl) (DuPont NEN Products, Boston, MA) was added to tested proteins at 1 nM labeled 18:l-LPA/45_(g protein. Mixtures were incubated in a bicarbonate buffered saline/EDTA solution at 37°C for 60 minutes. Incubated samples were mixed with native gel sample buffer (312_mM Tris pB 6.8, 50% glycerol, 0.05% bromophenol blue) and loaded entirely onto an 8% discontinuous native polyacrylamide gel.
  • native gel sample buffer 312_mM Tris pB 6.8, 50% glycerol, 0.05% bromophenol blue
  • proteins were fixed into the gels using an aqueous solution consisting of glacial acetic acid, 10% (vol/vol) and methanol 30% (vol./vol.) solution. Fixed gels were then incubated for 60 minutes in autoradiography enhancer (EN 3 HANCE, DuPontNEN Products, Boston, MA), and dried onto 3M chromatography paper (Whatman, Clifton, NJ) and exposed to autoradiography film (X-Omat, Kodak, Rochester, NY) for 72 hours at -80°C.
  • autoradiography enhancer EN 3 HANCE, DuPontNEN Products, Boston, MA
  • Size shift assay Size analysis of LPA both with and without protein was performed using a Superdex® S75 column (Pharmacia Biotech, Uppsala, Sweden). Soy LPA was combined in a chloroform solution with 0.5 Ci 3 B-labeled 18:l-LPA(DuPontNENProducts,Boston,MA),dried under vacuum and resuspended by sonication in a buffered solution (50 mM ammomum bicarbonate, 154 mM sodium chloride, pB 8.0) in the presence or absence of test protein. The ammonium bicarbonate/sodium chloride solution was used as the mobile phase at a flow rate of 1 ml/min. One-minute fractions were collected for 30 minutes following sample injection. Fractions were counted in scintiUant solution using a Uquid scintillation counter (Beckman
  • soy-LPA alone eluted at 22-26 minutes after sample injection, corresponding to a size of approximately 1.3 kD.
  • radioactivity eluted primarily in the 10 to 12-minute fractions, indicating that most of the LPA was bound to BSA.
  • a LP A/FAD soy protein mixture appUed to the column resulted in an elution profile with two peaks, one corresponding to the position of unbound LPA, and the other shifted.
  • a cytochrome c/LPA mixture gave an elution profile similar to that of soy-LPA alone, indicating that LPA does not bind to this protein.
  • the foUowing experiment was performed.
  • TPA 12-0-tetradecanoyl phorbol- 13 -acetate
  • AU non-adherent or loosely adherent cells were removed from the culture dish and counted by appropriate techniques, typicaUy counting by electronic particle counting instrument. These were the apoptotic cells, the serum deprived released cells (SDR), released by the action of cultivation in serum-free medium. Approximately 95% of these released cells were apoptotic as shown by both ultrastructure analysis and DNA fragmentation analysis.
  • SDR serum deprived released cells
  • the remaining adherent cells were exposed to a buffered, typicaUy pH_7.3, balanced salt solution such as Hanks Balanced Salt Solution without calcium and magnesium salts containing 0.05% trypsin and 0.53_mM ethylene maminetetraacetic acid (EDTA).
  • Each culture was incubated at either room temperature or 37°C on a rocking platform to ensure uniform distribution of the trypsin reagent over the culture surface.
  • typicaUy 10_minutes the released cells were removed from each culture dish and measured by the same means as described above, typicaUy electronic particle counting.
  • This ADH cell count was comprised of both trypsin resistant and trypsin sensitive cells as described in PCT Publication No._WO_94/25621.
  • Anti-apoptotic activity is expressed as the calculated concentration of material ((g/ml of media) required to save 50% of the cells released on serum free treatment.
  • Argon was dried in a glass tube with vacuum concentrator (Savant) with gentle heat. This was sonicated for 5 minutes under an Argon blanket in 1 mL of buffer (50 mM NB ⁇ CO ⁇ 104 mM
  • the septa Prior to use, the septa had been subjected to 2 hours of high vacuum then sealed in an aluminum bag filled with Argon in order to displace as much dissolved oxygen as possible.
  • the Five PhosphoUpid Mixture/PEG vials were labeled then individually packaged into aluminum bags filled with Argon and stored at 4°.
  • phospholipid mixtures were prepared by combining the desired phosphoUpids in solution with organic solvent then drying down the mixture with gentle heat (45 °C) and high vacuum foUowed by sonication into aqueous suspension at 5 to 20 mg/mL.
  • the phospholipids were then mixed with serum-free culture medium containing PEG of the indicated molecular weight, concentration and type of PEG noted in the figures and prepared as described above. The results obtained are depicted in Fig. 11 and Table 3.
  • Example 11 in vitro Cardiomyocyte Assays Isolation of rat neonatal cardiomvocvtes Cardiomyocytes were prepared from hearts of day-old Sprague Dawley rats by trypsinization and mechanical disaggregation (Simpson, (1985)) Circ. Res.56:884-894. The cells were resuspended in MEM, lxMEM vitamins (Gibco), 5% fetal bovine serum and 50 U/ml peniciUin-G and pre-plated for 30 minutes to reduce contamination of non-myocytes. The non-adherent cardiac myocytes were separated and seeded in 2 ml in 35 mm dishes at a density of 3.5xl0 5 viable cells per ml. The cells were aUowed to adhere for 16-24 hours in a 37°C/5% C0 2 humidified incubator.
  • the medium was replaced with fresh serum-free RPMI, whereas serum/glucose deprivation was performed using glucose-free RPMI.
  • the induction of cell death by adriamycin or C 2 -ceramide was accomplished by the addition of the agent prepared in serum-free RPMI.
  • cultures in serum and glucose free RPMI were placed in an airtight chamber and the latter was continuously perfused with oxygen-free gas overlay of 95% N 2 /5% C0 2 for 8 hours at 37°C.
  • 10% fetal bovine serum, 2 g/L of glucose were added and the cultures were returned to a normal oxygen gas overlay (37°C/5% CO 2 ) in a humidified incubator for 16 hours.
  • cardiomyocytes are terminally differentiated non-dividing cells, viability was determined by measurement of the decrease in the relative number of adherent cells. The measurement of non-adherent cells was found to be less reproducible because of their rapid lysis foUowing release from adhesion substrate.
  • Adherent cardiomyocytes were coUected from culture dishes using 0.25% Trypsin/0.05% EDTA and counted on Coulter Counter ZM and Coulter Channelyzer 256.
  • 15(M ceramide was added to cardiomyctes alone or in the presence of: (a) 0.3% PEG, (b) 2% PEG, (c) 5(M LPA presented as Five Phospholipid Mixture, (d) 5 (M LPA presented as Five PhosphoUd Mixture plus 0.4% PEG, and (e) 5 (M LPA presented as Five PhosphoUd Mixture plus 2% PEG.
  • LPA has anti-apototic activity as shown in the cardiomyocyte assay, and that the combination of LPA with other phosphoUpids retains that activity. Additionally, the presence of PEG improves the activity of LPA.
  • the experimental model used was a regional ischemic dog heart (mongrel male hound, approximately 20 kg) with the left anterior descending coronary artery clamped distal to the first diagonal branch rendering a portion ofthe left ventricle ischemic. The clamp was maintained for 90 minutes then released gradually over a 5 minute period.
  • a narrow canula was placed into the left main coronary artery through which Five Phospholipid Mixture (5 mL of 50 (g/mL in 10% PEG) was infused starting 75 minutes after the start ofthe ischemic period, and continuing for 75 minutes in total (approximately 67 (IJmin., approximately 3.3 (g Five PhosphoUpid Mixture/min.). The total dose was 250 (g Five PhosphoUpid Mixture and 500 mg PEG20L.
  • the atria and right ventricle are removed from the left ventricle. After removal ofthe apex, the portion ofthe left ventricle distal to the mitral apparatus is cut into 5 transverse sections of equal thickness.
  • Evans Blue dye injected into the circumflex vessel stains the circumflex territory blue.
  • TTC stains viable LAD territory red, while the infarcted myocardium will remain unstained (white). Sections are weighed and photographed for documentation and future verification of results .
  • Computer assisted planimetry is used to measure the areas ofthe circumflex, viable LAD and infarcted LAD territories. Calculation of infarct size is based upon the assumption that the area of infarction in the sectioned plane is representative of the mass of infarcted tissue in that plane.
  • PEG 100_mg/ml as a bolus plus infusion (at 4_n ⁇ 7hr) starting 15 min before reperfusion and going on for 60 min into the reperfusion period.
  • Placebogroup dogs receive placebo foUowing the same modalities described for the study drug group.
  • SOD-Catalase group dogs receive SOD plus catalase following the same modaUties described by Simpson et_al. (1987) Fed. Proc. 46:7, 2413-21.
  • Heart function was monitored by EKG and cardiac output was determined by contrast imaging. The animal was euthenized 1-3 days after the occlusion. The heart was removed for histology and examined. Infarct size was determined by a method of triphenyl tetrazolium chloride
  • TTC TTC staining for the area at risk and actual infarct.
  • the area not at risk was stained with a blue pigment or Evan's Blue dye.
  • LAD left anterior descending coronary artery
  • the chest was opened through the left fourth intercostal space. Then, the pericardium was incised and the heart was exposed. A large anterolateral branch ofthe circumflex artery, or the circumflex artery itself was identified and encircled with a 4-0 silk suture. The ends ofthe suture were threaded through a piece of flanged tubing, forming a snare, which was later used to occlude the artery. A catheter was then placed into the left atrial appendage to inject the control or test solution, the radioactive microspheres and blue pigment at the conclusion ofthe procedure.
  • the rabbits were stabilized for ten minutes. At this time, the rabbits were randomized into either the treatment or control group.
  • the treatment solution contained 1 mg/ml ofthe Five PhosphoUpid Mixture, described in Example 3, above, 8 mg/ml PEG 20L, in 10 mM citrate 110 mMNaCl.
  • the control solution contained 10 mM citrate and 110 mM NaCl.
  • a bolus dose of 1 mg/kg body weight of either the Five Phospholipid Mixture/PEG solution or the control solution was administered into the left atrium.
  • infusion was started at the rate of 1 mg/kg for one hour into the left atrium. Baseline hemodynamics and core body temperature were recorded.
  • the artery was occluded for 30 minutes. FoUowing this, the hearts were reperfused for three hours.
  • Heart rate and arterial blood pressure were recorded at 20 minutes of occlusion and at time points during reperfusion at a 25 mm/second paper speed.
  • Regional myocardial blood flow measurements were performed on two occasions; during coronary occlusion (20 minutes) to confirm no blood flow in the ischemic zone, and during early reperfusion (30 minutes) to confirm reflow in the same zone.
  • the coronary artery is reoccluded.
  • Unisperse blue (Ciby-Geigy, Hawthorne, NY) were infused through the left atrial catheter and allowed to circulate throughout the vascular system. The rabbit was then euthanized by an overdose intravenous injection of xylazine (300 mg) foUowed by 12 mEq of potassium chloride given into the left atrium.
  • Prospective exclusion criteria included an ischemic risk zone of less than 10% ofthe left ventricular weight, a regional blood flow of more than 0.2 ml/minute/g in the risk zone during coronary artery occlusion (lack of ischemia), or a regional blood flow of less than 0.4 ml/min/g in the risk zone at 30 minutes of reperfusion (failure to reperfuse).
  • Infarct size was evaluated as follows. The right ventricle was trimmed off and the left ventricle was sliced transversely into seven or eight sections, approximately two millimeters in thickness. These slices were photographed to identify the ischemic risk regions (uncolored by the blue pigment) and the non-ischemic regions (colored by the blue pigment). The slices were then incubated in a 1% solution of triphenyltetrazoUum chloride pre-heated to 37 °C for 10 minutes and rephotographed for analysis of area of necrosis. AU sections were later fixed in formalin. These photographic slides were projected and areas of risk (AR) and areas of necrosis (AN) were traced by planimetry. The planimetered areas of each sUce were multiplied by the weight ofthe sUce and then summed. Because infarct size is measured from photographic sUdes, the entire left ventricle was used for the analysis.
  • AR area of risk
  • AN necrosis
  • RMBF Regional myocardial blood flow
  • Radioactive microspheres were injected via the atrial catheter.
  • the catheter was then reprimed with approximately 0.2 ml ofthe drug treatment, and the catheter was reconnected to the pump and infusion continued.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention provides anti-apoptotic compositions containing lysophosphatidic acids and methods for making and using the compositions. Such compositions can also contain LPA potentiating agents, including proteins, lipid membrane structures and polymers such as polyethylene glycols. The compositions can additionally contain other pharmaceutically effective agents such as drugs, antibiotics, wound healing agents and antioxidants.

Description

COMPOSITIONS CONTAINING LYSOPHOSPHATIDIC ACIDS WHICH INfflBIT APOPTOSIS AND USES THEREOF
TECHNICAL FIELD This invention relates to therapeutically effective compositions of matter. More specifically, it encompasses compositions containing lysophosphatidic acid or analogs and derivatives thereof, (collectively "LPA") which have been found to exhibit anti-apoptotic activity and or to preserve or restore cell, tissue or organ function. This invention specifically relates to therapeutically effective reverse ester, reverse thioester, and bis-LPA analogs encompassed by the term LPA. Additionally, this invention relates to methods of use of these therapeutically effective compositions.
BACKGROUND OF THE INVENTION Phospholipids. Phospholipids are a class of amphipathic phosphorus-containing lipids which are essential constituents of biological membranes. Various phospholipid preparations have been used for cooking, drug delivery (liposomes), slow release delivery systems, carrier media for hydrophobic drugs, gene transfer and replacement therapy, sunscreens, emulsions, anti-foaming agents, replacement of damaged or absent pulmonary surfactants, detergents and membrane stabilization. Phosphatidic acid (PA), phosphatidylinositol (PI), lysophosphatidic acid, lysophosphatidylinositol
(LPI), and lysophosphatidylcholine (LPC) are found in a variety of plant and animal products. Lysophosphatidic acid analogs have been reported to have a variety of physiological activities including mitogenesis (i.e. prevention of hyperproliferative diseases), vasodilation, growth factor, wound healing and to be an anti-wrinkle agent. In addition, previous studies have shown that lysophosphatidic acid, when bound to serum albumin, can activate membrane currents v Xenopus oocytes and induce neurite retraction in PC 12 pheochromocytoma cells. Apoptosis.
Cell death can occur by necrosis and apoptosis. is generally results from catastrophic irreversible cell damage. It is characterized by early swelling of the cell and its cytoplasmic organelles, with subsequent rupture of the cellular membrane.
Apoptosis is a normal physiologic process that leads to individual cell death. This process of programmed cell death is involved in a variety of normal and pathogenic biological events and can be induced by a number of unrelated stimuli. Changes in the biological regulation of apoptosis also occur during aging and are responsible for many of the conditions and diseases related to aging. Apoptotic cell death appears to play a significant role in the tissue damage that occurs in association with, for example, ischemia, organ transplantation, and various gastrointestinal disorders.
Studies of apoptosis suggest that a common metabolic pathway leading to apoptosis can be initiated by a wide variety of signals, including hormones, serum growth factor deprivation, chemotherapeutic agents, ionizing radiation, and infection by human immunodeficiency virus
(HTV). Apoptosis can also be induced by mild, non-catastrophic cell injury and can be concomitant with adjacent necrosis. Agents that affect the biological control of apoptosis thus have therapeutic utility in numerous clinical indications.
Apoptotic cell death is characterized by morphologic changes such as cellular shrinkage, chromatin condensation and margination, cytoplasmic blebbing, and increased membrane permeability. Specific internucleosomal DNA fragmentation is a hallmark for many, but not all, instances of apoptosis.
Several genes and gene families involved in signal transduction and modulation of apoptosis have been described. Apoptosis, however, is an active cellular response to a physiologic or external signal and can be modulated by interfering with the apoptotic pathway. Conversely, by definition, necrosis can be prevented only by decreasing cell injury. Prevention of apoptosis by upregulation of bcl-2 and bcl-x expression, or by inhibitors of ICE-like proteases are typical examples of modulation of cell death. Vvnaxv-ty Molekylyarnaya Biologiya 30:285-295 (1996); VauxandStrasser N S93:2239-2244 (1996); Nunez etal. (1994) Immunol. Today 15:582-588; and Whyte (1996) Trends in Cell Biol. 6:245-148.
Ischemia and Reperfusion.
Ischemia is the result of decreased blood flow to a particular area or organ of the body. Ischemia is responsible for several important types of physiologic damage such as brain damage, spinal cord trauma and myocardial ischemia. Acute myocardial ischemia leads to the death of individual heart cells which can result in organ dysfunction. Although early reperfusion (i.e., restoration of blood flow) decrease heart damage caused by ischemia, cell death by apoptosis can occur upon reperfusion. In this instance, the cells that die are those that remained viable at the end of ischemia. Support for the role of apoptosis in heart injury induced by ischemia and subsequent reperfusion has been provided by numerous laboratories. Gottlieb et al. (1994) J. Clin. Invest. 94:16211628; Umansky et al. (1995) Cell Death and Differentiation 2:235241; Umansky et al.
(1996) Basic and Applied Myology 6:227235; and Itoh et al. (1995) Am. J. Pathol. 146: 13251331. Severe cell damage during prolonged ischemia appears to result in necrotic death of myocardial cells. However, if the ischemia is relatively limited in extent and duration, the apoptotic pathway is initiated. Restoration of blood flow (reperfusion) allows apoptosis to proceed. Insulin-like growth factors (IGF) and calpain inhibitors, which are capable of preventing apoptosis in different systems, also inhibited apoptosis of cardiomyocytes following ischemia and reperfusion both in vivo and in vitro. Umansky et al. (1995); and Buerke et al. (1995) Proc. Natl. Acad. Sci. USA 92:80318035. Organ preservation.
Transplantation of vital organs such as the heart, liver, kidney, pancreas, and lung has become increasingly successful and sophisticated in recent years. Because mammalian organs progressively lose their ability to function during storage, even at freezing temperatures, transplant operations need to be performed soon after obtaining a donor organ to minimize the period of time that the organ is without blood flow. This need diminishes the availabiUty of organs for transplantation.
In clinical practice, the two major situations in which cardiac preservation is required are heart transplantation and cardioplegia for open heart surgery. In heart transplantation, the donor heart is flushed with approximately 1 liter of cold organ preservation solution (OPS) to arrest the heart. Cooling is supplemented by surrounding the heart with iced saline. The chilled, arrested heart is then surgically excised, immersed in cold OPS, packed in ice and rushed to the recipient center.
After placing the recipient on cardiopulmonary bypass, the diseased heart is excised. The preserved donor heart is then removed from the OPS, trimmed appropriately and transplanted into the recipient. Blood is allowed to flow to the transplanted heart. The transplanted heart will then either resume beating spontaneously or will require chemical and/or electrical treatment to restore normal rhythm. When the heart is ready to take over the circulation, the cardiopulmonary bypass is discontinued and the recipient's chest closed.
Most non-transplant surgical procedures on the heart, such as coronary artery bypass grafting, require that the heart be arrested for a period ranging from 1 to 4_hours. During this time, the heart is kept cool by external cooling as well as by periodically reflushing an OPS through the coronary arteries. The OPS composition is designed to rapidly arrest the heart and to keep it in good condition during the period of standstill so that it will resume normal function when the procedure is finished. In cardioplegic procedures, the heart is exposed in the chest and, at a minimum, the aortic root is isolated. A vascular clamp is applied across the aorta and approximately 1 liter of cold OPS is flushed into the aortic root through a needle. Venting is provided through the left ventricle, pulmonary artery or the right atrium and the effluent, which can contain high levels of potassium, is suctioned out of the chest. This, together with external cooling, produces rapid cessation of contractions. During the period of arrest, the patient's circulation is maintained artificially using cardiopulmonary bypass.
After completion of the surgical procedure, blood flow is restored to the coronary circulation and heartbeat returns either spontaneously or after chemical and electric treatment. The ease with which stable function is restored depends to a large extent on the effectiveness of the
OPS to preserve the heart. Once the heart is beating satisfactorily, cardiopulmonary bypass is discontinued and the chest closed.
It is generally understood that "living" organs, including the heart, continue the process of metabolism after removal from the donor so that cell constituents are continuously metabolized to waste products. If the storage technique is inadequate, the accumulation of these metabolic waste products, depletion of cell nutrients and consequent derangement of cell composition lead to progressive loss of function and ultimately to cell death, with loss of adequate function after transplantation into the recipient. Several procedures have been explored to successfully enable organ preservation ex vivo for useful time periods. In one method, the donor organ is cooled rapidly by flushing cold solutions through the organ's vascular system and maintaining the organ at temperatures near 0°C for the purpose of greatly slowing the metabolic rate. In the case of the mammalian heart, the flush solution composition is designed to cause the heart to rapidly stop beating as well as to preserve it.
In 1988, University of Wisconsin (UW) solution was introduced. Belzer et al. (1988) Transplantation 45 : 673-676. This solution, capable of preserving the pancreas and kidney for 72 hours, and the liver for 30 hours, subsequently became the standard organ preservation solution (OPS) for transplant surgery and the benchmark against which other OPS compositions were measured. However, the heart is more recalcitrant to long-term storage than other organs, and UW solution is unreliable for storage of hearts for as short a period as 24 hours. Wicomb et al. (1989) Transplantation 47:733-734.
Improvements in the design of OPS compositions, as reviewed in Collins et al. (1992) Kidney International 42: S-l 97-S-202 and others described in the art, have proceeded along several paths, including: (1) modification and simplification of UW solution; (2) investigation of organ-specific requirements; (3) addition of pharmacologic agents, particularly calcium antagonists for control of acidosis; (5) the use of a terminal rinse solution; and (6) the use of solutions containing PEG.
Wicomb et al. reported the beneficial effects of a solution of PEG 8000 and horseradish peroxidase on rabbit hearts preserved by oxygenated low pressure perfusion for 24 hours. Wicombetal. (1989) Transplantation Proceedings 21:1366-1368. The substitution of PEG20M as the colloid for hydroxyethyl starch (HES) of the UW solution also yielded excellent cardiac function. PEG20M consists of two or more molecules of PEG 6000-8000 joined by a bisphenol epoxide linker (CAS # 37225-26-6; CAS name Oxirane, 2, 2' [(l-methyl-ethylidene)bis(4, l-phenyleneoxy methylene)]bis-, polymer with (-hydro-(-hydroxypoly(oxy-l,2-ethanediyl). The substitution of PEG20M for HES also allowed baboon heart storage up to 48 hours and increased cardiac output (CO) under conditions of microperfusion. Wicomb et al. (1986) J. Surg. Res. 40:276; and Wicomb et al. (1989) Transplantation 48:6-9. "Microperfusion" is a hypoxic, very-low-flow perfusion with flowrates such as 3 ml/g heart wt/24 hour, which is 1/500 of that typical of conventional continuous perfusion. Wicomb et al. (1989) Transplantation 48:6-9.
An improved OPS, Cardiosol™ heart preservation solution, contained the substitution of PEG20M for HES and eliminated five components of UW solution (penicillin, dexamethasone, insulin, allopurinol, and adenosine). Wicomb et al. (1990) Transplantation 49:261-264; and United States Patent No.4,938,961. Cardiosol™heart preservation solution contains 5% or 10% by weight PEG 20M (Union Carbide Chemicals and Plastics Co., Inc., Charleston, WV), 40 mM sodium, 125 mM potassium, 5 mM magnesium, 25 mM phosphate, 5 mM sulfate, 100 mM lactobionate, 30 mM raffinose, and 3 mM glutathione. Collins et al., The Lancet 338:890-891 (1991); and Wicomb etal. (1994) J. Heart Lung Transplantation 13:891-894. This solution was found to be superior to UW solution both for 4-hour hypothermic and 24-hour microperfusion storage. Collins et al. (1992).
Gastrointestinal Disorders.
A variety of food supplements containing, in part, partially processed plant extracts have been used to ameliorate the gastrointestinal disorders that often accompany chemotherapy, radiation and ADDS. The supplements generally contain carbohydrates, fat and plant protein hydrolysates. See, e.g., Tomei and Cope et al. in Apoptosis: The Molecular Basis of Cell Death
(1991) Cold Spring Harbor Laboratory Press. PCT Publication No. WO 95/15173, U.S. Patent Nos.5,620,885,5,567,425,5,635,186and5,624,672 describe plant-derived extracts thatproduce an anti-apoptotic effect. It has now been found that these extracts contain the following phospholipids: LPA, LPC, LPI, PA and PI in the ratios of approximately 2: 1 :2:20:20, by weight in addition to various optional protein and carbohydrate constituents.
A need exists for improved solutions and methods for preserving or restoring cell, tissue, or organ function, and/or preventing apoptosis for a variety of therapeutic uses, particularly organ preservation. The present invention satisfies this need and provides related advantages, as well. SUMMARY OF THE INVENTION
This invention relates to therapeutically effective compositions of matter. More specifically, it encompasses compositions containing lysophosphatidic acid or analogs and derivatives thereof, (collectively "LPA") which have been found to exhibit anti-apoptotic activity or to preserve or restore cell, tissue or organ function. Additionally, this invention relates to methods of use of these therapeutically effective compositions.
The present invention encompasses compositions, containing a compound of the following formula:
M
II
W-P-OH (I)
I
H
wherein M is O or S, each W is independently selected from one of the following structures II- V
X Z
II I
R-C -X-(CH2\ -CH-CH2-Y (H)
Z K-X- (CH2\ -CH-CH2-Y (m)
X V z
II I I
R -C-L -(CH2)n -(CH)m -CH-CH2 -Y (IV)
O Z
II I
R-C -NH-(CH2)n -CH-CH2-Y (V)
wherein Y is O or S; R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably 24-30, more preferably 24-28 and most preferably about 24 carbon atoms, or ((CH2)pO)q(CH2)pT where q is an integer from 1 to about 900
SUBSTΓΓUTE SHEET (RULE 26) and where each p is independently an integer from 2 to about 10 and T is OH, or 0(CH2)bCH3 where b is an integer from 0 to about 10; each V is independently OH, SH, H, NH2, halogen, OP03H2, or OSOjH; n is an integer from 0 to about 10; m is an integer from 0 to about 10; Z is OH, SH, NH2, halogen,
Figure imgf000009_0001
H, 0(CH2)bCH3 where b=0 to about 2, or S03H; L is independently O, S, CH2 ; and X is independently O or S; or a salt thereof. In specific embodiments, R is an alkyl having between about 10 and about 24 carbon atoms, wherein between 0 and 11, inclusive, of the carbon-carbon bonds are unsaturated. More specifically, R is an alkyl having 18 carbon atoms, wherein 1 or 2 of the carbon-carbon bonds are unsaturated, and mixtures thereof. Even more specifically, the composition comprises
Bis(3- -oleyl-2-0-methyl-rαc-glyceryl) thiophosphate, or a salt thereof.
In another embodiment of the present invention, a composition contains a compound of the following formula:
O
II
Q -M - W (VI)
W
wherein each W is independently SH, OH, OCH2CH(NH2)C02H, OCHCH3CH(NH2)C02H, OPO3H2, OP02HOP03H2 or Q, wherein when one W is Q, the other W is OH, and wherein Q is one of the following structures:
X Z R - (CH2\ -X - C - CH - CH2 - Y (VTl)
X V Z
R - (CH2\ -L - C ~ (CH)m - CH - CH2 - Y (vm)
wherein Y is O or S; R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably 24-30, more preferably 24-28 and most preferably about 24 carbon atoms, or ((CH^O^CH^T where q is an integer from 1 to about 900 and where each p is independently an integer from 2 to about 10 and T is OH, or 0(CH2)pCH3 where p is an integer from 0 to about 10; each V is independently OH, SH, H, NH , halogen, OP03H2, or OS03H; n is an integer from 0 to about 10; m is an integer from 0 to about 10; Z is OH, SH, NH2, halogen, OP03H2, H, 0(CHj)pCH3 where p=0 to about 2, or SO^; L is independently O, S, or CH2; X is independently O or S; and M is P or S, where when M is S, one W is (=0) and the other W is SH, OH, OCH2CH(NH2)C02H, OCHCH3CH(NH2)C02H, OPO^, or OPO^OPO^; or a salt thereof.
In another embodiment, the composition comprises a compound selected from the group consisting of reverse ester-LPA, reverse thioester-LPA or a salt of either.
In a specific embodiment, the composition comprises: 3 Oleyl l-thiophosphoryl-2-0- methyl-rαc-glycerate, or a salt thereof. The present invention also provides methods of treating apoptosis, preserving or restoring function in a cell, tissue or organ comprising administering in vivo or in vitro a therapeutically effective amount of a pharmaceutically acceptable composition of the present invention. The present composition can also contain a potentiating component including, for example, a polyethylene glycol, a protein, or a lipid membrane structure. In specific embodiments of the present invention, the compositions of the present invention also comprise pharmaceutically acceptable excipients, which can be, without limitation, selected from the group encompassing topical pharmaceutically acceptable carrier, cosmetic carrier, sterile solutions, sterile isotonic solutions, ingestable liquids, pharmaceutically acceptable aerosols and solutions for organ/tissue/cell preservation and/or transplantation. In specific embodiments of the present invention, the compositions of the present invention also comprise pharmaceutically effective agents, such as, without limitation, drugs, antibiotics, wound healing agents and antioxidants.
Also encompassed by the present invention are methods of making compositions of the present invention, and the compositions obtained therefrom, comprising the steps of: forming a lipid dispersion comprising LPA; providing at least one of said components; and combining the products of steps a) and b), and more specifically, wherein the lipid dispersion is formed by the steps of: a) dissolving LPA and any other lipids in organic solvent; b) removing the solvent to form dried lipid; and c) dispersing the dried lipid into aqueous media by the steps of: i) forming an even lipid dispersion; and ii) forming an even dispersion of lipid membrane structures. Additionally encompassed are methods of treating apoptosis or preserving or restoring function, in a cell, tissue or organ, comprising administering a therapeutically effective amount of a pharmaceutically acceptable composition of the compounds of the present invention. Such composition can further comprise a potentiating component. The compounds of the present invention can be administered to a patient suffering from a condition related to apoptosis, ischemia, traumatic injury or reperfusion damage (wherein the reperfusion damage can be, without limitation, associated with coronary artery obstruction, stroke, cerebral infarction, spinal/head trauma and concomitant severe paralysis, frostbite, coronary angioplasty, blood vessel attachment, limb attachment, organ attachment, or kidney reperfusion), and gastrointestinal perturbation (wherein the gastrointestinal perturbation can be caused by, without limitation, a stimulus such as viruses, chemotherapeutic agents, radiation, infectious diseases such as human immunodeficiency virus, inflammatory bowel disease, and diarrhea-causing organisms). Treatment can comprise administering to the patient at least one intravenous bolus of an effective amount of the composition. In another embodiment, methods of the present invention decrease apoptosis-related problems associated with immunosuppressing viruses, chemotherapeutic agents, or radiation and immunosuppressive drugs.
Also encompassed by the present invention are methods of culturing cells comprising treating cells with an amount of at least one of the compositions of the present invention effective to prevent apoptosis and/or preserve the cells, including preserving an organ.
BRIEF DESCRIPTION OF THE DRAWINGS Fig. 1 is a bar graph depicting the effect of various concentrations of 18: 1-LPA analog on protection of C3H/10T1/2 cells from serum deprivation. LPA was prepared with or without a potentiating component (bovine serum albumin or phosphatidyl glycerol vesicles). The open section of the bars represents adherent cells and the solid section of the bars represents non-adherent cells. Where not specified as a particular analog, "LPA" refers to 18: 1-LPA in all figures. In Figs. 1-8 and 11, the white sections represent adherent cells and the dark sections represent non-adherent cells. Fig. 2 is a bar graph depicting the percentage of adherent cells (open sections) and non-adherent cells (solid sections) in the C3H/10T1/2 assay after 24 hours exposure to serum-free medium to which 18: 1-LPA was added as a 10% (by weight) mixture in various phospholipid membrane structures. For all treatments, 18: 1-LPA was delivered at 0.25, 0.75, 2.25 or 6.75 (g/mL. The following list provides the compositions represented by each column in Fig. 2: BME Five Phospholipid Mixture (0.25 (g/ml LPA) Five Phospholipid Mixture (0.75 (g/ml LPA) Five Phospholipid Mixture (2.25 (g/ml LPA) Five Phospholipid Mixture (6.75 (g/ml LPA) 10% LPA in PG (0.25 (g/ml LPA) 10% LPA in PG (0.75 (g/ml LPA) 10% LPA in PG (2.25 (g/ml LPA) 10% LPA in PG (6.75 (g/ml LPA) PG only 67.5 (g/mL 10% LPA in PG/5% PE-PEG (0.25 (g/ml LPA) 10% LPA in PG/5% PE-PEG (0.75 (g/ml LPA) 10% LPA in PG/5% PE-PEG (2.25 (g/ml LPA) 10% LPA in PG/5% PE-PEG (6.75 (g/ml LPA) PG/5% PE-PEG only 67.5 (g/ml PG/5% PE-PEG only 22.5 (g/ml 10% LPA in PG/ 10%PE-PEG (0.25 (g/ml LPA) 10% LPA in PG/ 10% PE-PEG (0.75 (g/ml LPA) 10% LPA in PG/ 10% PE-PEG (2.25 (g/ml LPA) 10% LPA in PG/ 10% PE-PEG (6.75 (g/ml LPA) PG/10% PE-PEG only 67.5 (g/ml PG/10% PE-PEG only 22.5 (g/ml 10% LPA in PC (0.25 (g/ml LPA) 10% LPA in PC (0.75 (g/ml LPA) 10% LPA in PC (2.25 (g/ml LPA) 10% LPA in PC (6.75 (g/ml LPA) PC only 67.5 (g/ml PC only 22.5 (gml 10% LPA in PC/TAP (0.25 (g/ml LPA) 10% LPA in PC/TAP (0.75 (g/ml LPA) 10% LPA in PC TAP (2.25 (g/ml LPA) 10% LPA in PC/TAP (6.75 (g/ml LPA) PC/TAP only 67.5 (g/ml PC/TAP only 22.5 (g/ml 35 10% LPA in PS (0.25 (g/ml LPA)
36 10% LPA in PS (0.75 (g/ml LPA)
37 10% LPA in PS (2.25 (g/ml LPA)
38 10% LPA in PS (6.75 (g/ml LPA) 39 PS only 67.5 (g/ml
40 BME only
Fig. 3 is a bar graph depicting protection of serum-deprived C3H/10T1/2 cells by Five Phospholipid Mixture (referred to as "Elirex" in the figure) and 18: 1-LPA formulations in phosphotidyl glycerol (PG) and phosphotidyl glycerol/phosphotidylcholine (PG/PC) membrane structures.
Fig. 4 is a bar graph depicting protection of serum-deprived C3H/10T1/2 cells by 18: 1-LPA and lysophosphotidylserine (LPS).
Fig.5 is a bar graph depicting protection of C3H/10T1/2 cells from serum-deprivation by soy-derived, 18:1 and 16:0 LPA.
Fig. 6 is a bar graph depicting C3H/10T1/2 cell protection from serum-deprivation by 18:1 LPA and 18:0 LPA alone or in PG membrane structures.
Fig. 7 is abar graph depicting protection of C3H/10T1/2 cells from serum-deprivation by lysophosphatidic acid incorporated at different concentrations into Phosphatidic acid (PA)/phosphatidylinositol (PI) (1: 1) membrane structures. Controls include concentrations of 0.3,
1, 3, and 10 (g/ml of the Five Phospholipid Mixture (referred to in the figure as "Elirex").
Fig. 8 is a bar graph depicting protection of serum-deprived C3H/10T1/2 cells by 18: 1-LPA in Five Phospholipid Mixture (labeled "Elirex") and PC or chemically modified PC membrane structures. Fig. 9 depicts an autoradiograph of a non-denaturing polyacrylamide gel of proteins separated following incubation with 3H-labeled 18: 1-LPA.
Figs. 10A, 10B, 10C and 10D are graphs showing the elution profiles of 3H-18: 1-LPA, with and without various proteins, from a Superdex S75 column.
Fig. 11 is a bar graph depicting the effect of polyethylene glycol (PEG) molecular weight on protection of C3H/1 OT' i cells from serum deprivation at 1 mM in the presence and absence of
1.5 μM 18:1-LPA. PEG of molecular weights 200, 1,000, 3,000, 6,000, 8,000, 20,000 and 35,000 were used. Fig. 12 is a bar graph depicting the effect of the actual infarct size compared to the area at risk in the pig heart model treated with control (solid bar), PEG 20L (striped bar) and a dose of Five Phospholipid Mixture and PEG (gray bar - labeled "PEG + Elirex").
Fig. 13 is a graph depicting the prevention of cardiomyocyte death induced by serum/glucose deprivation by PEG, Five Phospholipid Mixture (referred to in the figure as
"Elirex") and a mixture of PEG and Five PhosphoUpid Mixture. The squares represent no PEG, the circles represent 0.3% PEG and the triangles represent 2% PEG.
Fig. 14 is a graph depicting the treatment of rat neonatal cardiomyocytes with PEG plus
Five Phospholipid Mixture (closed circles) and PEG plus 18: 1-LPA (open circles) following ischemia to evaluate the effect of treatment on cardiomyocyte death induced by ischemia and reperfusion. The amount of LPA corresponds to its quantity in the indicated amount of Five
Phospholipid Mixture.
Fig. 15 is a bar graph depicting the actual infarct size compared to the area at risk in the dog heart model treated with placebo (solid bar), a mixture of superoxide dismutase (SOD) and catalase (striped bar) and a dose of the combination of Five Phospholipid Mixture (12.5_(g/kg) and PEG (25_mg/kg) (white bar).
Fig. 16 is a bar graph depicting the actual infarct size compared to the area at risk in the rat heart model treated with placebo (soUd bar) or a dose of the combination of Five PhosphoUpid Mixture and PEG (gray bar, Usted in key as "APM/PEG").
DETAILED DESCRIPTION OF THE INVENTION
It has now been found that lysophosphatidic acid is the major active component of phosphoUpid mixtures extracted from plants that have anti-apoptotic activity. This invention encompasses methods of use of therapeutically effective compositions containing lysophosphatidic acid, its analogs and derivatives (collectively "LPA"), which have been found to exhibit anti-apoptotic activity and to preserve or restore cell, tissue and organ function.
'Treating apoptosis" is herein defined as adrninistering to a cell, tissue, organ or organism exhibiting apoptosis, or at risk of apoptosis, a treatment to effect beneficial or desired clinical results, including, but not limited to preventing or diminishing apoptosis. I. COMPOUNDS OF THE PRESENT INVENTION.
Although phosphoUpid structures are weU defined in the Uterature, they can vary with respect to Upid chain length and saturation. "Lysophosphatidic acids" or "LPAs" as used herein shall encompass the following structures and descriptions and will also include related structures known in the art provided they are effective in producing therapeutic response. A. Structures of compounds of the present invention.
LPA has the following general structure:
O OH O
R - C - 0 - CH2 - CH - CH2 - 0 - P - OH (LX)
0~
An LPA is an acid in which only one of the hydroxyl groups of the glycerol is esterified to a fatty acid. LPA is a phosphatidic acid in which the 2 carbon is not esterified and the 3 carbon is bound to the O-PO^ group, or, in the case of the salt, one or more hydrogens are replaced, for example with Na+. The 1 carbon will contain an acyl ester of fatty acids.
The term "UB" is used in reference to the various structures herein to describe the number of unsaturated carbon atoms in R. For example, if R is 18 and UB is 1, R contains 18 carbon atoms, with 1 unsaturated bond. Some LPA analogs are also referred to herein as R:UB-LPA (i.e.
18: 1-LPA, wherein R is 18 carbon atoms with 1 unsaturated bond).
As used herein, LPA includes LPA having any one of a variety of fatty acids esterified at the C 1 position. Examples include LPA wherein the fatty acid ester is lauryl, myristyl, palmityl, stearyl, palmitoleyl, oleyl or linoleyl. (In structure I, above, the composition where R is 18 and UB is 1, shall herein be referred to as "18: 1-LPA"). For a representative example of suitable phospholipids see any chemical catalog of a phospholipid suppUer, for instance, the (1994) Avanti Polar Lipids Inc. catalog, particularly pages 14 and 21.
R can be an unsubstituted or substituted, saturated or unsaturated, straight or branched chain alkyl having from about 10 to about 24 carbon atoms. For all of the structures referenced herein, R can have between 0 and (n-2)/2 unsaturated bonds, wherein n is the number of carbon atoms in R. Substitutions include, but are not limited to, halogen, hydroxy, phenyl, amino or acylamino.
As used herein, "LPA" also encompasses LPA analogs. Given the examples provided herein, it can be determined readily if an LPA analog exerts sufficient anti-apoptotic activity to be suitable for the uses contemplated herein. A wide variety of LPA analogs are known in the art and many of these can be purchased from commercial sources such as Avanti Polar Lipids Inc.
(Alabaster, AL), or they can be synthesized by methods known in the art.
LPA analogs include, but are not limited to, the foUowing structures: or a cyclic phosphate derivative thereof having the structure: or pharmaceutically acceptable salts thereof, wherein each X is independently O or S; M is P or S, where when M is S, one W is (=0); each W is independently SH, OH, OCH2CH(NH2)C02H, OCHCH3CH(NH2)C02H,
Figure imgf000016_0001
where if one W
X Z O
II I II
R - C -X - (CH2χ - CH -CH2 - Y -M - W (X)
W
is OP03H2 or OP02HOP03H2, the remaining W is OH; Z is OH, SH, NH2, halogen, OPOjH H,
Figure imgf000016_0002
O^H^Q , where b=0 to about 2, or SOjH; R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably 24-30, more preferably 24-28 and most preferably about 24 carbon atoms, or ((CH2)ιnO)p(CH2)I-W where p is an integer from 1 to about 900 and where each m is independently an integer from 2 to about 10 and W is OH, or 0(CH2)qCH3 where q is an integer from 0 to about 10; Y is O or S; and n is an integer from 0 to about 10. Preferably, R is between about 10 and 24, UB is 0-11, and mixtures thereof. More preferably, R is between about 14 and 20, UB is 0-6, and mixtures thereof. Even more preferably, R is between about 16 and 18, UB is 0-3, and mixtures thereof. Most preferably, R is 18, UB is 1 or 2, and mixtures thereof.
Methods of preparation of substitutions at the phosphate group of LPA have been described, and are included herein. Tokumura et al. (1981) J._Pharm._Exp._Therap. 219:219-224.
Also included in LPA analogs are ethers and thioethers at the Cl position having the structure: o
R -X - (CH2X - C IH - CH2 - Y -M - W (xπ)
W
or a cyclic phosphate derivative thereof having the structure:
Figure imgf000017_0001
or pharmaceutically acceptable salts thereof, wherein X is O, S, or CH2; M is P or S, where when M is S, one W is (=0); each W is independently SH, OH, OCH2CH(NH2)C02H, OCHCH3CH(NH2)C02H, OPO^, or OP02HOPθ3H2, where if one W is OPO^ or
OP02HOP03H2, the remaining W is OH; Z is OH, CH2OH, SH, NH2, halogen, OPOsHj, H or SOjH; R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably
Figure imgf000017_0002
where p is an integer from 1 to about 900 and where each m is independently an integer from 2 to about 10 and W is OH, or 0(CH2)qCH3 where q is an integer from 0 to about 10; Y is O or S; and n is an integer from 0 to about 10. Simon et al. (1982) Biochem. Biophys. Res. Comm. 108:1743-1750.
Preferably, R is between about 10 and 24, UB is 0-11, and mixtures thereof. More preferably, R is between about 14 and 20, UB is 0-6, and mixtures thereof. Even more preferably,
R is between about 16 and 18, UB is 0-3, and mixtures thereof. Most preferably, R is 18, UB is 1 or 2, and mixtures thereof.
Also included are glycero LPA analogs having the structure:
X V Z O
R - C II -X - (CH2χ - (C IH)m - CIH - CH2 - Y -M II - W (XTV)
W or a cyclic phosphate derivative thereof having the structure:
Figure imgf000018_0001
or pharmaceutically acceptable salts thereof, wherein each V is independently OH, SH, H, NH2, halogen, OPO^, or OSOsH; each X is independently O or S; M is P or S, where when M is S, one W is (=0); each W is independently SH, OH,
Figure imgf000018_0002
OCHCH3CH(NH2)C02H, OPOJHJ, or OPO^OPO^, where if one W is OPO^ or
OP02HOPθ3H2, the remaining W is OH; Z is OH, SH, NH2, halogen, OPO^, H or SO^; R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably 24-30, more preferably 24-28 and most preferably about 24 carbon atoms, or ((CH^pOJ^CH^pV where q is an integer from 1 to about 900 and where each p is independently an integer from 2 to about 10 and V is OH, or 0( Α^ Α3 where b is an integer from 0 to about 10; Y is O or S; n is an integer from 0 to about 10; and m is an integer from 0 to about 10. Preferably, R is between aboutlO and 24, UB is 0-11, and mixtures thereof. More preferably, R is between about 14 and 20, UB is 0-6, and mixtures thereof. Even more preferably, R is between about 16 and 18, UB is 0-3, and mixtures thereof. Most preferably, R is 18, UB is 1 or 2, and mixtures thereof.
Also included are LPA analogs containing an amide bond and having the structure:
O Z O
II I II
R - C -NH - (CH2)n - CH - CH2 - Y -M - W (XVI)
W or a cycUc phosphate derivative thereof having the structure:
Figure imgf000019_0001
or the reverse amide [structures XVI and XVII, having R-NH-C(=O)-(CH2)n- in place of R-C(=0)-NH-(CH2)n-]( NH-CH2)n-] or pharmaceutically acceptable salts thereof, wherein Z is OH, SH, NH2, halogen,
Figure imgf000019_0002
M is P or S, where when M is S, one W is (=0); each W is independently SH, OH, OCH2CH(NH2)C02H, OCHO CHOSIH^CO^, OP03H2, or
OP02HOPO3H2, where if one W is
Figure imgf000019_0003
the remaining W is OH; R is an amino acid side chain or a branched amino acid side chain, or an alkylated amino acid side chain, unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably 24-30, more preferably 24-28 and most preferably about 24 carbon atoms, or ((CH2)mO)p(CH2)I-W where p is an integer from 1 to about 900 and where each m is independently an integer from 2 to about 10 and W is OH, or 0(CH2)qCH3 where q is an integer from 0 to about 10; Y is O or S; and n is an integer from 0 to about 10. Preferably, R is between about 10 and 24, UB is 0-11, and mixtures thereof. More preferably, R is between about 14 and 20, UB is 0-6, and mixtures thereof. Even more preferably, R is between about 16 and 18, UB is 0-3, and mixtures thereof. Most preferably,
R is 18, UB is 1 or 2, and mixtures thereof.
Also included are "bis" LPA compounds, having the foUowing structure (I),
M
II
W- P -OH (I)
I w
wherein M is O or S, each W is independently selected from one of the following structures (XI, XH, Xffl, XTV): X Z
R-C-X-(CH2χ-CH-CH2 -Y (II)
Z
I
R -X-(CH2χ -CH-CH2 -Y (Tfl)
X V Z
II I I
R -C -L -(CH2X -(CH)m -CH-CH2 -Y (TV)
O Z
R-C -NH-(CH2X -CH-CH2-Y (V)
wherein Y is O or S; R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl having from preferably 5-7, more preferably 8-10 and most preferably, about 10 to preferably 24-30, more preferably 24-28 and most preferably about 23 carbon atoms, or ((CH2)pO)q(CH2)pT where q is an integer from 1 to about 900 and where each p is independently an integer from 2 to about 10 and T is OH, or
O(CH2)bCH3 where b is an integer from 0 to about 10; each V is independently OH, SH, H, NH2, halogen, OP03H2, or OSO3H; n is an integer from 0 to about 10; m is an integer from 0 to about 10; Z is OH, SH, NH2, halogen, OP03H2, H, OfCH^C^ where b=0 to about 2, or S03H; L is independently O, S, CH2 and X is independently O or S; or a salt thereof.
Also encompassed by the term LPA are the compounds herein referred to as "reverse ester-LPA" or "reverse thioester-LPA", wherein the R-C(=0)-0-CH2- group is replaced by
R-CH2-0-C(=0)- in Structure X, and wherein the R-C(=X)-X-(CH2)n- group is replaced by
R-(CH2)n-X-C(=X)- in Structure X, XI, XTV, XV, II, and IV. In all analogs containing W, it is preferred that W is not ethanolamine, glycerol, or choline.
In the above structures, where one W is SH, some molecules will exist as resonance structures, alternating between the (=O) and (=S) structures. Naturally occurring derivatives are also encompassed in the term "LPA." Such derivatives include, but are not limited to, PHYPLA or cLPA. Murakami-Murofushi et al. (1992) J. Biol. Chem.267:21512-21517. CycUc derivatives can also be synthesized by methods known in the art.
Pharmaceutically acceptable salts of the phosphoUpids encompassed by the present invention, include, but are not limited to, the free acid form, alkali metal salts, such as sodium and potassium; alkaline earth metal salts, such as calcium and magnesium; non-toxic heavy metal salts; ammonium salts; trialkylammonium salts, such as trimethyl-ammonium and triethylammonium; and alkoxyammonium salts, such as triethanolammonium, tri(2hydroxyethyl)ammonium, and fromethamine (tris(hydroxymemyl)aminomethane). Particularly preferred are sodium and ammomum salts.
B. Obtaining compounds of the present invention.
The phospholipids can be obtained from any source including, but not limited to, commercial, isolated from a variety of different plants (including plant organs) and animals or created synthetically. Preferably the plants are in the soybean family, but the phospholipids can be isolated from other plants including, but not limited to, those in the leguminosae (beans and peas etc.). The phospholipids can also be isolated from partially purified plant extracts including, but not limited to, soy molasses, lecithin (fluid, deoiled or other forms), partially purified protein concentrates, partially purified protein hydrolysates, defatted soy flakes, refined soy oils, soy grits, soy flours and other soy fractions from which lipids can be extracted. It is within the skill of one in the art, utilizing the methods described herein, to determine whether the phospholipids of the present invention can be isolated from a particular species of plant, plant extract or organ within a plant. In addition, U.S. Patent No. 3,365,440 describes extraction oflipids from soybeans. U.S. Patent Nos. 5,567,425; 5,602,885; 5,624,675; 5,635,186; 5,635,187 have further general descriptions of a variety of techniques useful for the present invention. The phospholipids can be obtained from plant sources by any method known in the art provided it results in purification of at least one of the phospholipids of the invention. A variety of methods are known in the art for purifying and analyzing phosphoUpids from plant sources. For review, see Bligh and Dyer (1959) Can. J. Biochem. Physiol. 37:911-917; Patton et al. (1982) Lipid Res. 23:190-196; Jungalwala (1985) Recent Developments in Techniques for PhosphoUpid Analysis, in Phospholipids in Nervous Tissues (ed. Eichberg) John Wiley and Sons, pp. 1-44;
Hamilton et al. (1992) in the series, A Practical Approach (Rickwood et al. eds.) IRL Press at Oxford University Press; and Kates (1986) Techniques of Lipidology: Isolation, Analysis and Identification in Laboratory Techniques in Biochemistry and Molecular Biology (Burdon et al. eds.) Elsevier. PhosphoUpids can also be derived from animal sources. Preferably, the animal is a mammal. Even more preferably, the phosphoUpids are derived from Uver cells. Such phosphoUpids are commercially available or can be purified from animal tissue by methods known in the art, for instance from animal and egg lecithin or from the compositions described in WO 95/15173. PhosphoUpids in general, and LPAs in particular, can also be derived from blood.
The phosphoUpids of the invention can also be synthesized by methods known in the art. Suitable semi-synthetic phospholipids and their synthesis are described in Kates, Techniques of Lipidology ( 1972). A synthesis of lysophosphatidic acid is described in W. Stoffel and G.D. Wolf, Chemische Synthese von l-0-[3H]Palmitoyl-L-glycerin-3-phosphate (L-3-Lysophosphatidsaure), ChemJBer., 347 (1966) 94-101. The synthesis of various cyclic phosphate LPAs is described in
A.J. Slotboom, et al., Synthesis of Lysophosphoglycerides, Chem. Phvs. Lipids. 1 (1967) 317-336; PCT Pubhcation No. WO 92/21323; and US Patent 5,565,439. The synthesis of a phosphonate analog of l-O-hexadecyl-2-O-methyl-glycero-phosphate is described in Z. Li, etal., Phosphonate isosteres of phosphoUpids, Tetrahedron Lett.. 34 (1993) 3539-3542. Procedures for synthesis of functionaUzed glycerol ether derivatives which can be used in the synthesis of compounds suitable for use in the present invention are described in K. Agarwal, et al. Synthesis of carbamyl and ether analogs of phosphatidylcholines, Chem. Phvs. Lipids. 39 ( 1984) 169- 177, and H. Eibl and P. WooUey, A general synthetic method for enantiomericaUy pure ester and ether lysophosphoUpids, Chem. Phvs. Lipids. 47 (1988) 63-68. The preparation of l-0-benzyl-2-deoxy-2-bromo glycerol, a starting material for the synthesis of 2-bromo LPA Compound 37, is described in W.L.F. Armarego, B.A. Milloy and W. Pendergast, A highly stereospecific synthesis of (R)- and (S)-[2-2Hl]glycine, J.C.S. Perkin I, (1976) 2229-2237.
The synthesis of 2-deoxy-2-bromo-phosphatidylchoUne is described in C.J. Lacey and L.M. Loew, PhosphoUpid synthesis based on new sequential phosphate and carboxylate ester bond formation steps, J. Ore. Chem.. 48 (1983) 5214-5221.
The synthesis of bisphosphatidic acid and its conversion to bis-lysophosphatidic acid using phosphoUpase A2 from pig pancreas is described in Q. Quan Dang, et al., Synthesis and identification of bis(diacylglycero)phosphoric acid and bis(monoacylglycero)phosphoric acid, Lipids. 17 (1982) 798-802, and Q. QuanDangandL. Douste-Blazy, Synthesis andstereochemical study of some biologicaUy relevant phosphoglycerides: dicarboxylic phosphatidyl choUnes and bis(diacylglycero)phosphoric acids, Phosphorus and Sulfur. 18 (1983) 377-380.
A method for the preparation of lysophosphatidic acid or lysophosphatidates by reacting glycidyl esters with anhydrous phosphoric acid is described in US Patent 3,423,440. A synthesis of lysothiophosphatidic acid is described in N.V. Heeb and K.P. Nambiar, Synthesis of (R)-lysothiophosphatidic acid and (R)-thiophosphatidic acid, Tetrahedron Lett.. 34 (1993) 6193-6196
The preparation of LPA amide analogs and 2-deoxy LPA plus various derivatives is described in D.W. Hopper, et al., Facile synthesis of lysophospholipids containing unsaturated fatty acid chains, Tetrahedron Lett.. 37 (1996) 7871-7874; and K.R. Lynch, et al.,
Structure activity relationships in lysophosphatidic acid: the 2-hydroxyl moiety, Mol. Pharmacol..
52 (1997) 75-81.
The following papers describe synthetic routes which can be used for the synthesis of additional LPA analogs: M. Fuji, et al., A stereoselective and highly practical synthesis of cytosolic phosphoUpase A2 substrate, 2-S aracbidonoyl-l-O-hexadecyl-sn-thioglycero-3-0- phosphocholine, J. Ore. Chem.. 62 (1997) 6804-6809; (Strategy for the preparation of 2-thioglycero phosphocholines and guidance for the synthesis of 2-deoxy-2-thiol LPA); and A. Markowska, et al., Etheranaloge der Thio- und DithiophosphoUpide mit C-S-P-Bindung, Liebies Ann. Chem.. (1993) 1327-1329 (synthesis of l-0-hexadecyl-2-O-methyl-3-thioglycero-3- phosphocholine and l-S-hexaάecyl-2-0-methyl-l,3-dithioglycero-3-phosphocholine, guidance for the preparation of LPA analogs containing sulfur linked phosphates).
Various degrees of purity of the phospholipids can be used. Purity can be assayed by any method known in the art such as two dimensional TLC or HPLC and assayed for total Upids, phospholipids or phosphate. Various suitable methods are outlined in Kates (1972). Preferably, the phosphoUpids must be of sufficient purity such that, when mixed at a total concentration of about 10 mg/rnL, the mixture can be sonicated as described below to provide a relatively translucent solution. Preferably, the phospholipids are at least 90% pure, more preferably, they are at least 95% pure and, most preferably, they are at least 99% pure. The references cited herein are incorporated by reference in their entirety.
C. Potentiating Components.
One factor that can influence the therapeutic activity of the compositions of the present invention is the presence of a potentiating component. Under certain conditions LPA alone has displayed mitogenic activity and a short half life in vivo and under certain storage conditions. The present invention provides novel compositions in which one or more potentiating components are combined with LPA that have been found to potentiate the therapeutic activity of LPA. A "potentiating component" is defined as a molecule which potentiates the therapeutic activity of LPA. Potentiating components include, but are not limited to, proteins, other phospholipids, polyethylene glycols (PEG), Upid membrane structure forming compounds, polypeptides, modified polypeptides and polymers.
In some cases, the presence of calcium has been found to inhibit the anti-apoptotic properties of LPA. However, certain potentiating components have proven to counteract the inhibition by calcium. Therefore, in one embodiment, LPA is presented in combination with a potentiating component such as BSA and/or PEG. Alternative potentiating components appropriate to protect the activity of LPA can be identified by performing a screen as described in Example 2, in the presence and absence of the potentiating component to be tested.
An appropriate potentiating component for use in the present invention can easily be selected by combining the composition to be evaluated with a therapeutic amount of LPA in a therapeutically acceptable solution and evaluating the combination for its mitogenic activity, storage stability, in vivo half life, and for anti-apoptotic activity by any method known in the art, including those described herein. If the mixture has acceptable stability, is not therapeutically unacceptable due to mitogenic activity, and displays anti-apoptotic activity, or preserves or restores cell, tissue or organ function, the potentiating component is appropriate for use in the present invention.
D. Other PhosphoUpids.
In one embodiment of the present invention, the phosphoUpids PA; PI; lysophosphatidic acid; LPI; and LPC are present in the composition in a range of ratios from 0-20:5-20:2-16:0-4:0-8, respectively. Preferably, these phosphoUpids are in a ratio of approximately 2-15:8-15:6-10:2-4:2-8, respectively. Most preferably, these phospholipids are in a ratio of approximately 10:10:8:2:4, respectively.
2. Polyethylene glycol.
In one embodiment of the present invention, LPA is combined with PEG prior to therapeutic use. PEGs constitute a diverse group of molecules. Only those that potentiate the therapeutic efficacy of LPA are encompassed herein. It is within the skill of one in the art to determine whether a particular PEG is suitable for use in the claimed compositions. Such a determination can be made, for instance, by the methods described herein. Polyethylene glycol ("PEG"), ((-Hydro-(-hydroxypoly (oxy-l,2-ethanediyl)), is known by numerous designations including macrogel; PEG; Carbowax; Jeffox; Nycoline; Pluracol E; Poly-G; Polyglycol E; and
Solbase. PEG refers to the Uquid and solid polymers of the general formula H(OCH2-CH2)„OH, where n is greater than or equal to 4. In general, each PEG is foUowed by a number which corresponds to its average MW. PEG syntheses are described for instance in Hibbert (1939) J. Am. Chem. Soc. 61:1905-1910. For review, see also, PoweU, III in Handbook of Water-Soluble Gums &Resins, R.L. Davidson ed. (McGraw-HiU, New York, 1980)pp. 18/1-18/31. PEGs have found use as water-soluble lubricants for rubber molds, textile fibers, and metal-forming operations, in food and food packaging, in hair preparations and in cosmetics in general and as ointment and suppository bases in pharmaceutical compositions. Typically, PEGs are clear, viscous Uquids or white soUds that dissolve in water to form transparent solutions. They are soluble in many organic solvents and readily soluble in aromatic hydrocarbons. They are only slightly soluble in aliphatic hydrocarbons. Typically, they do not hydrolyze on storage. PEGs have low toxicity. The molecular weights of PEG compositions Usted herein are given in number averages rather than weight averages. PEG20M consists of two or more molecules of PEG having approximate molecular weights of 6000-10,000 joined by a bisphenol epoxide linker (CAS # 37225-26-6; CAS name Oxirane, 2, 2' [(l-methyl-ethylidene)bis(4,l-phenyleneoxy methylene)]bis-, polymer with (-hydro-(-hydroxypoly(oxy-l,2-ethanediyl). PEG20L is a substantially linear PEG having an average molecular weight of about 20,000 Daltons (available from several commercial sources including, but not limited to, ClariantHoechst Celanese, Fluka and Nippon Oils and Fats). The molecular weights of PEG compositions listed herein are given in number averages rather than weight averages. Various other molecular weights of linear PEG are also available from several commercial sources.
More recently, PEG has been used in a number of pharmacologic applications. The conjugation of PEG to foreign proteins, such as cytokines and antibodies, reduces the immune response triggered when the proteins are administered into test mammals. U.S. Patent Nos. 5,447,722; 4,902,502; 5,089,261; 5,595,732; 5,559,213; and 4,732,863. Conjugation to PEG also increases the solubility and biological half-Ufe of cytokines. WO 8700056 and U.S. Patent No. 5,089,261. Conjugates of PEG and glucocerebroside have been formulated for treating Gaucher's disease. WO 9413311. PEG has also been conjugated to such enzymes as adenosine deaminase, amidase bovine and asparaginase, for therapeutic use. See Delgado et al. (1992) Crit. Rev. Ther. Drug. Carrier Syst. 9:249-304; and Burnham (1994) Am. J. Hosp. Pharm. 51:210-218, for review.
The invention further encompasses compositions comprising therapeutically effective amounts of LPA and PEG. In compositions comprising LPA and PEG, the PEG to LPA weight ratio is such that the LPA therapeutic activity is potentiated by the PEG. Typically, the PEG to
LPA weight ratio of the composition is preferably 1-100,000: 1 and most preferably 10-10,000: 1.
PEG can be in the molecular weight range from about 6,000 to about 500,000.
Preferably, the PEG has an average molecular weight of about 8,000 to about 40,000. More preferably, the PEG has an average molecular weight of from about 20,000 to about 40,000. Most preferably, the PEG has an average molecular weight of 20,000. Even more preferably, the PEG is 20L PEG. By "PEG20L" is meant a substantiaUy linear PEG having an average molecular weight of about 1,000 to 100,000 Daltons, preferably about 8,000 to 35,000 Daltons, and most preferably about 20,000 Daltons. By "linear PEG" or "linear polymer" is meant that each PEG molecule comprises a single polymeric subunit without molecular linkages such as those found in PEG20M. PEG of 35,000 molecular weight is also preferred. PEG of higher molecular weight may have clearance problems when administered in vivo. Thus, PEG of molecular weight greater than 35,000 is preferably used in compositions for topical deUvery. With reference to PEG20L, "linear PEG" or "linear polymer" means that each PEG molecule comprises a single subunit without molecular linkages. By "having an average molecular weight of about 20,000 Daltons" is meant that individual linear polymers can vary in length, but the average molecular weight is about 20,000 Daltons. Those skiUed in the art will appreciate that synthetic polymers such as PEG cannot be prepared practically to have exact molecular weights, and that the term "molecular weight" as used herein refers to the average molecular weight of a number of molecules in any given sample, as commonly used in the art. For example, a sample of PEG 2,000 might contain a statistical mixture of polymer molecules ranging in weight from, for example, 1,500 to 2,500 Daltons with one molecule differing slightly from the next over a range. Specification of a range of molecular weight indicates that the average molecular weight can be any value between the limits specified, and can include molecules outside those limits. The molecular weight distribution of a PEG can be determined by gel permeation chromatography (GPC), a technique known in the art, using, for example, a combination of columns to achieve resolution from 1,000,000 to 200 molecular weight. PEG standards from 100,000 to 1,400 molecular weight can be used for calibration. PEG20L is suppUed as a white flake, and is readily soluble in water. The oxidation rate of PEG is dependent on storage conditions including: (1) temperature; (2) exposure to light; and (3) the availabiUty of oxygen.
Selection of the appropriate PEG depends on the intended use and can be readily determined by those skilled in the art. By "anoxically" is meant the reduction of ambient oxygen, a condition which can be maintained by purging with argon or nitrogen gas, and then packaging in a gas-impervious container. The absence of oxygen need not be total. Preferably, it is below about 10% of the total gas present in the sample. More preferably, it is below about 1% of total gas present in the sample. Preferably, PEG is present in an effective concentration and is essentially free of impurities. By "impurities" is meant the products produced when PEG is oxidized. In addition, smaU molecular fragments are formed such as formate, methyl formate, formaldehyde, acetaldehyde, etc., all of which are defined here as impurities. These impurities can be removed by any method known in the art, including, but not limited to, dialysis, which removes not only small molecular impurities such as formaldehyde, but also removes hydroperoxides, as evidenced by spectrophotometry. In order to determine the effectiveness of dialysis, conductivity is measured. When dialysis is effective, conductivity drops markedly to 10-20 microsiemens.
Removal of impurities by dialysis can be performed through a CeUulose Acetate HoUow Fiber Dialyzer (Baxter Model CA 110) but the same procedure can also be performed by
Ultrafiltration using a Regenerated CeUulose Ultrafiltration Membrane or a Polyethersulphone Ultrafiltration Membrane or using other dialysis methods known in the art. The membrane should have a molecular weight cut off of approximately 20,000 Daltons to allow removal of impurities, metals and other contaminants. By "therapeutically effective amount" is meant an amount sufficient to effect beneficial or desired clinical results. A therapeutically effective amount can be administered in one or more administrations.
Preferably, PEG is present in a therapeutically effective amount. In the case of OPS, an effective amount is the amount required to improve the ability of an OPS to preserve organs. In the case of compositions comprising LPA and PEG, where the compositions include other phospholipids, the preferred composition is where phosphoUpids are combined in a ratio of about 10:10:8:2:4 by weight. A ratio of "about" means that the ratios of the phospholipids can range approximately up to 15% but preferably not more than 5%. More preferably, the ratios are within ± 0.5%. Just as PEG has been used as a potentiating component for therapeutic agents, so the capacity of other macromolecules and macromolecular structures to serve as potentiating components has been explored. Included among these are various lipid membrane structures and proteins.
3. Lipid Membrane Structures. Lipid membrane structures (LMSs), including Uposomes, micelles, multilamellar vesicles and cellular membrane isolates, have been used as vehicles for delivering therapeutic agents. U.S. Patents 5,045,530; 5,141,751; 5,100,662; 5,292,499; 5,213,804; 5,449,513; 5,190,822; 5,540,925; and 5,395,619. LMSs are lamellar lipid particles wherein polar head groups of a polar Upid are arranged to face an aqueous phase of an interface to form membrane structures. As used herein, a "Uposome" or "Upid vesicle" is a small vesicle bounded by at least one and possibly more than one bilayer Upid membrane. Liposomes are completely closed Upid bilayer membranes which contain entrapped aqueous volume. Liposomes are vesicles which can be unilameUar (single membrane) or multilamellar (onion-like structures characterized by multiple membrane bUayers, each separated from the next by an aqueous layer). The bilayer is composed of two Upid monolayers having a hydrophobic "tail" region and a hydrophilic "head" region. In the membrane bilayer, the hydrophobic (nonpolar) "tails" of the Upid monolayers orient toward the center of the bilayer, whereas the hydrophilic (polar) "heads" orient toward the aqueous phase. The basic structure of liposomes can be made by a variety of techniques known in the art. These Upid vesicles are made artificially from phospholipids, glycolipids, lipids, steroids such as cholesterol, bolaamphiles related molecules, or a combination thereof by any technique known in the art, including, but not limited to sonication, extrusion, or removal of detergent from lipiddetergent complexes. A Uposome can also optionally comprise additional components associated with the outer surface, such as a tissue targeting component. It is understood that a "Upid membrane" or "lipid bilayer" need not consist exclusively of lipids, but can additionally contain any percentage of other components, including, but not limited to, cholesterol and other steroids, proteins of any length, and other amphipathic molecules, providing the general structure of the membrane is a sheet of two hydrophilic surfaces sandwiching a hydrophobic core. For a general discussion of membrane structure, see The Encyclopedia of Molecular Biology by J. Kendrew (1994). Suitable lipids include, but are not limited to, those discussed in Lasic (1993)
"Liposomes: from Physics to Applications" Elsevier, Amsterdam.The lipid bilayer making up the Uposome can comprise phospholipids, glycolipids, steroids, and their equivalents . The lipid bilayer can also be made of amphipathic proteins and lipidsoluble chemicals. Preferably, a composition is chosen that allows the membrane to be formed with reproducible quaUties, such as diameter, and is stable in the presence of elements expected to occur where the Uposome is to be used, such as physiological buffers and circulating molecules. Preferably, the Uposome is resilient to effects of manipulation by storage, freezing, and mixing with pharmaceutical excipients.
Lipids suitable for incorporation into lipid membrane structures include, but are not Umited to, natural, semi-synthetic or synthetic mono- or di-glycerophospholipids including phosphatidylcholines, phosphatidylethanolamines, phosphatidylglycerols, phosphatidylinositols, phosphatidic acids, phosphatidylserines, glycero- and cardioUpins. Sphingolipids such as sphingomyelin and cerebrosides can also be incorporated. While natural phosphoUpids occur with the phospho moiety at the sn-3 position and hydrophobic chains at the sn-l and sn-2 positions, synthetic lipids can have alternative stereochemistry with, e.g., the phospho group at the sn-l or sn-2 positions. Furthermore, the hydrophobic chains can be attached to the glycerol backbone by acyl, ether, alkyl or other linkages. Derivatives of these Upids are also suitable for incorporation into Uposomes. Derivatives suitable for use include, but are not limited to, haloalkyl derivatives, including those in which all or some of the hydrogen atoms of the alkyl chains are substituted with, for example, fluorine. In addition, cholesterol and other amphipathic steroids, bolaamphiphiles
(Upids with polar moieties at either end of the molecule which form monolayer membranes) and polyglycerolmonoalkylthers can also be incorporated. Liposomes can be composed of a single Upid or mixtures of two or more different Upids.
In one preferred embodiment, the Upid bilayer of the Uposome is formed primarily from phospholipids. More preferably, the phospholipid composition is a complex mixture, comprising a combination of phosphatidylcholine (PC), phosphatidic acid (PA), phosphatidylethanolamine (PE), phosphatidylglycerol (PG) and sphingomyeUn (SM). The LMS can further comprise additional Upids such as phosphatidylinositol (PI), phosphatidylserine (PS), or cardioUpin (diphosphatidylglycerol). If desired, SM can be replaced with a greater proportion of PC, PE, or a combination thereof. PS can optionally be replaced with phosphatidylglycerol (PG). Preferably, at least PC and PE are included; more preferably, at least three of the group PC, PS, PE, and SM are included. The composition is chosen so as to confer upon the LMS stability during both storage and administration.Each phosphoUpid described above can vary in its structure depending on the fatty acid moieties that are esterified to the glycerol moiety of the phospholipid. Generally, most commercially available forms of a particular phosphoUpid can be used. However, phospholipids containing particular fatty acid moieties may be preferred for certain applications.
Preferably, the LMS also includes cholesterol or a related steroid to improve the rigidity of the membrane. Any amount of cholesterol can be used. A preferred ratio of total cholesterol to Upid is between about 0.5 and about 1.2 moles of cholesterol per mole of Upid. More preferred is a molar ratio of about 0.8 to about 1.2: 1; even more preferred is a molar ratio of about 0.9 to about 1.1:1; still more preferred is a molar ratio of about 1.0:1.0. Other molecules that can be used to increase the rigidity of the membrane include crossUnked phospholipids.
Other preferred liposomes for use in vivo are those with an enhanced ability to evade the reticuloendotheUal system, thereby giving them a longer period in which to reach the target cell. Effective Upid compositions in this regard are those with a large proportion of SM and cholesterol, or SM and PI. Liposomes with prolonged circulation time also include those that comprise the monosialogangUoside GMl, glucuronide, or PEG. For example, cholesterol can be added at the ratios indicated above to a lipid mixture consisting of any combination of SM, PI, glucuronide, PEG, and other suitable components. Methods of making LMSs are weU known in the art. A number of pubUcations describe a variety of methods for preparing Uposomes of different structure and Upid composition. Gregoriadis (1988) Liposomes as Drug Carriers Wiley, New York; Gregoriadis ( 1993) Liposome Technology 2nd Ed. Vol. I: Liposome Preparation and Related Techniques CRC Press, Boca Raton; Watwe et al. (1995) Curr. Sci. 68:715-724; Vemuri et al. (1995) Pharm. Acta Helvetiae
70:95-111; Elorza et al. (1993) J. Microencapsulation 10:237-248; and U.S. Patents 4,737,323 and 5,008,050.
Liposomes can also be provided with molecules at the surface that target them to the ceU of interest. Such smaU molecules can be attached by incorporating into the Upid bilayer a functionaUzed phosphoUpid (US . Patents 5,052,421 and 5,540,935) or a functionalized cholesterol
(U.S. Patent 4,544,545). Polypeptides can be attached covalently to the Upid bilayer (EP Patent 0036277), to a glycophosphohpid (U.S. Patent 5,374,548), to a carboxylated phosphoUpid (U.S. Patent 4,762,915), to a derivatized sterol (U.S. Patent 5,000,960), or to a peptide anchor (U.S. Patent 5,109,113). Alternatively, if the polypeptide comprises a hydrophobic domain, it can be incorporated directly into the lipid bilayer, either by forming the liposome in its presence, or by performing the liposome and inserting the polypeptide subsequently using a suitable detergent. Tranum-Jensenetal. (1994) J. Membrane Biol. 140: 215-23; EP Patent 0047480; and U.S. Patent 5,252,348.
Liposomes have been prepared with mammalian-derived peptides such as cytokines (U.S. Patent 5,258,499), transferrin (Stavridis et al.), antibody (Laukkanen et al. (1994) Biochem.
33:11664- 11670), asialofetuin and other galactose-terminated side chains (Ishihara ( 1990) Pharm. Res. 7:542-546; and Ghosh et al. (1991) In: Liver Diseases, Targeted Diagnosis and Therapy Using Specific Receptors and Ligands Wu et al., Ed. Marcel Dekker, New York), a fusogenic protein from rat brain microsomal membranes (Rakowska et al. (1994) J. Membrane Biol. 142:35-42), and surfactant protein A (Walther et al. (1993) Am. J. Physiol. 265:L330-339).
Liposomes have been prepared with artificial peptides, such as a 14-residue amphipathic sequence which is a fusogenic GALA-type peptide. Puyal et al. (1994) Biochim. Biophys. Acta 1195:259-266. Liposomes have also been prepared with viral components: for example, the F and G glycoprotein of respiratory syncytial virus (RSV) (U.S. Patent 5,252,348), reovirus M cell attachment protein (Rubas etal. (1990)^ Λ/croβ«cσpj«/ ή'o j 7:385-395), influenza virus surface protein (WO 92/19267; EP 0047480; and Nussbaum etal. (19897) J. F/ro/.61:2245-2252), viral membrane fusion proteins, particularly hemagglutinin (WO 95/32706), and the influenza hernagglutininD loop and K loop peptides (Friede etal. (1994) Vaccine 12:791-797).Rapid uptake of Uposomes in vivo by cells of the reticuloendothelial system has restricted their therapeutic utiUty. This problem has been overcome by incorporation of Upids derivatized with various synthetic polymers, for example, polyethylene glycol (PEG), polylactic acid, polyglycoUc acid, or combinations thereof. Woodle and Lasic (1992) Biochim. Biophys.Acta 1113:171-199; ZaUpsky et al. (1994) FEBS Letters 353:71-74; and U.S. Patent 5,395,619. The present invention also encompasses compositions comprising micelles and methods of using such compositions. MiceUes in aqueous solution, both non-ionic, cationic and anionic, have been described in the literature in numerous pubUcations. Mittal (1977) Micellization, Solubilization andMicroemulsions Plenum Press, New York; Mittal (1979) Solution Chemistry of Surfactants Plenum Press, New York; Menger ( 1977) In Biorganic Chemistry III. Macro- and Multicomponent Systems Van Tanelen, Ed. Academic Press, New York; and Menger ( 1979) Ace.
Chem. Res. 12:111-117. "Micelles" is a term appUed to aggregates which form from tenside molecules in aqueous solutions above a specific temperature or a characteristic concentration. This concentration is called the critical micellization concentration, or cmc. When the cmc is exceeded, the monomer concentration remains practically constant and the excess tenside molecules form micelles. They can occur in various shapes (spheres, rods, discs) depending on the chemical constitution of the tenside and on the temperature, concentration or ionic strength of the solution. The micelles have characteristic aggregation numbers with usually only a small distribution spread. Reaching the cmc is manifest by abrupt changes in the surface tension, the osmotic pressure, the electrical conductivity and the viscosity. Micelles are thermodynamically stable association colloids of surfactant substances in which the hydrophobic radicals of the monomers lie in the interior of the aggregates and are held together by hydrophobic interaction; the hydrophilic groups face the water and by solvation provide the solubility of the colloid.
A process for preparing liposomes containing LPA is as foUows. An aqueous dispersion of Uposomes is prepared from membrane components, such as phosphoUpids (e.g. phosphatidylcholine, phosphatidylglycerol, sphingomyelin and phosphatidylethanolamine) and glycoUpids according to known methods as disclosed. Ann. Rev. Biophys. Bioeng. 9:467 (1980). The Uposomes can further contain sterols (e.g., cholesterol and cholestanol), dialkylphosphates, diacylphosphatidic acids, stearylamine, (-tocopherol, etc., in the liposomal membrane.
An aqueous solution of LPA is added to the liposomal dispersion at a concentration sufficient to produce a therapeutically effective final product. The mixture is allowed to stand for a given period of time, preferably under warming at a temperature more than the phase transition temperature of the membrane or above 40°C, followed by cooling to produce liposomes containing LPA in the liposomal membrane. Alternatively, the desired liposomes can also be prepared by previously mixing the above-described membrane components and LPA and treating the mixture in accordance with known methods for preparing liposomes.
The Upid vesicles can be prepared by any suitable technique known in the art. Methods include, but are not Umited to, microencapsulation, microfluidization, LLC method, ethanol injection, freon injection, the "bubble" method, detergent dialysis, hydration, sonication, and reverse-phase evaporation (reviewed in Watwe et al.). For example, ultrasonication and dialysis methods generally produce small unilameUar vesicles, while extrusion and reverse-phase evaporation generally produce larger sized vesicles. Techniques can be combined in order to provide vesicles with the most desirable attributes. One particularly useful method is microfluidization.
The invention encompasses LMSs containing tissue or cellular targeting components. Such targeting components are components of a LMS that enhance its accumulation at certain tissue or cellular sites in preference to other tissue or ceUular sites when administered to an intact animal, organ, or ceU culture. A targeting component is generally accessible from outside the liposome and is, therefore, generally either bound to the outer surface or inserted into the outer lipid bilayer. A targeting component can be, for example, a peptide, a region of a larger peptide, an antibody or antigen binding fragment thereof, a nucleic acid, a carbohydrate, a region of a complex carbohydrate, a special lipid, or a small molecule such as a drug, hormone, or hapten, attached to any of the aforementioned molecules.
Attached to targeting components, the LMSs can be targeted to any cell type that is undergoing an inappropriate level of apoptosis. Such cells include, but are not limited to, cardiomyocytes, endothelial cells, neuronal cells, hepatocytes, glomerulocytes, lung cells, mucosal cells, skin cells, heart ceUs and cancer cells. LMSs can be targeted to such cell types in various ways. For example, a LMS can be modified to contain an antibody, or a fragment of an antibody, specific for a cell surface molecule, or marker, found solely or primarily on a given cell type. Antibodies with specificity toward cell type-specific cell surface markers are known in the art.
The compositions of the present invention can additionally comprise surfactants. Surfactants can be cationic, anionic, amphiphiUc, or nonionic. A preferred class of surfactants are nonionic surfactants; particularly preferred are those that are water soluble. Nonionic, water soluble surfactants include polyoxyethylene derivatives of fatty alcohols, fatty acid ester of fatty alcohols and glyceryl esters, wherein the polyoxyethylene group is coupled via an ether linkage to an alcohol group. Examples include, but are not Umited to, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene castor oil derivatives, polyoxyethylene hardened castor oU derivatives, fatty acid sodium salts, sodium cholates, polyexyethylene fatty acid ester and polyoxyethylene alkyl ethers.
4. Proteins. Proteins other than serum albumin have been characterized or impUcated as having fatty acid/Upid binding capabiUties. These include the fatty acid binding proteins (FABPs), the Upid binding proteins, long-chain fatty acyl-CoA (LCFA-CoA) binding proteins, phosphoUpid transfer proteins, and Ca +/lipid binding proteins. Lipid-binding proteins are a family of fatty acid and retinoid transport proteins. Some are intraceUular, for example, while others are secreted from the cell. IntraceUular lipid binding proteins include cellular retinoic acid binding proteins, CRABP
I and CRABP II, which belong to a family of small cytosoUc Upid binding proteins and appear to play a role in regulating transport and metabolism of retinoic acid in the developing embryo and throughout adult life. Banaszak et al. (1994) Adv. Prot. Chem. 45:89-151; and Donovan et al. (1995) J. Steroid Biochem. Mol. Biol. 53:459-465. The lipid-binding protein family includes many FABP. The FABP are relatively smaU proteins (13-15 kDa) capable of binding long-chain fatty acids (LCFA) and their coenzyme A and L-camitine esters . They are beUeved to have major functions in the metabolism of LCFA for energy production or storage, and are abundantly present in tissues such as the intestine, liver and heart, which are actively involved in the uptake or utilization of LCFA. Bass(1993) >/. Cell. Biochem. 123:191-202; and Glatzand van derVusse (1989)M>/. Cell. Biochem. 88:37-44. A Uver form of FABP has been shown to stimulate export of LPA from mitochondria. Vancura and Haldar (1992)./ Biol. Chem. 267:14353-14359.
LCFA-CoA play an important physiological role in intermediary metaboUsm of fatty acids, but recent data indicate that they also can be potent regulators of ceU functions . LCFA-CoA typically exist within a cell bound to membrane lipids and/or proteins. Proteins which bind LCFA-CoA include LCFA CoA binding protein, FABP and sterol carrier protein-2. Gossett et al. (1996) Lipids 31:895-918.
A protein that enhances a therapeutic effect of LPA can be a naturally-occurring or a synthetic protein, a protein fragment, or can contain lipid or sugar moieties. Preferably, the protein is a lipid-binding or carrier protein. The protein can contain other modifications including, but not Umited to glycosylation, phosphorylation, myristylation, sulfation and hydroxylation. The protein can be a hybrid protein, part of which confers the property of enhancing a therapeutic effect of LPA, and another part of which confers some other desirable property such as targeting to a particular cell type, enhanced in vivo stability, and the like. Preferred proteins include, but are not Umited to, albumin, soy and plant proteins, cytochrome C, low density Upoprotein, acyl carrier protein and alphafeto-protein.
Suitable polypeptides include any known in the art. Modified polypeptides contain any of a variety of modifications, including, but not Umited to glycosylation, phosphorylation, myristylation, sulfationandhydroxylation. As used herein, a suitable polypeptide is one that will protect LPA to preserve its activity. Examples of binding proteins include, but are not limited to, fatty acid binding proteins, albumins such as bovine serum albumin (BSA) and pea albumin.
Proteins active in enhancing anti-apoptotic activity of LPA can bind to LPA. "Binding" as used herein means that the LPA and the protein form non-covalent complexes with one another. Binding of a protein to LPA can be determined by a variety of methods known in the art, including a non-denaturing gel electrophoresis binding assay as described in Example_3 or a size shift assay as described in Example_3.
The protein/LPA compositions optionally exhibit a reduction in the mitogenic activity of LPA. Mitogenic activity can be measured by any method known in the art, for instance, measurement of uptake of 3H-thymidine by cells treated with LPA with or without the addition of protein.
LP Aprotein compositions of the present invention can be prepared in a variety of ways.
LPA and a protein can be suspended in any biocompatible buffer, for example, bicarbonate buffered saline, at a ratio of about 0.00001 % to 10%, more preferably about 0.001 % to 1 %. The mixtures can then be sonicated for about 5_minutes or until the mixture is clear or can be sterile filtered.
The protein concentration of the LP A/protein compositions can be from about 0.001 to about 50 mg/mL, more preferably from about 0.01 to about lO mg/mL, even more preferably from about 0.05 to about l.Ojmg/mL. The protein component of the LP A/protein compositions of the present invention are substantially pure, i.e., the protein is substantially free of lipids, other proteins, or any other material that might affect the activity of the LP A/protein complex. Preferably, the protein is at least about 75% pure, more preferably at least about 85% pure and still more preferably at least about 95% pure. 5. Other Optional Components.
Suitable polymers can be any known in the art of pharmaceuticals and include, but are not Umited to, naturaUy-occurring polymers such as dextrans, hydroxyethyl starch, and polysaccharides; and synthetic polymers. Examples of naturally occurring polymers include proteins, glycopeptides, polysaccharides, dextran and lipids. The additional polymer can be a synthetic polymer. Examples of synthetic polymers which are suitable for use in the present invention include, but are not Umited to, polyalkyl glycols (PAG) such as PEG, polyoxyethylated polyols (POP) such as polyoxyethylated glycerol (POG), polytrimethylene glycol (PTG) polypropylene glycol (PPG), polyhydroxyethyl methacrylate, polyvinyl alcohol (PVA), polyacryhc acid, polyethyloxazoUne, polyacrylamide, polyvinylpyrroUdone (PVP), polyamino acids, polyurethane and polyphosphazene. The synthetic polymers can also be linear or branched, substituted or unsubstituted, homopolymeric, copolymers, or block copolymers of two or more different synthetic monomers. The synthetic polymers can have the following generic structure:
Figure imgf000035_0001
where R, and R3 are the same or different and are H, H3C, OH, R2 or a reactive group (as described below); where R2 is a linear or branched substituted or unsubstituted alkyl group; where
X is O (in which case the synthetic polymer can be a polyoxyalkylene) or X is NH(C=0) (in which case the synthetic polymer can be a polyamine), or X is absent (in which case the synthetic polymer can be a polyalkylene); and a is an integer between 1 and about 1,000.
Biodegradable polymers can also be included in the compositions. These include, but are not Umited to poly(lactide), poly(glycoUde) poly(vinyl alcohol), crosslinked collagen. The polymers can also include polyglycolic acid, polyethylene terephthalate, polybutyl lactose, polycaprolactone, D-polylactic acid, L-polylactic acid and poly-L-lysine and polymeric mixtures thereof.
Suitable polymers also include polysaccharides. Suitable polysaccharides include, but are not limited to, trehalose, glucose, maltose, lactose, maltulose, iso-maltulose, lactulose, mono-reducing glycosides of polyhydroxy compounds selected from sugar alcohols, other straight chain polyalcohols, raffinose, stachyose, melezitose, dextran, sucrose and sugar alcohols thereof, maltitol, lactitol, iso-maltulose, palatinit, 2-D-glucopyranosyl- 1 -(-6-mannitol and their individual sugar alcohols. The compositions can further include pharmaceutically acceptable excipients.
Pharmaceutically acceptable excipients include, but are not limited to, topical pharmaceutically acceptable carrier, cosmetic carrier, sterile solutions, sterile isotonic solutions, ingestable liquids, pharmaceutically acceptable aerosols and other constituents used in solutions for organ/tissue/cell preservation and/or transplantation. The compositions can further include additional pharmaceutically effective agents. Suitable classes of pharmaceuticaUy effective agents include, but are not Umited to, drugs, antibiotics, wound healing agents and antioxidants.
Suitable drugs include, but are not Umited to, those listed in Table 1 and from the foUowing classes: antipyretic and anti-inflammatory drugs, analgesics, antiarthritics, antispasmodics, antidepressants, antipsychotics, tranquiUzers, antianxiety drugs, narcotic antagonists, antiparkinsonism agents, cholinergic antagonists, chemotherapeutic agents, immuno-suppressive agents, antiviral agents, parasiticides, appetite suppressants, antiemetics, antihistamines, antimigraine agents, coronary vasodilators, cerebral vasodUators, peripheral vasodilators, hormonal agents, contraceptives, antithrombotic agents, diuretics, antihypertensive agents, cardiovascular drugs, opioids, and vitamins are all included in the compositions described herein.
Table 1
Figure imgf000036_0001
Figure imgf000037_0001
Suitable antibiotics include, but are not limited to, ampicillin, tetracycline, chloramphenicol, erythromycin, amphotericin B and penicillin. Suitable wound healing agents include, but are not Umited to, transforming growth factors, platelet-derived growth factors, epidermal growth factors and fibroblast growth factors . Suitable antioxidants include, but are not limited to, naturaUy-occurring antioxidants such as tocopherols, (e.g., (-tocopherol, vitamin E), ascorbic acid (Vitamin_C), (-carotene (vitamin A), dihydroUpoamide and flavenoids; and synthetic antioxidants such as butylated hydroxytoluene, butylated hydroxyanisole, Trolox®, propyl gallate, other phenoUc antioxidants and phenothiazines; and chelators such as desferoxamide, HBED and
CP130.
II. Methods of Formulating Compositions for Use in the Present Invention. The invention further includes methods of formulating compositions for use in the present invention, some of which appear below and some of which appear throughout the text of the specification.
The compositions of the present invention can be in either a Uquid or soUd form. In the Uquid form, the LPA can be concentrated for dilution prior to use. Preferably, the components are in a concentration suitable for immediate use. In the case of a soUd, addition of a predetermined amount of an aqueous solution will result in the appropriate concentration of the components. The soUd can also be in powder form, for use in therapies including, but not Umited to inhalation therapies. In the case where the composition is a solution, the LPA is preferably present in an amount of from about 0.00001% to about 10% (weight/volume). More preferably, the LPA is present in an amount of from about 0.0001% to about 1% (weight/volume). Most preferably, the LPA is present in an amount of from about 0.005% to about 1% (weight/volume).
Where the composition is a solid, preferably the LPA is present in an amount of from about 0.00001% to 50% (weight/weight). More preferably, the LPA is present in an amount of from about 0.001% to 1% (weight/weight).
LPA can be suspended in any buffered solution that is preferably free of divalent cations having a pH range of 2-10, more preferably, about 4-8 and most preferably about 6-8. Suitable buffers include, but are not Umited to, D-PBS (phosphate buffered saline, free of calcium and magnesium salts; Gibco BRL) or 50 mM ammomum bicarbonate containing isotonic sodium chloride. When the compositions are to be used therapeutically, the buffered solution is preferably physiologically acceptable. A wide range ofpH values are effective. Preferably the pH is between about 5.5 to about 8. However, any pH at which the composition is at least minimally therapeutically effective is suitable for use. The mixture has been found to be most active at pH 8. Preferably, the phosphoUpids are suspended in 50 mM ammomum bicarbonate / 0.154 M sodium chloride, 250_(M EDTA with a pH of 7.7-8.0.
Preferably, if there is a mixture of phosphoUpidslipids, the mixture is dispersed in order to achieve maximal activity. Any method of dispersion that forms particles of about 5-450 microns is acceptable and about 30-100 microns is preferred. These methods include, but are not Umited to, microfluidization, extrusion and sonication, provided that the method does not denature or otherwise chemicaUy modify the phosphoUpids in such a manner as to render them toxic or of substantially diminished therapeutic activity. Typically, when small batches are prepared, the mixture is sonicated until optical clarity is attained although sonication can be continued beyond this point provided the mixture is not overheated. The preferred sonication parameters are those provided in the examples herein. As used herein, "optical clarity" indicates that the mixture changes from opaque to translucent. This change is readily monitored visually; no further measurements are necessary. However, "translucent" can be defined as when the mixture has an O.D. 600 of less than about 0.2 AU.Concentrations of up to approximately 50 mg/mL phosphoUpids can be prepared. Preferably, 10 mg/mL solutions are used. Typically, sonication, if used, is in 5 minute alternating cycles, with 5 minutes of sonication foUowed by 5 minutes of equihbration. However, this can be varied, depending on the volume of mixture being sonicated and the heat generated by sonication.
The total length of sonication depends on the concentration and volume of the mixture being sonicated and the power output of the sonicator. Sonication should proceed until the mixture has become translucent. Typically, mixtures are sonicated for 3 to 90 minutes. Preferably, sonication proceeds by several periods of 5 minutes each, 6 to 12 total periods, with 1 to 5 minutes between each period to allow equihbration and dissipation of heat. The temperature of the water bath should not exceed about 60°C. Preferably, the temperature of the water bath is not allowed to exceed 37°C. Preferably, the sonicated mixture is passed through a sterile filter before use. Preferably, the sterile filter has a 0.2 micron cut off.
The compositions can be sterilized at any point. Sterilization can be by any method known in the art, and encompasses, but is not limited to, heat sterilization, steam sterilization, ultrafiltration, sterile filtration and ultraviolet light sterilization. Sterilization is essential for most of the methods of treatment, although for certain applications, it may not be necessary, or the level of sterilization required can be reduced. The compositions can also be prepared and dried to form a solid. The soUd is suitable for use as a powder or pill, or in solution upon reconstitution. Any method of drying is suitable for use herein, including, but not limited to, freeze-drying, air drying, spray drying and fluidized bed evaporation, vacuum drying and rotary evaporation
Preferably, the compositions, both liquid and solid, are stored under anoxic conditions. Any method of such storage known in the art is suitable for use herein, including, but not Umited to, storage under an inert gas such as argon.
III. Prevention of Apoptosis and Preservation or Restoration of CeU, Tissue and Organ Function.
The anti-apoptotic activity of the compositions of the present invention can be measured in many anti-apoptosis assays known in the art. These include, but are not Umited to, the serum deprivation of the C3H/10T1/2 ceU assay described in detaU in Example 2. Furthermore, in vivo apoptosis inhibition can be measured by any method known in the art. Methods for determining therapeutic efficacy in treating an ischemic event are known in the art and described herein. Methods for determining efficacy in organ storage and transplantation are known in the art and described herein.
The therapeutic activity of the compositions described herein can be measured or determined by any method known in the art. For instance, there are a variety of wound healing assays described in the art and cited herein.
The invention further comprises any of the above-described compositions in combination with a pharmaceutically acceptable vehicle. The level of purity of the components necessary for the composition can be determined empirically and is within the skiU of one in the art. The compositions are suitable for use in a variety of disorders, as described below, and in both human and veterinary appUcations.
In general, the compositions are pharmaceutically acceptable due to their low toxicity in the therapeutic dosage range, stability and ability to be incoφorated into a wide variety of vehicles for numerous routes of administration. The compositions can be administered alone or in combination with other pharmaceutically effective agents including, but not limited to, antibiotics, wound healing agents, antioxidants and other therapeutic agents as previously described.
The compositions can contain at least a therapeutically effective amount of at least one of the above-described compositions and at least one physiologically acceptable carrier. A physiologically acceptable carrier is one that does not cause an adverse physical reaction upon administration and one in which the compositions are sufficiently soluble to deliver a therapeutically effective amount of the compound. The therapeutically effective amount of the compositions depends in part upon the manner of introduction and the indication to be treated and other criteria evident to one of ordinary skill in the art. Typically, a therapeutically effective amount is one sufficient to ameliorate or cure the condition being treated as evidenced by diminishment of the symptoms compared to a control. Typically, a therapeutically effective amount is from about 0.0001% or 1 μg/mL by weight ofthe phospholipid mixture although a wide range of effective amounts can be used for different indications and can be determined empiricaUy. The route(s) of administration useful in a particular indication are discussed below and are weU known to one of skiU in the art.
IV. Biological Materials Suitable for Treatment and Routes of Administration to These Materials.
Suitable cell types for treatment and/or preservation include, but are not limited to, eukaryotic and prokaryotic cells, such as bacterial cells, plant cells, yeast cells, fungi cells, insect ceUs, mammaUan cells, and human cells in particular. Mammalian cell types encompass cardiomyocytes, endotheUal cells, neuronal cells, hepatocytes, renal ceUs, lung cells, mucosal cells, pancreatic cells, gastrointestinal cells, corneal cells and skin cells. These cell types, and the tissues and organs they form, are suitable for treatment and/or preservation by the methods of the present invention. These cell types can be treated either in vivo or in vitro using methods of the present invention. Routes of administration include, but are not limited to, topical, transdermal, parenteral, gastrointestinal, transbronchial, transalveolar, and in vitro treatment of cells, tissues or organs foUowed by in vitro administration of treated ceUs, tissues or organs. Internal routes of administration encompass any method of in vivo administration other than solely by topical application to the skin. Surface administration is accomplished via application of a cream, gel, rinse, etc. containing a therapeutically effective amount of the compositions. Transdermal administration is accomplished by application of a cream, rinse, gel, etc. capable of allowing the active components to penetrate the skin and enter the blood stream. Parenteral routes of administration include, but are not Umited to, direct injection such as intravenous, intramuscular, intraperitoneal or subcutaneous injection. Gastrointestinal routes of administration include, but are not limited to, ingestion and rectal. Transbronchial and transalveolar routes of administration include, but are not Umited to, inhalation, either via the mouth or intranasally (for example, of a mist or a dry powder) and direct injection into an airway, such as through a tracheotomy. While the compositions can be topicaUy administered alone, it may be desirable to administer them in a mixture with a topical physiologically or cosmetically acceptable carrier. "Topical pharmaceutically acceptable carrier" as used herein is any substantiaUy non-toxic carrier conventionally useable for topical administration of pharmaceutical agents in which the compositions wiU remain stable and bioavailable when appUed directly to skin or mucosal surfaces . For example, the compositions can be dissolved in a liquid, dispersed or emulsified in a medium in a conventional manner to form a Uquid preparation or mixed with a semi-solid (gel) or soUd carrier to form a paste, powder, ointment, cream, lotion or the like.
Suitable topical pharmaceutically acceptable carriers include water, petroleum jeUy (Vaseline), petrolatum, mineral oil, vegetable oil, animal oil, organic and inorganic waxes, such as microcrystaUine, paraffin and ozocerite wax, natural polymers, such as xanthanes, gelatin, ceUulose, collagen, starch, or gum arabic, synthetic polymers, such as discussed below, alcohols, polyols, and the like. The carrier can be a water miscible carrier composition that is substantiaUy miscible in water. Such water miscible topical pbarmaceuticaUy acceptable carrier composition can include those made with one or more appropriate ingredients set forth above but can also include sustained or delayed release carriers, including water containing, water dispersible or water soluble compositions, such as liposomes, microsponges, microspheres or microcapsules, aqueous base ointments, water-in-oil or oil-in-water emulsions, gels or the like.
In one embodiment of the invention, the topical pharmaceutically acceptable carrier comprises a sustained release or delayed release carrier. The carrier is any material capable of sustained or delayed release of the compositions to provide a more efficient administration resulting in one or more of less frequent and/or decreased dosage of the compositions, ease of handling, and extended or delayed effects on dermatologic conditions. The carrier is capable of releasing the compositions when exposed to any oily, fatty, waxy, or moist environment on the area being treated or by diffusing or by release dependent on the degree of loading of the compositions to the carrier in order to obtain release thereof. Non-limiting examples of such carriers include liposomes, microsponges, microspheres, or microcapsules of natural and synthetic polymers and the like.
Examples of suitable carriers for sustained or delayed release in a moist environment include gelatin, gum arabic, xanthane polymers; by degree of loading include lignin polymers and the like; by oily, fatty or waxy environment include thermoplastic or flexible thermoset resin or elastomer including thermoplastic resins such as polyvinyl halides, polyvinyl esters, polyvinyUdene halides and halogenated polyolefins, elastomers such as brasiUensis, polydienes, and halogenated natural and synthetic rubbers, and flexible thermoset resins such as polyurethanes, epoxy resins and the like. Preferably, the sustained or delayed release carrier is a liposome, microsponge, microsphere or gel.
The compositions used in the method of treating dermatologic conditions of the invention are applied directly to the areas to be treated. While not required, it is desirable that the topical composition maintain the active components at the desired location for about 24 to 48 hours, or a length of time sufficient to exert therapeutic efficacy. If desired, one or more additional ingredients conventionally found in topical pharmaceutical or cosmetic compositions can be included with the carrier, such as a moisturizers, humectants, odor modifiers, buffers, pigments, preservatives, Vitamins such as A, C and E, emulsifiers, dispersing agents, wetting agents, odor-modifying agents, gelling agents, stabilizers, propeUants, antimicrobial agents, sunscreens, enzymes and the like. Those of skill in the art of topical pharmaceutical formulations can readily select the appropriate specific additional ingredients and amounts thereof. Suitable non-limiting examples of additional ingredients include superoxide dismutase, stearyl alcohol, isopropyl myristate, sorbitan monooleate, polyoxyethylene stearate, propylene glycol, water, alkali or alkaline earth lauryl sulfate, methylparaben, octyl dimethyl-p-amino benzoic acid (Padimate O), uric acid, reticuUn, polymucosaccharides, hydroxyethyl starch (such as, DuPont Pentafraction), hyaluronic acids, aloe vera, lecithin, polyoxyethylene sorbitan monooleate, Vitamin A or C, tocopherol (Vitamin E), alpha-hydroxy of alpha-keto acids such as pyruvic, lactic or glycolic acids, or any of the topical ingredients disclosed in U.S. patents 4,340,586, 4,695,590, 4,959,353 or 5,130,298 and 5,140,043. Because dermatologic conditions to be treated may be visible, the topical carrier can also be a topical cosmeticaUy acceptable carrier. By "topical cosmeticaUy acceptable carrier" as used herein is meant any substantiaUy non-toxic carrier conventionaUy Useable for topical administration of cosmetics in which the compositions will remain stable and bioavailable when appUed directly to the skin surface. Suitable cosmeticaUy acceptable carriers are known to those of skill in the art and include, but are not limited to, cosmeticaUy acceptable Uquids, creams, oUs, lotions, ointments, gels, or soUds, such as conventional cosmetic night creams, foundation creams, suntan lotions, sunscreens, hand lotions, make-up and make-up bases, masks and the like. Thus, to a substantial extent, topical cosmeticaUy acceptable carriers and pharmaceuticaUy acceptable carriers are similar, if not often identical, in nature so that most of the earlier discussion on pharmaceuticaUy acceptable carriers also appUes to cosmeticaUy acceptable carriers. The compositions can contain other ingredients conventional in cosmetics including perfumes, estrogen, Vitamins A, C or E, alpha-hydroxy or alpha-keto acids such as pyruvic, lactic or glycolic acids, lanolin, vaseline, aloe vera, methyl or propyl paraben, pigments and the like.
The effective amount of the compositions used to treat dermatologic conditions or diseases can vary depending on such factors as condition of the skin, age of the skin, the particular ratio of phospholipids or degree of the purity of phosphoUpids employed, the type of formulation and carrier ingredients used, frequency of administration, overall health of the individual being treated and the like. The precise amount for any particular patient use can be determined by those of skill in the dermatologic art taking into consideration these factors and the present disclosure. Preferably the composition is administered in at least two doses and no more than about six doses per day, or less when a sustained or delayed release form is used.
The compositions for topical, oral and parenteral administration usually contain from about 0.001% to about 10% by weight of the LPA compared to the total weight of the composition, preferably from about 0.01% to about 2% by weight of the mixture to the pharmaceutical composition, and especially from about 0.1% to about 1.5% by weight of the mixture to the pharmaceutical composition.
The topical composition is administered by applying a coating or layer to the skin or mucosal area desired to be treated. As a practical matter of convenience, the applied material is rubbed into the area. Applications need not be rubbed into the skin and the layer or coating can be left on the skin overnight.
The present invention provides compositions suitable for transdermal administration including, but not Umited to, pharmaceuticaUy acceptable lotions, suspensions, oils, creams, ointments, rinses, gels and liposomal carriers suspended in a suitable vehicle in which a therapeutically effective amount of the compositions has been admixed. Such compositions are appUed directly to the skin or incoφorated into a protective carrier such as a transdermal device (so-called "patch"). Examples of suitable creams, ointments etc. can be found, for instance, in the Physician's Desk Reference. Examples of suitable transdermal devices are described, for instance, in U.S. Patent No. 4,818,540 (Chien et al.).
The present invention includes compositions suitable for parenteral administration including, but not Umited to, pharmaceuticaUy acceptable sterile isotonic solutions. Such solutions include, but are not Umited to, saline and phosphate buffered saline for intravenous, intramuscular, intraperitoneal or subcutaneous injection. The present invention includes compositions suitable for gastrointestinal administration including, but not limited to, pharmaceuticaUy acceptable powders, pills or Uquids for ingestion and suppositories for rectal administration.
The present invention includes compositions suitable for transbronchial and transalveolar administration including, but not Umited to, various types of pharmaceutically acceptable aerosols for inhalation, both liquid and powder forms. An example of a drug administered in the form of an aerosol is pentamidine which is administered to AIDS patients by inhalation to prevent pneumonia caused by Pneumocystis carnii.
In some cases it may be desirable to perform internal delivery of LPA containing compositions in a localized area of the body, organ or tissue. The present invention encompasses methods of delivery including, but not Umited to, delivery by catheter inserted into a vessel. Where deUvery of the LPA containing compositions is desired to prevent or minimize damage resulting from cardiac ischemia the present invention encompasses intracoronary delivery by guide catheter.
The present invention further encompasses devices suitable for transbronchial and transalveolar administration of the compositions. Such devices include, but are not Umited to, atomizers and vaporizers. The present invention also includes devices for electrical or direct injection. Electrical injection, or iontophoresis, is the process of using a small electrical current to drive charged elements, compounds and drugs through the skin to deliver the therapeutic compound to the local tissues or to the whole body without breaking the skin. The present invention encompasses solutions suitable for flushing, perfusion, and storage of organs and tissues prior to or during transplantation. Such solutions are described in Chien et_al. (1993) "Hibernation Induction Trigger for Organ Preservation" in Medical InteUigence Unit,
R.G. Landes Co. Austin, TX. The compositions described herein can be used, for instance, to replace and improve on much more impure soy preparations currently in use.
By "organ preservation solution" (OPS) is meant an aqueous solution specifically designed to preserve organs. Preferably the organ is the heart. Preferably, the solutions are used in organ transplantation, but are also useful for use in cardioplegia during open heart surgery. The OPS can also be used to flush the organ to be transplanted either prior to or after harvesting, or both. Preferably, this solution contains between about 0.00001% to about 10%, preferably about
0.001% to about 1%, more preferably about 0.005% to about 0.1%. Preferably, this solution additionally contains between about 0.1% and 20% by weight PEG. More preferably, this solution contains between about 2% and 15% by weight PEG and most preferably, this solution contains about 8% by weight PEG. Preferably, the PEG is PEG20L. Preferably, this solution contains between about 2% and 15 % by weight PEG and can also contain effective amounts of: (a) a buffer such as NaOH, preferably about 30-40 mM, (or sufficient to result in pH of 7.2 to 7.9); (b) an impermeant anion such as Lactobionic acid, preferably about 100 mM; (c) a component providing phosphate such as KH2P04, preferably about 25 mM; (d) a component providing potassium such as KOH, preferably about 100 mM; and (e) a component controlling cell swelling such as Raffinose, preferably about 30 mM.
Optionally, the OPS also contains effective amounts of any component known in the art of organ preservation. These include, but are not Umited to glutathione, parahydroxyanisole (PHA), desferoxamine, and nitroglycerin.
The above-mentioned compositions are meant to describe, but not limit, the compositions suitable for use in the invention. The methods of producing the various compositions are within the ability of one skilled in the art and are not described in detail here. The methods of producing suitable devices for injection, topical application, atomizers and vaporizers are known in the art and will not be described in detail.
The invention further provides methods of treatment comprising administering an amount of the compositions effective to inhibit apoptosis or to preserve or restore cell, tissue or organ function. These methods entail administration of a therapeuticaUy effective amount of the above-described compositions.
Various indications which can be treated, include, but are not Umited to, those related to apoptosis, ischemia, traumatic injury and reperfusion damage. Those conditions related to apoptosis include, but are not Umited to, dermatological effects of aging, the effects of reperfusion after an ischemic event, immunosuppression, gastrointestinal perturbations, cardiovascular disorders, rejection of tissue transplantation, wound healing and Alzheimer's disease. The treatment can also diminish the apoptosis-related problems associated with immunosuppressing viruses, chemotherapeutic agents, or radiation and immunosuppressive drugs.
The compositions are also suitable for use in organ transplantation during aU phases of transplantation. The compositions can be used to prepare the organ by administering an amount of the compositions to the donor effective to stabilize or preserve the organ. The organ can be perfused and/or preserved in OPS containing the compositions. The organ recipient can then be administered an amount of the compositions effective to enhance organ stabiUty and function. The compositions are also particularly suitable for use in treating cardioplegia whether related to transplantation or other surgical intervention.
Apoptosis related problems are caused by a variety of stimuli which include, but are not Umited to, viruses including, but not Umited to, HTV, chemotherapeutic agents, and radiation. These stimuli trigger apoptosis in a variety of disorders, including, but not Umited to, those of the digestive tract tissues and associated gastrointestinal perturbations.
Gastrointestinal perturbations include, but are not Umited to, damage to the lining of the gut, severe chronic ulcers, colitis, radiation induced damage, chemotherapy induced damage, and the perturbation of the gastrointestinal tract caused by parasites, and diarrhea from any other cause. Various viral and bacterial infections are known to result in gastrointestinal perturbations; the compositions are also suitable for use in treatment of the side effects associated with these infections. The compositions are particularly suited for use in ameliorating the gastrointestinal disturbances associated with chemotherapy. Thus, the compositions are suitable for use not only in preventing the diarrhea associated with chemotherapy but also the nausea.The compositions are particularly suited to treatment of various gastrointestinal conditions in animals, including, but not
Umited to Uvestock and domesticated animals. Such conditions, particularly diarrhea, account for the loss of many calves and puppies to dehydration and malnutrition. Treatment of gastrointestinal conditions is preferably by gastrointestinal administration. In the case of cattle and domesticated animals, an effective amount of the compositions can be conveniently mixed in with the feed. In humans, administration can be by any method known in the art of gastrointestinal administration.
Preferably, administration is oral.
In addition, the compositions can be administered to immunodeficient patients, particularly HIV-positive patients, to prevent or at least mitigate apoptotic death of T ceUs associated with the condition, which results in the exacerbation of immunodeficiencies as seen in patients with AIDS. Preferably, administration to such patients is parenterally, but can also be transdermaUy or gastrointestinaUy.
The compositions can also be administered to treat apoptosis associated with reperfusion damage involved in a variety of conditions, including, but not Umited to, coronary artery obstruction; cerebral infarction; spinal/head trauma and concomitant severe paralysis; reperfusion damage due to other insults such as frostbite; coronary angioplasty, blood vessel attachment, limb attachment, organ attachment and kidney reperfusion.
Myocardial and cerebral infarctions (stroke) are caused generally by a sudden insufficiency of arterial or venous blood supply due to emboli, thrombi, or pressure that produces a macroscopic area of necrosis; the heart, brain, spleen, kidney, intestine, lung and testes are likely to be affected. CeU death occurs in tissue surrounding the infarct upon reperfusion of blood to the area; thus, the compositions are effective if administered at the onset of the infarct, during reperfusion, or shortly thereafter. The invention includes methods of treating reperfusion damage by administering a therapeutically effective amount of the compositions to a patient in need of such therapy.
The invention further encompasses a method of reducing the damage associated with myocardial and cerebral infarctions for patients with a high risk of heart attack and stroke by administering a therapeutically effective amount of the compositions to a patient in need of such therapy. Preferably, treatment of such damage is by parenteral administration of the compositions of the invention. Any other suitable method can be used, however, for instance, direct cardiac injection in the case of myocardial infarct. Devices for such injection are known in the art, for instance the Aboject cardiac syringe.
The invention further provides methods of limiting and preventing apoptosis in cells, or otherwise preserving cells, during the culture or maintenance of mammaUan organs, tissues, and ceUs, by the addition of an effective amount of the compositions to any media or solutions used in the art of culturing or maintaining mammaUan organs, tissues, and cells.
The invention further encompasses media and solutions known in the art of culturing and maintaining mammaUan organs, tissues and cells, which comprise an amount of the compositions effective to preserve or restore cell, tissue or organ function, or limit or prevent apoptosis of the ceUs in culture.
These aspects of the invention encompass mammaUan cell culture media comprising an effective amount of at least one composition and the use of such media to preserve or restore cell, tissue or organ function, or to limit or prevent apoptosis in mammalian cell culture. An effective amount is one which decreases the rate of apoptosis and/or preserves the cells, tissue or organ. The compositions have been found to limit or prevent apoptosis under circumstances in which cells are subjected to mild traumas which would normally stimulate apoptosis. Such traumas can include, but are not Umited to, low level irradiation, thawing of frozen cell stocks, rapid changes in the temperature, pH, osmolarity, or ion concentration of culture media, prolonged exposure to non-optimal temperature, pH, osmolarity, or ion concentration of the culture media, exposure to cytotoxins, disassociation of cells from an intact tissue in the preparation of primary cell cultures, serum deprivation (or growth in serum-free media).Thus, the invention encompasses compositions comprising tissue culture medium and an effective amount of the compositions. Serum-free media to which the compositions can be added as anti-apoptotic media supplements include, but are not limited to, AIM V® Media, Neuman and Tytell's Serumless Media, TroweU's T8 Media,
Waymouth's MB 752/1 and 705/1 Media, and Williams' Media E. In addition to serumfree media, suitable mammalian cell culture media to which the compositions can be added as anti-apoptotic media supplements include, but are not Umited to, Basal Media Eagle's, Fischer's Media, McCoy's Media, Media 199,RPMI Media 1630and 1640, Media based on F-10&F-12 Nutrient Mixtures, Leibovitz's L-15 Media, Glasgow Minimum Essential Media, and Dulbecco's Modified Eagle
Media. MammaUan cell culture media to which the compositions can be added further comprise any media supplement known in the art, including, but not Umited to, sugars, Vitamins, hormones, metaUoproteins, antibiotics, antimycotics, growth factors, lipoproteins and sera.
The invention further encompasses solutions for maintaining mammaUan organs prior to transplantation, which comprise an effective amount of the compositions, and the use of such solutions to preserve or restore organ function or to limit or prevent apoptosis in such mammaUan organs during their surgical removal and handling prior to transplantation. The solutions can be used to flush, perfuse and/or store the organs. In all cases, concentrations of the compositions required to limit or prevent damage to the organs can be determined empirically by one skiUed in the art by methods such as those found in the example provided below, as well as other methods known in the art.
It has now been found that the compositions can be topically appUed to the skin to treat a variety of dermatologic conditions. These conditions include, but are not limited to, hair loss and wrinkling due to age and/or photo damage. The present invention thus encompasses methods of treating dermatological conditions. Furthermore, hair loss can be caused by apoptosis of the ceUs of the hair follicles. Therefore, the compositions are suitable for use in topical treatment of the skin to prevent continued hair loss. Stenn et al. (1994) J.Jnvest. Dermatol. 103:107-111. As discussed above, these conditions are preferably treated by topical application of a composition comprising an effective amount of the compositions. An effective amount of the compositions is one which ameliorates or dirninishes the symptoms of the dermatologic conditions. Preferably, the treatment results in resolution of the dermatologic condition or restoration of normal skin function; however, any amelioration or lessening of symptoms is encompassed by the invention.
The following examples are provided to illustrate but not limit the invention. Note that references to specific LPAs using compound numbers in this document refer to the compound numbers assigned in Example 1, below.
EXAMPLE 1 Structure and Synthesis of Various Analogs of Lysophosphatidic Acid The following example sets forth the synthetic methodology and analytical data used in the construction and characterization of several lysophosphatidic acid analogs and derivatives. In addition experimental procedures and analytical data has been provided for intermediates used in the construction of these LPAs.
Contained at the end of the experimental procedures and analytical data are reaction schemes which show the synthetic routes used in compound construction. The following nomenclature and abbreviations are used in the naming of the compounds:
Bn benzyl
BSA bis(trimethylsilyl)acetamide t-BuOOH tert.-butylhydroperoxide
CNE cyanoethyl
DMAP N,N-dimethylaιnmopyridine
DMF N,N-dmιethylforrnamide lie L-isoleucine
Me methyl
Mel methyl iodide
MeOH methanol sat. saturated
TBAF tetra-butylammonium fluoride
TBS tert.-butyldimethylsilyl
TBTU 0-benzotriazol-l-yl-N,N,N',N'-tetiamethyluroniumtetrafluorO' borate
TEA triethylamine
THF tetrahydrofuran TLC thin layer chromatography
TMSBr trimethylsilyl bromide
Tr trityl
Ts tosyl
Val L-valine
Z benzyloxycarbonyl
1-O-Decyl-rαc-glycerol (Compound 1) To a stirring mixture of NaH (0.36 g, 15.1 mmol) in dry DMF (15 ml) under N2 was added solketal (0.94 ml, 7.6 mmol) dropwise over a 30 min period. After 30 min, Nal (0.30 g, 2.0 mmol) was added followed by the addition of
1-chlorodecane (1.34 g, 7.6 mmol) and stirring was continued at 50°C for 16 hours. The reaction mixture was diluted with diethyl ether (50 mL) and washed with H20 (2 x 50 mL), dried (MgS04) and concentrated to give a crude oU (1.48 g). The crude oil (1.06 g, 3.9 mmol) was treated with 1/1 2M HC1 THF (14 mL, v/v) for 2 hours, and the resulting solution was concentrated and redissolved in ethyl acetate (80 ml), washed with H20 (2 x 20 mL), dried (MgS04) and concentrated to give a yeUow oil. The oil was subjected to silica-gel column chromatography [eluent: ethyl acetate/hexane, 50/50, v/v] to give the title diol (Compound 1) (58 mg, 65%) as a colorless oil.
Η NMR (360 MHz; CDC13): ( 0.87 (3 H, t, J 7 Hz, Me), 1.26 (14 H, br s, 7 x O J, 1.53-1.59 (2 H, m, (-H2), 3.41-3.55 (4 H, m, (-H2 and 1-H2 or 1-Hj), 3.64 (1 H, dd, J 11.5 and
5 Hz, 1-H or 3-H), 3.72 (1 H, dd, J 11.5 and 4 Hz, 1-H or 3-H) and 3.83-3.88 (1 H, m, 2-H); ESI-MS (m/z, +ve): 231 (ΛΛT, 100%).
1-O-Tetradecyl-rβc-glycerol (Compound 2) To a mixture of solketal (5.0 mL, 40 mmol), 1-chlorotetradecane (10.9 mL, 40 mmol) and a catalytic amount of Nal in DMF (200 mL) was added NaH (3.1 g, 80 mmol) and the reaction was stirred for 16 hours at 50°C. After removal of the solvent in vacuo, the residue was redissolved in ethyl acetate (150 mL), washed with H20 (3 x 50 mL), dried (MgSO4) and evaporated to dryness. The obtained oil was treated with 1/1 2M HCl THF (150 mL, v/v) for 16 hours. The resulting mixture was concentrated and redissolved in ethyl acetate (150 mL), washed with H2O (3 x 50 mL), dried (MgS04) and evaporated to dryness. The crude product was purified by siUca-gel column chromatography [eluent: CH2Cl2(ethyl acetate/CH2Cl2, 50/50, v/v] to yield diol Compound 2 (8.4 g, 73%) as a white soUd.
'HNMR (360 MHz; CDC(): ( 0.87 (3 H, t, J7 Hz, Me), 1.25 (22 H, br s, 11 x CHJ, 1.57 (2 H, quintet, Jl Hz, f-H2), 2.38 and 2.77 (each 1 H, br s, 2 x OH), 3.46 (2 H, dt, J6.5 and 2 Hz, 1-H2 or (-H^, 3.49-3.55 (2 H, m, (-H2 or l-H^, 3.64 (1 H, dd, J 11.5 and 5 Hz, 3-H), 3.71 (1 H, dd, J 11.5 and 4 Hz, 3-H) and 3.85-3.86 (1 H, m, 2-H).
Dimethyl 3-0-tetradecyl-rαc-glycero-l-phosphate (Compound 3) To a solution of diol
Compound 2 (1.0 g, 3.5 mmol) and N-methylimidazole (0.45 mL, 5.6 mmol) in dry CH2C12 (25 mL) was added dimethyl chlorophosphate (0.42 mL, 3.9 mmol). After stirring for 3 days, the mixture was concentrated and purified by silica-gel column chromatography [eluent: hexane/ethyl acetate, 60/40, v/v] to yield phosphate Compound 3 (0.10 g, 7%) as an oU.
'HNMR (360 MHz; CDC13): ( 0.87 (3 H, t, Jl Hz, Me), 1.25 (22 H, br s, 11 x CHj), 1.55-1.58 (2 H, m, (-H^, 3.43-3.53 (4 H, m, 3-H2 and (-H^, 3.80 and 3.83 (each 3 H, s, 2 x OMe), 4.00-4.02 (1 H, m, 2-H) and 4.10-4.20 (2 H, m, 1-H2); ESI-MS (m/z, +ve): 397 (ΛΛT,
100%) and 419 (ΛΛT, 66).
3-O-Tetradecyl-rαc-glycero-l-phosphate (Compound 4) To a solution of diol 2 (1.0 g, 3.5 mmol) and dibenzyl N,N-diisopropylphosphoramidite (1.16 mL, 3.5 mmol) in dry CH3CN (30 mL) was added lH-tetrazole (0.12 g, 1.7 mmol). After stirring for 2 hours, t-BuOOH (2 mL) was added and after another 30 min the mixture was concentrated. The residue was purified by silica-gel column chromatography [eluent: hexane/ethyl acetate, 66/33, v/v) to give dibenzyl 3-0-tetradecyl-rac-glycero-l-phosphate (0.27 g, 14%) as an oil. Subsequent hydrogenolysis in MeOH (30 mL) in the presence of 10% Pd/C (0.5 g), foUowed by filtration over CeUte and evaporation to dryness, afforded the title phosphate (Compound 4) (0.18 g, 99%) as a glass. 'HNMR (360 MHz; CDC13): ( 0.87 (3 H, t, 6.5 Hz, Me), 1.25 (22 H, br s, 11 x CΑ ,
1.55 (2 H, br s, (-Hj), 3.45 (4 H, br s, (-H2 and 3-Hj) and 4.05-4.14 (3 H, m, 2-H and 1-Hj); 3,P NMR(146 MHz; CDC13): ( 1.11; ESI-MS (m/z, +ve): 367 (M-H+, 100%).
1- -Decyl-glycidol (Compound 6) To a stirring solution of glycidyl tosylate (2.0 g, 8.8 mmol) and decyl alcohol (1.34 mL, 7.0 mmol) in dry CH2C12 (40 mL) under N2 was added a BF3.0Et2 solution (-2.2 mL, -10% in CH2C12). After 48 hours, TLC analysis showed the reaction to be complete and the solvent was removed in vacuo to give l-0-decyl-rαc-glycero-3- »-toluenesulfonate (Compound 5) as an oil.
Crude Compound 5 was taken up in 50% aqueous MeOH (30 mL), NaOH (1.4 g, 35.0 mmol) was added and the mixture was left to stir for 16 hours. TLC analysis showed the reaction to be complete and the mixture was concentrated to remove MeOH. The remaining aqueous phase was extracted with diethyl ether (2 x 40 mL) and the combined organic phases were washed with H20 (30 mL), sat. NaHC03 (30 mL), dried (MgS0 ) and concentrated. The residue was subjected to siUca-gel column chromatography [eluent: ethyl acetate hexane, 91/9, v/v] to give epoxide Compound 6 (0.93 g, 62%) as a colourless oil. ,HNMR(360 MHz; CDCl3): (0.87 (3H,t,J7Hz, Me), 1.26 (14H, brs, 7 xCH2), 1.58
(2 H, m, (-Hj), 2.61 (1 H, dd, J5 and 2.5 Hz, 3-H), 2.80 (1 H, t, J4.5 Hz, 3-H), 3.13-3.17 (1 H, m, 2-H), 3.38 (1 H, dd,.πi.5 and 6 Hz, 1-H), 3.42-3.54 (2 H, m, (-H^ and 3.70 (l H, dd, 11.5 and 3 Hz, 1-H). 1-0-Tetradecyl-gIycidol (Compound 8) To a solution of glycidyl tosylate (2.0 g, 8.8 mmol) and 1-tetradecanol (1.5 g, 7.0 mmol) in dry CH2C1 (40 mL) under N2 was added a
BF3.OE , solution (-2.2 mL, -10% in CH2C1 . After 48 hours, TLC analysis showed the reaction to be complete and the solvent was removed in vacuo to give l-9-tetraιdecyl-roc-gly∞rcH3- >-tolueriesulfonate (Compound 7) as an oil. Crude Compound 7 was taken up in 50% aq. MeOH (30 mL), NaOH (1.4 g, 35.0 mmol) was added and the mixture was left to stir for 16 hours. TLC analysis showed the reaction to be complete and the mixture was concentrated to remove MeOH. The remaining aqueous phase was extracted with diethyl ether (2 x 40 mL) and the combined organic phases were washed with H20 (30 mL), sat. NaHC03 (30 mL), dried (MgS04) and evaporated to dryness. The residue was subjected to silica-gel column chromatography [eluent: ethyl acetate/hexane, 91/9, v/v] to give epoxide Compound 8 (1.57 g, 83%) as a colourless oil.
'HNMR (360 MHz; CDC13): ( 0.88 (3 H, t, J7 Hz, Me), 1.25 (22 H, m, 11 x CH^, 1.57 (2 H, m, (-H^, 2.61 (1 H, dd,J5.5 and 3 Hz, 3-H), 2.80 (1 H, t,J4.5 Hz, 3-H), 3.13-3.17 (1 H, m, 2-H), 3.38 (1 H, dd, Jl 1.5 and 6 Hz, 1-H), 3.42-3.54 (1 H, m, (-H^ and 3.70 (1 H, dd,./11.5 and 6 Hz, 1-H).
1-0-Oleyl-gIycidol (Compound 10) To a solution of glycidyl tosylate (2.0 g, 8.8 mmol) and oleyl alcohol (1.88 mL, 7.0 mmol) in dry CH2C12 (40 mL) under N2 was added a BF3.0Et2 solution (-2.2 mL, -10% in CH2C-2). After 48 hours, TLC analysis showed the reaction to be complete and the solvent was removed in vacuo to give 1 -O-oleyl-rac-glycero-3-p-toluenesulfonate (Compound 9) as an oU.
Crude Compound 9 was taken up in 50% aq. MeOH (30 mL) to whichNaOH(1.4 g, 35.0 mmol) was added. After 16 hours, TLC analysis showed the reaction to be complete and the mixture was concentrated to remove MeOH. The remaining aqueous phase was extracted with diethyl ether (2 x 40 mL) and the combined organic phases were washed with H20 (30 mL), sat. NaHC03 (30 mL), dried (MgS04) and concentrated. The residual oU was purified by silica-gel column chromatography [eluent: ethyl acetate hexane, 91/9, v/v] to furnish epoxide Compound 10 (1.65 g, 72%) as a colourless oil.
'H NMR (360 MHz; CDC13): ( 0.88 (3 H, t, J6.5 Hz, Me), 1.28 (22 H, apparent br d, separation 9 Hz,
Figure imgf000052_0001
1.58 (2 H, br s, (-Hj), 2.01 (4 H, apparent br d, separation 4.5 Hz, -CH2CH=CHCH2-), 2.61 (1 H, br s, 3-H), 2.80 (1 H, br t, J4 Hz, 3-H), 3.15 (1 H, br s, 2-H), 3.38 (1 H, dd, J 11.5 and 6 Hz, 1-H), 3.45-3.54 (2 H, m, (-Hj), 3.69-3.72 (1 H, m, 1-H) and 5.34 (2 H, br s, -CH2CH=CHCH2-).
3-£M)ecy--rαc- lycero-l-phosphate(Conιpound 11) A mixture of 98% phosphoric acid (0.18 g, 1.9mol) and decyl glycidol (Compound 6) (0.4 g, 1.9 mmol) in dry CH2C12 was refluxed for 2 hours until TLC analysis showed the reaction to be complete. Then, the reaction mixture was concentrated to afford phosphate Compound 11 (0.56 g, 78%) as a colourless oil.
'H NMR (360 MHz; CDC13): ( 0.87 (3 H, t, J6.5 Hz, Me), 1.25 (14 H, br s, 7 x CHj), 1.56 (2 H, br s, (-Hj), 3.44-3.53 (4 H, m, (-H2 and 3-Hj) and 3.61-4.13 (3 H, m, 2-H and 1-Hj); 31P NMR (146 MHz; CDC13): ( 1.41; ESI-MS (m/z, -ve): 311 (Λ/-JT, 100%).
3-0-Oleyl-rαc-gIycero-l-phosphate (Compound 12) A mixture of 98% phosphoric acid (0.18g, 1.9 mol) and oleyl glycidol (Compound 10) (0.4 g, 1.2 mmol) in dry CH2C12 was refluxed for 16 hours. TLC analysis showed the reaction to be complete and the mixture was concentrated to give the title phosphate (Compound 12) (0.501 g, 96%) as a colourless oil. Η NMR (360 MHz; CDC13): ( 0.87 (3 H, t J 6.5 Hz, Me), 1.27 (22 H, apparent br d, separation 6 Hz, -(CH^- and -(CH^-), 1.55 (2 H, br s, (-Hj), 1.98-2.03 (4 H, m, -C#2CH=CHC/72-), 3.44-3.51 (4 H, m, 3-H2 and (-H2), 3.53-4.12 (3 H, m, 2-H and 1-Hj), 5.30-5.38 (2 H, m, -CH2C77=07CH2-) and 6.43 (2 H, br s, 2 x OH); 3,P NMR (146 MHz; CDC13): ( 1.68; ESI-MS (m/z, -ve): 421 (M-W, 100%). Dimethyl 3-oleyioxypropyl-l-phosphate (Compound 15) To a suspension of NaH (.40 g, 13.1 mmol) and anhydrous Nal(1.98 g, 13.4 mmol) in dry DMF (30 mL) under N2 was added dropwise 1,3-propanediol (0.95 mL, 13.1 mmol) over a 30 min period. Next, oleyl tosylate (13) (5.6 g, 13.1 mmol) was added and stirring was continued for 16 hours at 50°C. The reaction mixture was quenched with water (50 mL) and extracted with diethyl ether (2 x 100 mL). The combined organic phases were dried (MgS04) and concentrated to give crude alcohol Compound
14.
To a solution of Compound 14 (0.51 g, 1.6 mmol) in dry CH2C1 (10 mL) was added N-methylimidazole (0.28 mL, 3.5 mmol) followed by the addition of dimethyl chlorophosphate (0.2 mL, 1.9 mmol). After 16 hours, the reaction was quenched with sat. KHSO (20 mL) and extracted with ethyl acetate (3 x 50 mL). The organic phases were washed with sat. NaCl (40 mL), dried
(MgS04) and concentrated to give protected phosphate Compound 15 as a yellow oil.
Η NMR (360 MHz; CDC13): ( 0.88 (3 H, t J 7 Hz, Me), 1.27 (22 H, apparent br d, separation 12 Hz, -(CH2)5- and -(CH^-), 1.55-1.57 (2 H, m, b-H2), 1.95-2.01 (6 H, m,
Figure imgf000053_0001
3.57 (2 H, t, J6.5 Hz, 3-Hj), 3.77 and 3.80 (each 3 H, s, 2 x OMe), 4.20 (2 H, m, l-H^ and 5.33-5.38 (2 H, m, -CH2C#=C H2-); ESI-MS (m/z, +ve): 435 (Mit, 100%).
3-0-OIeyloxypropyl-l-phosphate (Compound 16) To a solution of dimethyl phosphate Compound 15 (0.26 g, 0.59 mmol) in dry CH2C12 (8 mL) was added BSA (0.30 mL, 1.2 mmol) followed by the addition of TMSBr (0.24 mL, 1.8 mmol). After 15 min, TLC analysis showed complete consumption of the starting material and the reaction was quenched with 1/1 MeOH/H20 (2 mL, v/v) for 15 min, foUowed by the addition of sat. KHS04 (5 mL). The reaction mixture was subsequently extracted with ethyl acetate (2 x 30 mL), the organic extracts were combined, dried (MgS04) and concentrated to give the title phosphate (Compound 16) as an oU. Η NMR (360 MHz; CDC13): ( 0.88 (3 H, t, J 7 Hz, Me), 1.27 (22 H, apparent br d, separation 6 Hz,
Figure imgf000054_0001
1.95-2.01 (6 H, m, 2-H2 and -CH2CH=CHCH2-), 3.43 (2 H, t, J6 Hz, (-Ha), 3.57 (2 H, br s, 3-H2), 4.12 (2 H, br s, 1-H,), 5.33-5.38 (2 H, m, -CH2CH=CHCH2-) and 8.06 (2 H, br s, 2 x OH); 3,P NMR (146 MHz; CDC13): ( 1.95; ESI-MS (m/z, -ve): 405 (M-l , 100%). 3-Hydroxypropyl decyl ether (Compound 17) To a mixture of NaH (2.6 g, 70 mmol) and anhydrous Nal (9.9 g, 70 mmol) in dry DMF (80 mL) under N2 was added dropwise a solution of 1,3-propanediol (4.75 mL, 70 mmol) in DMF (20 mL). The mixture was stirred until hydrogen evolution had ceased, decyl chloride (1.34 g, 7.6 mmol) was added and stirring was continued at 50°C for 18 hours. Then, the reaction mixture was poured into H20 (300 mL) and extracted with diethyl ether (3 x 250 mL). The organic extracts were combined, washed with sat. NaCl (100 mL), dried (MgS04) and evaporated to dryness. The residue was subjected to silica-gel column chromatography [eluent: ethyl acetate/hexane, 30/70, v/v] to furnish decyl ether Compound 17 as a colourless oil.'H NMR (360 MHz; CDC13): ( 0.87 (3 H, t, J7 Hz, Me), 1.25 (14 H, br s, 7 x O J, 1.55 (2 H, quintet, J7 Hz, (-Hj), 1.82 (2 H, quintet, J5.5 Hz, 2-Ha), 2.62 (1 H, br s, OH), 3.41 (2 H, t, J7.5 Hz, (-H^, 3.60 (2 H, t, J5.5 Hz, 1-H,), and 3.77 (2 H, t, J5.5 Hz, 3-Hι);
ESI-MS (m/z, +ve): 217 (ΛΛT, 100%).
3-Decyloxypropyl-l-phosphate (Compound 19) To a solution of alcohol Compound 17 (0.5 g, 2.3 mmol) in dry CH2C12 (20 mL) was added TEA (0.48 mL, 3.5 mmol) foUowed by the addition of trimethyl phosphite (0.41 mL, 3.5 mmol). After 90 min, the reaction was cooled to -40°C, pyridinium tribromide (0.89 g, 2.8 mmol) was added and the reaction was allowed to warm to 20°C overnight. The mixture was subsequently quenched with sat. KHS04 (30 mL) and extracted with ethyl acetate (2 x 50 mL). The organic extracts were combined, washed with sat. NaCl (30 mL), dried (MgS04) and concentrated to give phosphate triester Compound 18.
SUBSTTTUTE SHEET (RULE 26) Η NMR (360 MHz; CDC13): (0.87 (3 H, t, J6.5 Hz, Me), 1.25 (14 H, br s, 7 x CH^, 1.50-1.56 (2 H, m, (-H^, 1.93 (2 H, quintet, J6 Hz, 2-^, 3.39 (2 H, t, J6.5 Hz, (-H2 or 3-Hj), 3.49 (2 H, t, J6 Hz, (-H2 or 3-Hj), 3.74 and 3.77 (each 3 H, s, 2 x OMe) and 4.14 (2 H, quartet, J6.5 Hz, 1-H,). To a stirring solution of phosphate triester Compound 18 (0.25 g, 0.72 mmol) in dry
CH2C12 (5 mL) was added BSA (0.25 mL, 1.0 mmol) foUowed by the addition of TMSBr (0.3 mL, 2.3 mmol). After 15 min, TLC analysis showed complete consumption of the starting material and the reaction was quenched with 1/1 MeOH/H20 (2 mL, v/v) for 15 min, foUowed by the addition of sat. KHS04 (5 mL). The reaction mixture was subsequently extracted with ethyl acetate (2 x 30 mL), and the combined organic extracts were dried (MgS04) and concentrated to give the title phosphate (Compound 19) as an oil.
Η NMR (360 MHz; CDC13): ( 0.88 (3 H, t, J 7 Hz, Me), 1.27 (14 H, br s, 7 x CH,), 1.55-1.58 (2 H, m, (-CH2), 1.95 (2 H, br s, 2-Hj), 3.42-3.48 (4 H, m, (-H2 and 3-H^, 4.10 (2 H, br s, 1-Hι) and 6.35 (2 H, br s, 2 x OH); ESI-MS (m/z, -ve): 295 (Λ -FT, 100%). Dimethyl l-O-oIeyl-2-O-methyl-rαc-glycero-l-phosphate (Compound 23) To a solution of tosyl chloride (19.1 g, 0.10 mol) in dry CH2C12 (200 mL) at 0°C was added TEA (15.3 mL, 0.10 mol) and oleyl alcohol (37.2 mL, 0.10 mol). The reaction mixture was allowed to warm to 20°C and was subsequently stirred for 3 days. Next, the solvent was removed in vacuo and the resulting residue was redissolved in ethyl acetate (400 mL), washed with sat. NaHC03 (40 mL), dried (MgS04), concentrated and subjected to sihca-gel column chromatography [eluent: hexane/CH2Cl2, 66:33, v/v] to give oleyl tosylate (Compound 13) as an oil.
To a mixture of NaH (4.8 g, 120 mmol) and Nal (0.2 g) in dry DMF (20 mL) was added solketal (5.0 mL, 40 mmol) dropwise over a 30 min period. Once the effervescence had stopped, oleyl tosylate (Compound 13) (16.9 g, 40 mmol) was added and the reaction mixture was left for 3 days at 50°C. Then, H20 (10 mL) was added and the mixture was extracted with diethyl ether
(2 x lOOmL), the combined organic phases were dried (MgS04) and evaporated to dryness. The residue was taken up in THF (150 mL), 2M HCl was added until the solution turned turbid and the reaction was left to stir for 16 hours. The mixture was concentrated and subjected to silica-gel column chromatography [eluent: ethyl acetate/hexane, 50/50, v/v] to give 1-O-oleyl-r c-glycerol (Compound 20) as an oil.
Diol Compound 20 (2.0 g, 5.97 mmol) was added dropwise to a solution of trityl chloride (1.66 g, 6.0 mmol) in anhydrous pyridine (10 mL). The reaction was left to stir for 16 hours after which the solvent was removed by co-evaporation with toluene (2 x 5 mL). Then, part of the residue (1.0 g, 1.7 mmol) was added dropwise to a suspension of NaH (0.14 g, 3.4 mmol) in THF (50 mL), followed by the addition of Mel (0.22 mL, 3.4 mmol). After 16 hours, TLC analysis showed that the reaction had gone to completion and H20 (5 mL) was added foUowed by concentration of the reaction mixture. The residue was partitioned between H20 (20 mL) and diethyl ether (100 mL), the organic extract was washed with sat. NaHC03 (10 mL), dried (MgS0 ) and concentrated to give l-(9-oleyl-2-9-methyl-3-0-tritylglycerol(Compound21)asanoU.Crude
Compound 21 (1.16 g) was dissolved in CH2C12 (10 mL) after which trifluoroacetic acid (2 mL) was added. The reaction mixture was stirred for 5 min and excess TEA was quenched by adding soUd NaHC03 until effervescence ceased. The mixture was diluted with H20 (10 mL), extracted with CH2C12 (2x50 mL) and the combined organic phases were dried (MgS0 ), concentrated and subjected to silica-gel column chromatography [eluent: hexane/ethyl acetate, 80/20, v/v] to give glycerol derivative Compound 22 (0.28 g, 42%) as an oil.
•H NMR (360 MHz; CDC13): ( 0.88 (3 H, t J 7 Hz, Me), 1.27 (22 H, apparent br d, separation 9.5 Hz, -(CH^- and -(CH^-), 1.53-1.60 (2 H, m, (-Hj), 1.98-2.01 (4 H, m, -C//2CH=CHC#2-), 3.37-3.46 (3 H, m, 2-H and (-Hj), 3.47 (3 H, s, OMe), 3.52 (1 H, dd, J 10 and 5 Hz, 1-H), 3.56 (1 H, dd, J 10 and 5 Hz, 1-H), 3.65 (1 H, dd, J 11.5 and 5.5 Hz, 3-H), 3.76
(1 H, dd, J 11.5 and 4 Hz, 3-H) and 5.30-5.39 (2 H, m, -CH2C#=C#CH2-).
To a solution of compound Compound 22 (0.26 g, 0.76 mmol) in dry CH2C12 (8 mL) was added N-methylimidazole (67 (1, 0.84 mmol) followed by the addition of dimethyl chlorophosphate (0.12 mL, 1.22 mmol). After 1 hours, TLC analysis showed the reaction to be complete, sat. KHS04 (20 mL) was added and the mixture was extracted with ethyl acetate (3 x 50 mL). The organic phases were combined and washed with sat. NaCl (40 mL), dried (MgS04), concentrated and subjected to sihca-gel column chromatography [eluent: ethyl acetate/hexanes, 50/50, v/v] to give the title dimethyl phosphate (Compound 23) as an oU.
Η NMR (360 MHz; CDC13): ( 0.88 (3 H, t J 7 Hz, Me), 1.27 (22 H, apparent br d, separation 12 Hz, -(CH^- and -(CH^-), 1.54-1.59 (2 H, m, (-H^, 1.98-2.01 (4 H, m,
^H2CΑ= ΑCH2-), 3.41-3.56 (5 H, m, 2-H, 3-H2 and (-H^, 3.47 (3 H, s, OMe), 3.77 (3 H, d, J2 Hz, OMe), 3.80 (3 H, d, J2 Hz, OMe), 4.08 (1 H, ddd, J 11, 7.0 and 5.5 Hz, 1-H), 4.19 (1 H, ddd, J 11, 7.0 and 4 Hz, 1-H) and 5.30-5.39 (2 H, m, -CH2C/7=CHCH2-); 31P NMR (146 MHz; CDC13): ( 2.60; ESI-MS (m/z, +ve): 465 (ΛΛT, 100%). 3-0-Oleyl-2-0-methyl-rαc-glycero-l-phosphate (Compound 24) To a stirring solution of protected phosphate Compound 23 (85 mg, 0.18 mmol) in dry CH2C12 (5 mL) was added BSA (140 (L, 0.55 mmol) and TMSBr (50 (L, 0.05 mmol). After 30 min, TLC analysis showed the reaction to be complete, 1/1 MeOH/H20 (1 mL, v/v) was added and left for 30 min. Then, sat. KHS04 (5 mL) was added and the mixture was extracted with ethyl acetate (2 x 30 mL), the combined organic extracts were dried (MgS0 ), evaporated to dryness and subjected to Sephadex LH-20 column chromatography [eluent: MeOH/CH2Cl2, 50/50, v/v] to give phosphate Compound 24 as an oil.
Η NMR (360 MHz; CDC13): ( 0.87 (3 H, t J 7 Hz, Me), 1.25 (22 H, apparent br d, separation 4 Hz, -(CH^- and -(CH^-), 1.55-1.56 (2 H, m, (-H^, 1.98-2.03 (4 H, m,
-C#2CH=CHCH2-), 3.40-3.58 (5 H, m, 2-H, 3-H2 and (-H^, 3.46 (3 H, s, OMe), 3.99-4.05 (2
H, m, 1-Hj), 5.30-5.37 (2 H, m, -CΑ2CH=CH Α2-) and 5.75 (2 H, br s, 2 x OH); 31P NMR (146
MHz; CDC13): ( 1.68; ESI-MS (m/z, -ve): 435 (Λ -rT, 100%).
Dimethyl 3-O-oleyl-r c-glycero-l-thiophosphate (Compound 25) To a solution of 1-O-Oleyl-glycerol (Compound 20) (1.5 g, 4.4 mmol) and dimethyl chlorothiophosphate (0.80 mL,
6.57 mmol), dried by azeotropic removal of H20 with CH3CN (2 x 15 mL), in CH2C12 (20 mL) under N2 was added N-methylimidazole (0.70 mL, 8.8 mmol). After 3 days, the mixture was concentrated and subjected to silica-gel column chromatography [eluent: hexane thyl acetate, 90/10, v/v] to give protected thiophosphate Compound 25 (0.17 g, 25%) as an oil. 'H ΝMR (360 MHz; CDC13): ( 0.87 (3 H, t, J6 Hz, Me), 1.27 (22 H, apparent br d, separation 12 Hz, -(CH^- and -(CH^-), 1.54-1.58 (2 H, m, (-H^, 1.98-2.04 (4 H, m, -CH2CH-~CHCH2-), 3.44-3.53 (2 H, m, 3-Hj), 3.75 and 3.79 (each 3 H, s, 2 x OMe), 3.97-4.03 (1 H, m, 2-H), 4.04-4.18 (2 H, m 1-H,) and 5.30-5.35 (2 H, m, -CH2Ci7=07CH2-); ESI-MS (m/z, +ve): 467 ( T, 52%), 489 (WΝa+, 100) and 505 (Mff, 20%). 3-Hydroxypropyl oleate (Compound 28) To a solution of 1,3-propanediol (3.61 mL,
50.0 mmol) in pyridine (100 mL) was added chlorotriphenylmethane (14 g, 50.0 mmol) and the mixture was stirred at 70°C for 16 hours. The solvent was removed by co-evaporation with dry toluene (2 x 50 mL) and the resulting residue was redissolved in diethyl ether (150 mL), washed with H20 (2 x 50 mL), dried (MgS0 ), concentrated and purified by silica-gel column chromatography [eluent: hexane/ethyl acetate, 90/10, v/v] to give monotritylated diol Compound
26.
Η NMR (360 MHz; CDC13): ( 1.82 (2 H, quintet, J6 Hz, 2-H2), 3.23 (2 H, t, J6 Hz, 1-H2 or 3-H2), 3.72 (2 H, t, J5.5 Hz, 1-H2 or 3-^, 7.18-7.28 (10 H, m, 2 x Ph) and 7.34-7.40 (5 H, m, Ph). To a solution of Compound 26 (3.2 g, 10 mmol) and DMAP (50 mg) in pyridine (10 mL) was added oleoyl chloride (3.3 g, 10 mmol). After 4 hours, pyridine was removed by co-evaporation with dry toluene (2 x 50 ml) and the obtained residue was partitioned between diethyl ether (150 mL) and H20 (50 mL). The diethyl ether layer was subsequently washed with sat. NaCl (50 mL), dried (MgS04) and evaporated to dryness. The residue was purified by siUca-gel column chromatography [eluent: hexane diethyl ether, 90/10, v/v] to give trityl ester 27. Treatment of a solution of Compound 27 (3.72 g, 6.39 mmol) in dry CH2C12 (100 mL) with trifluoroacetic acid (5 mL) resulted in a bright yeUow solution. Subsequently, the mixture was quenched by the addition of H20 ( 10 mL) and solid NaHC03 (5 g) . The product was extracted into
CH2C12 (2 x 100 mL), the organic phases were combined, dried (MgS04) and concentrated. Purification of the residue by silica-gel column chromatography [eluent: ethyl acetate/hexanes, 30/70, v/v] furnished the title alcohol (Compound 28) (2.1 g, 95%) as an oU.
'H NMR (360 MHz; CDC13): ( 0.87 (3 H, t, Jl Hz, Me), 1.28 (20 H, apparent br d, separation 13.5 Hz, -(CH^- and -(CH^-), 1.60-1.63 (2 H, m, (-H2), 1.86 (2 H, quintet, J 6 Hz,
2-H;), 1.98-2.01 (4 H, m, -CH2CH=CHCH2-), 2.31 (2 H, t, J7.5 Hz, -HJ, 3.68 (2 H, t, J6 Hz, 3-Hi), 4.24 (2 H, t, J6 Hz, 1-H,), and 5.29-5.39 (2 H, m, -CH2C#=C#CH2-).
Dimethyl l-(3-propyl oleoate)phosphate (Compound 29) To a solution of alcohol Compound 28 (0.52 g, 1.5 mmol) andN-methylimidazole (0.27 mL, 3.3 mmol) in dry CH2C12 (5 mL) was added dimethyl chlorophosphate (0.2 mL, 1.8 mmol). After 1 hours, TLC analysis showed the reaction to be complete, sat. KHS04 (20 mL) was added and the mixture was extracted with ethyl acetate (3 x50 mL), the combined organic phases were washed with sat. NaCl (40 mL), dried (MgS04) and concentrated. The residue was subjected to silica-gel column chromatography [eluent: ethyl acetate/hexanes, 50/50, v/v] to give protected phosphate Compound 29 as an oU. 'H NMR (360 MHz; CDC13): ( 0.87 (3 H, t, J 7 Hz, Me), 1.28 (20 H, apparent br d, separation 13 Hz, -(CH^- and -(CH^-), 1.59-1.64 (2 H, m, (-H2), 1.98-2.04 (6 H, m, 2-H2 and -CH2CH=CHCH2-), 2.30 (2 H, t, J7.5 Hz, (-HJ, 3.75 and 3.78 (each 3 H, s, 2 x OMe), 4.11-4.20 (4 H, m, 1-H2 and 3-H2), 5.29-5.39 (2 H, m, -CH2CH=CHCH2-); 31P NMR (146 MHz; CDC13): ( 2.54; ESI-MS (m/z, +ve): 449 (ΛΛT, 100%) and 466 ( +18+, 89). l-(3-propyl oleoate)phosphate (Compound 30) To a stirring solution of dimethyl phosphate 29 (0.31 g, 0.7 mmol) in dry CH2C12 (4 mL) was added BSA (0.52 mL, 2.1 mmol) foUowed by the addition of TMSBr (0.19 mL, 1.4 mmol). After 30 min, TLC analysis showed complete consumption of the starting material, the reaction was quenched with 1/1 MeOH/H20 (1 mL, 1/1) for 30 min, foUowed by the addition of sat. KHS04 (5 mL). The mixture was extracted with ethyl acetate (2 x 30 mL), the organic extracts were combined, dried (MgS0 ) and evaporated to dryness. Purification of the residue by Sephadex LH-20 column chromatography [eluent: MeOH/CH2Cl2, 50/50, v/v] gave the title phosphate (Compound 30) as an oil.
*H NMR (360 MHz; CDC13): ( 0.87 (3 H, t, J7 Hz, Me), 1.28 (20 H, apparent br d, separation 11 Hz, -(CH^- and -(CH^-), 1.57-1.61 (2 H, m, (-HJ, 2.00 (6 H, apparent br d, separation 3.5 Hz, -C-Y2CH=CHC#2- and 2-Hj), 2.30 (2 H, t, J7.5 Hz, (-H^, 4.11 (2 H, quartet, J6 Hz, 3-Ha), 4.19 (2 H, t, J6.5 Hz, 1-H^, 5.29-5.38 (2 H, m, -CH2CH=CHCH2-) and 9.24 (2 H, br s, 2 x OH); 31P NMR (146 MHz; CDC13): ( 1.80; ESI-MS (m/z, -ve): 419 (M-H+, 100%).
Dimethyl 3-0-oleoyl-2-deoxy-2-bromo-r c-glycero-l-phosphate (Compound 36) To a stirring solution of l-0-benzyl-2-deoxy-2-bromo-rac-glycerol (0.5 g, 2.0 mmol), dimethyl chlorophosphate (270 (L, 2.5 mmol) in dry CH2C12 (20 mL) under N2 was added N-methylimidazole (180 (L, 2.2 mmol). After 3 days, the mixture was concentrated and purified by silica-gel column chromatography [eluent: ethyl acetate hexane, 50/50, v/v] to yield dimethyl 3-0-benzyl-2-deoxy-2-bromo-rac-glycero-l-phosphate (Compound 34) (0.43 g, 59%) as an oU. 'H ΝMR (360 MHz; CDC13): ( 3.73-3.81 (2 H, m, 3-H^, 3.75 (3 H, apparent d, separation 13.5 Hz, OMe), 3.78 (3 H, apparent d, separation 14 Hz, OMe), 4.21 (1 H, quintet, J 5.5 Hz, 2-H), 4.28-4.40 (2 H, m, 1-H2), 4.57 (2 H, br s, -0O72Ph) and 7.26-7.37 (5 H, m, -Ph); 3,P ΝMR (146 MHz; CDC13): ( 1.88; ESI-MS (m/z, +ve): 353 and 355 (ΛΛT, 100%).
A mixture of bromo Compound 34 (0.43 g, 1.2 mmol) and 10% palladium on activated carbon (0.43 g) in ethyl acetate (50 mL) was evacuated using an aspirator pump and filled with hydrogen. After 2 hours, TLC analysis showed the reaction to be complete and the mixture was filtered through Celite. The filtrate was evaporated to dryness to furnish dimethyl
2-deoxy-2-bron wαc-glycero-l -phosphate (Compound 35) (0.28 g, 88%) as a colourless oU. lHΝMR (360 MHz; CDC13): ( 2.35 (1 H, br s, OH), 3.80 (3 H, d, J7 Hz, OMe), 3.83 (3 H, d, J 6.5 Hz, OMe), 3.89 (1 H, dd, J 12.5 and 6 Hz, 3-H), 3.94 (1 H, dd, J 12.5 and 5 Hz,
3-H), 4.13-4.19 (1 H, m, 2-H), 4.31 (1 H, ddd,J11.5, 9.5 and 6 Hz, 1-H) and 4.46 (1 H, ddd, J 11.5, 9 and 4.5 Hz, 1-H).
To a solution of alcohol Compound 35 (0.28 g, 1.1 mmol) in dry CH2C12 (5 mL) under N2 was added oleoyl chloride (0.39 mL, 1.2 mmol) foUowed by the addition of pyridine (0.09 mL, 1.2 mmol). After 1 hour, the mixture was concentrated and subjected to silica-gel column chromatography [eluent: ethyl acetate (20-50%) in hexane, v/v] to give phosphate Compound 36 (0.48 g, 85%) as an ou.Η NMR (360 MHz; CDC13): ( 0.86 (3 H, t, J 7 Hz, Me), 1.25 (20 H, apparent doublet, separation 14 Hz, -(CH2)4- and -(CH^-), 1.58-1.62 (2 H, m, (-H^, 1.97-1.98 (4 H, m, -Ci72CH=CHCH2-), 2.33 (2 H, t,J7.5 Hz, (-H2), 3.76 (3 H, d, J 1.5 Hz, OMe), 3.79 (3 H, d, J2 Hz, OMe), 4.20-4.31 (3 H, m, 2-H and 3-Hj), 4.36 (1 H, dd, J 12 and 5 Hz, 1-H), 4.40
(1 H, dd, J 12 and 5.5 Hz, 1-H) and 5.26-5.36 (2 H, m, -CH2C/7=CtfCH2-); 31P NMR (146 MHz; CDC13): ( 1.82; ESI-MS (m/z, +ve): 527 and 529 (ΛΛT, 29%), 549 and 551 (ΛΛT, 49) and 565 and 567 (ΛΛT, 100). 3-0-Oleoy I-2-deoxy-2-bromo-rαc-glycero-l-phosphate (Compound 37) To a solution of bromo alcohol Compound 36 (0.20 g, 0.38 mmol) in dry CH2C12 (20 mL) under N2 was added BSA (0.21 ml, 0.83 mmol) followed by the addition of TMSBr (0.11 mL, 0.83 mmol). The reaction was carefully monitored by TLC and after 85 min all the starting material had been consumed. Then, 1/1 MeOH/H20 (3 mL, v/v) was added and the mixture was stirred for a subsequent 15 min after which sat. KHS04 (5 mL) was added. The mixture was subsequently extracted with ethyl acetate (3 x 10 mL), the organic extracts were combined, dried (MgS0 ), concentrated and subjected to Sephadex LH-20 column chromatography [eluent: MeOH/CH2Cl2, 50/50, v/v] to give the title phosphate (Compound 37) (0.19 g, 99%) as a yellow oU. ,HNMR (360 MHz; CDCl3): ( 0.87 (3 H, t,J7 Hz, Me), 1.28 (20 H, apparent doublet- separation 13.5 Hz, -(CH^- and -(CB^-), 1.61-1.62 (2 H, m, (-H2), 1.99-2.01 (4 H, m,
Figure imgf000060_0001
2.36 (2 H, t, Jl Hz, (-HJ, 4.19-4.28 (3 H, m, 2-H and 3-B,), 4.37 (1 H, dd, J 12 and 5 Hz, 1-H), 4.47 (1 H, dd, J 12 and 5 Hz, 1-H), 5.29-5.38 (2 H, m, -CH2C#=C#CH2-) and 9.05 (2 H, s, 2 x OH); 31P NMR (146 MHz; CDC13): ( 1.08; ESI-MS (m/z, -ve): 499 and 501 (M-H+, 100%). l-0-OIeoyI-2-0-methyl-rαc-gIycerol (Compound 44) To a mixture of 1-0-benzyl-glycerol (27 mL, 0.16 mol) and t-butyldimethylsuyl chloride (25 g, 0.17 mol) in dry CH2C12 (250 mL) under N2 was added DMAP (0.8 g, 6.6 mmol) foUowed by the addition of TEA (23 mL, 0.17 mol). After 3 hours, the mixture was washed withH20 (2 x 100 mL), dried (MgS04) and concentrated to give l-O-benzyl-3-O- butyldimethylsilyl-rac-glycerol (40) (48.8 g, 100%) as a yellow oil.'H NMR (360 MHz; CDC13): ( 0.06 (6 H, s, MβjSi), 0.89 (9 H, s, Bu'Si), 3.48-3.56 (2 H, m, 1-H2 or 3-Hj), 3.64 (1 H, dd, J 10 and 5.5 Hz, 1-H or 3-H), 3.68 (1 H, dd, J 10 and 5 Hz, 1-H or 3-H), 3.86 (1 H, quintet, J 5.5 Hz, 2-H), 4.56 (2 H, s, -OC#2Ph) and 7.27-7.35 (5 H, m, -Ph). Alcohol Compound 40 (48.8 g, 0.16 mmol) was added to a mixture of NaH (4.0 g, 0.17 mol) in dry THF (500 mL). After 5 min, Mel (10 mL, 0.17 mol) was added and, after another 3 hours, the mixture was quenched with H20 (150 mL), extracted into diethyl ether (500 mL), dried (MgS04) and concentrated. Purification of the residue by silica-gel column chromatography [eluent: hexane/ethyl acetate, 90/10, v/v] yielded l- -benzyl-3-(9-t-bu1 ldimethylsilyl-2- -fliethyl- røc-glycerol (Compound 41) (37 g, 74%) as a yeUow oU.
ΗNMR (360 MHz; CDC13): ( 0.05 and 0.05 (each 3 H, s, MβjSi), 0.88 (9 H, s, Bu'Si), 3.42 (1 H, apparent quintet, separation 4.5 Hz, 2-H), 3.47 (3 H, s, OMe), 3.51 (1 H, dd, J 10 and 5.5 Hz, 1-H or 3-H), ), 3.61 (1 H, dd, J10and4Hz, 1-H or 3-H), 3.68 (2 H, <LJ5.5 Hz, 1-H or 3-H), 4.54 (2 H, apparent d, separation 12 Hz, -OCHflAr), 4.58 (2 H, apparent d, separation 12 Hz, -OCHHAr), 7.28 (1 H, m, p-H) and 7.34 (4 H, apparent d, separation 4.5 Hz, o-H2 and w-Hj); ESI-MS (m/z, +ve): 333 (MNa+, 100%).
A mixture of the protected glycerol derivative Compound 41 (5.0 g, 16 mmol) and 10% paUadium on activated carbon (wet Degussa type E101 NE/W) (0.43 g) in MeOH (50 mL) was evacuated using an aspirator pump and filled with hydrogen. After 2 hours, TLC analysis showed the reaction to be complete, the mixture was filtered through CeUte and the filtrate was evaporated to dryness to give l-O-t-butyldύnethylsilyl-2- -methyl-roc-glycerol (Compound 42) (99%) as a colourless oU.
'H NMR (360 MHz; CDC13): ( 0.07 (6 H, s, Me^i), 0.89 (9 H, s, Bu'Si), 3.30-3.36 (1 H, m, 2-H), 3.46 (3 H, s, OMe), 3.63 (1 H, dd, J 11.5 and 5.5 Hz, 1- or 3-H), 3.65 (1 H, dd, J
10.5 and 6.5 Hz, 1- or 3-H), 3.74 (1 H, dd,J10.5 and 5 Hz, 1- or 3-H) and 3.75 (1 H, dd,J11.5 and 4 Hz, 1- or 3-H).
To a solution of alcohol Compound 42 (1.1 g, 5.0 mmol) in CH2C12 (10 mL) was added oleoyl chloride (1.8 mL, 5.5 mmol) foUowed, after 5 min, by the addition of pyridine (0.44 mL, 5.5 mmol). After TLC analysis indicated that the reaction had gone to completion, the mixture was partitioned between ethyl acetate (100 mL) and H20 (30 mL), the organic extract was dried (MgS04) and concentrated to give l-0-t-butyloimethylsilyl-2-C)-methyl-3-0-oleoyl-rαc-glycerol (Compound 43) (2.4 g, 99%) as a dark brown oU.
'H NMR (360 MHz; CDC13): ( 0.06 (6 H, s, Me^i), 0.88 (3 H, t, J7 Hz, Me), 0.89 (9 H, s, Bu'Si), 1.28 (20 H, apparent br d, separation 11.5 Hz, -(CH2)4- and -(CH^-), 1.60-1.64 (2
H, m, (-Hj), 1.98-2.03 (4 H, m, -CH2CH=CHCH2-), 2.33 (2 H, t, J7.5 Hz, (-Hj), 3.41-3.46 (1
H, brt, 2-H), 3.45 (3 H, s, OMe), 3.64 (1 H, dd,J 10.5 and 6 Hz, 3-H), 3.68 (1 H, dd, J10.5 and
5.5 Hz, 3-H), 4.09 (1 H, dd, J 11.5 and 5.5 Hz, 1-H), 4.27 (1 H, dd, J 11.5 and 4 Hz, 1-H) and
5.32-5.36 (2 H, m, -CH2C#=C#CH2-). 1M TBAF (6 mL, 6.0 mmol) was added to a solution of compound Compound 43 (2.4 g,
5.0 mmol) in THF (50 mL). After 30 min, TLC analysis showed that aU the starting material had been consumed. Then, the reaction mixture was partitioned between diethyl ether (100 mL) and sat. NaCl solution (30 mL). The organic extract was dried (MgS04), concentrated and subjected to siUca-gel column chromatography [eluent: hexane/ethyl acetate, 75/25, v/v] to give glycerol derivative Compound 44 (1.52 g, 79%) as an oil
'H NMR (360 MHz; CDC13): ( 0.88 (3 H, t, J 7 Hz, Me), 1.28 (20 H, apparent br d, separation 13 Hz, -(CH2)4- and -(CHj)6-), 1.60-1.64 (2 H, m, (-HJ, 1.98-2.03 (4 H, m, -C/72CH=CHCH2-), 2.33 (2 H, t, J7.5 Hz, (-H , 3.45-3.51 (1 H, brt, 2-H), 3.47 (3 H, s, OMe), 3.61 (1 H, dd, J11.5 and όHz, 3-H), 3.69 (1 H, dd,J11.5 and4.5 Hz, 3-H), 4.20 (2 H, apparent d, separation 5 Hz, l-B and 5.29-5.39 (2 H, m, -CH2CH=CHCH2-).
Dimethyl 3-0-oIeoyI-2-0-methyl-rαc-glycero-l-phosphate (Compound 45) To a solution of alcohol Compound 44 (0.79 g, 2.2 mmol) in dry CH2C12 (8 mL) was added N-methylimidazole (0.2 mL, 2.4 mmol) followed by the addition of dimethyl chlorophosphate (0.26 mL, 2.4 mmol). After stirring for 1 hour, the mixture was quenched with sat. KHS04 (20 mL) and extracted into ethyl acetate (3 x50 mL). The combined organic phases were subsequently washed with sat. NaCl (40 mL), dried (MgS04), concentrated and subjected to silica-gel column chromatography [eluent: ethyl acetate/hexanes, 50/50, v/v] to give protected phosphate Compound 45 as an oU.
'H NMR (360 MHz; CDC13): ( 0.87 (3 H, t, J 7 Hz, Me), 1.28 (20 H, apparent br d, separation 12.5 Hz, -(CH2)4- and -(CHjλr), 1.60-1.64 (2 H, m, (-H2), 1.98-2.01 (4 H, m, -Ci/2CH=CHCH2-), 2.33 (2 H, t, J7.5 Hz, (-Hj), 3.47 (3 H, s, OMe), 3.61 (1 H, quintet, J5 Hz, 2-H), 3.76 (3 H, d, J2 Hz, OMe), 3.80 (3 H, d, J2 Hz, OMe), 4.05-4.19 (3 H, m, 1-H2 or 3-H2 and 3-H or 1-H), 4.25 (1 H, dd, J 12 and 4.5 Hz, 1-H or 3-H) and 5.29-5.39 (2 H, m,
-CH2C/7=CHCH2-); 31P NMR (146 MHz; CDC13): ( 2.52; ESI-MS (m/z, +ve): 479 ( T, 100%) and 496 (Λ +18+, 83).
3-0-Oleoyl-2-0-methyl-rσc-glycero-l-pb.osphate (Compound 46) To a solution of dimethyl phosphate Compound 45 (0.10 g, 0.22 mmol) in dry CH2C12 (5 mL) was added BSA (0.16 ml, 0.65 mmol) followed by the addition of TMSBr (60 (L, 1.4 mmol). After 30 min, the reaction was treated with 1/1 MeOH/H20 (1 mL, v/v) for 30 min after which sat. KHS04 (5 mL) was added. The mixture was subsequently extracted with ethyl acetate (2 x 30 mL), the combined organic extracts were dried (MgS04), concentrated and subjected to Sephadex LH-20 column chromatography [eluent: MeOH/CH2Cl2, 50/50, v/v] to give the title phosphate (Compound 46) as an oil.
•H NMR (360 MHz; CDC13): ( 0.86-0.87 (3 H, m, Me), 1.28 (20 H, apparent br d, separation 12 Hz, -(CH^- and -(CH^-), 1.61 (2 H, br s, (-H2), 2.00 (4 H, apparent br d, separation 5 Hz, -CH2CH=CHCH2-), 2.34 (2 H, t, J7.5 Hz, (-H^, 3.48 (3 H, s, OMe), 3.66 (1 H, br s, 2-H), 4.15 (3 H, m, 1-H2 or 3-H2 and 3-H or 1-H), 4.27 (1 H, m, 1-H or 3-H), 5.30-5.34 (2 H, m,
Figure imgf000062_0001
and 8.41 (1 H, br s, 2 x OH); 31P NMR (146 MHz; CDC13): ( 1.82;
ESI-MS (m/z, -ve): 449 (Λ/-IT, 100%) and 463 (MSfa+, 37).
Bis-(2-cyanoethyI)3-0-oleoyI-2-0-methyl-rαc-glycero-l-thiophosphate (Compound 47) 2-Cyanoethyl NNN'.N'-tetraisopropylphosphorodiamidite (0.53 mL, 1.7 mmol) was added under N2 to a solution of 3-hydroxypropionitrile (94 (1, 1.4 mmol) and 1 H-tetrazole (0.12 g, 1.7 mmol) in dry CH2C1 (10 mL). After stirring for 1 hour, a subsequent portion of 1 H-tetrazole (0.19 g, 2.8 mmol) was added followed by the addition of alcohol Compound 44 (0.51 g, 1.4 mmol). After an additional 30 min, elemental sulfur (1 g) and 1/1 CS2/pyridine (1 mL, v/v) was added. After 2 hours, the reaction mixture was filtered through a short silica-gel plug, concentrated and subjected to silica-gel column chromatography [eluent: ethyl acetate (20- 100%) in hexane, v/v] to yield protected thiophosphate Compound 47 (0.29 g, 37%) as a pale yellow oU.
Η NMR (360 MHz; CDC13): ( 0.88 (3 H, t, J 7 Hz, Me), 1.28 (20 H, apparent br d, separation 13.5 Hz, -(CH,),- and -(CH^-), 1.60-1.64 (2 H, m, (-H^, 1.98-2.01 (4 H, m, -CH2CH=CHCH2-), 2.34 (2 H, t, J7.5 Hz, (-BJ, 2.78 (4 H, t, J6 Hz, 2 x NCC T2CH2-), 3.47 (3 H, s, OMe), 4.11-4.48 (8 H, m, 1-H2, 3-H2, and 2 x NCCH2C//2-) and 5.29-5.39 (2 H, m,
-CH2Cff=C#CH2-); 31P NMR (146 MHz; CDC13): ( 69.08; ESI-MS (m/z, +ve): 595 (WNa+, 100%).
3-0-OleoyI-2-0-methyl-r c-glycero-l-thiophosphate (Compound 48) To a solution of Compound 47 (100 mg, 0.18 mmol) in CH3CN (1.5 mL) under N2 was added TEA (1.5 mL) foUowed by the addition of BSA (0.11 mL, 0.44 mmol). After 24 hours, the reaction mixture was concentrated and subjected to Sephadex LH-20 column chromatography [eluent: MeOH/CH2Cl2, 50/50, v/v] to give thiophosphate Compound 48 (75 mg, 92%) as a pale yellow oil.
'H NMR (360 MHz; CDC13): ( 0.87 (3 H, t, J7 Hz, Me), 1.28 (20 H, apparent br d, separation 12.5 Hz, -(CH^- and -(CB^-), 1.60-1.61 (2 H, m, (-Hj), 2.00-2.01 (4 H, m, -CH2CH=CHCH2-), 2.35 (2 H, t, J 7.5 Hz, (-H2), 3.51 (3 H, s, OMe), 3.69 (1 H, br t, 2-H),
4.13-4.31 (4 H, m, 1-H2 and 3-H,), 5.18 (2 H, br s, 2 x OH) and 5.29-5.39 (2 H, m, -CH2CH=CHCH2-); 31PNMR(146 MHz; CDC13): (56.72 ESI-MS (m/z, -ve): 465 ( -H\ 100%); ESI-MS (m/z, +ve): 467 (MH+, 100%).
3-HydroxypropyI oleoyl amide (Compound 51) To a solution of 3-amino-propan-l-ol (3.1 mL, 40 mmol) in dry THF (150 mL) was added oleoyl chloride (4.4 mL, 13 mmol). After 16 hours, the reaction mixture was dUuted with ethyl acetate (100 mL) and washed with sat. NBtCl (2 x 100 mL). The organic extract was dried (MgS04), concentrated and subjected to silica-gel column chromatography [eluent: ethyl acetate (25-100%) in hexane, v/v] to give amide Compound 51 (4.4 g, 95%) as a white soUd. »H NMR (360 MHz; CDC13): ( 0.88 (3 H, t, J 7 Hz, Me), 1.27 (20 H, apparent br d, separation 12.5 Hz, -(CH2)4- and -(CH^-), 1.60-1.69 (4 H, m, 2-H2 and (-Bj), 1.99-2.00 (4 H, m, -Ctf2CH=CHCH2-), 2.19 (2H,t,J7.5Hz, (-H^, 3.41 (2 H, quartet, J 6 Hz, 1-B>), 3.61 (2B, t, J5.5 Hz, 3-H2), 5.29-5.38 (2 H, m, -CH2CH=C#CB2-) and 5.90 (1 B, br s, N-H); ESI-MS (m/z, +ve): 340 (ΛΛT, 100%). Dibenzyl 3-amino-3-N-oleoyl-propyl phosphate (Compound 52) To a mixture of Compound 51 (0.34 g, 1.0 mmol) and 1 tf-tetrazole (0.14 g, 2.0 mmol) in dry CH2C12 (5 mL) under N2 was added dibenzyl NN-diisopropylphosphoramidite (1.1 mL, 3.0 mmol). After 45 min t-BuOOH (2 mL) was added and the mixture was stirred for a subsequent 35 min after which the mixture was concentrated and subjected to silica-gel column chromatography [eluent: ethyl acetate in hexane, 50/50, v/v] to give dibenzyl phosphate Compound 38 (0.27 g, 44%) as an oil.
IHΝMR (360 MHz; CDC13): ( 0.87 (3 H, t,J7 Hz, Me), 1.28 (20 H, br s, -(CH^-and -(CH^-), 1.57 (2 H, br s, (-BJ, 1.77-1.82 (2 H, m, 2-H2), 1.98-2.00 (4 H, m, -CH2CH=CBCi72-), 2.11 (2 H, t, J8 Hz, (-B , 3.29 (2 H, quartet, J6 Hz, 1-Hj), 4.10-4.15 (2 H, t, J5.5 Hz, 3-Bj), 4.99-5.09 (4 H, m, -OCi72Ph), 5.28-5.38 (2 B, m, -CH2CH=CHCH2-), 6.25 (1 H, br s, N-H) and
7.35 (10 H, br s, 2 x -Ph); 31P NMR (146 MHz; CDC13): ( 0.78 ESI-MS (m/z, +ve): 600 (ΛΛT, 100%).
Dimethyl 3-amino-3- V-oleoyl-propyl phosphate (Compound 53) To a mixture of amide Compound 51 (0.30 g, 0.9 mmol) and dimethyl chlorophosphate (0.14 mL, 1.3 mmol) in dry CH2Cl2(5 rnL)underN2wasaddedN-methylimidazole(0.14mL, 1.8 mmol). After 40 hours, TLC analysis showed the reaction to be complete and the mixture was partitioned between ethyl acetate (20 mL) and sat. NaCl (4 mL). The organic phase was dried (MgS04), concentrated and subjected to silica-gel column chromatography [eluent: 5% MeOH in CH2C12, v/v] to give dimethyl phosphate Compound 53 (0.35 g, 87%) as a pale yellow oU. 'H NMR (360 MHz; CDC13): ( 0.87 (3 H, t, J 7 Hz, Me), 1.28 (20 H, apparent br d, separation 11.5 Hz, -(CH2)4- and -(CB^-), 1.61-1.78 (2 H, m, (-H2), 1.88 (2 B, quintet, J6 Hz, 2-B 1.99-2.01 (4 H, m, -CH2CB=CHCH2-), 2.16 (2 H, t, J8 Hz, (-H2), 3.38 (2 H, quartet, J 6 Hz, 3-H^, 3.76 and 3.79 (each 3 H, s, 2 x OMe), 4.10-4.15 (2 H, t,J5.5 Hz, l-BJ, 5.33-5.35 (2 H, m, -CH2C#=C/7CH2-) and 6.25 (1 H, br s, N-H); 31P NMR (146 MHz; CDC13): ( 3.19; ESI-MS (m/z, +ve): 340 (MNa+, 32%) and (Λ/+83, 100).
1-O-Palmitoyl-rαc-glycidoI (Compound 57) To a solution of glycidol (0.45 mL, 6.7 mmol) in dry CH2C12 (50 ml) under N2 at -78°C was added palmitoyl chloride (2.0 mL, 7.3 mmol). After 5 min, pyridine (1.4 mL, 16.8 mmol) was added and the reaction mixture was allowed to stir for 1 hour at -78°C before being allowed to warm to room temperature over a 1.5 hours period. Then, the mixture was (United with CH2C12 (100 mL), washed with sat. KBS04 (10 mL), sat.
NaBC03 (10 mL) and sat. NaCl (20 mL), dried (MgS04) and concentrated to give an oil. The residue was subjected to silica-gel column chromatography [eluent: ethyl acetate/hexane, 10/90, v/v] to give epoxide Compound 57 (1.3 g, 63%) as a colourless oU. Η NMR (360 MHz; CDC13): ( 0.88 (3 H, t, J7 Hz, Me), 1.25 (24 H, br s, 12 x Q J,
1.63 (2 H, quintet, J7.5 Hz, (-BJ, 2.35 (2 H, t, J7.5 Hz, (-HJ, 2.65 (1 H, dd, J5 and 2.5 Hz, 3-H), 2.85 (1 H, t, J4.5 Hz, 3-H), 3.19-3.23 (1 H, m, 2-H), 3.91 (1 H, dd, J 12.5 and 6.5 Hz, 1-H) and 4.42 (1 H, dd, J 12.5 and 3 Hz, 1-H); ESI-MS (m/z, +ve): 335 (MNa+, 100%). 1-0-Palmitoleoyl-rαc-glycidoI (Compound 58) To a solution of glycidol (0.04 mL, 0.8 mmol) in dry CH2C12 (10 ml) under N2 at -78°C was added palmitoleoyl chloride (0.20 g, 0.7 mmol). After 5 min, pyridine (0.15 mL, 1.8 mmol) was added and the reaction mixture was allowed to stir for 1 hour at -78°C before being allowed to warm to room temperature over a 1.5 hours period. Next, the reaction mixture was diluted with CH2C12 (50 mL), washed with sat. KHS04 (5 mL), sat. NaHC03 (5 mL) and sat. NaCl (10 mL), dried (MgS04) and concentrated to give an oil. The residue was purified by silica-gel column chromatography [eluent: ethyl acetate hexane, 10/90, v/v] to furnish epoxide Compound 58 as a colourless oU.
*HNMR(360MHz; CDC13): (0.88 (3 H,t,J6.5 Hz, Me), 1.30 (16H, brs, -(CH^-and
-(CH^-), 1.61-1.65 (2 H, m, (-HJ, 2.00-2.01 (4 H, m, -CH2CH=CHCH2-), 2.35 (2 H, t, J7.5 Hz, (-Hj), 2.65 (1 H, dd, J4.5 and 2.5 Hz, 3-H), 2.85 (1 H, t,J4.5 Hz, 3-H), 3.20-3.22 (1 H, m,
2-H), 3.91 (1 B, dd, J 12.5 and 6.5 Hz, 1-B), 4.41 (1 H, dd, J 12.5 and 2.5 Hz, 1-H) and
5.29-5.39 (2 H, m, -CB2C. =C/7CH2-); ESI-MS (m/z, +ve): 311 (ΛΛT, 30%), 333 (WNa+, 100%).
Dimethyl 2-amino-2-N-oleoyl-ethyI phosphate (Compound 59) To a mixture of amide
Compound 51 (l.Og, 3.1 mmol) and dimethyl chlorophosphate (0.50 mL, 4.6 mmol) in dry CB2C12 (100 mL) under N2 was added N-methylimidazole (0.73 mL, 9.2 mmol). After 40 hours, TLC analysis showed the reaction to be complete and the mixture was washed with sat. NaHC03 (3 x
20 mL), dried (MgS04) and evaporated to dryness to give dimethyl phosphate Compound 59 (1.2 g, 89%) as a pale yellow oil.
Η NMR (360 MHz; CDC13): ( 0.75 (3 B, t, J 6.5 Hz, Me), 1.16 (20 H, apparent br d, separation 11.5 Hz, -(CH2)4- and -(CB^-), 1.50 (2 B, br s, (-HJ, 1.88 (4 B, apparent br d, separation 5.5 Hz, -CH2CH=CHCH2-), 2.08 (2 H, t, J7.5 Hz, (-HJ, 3.34-3.38 (2 H, m, 2-H^,
3.64 and 3.67 (each 3 H, s, 2 x OMe), 3.97-4.02 (2 H, m, 1-H^, 5.16-5.26 (2 H, m, -CH2C#=CHCH2-) and 6.79 (1 H, br s, N-H); 31P NMR(146 Mhz; CDC13): ( 2.49; ESI-MS (m/z, +ve): 434 ( T, 100%). Oi-tert -butyl 2-amino-2-N-oleoyl-propyl phosphate (Compound 60) To a mixture of
Compound 51 (1.0 g, 2.9 mmol) and 1 /7-tetrazole (0.41 g, 5.9 mmol) in dry CB2C12 (10 mL) under N2 was added di-tert. -butyl NN-diisopropylphosphoramidite (1.2 g, 4.4 mmol). After 1.5 hours, t-BuOOB (2 mL) was added and the mixture was stirred for a another 30 min. Next, the mixture was concentrated and subjected to silica-gel column chromatography [eluent: ethyl acetate in hexane, 25/75, v/v] to give di-tert. -butyl phosphate Compound 60 (1.3 g, 81%) as an oil.
'H NMR (360 MHz; CDC13): ( 0.87 (3 B, t, J 7 Hz, Me), 1.27 (20 H, apparent br d, separation 10 Hz, -(CH^- and -(CB^-), 1.49 (18H, s, 2 x BuO), 1.59-1.66 (2 H, m, (-HJ, 1.83 (2 H, quintet, J6 Hz, 2-^, 1.97-2.00 (4 H, m, -C/72CH=CHCH2-), 2.16 (2 H, t, J7.5 Hz, (-H^,
3.39 (2 H, quintet, J 6 Hz, 3-H,), 4.03 (2 H, dt, J 7.5 and 6 Hz, l-BJ, 5.32-5.35 (2 H, m,
Figure imgf000066_0001
CDC13): ( -7.47; ESI-MS (m/z, +ve): 420 ( -TBvtol? , 78%), 476 (AZ-BuM^lT, 30), 532 (ΛΛT, 100).
2-Amino-2- V-oIeoyl-propyl phosphate (Compound 61) A solution of the protected phosphate Compound 60 (250 mg, 0.47 mmol) was treated with 1/4 TFA/CH2C12 (15 mL, v/v) for 3 hours . Next, the reaction mixture was concentrated and the residue was purified by Sephadex LH-20 column chromatography [eluent: MeOH/CH2Cl2, 50/50, v/v] to give phosphate Compound 61 (130 mg, 67%) as a white solid.
'HNMR (360 MHz; CDC13): ( 0.81 (3 H, t, J6.7 Hz, Me), 1.20 (20 H, apparent br d, -(CB^- and -(CB^-), 1.51 (2 H, br s, (-HJ, 1.79 (2H, br s, 2-H2), 1.9 (4 H, m,
Figure imgf000066_0002
2.16 (2 H, br s, (-HJ, 3.3 (2 B, br s, 2-B2), 3.9 (2 H, m, l-HJ, 5.25 (2 H, m, -CH2CH=C#CB2-) and 7.2 (1 B, br s, N-H); 31P NMR(146 Mhz; CDC13): ( 1.10; ESI-MS (m/z, -ve): 418 (M-H 100%); ESI-MS (m/z, +ve): 420 (ΛΛT, 100%). l-0-OleyI-2-0-methyI glycerol (Compound 64) 2-0-Methyl glycerol (1.54 g, 14.5 mmol) was added to a suspension of NaH (0.38 g, 15.9 mmol) in dry DMF (20 mL) under N2.
After 2 min, oleyl bromide (2.4 g, 7.2 mmol) was added and stirring was continued for 0.5 hour. Then, the reaction mixture was quenched with water (20 mL) and extracted with diethyl ether (150 mL). The combined organic phases were dried (MgS04) and concentrated to give an oil which was subjected to sihca-gel column chromatography [eluent: EtOAc/Hexane, 50/50, v/v] to give alcohol Compound 64 (1.3 g, 50%). lH NMR (360 MHz; CDC13): ( 0.88-0.89 (3 H, m, Me), 1.29 (22 H, br s, -(CH^- and
Figure imgf000066_0003
3.44-3.76 (7 H, m, 3-H2, 1-H2, 2-H and (-BJ, 3.47 (3 H, s, OMe), and 5.34 (2 H, m, -CH2CH=CHCH2-); ESI-MS (m/z, +ve): 357 (ΛΛT, 100%), 379 (A Na+, 53). Bis-(2-cyanoethyl) 3-0-oleyl-2-0-methyl-rαc-glycero-l-thiophosphate (Compound 65)
To a solution of 3-hydroxypropionitrile (0.16 mL, 2.4 mmol) and 1 H-tetrazole (0.18 g, 2.5 mmol) in dry CH2C12 (8 mL) under N2 was added 2 -cyanoethyl N.NN'.N'-tetraisopropylphosphorodiamidite (0.71 mL, 2.2 mmol). After stirring for 1 hour, a second portion of 1 /7-tetrazole (0.20 g, 2.8 mmol) was added foUowed by the addition of alcohol Compound 64 (0.50 g, 1.4 mmol). After an additional 30 min, elemental sulfur (0.5 g) and 1/1
Figure imgf000067_0001
(0.5 mL, v/v) were added. After 14 hours, the reaction mixture was filtered through a plug of siUca-gel, concentrated and purified by silica-gel column chromatography [eluent: EtOAc/hexane, 25/75 v/v] to yield protected thiophosphate Compound 65 (0.33 g, 42%) as a pale yellow oU.
'HNMR (360 MHz; CDC13): ( 0.88 (3 H, t, J7 Hz, Me), 1.28 (22 H, m, -(CB^- and -(CB^-), 1.54-1.58 (2 B, m, (-B2), 1.98-2.03 (4 B, m, -CH2CH=CHCH2-), 2.78 (4 H, t, J6 Hz, 2 x NCCH2CH2-), 3.44 (2 H, t, J7 Hz, (-H2), 3.46 (3 H, s, OMe), 3.49-3.51 (2 H, m, 3-^, 3.53-3.58 (1 H, m, 2-B), 4.15 (1 H, dd, J15 and5 Hz, 1-H), 4.23-4.36 (5 H, m, 2 xNCCH2Ci72- and 1-H) and 5.30-5.39 (2 H, m, -CB2Ci7=C Η2-); 31P NMR (146 MHz; CDC13): ( 69.04;
ESI-MS (m/z, +ve): 581 (MSa+, 100%); ESI-MS (m/z, -ve): 504 (Λf-NCCtf2CH2, 100%).
3-0-OIeyl-2-0-methyl-rαc-glycero-l-thiophosphate (Compound 66) To a solution of compound Compound 65 (50 mg, 87 (mol) in CH3CN (0.5 mL) under N2 at 30°C was added TEA (0.5 mL) followed by the addition of BSA (54 (L, 0.22 mmol). After 24 hours, the reaction mixture was concentrated and subjected to Sephadex LB-20 column chromatography [eluent:
MeOH/CB2Cl2, 50/50, v/v] to give a mixture of thiophosphate Compound 66 and mono-deprotected Compound 65 (80 mg) in a 3/2 ratio.
31P NMR (146 MHz; CDC13): ( 59.24, 56.74 (ratio 3/2, respectively); ESI-MS (m/z, -ve): 451 (M-H 80%) and 504 (Λ/+NCCB2CB2-2B+, 100). Bis-(2-cyanoethyl) l-(3-propyI oleoate) thiophosphate (Compound 67) To a solution of 3-hydroxypropionitrile (0.16 mL, 2.4 mmol) and 1 /7-tetrazole (0.19 g, 2.6 mmol) in dry CB2C12 (7 mL) under N2 was added 2-cyanoethylN,NN',N'-tetraisopropylphosphorodiamidite (0.65 mL, 2.1 mmol). After stirring for 1 hour, a second portion of 1 /-tetrazole (0.21 g, 2.9 mmol) was added foUowed by the addition of alcohol Compound 28 (0.50 g, 1.5 mmol). After an additional 30 min, elemental sulfur (0.5 g) and 1/1 CS /pyridine (1 mL, v/v) were added. After 14 hours, the reaction mixture was filtered through a plug of silica-gel, concentrated and purified by sihca-gel column chromatography [eluent: EtOAc/hexane, 25/75 v/v] to yield protected thiophosphate Compound 67 (0.27 g, 34%) as a pale yellow oil.
'H ΝMR (360 MHz; CDC13): ( 0.88 (3 H, t, J6.5 Hz, Me), 1.28 (20 H, apparent br d, separation 13.5 Hz, -(CB^- and -(CB^-), 1.61 (2 B, br s, (-HJ, 2.02-2.05 (6 H, m,2-H2 and
-CH2CH=CHCH2-), 2.31 (2 H, t, J 7.5 Hz, (-HJ, 2.78 (4 H, t, J 6 Hz, 2 x ΝCCH2CB2-), 4.17-4.24 (4 H, m, 1-H2 and 3-B2 or 2 x NCCB2C772-), 4.17-4.24 (4 H, m, 1-H2 and 3-H2 or 2 x NCCB2C#2-), and 5.34 (2 B, br s, -CB2CH=C#CB2-); 31P NMR (146 MHz; CDC13): ( 68.19; ESI-MS (m/z, +ve): 543 (MB, 77%) and 565 (MNa+, 100). l-(3-propyl oleoate) thiophosphate (Compound 68) To a solution of Compound 67 (220 mg, 0.41 mmol) in CH3CN (2 mL) under N2 was added TEA (2 mL) foUowed by the addition of
BSA (0.25 mL, 1.01 mmol). After 66 hours, the reaction mixture was concentrated and subjected to Sephadex LB-20 column chromatography [eluent: MeOB/CB2Cl2, 50/50, v/v] to give thiophosphate Compound 68 (144 mg, 69%) as a pale yellow oil.
Η NMR (360 MHz; CDC13): ( 0.85 (3 H, t, J 7 Hz, Me), 1.25 (20 H, apparent br d, separation 11.5 Hz, -(CB2)4- and -(CB^-), 1.58 (2 B, br s, (-H^, 1.97-2.02 (6 B, m, 2-B2 and -CH2CH=CHCH2-), 2.30 (2 B, t, J7.5 Hz, (-H^, 4.11-4.17 (2 B, m, 1-B2 or 3-B^, 4.21 (2 B, t, J6 Hz, 1-B2 or 3-Hj), 5.31 (2 H, br s, -CH2C//=C Η2-) and 7.26 (2 H, br s, 2 x OB); 31P NMR (146 MHz; CDC13): ( 48.60 ESI-MS (m/z, -ve): 465 (M-B+, 100%); ESI-MS (m/z, +ve):
323 (100%) and 437 (MB, 95).
Oi-ter -butyl 3-0-oleyl-2-0-methyI-r c-glycero-l-thiophosphate (Compound 69) To a mixture of Compound 64 (0.3 g, 0.84 mmol) and 1 flr-tetrazole (0.12 g, 1.7 mmol) in dry CH2C12 (5 mL) under N2 was added di-tert. -butyl NN-dusopropylphosphoramidite (0.34 mL, 1.1 mmol). After 2 hours, elemental sulfur (0.5 g) and 1/1
Figure imgf000068_0001
the mixture was stirred for a another 2 hours after. Then, the mixture was filtered through a plug of silica-gel, concentrated and subjected to silica-gel column chromatography [eluent: ethyl acetate in hexane, 1/15, v/v] to give di-tert. -butyl thiophosphate Compound 69 (0.48 g, quant.) as an oil.
'BNMR (360 MHz; CDC13): ( 0.87 (3 B, t, J7 Hz, Me), 1.26-1.30 (22 H, m, -(CB^- and -(CB^-), 1.53 (20 B, m, 2 x BuO and (-HJ, 1.97-2.01 (4 B, m, -C^2CB=CBCH2-), 3.43
(2 B, t, J7 Hz, (-H2), 3.46 (3 B, s, OMe), 3.48-3.57 (3 H, m, 3-B2and 2-B), 4.01-4.14 (2 B, m,
1-B2) and 5.34 (2 H, apparent t, separation 5 Hz, -CH2C/7=C#CB2-); 31P NMR (146 MHz;
CDC13): ( 52.39; ESI-MS (m/z, -ve): 507 (M-Bu 100%); ESI-MS (m/z, +ve): 589 (MNa+, 100%).
Dimethyl erucyl phosphate (Compound 70) To a solution of erucyl alcohol (0.50 g, 1.5 rnmol)mdryCH2Cl2 (10nΛ)wasaddedN-methyliπιidazole(0.14mL, 1.7 mmol) followed by the addition of dimethyl chlorophosphate (0.20 mL, 1.9 mmol). After 20 hours, the reaction mixture was concentrated and subjected to silica-gel column chromatography [eluent: hexane/ethyl acetate,
66/33, v/v] to give dimethyl phosphate Compound 70 (0.59 g, 89%) as an oil.
*BNMR (360 MBz; CDC13): ( 0.88 (3 B, t, J7 Hz, Me), 1.26 (30 H, br s, -(CB^- and
Figure imgf000068_0002
2.01 (4 B, apparent br d, separation 5 Hz,
-C/72CH=CBC#2-), 3.75 and 3.78 (each 3 B, s, 2 x OMe), 4.04 (2 B, quartet, J7 Bz, (-B^, and
5.30-5.39 (2 B, m, -CB2CH=CHCB2-); 31P NMR(146 Mhz; CDC13): ( 2.65; ESI-MS (m/z, +ve):
433 (MB, 100). Erucyl phosphate (Compound 71) To a solution of protected phosphate Compound 70 (150 mg, 0.35 mmol) in dry CB2C12 (6 mL) was added BSA (0.19 mL, 0.76 mmol) followed by the addition of TMSBr (0.10 mL, 0.76 mmol). After 30 min, TLC analysis showed complete consumption of the starting material and the reaction was quenched with 1/1 MeOH/H20 (2 mL, v/v) for 15 min, foUowed by the addition of sat. KHS04 (5 mL). Subsequently, the reaction mixture was extracted with ethyl acetate (2 x 30 mL), the organic extracts were combined, dried (MgS04) and concentrated to give an oil which was subjected to Sephadex LH-20 column chromatography [eluent: MeOH/CH2Cl2, 50/50, v/v] to give phosphate Compound 71 (90 mg, 64%) as an oU. 'BNMR (360 MHz; CDC13): ( 0.88 (3 B, t, J7 Hz, Me), 1.26 (30 H, br s, - CB^- and
-(CB^-), 1.66 (2 B, quintet, J6.5 Bz (-B2), 1.98-2.03 (4 B, m, -C#2CB=CBCH2-), 3.94-4.00 (2 H, m, (-B2), 5.30-5.39 (2 B, m, -CB2CH=C/7CB2-) and 7.32 (2 H, br s, 2 x OH); 3,P NMR(146 Mhz; CDC13): ( 2.59; ESI-MS ( /z, -ve): 403 (M-ff, 100).
Oleyl 2-O-methyl-rαc-glycerate (Compound 76) To a solution of oxalyl chloride (87 (L, 1.0 mmol) in CB2C12 (7 mL) at -78°C was added DMSO (71 (L, 1.0 mmol). After 20 min, alcohol
Compound 42 (0.20 g, 0.91 mmol) was added followed, after another 20 min, by the addition of triethylamine (0.63 mL, 4.6 mmol). The reaction was warmed to room temperature over a 10 min period, filtered through a plug of silica-gel, which was washed with ethyl acetate (50 mL) and concentrated to give 3-0-t-butyld methylsUyl-2-6)-methyl-roc-glyceraldehyde Compound 73 (0.20 g, 100%) as a colourless oil.
*B NMR (360 MHz; CDC13): ( 0.07 (6 H, s, Me^i), 0.88 (9 B, s, Bu'Si), 3.51 (3 B, s, OMe), 3.67-3.68 (1 H, m, 2-H), 3.92 (2 H, br d, J6.5 Hz, 3-Bj), 9.72 (1 B, s, CBO).
To a vigorously stirring mixture of CB2C12 (20 mL), water (20 mL) and aldehyde Compound 73 (0.93 g, 4.3 mmol) at 0°C was added sulfamic acid (0.63 g, 6.8 mmol) and 2-methyl-2-butene (0.60 g, 8.5 mmol). After 5 min, sodium chlorite (0.77 g, 8.5 mmol) was added and the reaction was monitored by TLC until no starting material remained. The reaction mixture was extracted with CH2C12 (2 x 20 mL), and the combined organic layers were washed with sat. NaCl (2 x 20 mL), dried (MgS04) and concentrated to give a colourless oil. The oil was redissolved in CB2C12 (50 mL) after which sat. NaBC03 (10 mL) was added. The aqueous phase was isolated, CH2C12 (50 mL) was added and the mixture was acidified by adding 10% aqueous acetic acid (v/v). The organic phase was dried (MgS04) and concentrated to give 3-0-t-butyldimethylsUyl-2-0-methyl-rac-glyceric acid Compound 74 (0.39 g, 38%) as a colourless oil. 'B NMR (360 MHz; CDC13): ( 0.07 and 0.07 (each 3 B, s, Me^i), 0.88 (9 B, s, Bu'Si), 3.52 (3 B, s, OMe) and 3.87-3.99 (3 B m, 2-B and 3-B2).
To a mixture of oleyl alcohol (0.52 mL, 1.6 mmol), DIC (0.27 mL, 1.7 mmol) and glyceric acid Compound 74 (0.39 g, 1.6 mmol) in DMF (10 mL) was added DMAP (-20 mg). After 16 hours, the solvent was removed in vacuo and the residue was partitioned between EtOAc (50 mL) and water (10 mL). The organic phase was dried (MgS04) and concentrated to give an oU which was purified by silica-gel column chromatography [eluent: EtOAc/Hexane, 10/90, v/v] to give oleyl 3-0-t-butyldimethylsUyl-2-0-methyl-rac-glycerate Compound 75 (0.33 g, 41%) as a colourless oil. 'HNMR (360 MHz; CDC13): ( 0.05 (6 H, s, Me^i), 0.86-0.89 (12 B, m, Bu'Si and Me),
1.28 (22 B, apparent br d, separation 10.5 Hz, -(CB^- and -(CB^-), 1.61-1.67 (2 B, m, (-B^, 1.98-2.04 (4 B, m, -072CB=CHCH2-), 3.44 (3 H, s, OMe), 3.83-3.90 (3 B, m, 1-B and 2-B^, 4.15 (2 B, t, J7 Hz, (-H^, and 5.33-5.39 (2 B, m, -CB2Ctf=C/7CB2-).
1M TBAF (0.7 mL, 0.7 mmol) was added to a solution of Compound 75 in THF (8 mL). After 40 min, TLC analysis showed that aU the starting material had been consumed. Then, the reaction mixture was partitioned between diethyl ether (100 mL) and sat. NaCl solution (15 mL). The organic extract was dried (MgS04) and concentrated to give a yeUow oil which was subjected to siUca-gel column chromatography [eluent: hexane/ethyl acetate, 80/20, v/v] to give the title ester (Compound 76) (0.18 g, 71%). lH NMR (360 MHz; CDC13): ( 0.87 (3 B, t, J 7 Bz, Me), 1.28 (22 B, apparent br d, separation 11 Hz, -(CB^- and -(CB^-), 1.65 (2 B, quintet, J7 Hz, (-HJ, 2.01 (4 B, quintet, J 6.5 Hz, -CH2CH=CHCH2-), 2.24 (1 B, m, OB), 3.49 (3 B, s, OMe), 3.75-3.81 (1 H, m, 2-^,3.89 (2 H, quintet, J3.5 Hz, 3-H2 or (-B^, 4.11-4.22 (2 B, m, 3-B2 or (-HJ and 5.29-5.40 (2 B, m, -CB2CH=C#CB2-); ESI-MS (m/z, +ve): 393 (MNa+, 100), 763 (2MNa+, 20). Oleyl l-(bis-(2-cyanoethyl) thiophosphoryl)-2-0-methyl-rαc-glycerate (Compound 77)
To a solution of 3-hydroxypropionitrile (55 (L, 0.80 mmol) and 1 /y-tetrazole (60 mg, 0.85 mmol) in dry CB2C12 (4 mL) under N2 was added 2-cyanoethyl N,N,N',N'- tetraisopropylphosphorodiamidite (0.24 mL, 0.76 mmol). After stirring for 1 hour, a second portion of 1 H-tetrazole (66 mg, 0.94 mmol) was added followed by the addition of ester Compound 76 (175 mg, 0.47 mmol). After 1 hour, elemental sulfur (0.25 g) and 1/1 CSVpyridine
(0.5 mL, v/v) were added. After an additional 2 hours, the reaction mixture was filtered through a plug of sihca-gel, concentrated and purified by silica-gel column chromatography [eluent: EtOAc/hexane, 25/75 v/v] to yield protected thiophosphate 77 (78 mg, 29%) as a pale yellow oU and recovered ester Compound 76 (73 mg, 42%). XH NMR (360 MBz; CDC13): ( 0.86 (3 B, t, J 7 Bz, Me), 1.27 (22 B, apparent br d, separation 12.5 Bz, -(CB^- and -(CB^-), 1.65 (2 B, quintet, (-B^, 1.98-2.02 (4 B, m,
Figure imgf000071_0001
2.76 and 2.78 ( each 2 B t, J6.5 Hz, 2 xNCCi 2CH2-), 3.48 (3 B, s, OMe), 3.99 (IB, t, J 4.2 Hz, 2-H), 4.17 (2 B, t, J 7 Hz, (-H^, 4.22-4.47 (6 H, m, 3-B2 and 2 x NCCB2072-) and 5.28-5.37 (2H,m, -CB2C#=C#CB2-); 31P NMR (146 MHz; CDC13): ( 69.04;
ESI-MS (m/z, +ve): 573 (M-B+, 25%), 595 (MNa+, 100).
Oleyl l-thiophosphoryl-2-0-methyl-rac-glycerate (Compound 78). To a solution of Compound 77 (100 mg, 0.18 mmol) in CB3CN (0.5 mL) under N2 at 30°C was added TEA (0.5 mL) foUowed by the addition of BSA (0.11 mL, 0.45 mmol). After 24 hours, the reaction mixture was concentrated and subjected to Sephadex LB-20 column chromatography [eluent:
MeOH/CH2Cl2, 50/50, v/v] to give thiophosphate Compound 78 (40mg, quant.) as a pale yeUow oU.
'HNMR (360 MHz; CDC13): ( 0.87 (3 H, t, J7 Bz, Me), 1.24-1.35 (22 B, m, -(CBJ and -(CB^-), 1.65 (2 B, quintet, (-B^, 1.98-2.03 (4 B, m, - y2CB=CBCJy2-), 3.47 (3 H, s, OMe), 4.09 (IB, dd, J5 and 3.5 Hz, 2-H), 4.09-4.38 (4 B, m, 1-B2 and (-B^, 5.32-5.38 (2 B, m, -CH2CH=CHCH2-), 5.51 (2 B, br s, 2 x OB); 31P NMR (146 MHz; CDC13): ( 55.28; ESI-MS (m/z, -ve): 465 (M-B+, 100%).
3-0-Oleoyl-2-0-methyl-rαc-grycero-l-sulfate (Compound 81) To a solution of alcohol Compound 44 (0.30 g, 0.8 mmol) in DMF (10 mL) was added triethylamine sulfur trioxide complex (0.74 g, 4.1 mmol) and the mixture was stirred for 5.5 h at 40°C. Next, NaBC03 (0.3 g, 3.6 mmol) was added and ,after stirring for another 30 min, the mixture was filtered through a glass sinter. The filtrate was concentrated to give a light brown oil which was subjected to Sephadex LB-20 column chromatography [eluent: MeOH/CH2Cl2, 50/50, v/v] to furnish, after extensive drying in vacuo, sulfate Compound 81 (0.52 g) as its triethylamine salt. Η NMR (360 MHz; CDC13): ( 0.87 (3 B, t, J 7 Hz, Me), 1.27 (20 H, apparent br d, separation J 11 Hz, -(CHj),- and -(CB^-), 1.46 (15 B, t, (C/^CB^N, J7 Hz), 1.59 (2 H, m, (-HJ, 1.97-2.02 (4 B, m, -C/72CB=CBCiy2-), 2.31 (2 B, t, J7.6 Hz, (-HJ, 3.21-3.13 (10 B, m, (CH3CH2)3K), 3.45 (3 B, s, OMe), 3.69-3.74 (1 H, m, 2-H), 4.06-4.29 (4 H, m, 1-B2 and 3-Bj) and 5.28-5.37 (2 B, m, -CB2C77=CHCB2-); 31P NMR (146 MHz; CDC13): ( 63.69 (m); ESI-MS (m/z, +ve): 656 (MH++2TEA, 100%); ESI-MS (m/z, -ve): 449 (M-H*, 100%).
Bis(3-0-oleyl-2-0-methyl-rαc-glyceryl) thiophosphate (Compound 80) To a solution of 1 / -tetrazole (0.31 g, 4.0 mmol) and 2-cyanoemylNNN',N'-tetraisopropylphosphorodiamidite (0.53 mL, 1.7 mmol) in dry CH2C12 (5 mL) under N2 was added alcohol Compound 64 (0.38 g, 1.1 mmol). After stirring for 30 min, elemental sulfur (0.5 g) and 1/1 CS2/pyridine (0.5 mL, v/v) were added. After 2 h, the reaction mixture was filtered through a plug of silica-gel, concentrated and purified by sihca-gel column chromatography [eluent: EtOAc/hexane, 25/75 v/v] to yield 2-cyanoethyl bis(3-0-oleyl-2-0-methyl-rαc-glyceryl) thiophosphate (Compound 79) (0.15 g, 34%) as a pale yeUow oU. To a solution of compound 79 (0.15 g, 0.18 mmol) in CB3CN (1.5 mL) under N2 was added TEA (1.5 mL) foUowed by the addition of BSA (0.17 mL, 0.68 mmol). After 60 h, the reaction mixture was concentrated and subjected to Sephadex LB-20 column chromatography [eluent: MeOH CH2Cl2, 50/50, v/v] to give thiophosphate Compound 80 (0.12 g, 81%).
Η NMR (360 MHz; CDC13): ( 0.87 (6 B, t,J7 Hz, 2 x Me), 1.26 (44 H, apparent br d, separationJ6 Hz, 2 x -(CB^- and 2 x -(CB^-), 1.55 (4 B, m, 2 x (-HJ, 1.97-2.00 (8 B, m, 2 x
Figure imgf000072_0001
3.47 (6 B, 2 x s, 2 x OMe), 3.49-3.59 (6 B, m, 2 x 2-B and 2 x 3-Bj), 3.99^.20 (4 H, m, 2 x 1-B2) 5.33 (4 B, m, 2 x -CB2C#=CHCB2-) and 5.68 (2 B, br s, 2 x OB); 31P NMR (146 MHz; CDC13): ( 63.69 (m); ESI-MS (m/z, +ve): 792 (MB+, 100%); ESI-MS (m/z, -ve): 790 (M-B+, 100%).
Example 2 Anti-apoptotic Activity Assay In order to determine the apoptotic activity of the claimed invention, the following method of analysis was used. The cell assay is described in detail in U.S. Patent Nos. 5,637,486, 5,620,888, and 5,681,703, and Tomei et al. (1993) Proc Natl. Acad. Sci. 90:853-857. Briefly, mouse fibroblast C3B/10T1/2 cells (clone 8) were obtained from ATCC (RockviUe, MD) and were maintained in exponential growth phase in which the cell cycle is randomly distributed and no cells are arrested in Go, and in quiescence. Exponential growth phase was assured by seeding at 2000 cells per 1 ml (5 ml for a 60 mm culture plate) five days prior to the beginning of the experiment. Assays were performed on cells only up to passage 15. At T=0, cultures were transferred to serum-free medium, as an apoptosis stimulus, and seed extracts were added. Controls included 10"7 and 5 x 10"* M 12-0-tetradecanoyl phorbol-13-acetate (TPA) to ensure the responsiveness of the cell culture. The samples were added to serum free medium and sterile filtered prior to addition to the cultures. Assays were performed in tripUcate or quadrupUcate. Analyses of the cell responses were made between 18 and 28 hours of serum deprivation. Two assays were performed on each ceU culture plate consisting of differential cell counts.
1. AU non-adherent or loosely adherent cells were removed from the culture dish and counted by appropriate techniques, typically counting by electronic particle counting instrument. These are the apoptotic cells, the serum deprived released cells (SDR), released by the action of cultivation in serum-free medium. Approximately 95% of these released cells are apoptotic as shown by both ultrastructure analysis and DNA fragmentation analysis.
2. The remaining adherent cells (ADH) were exposed to a buffered, typicaUy pB_7.3, balanced salt solution such as Hanks Balanced Salt Solution without calcium and magnesium salts containing 0.05% trypsin and 0.53 mM ethylene cUaminetetraacetic acid (EDTA). Each culture was incubated at either room temperature or 37(C on a rocking platform to ensure uniform distribution of the trypsin reagent over the culture surface. After a standardized period of time, typicaUy 10 minutes, the released cells were removed from each culture dish and measured by the same means as described above, typically electronic particle counting. This ADH cell count is comprised of both trypsin resistant and trypsin sensitive ceUs as described in U.S. Patent Nos.
5,637,486, 5,620,888, and 5,681,703.
Anti-apoptotic activity is expressed in the following examples as the calculated concentration of material ((g/ml of media) required to save 50% of the ceUs released on serum free treatment.
Example 3 Preparation of Five PhosphoUpid Mixture Commercially available purified soy phosphoUpids containing lysophosphatidic acid and the following other phospholipids: PA, PI, LPI, LPC (available, for example, from Avanti® Polar Lipids, Inc.) were suspended in 50 mM ammomum bicarbonate pH 8.0 containing 154 mM NaCl or buffered aqueous solutions free of divalent cations having a pB range of 5 to 8. Total concentrations of phospholipids of greater than 10 mg/mL can be used provided that clarity is obtainable upon sonication. Total concentrations of up to about 50 mg/mL have been utilized. TypicaUy, the phosphoUpid mixtures are suspended in a buffer and the mixture is placed in a disposable borosilicate glass, preferably 1-2 mL in a 16 x 100 mm tube or 0.5-2 mL in a 13 x 100 mm tube, or up to 1 mL in a 12 x 75 mm tube. The combination of phosphoUpids is then sonicated. Preferably, a small bath sonicator is used, such as a that sold by Laboratory SuppUes, HicksviUe, NY. The temperature of the water bath is between about 21 and 50°C, preferably between about 21 °C and about 40°C. The optimal temperature depends on the phospholipids used and can be determined empirically. The water level is adjusted so that it is approximately the same height as the phosphoUpid mixture in the glass tube(s). Alternatively, a probe sonicator can be used (Fisher Scientific Sonic Dismembrator model 550), as long as care is taken to prevent overheating of the mixture. The mixture was sonicated for between 3 and 90 minutes, with alternating 5 minute intervals of sonication foUowed by 5 minutes of thermal equilibration, in a 1-2 ml volume until the mixture became translucent and passed readily through a filter attached to a 5 ml syringe with a pore size of 0.22 (m. Preferably, sonication is for 5-10 minutes.
The stability of the compositions at various temperature was determined. The compositions were stored for one week at 4°C, room temperature, and 65 °C . The results show loss of activity after storage at 65°C, while the compositions stored at 4°C or at room temperature do not have a significant loss of activity.
Optimization of each constituent phospholipid was determined by mixing the purified phosphoUpids in various ratios, varying one phosphoUpid at a time. Each mixture was analyzed for anti-apoptotic activity as described in Example 2. When the apparent optimized ratio was obtained, the ratio of the most active ingredient was varied to find the absolute optimized activity. Table 2 shows the final ratios tested (10: 10:8:2:4 is the "Five PhosphoUpid Mixture" referred to herein).
TABLE 2
Figure imgf000074_0001
The concentration of LPA was varied as was the chain length to determine the effects of these parameters on activity.
Example 4
Anti-apoptotic activity of LPA The ability of 18: 1-LPA and other LPAs to protect serum-starved cells from apoptotic death was measured using the C3H/10T1/2 cell assay, performed as described in Example 2. The effect of various compounds on the ability of lysophosphatidic acid to protect serum-starved cells from apoptotic death was also measured. Log phase cells were seeded in 60 mm Perri dishes at 175-350 ceUs per cm2 and maintained in Basal Medium Eagle (BME) supplemented with 10% Beat Inactivated Fetal Bovine Serum (HIFBS). On day 3 the cells were given fresh media. Treatment began on day 5 when the media containing serum was removed and replaced with the LPA mixture to be tested. After 24 hours of treatment, day 6, serum deprived released (SDR) cells (the apoptotic cell population) and adherent (ADH) cells were separated and counted using an electronic ceU counter (Coulter Corporation, Hialea, FL). SDR ceUs were counted with the lower threshold setting at 6.3 (m and were defined as the apoptotic cells dying in response to cultivation in serum free media. Approximately 95% of the SDR cells were confirmed to be apoptotic as previously shown by size, ultrastructure and DNA fragmentation analysis. Adherent cells were remove by treatment with 5 mL of Hanks Balanced Salt Solution (HBSS) without ions containing 0.05%trypsin and 0.53 mMEDTAand were counted with the lower threshold setting at 11.01 (m. AU samples were tested in tripUcate and serum-deprived controls (BME only) were assayed at both the beginning and end of each experiment.
To test the efficacy of each LPA to inhibit apoptosis or preserve function of serum-deprived cells, 2.5 ( Mol of dried LPA was dissolved (via approximately 5 minute sonication) in 1 mL of citrate-saline (10 mMNa Citrate, 110mMNaCl,pH6.5)togivea2.5 M stock solution and presented to the ceUs at four concentrations: 1, 3, 10 and 30 (M LPA. The results are shown in Fig. 1 (lysophosphatidic acid) and Table 3. The effects of LPA alone or in combination with various proteins or Uposomes are expressed as percent cells saved.
TABLE 3
Figure imgf000075_0001
10
15
20
25
Figure imgf000076_0001
Figure imgf000077_0001
For those compounds exhibiting toxicity, the dose shown is the lowest concentration at which cytotoxic effects were observed.
In medium alone, approximately 80% of serum-deprived 10T1/2 cells were non adherent following 24 hour serum deprivation. Bowever, 18: 1-LPA at various concentrations (1, 3, 10, 30
(M) showed anti-apoptotic activity, protecting 35% to approximately 53% of the cells. 18:1-LPA combined with polyethylene glycol or BSA also protects serum-deprived 10T1/2 ceUs from apoptosis.
When 18: 1-LPA (at the same various concentrations) was combined with BME (containing calcium) and filtered prior to presentation to serum-deprived 10T1/2 cells, the anti-apoptotic activity of 18: 1-LPA appeared to be inhibited. The addition of BSA apparently preserved the anti-apoptotic activity of 18: 1-LPA despite the presence of calcium, as seen when 18: 1-LPA (at the same various concentrations) was combined with BSA in BME (containing calcium) and presented to serum-deprived 10T1/2 cells.
To test the efficacy of LPA, presented in a Upid membrane structure, to inhibit apoptosis in serum-deprived cells, LPA was incorporated into Upid membrane structures of various Upid compositions . Except in the Five PhosphoUpid Mixture treatments, LPA was presented to the cells as 18: 1-LPA, and was tested in all preparations at four concentrations: 0.25, 0.75, 2.25 and 6.75
(g/ml. The results are shown in Fig. 2. In medium alone, approximately 50% of the cells died of serum deprivation-induced apoptosis. Phosphatidylserine (PS) alone was slightly toxic. When
18: 1-LPA at 0.75 (g/ml was added together with PS, approximately 75% of ceUs were protected.
Positively charged particles containing 18: 1-LPA combined with the neutral phosphoUpid phosphatidyl choline (PC) and 20 mol% of the positively charged lipid l,2-dioleoyl-3-trimethylammonium-propane (TAP) were efficacious in protecting 70 to 80 % of ceUs, compared with PC/TAP alone, offered no protection over control. Incorporation of
18:1 -LPA into the negatively charged phosphatidyl glycerol (PG) gave even better protection, with up to 90% of ceUs protected as compared to alone which offered no protection over control. Bowever, inclusion in the 18:1-LPA/PG particles of 5 to 10 mol% of the neutral phosphoUpid phosphatidylethanolamine (PE) with a long polyethylene glycol (2000 MW) covalently linked to the polar head group resulted in reduced efficacy of 18 : 1 -LPA. Taken together, these data suggest that LPA, presented in particles as a 10% by weight mixture with either PC or PG, afforded a degree of protection similar to that achieved with Five PhosphoUpid Mixture against serum deprivation-induced apoptosis.
LPA was then tested in two different weight ratios with PG, using 18: 1-LPA in concentrations representing LPA equivalents to Five PhosphoUpid Mixture at 1, 3, or 10 (g/ml. As shown in Fig. 3, LPA equivalent to 10 (g/ml Five PhosphoUpid Mixture incorporated into particles at 10% by weight with PG protected approximately 65% of ceUs from apoptosis. This degree of protection was roughly the same as that afforded by Five Phospholipid Mixture alone.
Increasing the weight percent of 18 : 1-LPA to 23.5 resulted in a reduction of the proportion of ceUs saved to about 50%. Incorporation of 18: 1-LPA into particles containing 1:1 ratio PG:PC with 23.5 % by weight 18: 1-LPA gave particles that protected about 57% of ceUs. In this assay, in medium alone, only about 30% of ceUs survived serum-deprivation, and when PC or PG/PC mixtures alone were added to the culture medium, only about 25% of cells survived.
Various other natural lysophosphoUpids were tested for ability to protect ceUs from serum deprivation-induced ceU death. Of the phosphoUpids tested, only lysophosphatidyl serine (LPS) showed any significant activity in the C3B10T1/2 assay. On a molar basis, LPS was approximately 50% as active as 18: 1-LPA (Fig. 4). Pa mitoyl-LPA (16:0 LPA) at 15 (M protected 63 % of ceUs, which was similar to the protection afforded by 15 (M 18: 1-LPA.
Bowever, differences were seen at lower concentrations of the two LPAs: 5 (M 18: 1-LPA protected about 62% of cells, while 5 (M palmitoyl-LPA protected only about 42% of ceUs, compared with a medium alone control of 30% protection (Fig. 5). Siimlarly, stearyl-LPA at 15 (M and at 5 (M protected 65 and 63% of cells, respectively, whUe 18:1 LPA at 15 (M and 5 (M protected 80% and 75%, respectively (Fig. 6). Stearyl-LPA and 16: 1-LPA as 10% dispersions in PG gave similar degrees of protection.
The efficacy of various mixtures of oleoyl-lysophosphatidic acid and phosphatidic acid (PA):phosphatidyl inositol (PI) was also tested, oleoyl-lysophosphatidic acid was mixed at 10, 20 and 30% by weight with PA:PI at a 1:1 (by weight) ratio and tested in the C3B10T1/2 serum deprivation assay. The results (Fig. 7) indicate that oleoyl-lysophosphatidic acid presented to
C3B10T1/2 ceUs at the time of serum deprivation as a 30% (by weight) mixture with 1:1 PA:PI in a final concentration equivalent to Five Phospholipid Mixture at 10 (g/ml afforded nearly the same degree of protection (about 82% of cells protected) as Five Phospholipid Mixture at 10 (g/ml (about 87% of ceUs protected), compared to a medium alone control value of 38%. 18: 1-LPA was presented to C3BT101/2 ceUs at the time of serum deprivation as a 10% (by weight) mixture with PC, PC containing 5 mol% phosphatidyl ethanolamine-N-polyethylene glycol (PE-PEG), or PC containing 20 mol% l,2-di-oleoyl-jn-glycero-3-ethylphosphochoUne (EtPC). The results, in Fig. 8, show that preparations containing LPA incorporated into PC, PC/PE-PEG, or PC/EtPC were aU effective in protecting ceUs from serum deprivation-induced ceU death, with 18: 1-LPA/PC/EtPC Uposomes providing as much protection (approximately 85% of cells protected) as Five PhosphoUpid Mixture.
The analogs and derivatives of lysophosphatidic acid that were tested (Table 3) generaUy showed activity similar to lysophosphatidic acid. One genus of LPAs, the phosphothionate analogs, (exemplified by the foUowing compositions: 3-0-01eoyl-2-0-methyl-rαc-glycero-l- thiophosphate, Oleyl l-thiophosphoryl-2-O-methyl-rac-glycerate, and3-0-01eyl-2-0-methyl-rac- glycero-1 -thiophosphate), showed activity significantly higher than lysophosphatidic acid.
Example 5 Preparation of Protein/LPA mixtures
Soy derived LPA (soy-LPA) (Avanti® Polar Lipids, Inc., Alabaster, AL) was stored in an organic solution and was dried immediately prior to assay using a Speed Vac (Savant Instruments, HicksvUle, NY). Tested proteins were resuspended at lOjmg/mL in a bicarbonate buffered saline EDTA solution (50_mM NH,BC03, 104 mM NaCl, 250_(M EDTA) prior to being added to dried soy-LPA at a 1 : 10_soy-LPA to protein (weightweight) ratio. The mixture was then sonicated for 5 minutes using a high power 80 Watt sonication bath (Laboratory SuppUes Co., Hicksville, NY). Following sonication, concentrated LP A/protein mixtures were left standing for 15 minutes at room temperature prior to dilution in cell culture assay media.
Example 6
Effects of protein on anti-apoptotic activity of LPA The effect of various proteins on the abiUty of LPA to protect serum-starved cells from apoptotic death was measured using the C3H 10T1/2 ceU assay, performed as described in Example 2, above. Log phase cells were seeded in 60 mm Petri dishes at 175-350 cells per cm2 and maintained in Basal Medium Eagle (BME) supplemented with 10% fetal calf serum (Hyclone,
Logan, UT). At time zero, serum containing media was replaced with sterile filtered BME containing the LP A/protein mixture to be tested. After 24 hours of treatment, serum deprived release (SDR) cells (the apoptotic cell population) and adherent (ADH) cells were separated and counted using an electronic cell counter (Coulter Corporation, Hialea, FL). SDR ceUs were counted with the lower threshold setting at 6.3_(m and were defined as the apoptotic ceUs dying in response to cultivation in serum-free-media. Approximately 95% of the SDR cells are confirmed to be apoptotic as previously shown by size, ultrastructure and DNA fragmentation analysis. Adherent cells were removed by treatment with a Hanks' balanced salt solution containing 0.05% trypsin and 0.53_mM EDTA and were counted with the lower threshold setting at 11.0 l_(m. AU samples were tested in tripUcate and serum-deprived controls (BME only) were assayed at both the beginning and end of each experiment.
The results are shown in Table_4. The effects of various proteins alone or in combination with LPA are expressed as percent ceUs saved from apoptosis. When C3H10T1/2 cells were treated with 10 (g/mL soy LPA without protein, 8.0% of the ceUs were saved. The degree of protection from apoptosis afforded by protein alone, added to the medium at a concentration of 0.1% (wt/vol.), varied from 0.9% to 27.7 % of cells saved from apoptosis. An unusually high percentage of cells were protected from serum-starvation-induced apoptosis by casein. This value may be artificially high, as casein preparations have been known to contain growth factors, including insulin-like growth factors (IGF) IGF 1 and IGF2. IGF2 is a mitogen that can mask the serum deprivation-induced apoptotic response in these ceUs.
When soy-LPA was added together with protein, the degree of protection from apoptosis was, for certain proteins, more than additive. Thus, fatty acid depleted (FAD) bovine serum albumin (BSA) alone protected 0.9% of cells from serum-induced apoptosis, soy-LPA alone protected 8.0% of cells, whϋe FAD BSA plus soy-LPA saved 58.7% of the cells. Similar effects were seen for mixtures of soy-LPA with FAD soy protein, cytochrome c and low density Upoprotein (LDL) and, to a lesser degree, acyl carrier protein, casein and myoglobin. Soybean trypsin inhibitor, ovalbumin, retinol binding protein and myelin basic protein did not enhance the anti-apoptotic activity of LPA, while bromolain was toxic to the ceUs.
TABLE 4
Figure imgf000080_0001
Figure imgf000081_0001
Example 7 Binding of LPA to protein
Binding of LPA to protein was examined using an electrophoresis radiobinding detection assay and a size shift analysis assay.
Electrophoresis radiobinding assay. Proteins were analyzed on native polyacrylamide gels foUowing incubation with 3B-labeled 18: 1-LPA. 3B-labeled lysophosphatidic acid (1 -oleyl) (DuPont NEN Products, Boston, MA) was added to tested proteins at 1 nM labeled 18:l-LPA/45_(g protein. Mixtures were incubated in a bicarbonate buffered saline/EDTA solution at 37°C for 60 minutes. Incubated samples were mixed with native gel sample buffer (312_mM Tris pB 6.8, 50% glycerol, 0.05% bromophenol blue) and loaded entirely onto an 8% discontinuous native polyacrylamide gel. Following electrophoresis, proteins were fixed into the gels using an aqueous solution consisting of glacial acetic acid, 10% (vol/vol) and methanol 30% (vol./vol.) solution. Fixed gels were then incubated for 60 minutes in autoradiography enhancer (EN3HANCE, DuPontNEN Products, Boston, MA), and dried onto 3M chromatography paper (Whatman, Clifton, NJ) and exposed to autoradiography film (X-Omat, Kodak, Rochester, NY) for 72 hours at -80°C.
The results, shown in Fig. 9, demonstrate that BSA and (-fetoprotein bind to 18: 1-LPA, whereas bromolain and acyl carrier protein did not yield detectable bands. In other experiments, ovalbumin was shown to produce a detectable band (see table_4). Other experiments indicate that FAD soy protein can bind LPA.
Size shift assay. Size analysis of LPA both with and without protein was performed using a Superdex® S75 column (Pharmacia Biotech, Uppsala, Sweden). Soy LPA was combined in a chloroform solution with 0.5 Ci3B-labeled 18:l-LPA(DuPontNENProducts,Boston,MA),dried under vacuum and resuspended by sonication in a buffered solution (50 mM ammomum bicarbonate, 154 mM sodium chloride, pB 8.0) in the presence or absence of test protein. The ammonium bicarbonate/sodium chloride solution was used as the mobile phase at a flow rate of 1 ml/min. One-minute fractions were collected for 30 minutes following sample injection. Fractions were counted in scintiUant solution using a Uquid scintillation counter (Beckman
Instruments, Irvine, CA). Calibration was performed using a standard solution containing purified proteins (BioRad, Bercules, CA).
As shown in Figs. 10A, 10B, 10C and 10D, soy-LPA alone eluted at 22-26 minutes after sample injection, corresponding to a size of approximately 1.3 kD. When a LPA/BSA mixture was appUed to the column, radioactivity eluted primarily in the 10 to 12-minute fractions, indicating that most of the LPA was bound to BSA. A LP A/FAD soy protein mixture appUed to the column resulted in an elution profile with two peaks, one corresponding to the position of unbound LPA, and the other shifted. A cytochrome c/LPA mixture gave an elution profile similar to that of soy-LPA alone, indicating that LPA does not bind to this protein.
Example 8
Anti-apoptotic activity of Five PhosphoUpid Mixture in an Organ Preservation Solution
In order to determine the apoptotic activity of the claimed invention, the foUowing experiment was performed. The cell assay is described in Example 2. Exponential growth phase was assured by seeding at 2000 cells per 1 ml (5 ml for a 60 mm culture plate) five days prior to the beginning of the experiment. At T=0, cultures were transferred to serum-free medium, as an apoptosis stimulus, and seed extracts were added. Controls included 10"7 and 5_x 10"8 M
12-0-tetradecanoyl phorbol- 13 -acetate (TPA) to ensure the responsiveness of the cell culture. The samples were added to serum free medium and sterile filtered prior to addition to the cultures. Assays were performed in triplicate or quadrupUcate. Analyses of the cell responses were made between 18 and 28 hours of serum deprivation with Five PhosphoUpid Mixture alone in Basal Medium Eagle's (BME) culture medium or supplemented with 5% or 25% of the Cardiosol™ organ preservation solution described in United States Patent No. 4,938,961. Two assays were performed on each cell culture plate consisting of differential cell counts.
1. AU non-adherent or loosely adherent cells were removed from the culture dish and counted by appropriate techniques, typicaUy counting by electronic particle counting instrument. These were the apoptotic cells, the serum deprived released cells (SDR), released by the action of cultivation in serum-free medium. Approximately 95% of these released cells were apoptotic as shown by both ultrastructure analysis and DNA fragmentation analysis.
2. The remaining adherent cells (ADH) were exposed to a buffered, typicaUy pH_7.3, balanced salt solution such as Hanks Balanced Salt Solution without calcium and magnesium salts containing 0.05% trypsin and 0.53_mM ethylene maminetetraacetic acid (EDTA). Each culture was incubated at either room temperature or 37°C on a rocking platform to ensure uniform distribution of the trypsin reagent over the culture surface. After a standardized period of time, typicaUy 10_minutes, the released cells were removed from each culture dish and measured by the same means as described above, typicaUy electronic particle counting. This ADH cell count was comprised of both trypsin resistant and trypsin sensitive cells as described in PCT Publication No._WO_94/25621. Anti-apoptotic activity is expressed as the calculated concentration of material ((g/ml of media) required to save 50% of the cells released on serum free treatment.
Example 9
Preparation and use of Five PhosphoUpid Mixture in PEG20L Twenty milligrams of Five PhosphoUpid Mixture stored in CHC13 (20 mg/mL) under
Argon was dried in a glass tube with vacuum concentrator (Savant) with gentle heat. This was sonicated for 5 minutes under an Argon blanket in 1 mL of buffer (50 mM NB^CO^ 104 mM
NaCl, 250 (M EDTA) 5 mL of which had been bubbled for 5 minutes with Argon.
250 (L (5 mg) of the Five PhosphoUpid Mixture sonicate was added under a stream of Argon to 100 mL of 10% PEG20L which had been prepared in water, ultrafiltered then sterilized by filtration (0.22 (M) and stored anaerobicaUy (under Argon in Ar-fiUed bags). The resulting 50 (g/mL Five PhosphoUpid Mixture in 10% PEG20L was packaged under a stream of Argon into 10 mL autoclaved amber glass vials in 2 mL and 10 mL amounts and sealed with autoclaved butyl rubber septa/aluminum rings. Prior to use, the septa had been subjected to 2 hours of high vacuum then sealed in an aluminum bag filled with Argon in order to displace as much dissolved oxygen as possible. The Five PhosphoUpid Mixture/PEG vials were labeled then individually packaged into aluminum bags filled with Argon and stored at 4°.
Example 10 Activity of LPA and PEG in in vitro C3H/10T1/2 assays
All the phospholipid mixtures were prepared by combining the desired phosphoUpids in solution with organic solvent then drying down the mixture with gentle heat (45 °C) and high vacuum foUowed by sonication into aqueous suspension at 5 to 20 mg/mL. In some cases, the phospholipids were then mixed with serum-free culture medium containing PEG of the indicated molecular weight, concentration and type of PEG noted in the figures and prepared as described above. The results obtained are depicted in Fig. 11 and Table 3.
Example 11 in vitro Cardiomyocyte Assays Isolation of rat neonatal cardiomvocvtes Cardiomyocytes were prepared from hearts of day-old Sprague Dawley rats by trypsinization and mechanical disaggregation (Simpson, (1985)) Circ. Res.56:884-894. The cells were resuspended in MEM, lxMEM vitamins (Gibco), 5% fetal bovine serum and 50 U/ml peniciUin-G and pre-plated for 30 minutes to reduce contamination of non-myocytes. The non-adherent cardiac myocytes were separated and seeded in 2 ml in 35 mm dishes at a density of 3.5xl05 viable cells per ml. The cells were aUowed to adhere for 16-24 hours in a 37°C/5% C02 humidified incubator.
Cardiomyocyte treatment
For serum deprivation, the medium was replaced with fresh serum-free RPMI, whereas serum/glucose deprivation was performed using glucose-free RPMI. The induction of cell death by adriamycin or C2-ceramide was accomplished by the addition of the agent prepared in serum-free RPMI. As a model of ischemia, cultures in serum and glucose free RPMI were placed in an airtight chamber and the latter was continuously perfused with oxygen-free gas overlay of 95% N2/5% C02 for 8 hours at 37°C. To model reperfusion of the ischemic ceUs, 10% fetal bovine serum, 2 g/L of glucose were added and the cultures were returned to a normal oxygen gas overlay (37°C/5% CO2) in a humidified incubator for 16 hours.
To investigate effects of LPA, LPA plus other phosphoUpids, PEG or their mixtures on ceU death, various combinations of these components were added to cells in serum free medium at the beginning of cytotoxic treatment. Measurement of ceU death
Because cardiomyocytes are terminally differentiated non-dividing cells, viability was determined by measurement of the decrease in the relative number of adherent cells. The measurement of non-adherent cells was found to be less reproducible because of their rapid lysis foUowing release from adhesion substrate. Adherent cardiomyocytes were coUected from culture dishes using 0.25% Trypsin/0.05% EDTA and counted on Coulter Counter ZM and Coulter Channelyzer 256.
The results were as foUows:
1. Five PhosphoUpid Mixture prevents cardiomyocyte death induced by serum/glucose deprivation or by ischemia/reperfusion (Fig. 13, 14).
2. 0.3-2% PEG with molecular weight 20,000 kDa enhances the protective effect of Five PhosphoUpid Mixture against serum/glucose deprivation induced ceU death (Fig.13).
3. Mixtures ofPEG with Five PhosphoUpid Mixture and 18: 1-LPA equaUy protect cardiomyocyte death induced by ischemia and reperfusion (Fig._14).
TABLE 5
Figure imgf000085_0001
Figure imgf000086_0001
ND: No protection detected.
4. Various LPAs prevent cardiomyocyte death induced by serum/glucose deprivation (results appear in Table 5).
5. Five Phospholipid Mixture reduces cardiomyocyte death induced by ceramide and the addition of PEG to the Five PhosphoUpid Mixture enhances this effect.
In these experiments, 10(M ceramide was added to ceU cultures of cardiomyctes alone or in the presence of: (a) 0.3% PEG, (b) 2% PEG, (c) 5 (M LPA presented as Five PhosphoUd Mixture, (d) 5 (M LPA presented as Five PhosphoUd Mixture plus 0.4% PEG, and (e) 5 (M LPA presented as Five PhosphoUd Mixture plus 2% PEG. Controls also included only serum and glucose without any active ingredients. In other experiments, 15(M ceramide was added to cardiomyctes alone or in the presence of: (a) 0.3% PEG, (b) 2% PEG, (c) 5(M LPA presented as Five Phospholipid Mixture, (d) 5 (M LPA presented as Five PhosphoUd Mixture plus 0.4% PEG, and (e) 5 (M LPA presented as Five PhosphoUd Mixture plus 2% PEG.
It is clear from these results that LPA has anti-apototic activity as shown in the cardiomyocyte assay, and that the combination of LPA with other phosphoUpids retains that activity. Additionally, the presence of PEG improves the activity of LPA.
Example 12
Regional ischemic dog heart model
The experimental model used was a regional ischemic dog heart (mongrel male hound, approximately 20 kg) with the left anterior descending coronary artery clamped distal to the first diagonal branch rendering a portion ofthe left ventricle ischemic. The clamp was maintained for 90 minutes then released gradually over a 5 minute period. A narrow canula was placed into the left main coronary artery through which Five Phospholipid Mixture (5 mL of 50 (g/mL in 10% PEG) was infused starting 75 minutes after the start ofthe ischemic period, and continuing for 75 minutes in total (approximately 67 (IJmin., approximately 3.3 (g Five PhosphoUpid Mixture/min.). The total dose was 250 (g Five PhosphoUpid Mixture and 500 mg PEG20L.
Determination of Infarct size
After staining and fixation, the atria and right ventricle are removed from the left ventricle. After removal ofthe apex, the portion ofthe left ventricle distal to the mitral apparatus is cut into 5 transverse sections of equal thickness. Evans Blue dye injected into the circumflex vessel stains the circumflex territory blue. TTC stains viable LAD territory red, while the infarcted myocardium will remain unstained (white). Sections are weighed and photographed for documentation and future verification of results . Computer assisted planimetry is used to measure the areas ofthe circumflex, viable LAD and infarcted LAD territories. Calculation of infarct size is based upon the assumption that the area of infarction in the sectioned plane is representative of the mass of infarcted tissue in that plane.
Study Protocols
1. Study-drug group; dogs are treated with Five PhosphoUpid Mixture 50_(g/ml and
PEG 100_mg/ml as a bolus plus infusion (at 4_nύ7hr) starting 15 min before reperfusion and going on for 60 min into the reperfusion period. 2. Placebogroup: dogs receive placebo foUowing the same modalities described for the study drug group.
3. SOD-Catalase group: dogs receive SOD plus catalase following the same modaUties described by Simpson et_al. (1987) Fed. Proc. 46:7, 2413-21.
The rationale for using a third group of dogs in the present protocol resides in the fact that the SOD + Catalase group represents a positive control. In fact it has been shown that the combination of oxygen radical scavenger is very efficacious in limiting myocardial reperfusion injury in the canine mode.
The results obtained in the dog heart experiments are depicted in Fig. 15.
Example 13
Evaluation of Infarct Measurement and EKG Alterations in a Regional Ischemic Pie Heart Model This example evaluates the anti-apoptotic activity of PEG20L in a regional ischemic pig heart model. Adult Yucatan miniature pigs (approx. 30 kg each) were treated with general anesthesia followed by balloon catheterization to create an occlusion ofthe first obtuse marginal branch ofthe circumflex coronary artery. After 105 minutes of occlusion treatment with control, infusion of PEG20L (100 mg/mL), or PEG20L and Five PhosphoUpid Mixture (100 mg/mL PEG + 50 (g/mL Five Phospholipid Mixture) solution was started through the guide catheter into the aortic root in close approximation to the occlusion. The solutions were introduced initially by a bolus equal to 25% ofthe hourly drug dose, foUowed by continuous infusion through the guide catheter for 135 minutes. The occlusion was removed after 120 minutes (15 minutes after the start of drug dehvery).
Heart function was monitored by EKG and cardiac output was determined by contrast imaging. The animal was euthenized 1-3 days after the occlusion. The heart was removed for histology and examined. Infarct size was determined by a method of triphenyl tetrazolium chloride
(TTC) staining for the area at risk and actual infarct. The area not at risk was stained with a blue pigment or Evan's Blue dye.
Pigs were treated with one ofthe following test regimens (displayed as total dose): a) 250 mg/kg PEG20L only (10% ultrafiltered in water) (n = 2); b) 125 μg/kg Five Phospholipid Mixture + 250 mg/kg PEG20L (n = 3). Four additional pigs were occluded but not treated with any solution, for use as controls. Of the four controls the mean infarct size (as % risk zone) was 34.5% ( 7.2 %. The controls also showed a marked unresolved S-T segment shift during the ischemic period which reflects severe myocardial ischemia and infarction.
In contrast to the controls, two effects were noted in the treatment groups. First, in some animals, within 2 minutes ofthe start of drug infusion there was a normalization ofthe EKG signal disturbances in the occluded hearts. During the ischemic period the normal EKG signal shifted to one in which there was an elevation in the S-T segment indicating an impairment of myocyte repolarization. This elevation disappeared in several pigs treated with PEG20L and Five
PhosphoUpid Mixture. Some pigs that were treated were not included in the infarct measurement data due to technical difficulties in determining the infarct size. However, some of these animals did exhibit a reduction in S-T segment elevation.
The second effect noted was a decrease in infarct size in the treated animals when compared to the control animals (Fig. 12). In three Five Phospholipid Mixture treated pigs with measurable infarcts, the infarct sizes were reduced to only 12.3 % ( 5.1 %, in contrast to 34.5 %in the controls. It is clear from this example that LPA and LPA in combination with PEG are effective therapeutics for the treatment of cardiac ischemia, as both protect tissue from infarct damage and reduce the level of S-T segment shift measured by electrocardiogram.
Example 14
Evaluation of Infarct Measurement in a Regional Ischemic Rat Heart Model
The left anterior descending coronary artery (LAD) of each Sprague dawley female rat
(225-250 grams) was occluded for 20 minutes followed by 2 hours of blood reperfusion. The heart was then removed and the non-ischemic area not at risk for developing an infarction was delineated by perfusing Evan's Blue through all but the LAD ofthe coronary vasculature. The infarct and area at risk for infarction was identified by perfusing the LAD with triphenyltetrazohum chloride which is metabolized to a red dye in viable tissues while remaining uncolored within the necrotic zone. The heart was then fixed and sliced in order to permit planimetry measurements of both the area at risk for developing the infarction and the actual infarct size itself. The results are expressed as infarct size as a proportion ofthe area at risk.
At start of reperfusion a mixture of Five PhosphoUpid Mixture (1 mg kg) (preparation described in Example 3) and PEG (8 mg/kg) was injected over an approximately two minute period into the left ventricle through a 35 gauge needle. Control rats received a similar volume injection of placebo containing 10 mM citrate/110 mM NaCl, pB 6.5. The results, in Fig. 16, show that treatment with LPA, in a phospholipid mixture, combined with PEG resulted in a 42% reduction in infarct size relative to placebo-treated rats (p=0.005, n=4 in both groups, two tailed T-test). The areas at risk in the two groups were similar (p=0.42).
Example 15
Evaluation of Infarct Measurement in a Regional Ischemic Rabbit Heart Model Male New Zealand white rabbits were initially anesthetized using a mixture of ketamine (400 mg per rabbit) and xylazine (20 mg per rabbit) administered intramuscularly in two doses, approximately 10 minutes apart. Throughout the study, a level of deep anesthesia is maintained using sodium pentobarbital given intraperitoneaUy at a dose of approximately 50 mg/hour. AU rabbits were intubated and mechanically ventilated using room air supplemented with oxygen. Fluid filled catheters were placed into the jugular vein to administer fluids. A catheter was also placed into the left carotid artery to measure heart rate and blood pressure and to obtain reference blood samples during regional myocardial blood flow measurements. The chest was opened through the left fourth intercostal space. Then, the pericardium was incised and the heart was exposed. A large anterolateral branch ofthe circumflex artery, or the circumflex artery itself was identified and encircled with a 4-0 silk suture. The ends ofthe suture were threaded through a piece of flanged tubing, forming a snare, which was later used to occlude the artery. A catheter was then placed into the left atrial appendage to inject the control or test solution, the radioactive microspheres and blue pigment at the conclusion ofthe procedure.
After the surgical preparation, the rabbits were stabilized for ten minutes. At this time, the rabbits were randomized into either the treatment or control group. The treatment solution contained 1 mg/ml ofthe Five PhosphoUpid Mixture, described in Example 3, above, 8 mg/ml PEG 20L, in 10 mM citrate 110 mMNaCl. The control solution contained 10 mM citrate and 110 mM NaCl. Five minutes before occlusion, a bolus dose of 1 mg/kg body weight of either the Five Phospholipid Mixture/PEG solution or the control solution was administered into the left atrium. Immediately following the bolus dose, infusion was started at the rate of 1 mg/kg for one hour into the left atrium. Baseline hemodynamics and core body temperature were recorded. Next, the artery was occluded for 30 minutes. FoUowing this, the hearts were reperfused for three hours.
Heart rate and arterial blood pressure were recorded at 20 minutes of occlusion and at time points during reperfusion at a 25 mm/second paper speed. Regional myocardial blood flow measurements were performed on two occasions; during coronary occlusion (20 minutes) to confirm no blood flow in the ischemic zone, and during early reperfusion (30 minutes) to confirm reflow in the same zone. At the end of three hours of reperfusion, the coronary artery is reoccluded. Next, 4 ml of
50% Unisperse blue (Ciby-Geigy, Hawthorne, NY) were infused through the left atrial catheter and allowed to circulate throughout the vascular system. The rabbit was then euthanized by an overdose intravenous injection of xylazine (300 mg) foUowed by 12 mEq of potassium chloride given into the left atrium. Prospective exclusion criteria included an ischemic risk zone of less than 10% ofthe left ventricular weight, a regional blood flow of more than 0.2 ml/minute/g in the risk zone during coronary artery occlusion (lack of ischemia), or a regional blood flow of less than 0.4 ml/min/g in the risk zone at 30 minutes of reperfusion (failure to reperfuse).
Infarct size was evaluated as follows. The right ventricle was trimmed off and the left ventricle was sliced transversely into seven or eight sections, approximately two millimeters in thickness. These slices were photographed to identify the ischemic risk regions (uncolored by the blue pigment) and the non-ischemic regions (colored by the blue pigment). The slices were then incubated in a 1% solution of triphenyltetrazoUum chloride pre-heated to 37 °C for 10 minutes and rephotographed for analysis of area of necrosis. AU sections were later fixed in formalin. These photographic slides were projected and areas of risk (AR) and areas of necrosis (AN) were traced by planimetry. The planimetered areas of each sUce were multiplied by the weight ofthe sUce and then summed. Because infarct size is measured from photographic sUdes, the entire left ventricle was used for the analysis.
Regional myocardial blood flow (RMBF) was measured as foUows. Just before measuring RMBF, during occlusion, the atrial catheter was disconnected from the treatment infusion pump.
Radioactive microspheres were injected via the atrial catheter. The catheter was then reprimed with approximately 0.2 ml ofthe drug treatment, and the catheter was reconnected to the pump and infusion continued.
Regional myocardial blood flow was measured with 11 (m radioactive microspheres labeled with 141Ce, * Nb or I03 Ru (New England Nuclear, North Billerica, MA), using approximately 500,000 per injection. These microspheres were injected into the left atrial catheter. At the same time, a reference blood sample was obtained from the carotid artery at 2.06 ml/minute. The blood removed during RMBF measurement was about 5 ml. These volume changes do not cause changes in systemic arterial pressure. At the end ofthe protocol, after the photographic sUdes had been taken and the heart weight, myocardial samples were cut from the center ofthe non-ischemic and the ischemic regions, weighed and counted with the reference blood samples in a weU gamma counter. Blood flows at each interval, for ischemic and non-ischemic tissues, were then computed and expressed in ml/minute/g.
The results were as follows. With a risk zone of approximately 25% ofthe left ventricle, treatment with the control solution resulted in infarct size of approximately 40% ofthe risk zone on average, as compared to an average of only approximately 25% ofthe risk zone in the subjects treated with the LPA containing PEG and other phospholipids.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be apparent to those skilled in the art that certain changes and modifications can be practiced. Therefore, the descriptions and examples should not be construed as limiting the scope ofthe invention, which is delineated by the appended claims.

Claims

Claims:
1. A composition, comprising a compound ofthe foUowing formula:
M
II
W-P-OH
I w
wherein M is O or S, each W is independently selected from one ofthe foUowing structures:
X Z
II I
R-C -X-(CH2X -CH-CH2-Y
Z R-X- (CH2)π -CH-CH2-Y
X V Z
R _C II _Z -(CH2χ -(C IH)m -C IH-CH2 -Y
O Z
II I
R-C -NH-(CH2χ -CH-CH2 -Y
wherein Y is O or S; R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl, or ((CB2)pO)q(CB2)pT where q is an integer from 1 to about 900 and where each p is independently an integer from 2 to about 10 and T is OB, or
Figure imgf000092_0001
where b is an integer from 0 to about 10; each V is independently OB, SB, B, NB2, halogen, OP03B2, or OSO^; n is an integer from 0 to about 10; m is an integer from 0 to about 10; Z is OB SB, NH2, halogen, OPOjBz, B, ©(CB^CB, where b=0 to about 2, or S03B; L is independently O, S, or
CB2 and X is independently O or S; or a salt thereof.
2. The composition according to claim 1, wherein R is an alkyl having between about 5 and about 24 carbon atoms, and mixtures thereof.
3. The composition according to claim 1, wherein R is an alkyl having 24 carbon atoms, wherein 1 or 2 ofthe carbon-carbon bonds are unsaturated, and mixtures thereof.
4. A composition, comprising: Bis(3-0-oleyl-2-0-methyl-røe-glyceryl) thiophosphate, or a salt thereof.
5. A composition, comprising a compound ofthe foUowing formula:
Figure imgf000093_0001
wherein each W is independently SB, OB OCB2CB(NB2)C02B, OCBCB3CB(NB2)C02B, OP03B2, 0P02 0P03B2 or Q, wherein when one W is Q, the other W is OB and wherein Q is one ofthe foUowing structures:
X Z
II I R ~ (CH2X -X - C - CH - CH2 - Y
X V Z
R - (CH2χ -L - C - (CH). - CH - CH2 - Y
wherein Y is O or S; R is unsubstituted or substituted, saturated or unsaturated, straight or branched-chain alkyl, or ((CB^pO^CBJpT where q is an integer from 1 to about 900 and where each p is independently an integer from 2 to about 10 and T is OB, or 0(CB2)bCB3 where b is an integer from 0 to about 10; each V is independently OH, SB B, NB2, halogen, OP03B2, or OSO3B; n is an integer from 0 to about 10; m is an integer from 0 to about 10; Z is O S NH2, halogen, OP03B2, O CB^CBa where b=0 to about 2, or SO3B; L is independently O, S, or CB2; X is independently O or S; and M is P or S, where when M is S, one W is (=0) and the other W is SB O OCB2CB(NB2)C02B OCHCB^BflSIB^CO^ OP03B2, or OP02BOP03B2; or a salt thereof.
6. A composition comprising a compound selected from the group consisting of reverse ester-LPA, reverse thioester-LPA and a salt of either compound.
7. A composition comprising 3 Oleyl l-thiophosphoryl-2-O-methyl-røc-glycerate, or a salt thereof.
8. A method of treating apoptosis, or preserving or restoring function in a cell, tissue or organ comprising administering in vivo a therapeutically effective amount of a pharmaceuticaUy acceptable composition of claims 1, 2, 3, 4, 5, 6, or 7.
9. The composition of Claims 1, 2, 3, 4, 5, 6, or 7, wherein said composition further comprises a potentiating component.
10. The composition of claim 9, wherein said component is a polyethylene glycol.
11. The composition of Claim 9, wherein said component is a protein.
12. The composition of Claim 9, wherein said component is a lipid membrane structure.
13. The composition according to claim 12, wherein the Upid membrane structure comprises at least one compound selected from the group consisting of a lipid, a phospholipid and a surfactant.
14. The composition according to claim 13, wherein the lipid is selected from the group consisting of phosphoUpids, glycolipids, steroids, bolaamphiles and a combination thereof.
15. The composition according to claim 13, wherein the surfactant is nonionic.
16. The composition according to claim 13, wherein the lipid membrane structure further comprises a tissue targeting compound.
17. The composition according to claim 16, wherein the tissue targeting compound is selected from the group consisting of an antibody, a cell surface receptor, a ligand for a ceU surface receptor, a polysaccharide, a drug, a hormone, a hapten, a special lipid and a nucleic acid.
18. The composition according to claim 13, wherein the composition further comprises a component selected from the group consisting of a polypeptide, a modified polypeptide and a polymer.
19. The composition according to claim 18, wherein the polypeptide is a fatty acid binding protein.
20. The composition according to claim 18, wherein the polymer is a naturally occurring polymer and is selected from the group consisting of dextrans, hydroxyethyl starch, and polysaccharides.
21. The composition according to claim 20, wherein the polysaccharide is selected from the group consisting of trehalose, glucose, maltose, lactose, maltulose, iso-maltulose, lacrulose, mono-reducing glycosides of polyhydroxy compounds selected from sugar alcohols, other straight chain polyalcohols, raffinose, stachyose, melezitose, dextran, sucrose and sugar alcohols thereof, maltitol, lactitol, iso-maltulose, palatinit, 2-D-glucopyranosyl- 1 (6-mannitol and their individual sugar alcohols.
22. The composition according to claim 18, wherein the polymer is synthetic and is selected from the group consisting of polyalkyl glycols, polyoxyethylated polyols, polyvinylpyrrolidone, polyhydroxyethyl methacrylate, polyvinyl alcohols, polyurethane, polytrimethylene glycol, polypropylene glycol, polyacryUc acid, polyethyloxazoUne, polyacrylamide, polyphosphazene, poly(lactic acid), poly(glycoUc acid), polyamino acids and polymeric mixtures thereof.
23. The composition according to claim 11 , wherein the protein comprises at least one compound selected from the group consisting of a lipid binding protein and a Upid carrier protein.
24. The composition according to claim 11, wherein the protein is selected from the group consisting of albumin, soy and plant protein, cytochrome c, low density lipoprotein, acyl carrier protein, and alphafeto protein.
25. The composition according to claim 12, wherein the weight ratio of PEG to LPA is 1-100,000 to 1.
26. The composition according to claim 12, wherein the PEG has an average molecular weight from about 8,000 to about 40,000.
27. The composition according to claim 12, wherein the PEG has an average molecular weight of about 20,000.
28. The composition according to claims 1, 2, 3, 4, 5, 6, or 7, further comprising pharmaceutically acceptable excipients.
29. The composition according to claims 1, 2, 3, 4, 5, 6, or 7, further comprising a pharmaceutically effective agent.
30. The composition according to claim 29, wherein the pharmaceutically effective agent is selected from the group consisting of a drug, an antibiotic, a wound healing agent and an antioxidant.
31. The composition according to claim 30, wherein the drug is selected from the group consisting of antipyretic and anti-inflammatory, analgesics, antiarthritics, antispasmodics, antidepressants, antipsychotics, tranquilizers, antianxiety drugs, narcotic antagonists, antiparkinsonism agents, choUnergic antagonists, chemotherapeutic agents, immuno-suppressive agents, antiviral agents, parasiticides, appetite suppressants, antiemetics, antihistamines, antimigraine agents, coronary vasodUators, cerebral vasodUators, peripheral vasodilators, hormonal agents, contraceptives, antithrombotic agents, diuretics, antihypertensive agents, cardiovascular drugs, opioids, and vitamins.
32. The composition according to claim 30, wherein the antibiotic is selected from the group consisting of ampicillin, tetracycline, chloramphemcol, erythromycin, amphotericin B and penicillin.
33. The composition according to claim 30, wherein the wound healing agent is selected from the group consisting of transforming growth factors, platelet-derived growth factors, epidermal growth factors and fibroblast growth factors.
34. The composition according to claim 30, wherein the antioxidant is selected from the group consisting of Vitamin_C, Vitamin_E , Vitamin A, dihydroUpoamide, flavenoids, butylated hydroxytoluene, butylated hydroxyanisole, Trolox®, propyl gallate, phenoUc antioxidants, phenothiazines, desferrioxamide, HBED and CP130.
35. A method of making the composition of Claim 9, comprising the steps of: a) forming a Upid dispersion comprising LPA; b) providing at least one of said components; and c) combining the products of steps a and b.
36. The method according to claim 35, wherein the Upid dispersion is formed by the steps of: a) dissolving LPA and any other Upids in organic solvent; b) removing the solvent to form dried Upid; and c) dispersing the dried lipid into aqueous media by the steps of: i) forming an even Upid dispersion; and ii) forming an even dispersion of lipid membrane structures.
37. The method according to claim 35, further comprising the step of d) sterilizing the dispersion.
38. A composition obtained according to a method according to claim 35.
39. The method of claim 8, wherein said pharmaceuticaUy acceptable composition further comprises a potentiating component.
40. The method of claim 39, wherein said potentiating component comprises a polyethylene glycol.
41. The method of claim 39, wherein said potentiating component comprises a protein.
42. The method of claim 39, wherein said potentiating component comprises a Upid membrane structure.
43. The method according to claim 8, comprising administering said composition to a patient suffering from a condition related to apoptosis, ischemia, traumatic injury or reperfusion damage.
44. The method according to claim 8, comprising administering said composition to a patient suffering from gastrointestinal perturbation.
45. The method according to claim 44, wherein the gastrointestinal perturbation is caused by a stimulus selected from the group consisting of viruses, chemotherapeutic agents, radiation, infectious diseases, inflammatory bowel disease, and diarrhea-causing organisms.
46. The method according to claim 45 , wherein the virus is human immunodeficiency virus.
47. The method according to claim 8, wherein the method diminishes apoptosis-related problems associated with immunosuppressing viruses, chemotherapeutic agents, or radiation and immunosuppressive drugs.
48. The method according to claim 43 , wherein the reperfusion damage is associated with coronary artery obstruction; stroke; cerebral infarction; spinal head trauma and concomitant severe paralysis; frostbite; coronary angioplasty; blood vessel attachment; limb attachment; organ attachment; and kidney reperfusion.
49. A method of culturing cells comprising treating ceUs with an amount of the composition according to claims 1, 2, 3, 4, 5, 6 or 7 effective to prevent apoptosis or preserve the cells.
50. The method according to claim 49, wherein the cells are part of a tissue or organ.
51. A method of preserving an organ comprising adding an effective amount of the composition according to claim 1, 2, 3, 4, 5, 6 or 7 to the solution with which the organ is treated.
52. A method of organ preservation comprising administeringto adonor organ at least one intravenous bolus of an effective amount ofthe composition according to claim 1, 2, 3, 4, 5, 6, or 7.
53. The method according to claim 8, wherein the patient is undergoing a condition selected from the group consisting of cardioplegia, congestive heart failure, angioplasty, and a valve operation.
54. A method of treating dermatologic conditions, comprising topicaUy administering a therapeutically effective amount of a pharmaceuticaUy acceptable composition comprising the composition according to claims 1, 2, 3, 4, 5, 6, or 7 to a patient in need of such treatment.
55. The method according to claim 54, wherein the dermatological condition is wrinkling, or hair loss.
56. A method of treating wounds comprising administering an effective amount ofthe composition according to claims 1, 2, 3, 4, 5, 6, or 7.
57. The method according to claim 55, wherein the wounds are burn wounds.
PCT/US1999/005943 1998-03-18 1999-03-17 Compositions containing lysophosphatidic acids which inhibit apoptosis and uses thereof WO1999047101A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP99913956A EP1069895A4 (en) 1998-03-18 1999-03-17 Compositions containing lysophosphatidic acids which inhibit apoptosis and uses thereof
US09/646,599 US6949528B1 (en) 1998-03-18 1999-03-17 Compositions containing lysophosphatidic acids which inhibit apoptosis and uses thereof
AU31915/99A AU3191599A (en) 1998-03-18 1999-03-17 Compositions containing lysophosphatidic acids which inhibit apoptosis and uses thereof
US11/236,429 US7259273B1 (en) 1998-03-18 2005-09-26 Compositions containing lysophosphotidic acids which inhabit apoptosis and uses thereof
US11/834,505 US20080076736A1 (en) 1998-03-18 2007-08-06 Compositions containing lysophosphatidic acids which inhibit apoptosis and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US7837598P 1998-03-18 1998-03-18
US60/078,375 1998-03-18

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US09646599 A-371-Of-International 1999-03-17
US11/236,429 Division US7259273B1 (en) 1998-03-18 2005-09-26 Compositions containing lysophosphotidic acids which inhabit apoptosis and uses thereof

Publications (2)

Publication Number Publication Date
WO1999047101A2 true WO1999047101A2 (en) 1999-09-23
WO1999047101A3 WO1999047101A3 (en) 1999-11-25

Family

ID=22143648

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/005943 WO1999047101A2 (en) 1998-03-18 1999-03-17 Compositions containing lysophosphatidic acids which inhibit apoptosis and uses thereof

Country Status (3)

Country Link
EP (1) EP1069895A4 (en)
AU (1) AU3191599A (en)
WO (1) WO1999047101A2 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001012838A2 (en) * 1999-08-18 2001-02-22 Atairgin Technologies, Inc. Compounds which modulate the activity of an lpa receptor
US6380177B1 (en) 1999-06-25 2002-04-30 Atairgin Technologies, Inc. LPA analogs as agonists of the Edg2 LPA receptor
EP1214928A1 (en) * 1999-08-18 2002-06-19 Kyowa Hakko Kogyo Co., Ltd. Hair growth stimulants
EP1232740A2 (en) * 2001-02-16 2002-08-21 Kyowa Hakko Kogyo Co., Ltd. Hair-growing agent
EP1248628A1 (en) * 1999-12-14 2002-10-16 Sky High, LLC Phospholipid compositions as anti-inflammation agents
JP2002308779A (en) * 2001-04-13 2002-10-23 Gencom Co Agent for promoting propagation, differentiation and/or survival of glia cell, containing cyclic phosphatidic acid
JP2002308778A (en) * 2001-04-13 2002-10-23 Gencom Co Nerve cell pro-survival agent containing cyclic phosphatidic acid derivative
US6492334B1 (en) 1999-12-30 2002-12-10 Robert James Generale Tri-compound analgesic for treating inflammation and pain
EP1263752A2 (en) * 2000-03-17 2002-12-11 The University Of Tennessee Research Corporation Lpa receptor agonists and antagonists and methods of use
EP1427424A2 (en) * 2001-09-17 2004-06-16 The University Of Tennessee Research Corporation Lpa receptor agonists and antagonists and methods of use
US6949529B2 (en) 1997-03-19 2005-09-27 Sky High, Llc Compositions containing lysophosphotidic acids which inhibit apoptosis and uses thereof
EP1750722A2 (en) * 2004-05-06 2007-02-14 University Of Virginia Patent Foundation Novel lysophosphatidic acid receptor selective antagonists
JP2007508324A (en) * 2003-10-09 2007-04-05 ユニバーシティ オブ テネシー リサーチ ファウンデーション LPA receptor agonists and antagonists and methods of use
EP1906972A2 (en) * 2005-07-19 2008-04-09 The University of Tennessee Research Foundation Lpa2 receptor agonist inhibitors of cftr
US7550449B2 (en) 2002-06-11 2009-06-23 Kimiko Murofushi Carba cyclic phosphatidic acid derivative
JP2010535193A (en) * 2007-08-01 2010-11-18 サーントゥル ナシオナル ドゥ ラ ルシェルシュ シャーンティフィク セエンエールエス Thiophosphine (thiophosphonic) acid derivatives and their therapeutic applications

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4938961A (en) 1989-04-28 1990-07-03 Geoffrey Collins Organ preservation solution containing pokyethylene gycol and method of performing cardioplegia
WO1995015173A1 (en) 1993-11-30 1995-06-08 Lxr Biotechnology Inc. Compositions which inhibit apoptosis, methods of purifying the compositions and uses thereof

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS492266B1 (en) * 1970-06-06 1974-01-19
JPS57167364A (en) * 1981-04-07 1982-10-15 Sankin Kogyo Kk Adhesive for hard tissue of human body
JPS5810559A (en) * 1981-07-10 1983-01-21 Ajinomoto Co Inc Preparation of alpha-chloro-beta-thiosulfo-propionic acid ester salt
JPS58104624A (en) * 1981-12-16 1983-06-22 Kao Corp Emulsified composition
JPS63265972A (en) * 1987-04-24 1988-11-02 Denki Kagaku Kogyo Kk Modified vinyl chloride resin solution
JP3142636B2 (en) * 1992-05-13 2001-03-07 三洋化成工業株式会社 Alkaline cleaning agent
AU1743495A (en) * 1994-02-04 1995-08-21 Cell Therapeutics, Inc. Composition for wound healing, neuron growth and vascularization
US6071528A (en) * 1997-02-19 2000-06-06 Ultradent Products, Inc. Adhesive antimicrobial and/or reparative dentin stimulating dental compositions and methods for forming and using such compositions
AU751876B2 (en) * 1997-03-19 2002-08-29 Sky High, Llc Compositions containing lysophosphotidic acids which inhibit apoptosis and uses thereof
AU9027298A (en) * 1997-08-20 1999-03-08 Lxr Biotechnology Inc. Compositions containing polyethylene glycol and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4938961A (en) 1989-04-28 1990-07-03 Geoffrey Collins Organ preservation solution containing pokyethylene gycol and method of performing cardioplegia
WO1995015173A1 (en) 1993-11-30 1995-06-08 Lxr Biotechnology Inc. Compositions which inhibit apoptosis, methods of purifying the compositions and uses thereof
US5567425A (en) 1993-11-30 1996-10-22 Lxr Biotechnology Inc. Compositions which inhibit apoptosis, methods of purifying the compositions and uses thereof
US5620885A (en) 1993-11-30 1997-04-15 Lxr Biotechnology Inc. Compositions which inhibit apoptosis, methods of purifying the compositions and uses thereof
US5624672A (en) 1993-11-30 1997-04-29 Lxr Biotechnology Inc. Compositions which inhibit apoptosis, methods of purifying the compositions and uses thereof
US5635186A (en) 1993-11-30 1997-06-03 Lxr Biotechnology Inc. Compositions which inhibit apoptosis, methods of purifying the compositions and uses thereof

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
CHIEN ET AL.: "Medical Intelligence Unit", 1993, R.G. LANDES CO., article "Hibernation Induction Trigger for Organ Preservation"
COLLINS ET AL., THE LANCET, vol. 338, 1991, pages 890 - 891
See also references of EP1069895A4
TOMEI; COPE ET AL.: "Apoptosis: The Molecular Basis of Cell Death", 1991, COLD SPRING HARBOR LABORATORY PRESS
WICOMB ET AL., J. HEART LUNG TRANSPLANTATION, vol. 13, 1994, pages 891 - 894
WICOMB ET AL., J. SURG. RES., vol. 40, 1986, pages 276
WICOMB ET AL., TRANSPLANTATION, vol. 48, 1989, pages 6 - 9
WICOMB ET AL., TRANSPLANTATION, vol. 49, 1990, pages 261 - 264

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6949529B2 (en) 1997-03-19 2005-09-27 Sky High, Llc Compositions containing lysophosphotidic acids which inhibit apoptosis and uses thereof
US6380177B1 (en) 1999-06-25 2002-04-30 Atairgin Technologies, Inc. LPA analogs as agonists of the Edg2 LPA receptor
WO2001012838A3 (en) * 1999-08-18 2001-10-04 Atairgin Technologies Inc Compounds which modulate the activity of an lpa receptor
EP1214928A1 (en) * 1999-08-18 2002-06-19 Kyowa Hakko Kogyo Co., Ltd. Hair growth stimulants
WO2001012838A2 (en) * 1999-08-18 2001-02-22 Atairgin Technologies, Inc. Compounds which modulate the activity of an lpa receptor
US7015209B2 (en) 1999-08-18 2006-03-21 Kyowa Hakko Kogyo Co., Ltd. Hair-growing agent
EP1214928A4 (en) * 1999-08-18 2004-03-24 Kyowa Hakko Kogyo Kk Hair growth stimulants
EP1248628A1 (en) * 1999-12-14 2002-10-16 Sky High, LLC Phospholipid compositions as anti-inflammation agents
EP1248628A4 (en) * 1999-12-14 2004-08-04 Sky High Llc Phospholipid compositions as anti-inflammation agents
US6492334B1 (en) 1999-12-30 2002-12-10 Robert James Generale Tri-compound analgesic for treating inflammation and pain
EP1918287A3 (en) * 2000-03-17 2008-08-20 The University Of Tennessee Research Corporation LPA receptor agonists and antagonists and methods of use
US6875757B2 (en) 2000-03-17 2005-04-05 University Of Tennessee Research Foundation LPA receptor agonists and antagonists and methods of use
EP1263752A4 (en) * 2000-03-17 2003-07-30 Univ Tennessee Res Corp Lpa receptor agonists and antagonists and methods of use
EP1263752A2 (en) * 2000-03-17 2002-12-11 The University Of Tennessee Research Corporation Lpa receptor agonists and antagonists and methods of use
EP1232740A3 (en) * 2001-02-16 2003-07-02 Kyowa Hakko Kogyo Co., Ltd. Hair-growing agent
EP1232740A2 (en) * 2001-02-16 2002-08-21 Kyowa Hakko Kogyo Co., Ltd. Hair-growing agent
WO2002083149A1 (en) * 2001-04-13 2002-10-24 Gencom Corporation Drugs fo rpromoting the proliferation, differentiation and/or survival of glial cells containing cyclic phosphatidic acid
WO2002083148A1 (en) * 2001-04-13 2002-10-24 Gencom Corporation Nerve cell survival promoters containing cyclic phosphatidic acid derivative
JP2002308778A (en) * 2001-04-13 2002-10-23 Gencom Co Nerve cell pro-survival agent containing cyclic phosphatidic acid derivative
JP2002308779A (en) * 2001-04-13 2002-10-23 Gencom Co Agent for promoting propagation, differentiation and/or survival of glia cell, containing cyclic phosphatidic acid
EP1427424A4 (en) * 2001-09-17 2006-12-13 Univ Tennessee Res Corp Lpa receptor agonists and antagonists and methods of use
EP1427424A2 (en) * 2001-09-17 2004-06-16 The University Of Tennessee Research Corporation Lpa receptor agonists and antagonists and methods of use
US7550449B2 (en) 2002-06-11 2009-06-23 Kimiko Murofushi Carba cyclic phosphatidic acid derivative
JP2007508324A (en) * 2003-10-09 2007-04-05 ユニバーシティ オブ テネシー リサーチ ファウンデーション LPA receptor agonists and antagonists and methods of use
KR101390040B1 (en) * 2003-10-09 2014-04-29 유니버시티 오브 테네시 리서치 파운데이션 Lpa receptor agonists and antagonists and methods of use
KR101216266B1 (en) * 2003-10-09 2012-12-28 유니버시티 오브 테네시 리서치 파운데이션 Lpa receptor agonists and antagonists and methods of use
EP1750722A2 (en) * 2004-05-06 2007-02-14 University Of Virginia Patent Foundation Novel lysophosphatidic acid receptor selective antagonists
US7820703B2 (en) 2004-05-06 2010-10-26 University Of Virginia Patent Foundation Lysophosphatidic acid receptor selective antagonists
EP1750722A4 (en) * 2004-05-06 2010-03-10 Univ Virginia Novel lysophosphatidic acid receptor selective antagonists
EP1906972B1 (en) * 2005-07-19 2013-09-11 The University of Tennessee Research Foundation LPA2 receptor agonist for treating diarrhea
EP1906972A2 (en) * 2005-07-19 2008-04-09 The University of Tennessee Research Foundation Lpa2 receptor agonist inhibitors of cftr
JP2010535193A (en) * 2007-08-01 2010-11-18 サーントゥル ナシオナル ドゥ ラ ルシェルシュ シャーンティフィク セエンエールエス Thiophosphine (thiophosphonic) acid derivatives and their therapeutic applications

Also Published As

Publication number Publication date
EP1069895A4 (en) 2002-07-31
WO1999047101A3 (en) 1999-11-25
EP1069895A2 (en) 2001-01-24
AU3191599A (en) 1999-10-11

Similar Documents

Publication Publication Date Title
US6949529B2 (en) Compositions containing lysophosphotidic acids which inhibit apoptosis and uses thereof
US7259273B1 (en) Compositions containing lysophosphotidic acids which inhabit apoptosis and uses thereof
AU2003229028B2 (en) A direct cellular energy delivery system
WO1999047101A2 (en) Compositions containing lysophosphatidic acids which inhibit apoptosis and uses thereof
CA2284142C (en) Compositions containing lysophosphotidic acids which inhibit apoptosis and uses thereof
US6004579A (en) Compositions which inhibit apoptosis, methods of making the compositions and uses thereof
US9066866B2 (en) Direct cellular energy delivery system
EP0342620B1 (en) Agent for prevention and remedy of injuries caused by ischemia
WO1999008514A1 (en) Compositions containing polyethylene glycol and uses thereof
Ahmad et al. Enhanced therapeutic effects of liposome-associated 1-O-octadecyl-2-O-methyl-sn-glycero-3-phosphocholine
Tretiakova et al. Liposomal formulation of a melphalan lipophilic prodrug: Studies of acute toxicity, tolerability, and antitumor efficacy
US20050008664A1 (en) Compositions and methods related to lipid:emodin formulations
US20060063738A1 (en) Compositions containing lysophosphotidic acids which inhibit apoptosis and uses thereof
AU2002309014B2 (en) Compositions containing Lysophosphotidic Acids Which Inhibit Apoptosis and uses Thereof
JPS63250323A (en) Novel anticancer agent
AU2006252244A1 (en) Compositions containing polyethylene glycol and uses thereof
ZA200409170B (en) A direct cellular energy delivery system.

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE 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 MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE 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 MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
NENP Non-entry into the national phase

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: 1999913956

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 09646599

Country of ref document: US

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1999913956

Country of ref document: EP