WO1995035094A1 - Sphingosomes for enhanced drug delivery - Google Patents

Sphingosomes for enhanced drug delivery Download PDF

Info

Publication number
WO1995035094A1
WO1995035094A1 PCT/CA1995/000363 CA9500363W WO9535094A1 WO 1995035094 A1 WO1995035094 A1 WO 1995035094A1 CA 9500363 W CA9500363 W CA 9500363W WO 9535094 A1 WO9535094 A1 WO 9535094A1
Authority
WO
WIPO (PCT)
Prior art keywords
liposomai
mol
liposomes
composition
vincristine
Prior art date
Application number
PCT/CA1995/000363
Other languages
French (fr)
Inventor
Murray S. Webb
Marcel B. Bally
Lawrence D. Mayer
James M. Miller
Paul G. Tardi
Original Assignee
Inex Pharmaceuticals Corp.
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 Inex Pharmaceuticals Corp. filed Critical Inex Pharmaceuticals Corp.
Priority to AU27094/95A priority Critical patent/AU2709495A/en
Priority to EP95922375A priority patent/EP0804159B1/en
Priority to DE69531701T priority patent/DE69531701T2/en
Priority to AT95922375T priority patent/ATE248586T1/en
Priority to JP50143496A priority patent/JP3270478B2/en
Priority to CA002193502A priority patent/CA2193502C/en
Publication of WO1995035094A1 publication Critical patent/WO1995035094A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids

Definitions

  • Liposomai formulations of therapeutically active drugs have significant advantages over drugs injected in free form. einstein, Liposomes: From Biophysics to Therapeutics, (Ostro, M.J., ed.), Marcel De ker, Inc., NY, pp. 277-338, (1987) .
  • liposomai formulations of the anti- cancer alkaloid vincristine have greater efficacy against L1210 leukemia cells than does free vincristine and have reduced collateral toxicity. Mayer et al., Cancer Chemother. Pharmacol. 33:17-24 (1993) and Mayer et al. , Cancer Res.
  • liposomai formulations of therapeutic agents with clinical and/or pharmaceutical potential depends on the liposome/drug combination possessing both biological efficacy and long-term chemical stability.
  • the efficacy of a liposomai agent can be improved by increasing both the liposome circulation lifetime and the ability of the liposome to retain the encapsulated drug. Mayer, ibid, and Boman et al., Cancer Res. 54: 2830-2833 (1994) . Therefore, much effort has focused on the development of liposomai formulations of therapeutic compounds having both extended circulation times and enhanced drug retention.
  • a wide variety of therapeutic agents can be loaded into liposomes with encapsulation efficiencies approaching 100% by using a transmembrane pH gradient. Mayer et al., Biochim. Biophys. Acta 1025:143-151 (1990) and Madden et al . , Chem. Phvs. Lipids 53: 37-46 (1990).
  • the chemical stability of these formulations i.e., the effective retention of the loaded drugs within the liposomes during circulation .in vivo. frequently requires that the intraliposomal pH be in the range between pH 2.0 to 4.0. Within this pH range however, acid hydrolysis of the acyl component of liposomes can destabilize the liposomai membranes and result in premature leakage of the drug.
  • vincristine can be loaded efficiently into liposomes by a pH gradient-dependent encapsulation procedure which employs an intraliposomal pH of 4.0. Mayer et al., Biochim. Biophys. Acta 1025:143-151 (1990) and Mayer et al.. Cancer Res. 50: 575-579 (1990).
  • the work with liposomai vincristine has been based on vesicles containing phosphatidylcholine (PC) , usually egg PC or distearoyl-PC, and cholesterol. Mayer et al., 1993, supra.
  • PC phosphatidylcholine
  • Liposomai formulations of therapeutic compounds having increased biological and chemical stability are needed in the art.
  • efficacy of liposomai agents may be improved by increasing the liposome circulation time and the ability of the liposome to retain the encapsulated drug
  • development of liposomai formulations having these properties would be valuable additions to clinical treatment regimens.
  • the present invention fulfills these and other related needs.
  • the present invention provides a liposomai composition for delivery of a therapeutic compound to a mammalian host.
  • the composition comprises a liposome having one or more membranes which comprise sphingomyelin and cholesterol, a liposomai interior having a pH less than that of the liposomai exterior, and a therapeutic compound contained in the liposome for delivery to the host.
  • the sphingomyelin and cholesterol are typically present at a molar ratio from 75/25 mol%/mol%, respectively, to 30/50 mol%/mol%, respectively and in a preferred example at a ratio of about 55/45, mol%/mol%, respectively.
  • the lipophilic therapeutic compound may be an alkaloid, such as vincristine, vinblastine, swainsonine, or etoposide or a prodrug thereof.
  • the drug such as vincristine, may be present at a drug to lipid ratio of approximately 0.01/1.0 to 0.2/1.0 (wt/wt) .
  • Swainsonine may be present at a drug to lipid ratio of 0.01:1.1 to 0.5:1.0 (mol:mol).
  • Targeting ligands and other lipids may also be present as components of the liposome so long as they do not adversely affect the stability of the drug and liposome.
  • the liposomes may be unilamellar or multilamellar, and will typically have mean diameters of about 0.05 microns to 0.45 microns, and more preferably about 0.05 microns to 0.2 microns.
  • the interior of the liposome will typically have at a pH of approximately pH 2 to pH 5, e.g., comprising a citrate buffer at about pH 4.
  • the invention provides liposomes for delivery of a therapeutic compound which are produced from a mixture which comprises sphingomyelin and cholesterol in a first buffered aqueous solution having an acidic pH greater than pH 2.
  • the liposome is then suspended in a second buffered solution having a pH which is greater than that of the first buffered aqueous solution, thereby forming a transmembrane pH gradient which facilitates the transfer of the therapeutic compound to the liposome.
  • other passive means of drug entrapment at a low intraliposomal pH can also be used in the process. These alternative processes are typically associated with a less efficient drug entrapment of drug and therefore an additional step of separating the liposome from the second buffer containing free drug may be necessary.
  • the invention also provides methods for enhanced delivery of a lipophilic therapeutic compound such as an alkaloid to a host for treatment.
  • a lipophilic therapeutic compound such as an alkaloid
  • the host in need of the treatment such as a patient suffering from a tumor, is administered the liposomai composition which comprises a liposome having one or more membranes which comprise sphingomyelin and cholesterol, a liposomai interior having a pH less than that of the liposomai exterior, and a therapeutic compound contained in the liposome for delivery to the host or a pharmaceutically acceptable salt thereof.
  • the pH gradient may be generated by a methylammonium or ethanolammonium concentration gradient.
  • the cholesterol will be present in the liposomai composition at a total molar proportion of 30% to 50%, and more preferably the sphingomyelin and cholesterol are present at a ratio of about 75/25 mol%/mol%, respectively to 30/50 mol%/mol%, respectively.
  • the delivery of an alkaloid compound such as vincristine or swainsonine is particularly suitable in these methods.
  • Vincristine and swainsonine may be present at a drug to lipid ratio of approximately 0.01/1.0 to 0.2/1.0 (wt/wt) and 0.01/1.0 to 0.5/1.0 (mol/mol) , respectively.
  • the liposomai composition containing the drug may be administered repeatedly to the host to maintain a concentration of the drug sufficient to inhibit or treat the disease, e.g. , a tumor, but less than an amount which causes unacceptable toxicity to the host.
  • Administration may be by a variety of routes, but the alkaloids are preferably given intravenously or parenterally.
  • Swainsonine is conveniently administered orally.
  • the liposomes administered to the host may be unilamellar, having a mean diameter of 0.05 to 0.45 microns, more preferably from 0.05 to 0.2 microns.
  • the invention also provides methods for delivering to a host an alkaloid immuno odulating compound in a liposomai composition.
  • the host may be suffering from chemotherapy induced immunosuppression and treated, for example, by a liposomai composition of swainsonine.
  • the liposomai composition comprises a liposome having one or more membranes which comprise sphingomyelin and cholesterol, a liposomai interior having a pH less than that of the liposomai exterior, and a therapeutic compound contained in the liposome for delivery to the host or a pharmaceutically acceptable salt thereof.
  • swainsonine is given orally, intravenously, or parenterally.
  • Fig. 1 illustrates the hydrolysis of large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) (O) SM/Chol (55/45, mol/mol) (•) at 37°C in 0.3 M citrate, pH 2.0.
  • Figs. 2A and Fig. 2B illustrate the amount of lipid remaining in circulation in BDF1 mice injected with large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) (O) , SM/Chol (55/45, mol/mol) (•) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) ( ⁇ ) .
  • Injected liposomes were either empty (Fig. 2A) or loaded with vincristine at a drug/1ipid ratio of approximately 0.1 (Fig. 2B) .
  • the injected dose of lipid was 20 mg/kg, corresponding to a total injection of approximately 430 ⁇ g of lipid.
  • Fig. 3 depicts the vincristine/lipid ratio in the plasma of BDF1 mice at various times after the injection of large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) (O) , SM/Chol (55/45, mol/mol) (•) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) ( ⁇ ) .
  • Fig. 4 shows the total vincristine remaining in the plasma of BDF1 mice at various times after the injection of large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) (O) , SM/Chol (55/45, mol/mol) (•) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) ( ⁇ ) .
  • mice were injected with liposomes at a vincristine/lipid ratio of approximately 0.1, corresponding to a lipid dose of 20 mg/kg and a vincristine dose of 2.0 mg/kg. Total amounts injected were approximately 430 ⁇ g of lipid and 43 ⁇ g of vincristine.
  • Fig. 5 shows the uptake of large unilamellar liposomes of SM/Chol (55/45, mol/mol) and DSPC/Chol (55/45, mol/mol) by peritoneal macrophages.
  • Liposomes containing the non-exchangeable and non-metabolized radiolabel 14 C-CHDE were injected parenterally at 100 mg/kg. After 4 hrs, macrophages were recovered by lavage and cells and lipid determined by hemocytometry and liquid scintillation counting, respectively.
  • Fig. 6 depicts the loading of vincristine in P388 tumors. Delivery of vincristine to peritoneal P388 tumors in BDF1 mice after i.v. injection of large unilamellar liposomes of DSPC/Chol (55/45. mol/mol) (O) , SM/Chol (55/45, mol/mol) (•) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) ( ⁇ ) containing vincristine at a drug/lipid ratio of 0.1 (wt/wt). Vincristine was injected at a dose of 20 mg/kg, representing a lipid dose of 20 mg/kg.
  • Figs. 7A-7C show collectively the anti-tumor efficacy of liposomai formulations of vincristine.
  • BDF1 mice containing P388 tumors were injected with large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) ( v) , SM/Chol (55/45, mol/mol) (D) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) ( ⁇ ) containing vincristine at a drug/lipid ratio of 0.1 (wt/wt).
  • Control mice received no injection (•) .
  • Liposome concentrations prior to injection were adjusted to achieve vincristine dosages of 1.0 (Fig. 7A) , 2.0 (Fig. 7B) and 4.0 (Fig. 7C) mg/kg.
  • Fig. 8 shows blood levels of radiolabeled swainsonine in Balb/c mice administered as either a liposomai formulation (L-Im) or as an aqueous solution of the free drug (F-Im) .
  • Formulations were administered orally by gavage (p.o.) r intraperitoneally (i.p.), or intravenously (i.v.). Blood was removed at 1 hr. , 3 hr. , 6 hr. , and 24 hrs. after the dose.
  • Fig. 9 shows the effects of GM-CSF and swainsonine on bone marrow cellularity 14 days after chemotherapeutic administration to C57BL/6 mice.
  • Figs. 10A and 10B illustrate, respectively, TNF production from LPS stimulated splenocytes, and IL-2 production from ConA stimulated splenocytes, collected from C57BL/6 mice 14 days after chemotherapeutic treatment.
  • Figs. 11A and 11B show vincristine levels in (A) plasma and (B) tumors after administration of free and liposomai vincristine in SCID mice bearing A431 tumors. SCID mice bearing two A431 tumors were injected i.v.
  • Vincristine was injected at a dose of 2.0 mg/kg, representing a lipid of dose of 20 mg/kg.
  • Data represent means ( ⁇ standard error) of three mice (6 tumors) ; where standard error bars are not visible, they are smaller than the size of the symbol.
  • Fig. 12 shows antitumor efficacy of free and liposomai vincristine in SCID mice bearing A431 tumors.
  • SCID mice bearing two A431 tumors received no treatment ( ⁇ ) or were injected i.v. with free vincristine (D) or with large unilamellar liposomes of DSPC/Chol (O) or SM/Chol (•) containing vincristine at a drug/lipid ratio of 0.1 (wt/wt).
  • Vincristine was injected at a dose of 2.0 mg/kg, representing a lipid of dose of 20 mg/kg.
  • Data represent the weight of A431 tumors (expressed as the percent of the tumor weight immediately prior to treatment) and are the means (+ standard error) of 8-10 tumors in 4-5 mice.
  • Fig. 13 shows the percent retention over time of swainsonine in liposomes incubated at 37°C at pH 2.
  • Swainsonine was loaded into liposomes at a drug to lipid ratio of 0.2/1.0 (mol/mol) using 0.3 M citrate pH 4 (except SM/Chol at pH 2).
  • EPC egg phosphotidyl choline
  • EPC/Chol egg phosphotidyl choline/cholesterol (55%/45%) (mol/mol)
  • SM/Chol sphingomyelin/cholesterol (55%/45%) (mol/mol) .
  • Fig. 14 shows the percent retention over time of swainsonine in liposomes incubated at 37°C in HEPES buffered saline (HBS) pH 7.5.
  • Fig. 15 shows the percent retention over time of swainsonine in liposomes incubated at 37°C in normal mouse serum.
  • the present invention provides compositions and methods for enhanced delivery of therapeutic compounds to a host.
  • the liposomai formulations of the invention have extended circulation lifetimes and/or enhanced drug retention.
  • the liposomes also referred to as "sphingosomes, " are comprised of sphingomyelin and cholesterol and have an acidic intraliposomal pH.
  • the liposomai formulations based on sphingomyelin and cholesterol have several advantages when compared to other formulations.
  • the sphingomyelin/cholesterol combination produces liposomes which are much more stable to acid hydrolysis, have significantly better drug retention characteristics, have better loading characteristics into tumors and the like, and show significantly better anti-tumor efficacy than other liposomai formulations which were tested.
  • Liposome vesicle will be understood to indicate structures having lipid-containing membranes enclosing an aqueous interior.
  • the structures may have one or more lipid membranes unless otherwise indicated, although generally the liposomes will have only one membrane.
  • Such single-layered liposomes are referred to herein as “unilamellar”.
  • Multilayer liposomes are referred to herein as “multilamellar”.
  • the liposome compositions of the present invention are comprised of sphingomyelin and cholesterol.
  • the ratio of sphingomyelin to cholesterol present in the liposome may vary, but generally is in the range of from 75/25 mol%/mol sphingomyelin/cholesterol to 30/50 mol%/mol% sphingomyelin/cholesterol, more preferably about 70/30 mol%/mol sphingomyelin/cholesterol to 40/45 mol%/mol% sphingomyelin/cholesterol, and most preferably is approximately 55/45 mol%/mol% sphingomyelin/cholesterol.
  • lipids may be present in the formulations as may be necessary, such as to prevent lipid oxidation or to attach ligands onto the liposome surface. Generally the inclusion of other lipids will result in a decrease in the sphingomyelin/ cholesterol ratio.
  • therapeutic compounds may be delivered by the liposomes and methods of the present invention.
  • “Therapeutic compound” is meant to include, e.g., nucleic acids, proteins, peptides, oncolytics, anti- infectives, anxiolytics, psychotropics, immunomodulators, ionotropes, toxins such as gelonin and inhibitors of eucaryotic protein synthesis, and the like.
  • Preferred among the therapeutic compounds for entrapment in the liposomes of the present invention are those which are lipophilic cations.
  • therapeutic agents of the class of lipophilic molecules which are able to partition into the lipid bilayer phase of the liposome, and which therefore are able to associate with the liposomes in a membrane form.
  • Representative drugs include prostaglandins, amphotericin B, methotrexate, cis-platin and derivatives, vincristine, vinblastine, progesterone, testosterone, estradiol, doxorubicin, epirubicin, beclomethasone and esters, vitamin E, cortisone, dexamethasone and esters, betamethasone valerete and other steroids, etc.
  • Particularly preferred therapeutic compounds for use in the present invention are the alkaloid compounds and their derivatives.
  • these are swainsonine and members of the vinca alkaloids and their semisynthetic derivatives, such as, e.g., vinblastine, vincristine, vindesin, etoposide, etoposide phosphate, and teniposide.
  • vinblastine and vincristine, and swainsonine are particularly preferred.
  • Swainsonine (Creaven and Mihich, Semin. Oncol. 4:147 (1977) has the capacity to stimulate bone marrow proliferation (White and Olden, Cancer Commun. 3:83 (1991)).
  • Swainsonine also stimulates the production of multiple cytokines including IL- 1, IL-2, TNF, GM-CSF and interferons (Newton, Cancer Commun. 1:373 (1989); Olden, K. , J. Natl. Cancer Inst.. 83:1149
  • liposomes are prepared which are able to entrap lipophilic cationic drugs in response to transmembrane pH gradients, yet which liposomes are resistant to drug leakage in the circulation.
  • liposomes containing sphingomyelin and cholesterol are prepared according to the desired molar ratio of sphingomyelin and cholesterol, e.g., 55/45 mol./mol., respectively.
  • An appropriate buffer for formation of the liposome, and thus for forming the liposomai interior is one which is physiologically acceptable and having an acid pH, typically about pH 2 to about pH 6, more preferably about pH 3 to pH 5, and most preferably at about pH 4.
  • An example of an appropriate entrapment buffer is citrate buffer, adjusted to approximately pH 4.
  • Other lipids can also be included in the preparation of the liposome.
  • lipids include phospholipids such as phosphatidylcholine, phosphatidylethanolamine, and phosphatidylserine, phosphatidylglycerol, phosphatidic acid, cardiolipin, and phosphatidylinositol, with varying fatty acyl compositions and of natural and/or (semi)synthetic origin, and dicetyl phosphate. Ceramide and various glycolipids, such as cerebrosides or gangliosides, may also be added. Cationic lipids may also be added. Additional lipids which may be suitable for use in the liposomes of the present invention are well known to persons skilled in the art.
  • the liposomes can be sized to a desired size range.
  • the liposomes should generally be less than about 1.0 microns in size, preferably approximately 0.05 to 0.45 microns, more preferably about 0.05 to 0.2 microns, which allows the liposome suspension to be sterilized by filtration.
  • a liposome suspension may be sonicated either by bath or probe down to small vesicles of less than about 0.05 microns in size. Homogenization may also be used to fragment large liposomes into smaller ones. In both methods the particle size distribution can be monitored by conventional laser-beam particle size discrimination or the like.
  • Extrusion of liposomes through a small-pore polycarbonate membrane or an asymmetric ceramic membrane is an effective method for reducing liposome sizes to a relatively well defined size distribution.
  • the suspension is cycled through the membrane one or more times until the desired liposome size distribution is achieved.
  • the liposomes can be extruded through successively smaller pore membranes to achieve a gradual reduction in liposome size.
  • the external pH of the liposome preparation is increased to about pH 7.0 to 7.5, by the addition of suitable buffer, e.g., 0.5 M Na 2 HP0 4 .
  • suitable buffer e.g., 0.5 M Na 2 HP0 4 .
  • the drug or drugs of choice are then admixed with the liposomes at an appropriate concentration, e.g., a vincristine/lipid ratio of 0.01/1.0 to 0.2/1.0 (wt/wt), for a time and under conditions sufficient to allow transmembrane uptake of the drug(s), e.g., from about 5 to 30 min. or more and at about 45-65°C (e.g., 10 min.
  • liposomes and therapeutic compound(s) should generally consist of a relatively uniform population of vesicles in terms of size and drug-lipid ratio. Procedures for passive entrapment of drugs other than the direct formation of pH transmembrane gradients can be used. In one embodiment internal/external concentration gradients are formed employing the charged amines: methylammonium or ethanolammonium. Liposomes are formed in the presence of an aqueous solution of the charged amine.
  • any number of pharmaceutically acceptable salts of the charged amine may be used to prepare the solution such as, but not limited to, fluoride, chloride, citrate, sulfate, phosphate, bromide, iodide, or acetate.
  • external charged amines are diluted or removed by, for example, dilution, filtration, dialysis or gel exclusion.
  • a internal/external pH gradient is thereby generated as uncharged amines leave the liposomai interior and leave behind a proton.
  • the size of the pH gradient will be proportional to the size of the concentration gradient formed.
  • the pH gradient is employed to load the liposome with a drug, such as swainsonine, per methods disclosed herein.
  • the liposomes can be conjugated to monoclonal antibodies or binding fragments thereof that bind to epitopes present only on specific cell types, such as cancer-related antigens, providing a means for targeting the liposomes following systemic administration.
  • ligands that bind surface receptors of the target cell types may also be bound to the liposomes.
  • Other means for targeting liposomes may also be employed in the present invention.
  • aqueous carriers may be used, e.g., water, buffered water, 0.4% saline, 0.3% glycine, and the like, and may include glycoproteins for enhanced stability, such as albumin, lipoprotein, globulin, etc.
  • compositions may be sterilized by conventional liposomai sterilization techniques, such as filtration.
  • the compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, etc.
  • These compositions may be sterilized techniques referred to above or produced under sterile conditions.
  • the resulting aqueous solutions may be packaged for use or filtered under aseptic conditions and lyophilized, the lyophilized preparation being combined with a sterile aqueous solution prior to administration.
  • the concentration of liposomes in the carrier may vary. Generally, the concentration will be about 20-200 mg/ml, usually about 50-150 mg/ml, and most usually about 75- 125 mg/ml, e.g., about 100 mg/ml. Persons of skill may vary these concentrations to optimize treatment with different liposome components or for particular patients. For example, the concentration may be increased to lower the fluid load associated with treatment.
  • the present invention also provides methods for introducing therapeutic compounds into cells of a host.
  • the methods generally comprise administering to the host a liposome containing the therapeutic compound, wherein the liposome has a membrane comprised of sphingomyelin and cholesterol and, optionally, other lipids, and an aqueous interior at a pH substantially below physiologic pH, e.g., pH 3 to about 5, and the therapeutic compound of interest.
  • the host may be a variety of animals, including humans, non-human primates, avian species, equine species, bovine species, swine, lagomorpha, rodents, and the like.
  • the cells of the host are usually exposed to the liposomai preparations of the invention by in vivo administration of the formulations, but ex vivo exposure of the cells to the liposomes is also feasible. In vivo exposure is obtained by administration of the liposomes to host.
  • the liposomes may be administered in many ways. These include parenteral routes of administration, such as intravenous, intramuscular, subcutaneous, and intraarterial. Generally, the liposomes will be administered intravenously or in some cases via inhalation. Often, the liposomes will be administered into a large central vein, such as the superior vena cava or inferior vena cava, to allow highly concentrated solutions to be administered into large volume and flow vessels.
  • the liposomes may be administered intraarterially following vascular procedures to deliver a high concentration directly to an affected vessel.
  • the liposomes may be administered orally or transdermally, although the advantages of the present invention are best realized by parenteral administration.
  • swainsonine is conveniently administered orally, but may be administered parenterally or intravenously.
  • the liposomes may also be incorporated into implantable devices for long duration release following placement.
  • the liposomes will generally be administered intravenously or via inhalation in the methods of the present invention. Often multiple treatments will be given to the patient. The dosage schedule of the treatments will be determined by the disease and the patient's condition.
  • Standard treatments with therapeutic compounds that are well known in the art may serve as a guide to treatment with liposomes containing the therapeutic compounds.
  • the duration and schedule of treatments may be varied by methods well known to those of skill, but the increased circulation time and decreased in liposome leakage will generally allow the dosages to be adjusted downward from those previously employed.
  • the dose of liposomes of the present invention may vary depending on the clinical condition and size of the animal or patient receiving treatment.
  • the standard dose of the therapeutic compound when not encapsulated may serve as a guide to the dose of the liposome-encapsulated compound.
  • the dose will typically be constant over the course of treatment, although in some cases the dose may vary. Standard physiological parameters may be assessed during treatment that may be used to alter the dose of the liposomes of the invention.
  • This Example demonstrates the stability of liposomes prepared with sphingomyelin and cholesterol to acid hydrolysis compared to liposomes prepared with distearoylphosphatidylcholine and cholesterol.
  • DSPC distearoylphosphatidylcholine
  • SM egg sphingomyelin
  • Cholesterol was obtained from Sigma Chemical Company, and PEG-PE was synthesized according to Parr et al., submitted, Biochim. Biophys. Acta (1994). Lipids were dissolved in CHC1 3 , or CHC1 3 with trace amounts of CH 3 OH, then mixed at molar ratios as indicated below and excess solvent removed under a stream of nitrogen gas. Residual solvent was removed from the lipid film under high vacuum for 3 to 16 hrs.
  • Lipids were dispersed by the addition of 0.3 M citrate buffer (pH 4.0 or 2.0) to achieve a final lipid concentration of either 50 or 100 mg/ml. Hydration of the lipid was facilitated by vortexing and heating to 65°C. Equilibration of the solute between the inside and outside of the liposomes was achieved by five freeze/thaw cycles between -196° and 60°C as described generally in Mayer et al. , Biochim. Biophys. Acta 817:193-196 (1985), incorporated herein by reference. Large unilamellar vesicles were produced by repeated extrusion of the multilamellar liposomes through two or three stacked O.l ⁇ m filters (Poretics, Livermore CA) held at 60-65°C in a
  • Themobarrel Extruder (Lipex Biomembranes, Vancouver, Canada) . Liposome size distributions were confirmed by quasi-elastic light scattering using a Nicomp Model 270 Submicron Particle Sizer; these preparations typically had mean diameters of 130 to 150 nm.
  • lipid liposomes of DSPC/Chol or SM/Chol were prepared as described above in 0.3 M citrate buffer at pH 2.0 and were then diluted to 3.2 mg/ml of lipid. The liposomes were incubated at 37 °C for various times then frozen prior to the determination of lipid hydrolysis. Lipid dispersions were thawed then the lipid extracted into CHCI 3 /CH 3 OH and concentrated under a stream of nitrogen gas. Known quantities of lipid were spotted onto K6F thin layer chromatography plates and developed in CHCl 3 /CH 3 /OH/H 2 0/NH 4 OH (65/25/4/0.3, by volume).
  • Lipids were visualized in iodine vapor then the appropriate regions of the plate were recovered and analyzed for phosphorous according to Bartlett, J. Biol. Chem. 234:466-468 (1959), incorporated herein by reference.
  • Total hydrolysis of DSPC was determined from the amount of MSPC present in the samples and then corrected to total hydrolysis; hydrolysis of sphingomyelin was calculated from the difference between the amount of lipid chromotographed and that recovered as non-hydrolyzed sphingomyelin. Calibration curves were determined for each of DSPC, MSPC and sphingomyelin.
  • liposomes composed of SM/Chol were significantly less susceptible to acid hydrolysis than were liposomes composed of DSPC/Chol (55/45, mol/mol) . That is, the rate of hydrolysis at 37°C and pH 2.0 was approximately 100-fold slower in SM/Chol liposomes than in DSPC/Chol liposomes. Similar results were observed during incubation of liposomes at pH 4.0 and at various temperatures between 4°C and 37°C. These results indicate that liposomes composed of SM/Chol were significantly more stable to acid hydrolysis than were identical liposomes composed of DSPC/Chol (Fig. 1) .
  • SM/Chol liposomes are more stable than any formulations based on lipids containing carboxyl-esterified fatty acids (i.e. any phospholipid-based formulations) .
  • Liposomes of DSPC/Chol (55/45) , SM/Chol (55/45) or SM/Chol/PEG-PE (55/40/5) containing the non-exchangeable and non-metabolized radiolabel 14 C-CHDE (cholesteryl-4-hexadecyl ether radiolabeled with 3 H or 14 C, as indicated, obtained from New England Nuclear) were prepared.
  • Empty liposomes or liposomes loaded with 3 H-vincristine (Amersham) were diluted to the indicated concentration with HBS then injected intravenously into BDF1 mice (8-10 weeks old; Charles River) at a vincristine dose of 2 mg/kg (lipid dose of 20 mg/kg) .
  • lipid and vincristine distributions were determined by liquid scintillation counting of known volumes of plasma and 10% homogenates of the tissues.
  • Fig. 2A The clearance of empty liposomes of DSPC/Chol and SM/Chol is shown in Fig. 2A. Liposomes composed of SM/Chol were removed from circulation at a slightly slower rate than were DSPC/Chol liposomes. This difference in clearance rates between DSPC/Chol liposomes and SM/Chol liposomes was also observed in formulations containing vincristine, as shown in Fig. 2B, although the overall clearance rates were slower in the presence of vincristine due to the effect of the drug on RES activity. The amount of SM/Chol remaining in circulation was typically 30-50% higher than for DSPC/Chol liposomes.
  • a further increase in the amount of lipid in circulation was achieved by the addition of 5 mol% PEG-PE to the SM/Chol mixtures; 24 hours after i.v. injection, 200 ⁇ g lipid/ml plasma remained in circulation for SM/Chol/PEG-PE liposomes compared with 100 ⁇ g/ml plasma for SM/Chol liposomes and 65 ⁇ g/ml plasma for DSPC/Chol liposomes (Fig. 2B) .
  • the drug retention characteristics of the liposomes were significantly altered by changes in the lipid composition of the vesicles.
  • Vincristine leakage from DSPC/Chol liposomes was very rapid, with only 50% of the originally encapsulated vincristine remaining entrapped after 4 hours in circulation, as shown in Fig. 3.
  • vincristine leakage from SM/Chol liposomes was much slower, with greater than 60% of the entrapped drug remaining in the liposomes 24 hours after injection (Fig. 3) .
  • Both sphingomyelin-based liposome formulations had identical amounts of vincristine remaining in circulation. This was a consequence of the higher vincristine/lipid ratio in SM/Chol than in SM/Chol/PEG- PE (Fig. 3) and the lower amount of lipid remaining in circulation in SM/Chol than in SM/Chol/PEG-PE (Fig. 2B) .
  • the uptake of liposomes by peritoneal macrophages was measured.
  • SM/Chol liposomes composed of SM/Chol had circulation lifetimes slightly longer than similar DSPC/Chol liposomes, both in the presence and absence of entrapped vincristine (Fig. 2) .
  • SM/Chol liposomes were dramatically better than DSPC/Chol liposomes at retaining vincristine that had been encapsulated using the transmembrane pH gradient method (Fig. 4) .
  • the addition of PEG-PE to SM/Chol liposomes significantly increased the circulation longevity of the liposomes, but PEG- PE also caused a significant increase in the leakage of vincristine from the liposomes.
  • SM/Chol and SM/Chol/PEG-PE liposomai formulations were a consequence of both improved drug retention in SM-containing liposomes (Fig. 3) and the increased circulation longevity of SM/Chol/PEG-PE liposomes (Fig. 2b) .
  • the increased circulation lifetimes of SM/Chol/PEG-PE liposomes were balanced by the lower drug retention by liposomes containing PEG-PE. Therefore, in SM-based liposomai formulations of vincristine, there was no improvement in vincristine circulation longevity by the addition of the lipid PEG-DSPE (Fig. 4) .
  • DSPC/Chol liposomes in P388 tumors had an early peak at 4 hours after liposome injection and was significantly lower at later times.
  • vincristine from formulations of both SM/Chol and SM/Chol/PEG-PE showed sustained delivery of vincristine for up to 24 to 48 hours after liposome injection. That is, SM/Chol and SM/Chol/PEG-PE formulations of vincristine delivered at least 30% more vincristine to P388 tumors than did DSPC/Chol liposomes.
  • BDF1 mice bearing P388 tumors were treated with liposomai formulations of DSPC/Chol (55/45) mol/mol) , SM/Chol (55/45, mol/mol) or SM/Chol/PEG-PE (55/40/5, mol,mol,mol) containing vincristine at a drug/lipid ratio of 0.1 (wt/wt).
  • both DSPC/Chol and SM/Chol/PEG-PE formulations increased survival to 30-31 days.
  • the SM/Chol formulation was significantly more effective; 60% of the mice were surviving at 60 days after administration of the P388 tumor (Fig. 7) .
  • both the DSPC/Chol and SM/Chol/PEG-PE formulations gave 40% of the mice surviving at 60 days after P388 tumor injection.
  • Formulations of SM/Chol were significantly more efficacious; apart from a single vincristine toxicity death, survival of the remaining mice at 60 days was 100% (Fig. 7) .
  • SM/Chol liposomes were significantly better than that of SM/Chol/PEG-PE liposomes (Fig. 7) despite the observation that the loading of vincristine to P388 tumors was identical in these two liposomai formulations (Fig. 6) .
  • This result suggests that the better vincristine retention properties of SM/Chol liposomes in circulation, compared to SM/Chol/PEG-PE liposomes (Fig. 3) , may also occur in the peritoneal cavity and result in improved vincristine uptake by the P388 tumor cells.
  • Formulations of SM/Chol were approximately two-fold more effective than were the formulations based on either DSPC/Chol or SM/Chol/PEG-PE. That is, survival achieved by DSPC/Chol and SM/Chol/PEG-PE formulations at vincristine dosages of 2.0 mg/kg were attained by SM/Chol at a dosage of 1.0 mg/kg. Similarly, the survival obtained by DSPC/Chol and SM/Chol/PEG- PE at a dose of 4.0 mg/kg of vincristine was very similar to that achieved by SM/Chol formulations at 2.0 mg/kg.
  • mice Female Balb/c mice, 5-6 weeks of age, were housed under standard conditions. The animals received free access to both food and water throughout the experiment after a one week acclimatization period prior to experimental manipulation.
  • Swainsonine (Toronto Res. Chem.) was radiolabeled with tritium. Tritiated swainsonine was administered as a lipid-based formulation (L-Im) and as an aqueous formulation containing the free drug (F-Im) .
  • Tritiated swainsonine was loaded into sphingomyelin/cholesterol (Avanti Polar Labs) sphingosomes using a citrate buffer pH 2 gradient at a drug-to-lipid ratio of 0.2:1 (mol:mol) and with an efficiency of loading of 80%.
  • Two hundred microliters of the lipid and aqueous swainsonine formulations were given orally by gavage (p.o.), intraperitoneally (i.p.), or intravenously (i.v.).
  • Fifty microliter blood samples were collected by retroorbital bleeds at 1, 3, 6, and 24 hours after administration. The blood samples were bleached and then counted in a scintillation counter.
  • results were expressed as the percentage of the administered dose in the blood at various time-points after administration.
  • the liposomai formulation (L-Im) of swainsonine has superior bioavailability and achieves higher blood levels when compared to the free aqueous formulation (F-Im) .
  • the oral bioavailability of swainsonine is about 60-65% when compared to swainsonine administered intravenously.
  • mice Female C57BL/6 mice (average weight 15.03 g) , were used and housed under standard conditions. The animals received free access to both food and water throughout the experiment after a one week acclimatization period prior to experimental manipulation. The mice were 6 weeks old on the beginning of the experiment and placed randomly in 12 groups of 5 mice per group. Forty mice were given a single bolus (i.p.) injection of either methotrexate (Mtx, 410 mg/kg) (Sigma Chemical Co.) or Doxorubicin (Dox, 14.9 mk/kg) (Adria Laboratories) . Two days after chemotherapy, administration of swainsonine (2 mg/kg i.p.
  • methotrexate Mtx, 410 mg/kg
  • Doxorubicin Dox, 14.9 mk/kg
  • recombinant murine GM-CSF granulocyte macrophage-colony stimulating factor
  • GM-CSF granulocyte macrophage-colony stimulating factor
  • PBS phosphate buffered saline
  • results in Table I show that when swainsonine is administered for 10 days after a LD 50 dose of chemotherapeutic (Mtx or Dox) , all animals administered swainsonine intravenously or by i.p. handled the cytotoxic insult and survived beyond 2 weeks after chemotherapy. Half of the animals treated with Mtx and half treated with Dox died within a few days after chemotherapy. Animals treated with an i.p. administration of recombinant murine GM-CSF for 10 days did not do as well as with swainsonine; about half of the animals treated with Mtx died within a few days after initiation of the 10 day dosing period.
  • chemotherapeutic Mtx or Dox
  • mice from each group of the immunomodulation study were randomly sacrificed and up to 1 ml of blood was obtained by cardiac puncture. Circulating peripheral WBCs were counted, blood smears (for neutrophil counts) were made and plasma samples were collected and tested for cytokine production (IL-1, IL-2, TNF) . Blood cytokine levels (TNF, IL- 1, and IL-2) were assayed by commercially available assay kits. The spleen, thymus and bone marrow were removed and single cell suspensions were prepared. The cellularity of these lymphoid organs was assessed by trypan blue exclusion test. Fig.
  • FIG. 9 shows the effects of GM-CSF and swainsonine on the bone marrow cellularity at 14 days after chemotherapy drug administration.
  • swainsonine given orally or by i.p. administration performed as effectively as GM-CSF given by i.p. for 10 consecutive days.
  • Cells from spleen, thymus and bone marrow were tested for their ability to respond to stimulation from different mitogens: ConA (1, 2.5 and 5 ⁇ g/ml) (Sigma), phytohemagglutinin (PHA) (1, and 2.5 ⁇ g/ml) (Sigma) and LPS (2.5 and 5 ⁇ g/ml) (Difco) . After 72 hours of stimulation the proliferative response was measured using CellTiter 96
  • IL-2 production using the direct ELISA method.
  • Supernatants from unstimulated cells served as controls. Results are expressed as pg/ml of TNF- ⁇ ? (sensitivity of the assay is ⁇ 25 pg/ml) or IL-2 (sensitivity of the assay is ⁇ 3 pg/ml) .
  • TNF and IL-2 levels in ConA and LPS stimulated splenocytes were significantly elevated in swainsonine-treated animals compared to chemotherapy-treated and PBS (no treatment) controls.
  • swainsonine was incubated .in vitro in simulated gastric contents for various periods of time (1, 2, 4, 24, 48 and 72 hrs.) and the "oral stability" of swainsonine was determined.
  • Swainsonine was also incubated in hydrochloric acid (pH2) containing the main gastric digestive enzyme, pepsin.
  • the j_n vitro stability tests have shown swainsonine to be stable under these harsh conditions for up to 72 hours.
  • DSPC/Chol and SM/Chol liposomai formulations of vincristine were determined in mice bearing solid human A431 squamous cell xenograft tumors. These experiments were undertaken to ensure that the positive results observed in the murine ascitic P388 tumor model were representative of other tumor types.
  • SCID mice bearing 100-200 mg solid human A431 tumors were injected i.v. with free vincristine or with liposomes of either DSPC/Chol or SM/Chol containing vincristine.
  • Vincristine encapsulated DSPC/Chol and SM/Chol liposomes were prepared as in Example II.
  • Encapsulation of vincristine in DSPC/Chol and SM/Chol liposomes increased the amount of vincristine remaining in circulation 24 hours after administration by 28- and 87-fold, respectively, compared with free vincristine (Fig. 11A) .
  • the amount of vincristine remaining in the circulation in SM/Chol liposomes at 24 hours after injection was approximately 3-fold greater than for vincristine encapsulated in DSPC/Chol liposomes (Fig. 11A) .
  • FIG. 11A Improved vincristine circulation longevity correlated with increases in the loading of vincristine in the A431 tumors.
  • free vincristine levels in the A431 tumors were highest (0.856 mg/g tumor) at 0.5 hours after injection and decreased to 0.32 mg/g tumor at 24 hours (Fig. 11B) .
  • Encapsulation of vincristine in DSPC/Chol liposomes increased the amount of vincristine in A431 tumors at 4 to 48 hours after administration to 1.3-1.55 mg/g tumor, respectively (Fig. 11B) .
  • Encapsulation of vincristine in SM/Chol liposomes resulted in a further increase in vincristine delivery to A431 tumors at 24 to 48 hours after injection to 2.8-3.2 mg/g tumor, representing a 2-fold increase in the delivery obtained with DSPC/Chol liposomes.
  • the vincristine/lipid observed in the solid human A431 tumors were very similar to those observed in the plasma.
  • the vincristine/lipid (wt/wt) ratios at 24 hours after injection were 0.022 in the plasma and 0.029 in the tumor
  • the vincristine/lipid ratios were 0.055 in the plasma and 0.050 in the tumor.
  • the antitumor efficacy of free and liposomai vincristine against A431 was closely correlated with vincristine accumulation at the tumor site (Fig. 12) .
  • SCID mice bearing the A431 tumors that received no treatment showed a 100% increase in tumor weight within 4-5 days after treatment was initiated and required termination within 10 days when the tumor exceeded 10% of the total body weight.
  • Tumor bearing SCID mice treated with free vincristine at 2.0 mg/kg had a brief delay in tumor growth (100% increase in tumor weight achieved within 6-8 days) but required termination between 10-12 days.
  • treatment with vincristine encapsulated in DSPC/Chol liposomes resulted in a significant delay in tumor growth (100% increase in tumor weight at 15-20 days, termination at 21 days after treatment) .
  • This therapy was further enhanced by a single treatment of vincristine encapsulated in SM/Chol liposomes.
  • This treatment group a small but consistent decrease in tumor size was observed.
  • several tumors were palpable but unmeasurable and by 33 days after treatment several tumors were not palpable.
  • SM/Chol liposomai vincristine 1 animal was terminated early due to tumor ulceration, not due to tumor growth.
  • histological analysis indicated that all eight tumors were actively dividing squamous cell carcinomas of a mass undetectable by physical examination. Therefore, treatment with SM/Chol liposomai vincristine effected a significant reduction in tumor growth, although none of the original tumors were cured.
  • the present invention demonstrates that liposomai formulations of vincristine and other alkaloids based on sphingomyelin/cholesterol vesicles have several significant advantages over formulations based on DSPC/cholesterol vesicles. Specifically, formulations based on sphingomyelin/cholesterol: (1) are much more stable to acid hydrolysis, (2) have significantly better drug retention characteristics, (3) have better tumor loading characteristics, and (4) show significantly better anti-tumor efficacy than do comparable liposomes composed of DSPC/Chol or SM/Chol/PEG-PE.

Abstract

Liposomal formulations having extended circulation time (in vivo and increased drug retention are comprised of sphingomyelin and cholesterol and have an acidic intraliposomal pH. The formulations have enhanced stability and thus are used in methods which provide improved drug delivery and more effective treatment. The delivery of alkaloid drugs, particularly swainsonine, vincristine and vinblastine, is significantly improved.

Description

SPHINGOSOMES FOR ENHANCED DRUG DELIVERY
Related Application This application is a continuation-in-part of U.S, Serial No. 08/263,603, filed June 20, 1994, which is incorporated herein by reference.
Background of the Invention Liposomai formulations of therapeutically active drugs have significant advantages over drugs injected in free form. einstein, Liposomes: From Biophysics to Therapeutics, (Ostro, M.J., ed.), Marcel De ker, Inc., NY, pp. 277-338, (1987) . For example, liposomai formulations of the anti- cancer alkaloid vincristine have greater efficacy against L1210 leukemia cells than does free vincristine and have reduced collateral toxicity. Mayer et al., Cancer Chemother. Pharmacol. 33:17-24 (1993) and Mayer et al. , Cancer Res.
50:575-579 (1990). The development of liposomai formulations of therapeutic agents with clinical and/or pharmaceutical potential depends on the liposome/drug combination possessing both biological efficacy and long-term chemical stability. In general, the efficacy of a liposomai agent can be improved by increasing both the liposome circulation lifetime and the ability of the liposome to retain the encapsulated drug. Mayer, ibid, and Boman et al., Cancer Res. 54: 2830-2833 (1994) . Therefore, much effort has focused on the development of liposomai formulations of therapeutic compounds having both extended circulation times and enhanced drug retention.
A wide variety of therapeutic agents can be loaded into liposomes with encapsulation efficiencies approaching 100% by using a transmembrane pH gradient. Mayer et al., Biochim. Biophys. Acta 1025:143-151 (1990) and Madden et al . , Chem. Phvs. Lipids 53: 37-46 (1990). The chemical stability of these formulations, i.e., the effective retention of the loaded drugs within the liposomes during circulation .in vivo. frequently requires that the intraliposomal pH be in the range between pH 2.0 to 4.0. Within this pH range however, acid hydrolysis of the acyl component of liposomes can destabilize the liposomai membranes and result in premature leakage of the drug.
For example, vincristine can be loaded efficiently into liposomes by a pH gradient-dependent encapsulation procedure which employs an intraliposomal pH of 4.0. Mayer et al., Biochim. Biophys. Acta 1025:143-151 (1990) and Mayer et al.. Cancer Res. 50: 575-579 (1990). The work with liposomai vincristine has been based on vesicles containing phosphatidylcholine (PC) , usually egg PC or distearoyl-PC, and cholesterol. Mayer et al., 1993, supra. Increased anti-tumor efficacy of liposomai vincristine occurs when the jLn vivo retention of vincristine in the liposomes is increased using a 100-fold larger transmembrane pH gradient (i.e. intraliposomal pH = 2.0). Boman et al., supra. However, at this pH the acid- hydrolysis of the PC component of the liposomes occurs at a significant rate and severely limits the chemical stability of the liposomes. In particular, the fatty acid carboxyl esters at positions sn-1 and sn-2 are especially susceptible to acid- hydrolysis to produce lyso-PC and free fatty acids. Grit et al., Chem. Phvs. Lipids 64:3-18 (1993). Liposomes containing significant proportions of lyso-PC are more permeable to solutes, and therefore would be unsuitable as drug delivery vehicles.
It has been reported that sphingomyelin imparts an increase in the circulation lifetime of liposomes. Allen et al., Biochim. Biophvs. Acta 981:27-35 (1989) and Allen et al., FEBS Lett. 223:42-46 (1987). However, these studies employed an entrapped aqueous solute (125I-tyraminylinulin) as a marker for liposome distribution, and the apparent increase in liposome longevity in the presence of sphingomyelin may have resulted at least in part from increased solute retention by sphingomyelin. There have also been several reports that sphingomyelin-containing liposomes are more toxic than PC- containing liposomes. Weereratne et al., Brit. J. Exp. Pathol■ 64:670-676 (1983), Allen et al. , J. Pharmacol. Exp. Therap. 229:267-275 (1984) , and Allen et al. , Res. Co mun. Chem. Pathol. Pharmacol. 50:281-290 (1985). Although more conclusive studies are not available, the perception is that sphingomyelin-containing liposomes are associated with an increased risk of toxicity.
Liposomai formulations of therapeutic compounds having increased biological and chemical stability are needed in the art. As the efficacy of liposomai agents may be improved by increasing the liposome circulation time and the ability of the liposome to retain the encapsulated drug, the development of liposomai formulations having these properties would be valuable additions to clinical treatment regimens. Quite surprisingly, the present invention fulfills these and other related needs.
Summary of the Invention The present invention provides a liposomai composition for delivery of a therapeutic compound to a mammalian host. The composition comprises a liposome having one or more membranes which comprise sphingomyelin and cholesterol, a liposomai interior having a pH less than that of the liposomai exterior, and a therapeutic compound contained in the liposome for delivery to the host. The sphingomyelin and cholesterol are typically present at a molar ratio from 75/25 mol%/mol%, respectively, to 30/50 mol%/mol%, respectively and in a preferred example at a ratio of about 55/45, mol%/mol%, respectively. The lipophilic therapeutic compound may be an alkaloid, such as vincristine, vinblastine, swainsonine, or etoposide or a prodrug thereof. The drug, such as vincristine, may be present at a drug to lipid ratio of approximately 0.01/1.0 to 0.2/1.0 (wt/wt) . Swainsonine may be present at a drug to lipid ratio of 0.01:1.1 to 0.5:1.0 (mol:mol). Targeting ligands and other lipids may also be present as components of the liposome so long as they do not adversely affect the stability of the drug and liposome. The liposomes may be unilamellar or multilamellar, and will typically have mean diameters of about 0.05 microns to 0.45 microns, and more preferably about 0.05 microns to 0.2 microns. The interior of the liposome will typically have at a pH of approximately pH 2 to pH 5, e.g., comprising a citrate buffer at about pH 4.
In other embodiments the invention provides liposomes for delivery of a therapeutic compound which are produced from a mixture which comprises sphingomyelin and cholesterol in a first buffered aqueous solution having an acidic pH greater than pH 2. The liposome is then suspended in a second buffered solution having a pH which is greater than that of the first buffered aqueous solution, thereby forming a transmembrane pH gradient which facilitates the transfer of the therapeutic compound to the liposome. In some embodiments other passive means of drug entrapment at a low intraliposomal pH can also be used in the process. These alternative processes are typically associated with a less efficient drug entrapment of drug and therefore an additional step of separating the liposome from the second buffer containing free drug may be necessary.
The invention also provides methods for enhanced delivery of a lipophilic therapeutic compound such as an alkaloid to a host for treatment. The host in need of the treatment, such as a patient suffering from a tumor, is administered the liposomai composition which comprises a liposome having one or more membranes which comprise sphingomyelin and cholesterol, a liposomai interior having a pH less than that of the liposomai exterior, and a therapeutic compound contained in the liposome for delivery to the host or a pharmaceutically acceptable salt thereof. The pH gradient may be generated by a methylammonium or ethanolammonium concentration gradient. Typically the cholesterol will be present in the liposomai composition at a total molar proportion of 30% to 50%, and more preferably the sphingomyelin and cholesterol are present at a ratio of about 75/25 mol%/mol%, respectively to 30/50 mol%/mol%, respectively. The delivery of an alkaloid compound such as vincristine or swainsonine is particularly suitable in these methods. Vincristine and swainsonine may be present at a drug to lipid ratio of approximately 0.01/1.0 to 0.2/1.0 (wt/wt) and 0.01/1.0 to 0.5/1.0 (mol/mol) , respectively. In any event, the liposomai composition containing the drug may be administered repeatedly to the host to maintain a concentration of the drug sufficient to inhibit or treat the disease, e.g. , a tumor, but less than an amount which causes unacceptable toxicity to the host. Administration may be by a variety of routes, but the alkaloids are preferably given intravenously or parenterally. Swainsonine is conveniently administered orally. The liposomes administered to the host may be unilamellar, having a mean diameter of 0.05 to 0.45 microns, more preferably from 0.05 to 0.2 microns.
The invention also provides methods for delivering to a host an alkaloid immuno odulating compound in a liposomai composition. The host may be suffering from chemotherapy induced immunosuppression and treated, for example, by a liposomai composition of swainsonine. The liposomai composition comprises a liposome having one or more membranes which comprise sphingomyelin and cholesterol, a liposomai interior having a pH less than that of the liposomai exterior, and a therapeutic compound contained in the liposome for delivery to the host or a pharmaceutically acceptable salt thereof. Preferably, swainsonine is given orally, intravenously, or parenterally.
Brief Description of the Figures
Fig. 1 illustrates the hydrolysis of large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) (O) SM/Chol (55/45, mol/mol) (•) at 37°C in 0.3 M citrate, pH 2.0. Figs. 2A and Fig. 2B illustrate the amount of lipid remaining in circulation in BDF1 mice injected with large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) (O) , SM/Chol (55/45, mol/mol) (•) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) (■) . Injected liposomes were either empty (Fig. 2A) or loaded with vincristine at a drug/1ipid ratio of approximately 0.1 (Fig. 2B) . The injected dose of lipid was 20 mg/kg, corresponding to a total injection of approximately 430 μg of lipid. Fig. 3 depicts the vincristine/lipid ratio in the plasma of BDF1 mice at various times after the injection of large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) (O) , SM/Chol (55/45, mol/mol) (•) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) (■) . Mice were injected with liposomes at a vincristine/lipid ratio of approximately 0.1, corresponding to a lipid dose of 20 mg/kg and a vincristine dose of 2.0 mg/kg. Total amounts injected were approximately 430 μg of lipid and 43 μg of vincristine. Fig. 4 shows the total vincristine remaining in the plasma of BDF1 mice at various times after the injection of large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) (O) , SM/Chol (55/45, mol/mol) (•) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) (■) . Mice were injected with liposomes at a vincristine/lipid ratio of approximately 0.1, corresponding to a lipid dose of 20 mg/kg and a vincristine dose of 2.0 mg/kg. Total amounts injected were approximately 430 μg of lipid and 43 μg of vincristine.
Fig. 5 shows the uptake of large unilamellar liposomes of SM/Chol (55/45, mol/mol) and DSPC/Chol (55/45, mol/mol) by peritoneal macrophages. Liposomes containing the non-exchangeable and non-metabolized radiolabel 14C-CHDE were injected parenterally at 100 mg/kg. After 4 hrs, macrophages were recovered by lavage and cells and lipid determined by hemocytometry and liquid scintillation counting, respectively.
Fig. 6 depicts the loading of vincristine in P388 tumors. Delivery of vincristine to peritoneal P388 tumors in BDF1 mice after i.v. injection of large unilamellar liposomes of DSPC/Chol (55/45. mol/mol) (O) , SM/Chol (55/45, mol/mol) (•) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) (■) containing vincristine at a drug/lipid ratio of 0.1 (wt/wt). Vincristine was injected at a dose of 20 mg/kg, representing a lipid dose of 20 mg/kg.
Figs. 7A-7C show collectively the anti-tumor efficacy of liposomai formulations of vincristine. BDF1 mice containing P388 tumors were injected with large unilamellar liposomes of DSPC/Chol (55/45, mol/mol) ( v) , SM/Chol (55/45, mol/mol) (D) or SM/Chol/PEG-PE (55/40/5, mol/mol/mol) (Δ ) containing vincristine at a drug/lipid ratio of 0.1 (wt/wt). Control mice received no injection (•) . Liposome concentrations prior to injection were adjusted to achieve vincristine dosages of 1.0 (Fig. 7A) , 2.0 (Fig. 7B) and 4.0 (Fig. 7C) mg/kg.
Fig. 8 shows blood levels of radiolabeled swainsonine in Balb/c mice administered as either a liposomai formulation (L-Im) or as an aqueous solution of the free drug (F-Im) . Formulations were administered orally by gavage (p.o.)r intraperitoneally (i.p.), or intravenously (i.v.). Blood was removed at 1 hr. , 3 hr. , 6 hr. , and 24 hrs. after the dose.
Fig. 9 shows the effects of GM-CSF and swainsonine on bone marrow cellularity 14 days after chemotherapeutic administration to C57BL/6 mice.
Figs. 10A and 10B illustrate, respectively, TNF production from LPS stimulated splenocytes, and IL-2 production from ConA stimulated splenocytes, collected from C57BL/6 mice 14 days after chemotherapeutic treatment. Figs. 11A and 11B show vincristine levels in (A) plasma and (B) tumors after administration of free and liposomai vincristine in SCID mice bearing A431 tumors. SCID mice bearing two A431 tumors were injected i.v. with free vincristine (D) or with large unilamellar liposomes of DSPC/Chol (O) or SM/Chol (•) containing vincristine at a drug/lipid ratio of 0.1 (wt/wt). Vincristine was injected at a dose of 2.0 mg/kg, representing a lipid of dose of 20 mg/kg. Data represent means (± standard error) of three mice (6 tumors) ; where standard error bars are not visible, they are smaller than the size of the symbol.
Fig. 12 shows antitumor efficacy of free and liposomai vincristine in SCID mice bearing A431 tumors. SCID mice bearing two A431 tumors received no treatment (■) or were injected i.v. with free vincristine (D) or with large unilamellar liposomes of DSPC/Chol (O) or SM/Chol (•) containing vincristine at a drug/lipid ratio of 0.1 (wt/wt). Vincristine was injected at a dose of 2.0 mg/kg, representing a lipid of dose of 20 mg/kg. Data represent the weight of A431 tumors (expressed as the percent of the tumor weight immediately prior to treatment) and are the means (+ standard error) of 8-10 tumors in 4-5 mice.
Fig. 13 shows the percent retention over time of swainsonine in liposomes incubated at 37°C at pH 2.
Swainsonine was loaded into liposomes at a drug to lipid ratio of 0.2/1.0 (mol/mol) using 0.3 M citrate pH 4 (except SM/Chol at pH 2). EPC, egg phosphotidyl choline; EPC/Chol, egg phosphotidyl choline/cholesterol (55%/45%) (mol/mol) ; SM/Chol, sphingomyelin/cholesterol (55%/45%) (mol/mol) .
Fig. 14 shows the percent retention over time of swainsonine in liposomes incubated at 37°C in HEPES buffered saline (HBS) pH 7.5.
Fig. 15 shows the percent retention over time of swainsonine in liposomes incubated at 37°C in normal mouse serum.
Description of Specific Embodiments The present invention provides compositions and methods for enhanced delivery of therapeutic compounds to a host. The liposomai formulations of the invention have extended circulation lifetimes and/or enhanced drug retention. The liposomes, also referred to as "sphingosomes, " are comprised of sphingomyelin and cholesterol and have an acidic intraliposomal pH. The liposomai formulations based on sphingomyelin and cholesterol have several advantages when compared to other formulations. The sphingomyelin/cholesterol combination produces liposomes which are much more stable to acid hydrolysis, have significantly better drug retention characteristics, have better loading characteristics into tumors and the like, and show significantly better anti-tumor efficacy than other liposomai formulations which were tested.
"Liposome", "vesicle" and "liposome vesicle" will be understood to indicate structures having lipid-containing membranes enclosing an aqueous interior. The structures may have one or more lipid membranes unless otherwise indicated, although generally the liposomes will have only one membrane. Such single-layered liposomes are referred to herein as "unilamellar". Multilayer liposomes are referred to herein as "multilamellar".
The liposome compositions of the present invention are comprised of sphingomyelin and cholesterol. The ratio of sphingomyelin to cholesterol present in the liposome may vary, but generally is in the range of from 75/25 mol%/mol sphingomyelin/cholesterol to 30/50 mol%/mol% sphingomyelin/cholesterol, more preferably about 70/30 mol%/mol sphingomyelin/cholesterol to 40/45 mol%/mol% sphingomyelin/cholesterol, and most preferably is approximately 55/45 mol%/mol% sphingomyelin/cholesterol. Other lipids may be present in the formulations as may be necessary, such as to prevent lipid oxidation or to attach ligands onto the liposome surface. Generally the inclusion of other lipids will result in a decrease in the sphingomyelin/ cholesterol ratio.
A wide variety of therapeutic compounds may be delivered by the liposomes and methods of the present invention. "Therapeutic compound" is meant to include, e.g., nucleic acids, proteins, peptides, oncolytics, anti- infectives, anxiolytics, psychotropics, immunomodulators, ionotropes, toxins such as gelonin and inhibitors of eucaryotic protein synthesis, and the like. Preferred among the therapeutic compounds for entrapment in the liposomes of the present invention are those which are lipophilic cations. Among these are therapeutic agents of the class of lipophilic molecules which are able to partition into the lipid bilayer phase of the liposome, and which therefore are able to associate with the liposomes in a membrane form. Representative drugs include prostaglandins, amphotericin B, methotrexate, cis-platin and derivatives, vincristine, vinblastine, progesterone, testosterone, estradiol, doxorubicin, epirubicin, beclomethasone and esters, vitamin E, cortisone, dexamethasone and esters, betamethasone valerete and other steroids, etc.
Particularly preferred therapeutic compounds for use in the present invention are the alkaloid compounds and their derivatives. Among these are swainsonine and members of the vinca alkaloids and their semisynthetic derivatives, such as, e.g., vinblastine, vincristine, vindesin, etoposide, etoposide phosphate, and teniposide. Among this group, vinblastine and vincristine, and swainsonine are particularly preferred. Swainsonine (Creaven and Mihich, Semin. Oncol. 4:147 (1977) has the capacity to stimulate bone marrow proliferation (White and Olden, Cancer Commun. 3:83 (1991)). Swainsonine also stimulates the production of multiple cytokines including IL- 1, IL-2, TNF, GM-CSF and interferons (Newton, Cancer Commun. 1:373 (1989); Olden, K. , J. Natl. Cancer Inst.. 83:1149
(1991)). It also reportedly induces B- and T-cell immunity, natural killer T-cell and macrophage-induced destruction of tumor cells in vitro and, when combined with interferon, has direct anti-tumor activity against colon cancer and melanoma cancers in vivo (Dennis, J., Cancer Res.. 50:1867 (1990); Olden, K. , Pharm. Ther. 44:85 (1989); White and Olden, Anticancer Res., 10:1515 (1990)). Other alkaloids useful in the present invention include paclitaxel (taxol) and synthetic derivatives thereof. A representative method for producing the liposomes of the invention is now described, although it will be understood that the procedure can be subjected to modifications in various aspects without affecting the outcome. As described more fully below in the experimental section, liposomes are prepared which are able to entrap lipophilic cationic drugs in response to transmembrane pH gradients, yet which liposomes are resistant to drug leakage in the circulation. Initially, liposomes containing sphingomyelin and cholesterol are prepared according to the desired molar ratio of sphingomyelin and cholesterol, e.g., 55/45 mol./mol., respectively. An appropriate buffer for formation of the liposome, and thus for forming the liposomai interior, is one which is physiologically acceptable and having an acid pH, typically about pH 2 to about pH 6, more preferably about pH 3 to pH 5, and most preferably at about pH 4. An example of an appropriate entrapment buffer is citrate buffer, adjusted to approximately pH 4. Other lipids can also be included in the preparation of the liposome. These lipids include phospholipids such as phosphatidylcholine, phosphatidylethanolamine, and phosphatidylserine, phosphatidylglycerol, phosphatidic acid, cardiolipin, and phosphatidylinositol, with varying fatty acyl compositions and of natural and/or (semi)synthetic origin, and dicetyl phosphate. Ceramide and various glycolipids, such as cerebrosides or gangliosides, may also be added. Cationic lipids may also be added. Additional lipids which may be suitable for use in the liposomes of the present invention are well known to persons skilled in the art.
Once the liposomes are prepared with the entrapped acidic buffer the liposomes can be sized to a desired size range. The liposomes should generally be less than about 1.0 microns in size, preferably approximately 0.05 to 0.45 microns, more preferably about 0.05 to 0.2 microns, which allows the liposome suspension to be sterilized by filtration. For sizing liposomes, a liposome suspension may be sonicated either by bath or probe down to small vesicles of less than about 0.05 microns in size. Homogenization may also be used to fragment large liposomes into smaller ones. In both methods the particle size distribution can be monitored by conventional laser-beam particle size discrimination or the like. Extrusion of liposomes through a small-pore polycarbonate membrane or an asymmetric ceramic membrane is an effective method for reducing liposome sizes to a relatively well defined size distribution. Typically the suspension is cycled through the membrane one or more times until the desired liposome size distribution is achieved. The liposomes can be extruded through successively smaller pore membranes to achieve a gradual reduction in liposome size.
Before or after sizing, the external pH of the liposome preparation is increased to about pH 7.0 to 7.5, by the addition of suitable buffer, e.g., 0.5 M Na2HP04. The drug or drugs of choice are then admixed with the liposomes at an appropriate concentration, e.g., a vincristine/lipid ratio of 0.01/1.0 to 0.2/1.0 (wt/wt), for a time and under conditions sufficient to allow transmembrane uptake of the drug(s), e.g., from about 5 to 30 min. or more and at about 45-65°C (e.g., 10 min. at 60°C in the case of the liposomai vincristine preparations described in the Examples below) , although one of ordinary skill in the art will understand that the conditions may be adjusted and uptake monitored accordingly. The formulation of liposomes and therapeutic compound(s) should generally consist of a relatively uniform population of vesicles in terms of size and drug-lipid ratio. Procedures for passive entrapment of drugs other than the direct formation of pH transmembrane gradients can be used. In one embodiment internal/external concentration gradients are formed employing the charged amines: methylammonium or ethanolammonium. Liposomes are formed in the presence of an aqueous solution of the charged amine. Any number of pharmaceutically acceptable salts of the charged amine may be used to prepare the solution such as, but not limited to, fluoride, chloride, citrate, sulfate, phosphate, bromide, iodide, or acetate. After formation of the liposome, external charged amines are diluted or removed by, for example, dilution, filtration, dialysis or gel exclusion. A internal/external pH gradient is thereby generated as uncharged amines leave the liposomai interior and leave behind a proton. The size of the pH gradient will be proportional to the size of the concentration gradient formed. The pH gradient is employed to load the liposome with a drug, such as swainsonine, per methods disclosed herein.
Additional components may be added to the liposomes to target them to specific cell types. For example, the liposomes can be conjugated to monoclonal antibodies or binding fragments thereof that bind to epitopes present only on specific cell types, such as cancer-related antigens, providing a means for targeting the liposomes following systemic administration. Alternatively, ligands that bind surface receptors of the target cell types may also be bound to the liposomes. Other means for targeting liposomes may also be employed in the present invention. Following a separation step as may be necessary to remove free drug from the medium containing the liposome, the liposome suspension is brought to a desired concentration in a pharmaceutically acceptable carrier for administration to the patient or host cells. Many pharmaceutically acceptable carriers may be employed in the compositions and methods of the present invention. A variety of aqueous carriers may be used, e.g., water, buffered water, 0.4% saline, 0.3% glycine, and the like, and may include glycoproteins for enhanced stability, such as albumin, lipoprotein, globulin, etc.
Generally, normal buffered saline (135-150 mM NaCl) will be employed as the pharmaceutically acceptable carrier, but other suitable carriers will suffice. These compositions may be sterilized by conventional liposomai sterilization techniques, such as filtration. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, etc. These compositions may be sterilized techniques referred to above or produced under sterile conditions. The resulting aqueous solutions may be packaged for use or filtered under aseptic conditions and lyophilized, the lyophilized preparation being combined with a sterile aqueous solution prior to administration.
The concentration of liposomes in the carrier may vary. Generally, the concentration will be about 20-200 mg/ml, usually about 50-150 mg/ml, and most usually about 75- 125 mg/ml, e.g., about 100 mg/ml. Persons of skill may vary these concentrations to optimize treatment with different liposome components or for particular patients. For example, the concentration may be increased to lower the fluid load associated with treatment.
The present invention also provides methods for introducing therapeutic compounds into cells of a host. The methods generally comprise administering to the host a liposome containing the therapeutic compound, wherein the liposome has a membrane comprised of sphingomyelin and cholesterol and, optionally, other lipids, and an aqueous interior at a pH substantially below physiologic pH, e.g., pH 3 to about 5, and the therapeutic compound of interest. The host may be a variety of animals, including humans, non-human primates, avian species, equine species, bovine species, swine, lagomorpha, rodents, and the like.
The cells of the host are usually exposed to the liposomai preparations of the invention by in vivo administration of the formulations, but ex vivo exposure of the cells to the liposomes is also feasible. In vivo exposure is obtained by administration of the liposomes to host. The liposomes may be administered in many ways. These include parenteral routes of administration, such as intravenous, intramuscular, subcutaneous, and intraarterial. Generally, the liposomes will be administered intravenously or in some cases via inhalation. Often, the liposomes will be administered into a large central vein, such as the superior vena cava or inferior vena cava, to allow highly concentrated solutions to be administered into large volume and flow vessels. The liposomes may be administered intraarterially following vascular procedures to deliver a high concentration directly to an affected vessel. In some instances, the liposomes may be administered orally or transdermally, although the advantages of the present invention are best realized by parenteral administration. For example, swainsonine is conveniently administered orally, but may be administered parenterally or intravenously. The liposomes may also be incorporated into implantable devices for long duration release following placement. As described above, the liposomes will generally be administered intravenously or via inhalation in the methods of the present invention. Often multiple treatments will be given to the patient. The dosage schedule of the treatments will be determined by the disease and the patient's condition. Standard treatments with therapeutic compounds that are well known in the art may serve as a guide to treatment with liposomes containing the therapeutic compounds. The duration and schedule of treatments may be varied by methods well known to those of skill, but the increased circulation time and decreased in liposome leakage will generally allow the dosages to be adjusted downward from those previously employed. The dose of liposomes of the present invention may vary depending on the clinical condition and size of the animal or patient receiving treatment. The standard dose of the therapeutic compound when not encapsulated may serve as a guide to the dose of the liposome-encapsulated compound. The dose will typically be constant over the course of treatment, although in some cases the dose may vary. Standard physiological parameters may be assessed during treatment that may be used to alter the dose of the liposomes of the invention.
The following examples are offered by way of illustration and not limitation.
EXAMPLE I Acid Stability of DSPC/Chol vs. SM/Chol Liposomes
This Example demonstrates the stability of liposomes prepared with sphingomyelin and cholesterol to acid hydrolysis compared to liposomes prepared with distearoylphosphatidylcholine and cholesterol.
For liposome preparation, distearoylphosphatidylcholine (DSPC) and egg sphingomyelin (SM) were obtained from Avanti Polar Lipids and used without further purification. Cholesterol was obtained from Sigma Chemical Company, and PEG-PE was synthesized according to Parr et al., submitted, Biochim. Biophys. Acta (1994). Lipids were dissolved in CHC13, or CHC13 with trace amounts of CH3OH, then mixed at molar ratios as indicated below and excess solvent removed under a stream of nitrogen gas. Residual solvent was removed from the lipid film under high vacuum for 3 to 16 hrs. Lipids were dispersed by the addition of 0.3 M citrate buffer (pH 4.0 or 2.0) to achieve a final lipid concentration of either 50 or 100 mg/ml. Hydration of the lipid was facilitated by vortexing and heating to 65°C. Equilibration of the solute between the inside and outside of the liposomes was achieved by five freeze/thaw cycles between -196° and 60°C as described generally in Mayer et al. , Biochim. Biophys. Acta 817:193-196 (1985), incorporated herein by reference. Large unilamellar vesicles were produced by repeated extrusion of the multilamellar liposomes through two or three stacked O.lμm filters (Poretics, Livermore CA) held at 60-65°C in a
Themobarrel Extruder (Lipex Biomembranes, Vancouver, Canada) . Liposome size distributions were confirmed by quasi-elastic light scattering using a Nicomp Model 270 Submicron Particle Sizer; these preparations typically had mean diameters of 130 to 150 nm.
Large unilamellar liposomes of DSPC/Chol or SM/Chol were prepared as described above in 0.3 M citrate buffer at pH 2.0 and were then diluted to 3.2 mg/ml of lipid. The liposomes were incubated at 37 °C for various times then frozen prior to the determination of lipid hydrolysis. Lipid dispersions were thawed then the lipid extracted into CHCI3/CH3OH and concentrated under a stream of nitrogen gas. Known quantities of lipid were spotted onto K6F thin layer chromatography plates and developed in CHCl3/CH3/OH/H20/NH4OH (65/25/4/0.3, by volume). Lipids were visualized in iodine vapor then the appropriate regions of the plate were recovered and analyzed for phosphorous according to Bartlett, J. Biol. Chem. 234:466-468 (1959), incorporated herein by reference. Total hydrolysis of DSPC was determined from the amount of MSPC present in the samples and then corrected to total hydrolysis; hydrolysis of sphingomyelin was calculated from the difference between the amount of lipid chromotographed and that recovered as non-hydrolyzed sphingomyelin. Calibration curves were determined for each of DSPC, MSPC and sphingomyelin.
As shown in Fig. 1, liposomes composed of SM/Chol (55/45, mol/mol) were significantly less susceptible to acid hydrolysis than were liposomes composed of DSPC/Chol (55/45, mol/mol) . That is, the rate of hydrolysis at 37°C and pH 2.0 was approximately 100-fold slower in SM/Chol liposomes than in DSPC/Chol liposomes. Similar results were observed during incubation of liposomes at pH 4.0 and at various temperatures between 4°C and 37°C. These results indicate that liposomes composed of SM/Chol were significantly more stable to acid hydrolysis than were identical liposomes composed of DSPC/Chol (Fig. 1) . As the primary degradation product in DSPC/Chol liposomes is the lyso-PC (MSPC) , it is very likely that SM/Chol liposomes are more stable than any formulations based on lipids containing carboxyl-esterified fatty acids (i.e. any phospholipid-based formulations) .
EXAMPLE II
Lipid and Drug Pharmacokinetics
Uptake of vincristine into large unilamellar liposomes was achieved using a pH gradient-dependent procedure described by Mayer et al., Cancer Chemother. Pharmacol. 33:17- 24 (1993) , incorporated herein by reference. Briefly, a solution of vincristine sulfate (Oncovin®, Eli Lilly, Indianapolis, IN) was added to liposomes at a drug/lipid ratio of 0.1/1 (wt/wt) and equilibrated at 60°C for 5 to 10 minutes. Vincristine uptake in response to a transmembrane pH gradient was initiated by the addition of 0.5 M Na2HP04 to bring the external pH to 7.2-7.6. Uptake was allowed to proceed for 10 minutes at 60°C and typically had a trapping efficiency of approximately 95% (Mayer et al., Cancer Chemother. Pharmacol. 33:17-24 (1993)).
Liposomes of DSPC/Chol (55/45) , SM/Chol (55/45) or SM/Chol/PEG-PE (55/40/5) containing the non-exchangeable and non-metabolized radiolabel 14C-CHDE (cholesteryl-4-hexadecyl ether radiolabeled with 3H or 14C, as indicated, obtained from New England Nuclear) were prepared. Empty liposomes or liposomes loaded with 3H-vincristine (Amersham) were diluted to the indicated concentration with HBS then injected intravenously into BDF1 mice (8-10 weeks old; Charles River) at a vincristine dose of 2 mg/kg (lipid dose of 20 mg/kg) . At various times following the liposome injection, blood was obtained by heart puncture and liver, spleen and muscle recovered. In all cases, lipid and vincristine distributions were determined by liquid scintillation counting of known volumes of plasma and 10% homogenates of the tissues.
The clearance of empty liposomes of DSPC/Chol and SM/Chol is shown in Fig. 2A. Liposomes composed of SM/Chol were removed from circulation at a slightly slower rate than were DSPC/Chol liposomes. This difference in clearance rates between DSPC/Chol liposomes and SM/Chol liposomes was also observed in formulations containing vincristine, as shown in Fig. 2B, although the overall clearance rates were slower in the presence of vincristine due to the effect of the drug on RES activity. The amount of SM/Chol remaining in circulation was typically 30-50% higher than for DSPC/Chol liposomes. A further increase in the amount of lipid in circulation was achieved by the addition of 5 mol% PEG-PE to the SM/Chol mixtures; 24 hours after i.v. injection, 200 μg lipid/ml plasma remained in circulation for SM/Chol/PEG-PE liposomes compared with 100 μg/ml plasma for SM/Chol liposomes and 65 μg/ml plasma for DSPC/Chol liposomes (Fig. 2B) .
The drug retention characteristics of the liposomes were significantly altered by changes in the lipid composition of the vesicles. Vincristine leakage from DSPC/Chol liposomes was very rapid, with only 50% of the originally encapsulated vincristine remaining entrapped after 4 hours in circulation, as shown in Fig. 3. In contrast, vincristine leakage from SM/Chol liposomes was much slower, with greater than 60% of the entrapped drug remaining in the liposomes 24 hours after injection (Fig. 3) . Furthermore, additional increases in the retention of vincristine in SM/Chol liposomes were not observed in the presence of a two-fold greater transmembrane pH gradient (i.e., pHi=2.0). The presence of 5 mol% PEG-PE in SM/Chol liposomes caused a significant increase in the permeability of vincristine; approximately 30% of the entrapped vincristine remained in the liposomes after 24 hours in circulation, as shown in Fig. 3. Anti-tumor efficacy of liposomai vincristine is a function of the amount of the drug remaining in circulation and, therefore, is a consequence of both liposome longevity in circulation and drug retention within the liposomes. The total amount of vincristine remaining in circulation was significantly lower in the liposomai DSPC/Chol formulations than in either the liposomai SM/Chol or SM/Chol/PEG-PE formulations, as shown in Fig. 4. Both sphingomyelin-based liposome formulations had identical amounts of vincristine remaining in circulation. This was a consequence of the higher vincristine/lipid ratio in SM/Chol than in SM/Chol/PEG- PE (Fig. 3) and the lower amount of lipid remaining in circulation in SM/Chol than in SM/Chol/PEG-PE (Fig. 2B) . To determine whether the extended circulation lifetime of SM/Chol liposomes was a consequence of reduced uptake of the SM/Chol liposomes by macrophages, the uptake of liposomes by peritoneal macrophages was measured. Empty DSPC/Chol and SM/Chol liposomes containing 14C-CHDE were prepared as described above and the external pH brought to 7.2 to 7.6 with 0.5 M Na2HP04. Liposomes were injected i.p. into CD1 mice (8-10 weeks old) (Charles River) at 100 mg lipid/kg in a volume of 0.5 ml. After 4 hrs, peritoneal macrophages were recovered by lavage, purified by repeated centrifugation and then macrophages counted with a hemocytometer and the amount of lipid taken up by the macrophages was determined by liquid scintillation counting.
For serum protein binding assays, 10 mg of either DSPC/Chol or SM/Chol liposomes labelled with 14C-CHDE were brought to external pH of 7.2-7.6, then diluted to 20 mg/ml with HBS. Liposomes were incubated with 500 μl of fetal bovine serum (ICN Biomedicals) (pre-filtered through a 0.22 μm filter) for 30 mins at 37°C. Serum protein that was not bound to the liposomes was removed by passing the sample over a 1 cm (internal diameter) x 18 cm BioGel A-15m column (Bio-Rad
Laboratories) (in HBS) at 35 ml/hr. Fractions (1 ml) were assayed for protein (Sigma bicinchoninic acid protein assay kit) and lipid (LSC) and the adsorbed protein was calculated after correction for co-eluting serum protein. The uptake of i.p.-injected SM/Chol liposomes into macrophages was 50% lower than the uptake of DSPC/Chol liposomes, as shown in Fig. 5. It is likely that the reduced uptake of SM/Chol liposomes by macrophages and their extended circulation longevity was a consequence of lowered protein opsonization to the surface of SM/Chol liposomes than to DSPC/Chol liposomes. Measurement of the adsorption of fetal bovine serum proteins to SM/Chol and DSPC/Chol liposomes indicated that the DSPC/Chol liposomes adsorbed 13.7 μg protein/mg lipid. In contrast, significant adsorption of fetal bovine serum proteins to SM/Chol liposomes was not detected.
Thus, from this Example it can be seen that liposomes composed of SM/Chol had circulation lifetimes slightly longer than similar DSPC/Chol liposomes, both in the presence and absence of entrapped vincristine (Fig. 2) . SM/Chol liposomes were dramatically better than DSPC/Chol liposomes at retaining vincristine that had been encapsulated using the transmembrane pH gradient method (Fig. 4) . The addition of PEG-PE to SM/Chol liposomes significantly increased the circulation longevity of the liposomes, but PEG- PE also caused a significant increase in the leakage of vincristine from the liposomes. The increased levels of vincristine remaining in circulation in SM/Chol and SM/Chol/PEG-PE liposomai formulations (Fig. 4) was a consequence of both improved drug retention in SM-containing liposomes (Fig. 3) and the increased circulation longevity of SM/Chol/PEG-PE liposomes (Fig. 2b) . However, the increased circulation lifetimes of SM/Chol/PEG-PE liposomes were balanced by the lower drug retention by liposomes containing PEG-PE. Therefore, in SM-based liposomai formulations of vincristine, there was no improvement in vincristine circulation longevity by the addition of the lipid PEG-DSPE (Fig. 4) . Furthermore, since there was no improvement in vincristine retention _Ln vivo by the use of a pHi=2.0, the optimal vincristine retention in circulation was achieved with a relatively simple liposomai formulation comprised of only sphingomyelin, cholesterol and citrate buffer (pH 4.0). EXAMPLE III Tumor Loading Of Liposomai Vincristine
To determine whether increased vincristine longevity in circulation, as shown in Fig. 4, resulted in increased drug delivery to tumors, the loading of liposomai vincristine into P388 tumors was examined. For tumor loading experiments, BDF1 mice were injected i.p. with 106 P388 cells (obtained from National Cancer Institute, Bethesda, MD) (passaged weekly in BDF1 mice) 24 hrs prior to the liposome injection. At various times following the liposome injection the tumor was recovered by peritoneal lavage. In all cases, lipid and vincristine distributions were determined by liquid scintillation counting of known volumes of lavage. As shown in Fig. 6, accumulation of vincristine from
DSPC/Chol liposomes in P388 tumors had an early peak at 4 hours after liposome injection and was significantly lower at later times. In contrast, vincristine from formulations of both SM/Chol and SM/Chol/PEG-PE showed sustained delivery of vincristine for up to 24 to 48 hours after liposome injection. That is, SM/Chol and SM/Chol/PEG-PE formulations of vincristine delivered at least 30% more vincristine to P388 tumors than did DSPC/Chol liposomes.
The increased levels of vincristine remaining in circulation in the plasma using SM-based liposomai formulations (Fig. 4) was reflected in greater amounts of vincristine loaded to P388 tumors (Fig. 6) . This relationship suggests, for P388 tumors in BDF1 mice, that liposomes containing DSPC, SM and/or PEG-PE are not significantly different in their ability to extravasate from circulation to the peritoneal tumor.
EXAMPLE IV In Vivo Efficacy of Liposomai Vincristine Against P388 Tumors
To determine whether increased delivery of vincristine to P388 tumors by SM/Chol and SM/Chol/PEG-PE liposomes, as shown in Example III, resulted in increased anti-tumor activity, the efficacy of liposomai formulations of vincristine was determined.
BDF1 mice bearing P388 tumors were treated with liposomai formulations of DSPC/Chol (55/45) mol/mol) , SM/Chol (55/45, mol/mol) or SM/Chol/PEG-PE (55/40/5, mol,mol,mol) containing vincristine at a drug/lipid ratio of 0.1 (wt/wt).
Large unilamellar liposomes of DSPC/Chol (55/45) , SM/Chol (55/45) and SM/Chol/PEG-PE (55/40/5) were prepared as described above and loaded with vincristine at a vincristine/lipid ratio of 0.1/1 (wt/wt). Liposomai vincristine was injected i.v. into BDF1 mice that had been administered 24 hours earlier with an i.p. injection of 106P388 cells. Liposome concentration was adjusted to achieve vincristine dosages of 1.0, 2.0 and 4.0 mg/kg, then animal weights and survival was followed during the subsequent 60 days. Animals surviving for 60 days were re-injected with 106P388 cells to evaluate the immune component of long-term survival.
As shown in Fig. 7, control mice that received no injection of liposomai vincristine survived 10-11 days after administration of the P388 tumor. Treatment with either DSPC/Chol or SM/Chol/PEG-PE formulations at a vincristine dosage of 1 mg/kg increased the survival time to 17 and 19 days, respectively. Treatment with SM/Chol formulations at the same vincristine dosage gave a slight improvement in survival, 23 days.
At a vincristine dosage of 2 mg/kg, both DSPC/Chol and SM/Chol/PEG-PE formulations increased survival to 30-31 days. In contrast, at this vincristine dosage, the SM/Chol formulation was significantly more effective; 60% of the mice were surviving at 60 days after administration of the P388 tumor (Fig. 7) . At a vincristine dosage of 4 mg/kg, both the DSPC/Chol and SM/Chol/PEG-PE formulations gave 40% of the mice surviving at 60 days after P388 tumor injection. Formulations of SM/Chol were significantly more efficacious; apart from a single vincristine toxicity death, survival of the remaining mice at 60 days was 100% (Fig. 7) . Thus, the antitumor efficacy of SM/Chol liposomes was significantly better than that of SM/Chol/PEG-PE liposomes (Fig. 7) despite the observation that the loading of vincristine to P388 tumors was identical in these two liposomai formulations (Fig. 6) . This result suggests that the better vincristine retention properties of SM/Chol liposomes in circulation, compared to SM/Chol/PEG-PE liposomes (Fig. 3) , may also occur in the peritoneal cavity and result in improved vincristine uptake by the P388 tumor cells. Formulations of SM/Chol were approximately two-fold more effective than were the formulations based on either DSPC/Chol or SM/Chol/PEG-PE. That is, survival achieved by DSPC/Chol and SM/Chol/PEG-PE formulations at vincristine dosages of 2.0 mg/kg were attained by SM/Chol at a dosage of 1.0 mg/kg. Similarly, the survival obtained by DSPC/Chol and SM/Chol/PEG- PE at a dose of 4.0 mg/kg of vincristine was very similar to that achieved by SM/Chol formulations at 2.0 mg/kg.
EXAMPLE V Bioavailability of Liposomai Swainsonine
Female Balb/c mice, 5-6 weeks of age, were housed under standard conditions. The animals received free access to both food and water throughout the experiment after a one week acclimatization period prior to experimental manipulation. Swainsonine (Toronto Res. Chem.) was radiolabeled with tritium. Tritiated swainsonine was administered as a lipid-based formulation (L-Im) and as an aqueous formulation containing the free drug (F-Im) . Tritiated swainsonine was loaded into sphingomyelin/cholesterol (Avanti Polar Labs) sphingosomes using a citrate buffer pH 2 gradient at a drug-to-lipid ratio of 0.2:1 (mol:mol) and with an efficiency of loading of 80%. Two hundred microliters of the lipid and aqueous swainsonine formulations were given orally by gavage (p.o.), intraperitoneally (i.p.), or intravenously (i.v.). Fifty microliter blood samples were collected by retroorbital bleeds at 1, 3, 6, and 24 hours after administration. The blood samples were bleached and then counted in a scintillation counter. Results were expressed as the percentage of the administered dose in the blood at various time-points after administration. As seen in Fig. 8, the liposomai formulation (L-Im) of swainsonine has superior bioavailability and achieves higher blood levels when compared to the free aqueous formulation (F-Im) . The oral bioavailability of swainsonine is about 60-65% when compared to swainsonine administered intravenously.
EXAMPLE VI Efficacy of Swainsonine and GM-CSF
Female C57BL/6 mice (average weight 15.03 g) , were used and housed under standard conditions. The animals received free access to both food and water throughout the experiment after a one week acclimatization period prior to experimental manipulation. The mice were 6 weeks old on the beginning of the experiment and placed randomly in 12 groups of 5 mice per group. Forty mice were given a single bolus (i.p.) injection of either methotrexate (Mtx, 410 mg/kg) (Sigma Chemical Co.) or Doxorubicin (Dox, 14.9 mk/kg) (Adria Laboratories) . Two days after chemotherapy, administration of swainsonine (2 mg/kg i.p. or p.o.), recombinant murine GM-CSF (granulocyte macrophage-colony stimulating factor) (1 μg/ ouse/day i.p., 5 x 104 U/μg activity) (R & D Systems) or phosphate buffered saline (PBS) (200 μl i.p.) was provided for 10 consecutive days (once per day) . The number of deaths was recorded for each treatment group over an observation period of 14 days.
Results in Table I (below) show that when swainsonine is administered for 10 days after a LD50 dose of chemotherapeutic (Mtx or Dox) , all animals administered swainsonine intravenously or by i.p. handled the cytotoxic insult and survived beyond 2 weeks after chemotherapy. Half of the animals treated with Mtx and half treated with Dox died within a few days after chemotherapy. Animals treated with an i.p. administration of recombinant murine GM-CSF for 10 days did not do as well as with swainsonine; about half of the animals treated with Mtx died within a few days after initiation of the 10 day dosing period. Animals dosed with Dox and 10 days of GM-CSF survived the two week recovery period. Swainsonine was administered orally for 10 days to the che otherapeutic-treated animals and all but one animal (in the Mtx-treated group) survived the cytotoxic treatment.
TABLE I
Test Agent Administration Survival (%)
1. PBS i.p. 100
2. MTX i.p. 40
3. DOX i.p. 60
4. L-SW i.p. 100
5. MTX/L-SW i.p. 100
6. DOX/L-SW i.p. 100
7. GM-CSF i.p. 100
8. MTX/GM-CSF i.p. 60
9. DOX/GM-CSF i.p. 100
10. L-SW p.o. 100
11. MTX/L-SW p.o. 80
12. DOX/L-SW p.o. 100
EXAMPLE VII Recovery from Chemotherapeutic Induced Leukopenia
On the 15th day after the initial chemotherapeutic dose, 4 mice from each group of the immunomodulation study (Example 6) were randomly sacrificed and up to 1 ml of blood was obtained by cardiac puncture. Circulating peripheral WBCs were counted, blood smears (for neutrophil counts) were made and plasma samples were collected and tested for cytokine production (IL-1, IL-2, TNF) . Blood cytokine levels (TNF, IL- 1, and IL-2) were assayed by commercially available assay kits. The spleen, thymus and bone marrow were removed and single cell suspensions were prepared. The cellularity of these lymphoid organs was assessed by trypan blue exclusion test. Fig. 9 shows the effects of GM-CSF and swainsonine on the bone marrow cellularity at 14 days after chemotherapy drug administration. As shown, swainsonine, given orally or by i.p. administration performed as effectively as GM-CSF given by i.p. for 10 consecutive days. Cells from spleen, thymus and bone marrow were tested for their ability to respond to stimulation from different mitogens: ConA (1, 2.5 and 5 μg/ml) (Sigma), phytohemagglutinin (PHA) (1, and 2.5 μg/ml) (Sigma) and LPS (2.5 and 5 μg/ml) (Difco) . After 72 hours of stimulation the proliferative response was measured using CellTiter 96
(Promega) . Simultaneously, mitogen stimulated splenocytes were set up for cytokine production after ConA (2.5 μg/ml) and lipopolysaccharide (LPS) (2.5 μg/ml) stimulation. The supernatants were collected after 24 hr. and 48 hr. and tested for Tumor Necrosis Factor-α (TNF-c.) and Interleukin-2
(IL-2) production using the direct ELISA method. Supernatants from unstimulated cells served as controls. Results are expressed as pg/ml of TNF-α? (sensitivity of the assay is < 25 pg/ml) or IL-2 (sensitivity of the assay is < 3 pg/ml) . As shown in Figs. 10A and 10B, respectively, TNF and IL-2 levels in ConA and LPS stimulated splenocytes were significantly elevated in swainsonine-treated animals compared to chemotherapy-treated and PBS (no treatment) controls.
To establish an advantage for the oral route of administration, swainsonine was incubated .in vitro in simulated gastric contents for various periods of time (1, 2, 4, 24, 48 and 72 hrs.) and the "oral stability" of swainsonine was determined. Swainsonine was also incubated in hydrochloric acid (pH2) containing the main gastric digestive enzyme, pepsin. The j_n vitro stability tests have shown swainsonine to be stable under these harsh conditions for up to 72 hours. Example VIII Pharmacokinetics, Tumor Loading and Therapy in SCID Mice
Bearing A431 Tumors
Tumor loading and antitumor efficacy properties of
DSPC/Chol and SM/Chol liposomai formulations of vincristine were determined in mice bearing solid human A431 squamous cell xenograft tumors. These experiments were undertaken to ensure that the positive results observed in the murine ascitic P388 tumor model were representative of other tumor types. SCID mice bearing 100-200 mg solid human A431 tumors were injected i.v. with free vincristine or with liposomes of either DSPC/Chol or SM/Chol containing vincristine. Vincristine encapsulated DSPC/Chol and SM/Chol liposomes were prepared as in Example II. Encapsulation of vincristine in DSPC/Chol and SM/Chol liposomes increased the amount of vincristine remaining in circulation 24 hours after administration by 28- and 87-fold, respectively, compared with free vincristine (Fig. 11A) . As observed in BDF1 mice bearing P388 tumors, the amount of vincristine remaining in the circulation in SM/Chol liposomes at 24 hours after injection was approximately 3-fold greater than for vincristine encapsulated in DSPC/Chol liposomes (Fig. 11A) .
Improved vincristine circulation longevity correlated with increases in the loading of vincristine in the A431 tumors (Fig. 11A) . Specifically, free vincristine levels in the A431 tumors were highest (0.856 mg/g tumor) at 0.5 hours after injection and decreased to 0.32 mg/g tumor at 24 hours (Fig. 11B) . Encapsulation of vincristine in DSPC/Chol liposomes increased the amount of vincristine in A431 tumors at 4 to 48 hours after administration to 1.3-1.55 mg/g tumor, respectively (Fig. 11B) . Encapsulation of vincristine in SM/Chol liposomes resulted in a further increase in vincristine delivery to A431 tumors at 24 to 48 hours after injection to 2.8-3.2 mg/g tumor, representing a 2-fold increase in the delivery obtained with DSPC/Chol liposomes. As observed in the murine ascitic tumor model, the vincristine/lipid observed in the solid human A431 tumors were very similar to those observed in the plasma. That is, for vincristine encapsulated in DSPC/Chol liposomes, the vincristine/lipid (wt/wt) ratios at 24 hours after injection were 0.022 in the plasma and 0.029 in the tumor, while for vincristine encapsulated in SM/Chol liposomes the vincristine/lipid ratios were 0.055 in the plasma and 0.050 in the tumor.
The antitumor efficacy of free and liposomai vincristine against A431 was closely correlated with vincristine accumulation at the tumor site (Fig. 12) . SCID mice bearing the A431 tumors that received no treatment showed a 100% increase in tumor weight within 4-5 days after treatment was initiated and required termination within 10 days when the tumor exceeded 10% of the total body weight. Tumor bearing SCID mice treated with free vincristine at 2.0 mg/kg had a brief delay in tumor growth (100% increase in tumor weight achieved within 6-8 days) but required termination between 10-12 days. In contrast, treatment with vincristine encapsulated in DSPC/Chol liposomes resulted in a significant delay in tumor growth (100% increase in tumor weight at 15-20 days, termination at 21 days after treatment) . This therapy was further enhanced by a single treatment of vincristine encapsulated in SM/Chol liposomes. In this treatment group a small but consistent decrease in tumor size was observed. At 15 days after injection, several tumors were palpable but unmeasurable and by 33 days after treatment several tumors were not palpable. Of the five mice (total of 10 tumors) treated with SM/Chol liposomai vincristine, 1 animal was terminated early due to tumor ulceration, not due to tumor growth. Of the eight tumors remaining at 40 days after liposome injection, histological analysis indicated that all eight tumors were actively dividing squamous cell carcinomas of a mass undetectable by physical examination. Therefore, treatment with SM/Chol liposomai vincristine effected a significant reduction in tumor growth, although none of the original tumors were cured.
In summary, the present invention demonstrates that liposomai formulations of vincristine and other alkaloids based on sphingomyelin/cholesterol vesicles have several significant advantages over formulations based on DSPC/cholesterol vesicles. Specifically, formulations based on sphingomyelin/cholesterol: (1) are much more stable to acid hydrolysis, (2) have significantly better drug retention characteristics, (3) have better tumor loading characteristics, and (4) show significantly better anti-tumor efficacy than do comparable liposomes composed of DSPC/Chol or SM/Chol/PEG-PE.
All publications and patents mentioned in this specification are herein incorporated by reference into the specification to the same extent as if each individual publication or patent was specifically and individually indicated to be incorporated herein by reference.
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 that certain changes and modifications may be practiced within the scope of the appended claims.

Claims

THE EMBODIMENTS OF THE INVENTION IN WHICH AN EXCLUSIVE PROPERTY OR PRIVILEGE IS CLAIMED ARE DEFINED AS FOLLOWS:
1. A liposomai composition for delivery of a therapeutic compound to a mammalian host which comprises a liposome having one or more membranes which comprise sphingomyelin and cholesterol, a liposomai interior having a pH less than that of the liposomai exterior, and a therapeutic compound contained in said liposome for delivery to the host .
2. The liposomai composition of claim 1, wherein the sphingomyelin and cholesterol are present at a molar ratio from 75/25 mol%/mol% sphingomyelin/cholesterol to 30/50 mol%/mol% sphingomyelin/cholesterol.
3. The liposomai composition of claim 2 , wherein the sphingomyelin and cholesterol are present at a molar ratio from 70/30 mol%/mol% sphingomyelin/cholesterol to 40/45 mol%/mol% sphingomyelin/cholesterol.
4. The liposomai composition of claim 3, wherein the sphingomyelin and cholesterol are present at a ratio of approximately 55/45 mol%/mol% sphingomyelin/cholesterol.
5. The liposomai composition of claim 1, wherein the lipophilic therapeutic compound is an alkaloid.
6. The liposomai composition of claim 5 wherein the
SUBSTITUTESHEET alkaloid is selected from vincristine, vinblastine, swainsonine or etoposide or prodrugs thereof.
7. The liposomai composition of claim 6, wherein the alkaloid is vincristine.
8. The liposomai composition of claim 6, wherein the alkaloid is swainsonine.
9. The liposomai composition of claim 6, wherein vincristine is present at a drug to lipid ratio of approximately 0.01/1.0 to 0.2/1.0 (wt/wt) and swainsonine is present at a drug to lipid ratio of 0.01/1.0 to 0.5/1.0 (mol/mol) .
10. The liposomai composition of claim 1, further comprising at least one lipid selected from a phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidyIglycerol, phosphatidle acid, cardiollpin, phosphatidylinosi ol, ceramide, cerebroslde and ganglioside.
11. The liposomai composition of claim 1, wherein the liposomes are unilamellar.
12. The liposomai composition of claim 1, wherein the liposomes have mean diameters of about 0.05 microns to 0.45 microns.
UBSTITUTESHEET
13. The liposomai composition of claim 1, wherein the liposomes have mean diameters of about 0.05 microns to 0.2 microns.
14. The liposomai composition of claim 1 wherein the interior of said liposome is pH 2 to pH 5.
15. The liposomai composition of claim 1, wherein the interior comprises a citrate buffer at about pH 4.0.
16. A liposome for delivery of an alkaloid therapeutic compound, produced by the process of« forming a liposome from a mixture which comprises sphingomyelin and cholesterol, in a first buffered aqueous solution having an acidic pH greater than pH 2> and suspending the liposome in a second buffered solution having a pH which is greater than that of the first buffered aqueous solution, whereby a transmembrane pH gradient is formed which facilitates the transfer of the therapeutic compound to the liposome.
17. The liposome produced by the process of claim 16, wherein the process further comprises the step of separating the liposome containing the therapeutic compound from the second buffer containing therapeutic compound which has not been entrapped by the liposome.
18. A method for delivering an alkaloid therapeutic
SUBSTITUTE SHEET compound to a tumor susceptible to inhibition by said compound, comprising: administering to a host containing said tumor, a liposomai composition according to claim 1 which comprises said compound or a pharmaceutically acceptable salt thereof.
19. The method of claim 18, wherein the cholesterol is present in the liposomai composition at a total molar proportion of 30% to 50%.
20. The method of claim 19, wherein the sphingomyelin and cholesterol are present at a ratio of about 55/45, mol%/mol%, respectively.
21. The method of claim 18, wherein the alkaloid compound is vincristine or swainsonine.
22. The method of claim 21, wherein the alkaloid compound is vincristine.
23. The method of claim 21, wherein the alkaloid compound is swainsonine.
24. The method of claim 21, wherein vincristine is present in the liposomai composition at a drug to lipid ratio of approximately 0.01/1.0 to 0.2/1.0 (wt/wt) and swainsonine is present at a drug to lipid ratio of 0.01/1.0 to 0.5/1.0 (mol/mol) .
SUBSTITUTE SHEET
25. The method of claim 18, wherein the liposomai composition containing said alkaloid compound is administered repeatedly to the host to maintain a concentration of said compound sufficient to inhibit the tumor but less than an amount which causes unacceptable toxicity to the host.
26. The method of claim 18, wherein the liposomai composition containing said alkaloid compound is administered Intravenously.
27. The method of claim 18, wherein the liposomai composition containing said alkaloid compound is administered parenterally.
28. The method of claim 18, wherein the liposomai composition containing said alkaloid is administered orally.
29. The method of claim 18, wherein the liposomes of the liposomai composition administered to the host are unilamellar.
30. The method of claim 29, wherein the unilamellar liposomes of said composition have a mean diameter of 0.05 microns to 0.45 microns.
31. The method of claim 30, wherein the unilamellar liposomes of said composition have a mean diameter of 0.05 microns to 0.2 microns.
SUBSTITUTE SHEET
32. A method for delivering an alkaloid immunomodulatlng compound, comprising: administering to a host a liposomai composition according to claim 1 comprising said immunomodulator or a pharmaceutically acceptable salt thereof.
33. The method of claim 32, wherein the compound is swainsonine.
34. The method of claim 33, wherein swainsonine is delivered parenterally or orally.
35. The method of claim 34, wherein swainsonine is delivered intravenously or orally.
36. The method of claim 33, wherein swainsonine is present in the liposomai composition at a drug to lipid ratio of approximately 0.01/1.0 to 0.5/1.0 (mol/mol).
37. A method of loading an alkaloid therapeutic compound, comprising: forming a liposome from a mixture which comprises sphingomyelin and cholesterol, in a first aqueous solution of ethanolammonium or methylammonium, subsequently decreasing the external concentration of ethanolammonium or methylammonium to form a liposomai transmembrane pH gradient wherein the pH gradient facilitates uptake of the therapeutic into the liposomai interior? and
TIT TE EET admixing the therapeutic compound with the liposomes to allow transmembrane uptake.
38. The method of claim 37, wherein the interior of the liposome is between pH 2 and pH 5.
39. The method of claim 37, wherein the therapeutic compound is vincristine or swainsonine.
SUBSTITUTE SHEET
PCT/CA1995/000363 1994-06-20 1995-06-19 Sphingosomes for enhanced drug delivery WO1995035094A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
AU27094/95A AU2709495A (en) 1994-06-20 1995-06-19 Sphingosomes for enhanced drug delivery
EP95922375A EP0804159B1 (en) 1994-06-20 1995-06-19 Sphingosomes for enhanced drug delivery
DE69531701T DE69531701T2 (en) 1994-06-20 1995-06-19 SPHINGOSOME WITH IMPROVED DRUG RELEASE
AT95922375T ATE248586T1 (en) 1994-06-20 1995-06-19 SPHINGOSOMES WITH IMPROVED DRUG DELIVERY
JP50143496A JP3270478B2 (en) 1994-06-20 1995-06-19 Sphingosomes for enhanced drug delivery
CA002193502A CA2193502C (en) 1994-06-20 1995-06-19 Sphingosomes for enhanced drug delivery

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/263,603 US5543152A (en) 1994-06-20 1994-06-20 Sphingosomes for enhanced drug delivery
US08/263,603 1994-06-20

Publications (1)

Publication Number Publication Date
WO1995035094A1 true WO1995035094A1 (en) 1995-12-28

Family

ID=23002474

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA1995/000363 WO1995035094A1 (en) 1994-06-20 1995-06-19 Sphingosomes for enhanced drug delivery

Country Status (10)

Country Link
US (1) US5543152A (en)
EP (1) EP0804159B1 (en)
JP (1) JP3270478B2 (en)
AT (1) ATE248586T1 (en)
AU (1) AU2709495A (en)
CA (1) CA2193502C (en)
DE (1) DE69531701T2 (en)
ES (1) ES2206510T3 (en)
PT (1) PT804159E (en)
WO (1) WO1995035094A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996032930A1 (en) * 1995-04-18 1996-10-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem Liposome drug-loading method and composition
US5962467A (en) * 1995-06-07 1999-10-05 Glycodesign, Inc. Derivatives of swainsonine and their use as therapeutic agents
US6048870A (en) * 1996-10-01 2000-04-11 Glycodesign 3, 5, and/or 6 substituted analogues of swainsonine processes for their preparation and their use as therapeutic agents
US6051711A (en) * 1997-10-24 2000-04-18 Glycodesign Inc. Synthesis of swainsonine salts
US6093816A (en) * 1996-06-27 2000-07-25 Isis Pharmaceuticals, Inc. Cationic lipids
US6395745B1 (en) 1997-04-15 2002-05-28 Glycodesign, Inc. Alkaloid halide salts of swainsonine and methods of use
FR2829391A1 (en) * 2001-09-07 2003-03-14 Coreana Cosmetics Co Ltd COSMETIC COMPOSITION CONTAINING FULLY ENCAPSULATED RETINOL
US7887836B2 (en) 1999-04-01 2011-02-15 Hana Biosciences, Inc. Compositions and methods for treating lymphoma
EP1933813A4 (en) * 2005-10-11 2013-02-27 Univ Pittsburgh Sphingomyelin liposomes for the treatment of hyperactive bladder disorders
WO2016045732A1 (en) * 2014-09-25 2016-03-31 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
US9801874B2 (en) 2012-11-20 2017-10-31 Spectrum Pharmaceuticals Method for the preparation of liposome encapsulated vincristine for therapeutic use
US11559486B2 (en) 2015-07-22 2023-01-24 Acrotech Biopharma, LLC Ready-to-use formulation for Vincristine Sulfate Liposome Injection

Families Citing this family (362)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030068362A1 (en) * 1993-02-22 2003-04-10 American Bioscience, Inc. Methods and formulations for the delivery of pharmacologically active agents
US20030133955A1 (en) * 1993-02-22 2003-07-17 American Bioscience, Inc. Methods and compositions useful for administration of chemotherapeutic agents
US6537579B1 (en) 1993-02-22 2003-03-25 American Bioscience, Inc. Compositions and methods for administration of pharmacologically active compounds
US6749868B1 (en) 1993-02-22 2004-06-15 American Bioscience, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US6096331A (en) 1993-02-22 2000-08-01 Vivorx Pharmaceuticals, Inc. Methods and compositions useful for administration of chemotherapeutic agents
US6753006B1 (en) * 1993-02-22 2004-06-22 American Bioscience, Inc. Paclitaxel-containing formulations
US5741516A (en) * 1994-06-20 1998-04-21 Inex Pharmaceuticals Corporation Sphingosomes for enhanced drug delivery
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US20030026782A1 (en) * 1995-02-07 2003-02-06 Arthur M. Krieg Immunomodulatory oligonucleotides
US5820873A (en) * 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
CN1245423A (en) 1996-12-30 2000-02-23 巴特勒纪念研究院 Formulation and method for treating neoplasms by inhalation
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6083923A (en) * 1997-10-31 2000-07-04 Isis Pharmaceuticals Inc. Liposomal oligonucleotide compositions for modulating RAS gene expression
US6410328B1 (en) 1998-02-03 2002-06-25 Protiva Biotherapeutics Inc. Sensitizing cells to compounds using lipid-mediated gene and compound delivery
US20030022854A1 (en) 1998-06-25 2003-01-30 Dow Steven W. Vaccines using nucleic acid-lipid complexes
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
US7683044B2 (en) * 1999-03-25 2010-03-23 Center For Molecular Medicine And Immunology Sphingomyelin therapy of autoimmune disease
AU3740000A (en) * 1999-03-25 2000-10-16 Center For Molecular Medicine And Immunology Sphingomyelin containing preparation for the enhancement of tumor therapy
ES2524141T3 (en) * 1999-04-01 2014-12-04 Talon Therapeutics, Inc. Compositions to treat cancer
US7244450B2 (en) * 1999-04-01 2007-07-17 Inex Pharmaceuticals Corporation Compositions and methods for treating lymphoma
US7311924B2 (en) * 1999-04-01 2007-12-25 Hana Biosciences, Inc. Compositions and methods for treating cancer
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
AUPQ259399A0 (en) 1999-09-01 1999-09-23 Lustre Investments Pte Ltd Therapeutic agents
JP4776843B2 (en) 1999-10-01 2011-09-21 イムノゲン インコーポレーティッド Cancer composition and method using immunoconjugate and chemotherapeutic agent
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
WO2001095935A1 (en) * 2000-01-20 2001-12-20 Ottawa Health Research Institute Immunostimulatory nucleic acids for inducing a th2 immune response
GB0007150D0 (en) * 2000-03-24 2000-05-17 Lamellar Therapeutics Limited Immunotherapeutic methods and compositions
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
EP1299085B1 (en) * 2000-06-30 2005-11-16 Inex Pharmaceuticals Corp. Improved liposomal camptothecins and uses thereof
US7452550B2 (en) 2000-06-30 2008-11-18 Hana Biosciences, Inc. Liposomal antineoplastic drugs and uses thereof
WO2002089772A1 (en) * 2001-05-09 2002-11-14 Inex Pharmaceuticals Corporation Anti-angiogenic therapy using liposome-encapsulated chemotherapeutic agents
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
DK2000545T3 (en) 2001-06-20 2011-11-28 Genentech Inc Compositions and methods for diagnosis and treatment of lung tumor
CA2451643C (en) 2001-06-21 2012-11-13 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US20030096772A1 (en) 2001-07-30 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US20030054042A1 (en) * 2001-09-14 2003-03-20 Elaine Liversidge Stabilization of chemical compounds using nanoparticulate formulations
EP2174953A1 (en) 2001-09-18 2010-04-14 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
NZ566396A (en) 2001-10-09 2009-07-31 Isis Pharmaceuticals Inc Antisense modulation of insulin-like growth factor binding protein 5 expressions
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
IL161733A0 (en) 2001-11-02 2005-11-20 Insert Therapeutics Inc Methods and compositions for therapeutic use of rna interference
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
AU2002367318B2 (en) 2002-01-02 2007-07-12 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20040009126A1 (en) * 2002-03-05 2004-01-15 Transave, Inc. Inhalation system for prevention and treatment of intracellular infections
US20030180712A1 (en) 2002-03-20 2003-09-25 Biostratum Ab Inhibition of the beta3 subunit of L-type Ca2+ channels
WO2003080027A1 (en) * 2002-03-20 2003-10-02 Elan Pharma International, Ltd. Nanoparticulate compositions of angiogenesis inhibitors
JP2005536190A (en) 2002-04-16 2005-12-02 ジェネンテック・インコーポレーテッド Compositions and methods for tumor diagnosis and treatment
US7199107B2 (en) 2002-05-23 2007-04-03 Isis Pharmaceuticals, Inc. Antisense modulation of kinesin-like 1 expression
CN1694959B (en) * 2002-09-13 2013-09-18 雷普利瑟公司 Non-sequence complementary antiviral oligonucleotides
US20050196382A1 (en) * 2002-09-13 2005-09-08 Replicor, Inc. Antiviral oligonucleotides targeting viral families
US7229976B2 (en) 2002-09-26 2007-06-12 Isis Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
US7879351B2 (en) * 2002-10-29 2011-02-01 Transave, Inc. High delivery rates for lipid based drug formulations, and methods of treatment thereof
ES2686361T3 (en) * 2002-10-29 2018-10-17 Insmed Incorporated Liposomes comprising an aminoglycoside for the treatment of lung infections
US7718189B2 (en) 2002-10-29 2010-05-18 Transave, Inc. Sustained release of antiinfectives
AU2003290598A1 (en) 2002-11-05 2004-06-03 Isis Pharmaceuticals, Inc. Modified oligonucleotides for use in rna interference
AU2003287505A1 (en) 2002-11-05 2004-06-03 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
CA2505801A1 (en) 2002-11-13 2004-05-27 Rosanne Crooke Antisense modulation of apolipoprotein b expression
ES2420914T3 (en) 2002-11-13 2013-08-27 Genzyme Corporation Antisense modulation of apolipoprotein B expression
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
CA2502015A1 (en) 2002-12-11 2004-06-24 Coley Pharmaceutical Group, Inc. 5' cpg nucleic acids and methods of use
US7468356B2 (en) 2003-02-11 2008-12-23 Antisense Therapeutics Ltd. Modulation of insulin like growth factor I receptor expression
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
CA2522184A1 (en) * 2003-04-17 2004-11-04 The Trustees Of Columbia University In The City Of New York Desmoglein 4 is a novel gene involved in hair growth
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
WO2005002507A2 (en) 2003-06-03 2005-01-13 Isis Pharmaceuticals, Inc. Modulation of survivin expression
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
NZ545134A (en) 2003-09-18 2009-06-26 Lilly Co Eli Modulation of eIF4E expression
EP1678194B1 (en) 2003-10-10 2013-06-26 Alchemia Oncology Pty Limited The modulation of hyaluronan synthesis and degradation in the treatment of disease
WO2005041870A2 (en) 2003-10-24 2005-05-12 Ader Enterprises, Inc. Composition and method for the treatment of eye disease
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
ES2493016T3 (en) 2003-11-17 2014-09-11 Genentech, Inc. Compositions comprising antibodies against CD79b conjugated to a growth inhibitory agent or a cytotoxic agent and methods for the treatment of tumors of hematopoietic origin
JP2007520222A (en) 2004-01-20 2007-07-26 アイシス ファーマシューティカルズ インコーポレイテッド Regulation of glucocorticoid receptor expression
US7468431B2 (en) 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
JP4937899B2 (en) * 2004-03-12 2012-05-23 アルナイラム ファーマシューティカルズ, インコーポレイテッド IRNA substances targeting VEGF
US8790919B2 (en) 2004-03-15 2014-07-29 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of RNA by RNase H
CA2561221C (en) * 2004-03-26 2016-09-20 Curis, Inc. Rna interference modulators of hedgehog signaling and uses thereof
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
CA3006109A1 (en) 2004-05-03 2005-11-17 Ipsen Biopharm Ltd. Drug delivery liposomes containing anionic polyols or anionic sugars
AU2005251691A1 (en) * 2004-05-17 2005-12-22 Tekmira Pharmaceuticals Corporation Liposomal formulations comprising dihydrosphingomyelin and methods of use thereof
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
ATE514776T1 (en) 2004-10-05 2011-07-15 California Inst Of Techn APTAMER-REGULATED NUCLEIC ACIDS AND USES THEREOF
JP4990786B2 (en) * 2004-11-05 2012-08-01 イネックス ファーマシューティカルズ コーポレイション Compositions and methods for stabilizing drug liposome formulations
EP1855694B1 (en) 2005-02-09 2020-12-02 Sarepta Therapeutics, Inc. Antisense composition for treating muscle atrophy
AU2006223498A1 (en) 2005-03-10 2006-09-21 Genentech, Inc. Methods and compositions for modulating vascular integrity
JP2008537551A (en) 2005-03-31 2008-09-18 カランド ファーマシューティカルズ, インコーポレイテッド Inhibitors of ribonucleotide reductase subunit 2 and uses thereof
US7700567B2 (en) 2005-09-29 2010-04-20 Supergen, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
JP2009513716A (en) 2005-11-01 2009-04-02 アルナイラム ファーマシューティカルズ インコーポレイテッド Inhibition of influenza virus replication by RNAi
EP1966377A2 (en) 2005-11-21 2008-09-10 Isis Pharmaceuticals, Inc. Modulation of eif4e-bp2 expression
CA2896083A1 (en) 2005-12-08 2007-06-14 Insmed Incorporated Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US7951934B2 (en) 2006-01-26 2011-05-31 Isis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
CA2873833A1 (en) 2006-03-31 2007-10-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of eg5 gene
EP2614839A3 (en) 2006-04-05 2015-01-28 Genentech, Inc. Method for using BOC/CDO to modulate hedgehog signaling
JP4812874B2 (en) * 2006-04-28 2011-11-09 アルナイラム ファーマシューティカルズ, インコーポレイテッド Composition and method for suppressing expression of JC virus gene
WO2007125173A2 (en) 2006-05-03 2007-11-08 Baltic Technology Development, Ltd. Antisense agents combining strongly bound base - modified oligonucleotide and artificial nuclease
US7812150B2 (en) 2006-05-19 2010-10-12 Alnylam Pharmaceuticals, Inc. RNAi modulation of Aha and therapeutic uses thereof
US7888498B2 (en) * 2006-05-22 2011-02-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of IKK-B gene
US8598333B2 (en) * 2006-05-26 2013-12-03 Alnylam Pharmaceuticals, Inc. SiRNA silencing of genes expressed in cancer
US8198253B2 (en) 2006-07-19 2012-06-12 Isis Pharmaceuticals, Inc. Compositions and their uses directed to HBXIP
US8158595B2 (en) 2006-11-09 2012-04-17 California Institute Of Technology Modular aptamer-regulated ribozymes
US8048998B2 (en) * 2007-01-19 2011-11-01 Exiqon A/S Mediated cellular delivery of LNA oligonucleotides
US20100093836A1 (en) 2007-01-29 2010-04-15 Isis Pharmaceuticals, Inc Compounds and methods for modulating protein expression
CN101663407B (en) 2007-02-22 2017-08-08 健泰科生物技术公司 method for detecting inflammatory bowel disease
AP3018A (en) 2007-03-29 2014-10-31 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expressionof a gene from the ebola
US20100196455A1 (en) * 2007-05-04 2010-08-05 Transave, Inc. Compositions of Multicationic Drugs for Reducing Interactions with Polyanionic Biomolecules and Methods of Use Thereof
US9114081B2 (en) 2007-05-07 2015-08-25 Insmed Incorporated Methods of treating pulmonary disorders with liposomal amikacin formulations
US9119783B2 (en) 2007-05-07 2015-09-01 Insmed Incorporated Method of treating pulmonary disorders with liposomal amikacin formulations
US9333214B2 (en) 2007-05-07 2016-05-10 Insmed Incorporated Method for treating pulmonary disorders with liposomal amikacin formulations
AR066984A1 (en) * 2007-06-15 2009-09-23 Novartis Ag INHIBITION OF THE EXPRESSION OF THE ALFA SUBUNITY OF THE SODIUM EPITELIAL CHANNEL (ENAC) THROUGH ARNI (INTERFERENCE RNA)
US20090082217A1 (en) * 2007-07-16 2009-03-26 California Institute Of Technology Selection of nucleic acid-based sensor domains within nucleic acid switch platform
US20120165387A1 (en) 2007-08-28 2012-06-28 Smolke Christina D General composition framework for ligand-controlled RNA regulatory systems
US8367815B2 (en) * 2007-08-28 2013-02-05 California Institute Of Technology Modular polynucleotides for ligand-controlled regulatory systems
US8865667B2 (en) * 2007-09-12 2014-10-21 California Institute Of Technology Higher-order cellular information processing devices
MX2010003465A (en) 2007-10-02 2010-07-05 Amgen Inc Increasing erythropoietin using nucleic acids hybridizable to micro-rna and precursors thereof.
US9029524B2 (en) * 2007-12-10 2015-05-12 California Institute Of Technology Signal activated RNA interference
US7871985B2 (en) 2007-12-10 2011-01-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor VII gene
CN102014932B (en) * 2008-03-05 2015-11-25 阿尔尼拉姆医药品有限公司 For suppressing compositions and the method for Eg5 and VEGF gene expression
JP2011516065A (en) 2008-04-04 2011-05-26 カランド ファーマシューティカルズ, インコーポレイテッド Compositions and uses of EPAS1 inhibitors
ITMI20081052A1 (en) * 2008-06-10 2009-12-11 Univ Milano Bicocca LIPOSOMAS ABLE TO EFFECTIVELY TIE THE BETA-AMYLOID PEPTIDE
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
EP3081648A1 (en) 2008-08-25 2016-10-19 Excaliard Pharmaceuticals, Inc. Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
EP2331690B1 (en) 2008-09-02 2016-01-13 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of mutant egfr gene
US10138485B2 (en) 2008-09-22 2018-11-27 Rxi Pharmaceuticals Corporation Neutral nanotransporters
EP2334793B1 (en) 2008-09-25 2016-04-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of serum amyloid a gene
NZ592867A (en) 2008-10-20 2013-05-31 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of transthyretin
CN102317458B (en) 2008-12-04 2018-01-02 库尔纳公司 Pass through treatment of the suppression of erythropoietin(EPO) (EPO) natural antisense transcript to EPO relevant diseases
KR101749352B1 (en) 2008-12-04 2017-06-20 큐알엔에이, 인크. Treatment of sirtuin 1(sirt1) related diseases by inhibition of natural antisense transcript to sirtuin 1
CA2745811C (en) 2008-12-04 2021-07-13 Joseph Collard Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene
EP3225281A1 (en) 2008-12-10 2017-10-04 Alnylam Pharmaceuticals, Inc. Gnaq targeted dsrna compositions and methods for inhibiting expression
WO2010078536A1 (en) 2009-01-05 2010-07-08 Rxi Pharmaceuticals Corporation Inhibition of pcsk9 through rnai
EP3009150B1 (en) 2009-02-12 2019-11-13 CuRNA, Inc. Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
KR101805199B1 (en) 2009-02-12 2017-12-05 큐알엔에이, 인크. Treatment of glial cell derived neurotrophic factor (gdnf) related diseases by inhibition of natural antisense transcript to gdnf
US8329882B2 (en) 2009-02-18 2012-12-11 California Institute Of Technology Genetic control of mammalian cells with synthetic RNA regulatory systems
US20120041051A1 (en) 2009-02-26 2012-02-16 Kevin Fitzgerald Compositions And Methods For Inhibiting Expression Of MIG-12 Gene
US20110319317A1 (en) 2009-03-04 2011-12-29 Opko Curna, Llc Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirt1
AU2010223967B2 (en) 2009-03-12 2015-07-30 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of Eg5 and VEGF genes
WO2010107733A2 (en) 2009-03-16 2010-09-23 Curna, Inc. Treatment of nuclear factor (erythroid-derived 2)-like 2 (nrf2) related diseases by inhibition of natural antisense transcript to nrf2
EP2408920B1 (en) 2009-03-17 2017-03-08 CuRNA, Inc. Treatment of delta-like 1 homolog (dlk1) related diseases by inhibition of natural antisense transcript to dlk1
US9145555B2 (en) 2009-04-02 2015-09-29 California Institute Of Technology Integrated—ligand-responsive microRNAs
EP3248618A1 (en) 2009-04-22 2017-11-29 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
ES2661787T3 (en) 2009-05-01 2018-04-04 Curna, Inc. Treatment of hemoglobin-related diseases (hbf / hbg) by inhibition of natural antisense transcript for hbf / hbg
KR101722541B1 (en) 2009-05-06 2017-04-04 큐알엔에이, 인크. Treatment of tristetraproline(ttp) related diseases by inhibition of natural antisense transcript to ttp
CN103223177B (en) 2009-05-06 2016-08-10 库尔纳公司 By suppression therapy lipid transfer and the metabolic gene relevant disease of the natural antisense transcript for lipid transfer and metabolic gene
CN102575251B (en) 2009-05-18 2018-12-04 库尔纳公司 The relevant disease of the reprogramming factor is treated by inhibiting the natural antisense transcript for the reprogramming factor
KR101703695B1 (en) 2009-05-22 2017-02-08 큐알엔에이, 인크. Treatment of transcription factor e3 (tfe3) and insulin receptor substrate 2 (irs2) related diseases by inhibition of natural antisense transcript to tfe3
EP2435571B1 (en) 2009-05-28 2016-12-14 CuRNA, Inc. Treatment of antiviral gene related diseases by inhibition of natural antisense transcript to an antiviral gene
JP5638204B2 (en) * 2009-05-29 2014-12-10 国立大学法人 岡山大学 Liposome preparation for oral administration and production method thereof
EP2443238B1 (en) 2009-06-16 2017-03-22 CuRNA, Inc. Treatment of paraoxonase 1 (pon1) related diseases by inhibition of natural antisense transcript to pon1
JP5944311B2 (en) 2009-06-16 2016-07-05 クルナ・インコーポレーテッド Treatment of collagen gene-related diseases by suppression of natural antisense transcripts against collagen genes
CN102597238B (en) 2009-06-24 2016-06-29 库尔纳公司 The relevant disease of TNFR2 is treated by suppressing for the natural antisense transcript of tumor necrosis factor receptor 2 (TNFR2)
WO2010151674A2 (en) 2009-06-26 2010-12-29 Curna, Inc. Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
JP6128848B2 (en) 2009-08-05 2017-05-17 クルナ・インコーポレーテッド Treatment of insulin gene (INS) -related diseases by suppression of natural antisense transcripts against the insulin gene (INS)
EP2464336A4 (en) 2009-08-14 2013-11-20 Alnylam Pharmaceuticals Inc Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
WO2011031482A2 (en) 2009-08-25 2011-03-17 Curna, Inc. Treatment of 'iq motif containing gtpase activating protein' (iqgap) related diseases by inhibition of natural antisense transcript to iqgap
CA2772715C (en) 2009-09-02 2019-03-26 Genentech, Inc. Mutant smoothened and methods of using the same
EP2488210A4 (en) 2009-10-12 2014-04-30 Smith Holdings Llc Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
CN102711826B (en) 2009-10-22 2017-03-29 霍夫曼-拉罗奇有限公司 For the method and composition that the HEPSIN for regulating and controlling macrophage-stimulating albumen is activated
WO2011056234A1 (en) 2009-11-06 2011-05-12 Fibrogen, Inc. Treatment for radiation-induced disorders
CA2781887C (en) 2009-11-30 2018-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US8901129B2 (en) 2009-12-11 2014-12-02 Genecode As Methods of facilitating neural cell survival using GDNF family ligand (GFL) mimetics or RET signaling pathway activators
WO2011084455A2 (en) 2009-12-16 2011-07-14 Opko Curna, Llc. Treatment of membrane bound transcription factor peptidase, site 1 (mbtps1) related diseases by inhibition of natural antisense transcript to mbtps1
KR101793753B1 (en) 2009-12-23 2017-11-03 큐알엔에이, 인크. Treatment of uncoupling protein 2 (ucp2) related diseases by inhibition of natural antisense transcript to ucp2
CN102869776B (en) 2009-12-23 2017-06-23 库尔纳公司 HGF relevant diseases are treated by suppressing the natural antisense transcript of HGF (HGF)
WO2011090741A2 (en) 2009-12-29 2011-07-28 Opko Curna, Llc TREATMENT OF TUMOR PROTEIN 63 (p63) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO p63
EP2519633B1 (en) 2009-12-29 2017-10-25 CuRNA, Inc. Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
NO2521784T3 (en) 2010-01-04 2018-05-05
US8912157B2 (en) 2010-01-06 2014-12-16 Curna, Inc. Treatment of pancreatic developmental gene related diseases by inhibition of natural antisense transcript to a pancreatic developmental gene
DK2524039T3 (en) 2010-01-11 2018-03-12 Curna Inc TREATMENT OF GENDER HORMON-BINDING GLOBULIN (SHBG) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPTS TO SHBG
EP2525808A2 (en) 2010-01-19 2012-11-28 The Trustees Of Columbia University In The City Of New York Osteocalcin as a treatment for male reproductive disorders
WO2011091390A2 (en) 2010-01-25 2011-07-28 Opko Curna, Llc Treatment of rnase h1 related diseases by inhibition of natural antisense transcript to rnase h1
WO2011103528A2 (en) 2010-02-22 2011-08-25 Opko Curna Llc Treatment of pyrroline-5-carboxylate reductase 1 (pycr1) related diseases by inhibition of natural antisense transcript to pycr1
EP2538981B1 (en) 2010-02-23 2017-12-20 F. Hoffmann-La Roche AG Compositions and methods for the diagnosis and treatment of tumor
US20130195961A1 (en) 2010-03-09 2013-08-01 Kejian Yang Novel mucosal vaccination approach for herpes simplex virus type-2
ES2893199T3 (en) 2010-03-29 2022-02-08 Alnylam Pharmaceuticals Inc dsRNA therapy for transthyretin (TTR)-related ocular amyloidosis
US9044494B2 (en) 2010-04-09 2015-06-02 Curna, Inc. Treatment of fibroblast growth factor 21 (FGF21) related diseases by inhibition of natural antisense transcript to FGF21
WO2011139917A1 (en) 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Modulation of transthyretin expression
MA34291B1 (en) 2010-05-03 2013-06-01 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSING AND TREATING A TUMOR
EP2566966A4 (en) 2010-05-03 2013-12-11 Curna Inc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
CN103429739B (en) 2010-05-12 2018-11-13 哥伦比亚大学纽约管理委员会 Prepare the method generated with the enteroendocrine cell of excreting insulin
TWI586356B (en) 2010-05-14 2017-06-11 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
CA2799207C (en) 2010-05-26 2019-03-26 Curna, Inc. Treatment of atonal homolog 1 (atoh1) related diseases by inhibition of natural antisense transcript to atoh1
WO2011153323A2 (en) 2010-06-02 2011-12-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
US8980860B2 (en) 2010-07-14 2015-03-17 Curna, Inc. Treatment of discs large homolog (DLG) related diseases by inhibition of natural antisense transcript to DLG
US8481680B2 (en) 2010-10-05 2013-07-09 Genentech, Inc. Mutant smoothened and methods of using the same
US8993533B2 (en) 2010-10-06 2015-03-31 Curna, Inc. Treatment of sialidase 4 (NEU4) related diseases by inhibition of natural antisense transcript to NEU4
CA2815212A1 (en) 2010-10-22 2012-04-26 Curna, Inc. Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
CN103201387B (en) 2010-10-27 2018-02-02 库尔纳公司 IFRD1 relevant diseases are treated by suppressing the natural antisense transcript of interferon correlative development regulatory factor 1 (IFRD1)
WO2012061811A2 (en) 2010-11-05 2012-05-10 Fibrogen, Inc. Treatment method for lung remodeling diseases
CN110123830A (en) 2010-11-09 2019-08-16 阿尔尼拉姆医药品有限公司 Composition and method for inhibiting the lipid of the expression of Eg5 and VEGF gene to prepare
WO2012071238A2 (en) 2010-11-23 2012-05-31 Opko Curna Llc Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
WO2012078967A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for increasing erythropoietin (epo) production
EP2654779B1 (en) 2010-12-23 2018-02-28 Ludwig Institute for Cancer Research, Ltd. Liposomal formulation of nonglycosidic ceramides and uses thereof
SI2670411T1 (en) 2011-02-02 2019-06-28 Excaliard Pharmaceuticals, Inc. Antisense compounds targeting connective tissue growth factor (ctgf) for use in a method of treating keloids or hypertrophic scars
WO2012109495A1 (en) 2011-02-09 2012-08-16 Metabolic Solutions Development Company, Llc Cellular targets of thiazolidinediones
SG193923A1 (en) 2011-03-29 2013-11-29 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of tmprss6 gene
CN103547588B (en) 2011-04-13 2016-06-29 Isis制药公司 The antisense that PTP1B expresses regulates
KR20190042747A (en) 2011-06-02 2019-04-24 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Methods and uses for ex vivo tissue culture systems
RU2620980C2 (en) 2011-06-09 2017-05-30 Курна, Инк. Treatment of diseases associated with frataxin (fxn), by inhibiting natural antisense fxn transcript
EP2721156B1 (en) 2011-06-16 2016-12-21 Ionis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
WO2012177949A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of protein c (proc) genes
KR102395085B1 (en) 2011-06-21 2022-05-09 알닐람 파마슈티칼스 인코포레이티드 Angiopoietin-like 3(angptl3) irna compostions and methods of use thereof
CA2839896A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
CN112111492A (en) 2011-06-21 2020-12-22 阿尔尼拉姆医药品有限公司 Compositions and methods for inhibiting expression of apolipoprotein C-III (APOC3) gene
WO2012178033A2 (en) 2011-06-23 2012-12-27 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
WO2013003112A1 (en) 2011-06-27 2013-01-03 The Jackson Laboratory Methods and compositions for treatment of cancer and autoimmune disease
EP2739735A2 (en) 2011-08-01 2014-06-11 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
EP3401401B1 (en) 2011-09-20 2020-04-15 Ionis Pharmaceuticals, Inc. Antisense modulation of gcgr expression
US20130085139A1 (en) 2011-10-04 2013-04-04 Royal Holloway And Bedford New College Oligomers
CN103906838A (en) 2011-10-25 2014-07-02 Isis制药公司 Antisense modulation of GCCR expression
JP2015511494A (en) 2012-03-15 2015-04-20 キュアナ,インク. Treatment of BDNF-related diseases by inhibition of natural antisense transcripts against brain-derived neurotrophic factor (BDNF)
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
CA2870860C (en) 2012-05-21 2021-07-27 Insmed Incorporated Systems for treating pulmonary infections
AU2013202947B2 (en) 2012-06-13 2016-06-02 Ipsen Biopharm Ltd. Methods for treating pancreatic cancer using combination therapies comprising liposomal irinotecan
US9717724B2 (en) 2012-06-13 2017-08-01 Ipsen Biopharm Ltd. Methods for treating pancreatic cancer using combination therapies
CN104602672B (en) * 2012-06-14 2018-12-25 伯尔尼大学 For treating the customization liposome of bacterium infection
US20160136159A1 (en) 2012-09-17 2016-05-19 Chemedest Ltd. Method for Treating Peripheral Neuropathy
RU2675859C2 (en) 2012-11-29 2018-12-25 Инсмед Инкорпорейтед Stabilised vancomycin formulations
ES2657608T3 (en) 2012-12-05 2018-03-06 Alnylam Pharmaceuticals, Inc. Compositions of pcsk9 arni and methods of use thereof
WO2014107731A1 (en) 2013-01-07 2014-07-10 Biomedical Research Models, Inc. Therapeutic vaccines for treating herpes simplex virus type 2 infections
KR102605775B1 (en) 2013-03-14 2023-11-29 알닐람 파마슈티칼스 인코포레이티드 Complement component c5 irna compositions and methods of use thereof
WO2014152497A2 (en) 2013-03-15 2014-09-25 The Trustees Of Columbia University In The City Of New York Osteocalcin as a treatment for cognitive disorders
EP2978446B1 (en) 2013-03-27 2020-03-04 The General Hospital Corporation Anti-cd33 antibody for use in treating alzheimer's disease
EP2994167B1 (en) 2013-05-06 2020-05-06 Alnylam Pharmaceuticals, Inc. Dosages and methods for delivering lipid formulated nucleic acid molecules
TWI727917B (en) 2013-05-22 2021-05-21 美商阿尼拉製藥公司 TMPRSS6 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
RS57418B1 (en) 2013-05-22 2018-09-28 Alnylam Pharmaceuticals Inc Serpina1 irna compositions and methods of use thereof
AU2014280847B2 (en) 2013-06-13 2019-07-04 Antisense Therapeutics Ltd Combination therapy
AU2014317961B2 (en) 2013-09-05 2020-07-30 Murdoch University Antisense-induced exon2 inclusion in acid alpha-glucosidase
US10036020B2 (en) 2013-09-19 2018-07-31 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Compositions and methods for inhibiting JC virus (JCV)
CN105793423A (en) 2013-10-02 2016-07-20 阿尔尼拉姆医药品有限公司 Compositions and methods for inhibiting expression of the LECT2 gene
JP6613227B2 (en) 2013-10-04 2019-11-27 アルナイラム ファーマシューティカルズ, インコーポレイテッド Composition and method for inhibiting the expression of ALAS1 gene
IL282401B (en) 2013-12-12 2022-08-01 Alnylam Pharmaceuticals Inc Complement component irna compositions and methods of use thereof
WO2015116902A1 (en) 2014-01-31 2015-08-06 Genentech, Inc. G-protein coupled receptors in hedgehog signaling
EP3102197B1 (en) 2014-02-04 2018-08-29 Genentech, Inc. Mutant smoothened and methods of using the same
WO2015123264A1 (en) 2014-02-11 2015-08-20 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
WO2015171918A2 (en) 2014-05-07 2015-11-12 Louisiana State University And Agricultural And Mechanical College Compositions and uses for treatment thereof
ES2926985T3 (en) 2014-05-15 2022-10-31 Insmed Inc Methods for treating nontuberculous mycobacterial lung infections
EA201692370A1 (en) 2014-05-22 2017-03-31 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS of mRNA ANGIOTENZINOGENA (AGT) AND METHODS OF THEIR USE
EP3160503B1 (en) 2014-06-26 2021-02-17 The Trustees of Columbia University in the City of New York Inhibition of serotonin expression in gut enteroendocrine cells results in conversion to insulin-positive cells
WO2016033424A1 (en) 2014-08-29 2016-03-03 Genzyme Corporation Methods for the prevention and treatment of major adverse cardiovascular events using compounds that modulate apolipoprotein b
EP3191591A1 (en) 2014-09-12 2017-07-19 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
EP3207138B1 (en) 2014-10-17 2020-07-15 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
EP3212794B1 (en) 2014-10-30 2021-04-07 Genzyme Corporation Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
CN107250362B (en) 2014-11-17 2021-10-22 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
US11191811B2 (en) 2014-11-19 2021-12-07 The Trustees Of Columbia University In The City Of New York Osteocalcin as a treatment for frailty associated with aging
TW201702218A (en) 2014-12-12 2017-01-16 美國杰克森實驗室 Compositions and methods relating to the treatment of cancer, autoimmune disease, and neurodegenerative disease
JP2018504380A (en) 2014-12-18 2018-02-15 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. REVERSIR ™ compounds
EP3797789A1 (en) 2015-01-20 2021-03-31 The Children's Medical Center Corporation Anti-net compounds for treating and preventing fibrosis and for facilitating wound healing
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
MA41795A (en) 2015-03-18 2018-01-23 Sarepta Therapeutics Inc EXCLUSION OF AN EXON INDUCED BY ANTISENSE COMPOUNDS IN MYOSTATIN
WO2016164746A1 (en) 2015-04-08 2016-10-13 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
KR20180002688A (en) 2015-05-06 2018-01-08 알닐람 파마슈티칼스 인코포레이티드 (F12), calichein B, plasma (Fletcher factor) 1 (KLKB1) and kininogen 1 (KNG1) iRNA compositions and methods for their use
US11318131B2 (en) 2015-05-18 2022-05-03 Ipsen Biopharm Ltd. Nanoliposomal irinotecan for use in treating small cell lung cancer
MA50829A (en) 2015-06-01 2018-04-11 Sarepta Therapeutics Inc EXCLUSION OF EXON INDUCED BY ANTISEN TECHNOLOGY IN TYPE VII COLLAGEN
EP3307316A1 (en) 2015-06-12 2018-04-18 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
WO2017004538A1 (en) 2015-07-02 2017-01-05 Otsuka Pharmaceutical Co., Ltd. Lyophilized pharmaceutical compositions
US10494632B2 (en) 2015-07-10 2019-12-03 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (IGFALS) compositions and methods of use thereof
EP3919619A1 (en) 2015-07-17 2021-12-08 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
CN108348480A (en) 2015-08-20 2018-07-31 益普生生物制药有限公司 Use liposome Irinotecan and PARP inhibitor combination treatment use for cancer treatment
ES2826826T3 (en) 2015-08-21 2021-05-19 Ipsen Biopharm Ltd Pharmaceutical combination comprising liposomal irinotecan, oxaliplatin, 5-fluorouracil and leucovorin for the treatment of metastatic pancreatic cancer
CN114525280A (en) 2015-09-02 2022-05-24 阿尔尼拉姆医药品有限公司 iRNA compositions of programmed cell death 1 ligand 1(PD-L1) and methods of use thereof
WO2017048620A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting patatin-like phospholipase domain containing 3 (pnpla3) and methods of use thereof
WO2017048843A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
US10954300B2 (en) 2015-09-28 2021-03-23 The Trustees Of Columbia University In The City Of New York Use of pentoxifylline with immune checkpoint-blockade therapies for the treatment of melanoma
WO2017062835A2 (en) 2015-10-09 2017-04-13 Sarepta Therapeutics, Inc. Compositions and methods for treating duchenne muscular dystrophy and related disorders
SG10201912568PA (en) 2015-10-16 2020-02-27 Ipsen Biopharm Ltd Stabilizing camptothecin pharmaceutical compositions
WO2017075670A1 (en) 2015-11-05 2017-05-11 Children's Hospital Los Angeles "mobilizing leukemia cells"
JP2018536689A (en) 2015-12-10 2018-12-13 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Sterol regulatory element binding protein (SREBP) chaperone (SCAP) iRNA compositions and methods of use thereof
AU2017213826A1 (en) 2016-02-04 2018-08-23 Curis, Inc. Mutant smoothened and methods of using the same
CN109414511B (en) 2016-04-18 2023-05-23 萨勒普塔医疗公司 Antisense oligomers for treating diseases associated with acid alpha-glucosidase genes and methods of use thereof
US20190256845A1 (en) 2016-06-10 2019-08-22 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
MX2019004783A (en) 2016-11-02 2019-08-12 Ipsen Biopharm Ltd Treating gastric cancer using combination therapies comprising liposomal irinotecan, oxaliplatin, 5-fluoruracil (and leucovorin).
TWI788312B (en) 2016-11-23 2023-01-01 美商阿尼拉製藥公司 SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
KR20230166146A (en) 2016-12-16 2023-12-06 알닐람 파마슈티칼스 인코포레이티드 Methods for treating or preventing ttr-associated diseases using transthyretin(ttr) irna compositions
CN116549393A (en) * 2017-03-02 2023-08-08 康柏辛股份有限公司 Liposome for inhibiting formation of biological membrane
CN114224840A (en) 2017-03-31 2022-03-25 富士胶片株式会社 Liposome composition and pharmaceutical composition
AU2018254437A1 (en) 2017-04-18 2019-11-28 Alnylam Pharmaceuticals, Inc. Methods for the treatment of subjects having a hepatitis B virus (HBV) infection
WO2018209288A1 (en) 2017-05-12 2018-11-15 Massachusetts Institute Of Technology Argonaute protein-double stranded rna complexes and uses related thereto
PE20200746A1 (en) 2017-07-06 2020-07-24 Arrowhead Pharmaceuticals Inc IARN AGENTS FOR THE INHIBITION OF THE EXPRESSION OF ALFA-ENAC AND METHODS OF USE
AU2018310857A1 (en) 2017-08-03 2020-02-13 Otsuka Pharmaceutical Co., Ltd. Drug compound and purification methods thereof
JP7394753B2 (en) 2017-10-18 2023-12-08 サレプタ セラピューティクス, インコーポレイテッド antisense oligomer compounds
WO2019089922A1 (en) 2017-11-01 2019-05-09 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
EP3714054A1 (en) 2017-11-20 2020-09-30 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
CN111757886A (en) * 2018-03-27 2020-10-09 日油株式会社 Sphingomyelin-derived lipids and methods for producing same
WO2019191627A1 (en) 2018-03-30 2019-10-03 Insmed Incorporated Methods for continuous manufacture of liposomal drug products
CN116536272A (en) 2018-04-06 2023-08-04 儿童医疗中心有限公司 Compositions and methods for somatic reprogramming and imprinting
US20210348162A1 (en) 2018-08-16 2021-11-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
US20210332367A1 (en) 2018-09-18 2021-10-28 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
CN113454216A (en) 2018-11-29 2021-09-28 旗舰先锋创新V股份有限公司 Methods of modulating RNA
WO2020132521A1 (en) 2018-12-20 2020-06-25 Praxis Precision Medicines, Inc. Compositions and methods for the treatment of kcnt1 related disorders
WO2021030522A1 (en) 2019-08-13 2021-02-18 Alnylam Pharmaceuticals, Inc. SMALL RIBOSOMAL PROTEIN SUBUNIT 25 (RPS25) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
EP4025694A1 (en) 2019-09-03 2022-07-13 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
US20220389429A1 (en) 2019-10-04 2022-12-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
TW202132567A (en) 2019-11-01 2021-09-01 美商阿尼拉製藥公司 Huntingtin (htt) irna agent compositions and methods of use thereof
EP4051796A1 (en) 2019-11-01 2022-09-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
US20230056569A1 (en) 2019-11-22 2023-02-23 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
MX2022006433A (en) 2019-12-13 2022-06-23 Alnylam Pharmaceuticals Inc Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof.
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
CN115427571A (en) 2020-02-10 2022-12-02 阿尔尼拉姆医药品有限公司 Compositions and methods for silencing VEGF-A expression
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
WO2021195307A1 (en) 2020-03-26 2021-09-30 Alnylam Pharmaceuticals, Inc. Coronavirus irna compositions and methods of use thereof
US20230190785A1 (en) 2020-03-30 2023-06-22 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajc15 gene expression
US20230295622A1 (en) 2020-04-06 2023-09-21 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
EP4133077A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
CN116134135A (en) 2020-04-07 2023-05-16 阿尔尼拉姆医药品有限公司 Compositions and methods for silencing SCN9A expression
WO2021206917A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
BR112022021813A2 (en) 2020-04-27 2023-01-17 Alnylam Pharmaceuticals Inc APOLIPOPROTEIN AND (APOE) IRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2021231673A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
EP4150076A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
WO2021231685A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
EP4150087A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
EP4150088A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
WO2021231691A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rsi)
EP4150090A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
EP4153746A1 (en) 2020-05-21 2023-03-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
AR122534A1 (en) 2020-06-03 2022-09-21 Triplet Therapeutics Inc METHODS FOR THE TREATMENT OF NUCLEOTIDE REPEAT EXPANSION DISORDERS ASSOCIATED WITH MSH3 ACTIVITY
EP4162050A1 (en) 2020-06-09 2023-04-12 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
JP2023540429A (en) 2020-07-10 2023-09-25 アンセルム(アンスティチュート・ナシオナル・ドゥ・ラ・サンテ・エ・ドゥ・ラ・ルシェルシュ・メディカル) Methods and compositions for treating epilepsy
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
TW202229552A (en) 2020-10-05 2022-08-01 美商艾拉倫製藥股份有限公司 G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
EP4232581A1 (en) 2020-10-21 2023-08-30 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
EP4232582A1 (en) 2020-10-23 2023-08-30 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
CA3201452A1 (en) 2020-12-01 2022-06-09 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
TW202305131A (en) 2021-02-12 2023-02-01 美商艾拉倫製藥股份有限公司 SUPEROXIDE DISMUTASE 1 (SOD1) iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING OR PREVENTING SUPEROXIDE DISMUTASE 1- (SOD1-) ASSOCIATED NEURODEGENERATIVE DISEASES
CN117222739A (en) 2021-02-25 2023-12-12 阿尔尼拉姆医药品有限公司 Prion protein (PRNP) IRNA compositions and methods of use thereof
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
JP2024512635A (en) 2021-03-29 2024-03-19 アルナイラム ファーマシューティカルズ, インコーポレイテッド Huntingtin (HTT) iRNA agent composition and method of use thereof
AU2022261124A1 (en) 2021-04-22 2023-10-05 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating cancer
WO2022232343A1 (en) 2021-04-29 2022-11-03 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
WO2022245583A1 (en) 2021-05-18 2022-11-24 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
EP4341405A1 (en) 2021-05-20 2024-03-27 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
WO2022256290A2 (en) 2021-06-04 2022-12-08 Alnylam Pharmaceuticals, Inc. HUMAN CHROMOSOME 9 OPEN READING FRAME 72 (C9ORF72) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
WO2023283403A2 (en) 2021-07-09 2023-01-12 Alnylam Pharmaceuticals, Inc. Bis-rnai compounds for cns delivery
IL309905A (en) 2021-07-23 2024-03-01 Alnylam Pharmaceuticals Inc Beta-catenin (ctnnb1) irna compositions and methods of use thereof
WO2023069603A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
WO2023122762A1 (en) 2021-12-22 2023-06-29 Camp4 Therapeutics Corporation Modulation of gene transcription using antisense oligonucleotides targeting regulatory rnas
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023240277A2 (en) 2022-06-10 2023-12-14 Camp4 Therapeutics Corporation Methods of modulating progranulin expression using antisense oligonucleotides targeting regulatory rnas
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988004924A1 (en) * 1986-12-24 1988-07-14 Liposome Technology, Inc. Liposomes with enhanced circulation time
WO1988006442A1 (en) * 1987-03-05 1988-09-07 The Liposome Company, Inc. High drug:lipid formulations of liposomal-antineoplastic agents
WO1990014105A1 (en) * 1989-05-15 1990-11-29 The Liposome Company, Inc. Accumulation of drugs into liposomes by a proton gradient

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL91664A (en) * 1988-09-28 1993-05-13 Yissum Res Dev Co Ammonium transmembrane gradient system for efficient loading of liposomes with amphipathic drugs and their controlled release

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988004924A1 (en) * 1986-12-24 1988-07-14 Liposome Technology, Inc. Liposomes with enhanced circulation time
WO1988006442A1 (en) * 1987-03-05 1988-09-07 The Liposome Company, Inc. High drug:lipid formulations of liposomal-antineoplastic agents
WO1990014105A1 (en) * 1989-05-15 1990-11-29 The Liposome Company, Inc. Accumulation of drugs into liposomes by a proton gradient

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5939096A (en) * 1995-04-18 1999-08-17 Yissum Research Development Company Of The Hebrew University Of Jerusalem Liposome drug-loading method and composition
WO1996032930A1 (en) * 1995-04-18 1996-10-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem Liposome drug-loading method and composition
US5962467A (en) * 1995-06-07 1999-10-05 Glycodesign, Inc. Derivatives of swainsonine and their use as therapeutic agents
US6610664B2 (en) 1996-06-27 2003-08-26 Isis Pharmaceuticals, Inc. Cationic lipids
US6093816A (en) * 1996-06-27 2000-07-25 Isis Pharmaceuticals, Inc. Cationic lipids
US6437121B1 (en) 1996-06-27 2002-08-20 Isis Pharmaceuticals, Inc. Cationic lipids
US6048870A (en) * 1996-10-01 2000-04-11 Glycodesign 3, 5, and/or 6 substituted analogues of swainsonine processes for their preparation and their use as therapeutic agents
US6395745B1 (en) 1997-04-15 2002-05-28 Glycodesign, Inc. Alkaloid halide salts of swainsonine and methods of use
US6051711A (en) * 1997-10-24 2000-04-18 Glycodesign Inc. Synthesis of swainsonine salts
US7887836B2 (en) 1999-04-01 2011-02-15 Hana Biosciences, Inc. Compositions and methods for treating lymphoma
FR2829391A1 (en) * 2001-09-07 2003-03-14 Coreana Cosmetics Co Ltd COSMETIC COMPOSITION CONTAINING FULLY ENCAPSULATED RETINOL
EP1933813A4 (en) * 2005-10-11 2013-02-27 Univ Pittsburgh Sphingomyelin liposomes for the treatment of hyperactive bladder disorders
US9801874B2 (en) 2012-11-20 2017-10-31 Spectrum Pharmaceuticals Method for the preparation of liposome encapsulated vincristine for therapeutic use
WO2016045732A1 (en) * 2014-09-25 2016-03-31 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
WO2016046060A1 (en) * 2014-09-25 2016-03-31 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
KR20170063780A (en) * 2014-09-25 2017-06-08 비온테크 알엔에이 파마슈티컬스 게엠베하 Stable formulations of lipids and liposomes
AU2015320980B2 (en) * 2014-09-25 2021-02-18 BioNTech SE Stable formulations of lipids and liposomes
KR102264820B1 (en) 2014-09-25 2021-06-14 비온테크 알엔에이 파마슈티컬스 게엠베하 Stable formulations of lipids and liposomes
US11173120B2 (en) 2014-09-25 2021-11-16 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
US11559486B2 (en) 2015-07-22 2023-01-24 Acrotech Biopharma, LLC Ready-to-use formulation for Vincristine Sulfate Liposome Injection

Also Published As

Publication number Publication date
ATE248586T1 (en) 2003-09-15
CA2193502A1 (en) 1995-12-28
AU2709495A (en) 1996-01-15
US5543152A (en) 1996-08-06
CA2193502C (en) 2005-09-13
EP0804159A1 (en) 1997-11-05
EP0804159B1 (en) 2003-09-03
JPH10501534A (en) 1998-02-10
JP3270478B2 (en) 2002-04-02
DE69531701T2 (en) 2004-03-25
PT804159E (en) 2004-02-27
DE69531701D1 (en) 2003-10-09
ES2206510T3 (en) 2004-05-16

Similar Documents

Publication Publication Date Title
EP0804159B1 (en) Sphingosomes for enhanced drug delivery
US5741516A (en) Sphingosomes for enhanced drug delivery
CA2584279C (en) Compositions and methods for stabilizing liposomal drug formulations
JP4885715B2 (en) Irinotecan formulation
US7311924B2 (en) Compositions and methods for treating cancer
US6787132B1 (en) Combined chemo-immunotherapy with liposomal drugs and cytokines
Harrington et al. Liposomes as vehicles for targeted therapy of cancer. Part 1: preclinical development
JP2006514016A (en) Liposome formulation
US10085940B2 (en) Liposomes co-encapsulating a bisphosphonate and an amphipathic agent
JP5199666B2 (en) Compositions and methods for treating leukemia
EP1448165B1 (en) Lipid carrier compositions and methods for improved drug retention
US20030129224A1 (en) Lipid carrier compositions and methods for improved drug retention
US20220296520A1 (en) Liposome composition and preparation method thereof
US20070298094A1 (en) Medicinal Composition, Preparation and Combined Preparation
AU756109B2 (en) Methods for antitumor therapy
WO2000009071A2 (en) A novel liposomal formulation useful in treatment of cancer and other proliferation diseases
JP2001026544A (en) Liposome formulation of oil-soluble antitumor agent
KR100768265B1 (en) Heparin coated liposomes to prolong circulation time in bloodstream and preparation method thereof
IL136343A (en) Combined chemo - immunotherapy with liposomal drugs and cytokines

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LT LU LV MD MG MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TT UA UG UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN 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)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2193502

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1995922375

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1995922375

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1995922375

Country of ref document: EP