WO1991005545A1 - Liposome microreservoir composition and method - Google Patents

Liposome microreservoir composition and method Download PDF

Info

Publication number
WO1991005545A1
WO1991005545A1 PCT/US1990/006034 US9006034W WO9105545A1 WO 1991005545 A1 WO1991005545 A1 WO 1991005545A1 US 9006034 W US9006034 W US 9006034W WO 9105545 A1 WO9105545 A1 WO 9105545A1
Authority
WO
WIPO (PCT)
Prior art keywords
liposome
liposomes
composition
compound
vesicle
Prior art date
Application number
PCT/US1990/006034
Other languages
French (fr)
Inventor
Frank J. Martin
Martin C. Woodle
Carl Redemann
Rhada Radhakrishnan
Annie Yau-Young
Original Assignee
Liposome Technology, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23685679&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1991005545(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Liposome Technology, Inc. filed Critical Liposome Technology, Inc.
Priority to KR1019920700918A priority Critical patent/KR0134982B1/en
Priority to EP90916409A priority patent/EP0496813B1/en
Priority to KR1019920700919A priority patent/KR920703013A/en
Priority to DE69015207T priority patent/DE69015207T2/en
Publication of WO1991005545A1 publication Critical patent/WO1991005545A1/en
Priority to NO92921213A priority patent/NO921213L/en
Priority to FI921763A priority patent/FI921763A0/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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/5537Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom the heteroring containing the structure -C(=O)-N-C(=O)- (both carbon atoms belong to the heteroring)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to a liposome composition and method for administering a therapeutic compound into the bloodstream over an extended period.
  • Liposomes have been proposed as a carrier for intraven ⁇ ously (IV) administered compounds.
  • IV intraven ⁇ ously
  • the use of lipo ⁇ somes for slow release of liposome-entrapped material into the bloodstream has been severely restricted by the rapid clear ⁇ ance of liposomes from the bloodstream by cells of the reti- culoendothelial system (RES) .
  • the RES will remove 80-95% of IV injected liposomes within one hour, and effec ⁇ tively remove circulating liposomes from the bloodstream within of 4-6 hours.
  • the invention includes, in one aspect, a liposome compo ⁇ sition effective to extend to at least 24 hours, the period of effective activity of an therapeutic compound which can be administered intravenously in a therapeutically effective amount, and which has a blood halflife, in free form, of less than about 4 hours.
  • the composition includes liposomes (i) composed of vesicle-forming lipids and between 1-20 mole per ⁇ cent of a vesicle-forming lipid derivatized with a biocompa- tible hydrophilic polymer, and (ii) having a selected mean particle diameter in the size range between about 0.1 to 0.4 microns, and the compound in liposome-entrapped form.
  • the composition is intended for intravenous administration at a dose which contains an amount of the liposome-entrapped compound which is at least three times the therapeutically effective dose for the compound in free form.
  • the hydrophilic polymer is polyethyleneglycol having a molecular weight between about 1,000-5,000 daltons, and the polymer is derivatized with the polar head roup of a phospholipid, such a phosphatidylethanol- amine (PE) .
  • a phospholipid such as a phosphatidylethanol- amine (PE)
  • the polymer may be other suitable biocompatible hydrophilic polymers, such as polylactic acid and polyglycolic acid.
  • the composition is effective to extend to at least 48 hours, the period of thera ⁇ Chamberic activity of an intravenously injected polypeptide which can be administered intravenously in a therapeutically effec ⁇ tive amount.
  • the polypeptide may be a peptide or protein, such as superoxide dismutase, glucocerebrosidase, asparagi- nase, adenosine deaminase, interferons (alpha, beta, and gamma), interleukin (1,2,3,4,5,6,7), tissue necrosis factor
  • TNF - alpha, beta colony stimulating factors
  • M-CSF macro- phage
  • G-CSF granulocyte
  • GM-CSF granulocyte, macrophage
  • TPA prourokinase
  • urokinase HIV-1 vaccine
  • hepatitis B vaccine hepatitis B vaccine
  • malaria vaccine hepatitis B vaccine
  • melanoma vaccine erythropoietin
  • EPO ribosome inhibitor pro- tein
  • factor VIII factor VIII
  • bone growth factor fibroblast growth factor
  • insulin-like growth factor nerve growth factor
  • platelet-derived growth factor tumor growth factors (alpha, beta), somatomedin C (IGF-1)
  • IGF-1 tumor growth factors
  • ribosome inhibitor pro- tein ribosome inhibitor pro- tein
  • the composition is effective to deliver a therapeutically effective amount of the peptide into the bloodstream for a period of between 5-10 days.
  • Also forming part of the invention is a method for exten ⁇ ding to at least 24 hours, the period of effective activity of an therapeutic compound which can be administered intravenous ⁇ ly in a therapeutically effective amount, and which has a halflife in the blood, in free form, of less than about 4 hours.
  • a liposome composition of the type described above is administered intravenously to a subject at a dose which contains an amount of the compound which is at least three times such therapeutically effective amount.
  • a liposome composition effective to extend to at least one week, the period of effective activity of an therapeutic compound which can be administered intra ⁇ venously in a therapeutically effective amount.
  • the composi- tion includes liposomes (i) composed of vesicle-forming lipids and between 1-20 mole percent of a vesicle-forming lipid deri ⁇ vatized with a biocompatible hydrophilic polymer, and (ii) having a selected mean particle diameter in the size range between about 0.07-.15 microns, and the compound in liposome- entrapped form.
  • the composition is intended for subcutaneous administration at a dose which contains an amount of the lipo ⁇ some-entrapped compound which is at least ten times such therapeutically effective intravenously administered amount.
  • the liposome composition is used in a method for extending the period of release of a therapeutic compound, preferably a polypeptide, in a therapeutically active amount, for a period of at least 2 weeks.
  • the invention includes a liposome composition composed of vesicle-forming lipids and a vesicle- forming lipid derivatized with polylactic acid or polyglycolic acid, and a lipid composition composed of a vesicle-forming lipid having a polar head group, and a polylactic acid or polyglycolic acid moiety derivatized to the lipid's head group.
  • Figure 1 illustrates a general reaction scheme for derivatizing a vesicle-forming lipid amine with a polyalkyl- 5 ether
  • Figure 2 is a reaction scheme for preparing phosphati- dylethanolamine (PE) derivatized with polyethyleneglycol via a cyanuric chloride linking agent;
  • PE phosphati- dylethanolamine
  • Figure 3 illustrates a reaction scheme for preparing 10 phosphatidylethanolamine (PE) derivatized with polyethylene ⁇ glycol by means of a diimidazole activating reagent
  • Figure 4 illustrates a reaction scheme for preparing phosphatidylethanolamine (PE) derivatized with polyethylene ⁇ glycol by means of a trifluoromethane sulfonate reagent
  • Figure 5 illustrates a vesicle-forming lipid derivatized with polyethyleneglycol through a peptide (A) , ester (B) , and disulfide (C) linkage;
  • Figure 6 illustrates a reaction scheme for preparing phosphatidylethanolamine (PE) derivatized with polylactic 20 acid
  • Figure 7 is a plot of liposome retention time in the blood, expressed in terms of percent injected dose as a function of hours after IV injection, for PEG-PE liposomes containing different amounts of phosphatidylglycerol; 25 Figure 8 is a plot similar to that of Figure 7, showing retention times in the blood of liposomes composed of predomi ⁇ nantly unsaturated phospholipid components;
  • Figure 9 is a plot similar to that of Figure 1 , showing retention times in the blood of PEG liposomes (solid tri ⁇
  • Figure 10 is a plot of blood lifetimes of PEG-liposomes sized by extrusion through 0.1 micron (solid squares), 0.2 micron (solid circles), and 0.4 micron (solid triangles) polycarbonate membranes;
  • Figure 11 is a plot of blood retention times in liposomes containing a vesicle-forming lipid derivatized with polylactic acid (solid squares) and polyglycolic acid (open triangles) ;
  • Figure 12 shows urine flow rates in rats, as a percentage of predosage rate, after surgery and IV administration of saline (control, open circles) and of aqueous solutions of vasopressin at total doses of 0.2 ⁇ g (closed squares), 0.8 ⁇ g (closed triangles) , and 2 ⁇ g (closed circles) ;
  • Figure 13 shows urine flow rates in rats, as a percentage of predosage rate, after surgery and IV administration of saline (control, open circles) and of PEG-liposomes containing entrapped vasopressin at total doses of 2 ⁇ g (closed squares) , 8 ⁇ g (closed triangles) , and 24 ⁇ g (closed circles) ;
  • Figure 14 shows urine
  • Figure 15 shows the blood clearance kinetics of free macrophage-colony stimulating factor (M-CSF) (solid triangles) , PEG-liposomes containing 30 mole percent cholesterol (solid triangles) , and M-CSF associated with the PEG-liposomes (solid circles) ;
  • M-CSF free macrophage-colony stimulating factor
  • Figure 16 shows the blood clearance kinetics of free M-CSF (solid triangles) , cholesterol-free PEG-liposomes (solid triangles) , and M-CSF associated with the PEG-liposomes (solid circles) ;
  • Figure 17 is a plot of percent release of M-CSF into the blood from PEG liposomes containing 30 (solid, circles) and 0 (solid triangles) mole percent cholesterol;
  • Figure 18A shows urine flow rates in rats, as a percentage of predosage rate, after surgery and subcutaneous administra ⁇ tion of saline (control, open circles) and free vasopressin, in an amount 2 ⁇ g (solid triangles) , 25 ⁇ g " (solid circles) , and 50 ⁇ g (solid diamonds) ; and
  • Figure 18B shows urine flow rates in rats, as a percentage of predosage rate, after surgery and subcutaneous administra ⁇ tion of saline (control, open circles) and vasopressin en ⁇ trapped in PEG-liposomes, in an amount 25 ⁇ g (solid triang- les) , 100 ⁇ g (solid circles) , and 400 ⁇ g (solid diamonds) .
  • FIG. 1 shows a general reaction scheme for preparing vesicle-forming lipid derivatized a biocompatible, hydrophili polymer, as exemplified by polyethylene glycol (PEG) , poly lactic acid, and polyglycolic acid, all of which are readil water soluble, can be coupled to vesicle-forming lipids, an are tolerated in vivo without toxic effects.
  • the hydrophili polymer which is employed, e.g., PEG is preferably capped b a methoxy, ethoxy or other unreactive group at one end, or i one in which one end is more reactive than the other, such a polylactic acid.
  • the polymer is activated at one end by reaction with suitable activating agent, such as cyanuric acid, diimadozle, anhydride reagent, or the like, as described below. Th activated compound is then reacted with a vesicle-formin lipid, such as phosphatidylethanol (PE) , to produce th derivatized lipid.
  • suitable activating agent such as cyanuric acid, diimadozle, anhydride reagent, or the like
  • Th activated compound is then reacted with a vesicle-formin lipid, such as phosphatidylethanol (PE) , to produce th derivatized lipid.
  • PE phosphatidylethanol
  • the polar group in the vesicle-formin lipid may be activated for reaction with the polymer, or th two groups may be joined in a concerted coupling reaction, according to known coupling methods.
  • PEG capped at one end with a methoxy or ethoxy group can be obtained commercially in a variety of polymer sizes, e.g., 500-20,000 ' dalton molecular weights.
  • the vesicle-forming lipid is preferably one having two hydrocarbon chains, typically acyl chains, and a polar head group. Included in this class are the phospholipids, such as phosphatidylcholine (PC) , PE, phosphatidic acid (PA) , phospha- tidylinositol (PI) , and sphingomyelin (SM) , where the two hydrocarbon chains are typically between about 14-22 carbon atoms in length, and have varying degrees of unsaturation. Also included in this class are the glycolipids, such as cere- broside and gangliosides. Another vesicle-forming lipid which may be employed is cholesterol and related sterols.
  • PC phosphatidylcholine
  • PA phosphatidic acid
  • PI phospha- tidylinositol
  • SM sphingomyelin
  • glycolipids such as cere- broside and gangliosides.
  • cholesterol may be less tightly anchored to a lipid bilayer membrane, particu ⁇ larly when derivatized with a high molecular weight polyalkyl- ether, and therefore be less effective in promoting liposome evasion of the RES in the bloodstream.
  • vesicle-forming lipid is intended to include any amphipathic lipid having hydrophobic and polar head group moieties, and which (a) by itself can form spontaneously into bilayer vesicles in water, as exemplified by phospholipids, or (b) is stably incorporated into lipid bilayers in combination with phospholipids, with its hydrophobic moiety in contact with the interior, hydro- phobic region of the bilayer membrane, and its polar head group moiety oreinted toward the exterior, polar surface of the membrane.
  • An example of a latter type of vesicle-forming lipid is cholesterol and cholesterol derivatives, such as cholesterol sulfate and cholesterol hemisuccinate.
  • the vesicle-forming lipid may be a relatively fluid lipid, meaning that the lipid phase has a relatively low liquid-to-liquid crystal phase-transition temperature, e.g., at or below room temperature, or relatively rigid lipid, meaning that the lipid has a relatively high melting temperature, e.g., up to 50°C.
  • relatively rigid lipid meaning that the lipid has a relatively high melting temperature, e.g., up to 50°C.
  • the more rigid, i.e., saturated lipids contribute to greater membrane rigidity in a lipid bilayer structure and also contribute to greater bilayer stability in serum.
  • Other lipid components, such as cholesterol are also known to contribute to membrane rigidity and stability in lipid bilayer structures.
  • Phospholipids whose acyl chains have a variety of degrees of saturation can be obtained commercially, or prepared according to published methods.
  • FIG. 2 shows a reaction scheme for producing a PE-PEG lipid in which the PEG is derivatized to PE through a cyanuric chloride group. Details of the reaction are provided in Example 1. Briefly, ethoxy-capped PEG is activated with cyanuric chloride in the presence in sodium carbonate under conditions which produced the activated PEG compound IV in the figure. This material is purified to remove unreacted cyanuric acid. The activated PEG compound is reacted with PE in the presence of triethyl a ine to produce the desired PE- PEG compound shown at VII in the figure. The yield is about 8-10% with respect to initial quantities of PEG. The method just described may be applied to a variety of lipid amines, including PE, cholesteryl amine, and glycolipids with sugar-amine groups.
  • lipid amines including PE, cholesteryl amine, and glycolipids with sugar-amine groups.
  • a second method of coupling a polyalkylether, such as capped PEG to a lipid amine is illustrated in Figure 3.
  • the capped PEG is activated with a carbonyl diimidazole coupling reagent, to form the activated imidazole compoun shown at X in Figure 3.
  • Reaction with a lipid amine, such as PE leads to PEG coupling to the lipid through an amide linkage, as illustrated in the PEG-PE compound shown at XI in the figure. Details of the reaction are given in Example 2.
  • a third reaction method for coupling a capped poly ⁇ alkylether to a lipid amine is shown in Figure " 4.
  • PEG is first protected at its OH end by a trimethylsilane group.
  • the end-protection reaction is shown at A in the figure, and involves the reaction of trimethylsilylchloride with PEG in the presence of triethylamine.
  • the protected PEG is then reacted with the anhydride of trifluoromethyl sulfonate (compound XVI in Figure 4) to form the PEG compound activated with trifluoromethyl sulfonate.
  • the trimethylsilyl protective group can be released by acid treatment, as indicated at H+ in the figure, or by reaction with a quaternary amine fluoride salt, such as the fluoride salt of tetrabutylamine. It will be appreciated that a variety of known coupling reactions, in addition to those just described, are suitable for preparing vesicle-forming lipids derivatized with hydro ⁇ philic polymers such as PEG.
  • the sulfonate anhydride coupling reagent illustrated in Figure 4 can be used to join an activated polyalkylether to the hydroxyl group of an amphipathic lipid, such as the 5'-OH of cholesterol.
  • Other reactive lipid groups such as an acid or ester lipid group may also be used for coupling, according to known coupling methods.
  • the acid group of phosphatidic acid can be activated to form an active lipid anhydride, by reaction with a suitable anhydride, such as acetic anhydride, and the reactive lipid can then be joined to a protected polyalk 1- amine by reaction in the presence of an isothiocyanate reagent.
  • the derivatized lipid component are prepared to include a labile lipid-polymer linkage, suc as a peptide, ester, or disulfide linkage; which can b cleaved under selective physiological conditions, such as i the presence of peptidase or esterase enzymes present in th bloodstream.
  • Figure 5 shows exemplary lipids which are linke through (A) peptide, (B) , ester, and (C) , disulfide containin linkages.
  • the peptide-linked compound can be prepared, fo example, by first coupling a polyalkylether with the N terminal amine of the tripeptide shown, e.g., via the reactio shown in Figure 3.
  • the peptide carboxyl group can then b coupled to a lipid amine group through a carbodiimide couplin reagent conventionally.
  • the ester linked compound can b prepared, for example, by coupling a lipid acid, such a phosphatidic acid, to the terminal alcohol group of a poly alkylether, using alcohol via an anhydride coupling agent.
  • a lipid acid such as a phosphatidic acid
  • an anhydride coupling agent Alternatively, an short linkage fragment containing an inter nal ester bond an suitable end groups, such as primary amin groups can be used to couple the polyalkylether to the amphi pathic lipid through amide or carbamate linkages.
  • the linkage fragment may contain an internal disulfide link age, for use in forming the compound shown at C in Figure 5.
  • Figure 6 illustrates a method for derivatizing polylacti acid with PE.
  • the polylactic acid is reacted, in the presenc of PE, with dicyclohexylcarboimide (DCCI) , as detailed i Example 4.
  • DCCI dicyclohexylcarboimide
  • a vesicle-forming lipid derivatize with polyglycolic acid may be formed by reaction of polygly colic acid or glycolic acid with PE in the presence of suitable coupling agent, such as DCCI, also as detailed i Example 4.
  • the vesicle-forming lipids derivatized with eithe polylactic acid or polyglycolic acid form part of the inven tion herein.
  • liposome containing these derivatized lipids in a 1-20 mole percent.
  • lipid components used in forming the liposomes of the invention may be selected from a variety of vesicle-forming lipids, typically including phospholipids and sterols. As will be seen, one requirement of the liposomes of the present invention is long blood circulation lifetime. It is therefore useful to establish a standardized measure of blood lifetime which can be used for evaluating the effect of lipid component on blood halflife.
  • One method used for evaluating liposome circulation time in vivo measures the distribution of IV injected liposomes in the bloodstream and the primary organs of the RES at selected times after injection.
  • RES uptake is measured by the ratio of total liposomes in the bloodstream to total liposomes in the liver and spleen, the principal organs of the RES.
  • age and sex matched mice are injected intravenously (IV) through the tail vein with a radiolabeled liposome composition, and each time point is determined by measuring total blood and combined liver and spleen radiolabel counts, as detailed in Example 6.
  • the liposomes of the present invention include 1-20 mole percent of the vesicle-forming lipid derivatized with a hydro ⁇ philic polymer, described in Section I.
  • the phospholipid components may be composed of predominantly of fluidic, relatively unsaturated, acyl chains, or of more saturated, rigidifying acyl chain components.
  • Example 7 which examines blood/RES ratios in liposomes formed with PEG-PE, cholesterol, and PC having varying degrees of saturation (Table 4) .
  • the vesicle-forming lipids may be selected to achieve a selected degree of fluidity or rigidity, to control the stability of the liposomes in serum and the rate of release of entrapped drug from the liposomes in the blood ⁇ stream and/or tumor.
  • the vesicle-forming lipids may also be selected, in lipid saturation characteristics, to achieve desired liposome preparation properties. It is generally the case, for example, that more fluidic lipids are easier to formulate and down size by extrusion or homogenization than more rigid lipid components. In general, more fluidic lipids
  • Cholesterol, or related cholesterol derivatives may be important, however, in regulating the rate of release of lipo ⁇ some entrapped therapeutic compounds into the bloodstream.
  • PEG-liposome formulations containing high cholesterol (e.g., 30 mole percent or greater) released very little peptide or protein into serum in vitro, whereas decreasing amounts of cholesterol producing increasing loss of encapsulated poly- peptide.
  • the rate of release of compound from long-circulating liposomes can be controlled by the percent cholesterol included in the liposomes.
  • the vesicle-forming lipid derivatized with a hydrophilic polymer is present in an amount preferably between about 1-20 mole percent, on the basis of moles of derivatized lipid as a percentage of total moles of vesicle-forming lipids. It will be appreciated that a lower mole ratio, such as 0.1 mole percent, may be appropriate for a lipid derivatized with a large molecular weight polymer, such as one having a molecular weight greater than 100 kilodaltons.
  • the hydrophilic polymer in the derivatized lipid preferably has a molecular weight between about 200-20,000 daltons, and more preferably between about 1,000-5,000 daltons.
  • Example 8B which examines the effect of very short ethoxy ether moieties on blood/RES ratios indicates that polyether moieties of greater than about 5 carbon ether are required to achieve significant enhancement of blood/RES ratios.
  • the liposomes may be prepared by a variety of techniques, such as those detailed in Szoka et al, 1980.
  • One method for preparing drug-containing liposomes is the reverse phase evaporation method described by Szoka et al and in U.S. Patent No. 4,235,871.
  • the reverse phase evaporation vesicles (REVs) have typical average sizes between about 2-4 microns and are predominantly oligolamellar, that is, contain one or a few lipid bilayer shells.
  • the method is detailed in Example 5A. This method is generally preferred for preparing liposomes with encapsulated proteins high encapsulation efficiencies (up to 50%) are possible, and thus protein loss or problems of recovery and purification of non-encapsulated protein are reduced.
  • Multilamellar vesicles can be formed by simple lipid-film hydration techniques. In this procedure, a mixtur of liposome-forming lipids of the type detailed above dis solved in a suitable solvent is evaporated in a vessel to for a thin film, which is then covered by an aqueous medium, a detailed in Example 5B. The lipid film hydrates to form MLVs, typically with sizes between about 0.1 to 10 microns.
  • the liposomes for intravenous injection are prepared to have substantially homogeneous sizes in a selected size range between about 0.1 and 0.4, and preferably 0.1 to 0.2 micron size ranges. Liposomes in this size range have sufficiently high encapsulation volumes for carrying therapeutically effec ⁇ tive amounts of the compound to be administered. At lower liposome sizes, the ratio of liposome-encapsulated compound to free compound may too low to achieve a requisite initial dose level of liposome-encapsulated compound in the bloodstream or may not remain in circulation due to extravasation. At the same time, 0.1-0.4 micron liposomes are small enough to give long blood circulation times, as discussed below, and also to allow sterilization by sterile filtration.
  • One effective sizing method for REVs and MLVs involves extruding an aqueous suspension of the liposomes through a polycarbonate membrane having a selected uniform pore size, typically 0.4, 0.2, and/or 0.1 micron pore sizes.
  • the pore size of the membrane is proportional to the largest sizes of the liposomes which are produced, particularly where the pre ⁇ paration is extruded two or more times through the same size membrane.
  • This method of liposome sizing is used in preparing homogeneous-size REV and MLV compositions described in the examples below.
  • a more recent method involves extrusion through an asymmetric ceramic filter. The method is detailed in U.S. patent application for Liposome Extrusion Method, Serial No. 829,710, filed February 13, 1986. Homogenization methods are also useful for down-sizing liposomes.
  • the composition of the invention is used for slow-release delivery into the bloodstream or a poly ⁇ peptide (peptide or protein) which is therapeutically active in the bloodstream when administered IV, but which in free form has a short blood halflife, typically 4 ' hours or less.
  • polypeptides examples include hormones, such as vaso ⁇ pressin, calcitonin, oxytocin, somatotropin, human growth hormone, atrial naturectic factor (ANF) , and insulin; enzymes, such as superoxide dismutase, glucocerebrosidase, asparagin- ase, and adenosine deaminase; immunomodulators, such as inter- ferons (alpha, beta, and gamma), interleukin (1,2,3,4,5,6,7), tissue necrosis factor (TNF - alpha, beta) , and colony stimu ⁇ lating factors (M-CSF (macrophage) , G-CSF (granulocyte) , GM- CSF (granulocyte, macrophage) ; anticoagulants, such as TPA, prourokinase, and urokinase; vaccines, such as HIV-1 vaccine, hepatitis B vaccine, malaria vaccine
  • polypeptides useful in the invention typically hav relatively short blood halflives, on the order of 2-4 hours o less, and are active in the picogram/ml to nanogram/ml concen tration range in the blood.
  • polypeptide is preferably loaded passivel by the reverse-phase emulsion method for preparing liposomes, although many other methods, such as solvent injection o lipid hydration may be employed.
  • liposome formation an sizing, free (unbound) drug can be removed by a variety o methods, for example, by gel filtration or ion exchang chromatography or diafiltration.
  • the amount of fre peptide in the final sterilized composition is less than abou 20%, and preferably less than 10% of the total polypeptide contained in the composition.
  • the encapsulated compound is preferably present in an amount which, in a selected liposome dose, is between 3-20 times the amount of compound which would be given as a therapeutic dose in free form by IV administration.
  • a selected liposome dose will pre ⁇ ferably contain between about 3-20 ⁇ g of the peptide. Since 10-20% of this compound, e.g., 2-4 ⁇ g, may be in non-encapsu ⁇ lated form, it will be appreciated that the total amount of liposome which can be administered may be limited by the maxi- mum tolerated dose of the free compound.
  • liposomes can be administered by achieving higher ratios of encapsulated to non-encapsulated compound. In general, this ratio is increased with larger liposomes, more complete free drug removal from the liposome composition, and greater liposome stability on storage.
  • composition is also useful for slow-release delivery of a variety of non-peptide, water-soluble compounds which are effective in treating circulating cancers such as leukemias, for example, cytarabine, cyclophosphamide, carmustine, thio- guanine, bleomycin, daunorubicin, vinblastine, vincristine, and asparaginase.
  • leukemias for example, cytarabine, cyclophosphamide, carmustine, thio- guanine, bleomycin, daunorubicin, vinblastine, vincristine, and asparaginase.
  • antibiotics such as gentamicin, tobramycin, amikacin, netilmicin, kanamycin, and streptomycin, cefotaxime, ceftizoxi e, and ceftriaxone
  • anti-viral agents such as AZT (zidovidine) , DDI (dideoxyinosine) , DDC (didioxycytidine) , ganciclovir, D4T (didehydrodeoxythymidine) , phosphonoformate, ribavirin, and acyclovir.
  • anti-viral agents such as AZT (zidovidine) , DDI (dideoxyinosine) , DDC (didioxycytidine) , ganciclovir, D4T (didehydrodeoxythymidine) , phosphonoformate, ribavirin, and acyclovir.
  • Such compounds may be encapsulated by passive loading, as above, during liposome
  • drugs which form weak bases at physiological pH may be actively loaded into the liposomes at high drug concentration in the liposomes.
  • One method for active loading drugs into liposomes is described in co-owned U.S. patent application Serial No. 413,037, filed September 28, 1988.
  • liposomes are prepared in the presence of a relatively high ammonium ion, such as 0.125 M ammonium sulfate. After sizing the liposomes to a desired size, the liposome suspension is treated to create an inside- to-outside ammonium ion gradient across the liposomal membranes.
  • the gradient may be created by dialysis against a non-ammonium containing medium, such as an isotonic glucose medium, or by gel filtration, such as on a Sephadex G-50 column equilibrated with 0.15 M NaCl or KC1, effectively replacing ammonium ions in the exterior phase with sodium or potassium ions.
  • a non-ammonium containing medium such as an isotonic glucose medium
  • gel filtration such as on a Sephadex G-50 column equilibrated with 0.15 M NaCl or KC1
  • ammonium concentration inside the liposomes is preferably at least 10 times, and more preferably at least 100 to 1000 times that in the external liposome phase.
  • a rela ⁇ tively dilute suspension of the liposomes e.g., less than about 50 mM lipid
  • an aqueous solution of the drug e.g., less than about 50 mM lipid
  • the mixture is allowed to equilibrate over an extended period, e.g., 24 hours at room temperature.
  • a suspension of liposomes having a lipid con ⁇ centration of 25 mg/ml is mixed with an equal volume of anthracycline drug at a concentration of about 10 mg/ml. At the end of the incubation period, the suspension is treated to remove free (unbound) drug,
  • A. Extended Lifetime in the Bloodstream One of the requirements for extended compound release into the bloodstream, in accordance with the invention, is an extended liposome lifetime in the bloodstream with IV liposome administration.
  • One measure of liposome lifetime in the bloodstream in the blood/RES ratio determined at a selected time after liposome administration, as discussed above.
  • Blood/RES ratios for a variety of liposome compositions are given in Table 3 of Example 6. In the absence of PEG- derivatized lipids, blood/RES ratios were 0.03 or less.
  • the blood/RES ratio ranged from 0.2, for low-molecular weight PEG, to between 1.7- 4 for several of the formulations, one of which lacks cholesterol, and three of which lack a charged phospholipid (e.g., PG) .
  • the blood/RES values reported above can be compared with blood/RES values reported in co-owned U.S. Patent No. 4,920,016, which used blood/RES measurement methods identical to those used in for the data presented in Tables 3 and 5.
  • the best 24-hour blood/RES ratios which were reported in the above-noted patent was 0.9, for a formulation composed of GM lf saturated PC, and cholesterol.
  • the next best formulations gave 24-hour blood/RES values of about 0.5.
  • typical 24- hour blood/RES ratios obtained in a number of the current formulations were more than twice as high as the best formula ⁇ tions reported which have been reported to date.
  • ability to achieve high blood/RES with GM X or HPI lipids was dependent on the presence of predominantly saturated lipids in the liposomes.
  • Plasma kinetics of a liposomal marker in the bloodstream can provide another measure of the enhanced liposome lifetime which is achieved by the liposome formulations of the present invention.
  • Figure 7 and 8 discussed above show the slow loss of liposomal marker over a 24 hour period in typical PEG-lipo- some formulations, substantially independent of whether the marker is a lipid or an encapsulated water-soluble compound
  • Figure 9 shows the kinetics of liposome loss from the bloodstream for a typical PEG-lipsome formulation and the same liposomes in the absence of a PEG-derivatized lipid. After 24 hours, the percent marker remaining in the PEG-liposomes was greater than about 20%, whereas the conventional liposomes showed less than 5% retention in the blood after 3 hours, and virtually no detectable marker at 24 hours.
  • liposome size was been investigated by comparing loss of liposomal marker in intra- venously injected liposomes having selected sizes between about 0.1 and 0.25 microns. Experimental details are given in Example 10. The results, given in Figure 10, show about 10% or greater liposome marker present in the blood after 24 hours for each of the liposome formulations. Highest blood life- times were achieved with the smallest liposomes. Thus, although all of the liposome preparations give high blood lifetimes, it is also clear that liposome size can be selected to produce desired increase or decrease in total drug release time.
  • the liposome size between 0.1 and 0.4 microns allows relatively high compound loading in the lipo ⁇ somes, for effective compound release as the percentage of liposomes in the bloodstream even at relatively low liposome concentrations in the bloodstream. This is typically an important consideration since the total amount quantity of liposomes which can be administered will be limited, due to the unavoidable presence of some free compound in injected composition.
  • Example 16 The study reported in Example 16 demonstrates a simila ability to control release rates of large proteins from long- circulating liposomes.
  • PEG-liposomes containing encapsu ⁇ lated M-CSF (a 55 kilodalton protein) were examined for per- cent retention in the bloodstream, of both lipid and protein components.
  • the data plotted in Figure 15 show liposome lipi (solid triangles) and protein (solid circles) kinetics 24 hours after IV injection for a PEG-lipsome formulation con ⁇ taining 30 mole percent cholesterol. The data indicate about 20% loss of encapsulated material over 24 hours, as discusse in Example 16.
  • Figure 17 shows a similar plot for PEG-liposomes without cholesterol.
  • the data here show a 40-50% loss of M-CSF after 24 hours.
  • the plots o protein release from cholesterol (solid circles) and no cholesterol (solid triangles) PEG-liposomes are obtained.
  • the plots illustrate the markedly different protein release kinetics which can be obtained with long circulating lipo- somes, by varying cholesterol content of the liposomes.
  • Another feature of the data shown in Figure 17 is th relatively high percent (at least 3%) of initially injecte protein which is released at 24 hours in both formulations. IV. Intravenous Liposome Treatment
  • the invention includes, in one aspect, a method for extending to at least 24 hours, the period of effective activity of an therapeutic compound.
  • the compound is one which can be administered intravenously in a therapeutically effective amount, and which has a halflife in the bloodstream, in free form, of less than about 4 hours.
  • a liposome composition such as described above, containing the compound in liposome-entrapped form.
  • the size, lipid composition, and extent compound loading of the drug are selected, according to the desired release rates and total release times, as con ⁇ sidered in Section III above.
  • the composition is injected intravenously to a subject at a dose which contains an amount of the compound which is at least 3, and typically 5-20 times the therapeutically effec ⁇ tive dose.
  • a therapeutically effective dose of a compound is 10 ⁇ g/Kg body weight
  • the liposome com- position would be injected at a liposome dose of at least 30 ⁇ g compound/Kg, and typically between 50-200 ⁇ g compound/Kg body weight.
  • the amount of material which can be injected is generally limited by the maximum tolerated dose of free compound, since 5-20% or more of the compound may be in free (non-entrapped) form when the composition is admi ⁇ nistered.
  • the maximum tolerated dose of a compound in free form is 2 ⁇ g/Kg body weight
  • the lipo ⁇ some composition contains 10% non-entrapped compound
  • the highest dose of liposome composition which can be given is 20 ⁇ g/Kg body weight.
  • Figure 13 shows the effect, measured in percent predosage urine flow, of IV injection of free vaso ⁇ pressin at dose levels of 0.2 ⁇ g (solid squares), 0.8 ⁇ g (solid triangles) and 2.0 ⁇ g (solid circles) .
  • the reduction in urine flow is maximum after 1 day, but returns to control levels (open circles) by day 2.
  • Figure 14 shows the effect on urine flow by vasopressin administered IV in PEG-liposomes, at dosage levels of 2 ⁇ g (solid squares) , 8 ⁇ g (solid triangles) and 24 ⁇ g (solid cir ⁇ cles) .
  • a pronounced reduction in urine flow was observed within one day, with a slight rebound toward control values by day 2 — an effect likely due, in part, to free peptide in the liposome formulation.
  • a marked reduction in urine flow was observed for at least 7 days following IV administration. It is noted that the long-term therapeutic effect is saturated at an 8 ⁇ g dose, with no further effect observed at 24 ⁇ g.
  • the long-live liposome composition of the invention is also effective for slow release of a liposome- entrapped compound from a subcutaneous (SubQ) site into the bloodstream.
  • a subcutaneous site into the bloodstream.
  • therapeutic effects of up to 3 weeks or more can be achieved by a single subcutaneous injection of the liposome composition of the invention.
  • the experimental model used to demonstrate this method is the vasopressin model described above, and detailed in Example 17. Briefly, a treatment method involving SubQ injection of free vasopressin was compared, for duration of physiological effect, with vasopressin administered SubQ in PEG-liposomes.
  • Figure 18A shows the depression in urine production observed with SubQ administration of free vasopressin at doses of 2 ⁇ g
  • FIG. 18B shows the effect of SubQ administration of the peptide in PEG liposomes. The lowest does administered, 25 ⁇ g (closed triangles) gave a slight prolongation of activity, to about 8 days. A vasopressin dose of 100 ⁇ g (closed circles) showed a significant reduction in urine output out to about 20 days. An even longer effect, of at least 24 days, was observed at a dose of 400 ⁇ g (closed squares) .
  • the treatment method employing vasopressin is designed for treatment of diabetes insipidus, by long-term administration of vasopressin from a SubQ site. More generally, the method is designed for extending to at least one week, the period of effective activity of an therapeutic compound which can be administered intravenously in a therapeutically effective amount.
  • the method utilizes liposomes (i) composed of vesicle-forming lipids and between 1-20 mole percent of a vesicle-forming lipid derivatized with a hydrophilic polymer, and (ii) having a selected mean particle diameter in the size range preferably between about 0.07-.15 microns, and having the compound in liposome-entrapped form.
  • the liposome composition is administered subcutaneously at a dose of the composition which contains an amount of the liposome-entrapped compound which is at least ten times the therapeutically effective intravenously administered amount.
  • the method can be used for therapeutic delivery of the compound over a several- week period.
  • the ability of the liposome composition of the invention to produce a long-term therapeutic effect from a SubQ site suggests that the liposomes taken up from this site through the lymphatics may be able to successfully evade the normal lymphatic clearance mechanisms, including Kupfer cells.
  • This mechanism is supported by studies on araC administration by the interperitoneal (IP) route by PEG-liposomes. Briefly, it was found that PEG-liposomes containing entrapped araC and administered by IP route produced a 250-300% increase in survial in animals having L1210 leukemias, compared with about 120% increase in survival with the free drug given by the IP route. The results suggests that PEG-liposomes are capable of migrating from the peritoneum through the lymphatics into the bloodstream.
  • Cholesterol Cholesterol (Choi) was obtained from Sigma (St. Louis, MO) .
  • Sphingomyelin (SM) egg phosphatidylcholine (lecithin or PC), partially hydrogenated PC having the composition IV40, IV30, IV20, IV10, and IV1, phosphatidylglycerol (PG) , phospha- tidylethanolamine (PE) , dipalmitoyl-phosphatidyl glycerol (DPPG) , dipalmitoyl PC (DPPC) , dioleyl PC (DOPC) and distear- oyl PC (DSPC) were obtained from Avanti Polar Lipids (Birming ⁇ ham, AL) or Austin Chemical Co (Chicago, II) .
  • Cyanuric chloride (5.5 g; 0.03 mol) was dissolved in 400 ml of anhydrous benzene containing 10 g of anhydrous sodium carbonate, and PEG-1900 (19 g; 0.01 mol) was added and the mixture was stirred overnight at room temperature. The solu ⁇ tion was filtered, and 600 ml of petroleum ether (boiling range, 35-60°) was added slowly with stirring. The finely divided precipitate was collected on a filter and redissolved in 400 ml of benzene.
  • the activated PEG was analyzed for nitrogen and an appropriate correction was applied in selecting the quantity of reactant to use in further synthetic steps.
  • the quantity of material used in the next synthetic step was increased by 100/20, or 5-fold.
  • the product contained 50% of the theoretical amount of nitrogen only 100/50 or a 2-fold increase was needed.
  • the solid compound was taken up in 24 ml of ethanol/- chloroform; 50/50 chloroform and centrifuged to remove insoluble material. Evaporation of the clarified solution to dryness under vacuum afforded 21 mg (7.62 ' micromoles) of colorless solid.
  • the reaction mixture was cooled and the clear solution formed at room temperature.
  • the solution was diluted to 50.0 ml with dry benzene and stored in the refrigerator as a 100 micromole/ml stock solution of the imidazole carbamate of PEG ether 1900.
  • the solvent from the remaining reaction mixture was evapo rated under vacuum.
  • the residue was taken up in 10 m methylene chloride and placed at the top of a 21 mm x 270 m chromatographic absorption column packed with Merck Kieselge 60 (70-230 mesh silica gel) , which has been first rinsed wit methylene chloride.
  • the mixture was passed through th column, in sequence, using the following solvents.
  • Air from the reaction vessel was displaced with nitrogen.
  • the vessel was closed and heated in a sand bath 110°C for 22 hours.
  • the solvent was evaporated under vacuum to obtain 1.58 grams of brownish- colored oil.
  • the crude product was dissolved in 3 ml of the same solvent and transferred to the top of the chromatography column.
  • the chromatogram was developed with the same solvent and sequential 30 ml portions of effluent were assayed each by TLC.
  • the TLC assay system used silica gel coated glass plates, with solvent combination butanone/acetic acid/water; 40/25/5; v/v/v. Iodine vapor absorption served for visualization.
  • the desired N-1-trimethylsilyloxy polyethylene glycol 1500 PE was a chief constituent of the 170-300 ml portions of column effluent. When evaporated to dryness under vacuum these portions afforded 111 mg of pale yellow oil of compound.
  • PE compound was dissolved in 2 ml of tetrahydrofuran. To this, 6 ml acetic acid and 2 ml water was added. The resulting solution was let to stand for 3 days at 23°C. The solvent from the reaction mixture was evaporated under vacuum and dried to constant weight to obtain 75 mg of pale yellow wax. TLC on Si-C18 reversed-phase plates. developed with a mixture of 4 volumes ethanol, 1 volume water indicated that some free PE and some polyglycol-like materia formed during the hydrolysis.
  • N-Polyethylene glycol 1500 P.E. by fluorid deprotection.
  • 500 mg of crude N-1-trimethylsilyloxy polyethylene glyco PE was dissolved in 5 ml tetrahydrofuran and 189 mg (0.60 millimoles) of tetrabutyl ammonium fluoride was added an agitated until dissolved.
  • the reactants were let to stan over night at 20°C.
  • the solvent was evaporated under reduced pressure and th residue was dissolved in 10 ml chloroform, washed with tw successive 10 ml portions of water, and centrifuged t separate chloroform and water phases.
  • the chloroform phas was evaporated under vacuum to obtain 390 mg of orange-brown wax, which was determined to be impure N-polyethylene glycol 1500 PE compound.
  • the wax was re-dissolved in 5 ml chloro ' form and trans- ferred to the top of a 21 X 270 mm column of silica gel moistened with chloroform.
  • the column was developed by passing 100 ml of solvent through the column.
  • the Table 2 solvents were used in sequence:
  • the tube was cooled to room temperature, and 6 ml of chloroform added.
  • the chloroform solution wa washed with three successive 6 ml volumes of water, centrifuged after each wash, and the phases separated with Pasteur pipette.
  • the remaining chloroform phase was filtere with suction to remove suspended distearolyphosphatidyl ethanola ine.
  • the filtrate was dried under vacuum to obtai 212 mg of semi-crystalline solid.
  • This solid was dissolved in 15 ml of a mixture of volumes ethanol with 1 volume water and passed through a 50 m deep and 21 mm diameter bed of H + Dowex 50 cation exchang resin, and washed with 100 ml of the same solvent.
  • Phosphate analysis suggests a molecular weight of 924,000.
  • Example 5 Preparation of REVs and MLVs A. Sized REVs A total of 15 ⁇ moles of the selected lipid components, in the mole ratios indicated in the examples below, were dis ⁇ solved in chloroform and dried as a thin film by rotary evapo ⁇ ration. This lipid film was dissolved in 1 ml of diethyl ether washed with distilled water. To this lipid solution was added 0.34 ml of an aqueous buffer solution containing 5 ⁇ i Tris, 100 mM NaCl, 0.1 mM EDTA, pH 7.4, and the mixture was emulsified by sonication for 1 minute, maintaining the tempe rature of the solution at or below room temperature. Wher the liposomes were prepared to contain encapsulated [1251] tyraminyl-inulin, such was included in the phosphate buffer at a concentration of about 4 ⁇ Ci/ml buffer.
  • the ether solvent was removed under reduced pressure at room temperature, and the resulting gel was taken up in 0.1 ml of the above buffer, and shaken vigorously.
  • the resulting REV suspension had particle sizes, as determined by microscopic examination, of between about 0.1 to 20 microns, and was com ⁇ posed predominantly of relatively large (greater than 1 micron) vesicles having one or only a few bilayer lamellae.
  • the liposomes were extruded twice through a polycarbonate filter (Szoka, 1978), having a selected pore size of 0.4 microns or 0.2 microns. Liposomes extruded through the 0.4 micron filter averaged 0.17+ (0.05) micron diameters, and through the 0.2 micron filter, 0.16 (0.05) micron diameters. Non-encapsulated [1251] tyraminyl-inulin was removed by passing the extruded liposomes through Sephadex G-50 (Phar ⁇ macia) .
  • Multilamellar vesicle (MLV) liposomes were prepared according to standard procedures by dissolving a mixture of lipids in an organic solvent containing primarily CHC1 3 and drying the lipids as a thin film by rotation under reduced pressure.
  • a radioactive label for the lipid phase was added to the lipid solution before drying.
  • the lipid film was hydrated by addition of the desired aqueous phase and 3 mm glass beads followed by agitation with a vortex and shaking above the phase transition temperature of the phospholipid component for at least 1 hour.
  • a radioactive label for the aqueous phase was included in the buffer.
  • the hydrated lipid was repeatedly frozen and thawed three times to provide for ease of the following extrusion step.
  • the size of the liposome samples was controlled by extru ⁇ sion through defined pore polycarbonate filters using pres ⁇ surized nitrogen gas.
  • the ' liposomes were extruded one time through a filter with pores of 0.4 ⁇ m and then ten times through a filter with pores of 0.1 ⁇ m.
  • the liposomes were extruded three times through a filter with 0.2 ⁇ m pores followed by repeated extrusion with 0.05 ⁇ m pores until the mean diameter of the particles was below 100 nm as determined by DLS.
  • Unencapsu- lated aqueous components were removed by passing the extruded sample through a gel permeation column separating the lipo ⁇ somes in the void volume from the small molecules in the included volume.
  • PEG-PE composed of methoxy PEG, molecular weight 1900 and l-palmitoyl-2-oleyl-PE (POPE) was prepared as in Example 2.
  • the PEG-POPE lipid was combined with and partially hydrogen- ated egg PC (PHEPC) in a lipid:lipid mole ratio of about 0.1:2, and the lipid mixture was hydrated and extruded through a 0.1 micron polycarbonate membrane, as described in Example 4, to produce MLVs with average size about 0.1 micron.
  • the MLV lipids included a small amount of radiolabeled lipid marker 14 C-cholesteryl oleate, and the encapsulated marker 3 H- inulin.
  • the liposome composition was injected and the percent initial injected dose in mice was determined as described in Example 4, at 1, 2, 3, 4, and 24 after injection.
  • the time course of loss of radiolabeled material is seen in Figure which is a plot of percent injected dose for encapsulate inulin (solid circles), inulin marker corrected to the initia injection point of 100% (open circles) , and lipid marke (closed triangles), over a 24-hour period post injection. A seen, both lipid and encapsulated markers showed greater tha 10% of original injected dose after 24 hours.
  • PEG-PE composed of methoxy PEG, molecular weight 1900 and distearylPE (DSPE) was prepared as in Example 2.
  • the PEG-PE lipids were formulated with selected lipids from among sphin- gomyelin (SM) , fully hydrogenated soy PC (PC) , cholesterol (Choi) , partially hydrogenated soy PC (PHSPC) , and partially hydrogenated PC lipids identified as PC IV1, IV10, IV20, IV30, and IV40 in Table 4.
  • SM sphin- gomyelin
  • PC fully hydrogenated soy PC
  • PHSPC partially hydrogenated soy PC
  • PC lipids identified as PC IV1, IV10, IV20, IV30, and IV40 in Table 4.
  • the lipid components were mixed in the molar ratios shown at the left in Table 5, and used to form MLVs sized to 0.1 micron as described in Example 4.
  • the blood and L+S (RES) values were used to calculate a blood/RES value for each composition.
  • the column at the right in Table 4 shows total amount of radioactivity recovered.
  • the two low total recovery values in the table indicate anomalous clearance behavior.
  • the results from the table demonstrate that the blood/RES ratios are largely independent of the fluidity, or degree of saturation of the phospholipid components forming the lipo ⁇ somes. In particular, there was no systematic change in blood/RES ratio observed among liposomes containing largely saturated PC components (e.g., IV1 and IV10 PC's), largely unsaturated PC components (IV40) , and intermediate-saturation components (e.g., IV20) .
  • largely saturated PC components e.g., IV1 and IV10 PC's
  • IV40 largely unsaturated PC components
  • IV20 intermediate-saturation components
  • the PEG-PE lipids were formulated with selected lipids from among sphin- gomyelin (SM) , fully hydrogenated soy PC (PC) , and cholesterol
  • Methoxy-ethyoxy-cholesterol was prepared by coupling methoxy ethanol to cholesterol via the trifluorosulfonate coupling method described in Section I.
  • PEG-PE composed of methoxy PEG, molecular weight 1900 and was derivatized DSPE as described in Example 6.
  • the PEG-PE lipids were formulated with selected lipids from among distearylPC (DSPC) , partially hydrogenated soy PC (PHSPC) , cholesterol, and ethoxylated cholesterol, as indicated at the right in Table 7.
  • DSPC distearylPC
  • PHSPC partially hydrogenated soy PC
  • cholesterol ethoxylated cholesterol
  • Example 9 Effect of Charged Lipid Components on Blood/RES Ratios in PEG-PE Liposomes
  • PEG-PE composed of methoxy PEG, molecular weight 1900 and was derivatized DSPE as described in Example 6.
  • the PEG- PE lipids were formulated with lipids selected from among egg PG (PG) , partially hydrogenated egg PC (PHEPC) , and choleste ⁇ rol (Choi), as indicated in the Figure 6.
  • the two formula ⁇ tions shown in the figure contained about 4.7 mole percent (triangles) or 14 mole percent (circles) PG.
  • the lipids were prepared as MLVs, sized to 0.1 micron as in Example 4.
  • the percent of injected liposome dose present 0.25, 1, 2, 4, and 24 hours after injection are plotted for both formula ⁇ tions in Figure 6. As seen, the percent PG in the composition had little or no effect on liposome retention in the bloodstr ⁇ eam. The rate of loss of encapsulated marker seen is also similar to that observed for similarly prepared liposomes containing no PG.
  • PEG-DSPE prepared as above with PEG-1900, was formulated with partially hydrogenated egg PC (PHEPC) , and cholesterol (Choi), at a mole ratio of 0.15: 1.85: 1.
  • PHEPC partially hydrogenated egg PC
  • Choi cholesterol
  • the liposomes were sized by extrusion through 0.25, 0.15 or 0.1 micron polycarbo ⁇ nate filters, to produce liposome sizes of about 0.4, 0.2, and 0.1 microns, respectively.
  • Non-encapsulated 3 H-inulin was removed by gel filtration.
  • each of the three liposome were injected intravenously, and the percent of injected liposome marker in the blood was measured at 1, 2, 3, 4, and 24 hours, with the results shown in Figure 10. All three formulations show long blood half- lives, as evidence by at least about 10% liposome marker remaining after 24 hours.
  • the 0.1 micron formulation (solid squares) is longer lived than the 0.2 micron formulation (solid circles) which is in turn longer lived than the 0.4 micron formulation.
  • Example 11 Effect of Other Hydrophilic Polymers on Blood Lifetime Polylactic acid-DSPE or polyglycolic acid-DSPE, prepared as above, was formulated with lipids selected from among hydrogenated soy PC (HSPC) , and cholesterol (Choi) , at a weight ratio of either 2: 3.5: 1 or 1: 3.5: 1.
  • the liposomes were sized by extrusion through a 0.1 micron polycarbonate filter.
  • the liposomes were labeled by Ga-DF as described in Example 5.
  • These liposome formulations were injected intravenously, and the percent of injected liposome marker in the blood was measured at 1, 2, 3, 4, and 24 hours, with the results shown in Figure 11.
  • PEG-PE composed of methoxy PEG, molecular weight 1900 and distearyl PE (DSPE) was prepared as in Example 2.
  • the PEG-PE lipids were formulated with PHEPC, and cholesterol, in a mole ratio of 0.15:1.85:1.
  • a second lipid mixture contained the same lipids, but without PEG-PE.
  • Liposomes were prepared from the two lipid mixtures as described in Example 5, by lipid hydration in the presence of DF followed by sizing to 0.1 micron, removal of non-entrapped DF by gel filtration, and Ga labeling as described in Example 5. Both compositions contained 10 ⁇ M lipid/ml in 0.15 M NaCl, 0.5 mM desferal.
  • the two liposome compositions (0.4 ml) were injected IV in animals, as described in Example 6. At time 0.25, 1, 3 or 5, and 24 hours after injection, blood samples were removed and assayed for amount inulin remaining in the blood, expressed as a percentage of the amount measured immediately after injection. The results are shown in Figure 9. As seen, the PEG-coated liposomes have a blood halflife of about 11 hours, and nearly 30% of the injected material is present in the blood after 24 hours. By contrast, uncoated liposomes showed a halflife in the blood of less than 1 hour. At 24 hours, the amount of injected material was undetectable.
  • the vein was nicked and a bevelled 6-inch section of poly ⁇ ethylene catheter tubing (PE_50 Clay-Adams) filled with normal saline (0.9%) or 5% dextrose was inserted into the vein. Correct placement was verified by venous drawback and flush using a 1 cc syringe. The catheter was anchored loosely to the vein by passing a small tie under the vessel and knotting it around the cannula.
  • PE_50 Clay-Adams poly ⁇ ethylene catheter tubing
  • test solution 150-1000 ⁇ l of test solution was administered intravenously.
  • the dose volume administered was adjusted to give the desired total amount of vasopressin, for example 0.2, 0.8, and 3 ⁇ g of aqueous vaso- pressin.
  • the venous catheter was then removed, the femoral vein was tied off, and both leg incisions were closed. The animals were allowed to regain consciousness.
  • Figure 12 shows the percent predosage urine flow in animals treated with saline (open circles) or 0.2 ⁇ g (solid squares) 0.8 ⁇ g (solid triangles), and 2 ⁇ g (solid circles) vasopressin solution.
  • urine production showed a dose-dependent drop.
  • urine production was substantially back to control levels.
  • lipid composition used in preparing the liposomes was PEG-DSPE, PC, sphingomyelin, and cholesterol, in a mole ratio 0.2: 1: 1: 1.
  • Lipid mixtures for a 1 ml final volume of liposomes with a phospholipid concentration of 10 ⁇ mol/ml were dissolved in chloroform and evaporated to dryness under reduced pressure. After residual solvent was removed by high vacuum for 1 hour the mixture was dissolved in ethyl ether freshly washed with phosphate buffer at pH 7.
  • aqueous buffer (5 mM Tris, 100 mM NaCl, 0.1 mM EDTA) containing sufficient arginine vasopressin to give a final drug concentration of 510 ⁇ g/ml was added to the lipids in ether.
  • a trace amount of 3 H-labeled vasopressin was added to the vasopressin solution for determination of protein concentrations.
  • the ether was removed by controlled rotoevaporation, and additional drug-free buffer added to give a 1 ml solution immediately after gel dispersion.
  • the vasopressin content of the resulting liposomes was determined by separating free from liposome-bound by gel filtration on Bio-Gel A15.
  • the liposomes were extruded through 0.4 ⁇ m Nuclepore filters (Olsen et al., 1979) and particle size distribution measured by dynamic light scattering with a Nicomp model 200. Mean diameter ranged between 0.2 ⁇ m and 0.5 ⁇ m and showed a low polydispersity. Unbound vasopressin was removed by dialysis and monitored by gel chromatography as above.
  • PEG-liposome preparations from above were administered intravenously to rats prepared as in Example 13, at liposomal doses 2 ⁇ g (solid squares) 8 ⁇ g (solid triangles) , and 24 ⁇ g (closed circles) vasopressin solution.
  • Percent predosage urine flow was measured as above, with the results shown in Figure 14.
  • the data show the substantially the same dose- dependent depression in urine production in the first day after drug administration, presumably resulting predominantly from non-entrapped vasopressin in the lipsome formulations. In contrast to free peptide administration, however, all three formulation produced a significant inhibition in urine production with respect to control (open circles) over a 2-8 day period after liposome administration.
  • Example 15 Treatment with PEG-Liposomal Vasopressin: Effect of Liposome Cholesterol Concentration Large unilamellar liposomes were prepared as in Example 14, with liposomes containing either 33, 16, or 0 mole percent cholesterol. Vasopressin encapsulation, lipsome sizing, and free peptide removal was carried out as in Example 14.
  • the three PEG-liposome preparations were administered intravenously to rats prepared as in Example 13, at liposomal doses giving 8 ⁇ g vasopressin, and the percent predosage urine production was measured over a 9 day period following liposome administration.
  • the results are shown in Figure 15, for saline control (open circles) and PEG-liposomes with 0 (solid squares) , 16 (solid triangles) , and 33 (solid circles) mole percent cholesterol.
  • the day-1 response shows a marked dependence on percent cholesterol, with the greatest effect on urine flow being produced in liposomes with the lowest mole ratio of choleste ⁇ rol.
  • This result is consistent with in vitro stability studies of vasopressin release from PEG-liposomes in serum: In the presence of serum, little release of vasopressin was seen in liposomes containing greater than 30 mole percent cholesterol. By contrast, formulations containing reduced amounts of cholesterol showed increasingly higher release rates of encapsulated peptide. Thus, it would appear that the significantly higher diuretic effect seen after 1 day with low and no cholesterol formulations is due to the presence of free peptide released from the liposomes in serum.
  • Example 16 Blood Clearance Kinetics of M-CSF from PEG-Liposomes A lipid film containing PEG-DSPE, PHEPC IV-40, cholesterol, and ⁇ -tocopherol, in a mole ratio 5:61:33:1, was hydrated with distilled water, and the resulting MLVs were sonicated for 30 minutes to form SUVs. M-CSF was concentrated and a portion of the protein was labeled with U125I-iodine.
  • Equal volumes (2 ml) of the protein solution and SUVs were mixed, and the mixture was frozen in an acetone/dry bath, and lyophilized overnight.
  • the dried material was rehydrated in 0.8 ml of distilled water, and the resulting liposome suspension was extruded 1 time through a 0.4 ⁇ polycarbonate filter, and 3 times through a 0.2 ⁇ filter.
  • the sized liposomes were diluted to 10 ml in distilled water, washed washed two times with high speed centrifugation, and the washed pellet was resuspended in 0.9 ⁇ l of sterile buffer, to a final concentration of 40 ⁇ ol lipid/ml and between 0.5 and 1.25 mg protein/ml.
  • PEG-liposome preparations from above were administered intravenously to animals as in Example 6, and the blood levels of M-CSF were measured at 1, 2, 4, and 24 hours after rats prepared as in Example 14, at liposomal doses 2 ⁇ g (solid squares) 8 ⁇ g (solid triangles) , and 24 ⁇ g (closed circles) vasopressin solution. Similar measurements were made for an equivalent amount of M-CSF administered in solution form.
  • the plasma kinetics for the PEG-liposome formulation containing 30 mole percent cholesterol are shown in Figure 15. The data show rapid clearance of free protein (solid triangles) with less than 1% protein remaining in the blood at 24 hours, compared with about 8% for liposome-associated protein (solid circles) .
  • Percent liposomes remaining in the bloodstream was slightly greater than 10%.
  • a comparison of the clearance of rates of the lipid and pro ⁇ tein markers indicates that about 20% of the protein marker was released from the liposomes by 24 hours post injection.
  • the plasma kinetics obtained with cholesterol-free PEG- liposomes is shown in Figure 16.
  • Percent liposomes remaining after 24 hours was about 8.5 (solid triangles) compared with about 4.5% for liposome-associated M-CSF.
  • the radioactive counts in the liposome lipid marker and in the encapsulated protein were normalized to 100% initial values, and the percent injected dose released into the blood ⁇ stream over time was then determined from the ' difference be- tween the normalized protein and normalized liposome marker radioactivity levels.
  • a plot of the calculated values of percent protein released at 1, 2, 4, and 24 hours post injec ⁇ tion is shown in Figure 17.
  • the plot for the cholesterol-free formulation shows a protein release peak at 2 hours, with a gradual decline in amount released in the 2-24 hour period in the no-cholesterol formulation (solid triangles) .
  • the amount of protein released from the liposomes at 24 hours was between 3-4 percent of the total administered.
  • the plot for the formulation containing 30 mole percent cholesterol shows a gradual increase in release protein release rate over 24 hours.
  • the amount of protein released from the liposomes at 24 hours was about 3 percent of the total administered.
  • both formulations showed relatively high levels of protein release (3% or greater) at 24 hours.
  • Example 17 Subcutaneously Administered Liposomes
  • the lipid composition of the thin film was PEG- DSPE, HEPC, and cholesterol, in a mole ratio 0.15:1.85:1.
  • the thin film was hydrated with an aqueous buffer (5 mM Tris, 100 mM NaCl, 0.1 mM EDTA) containing arginine vasopressin at 7.5 mgs/ml.
  • the MLVs were sized by repeated extrusion through a 0.1 micron polycarbonate membrane, and free (non-encapsulated) peptide was removed by gel filtration, as in Example 15. The final concentration of PEG-liposomes in the suspension was 100 ⁇ M/ml.
  • Vasopressin in free form was administered subcutaneously (1 ml) to Brattleboro rats, as in Example 13.
  • the site of subcutaneous injection was the dorsal neck region.
  • the doses administered were 2 ⁇ g (solid triangles) , 25 ⁇ g (solid circles) , 50 ⁇ g (solid squares) , and 100 ⁇ g (solid diamonds) .
  • the percent of predosage urine flow observed is plotted in Figure 18A.
  • the percent of predosage urine flow observed is plotted in Figure 18B.

Abstract

A liposome composition for extended release of a therapeutic compound into the bloodstream. The liposomes are composed of vesicle-forming lipids and between 1-20 mole percent of a vesicle-forming lipid derivatized with hydrophilic polymer, have sizes in a selected size range between 0.1 and 0.4 microns, and contain the therapeutic compound in liposome-entrapped form. The dosage form of the composition contains at least about three times the dose of the compound required for intravenous injection in free form. Also disclosed is a method for extending to at least 24 hours the period in which an intravenously administered therapeutic compound is therapeutically active in the blood-stream, and novel liposomes compositions for practicing the method.

Description

LIPOSOIIE MICRORESERVOIR COMPOSITION AMD METHOD
1. Field of the Invention The present invention relates to a liposome composition and method for administering a therapeutic compound into the bloodstream over an extended period.
2. References Allen, T.M., (1981) Biochem. Biophys. Acta 640. 385397. Allen, T.M., and Everest, J. (1983) J. Pharmacol. Exp. Therap. 226. 539-544.
Ashwell, G., and Morell, A.G. (1974) Adv. Enzymology 41, 99-128. Banga, A.K., et al., Int J Pharm, 48:15 (1988).
Czop, J.K. (1978) Proc. Natl. Acad. Sci. USA 75:3831. Durocher, J.P., et al. (1975) Blood 45:11. Ellens, H., et al. (1981) Biochim. Biophys. Acta 674: 10-18. Gregoriadis, G., and Ryman, B.E. (1972) Eur. J. Biochem. 24_, 485-491.
Gregoriadis, G., and Neerunjun, D. (1974) Eur. J. Biochem. £7, 179-185.
Gregoriadis, G., and Senior, J. (1980) FEBS Lett. 119, 43-46.
Greenberg, J.P., et al (1979) Blood 53:916. Hakomori, S. (1981) Ann. Rev. Biochem. 50, 733-764. Hwang, K.J., et al. (1980) Proc. Natl. Acad. Sci. USA 72:4030.
Jonah, M.M., et al. (1975) Biochem. Biophys. Acta 401, 336-348. Juliano, R.L., and Stamp, D. (1975) Biochem. Biophys. Res. Commun. 63. 651-658.
Karlsson, K.A. (1982) In: Biological Membranes, Vol. 4, D. Chapman (ed.) Academic Press, N.Y., pp. 1-74.
Kimelberg, H.K., et al. (1976) Cancer Res. 36,2949-2957. Lee, K.C., et al., J. Immunology 125:86 (1980).
Lee, V.H.L., Phar Int, 7:208 (1986).
Lee, V.H.L., Biopharm Manuf, 1:24 (1988).
Lopez-Berestein, G., et al. (1984) Cancer Res. 44, 375-378. Okada, N. (1982) Nature 299:261.
Poste, G., et al., in "Liposome Technology" Volume 3, page 1 (Gregoriadis, G., et al, eds.), CRC Press, Boca Raton (1984);
Poznansky, M.J., and Juliano, R.L. (1984) Pharmacol. Rev.
Figure imgf000004_0001
Richardson, V.J., et al. (1979) Br. J. Cancer 40_, 3543.
Scherphof, T., et al. (1978) Biochim.Biophys. Acta 542, 296-307.
Senior, J., and Gregoriadis, G. (1982) FEBS Lett. 145, 109-114.
Senior, J., et al. (1985) Biochim. Biophys. Acta 839, 1-8.
Szoka, F., Jr., et al. (1978) Proc. Natl. Acad. Sci. USA 75:4194. Szoka, F., Jr., et al. (1980) Ann. Rev. Biophys. Bioeng. : 67.
Woodruff, J.J., et al. (1969) J. Exp. Med. 129:551. 3. Background of the Invention
With recent advances in biotechnology, the development of medicinal peptides or proteins has become an integral parti of the pharmaceutical industry (Lee, 1986, 1988) .' Several thera- peutic proteins have been successfully produced through recom- binant DNA technology, such as human growth hormone, human insulin, α-interferon, interleukin-2, TPA, and a variety of peptide vaccines, all of which are now commercially available (Banga) . As oral administration generally does not result in therapeutic responses, the parenteral route is preferred. However, when administered parenterally, most peptides and proteins have an extremely short half-life in the bloodstream, typically less than 2 hours, and thus require large doses and multiple daily injections or infusions. Often, the therapeu- tic regimens employed require close medical supervision and are difficult for most patients to accept.
Liposomes have been proposed as a carrier for intraven¬ ously (IV) administered compounds. However, the use of lipo¬ somes for slow release of liposome-entrapped material into the bloodstream has been severely restricted by the rapid clear¬ ance of liposomes from the bloodstream by cells of the reti- culoendothelial system (RES) . Typically, the RES will remove 80-95% of IV injected liposomes within one hour, and effec¬ tively remove circulating liposomes from the bloodstream within of 4-6 hours.
A variety of factors which influence the rate of RES uptake of liposomes have been reported (e.g., Gregoriadis, 1974; Jonah; Gregoriadis, 1972; Juliano; Allen, 1983; Kimelberg, 1976; Richardson; Lopez-Berestein; Allen, 1981; Scherphof; Gregoriadis, 1980; Hwang; Patel, 1983; Senior, 1985; Allen, 1983; Ellens; Senior, 1982; Hwang; Ashwell; Hakomori; Karlsson; Schauer; Durocher; Greenberg; Woodruff; Czop; and Okada) . Briefly, liposome size, charge, degree of lipid saturation, and surface moieties have all been impli¬ cated in liposome clearance by the RES. However, no single factor identified to date has been effective to provide long blood halflife, and more particularly, a relatively high per- centage of liposomes in the bloodstream than 1 day or more after IV administration.
One factor which does favor longer liposome lifetime in the bloodstream is small liposome size, typically in the size range of small unilamellar vesicles (SUVs) : 0.03-0.07 microns. However, the intravesicular volume of SUVs is quite limited, to the extent that loading SUVs with a peptides or proteins in a therapeutically effective dose range is not practical for parenteral administration.
4. Summary of the Invention
It is therefore one general object of the invention to provide a liposome composition and method for administering a therapeutic compound for an extended period in the blood¬ stream. The invention includes, in one aspect, a liposome compo¬ sition effective to extend to at least 24 hours, the period of effective activity of an therapeutic compound which can be administered intravenously in a therapeutically effective amount, and which has a blood halflife, in free form, of less than about 4 hours. The composition includes liposomes (i) composed of vesicle-forming lipids and between 1-20 mole per¬ cent of a vesicle-forming lipid derivatized with a biocompa- tible hydrophilic polymer, and (ii) having a selected mean particle diameter in the size range between about 0.1 to 0.4 microns, and the compound in liposome-entrapped form. The composition is intended for intravenous administration at a dose which contains an amount of the liposome-entrapped compound which is at least three times the therapeutically effective dose for the compound in free form.
In one preferred embodiment, the hydrophilic polymer is polyethyleneglycol having a molecular weight between about 1,000-5,000 daltons, and the polymer is derivatized with the polar head roup of a phospholipid, such a phosphatidylethanol- amine (PE) . Alternatively, the polymer may be other suitable biocompatible hydrophilic polymers, such as polylactic acid and polyglycolic acid.
Also in one preferred embodiment, the composition is effective to extend to at least 48 hours, the period of thera¬ peutic activity of an intravenously injected polypeptide which can be administered intravenously in a therapeutically effec¬ tive amount. The polypeptide may be a peptide or protein, such as superoxide dismutase, glucocerebrosidase, asparagi- nase, adenosine deaminase, interferons (alpha, beta, and gamma), interleukin (1,2,3,4,5,6,7), tissue necrosis factor
(TNF - alpha, beta) , colony stimulating factors (M-CSF (macro- phage) , G-CSF (granulocyte) , GM-CSF (granulocyte, macrophage) ,
TPA, prourokinase, and urokinase, HIV-1 vaccine, hepatitis B vaccine, malaria vaccine, and melanoma vaccine, erythropoietin
(EPO) , factor VIII, bone growth factor, fibroblast growth factor, insulin-like growth factor, nerve growth factor, platelet-derived growth factor, tumor growth factors (alpha, beta), somatomedin C (IGF-1) , and a ribosome inhibitor pro- tein, which is therapeutically active when administered intra¬ venously. Where the polypeptide is active in the picogram/ml range, such as is vasopressin, the composition is effective to deliver a therapeutically effective amount of the peptide into the bloodstream for a period of between 5-10 days. Also forming part of the invention is a method for exten¬ ding to at least 24 hours, the period of effective activity of an therapeutic compound which can be administered intravenous¬ ly in a therapeutically effective amount, and which has a halflife in the blood, in free form, of less than about 4 hours. In this method, a liposome composition of the type described above is administered intravenously to a subject at a dose which contains an amount of the compound which is at least three times such therapeutically effective amount.
Also disclosed is a liposome composition effective to extend to at least one week, the period of effective activity of an therapeutic compound which can be administered intra¬ venously in a therapeutically effective amount. The composi- tion includes liposomes (i) composed of vesicle-forming lipids and between 1-20 mole percent of a vesicle-forming lipid deri¬ vatized with a biocompatible hydrophilic polymer, and (ii) having a selected mean particle diameter in the size range between about 0.07-.15 microns, and the compound in liposome- entrapped form. The composition is intended for subcutaneous administration at a dose which contains an amount of the lipo¬ some-entrapped compound which is at least ten times such therapeutically effective intravenously administered amount. The liposome composition is used in a method for extending the period of release of a therapeutic compound, preferably a polypeptide, in a therapeutically active amount, for a period of at least 2 weeks.
In another aspect, the invention includes a liposome composition composed of vesicle-forming lipids and a vesicle- forming lipid derivatized with polylactic acid or polyglycolic acid, and a lipid composition composed of a vesicle-forming lipid having a polar head group, and a polylactic acid or polyglycolic acid moiety derivatized to the lipid's head group. These and other objects and features of the present invention will become more fully apparent when the following detailed description of the invention is read in conjunction with the accompanying drawings. Brief Description of the Drawings
Figure 1 illustrates a general reaction scheme for derivatizing a vesicle-forming lipid amine with a polyalkyl- 5 ether;
Figure 2 is a reaction scheme for preparing phosphati- dylethanolamine (PE) derivatized with polyethyleneglycol via a cyanuric chloride linking agent;
Figure 3 illustrates a reaction scheme for preparing 10 phosphatidylethanolamine (PE) derivatized with polyethylene¬ glycol by means of a diimidazole activating reagent;
Figure 4 illustrates a reaction scheme for preparing phosphatidylethanolamine (PE) derivatized with polyethylene¬ glycol by means of a trifluoromethane sulfonate reagent; 15 Figure 5 illustrates a vesicle-forming lipid derivatized with polyethyleneglycol through a peptide (A) , ester (B) , and disulfide (C) linkage;
Figure 6 illustrates a reaction scheme for preparing phosphatidylethanolamine (PE) derivatized with polylactic 20 acid;
Figure 7 is a plot of liposome retention time in the blood, expressed in terms of percent injected dose as a function of hours after IV injection, for PEG-PE liposomes containing different amounts of phosphatidylglycerol; 25 Figure 8 is a plot similar to that of Figure 7, showing retention times in the blood of liposomes composed of predomi¬ nantly unsaturated phospholipid components;
Figure 9 is a plot similar to that of Figure 1 , showing retention times in the blood of PEG liposomes (solid tri¬
3(1 angles) and conventional liposomes (solid circles) ;
Figure 10 is a plot of blood lifetimes of PEG-liposomes sized by extrusion through 0.1 micron (solid squares), 0.2 micron (solid circles), and 0.4 micron (solid triangles) polycarbonate membranes;
Figure 11 is a plot of blood retention times in liposomes containing a vesicle-forming lipid derivatized with polylactic acid (solid squares) and polyglycolic acid (open triangles) ; Figure 12 shows urine flow rates in rats, as a percentage of predosage rate, after surgery and IV administration of saline (control, open circles) and of aqueous solutions of vasopressin at total doses of 0.2 μg (closed squares), 0.8 μg (closed triangles) , and 2 μg (closed circles) ; Figure 13 shows urine flow rates in rats, as a percentage of predosage rate, after surgery and IV administration of saline (control, open circles) and of PEG-liposomes containing entrapped vasopressin at total doses of 2 μg (closed squares) , 8 μg (closed triangles) , and 24 μg (closed circles) ; Figure 14 shows urine flow rates in rats, as a percentage of predosage rate, after surgery and IV administration of saline (control, open circles) and of PEG-liposomes containing entrapped vasopressin at a total dose of 8 μg and mole percent of cholesterol in the liposomes of 33% (closed circles) , 16% (closed triangles) , and 0% (closed squares) ;
Figure 15 shows the blood clearance kinetics of free macrophage-colony stimulating factor (M-CSF) (solid triangles) , PEG-liposomes containing 30 mole percent cholesterol (solid triangles) , and M-CSF associated with the PEG-liposomes (solid circles) ;
Figure 16 shows the blood clearance kinetics of free M-CSF (solid triangles) , cholesterol-free PEG-liposomes (solid triangles) , and M-CSF associated with the PEG-liposomes (solid circles) ; Figure 17 is a plot of percent release of M-CSF into the blood from PEG liposomes containing 30 (solid, circles) and 0 (solid triangles) mole percent cholesterol; and Figure 18A shows urine flow rates in rats, as a percentage of predosage rate, after surgery and subcutaneous administra¬ tion of saline (control, open circles) and free vasopressin, in an amount 2 μg (solid triangles) , 25 μg "(solid circles) , and 50 μg (solid diamonds) ; and
Figure 18B shows urine flow rates in rats, as a percentage of predosage rate, after surgery and subcutaneous administra¬ tion of saline (control, open circles) and vasopressin en¬ trapped in PEG-liposomes, in an amount 25 μg (solid triang- les) , 100 μg (solid circles) , and 400 μg (solid diamonds) .
Detailed Description of the Invention I. Preparation of Derivatized Lipids Figure 1 shows a general reaction scheme for preparing vesicle-forming lipid derivatized a biocompatible, hydrophili polymer, as exemplified by polyethylene glycol (PEG) , poly lactic acid, and polyglycolic acid, all of which are readil water soluble, can be coupled to vesicle-forming lipids, an are tolerated in vivo without toxic effects. The hydrophili polymer which is employed, e.g., PEG, is preferably capped b a methoxy, ethoxy or other unreactive group at one end, or i one in which one end is more reactive than the other, such a polylactic acid. The polymer is activated at one end by reaction with suitable activating agent, such as cyanuric acid, diimadozle, anhydride reagent, or the like, as described below. Th activated compound is then reacted with a vesicle-formin lipid, such as phosphatidylethanol (PE) , to produce th derivatized lipid.
Alternatively, the polar group in the vesicle-formin lipid may be activated for reaction with the polymer, or th two groups may be joined in a concerted coupling reaction, according to known coupling methods. PEG capped at one end with a methoxy or ethoxy group can be obtained commercially in a variety of polymer sizes, e.g., 500-20,000 'dalton molecular weights.
The vesicle-forming lipid is preferably one having two hydrocarbon chains, typically acyl chains, and a polar head group. Included in this class are the phospholipids, such as phosphatidylcholine (PC) , PE, phosphatidic acid (PA) , phospha- tidylinositol (PI) , and sphingomyelin (SM) , where the two hydrocarbon chains are typically between about 14-22 carbon atoms in length, and have varying degrees of unsaturation. Also included in this class are the glycolipids, such as cere- broside and gangliosides. Another vesicle-forming lipid which may be employed is cholesterol and related sterols. In general, cholesterol may be less tightly anchored to a lipid bilayer membrane, particu¬ larly when derivatized with a high molecular weight polyalkyl- ether, and therefore be less effective in promoting liposome evasion of the RES in the bloodstream.
More generally, and as defined herein, "vesicle-forming lipid" is intended to include any amphipathic lipid having hydrophobic and polar head group moieties, and which (a) by itself can form spontaneously into bilayer vesicles in water, as exemplified by phospholipids, or (b) is stably incorporated into lipid bilayers in combination with phospholipids, with its hydrophobic moiety in contact with the interior, hydro- phobic region of the bilayer membrane, and its polar head group moiety oreinted toward the exterior, polar surface of the membrane. An example of a latter type of vesicle-forming lipid is cholesterol and cholesterol derivatives, such as cholesterol sulfate and cholesterol hemisuccinate.
According to one important feature of the invention, the vesicle-forming lipid may be a relatively fluid lipid, meaning that the lipid phase has a relatively low liquid-to-liquid crystal phase-transition temperature, e.g., at or below room temperature, or relatively rigid lipid, meaning that the lipid has a relatively high melting temperature, e.g., up to 50°C. As a rule, the more rigid, i.e., saturated lipids, contribute to greater membrane rigidity in a lipid bilayer structure and also contribute to greater bilayer stability in serum. Other lipid components, such as cholesterol, are also known to contribute to membrane rigidity and stability in lipid bilayer structures. Phospholipids whose acyl chains have a variety of degrees of saturation can be obtained commercially, or prepared according to published methods.
Figure 2 shows a reaction scheme for producing a PE-PEG lipid in which the PEG is derivatized to PE through a cyanuric chloride group. Details of the reaction are provided in Example 1. Briefly, ethoxy-capped PEG is activated with cyanuric chloride in the presence in sodium carbonate under conditions which produced the activated PEG compound IV in the figure. This material is purified to remove unreacted cyanuric acid. The activated PEG compound is reacted with PE in the presence of triethyl a ine to produce the desired PE- PEG compound shown at VII in the figure. The yield is about 8-10% with respect to initial quantities of PEG. The method just described may be applied to a variety of lipid amines, including PE, cholesteryl amine, and glycolipids with sugar-amine groups.
A second method of coupling a polyalkylether, such as capped PEG to a lipid amine is illustrated in Figure 3. Here the capped PEG is activated with a carbonyl diimidazole coupling reagent, to form the activated imidazole compoun shown at X in Figure 3. Reaction with a lipid amine, such as PE leads to PEG coupling to the lipid through an amide linkage, as illustrated in the PEG-PE compound shown at XI in the figure. Details of the reaction are given in Example 2.
A third reaction method for coupling a capped poly¬ alkylether to a lipid amine is shown in Figure" 4. Here PEG is first protected at its OH end by a trimethylsilane group. The end-protection reaction is shown at A in the figure, and involves the reaction of trimethylsilylchloride with PEG in the presence of triethylamine. The protected PEG is then reacted with the anhydride of trifluoromethyl sulfonate (compound XVI in Figure 4) to form the PEG compound activated with trifluoromethyl sulfonate. Reaction of the activated compound with a lipid amine, such as PE, in the presence of triethylamine, gives the desired derivatized lipid product, such as the PEG-PE compound, in which the lipid amine group is coupled to the polyether through the terminal methylene carbon in the polyether polymer. The trimethylsilyl protective group can be released by acid treatment, as indicated at H+ in the figure, or by reaction with a quaternary amine fluoride salt, such as the fluoride salt of tetrabutylamine. It will be appreciated that a variety of known coupling reactions, in addition to those just described, are suitable for preparing vesicle-forming lipids derivatized with hydro¬ philic polymers such as PEG. For example, the sulfonate anhydride coupling reagent illustrated in Figure 4 can be used to join an activated polyalkylether to the hydroxyl group of an amphipathic lipid, such as the 5'-OH of cholesterol. Other reactive lipid groups, such as an acid or ester lipid group may also be used for coupling, according to known coupling methods. For example, the acid group of phosphatidic acid can be activated to form an active lipid anhydride, by reaction with a suitable anhydride, such as acetic anhydride, and the reactive lipid can then be joined to a protected polyalk 1- amine by reaction in the presence of an isothiocyanate reagent.
In another embodiment, the derivatized lipid component are prepared to include a labile lipid-polymer linkage, suc as a peptide, ester, or disulfide linkage; which can b cleaved under selective physiological conditions, such as i the presence of peptidase or esterase enzymes present in th bloodstream. Figure 5 shows exemplary lipids which are linke through (A) peptide, (B) , ester, and (C) , disulfide containin linkages. The peptide-linked compound can be prepared, fo example, by first coupling a polyalkylether with the N terminal amine of the tripeptide shown, e.g., via the reactio shown in Figure 3. The peptide carboxyl group can then b coupled to a lipid amine group through a carbodiimide couplin reagent conventionally. The ester linked compound can b prepared, for example, by coupling a lipid acid, such a phosphatidic acid, to the terminal alcohol group of a poly alkylether, using alcohol via an anhydride coupling agent. Alternatively, an short linkage fragment containing an inter nal ester bond an suitable end groups, such as primary amin groups can be used to couple the polyalkylether to the amphi pathic lipid through amide or carbamate linkages. Similarly, the linkage fragment may contain an internal disulfide link age, for use in forming the compound shown at C in Figure 5. Figure 6 illustrates a method for derivatizing polylacti acid with PE. The polylactic acid is reacted, in the presenc of PE, with dicyclohexylcarboimide (DCCI) , as detailed i Example 4. Similarly, a vesicle-forming lipid derivatize with polyglycolic acid may be formed by reaction of polygly colic acid or glycolic acid with PE in the presence of suitable coupling agent, such as DCCI, also as detailed i Example 4. The vesicle-forming lipids derivatized with eithe polylactic acid or polyglycolic acid form part of the inven tion herein. Also forming part of the invention are liposome containing these derivatized lipids, in a 1-20 mole percent.
II. Preparation of Liposome Composition A. Lipid Components The lipid components used in forming the liposomes of the invention may be selected from a variety of vesicle-forming lipids, typically including phospholipids and sterols. As will be seen, one requirement of the liposomes of the present invention is long blood circulation lifetime. It is therefore useful to establish a standardized measure of blood lifetime which can be used for evaluating the effect of lipid component on blood halflife.
One method used for evaluating liposome circulation time in vivo measures the distribution of IV injected liposomes in the bloodstream and the primary organs of the RES at selected times after injection. In the standardized model which is used herein, RES uptake is measured by the ratio of total liposomes in the bloodstream to total liposomes in the liver and spleen, the principal organs of the RES. In practice, age and sex matched mice are injected intravenously (IV) through the tail vein with a radiolabeled liposome composition, and each time point is determined by measuring total blood and combined liver and spleen radiolabel counts, as detailed in Example 6. Since the liver and spleen account for nearly 100% of the initial uptake of liposomes by the RES, the blood/RES ratio just described provides a good approximation of the extent of uptake from the blood to the RES in vivo. For example, a ratio of about 1 or greater indicates a predominance of injected liposomes remaining in the bloodstream, and a ratio below about 1, a predominance of liposomes in the RES. For most of the lipid compositions of interest, blood/RES ratios were calculated at 1,2, 3, 4, and 24 hours. The liposomes of the present invention include 1-20 mole percent of the vesicle-forming lipid derivatized with a hydro¬ philic polymer, described in Section I. According to one aspect of the invention, it has been discovered that blood circulation halflives in these liposomes is largely indepen¬ dent of the degree of saturation of the phospholipid compo¬ nents making up the liposomes. That is, the phospholipid components may be composed of predominantly of fluidic, relatively unsaturated, acyl chains, or of more saturated, rigidifying acyl chain components. This feature of the invention is seen in Example 7, which examines blood/RES ratios in liposomes formed with PEG-PE, cholesterol, and PC having varying degrees of saturation (Table 4) . As seen from the data in Table 5 in the example, high blood/RES ratios were achieved with in substantially all of the liposome formula¬ tions, independent of the extent of lipid unsaturation in the bulk PC phospholipid, and no systematic trend, as a function of degree of lipid saturation, was observed.
Accordingly, the vesicle-forming lipids may be selected to achieve a selected degree of fluidity or rigidity, to control the stability of the liposomes in serum and the rate of release of entrapped drug from the liposomes in the blood¬ stream and/or tumor. The vesicle-forming lipids may also be selected, in lipid saturation characteristics, to achieve desired liposome preparation properties. It is generally the case, for example, that more fluidic lipids are easier to formulate and down size by extrusion or homogenization than more rigid lipid components. In general, more fluidic lipids
(low transition temperature) are preferred because of high compound-release rates in the bloodstream.
Similarly, it has been found that the percentage of cholesterol in the liposomes may be varied over a wide range without significant effect on observed blood/RES ratios. The studies presented in Example 8A, with reference to Table 6 therein, show virtually no change in blood/RES ratios in the range of cholesterol between 0-30 mole percent.
Cholesterol, or related cholesterol derivatives may be important, however, in regulating the rate of release of lipo¬ some entrapped therapeutic compounds into the bloodstream. The studies reported in Examples 15 and 16, for example, indicate that the rate of release of encapsulated polypeptide (peptide or protein) from liposomes in vitro in human serum or in vivo is strongly dependent on cholesterol concentration. PEG-liposome formulations containing high cholesterol (e.g., 30 mole percent or greater) released very little peptide or protein into serum in vitro, whereas decreasing amounts of cholesterol producing increasing loss of encapsulated poly- peptide. Similarly, and as described below, increased chol¬ esterol in intravenously administered PEG-liposomes produced reduced release of encapsulated compound into the bloodstream (Example 18) and reduced physiological effect (Example 16) . Thus, in accordance with one feature of the invention, the rate of release of compound from long-circulating liposomes can be controlled by the percent cholesterol included in the liposomes.
It has also been found, in studies conducted in support of the invention, that blood/RES ratios are also relatively unaffected by the presence of charged lipid components, such as phosphatidylglycerol (PG) . This can be seen from Figure 7, which plots percent loss of encapsulated marker for PEG-PE liposomes containing either 4.7 mole percent PG (triangles) or 14 mole percent PG (circles) . Virtually no difference in liposome retention in the bloodstream over a 24 hour period was observed.
The vesicle-forming lipid derivatized with a hydrophilic polymer is present in an amount preferably between about 1-20 mole percent, on the basis of moles of derivatized lipid as a percentage of total moles of vesicle-forming lipids. It will be appreciated that a lower mole ratio, such as 0.1 mole percent, may be appropriate for a lipid derivatized with a large molecular weight polymer, such as one having a molecular weight greater than 100 kilodaltons. As noted in Section I, the hydrophilic polymer in the derivatized lipid preferably has a molecular weight between about 200-20,000 daltons, and more preferably between about 1,000-5,000 daltons. Example 8B, which examines the effect of very short ethoxy ether moieties on blood/RES ratios indicates that polyether moieties of greater than about 5 carbon ether are required to achieve significant enhancement of blood/RES ratios.
B. Preparing the Liposome Composition
The liposomes may be prepared by a variety of techniques, such as those detailed in Szoka et al, 1980. One method for preparing drug-containing liposomes is the reverse phase evaporation method described by Szoka et al and in U.S. Patent No. 4,235,871. The reverse phase evaporation vesicles (REVs) have typical average sizes between about 2-4 microns and are predominantly oligolamellar, that is, contain one or a few lipid bilayer shells. The method is detailed in Example 5A. This method is generally preferred for preparing liposomes with encapsulated proteins high encapsulation efficiencies (up to 50%) are possible, and thus protein loss or problems of recovery and purification of non-encapsulated protein are reduced.
Multilamellar vesicles (MLVs) can be formed by simple lipid-film hydration techniques. In this procedure, a mixtur of liposome-forming lipids of the type detailed above dis solved in a suitable solvent is evaporated in a vessel to for a thin film, which is then covered by an aqueous medium, a detailed in Example 5B. The lipid film hydrates to form MLVs, typically with sizes between about 0.1 to 10 microns.
In accordance with one important aspect of the invention, the liposomes for intravenous injection are prepared to have substantially homogeneous sizes in a selected size range between about 0.1 and 0.4, and preferably 0.1 to 0.2 micron size ranges. Liposomes in this size range have sufficiently high encapsulation volumes for carrying therapeutically effec¬ tive amounts of the compound to be administered. At lower liposome sizes, the ratio of liposome-encapsulated compound to free compound may too low to achieve a requisite initial dose level of liposome-encapsulated compound in the bloodstream or may not remain in circulation due to extravasation. At the same time, 0.1-0.4 micron liposomes are small enough to give long blood circulation times, as discussed below, and also to allow sterilization by sterile filtration.
One effective sizing method for REVs and MLVs involves extruding an aqueous suspension of the liposomes through a polycarbonate membrane having a selected uniform pore size, typically 0.4, 0.2, and/or 0.1 micron pore sizes. The pore size of the membrane is proportional to the largest sizes of the liposomes which are produced, particularly where the pre¬ paration is extruded two or more times through the same size membrane. This method of liposome sizing is used in preparing homogeneous-size REV and MLV compositions described in the examples below. A more recent method involves extrusion through an asymmetric ceramic filter. The method is detailed in U.S. patent application for Liposome Extrusion Method, Serial No. 829,710, filed February 13, 1986. Homogenization methods are also useful for down-sizing liposomes.
C. Compound Loading
In one embodiment, the composition of the invention is used for slow-release delivery into the bloodstream or a poly¬ peptide (peptide or protein) which is therapeutically active in the bloodstream when administered IV, but which in free form has a short blood halflife, typically 4' hours or less. Examples of such polypeptides include hormones, such as vaso¬ pressin, calcitonin, oxytocin, somatotropin, human growth hormone, atrial naturectic factor (ANF) , and insulin; enzymes, such as superoxide dismutase, glucocerebrosidase, asparagin- ase, and adenosine deaminase; immunomodulators, such as inter- ferons (alpha, beta, and gamma), interleukin (1,2,3,4,5,6,7), tissue necrosis factor (TNF - alpha, beta) , and colony stimu¬ lating factors (M-CSF (macrophage) , G-CSF (granulocyte) , GM- CSF (granulocyte, macrophage) ; anticoagulants, such as TPA, prourokinase, and urokinase; vaccines, such as HIV-1 vaccine, hepatitis B vaccine, malaria vaccine, and melanoma vaccine; and other polypeptides (peptides and proteins) , such as erythropoietin (EPO) , factor VIII, bone growth factor, fibro blast growth factor, insulin-like growth factor, nerve growt factor, platelet-derived growth factor, tumor growth factors (alpha, beta) , and somatomedin C (IGF-1) ; and ribosome-inhibi tor proteins, such as pokeweed antiviral protein and gelonin.
The polypeptides useful in the invention typically hav relatively short blood halflives, on the order of 2-4 hours o less, and are active in the picogram/ml to nanogram/ml concen tration range in the blood.
As noted above, polypeptide is preferably loaded passivel by the reverse-phase emulsion method for preparing liposomes, although many other methods, such as solvent injection o lipid hydration may be employed. After liposome formation an sizing, free (unbound) drug can be removed by a variety o methods, for example, by gel filtration or ion exchang chromatography or diafiltration. Typically the amount of fre peptide in the final sterilized composition is less than abou 20%, and preferably less than 10% of the total polypeptide contained in the composition.
At the same time, the encapsulated compound is preferably present in an amount which, in a selected liposome dose, is between 3-20 times the amount of compound which would be given as a therapeutic dose in free form by IV administration. Thus, if the therapeutic dose of a peptide in free form is 1 μg for IV administration, a selected liposome dose will pre¬ ferably contain between about 3-20 μg of the peptide. Since 10-20% of this compound, e.g., 2-4 μg, may be in non-encapsu¬ lated form, it will be appreciated that the total amount of liposome which can be administered may be limited by the maxi- mum tolerated dose of the free compound. It is clear that larger doses of liposomes can be administered by achieving higher ratios of encapsulated to non-encapsulated compound. In general, this ratio is increased with larger liposomes, more complete free drug removal from the liposome composition, and greater liposome stability on storage.
The composition is also useful for slow-release delivery of a variety of non-peptide, water-soluble compounds which are effective in treating circulating cancers such as leukemias, for example, cytarabine, cyclophosphamide, carmustine, thio- guanine, bleomycin, daunorubicin, vinblastine, vincristine, and asparaginase. Also useful in therapeutic delivery by the present invention are antibiotics, such as gentamicin, tobramycin, amikacin, netilmicin, kanamycin, and streptomycin, cefotaxime, ceftizoxi e, and ceftriaxone, and anti-viral agents, such as AZT (zidovidine) , DDI (dideoxyinosine) , DDC (didioxycytidine) , ganciclovir, D4T (didehydrodeoxythymidine) , phosphonoformate, ribavirin, and acyclovir. Such compounds may be encapsulated by passive loading, as above, during liposome formation by reverse evaporation phase, lipid hydration, solvent injection, or other liposome forma¬ tion methods, and removed, after sizing by gel filtration or the like.
Alternatively, drugs which form weak bases at physiological pH may be actively loaded into the liposomes at high drug concentration in the liposomes. One method for active loading drugs into liposomes is described in co-owned U.S. patent application Serial No. 413,037, filed September 28, 1988. In this method, liposomes are prepared in the presence of a relatively high ammonium ion, such as 0.125 M ammonium sulfate. After sizing the liposomes to a desired size, the liposome suspension is treated to create an inside- to-outside ammonium ion gradient across the liposomal membranes. The gradient may be created by dialysis against a non-ammonium containing medium, such as an isotonic glucose medium, or by gel filtration, such as on a Sephadex G-50 column equilibrated with 0.15 M NaCl or KC1, effectively replacing ammonium ions in the exterior phase with sodium or potassium ions. Alternatively, the liposome suspension may be diluted with a non-ammonium solution, thereby reducing the exterior-phase concentration of ammonium ions. The ammonium concentration inside the liposomes is preferably at least 10 times, and more preferably at least 100 to 1000 times that in the external liposome phase.
The ammonium ion gradient across the liposomes in turn creates a pH gradient, as ammonia is released across the lipo¬ some membrane, and protons are trapped in the internal lipo- some phase. To load liposomes with the selected drug, a rela¬ tively dilute suspension of the liposomes, e.g., less than about 50 mM lipid, is mixed with an aqueous solution of the drug, and the mixture is allowed to equilibrate over an extended period, e.g., 24 hours at room temperature. In one typical method, a suspension of liposomes having a lipid con¬ centration of 25 mg/ml is mixed with an equal volume of anthracycline drug at a concentration of about 10 mg/ml. At the end of the incubation period, the suspension is treated to remove free (unbound) drug,
III. Intravenous Administration
A. Extended Lifetime in the Bloodstream One of the requirements for extended compound release into the bloodstream, in accordance with the invention, is an extended liposome lifetime in the bloodstream with IV liposome administration. One measure of liposome lifetime in the bloodstream in the blood/RES ratio determined at a selected time after liposome administration, as discussed above. Blood/RES ratios for a variety of liposome compositions are given in Table 3 of Example 6. In the absence of PEG- derivatized lipids, blood/RES ratios were 0.03 or less. In the presence of PEG-derivatized lipids, the blood/RES ratio ranged from 0.2, for low-molecular weight PEG, to between 1.7- 4 for several of the formulations, one of which lacks cholesterol, and three of which lack a charged phospholipid (e.g., PG) .
The data presented in Table 5 in Example 7 show blood/RES ratios (excluding two points with low percent recovery) between about 1.26 and 3.27, consistent with the data given in Table 3. As noted in Section II above, the blood lifetime values are substantially independent of degree of saturation of the liposome lipids, presence of cholesterol, and presence of charged lipids.
The blood/RES values reported above can be compared with blood/RES values reported in co-owned U.S. Patent No. 4,920,016, which used blood/RES measurement methods identical to those used in for the data presented in Tables 3 and 5. The best 24-hour blood/RES ratios which were reported in the above-noted patent was 0.9, for a formulation composed of GMlf saturated PC, and cholesterol. The next best formulations gave 24-hour blood/RES values of about 0.5. Thus, typical 24- hour blood/RES ratios obtained in a number of the current formulations were more than twice as high as the best formula¬ tions reported which have been reported to date. Further, ability to achieve high blood/RES with GMX or HPI lipids was dependent on the presence of predominantly saturated lipids in the liposomes.
Plasma kinetics of a liposomal marker in the bloodstream can provide another measure of the enhanced liposome lifetime which is achieved by the liposome formulations of the present invention. Figure 7 and 8 discussed above show the slow loss of liposomal marker over a 24 hour period in typical PEG-lipo- some formulations, substantially independent of whether the marker is a lipid or an encapsulated water-soluble compound
(Figure 8) . In both plots, the amount of liposomal marker present 24 hours after liposome injection is greater than 10% of the originally injected material.
Figure 9 shows the kinetics of liposome loss from the bloodstream for a typical PEG-lipsome formulation and the same liposomes in the absence of a PEG-derivatized lipid. After 24 hours, the percent marker remaining in the PEG-liposomes was greater than about 20%, whereas the conventional liposomes showed less than 5% retention in the blood after 3 hours, and virtually no detectable marker at 24 hours.
The results seen in Figures 7-9 are consistent with 24 hour blood liposome values measured for a variety of liposome formulations, and reported in Tables 3 and 5-7 in Example 6-9 below. As seen Table 3 in Example 6, the percent dose remaining at 24 hours was less than 1% for conventional lipo- somes, versus at least 5% for the PEG-liposomes. In the best formulations, values between about 20-40% were obtained. Similarly in Table 5 from Example 7, liposome levels in the blood after 24 hours (again neglecting two low recovery values) were between 12 and about 25 percent of total dose given. Similar results are reported in Tables 6 and 7 of Example 8.
The effect of liposome size on blood lifetime was been investigated by comparing loss of liposomal marker in intra- venously injected liposomes having selected sizes between about 0.1 and 0.25 microns. Experimental details are given in Example 10. The results, given in Figure 10, show about 10% or greater liposome marker present in the blood after 24 hours for each of the liposome formulations. Highest blood life- times were achieved with the smallest liposomes. Thus, although all of the liposome preparations give high blood lifetimes, it is also clear that liposome size can be selected to produce desired increase or decrease in total drug release time. The enhancement in liposome blood circulation time achieved with two other biocompatible hydrophilic polymers, polylactic acid and polyglycolic acid, is seen in Figure 11, which shows loss of liposome marker during the 24-hour period after intravenous liposome injection. The percent marker remaining at 24 hours is about 4.3 percent for polylactic acid (solid squares) , and about 6% for polyglycolic acid (open triangles). These values compare with the 0.1-1% retention seen in conventional liposomes after 24 hours.
The data relating to both blood/RES ratios and to liposome retention time in the bloodstream which were obtained from an model animal system can be reasonably extrapolated to humans and veterinary animals of interest. This is because uptake of liposomes by liver and spleen has been found to occur at similar rates in several mammalian species, including mouse, rat monkey, and human (Gregoriadis, 1974; Kimelberg, 1976; Juliano; Richardson; Lopez-Berestein) . This result likely reflects the fact that the biochemical factors' which appear to be most important in liposome uptake by the RES — including opsinization by serum lipoproteins, size-dependent uptake effects, and cell shielding by surface moieties — are common features of all mammalian species which have been examined.
B. Compound Release in the Bloodstream
In addition to long circulating halflives, another important property of the liposomes of the present invention is the ability to release entrapped compound, at a thera¬ peutically effective dose rate in the bloodstream. As discussed above, the liposome size between 0.1 and 0.4 microns allows relatively high compound loading in the lipo¬ somes, for effective compound release as the percentage of liposomes in the bloodstream even at relatively low liposome concentrations in the bloodstream. This is typically an important consideration since the total amount quantity of liposomes which can be administered will be limited, due to the unavoidable presence of some free compound in injected composition.
Another consideration is the ability of entrapped compound to be released from the liposomes during circulation in the blood. This feature is illustrated in the studies described in Example 15 below. Here PEG-liposomes containing entrapped vasopressin (a 1 kilodalton peptide) were prepared with increasing concentrations of cholesterol, from 0 to about 30 mole percent. In vitro measurements on peptide release from the liposomes in serum showed substantially less peptide release with greater amounts of cholesterol. The rate of release of the peptide hormone in vivo was determined by diuretic effect, as measured by decreased urine output, in period 1-8 days following intravenous administration of th liposomes. Details of the study are given in Example 15. A seen in Figure 14, the short-term effect on" urine flow was dependent on cholesterol content, the PEG-liposomes wit highest cholesterol producing the greatest hormone effect. Similarly, in the period 2-8 days following IV administratio of the liposomes, the two PEG-liposome formulations having th lowest cholesterol concentration gave the greatest hormone effect, indicating higher release rates from the liposomes.
The study reported in Example 16 demonstrates a simila ability to control release rates of large proteins from long- circulating liposomes. Here PEG-liposomes containing encapsu¬ lated M-CSF (a 55 kilodalton protein) were examined for per- cent retention in the bloodstream, of both lipid and protein components. The data plotted in Figure 15 show liposome lipi (solid triangles) and protein (solid circles) kinetics 24 hours after IV injection for a PEG-lipsome formulation con¬ taining 30 mole percent cholesterol. The data indicate about 20% loss of encapsulated material over 24 hours, as discusse in Example 16.
Figure 17 shows a similar plot for PEG-liposomes without cholesterol. The data here show a 40-50% loss of M-CSF after 24 hours. When normalized lipid and protein markers are compared at selected time points over 24 hours, the plots o protein release from cholesterol (solid circles) and no cholesterol (solid triangles) PEG-liposomes are obtained. The plots illustrate the markedly different protein release kinetics which can be obtained with long circulating lipo- somes, by varying cholesterol content of the liposomes. Another feature of the data shown in Figure 17 is th relatively high percent (at least 3%) of initially injecte protein which is released at 24 hours in both formulations. IV. Intravenous Liposome Treatment
The invention includes, in one aspect, a method for extending to at least 24 hours, the period of effective activity of an therapeutic compound. The compound is one which can be administered intravenously in a therapeutically effective amount, and which has a halflife in the bloodstream, in free form, of less than about 4 hours.
In practicing the method, there is provided a liposome composition such as described above, containing the compound in liposome-entrapped form. The size, lipid composition, and extent compound loading of the drug are selected, according to the desired release rates and total release times, as con¬ sidered in Section III above. The composition is injected intravenously to a subject at a dose which contains an amount of the compound which is at least 3, and typically 5-20 times the therapeutically effec¬ tive dose. Thus, for example, if a therapeutically effective dose of a compound is 10 μg/Kg body weight, the liposome com- position would be injected at a liposome dose of at least 30 μg compound/Kg, and typically between 50-200 μg compound/Kg body weight. As noted above, the amount of material which can be injected is generally limited by the maximum tolerated dose of free compound, since 5-20% or more of the compound may be in free (non-entrapped) form when the composition is admi¬ nistered. Thus, for example, if the maximum tolerated dose of a compound in free form is 2 μg/Kg body weight, and the lipo¬ some composition contains 10% non-entrapped compound, the highest dose of liposome composition which can be given is 20 μg/Kg body weight.
Studies on the treatment of L1210 leukemia in mice with cytosine arabinoside (araC) , in free form and entrapped in PEG-liposomes indicates that the liposome composition produced about a 250-300% increase in survival time when administered in PEG-liposomal form, compared with about a 120% increase in survival time for the drug in free form. The ability to achieve long drug release times, on the order of several days, by the method of the invention is illu¬ strated by the studies on IV vasopressin administration repor¬ ted in Examples 13-15. The studies were carried on Brattle- boro rats which are genetically diabetic by virtue of vaso- pressin deficiency. Figure 13 shows the effect, measured in percent predosage urine flow, of IV injection of free vaso¬ pressin at dose levels of 0.2 μg (solid squares), 0.8 μg (solid triangles) and 2.0 μg (solid circles) . The reduction in urine flow is maximum after 1 day, but returns to control levels (open circles) by day 2.
Figure 14 shows the effect on urine flow by vasopressin administered IV in PEG-liposomes, at dosage levels of 2 μg (solid squares) , 8 μg (solid triangles) and 24 μg (solid cir¬ cles) . A pronounced reduction in urine flow was observed within one day, with a slight rebound toward control values by day 2 — an effect likely due, in part, to free peptide in the liposome formulation. In addition, a marked reduction in urine flow was observed for at least 7 days following IV administration. It is noted that the long-term therapeutic effect is saturated at an 8 μg dose, with no further effect observed at 24 μg. The same long-term therapeutic effect of vasopressin was observed in PEG-liposomes containing various amounts of cholesterol, as seen in Figure 14, and as discussed above. The data in this figure also illustrate increased short-term (1-2 days) and decreased long-term (2-8 days) effect seen with low cholesterol liposomes having the highest peptide release rates. More generally, it will be appreciated that a variety of polypeptides which are active in the picogram-to-nanogram/ml range may be administered over a several day period by the method of the invention. The data presented in Figures 15-17, for example, demonstrate that (a) large proteins may be se¬ questered in the bloodstream, for slow release in therapeutic amounts over an extended period and (b) the rate of release of the protein into the bloodstream can be selectively controlled by liposome composition.
V. Subcutaneous Liposome Treatment
In accordance with another aspect of the invention, it has been discovered that the long-live liposome composition of the invention is also effective for slow release of a liposome- entrapped compound from a subcutaneous (SubQ) site into the bloodstream. In particular, it has been discovered that therapeutic effects of up to 3 weeks or more can be achieved by a single subcutaneous injection of the liposome composition of the invention. The experimental model used to demonstrate this method is the vasopressin model described above, and detailed in Example 17. Briefly, a treatment method involving SubQ injection of free vasopressin was compared, for duration of physiological effect, with vasopressin administered SubQ in PEG-liposomes. Figure 18A shows the depression in urine production observed with SubQ administration of free vasopressin at doses of 2 μg
(solid triangles) , 25 μg (solid circles) , 50 μg (solid squares) , and 100 μg (solid diamonds) . With the higher doses, a pronounced reduction in urine production at day 1 was observed, with a rebound to control levels (open circles) by day 4. Figure 18B shows the effect of SubQ administration of the peptide in PEG liposomes. The lowest does administered, 25 μg (closed triangles) gave a slight prolongation of activity, to about 8 days. A vasopressin dose of 100 μg (closed circles) showed a significant reduction in urine output out to about 20 days. An even longer effect, of at least 24 days, was observed at a dose of 400 μg (closed squares) .
The treatment method employing vasopressin is designed for treatment of diabetes insipidus, by long-term administration of vasopressin from a SubQ site. More generally, the method is designed for extending to at least one week, the period of effective activity of an therapeutic compound which can be administered intravenously in a therapeutically effective amount. The method utilizes liposomes (i) composed of vesicle-forming lipids and between 1-20 mole percent of a vesicle-forming lipid derivatized with a hydrophilic polymer, and (ii) having a selected mean particle diameter in the size range preferably between about 0.07-.15 microns, and having the compound in liposome-entrapped form. The liposome composition is administered subcutaneously at a dose of the composition which contains an amount of the liposome-entrapped compound which is at least ten times the therapeutically effective intravenously administered amount.
For compounds, such as vasopressin, which are active in the bloodstream in the picogram-to-nanogram/ml concentration, such as a variety of peptide and proteins, the method can be used for therapeutic delivery of the compound over a several- week period.
The ability of the liposome composition of the invention to produce a long-term therapeutic effect from a SubQ site suggests that the liposomes taken up from this site through the lymphatics may be able to successfully evade the normal lymphatic clearance mechanisms, including Kupfer cells. This mechanism is supported by studies on araC administration by the interperitoneal (IP) route by PEG-liposomes. Briefly, it was found that PEG-liposomes containing entrapped araC and administered by IP route produced a 250-300% increase in survial in animals having L1210 leukemias, compared with about 120% increase in survival with the free drug given by the IP route. The results suggests that PEG-liposomes are capable of migrating from the peritoneum through the lymphatics into the bloodstream.
The following examples are intended to illustrate, but not limit, the scope of the invention.
Materials
Cholesterol (Choi) was obtained from Sigma (St. Louis, MO) . Sphingomyelin (SM) , egg phosphatidylcholine (lecithin or PC), partially hydrogenated PC having the composition IV40, IV30, IV20, IV10, and IV1, phosphatidylglycerol (PG) , phospha- tidylethanolamine (PE) , dipalmitoyl-phosphatidyl glycerol (DPPG) , dipalmitoyl PC (DPPC) , dioleyl PC (DOPC) and distear- oyl PC (DSPC) were obtained from Avanti Polar Lipids (Birming¬ ham, AL) or Austin Chemical Co (Chicago, II) .
[15I]-tyraminyl-inulin was made according to published procedures. "Gallium citrate was supplied by NEN Neoscan (Boston, MA) . Vasopressin and macrophage colony stimulating factor were obtained from Sigma (St. Louis,Mo) , and Cetus (Emeryville,Ca) , respectively.
Example 1
Preparation of PEG-PE Linked by Cyanuric Chloride A. Preparation of activated PEG
2-0-Methoxypolyethylene glycol 1900-4, 6-dichloro-l,3,5 triazine previously called activated PEG was prepared as described in J. Biol. Chem., 252:3582 (1977) with the fol¬ lowing modifications.
Cyanuric chloride (5.5 g; 0.03 mol) was dissolved in 400 ml of anhydrous benzene containing 10 g of anhydrous sodium carbonate, and PEG-1900 (19 g; 0.01 mol) was added and the mixture was stirred overnight at room temperature. The solu¬ tion was filtered, and 600 ml of petroleum ether (boiling range, 35-60°) was added slowly with stirring. The finely divided precipitate was collected on a filter and redissolved in 400 ml of benzene. The precipitation and filtration pro¬ cess was repeated several times until the petroleum ether was free of residual cyanuric chloride as determined by high pres¬ sure liquid chromatography on a column (250 x 3.2 mm) of 5-m "LiChrosorb" (E. Merck) , developed with hexane, and detected with an ultraviolet detector. Titration of activated PEG-1900 with silver nitrate after overnight hydrolysis in aqueous buffer at pH 10.0, room temperature, gave a value of 1.7 mol of chloride liberated/mol of PEG. TLC analysis of the product was effected with TLC reversed-phase plates obtained from Baker using methanol:- water, 4:1 (v/v) as developer and exposure to iodine vapor for visualization. Under these conditions, the starting methoxy polyglycol 1900 appeared at Rf=0.54 to 0.60. The activated PEG appeared at Rf=0.41. Unreacted cyanuric chloride appeared at Rf=0.88 and was removed.
The activated PEG was analyzed for nitrogen and an appropriate correction was applied in selecting the quantity of reactant to use in further synthetic steps. Thus, when the product contained only 20% of the theoretical amount of nitrogen, the quantity of material used in the next synthetic step was increased by 100/20, or 5-fold. When the product contained 50% of the theoretical amount of nitrogen, only 100/50 or a 2-fold increase was needed.
B. Preparation of N-(4-Chloro-polyglycol 1900)-1,3,5-triazinyl egg phosphatidylethanolamine. In a screw-capped test tube, 0.74 ml of a 100 mg/ml (0.100 mmole) stock solution of egg phosphatidylethanolamine in chloroform was evaporated to dryness under a stream of nitrogen and was added to the residue of the activated PEG described in section A, in the amount to provide 205 mg (0.100 mmole) . To this mixture, 5 ml anhydrous dimethyl formamide was added. 27 microliters (0.200 mmole) triethylamine was added to the mixture, and the air was displaced with nitrogen gas. The mixture was heated overnight in a sand bath maintained at 110°C. The mixture was then evaporated to dryness under vacuum and a pasty mass of crystalline solid was obtained. This solid was dissolved in 5 ml of a mixture of 4 volumes of acetone and 1 volume of acetic acid. The resulting mixture was placed at the top of a 21 mm X 240 mm chromatographic absorption column packed with silica gel (Merck Kieselgel 60, 70-230 mesh) which had first been moistened with a solvent composed of acetone acetic acid, 80/20; v/v.
The column chromatography was developed with the same sol¬ vent mixture, and separate 20 to 50 ml aliquots of effluent were collected. Each portion of effluent was assayed by TLC on silica gel coated plates, using 2-butanone/acetic acid/water; 40/25/5; v/v/v as developer and iodine vapor exposure for visualization. Fractions containing only material of Rf=about 0.79 were combined and evaporated to dryness under vacuum. Drying to constant weight under high vacuum afforded 86 mg (31.2 micromoles) of nearly colorless solid N-(4-chloro-polyglycol 1900)-1,3,5-triazinyl eg phosphatidylethanolamine containing phosphorous. The solid compound was taken up in 24 ml of ethanol/- chloroform; 50/50 chloroform and centrifuged to remove insoluble material. Evaporation of the clarified solution to dryness under vacuum afforded 21 mg (7.62' micromoles) of colorless solid.
Example 2 Preparation of the Carbamate-Linked PEG-PE
A. Preparation of the imidazole carbamate of polyethylene glycol methyl ether 1900.
9.5 grams (5 mmoles) of polyethylene glycol methyl ether 1900 obtained from Aldrich Chemical Co. was dissolved in 45 ml benzene which has been dried over molecular sieves. 0.89 grams (5.5 mmoles) of pure carbonyl diimidazole was added. The purity was checked by an infra-red spectrum. The air in the reaction vessel was displaced with nitrogen. Vessel was enclosed and heated in a sand bath at 75°C for 16 hours.
The reaction mixture was cooled and the clear solution formed at room temperature. The solution was diluted to 50.0 ml with dry benzene and stored in the refrigerator as a 100 micromole/ml stock solution of the imidazole carbamate of PEG ether 1900.
B. Preparation of the phosphatidylethanolamine carbamate of polyethylene glycol methyl ether 1900.
10.0 ml (lmmol) of the 100 mmol/ml stock solution of the imidazole carbamate of polyethylene glycol methyl ether 1900 (compound X) was pipetted into a 10 ml pear-shaped flask. The solvent was removed under vacuum. 3.7 ml of a 100 mg/ml solution of egg phosphatidyl ethanolamine (V) in chlorofor (0.5 mmol) was added. The solvent was evaporated unde vacuum. 2 ml of 1,1,2,2-tetrachloroethylene and 139 micro liters (1.0 mmol) of triethylamine VI was added. The vessel was closed and heated in a sand bath maintained at 95°C for hours. At this time, thin-layer chromatography was performe with fractions of the above mixture to determine an extent o conjugation on Si02 coated TLC plates, using" butanone/aceti acid/water; 40/5/5; v/v/v; was performed as developer. 1 vapor visualization revealed that most of the free phosphati dyl ethanolamine of Rf=0.68, had reacted, and was replaced b a phosphorous-containing lipid at Rf=0.78 to 0.80.
The solvent from the remaining reaction mixture was evapo rated under vacuum. The residue was taken up in 10 m methylene chloride and placed at the top of a 21 mm x 270 m chromatographic absorption column packed with Merck Kieselge 60 (70-230 mesh silica gel) , which has been first rinsed wit methylene chloride. The mixture was passed through th column, in sequence, using the following solvents.
Table 1
Figure imgf000037_0001
50 ml portions of effluent were collected and each portio was assayed by TLC on Si02 - coated plates, using 12 vapo absorption for visualization after development with chloro form/methanol/water/concentrated ammonium hydroxide 130/70/8/0.5%; v/v/v/v. Most of the phosphates were found i fractions 11, 12, 13 and 14.
These fractions were combined, evaporated to dryness unde vacuum and dried in high vacuum to constant weight. The yielded 669 mg of colorless wax of phosphatidyl etha-nolamin carbamate of polyethylene glycol methyl ether. Thi represented 263 micromoles and a yield of 52.6% based on the phosphatidyl ethanolamine.
An NMR spectrum of the product dissolved in deutero— chloroform showed peaks corresponding to the spectrum for egg PE, together with a strong singlet due to the methylene groups of the ethylene oxide chain at Delta = 3.4 ppm. The ratio of methylene protons from the ethylene oxide to the terminal methyl protons of the PE acyl groups was large enough to confirm a molecular weight of about 2000 for the polyethylene oxide portion of the molecule of the desired product polyethylene glycol conjugated phosphatidyethanolamine carbamate, M.W. 2,654.
Example 3 Preparation of Ethylene-Linked PEG-PE
A. Preparation of I-trimethylsilyloxy-polyethylene glycol is illustrated in Reaction Scheme 3A.
15.0 gm (10 mmoles) of polyethylene glycol) M.Wt. 1500, (Aldrich Chemical) was dissolved in 80 ml benzene. 1.40 ml (11 mmoles) of chlorotrimethyl silane (Aldrich Chemical Co.) and 1.53 ml (Immoles) of triethylamine was added. The mixture was stirred at room temperature under an inert atmosphere for 5 hours.
The mixture was filtered with suction to separate crystals of triethylammonium chloride and the crystals were washed with 5 ml benzene. Filtrate and benzene wash liquids were combined. This solution was evaporated to dryness under vacuum to provide 15.83 grams of colorless oil which solidi¬ fied on standing. TLC of the product on Si-C18 reversed-phase plates using a mixture of 4 volumes of ethanol with 1 volume of water as developer, and iodine vapor visualization, revealed that all the polyglycol 1500 (Rf=0.93) has been consumed, and was replaced by a material of Rf=0.82. An infra-red spectrum revealed absorption peaks characteristic only of polyglycols.
Yield of I-trimethylsilyoxypolyethylene glycol, M.W. 1500 was nearly quantitative. B. Preparation of trifluoromethane sulfonyl ester of ltrimethylsilyloxy-polyethylene glycol.
15.74 grams (10 mmol) of the crystalline I-trimethyl- silyloxy polyethylene glycol obtained above was dissolved in 40 ml anhydrous benzene and cooled in a bath of crushed ice. 1.53 ml (11 mmol) triethylamine and 1.85 ml (11 mmol) of trifluoromethanesulfonic anhydride obtained from Aldrich Chemical Co. were added and the mixture was stirred over night under an inert atmosphere until the reaction mixture changed to a brown color. The solvent was then evaporated under reduced pressure and the residual syrupy paste was diluted to 100.0 ml with methylene chloride. Because of the great reactivity of tri¬ fluoromethane sulfonic esters, no further purification of the trifluoromethane sulfonyl ester of I-trimethylsilyloxy poly- ethylene glycol was done.
C. Preparation of N-1-trimethylsilyloxy polyethylene glycol 1500 PE.
10 ml of the methylene chloride stock solution of the trifluoromethane sulfonyl ester of 1-trimethylsilyloxy poly¬ ethylene glycol was evaporated to dryness under vacuum to obtain about 1.2 grams of residue (approximately 0.7 mmoles). To this residue, 3.72 ml of a chloroform solution containing 372 mg (0.5 mmoles) egg PE was added. To the resulting solu- tion, 139 icroliters (1.0 mmole) of triethylamine was added and the solvent was evaporated under vacuum. To the obtained residue, 5 ml dry dimethyl formamide and 70 microliters (0.50 mmoles) triethylamine (VI) was added. Air from the reaction vessel was displaced with nitrogen. The vessel was closed and heated in a sand bath 110°C for 22 hours. The solvent was evaporated under vacuum to obtain 1.58 grams of brownish- colored oil. A 21 X 260 mm chro atographic absorption column filled with Kieselgel 60 silica 70-230 mesh, was prepared and rinsed with a solvent composed of 40 volumes of butanone, 25 volumes acetic acid and 5 volumes of water. The crude product was dissolved in 3 ml of the same solvent and transferred to the top of the chromatography column. The chromatogram was developed with the same solvent and sequential 30 ml portions of effluent were assayed each by TLC.
The TLC assay system used silica gel coated glass plates, with solvent combination butanone/acetic acid/water; 40/25/5; v/v/v. Iodine vapor absorption served for visualization. In this solvent system, the N-1-trimethylsilyloxy polyethylene glycol 1500 PE appeared at Rf=0.78. Unchanged PE appeared at Rf=0.68.
The desired N-1-trimethylsilyloxy polyethylene glycol 1500 PE was a chief constituent of the 170-300 ml portions of column effluent. When evaporated to dryness under vacuum these portions afforded 111 mg of pale yellow oil of compound.
D. Preparation of N-polyethylene glycyl 1500: phospha¬ tidyl-ethanolamine acetic acid deprotection.
Once-chromatographed, PE compound was dissolved in 2 ml of tetrahydrofuran. To this, 6 ml acetic acid and 2 ml water was added. The resulting solution was let to stand for 3 days at 23°C. The solvent from the reaction mixture was evaporated under vacuum and dried to constant weight to obtain 75 mg of pale yellow wax. TLC on Si-C18 reversed-phase plates. developed with a mixture of 4 volumes ethanol, 1 volume water indicated that some free PE and some polyglycol-like materia formed during the hydrolysis.
The residue was dissolved in 0.5 ml tetfahydrofuran an diluted with 3 ml of a solution of ethanol water; 80:20; v:v The mixture was applied to the top of a 10 mm X 250 mm chroma tographic absorption column packed with octadecyl bonded phas silica gel and column was developed with ethanol water 80:20 by volume, collecting sequential 20 ml portions of effluent The effluent was assayed by reversed phase TLC. Fraction containing only product of Rf=0.08 to 0.15 were combined This was typically the 20-100 ml portion of effluent. Whe evaporated to dryness, under vacuum, these portions afforde 33 mg of colorless wax PEG-PE corresponding to a yield of onl 3%, based on the starting phosphatidyl ethanolamine.
NMR analysis indicated that the product incorporated bot PE residues and polyethylene glycol residues, but that i spite of the favorable-appearing elemental analysis, the chai length of the polyglycol chain has been reduced to about thre to four ethylene oxide residues. The product prepared wa used for a preparation of PEG-PE liposomes.
E. Preparation of N-Polyethylene glycol 1500 P.E. by fluorid deprotection. 500 mg of crude N-1-trimethylsilyloxy polyethylene glyco PE was dissolved in 5 ml tetrahydrofuran and 189 mg (0.60 millimoles) of tetrabutyl ammonium fluoride was added an agitated until dissolved. The reactants were let to stan over night at 20°C. The solvent was evaporated under reduced pressure and th residue was dissolved in 10 ml chloroform, washed with tw successive 10 ml portions of water, and centrifuged t separate chloroform and water phases. The chloroform phas was evaporated under vacuum to obtain 390 mg of orange-brown wax, which was determined to be impure N-polyethylene glycol 1500 PE compound.
The wax was re-dissolved in 5 ml chloro'form and trans- ferred to the top of a 21 X 270 mm column of silica gel moistened with chloroform. The column was developed by passing 100 ml of solvent through the column. The Table 2 solvents were used in sequence:
Table 2
Volume % Volume % Methanol Containing
Chloroform 2% Cone. Ammonium Hydroxide/methanol 100% 0%
95% 5%
90% 10%
85% 15%
80% 20% 70% 30%
60% 40%
50% 50%
0% 100%
Separated 50 ml fractions of column effluent were saved. The fractions of the column were separated by TLC on Si-C18 reversed-phase plates. TLC plates were developed with 4 volumes of ethanol mixed with 1 volume of water. Visualiza- tion was done by exposure to iodine vapor.
Only those fractions containing an iodine-absorbing lipid of Rf about 0.20 were combined and evaporated to dryness under vacuum and dried in high vacuum to constant weight. In this way 94 mg of waxy crystalline solid was obtained of M.W. 2226. The proton NMR spectrum of this material dissolved in deutero- chloroform showed the expected peaks due to the phosphatidyl ethanolamine portion of the molecule, together with a few methylene protons attributable to polyethylene glycol. (Delta = 3 .7) .
Example 4 Preparation of PE-Hydroph lic Polymers A. Preparation of PE polylactic acid.
200 mg (0.1 mmoles) poly (lactic acid), m. wt. = 2,00 (ICN, Cleveland, Ohio) was dissolved in 2.0 ml dimethy sulfoxide by heating while stirring to dissolve the materia completely. Then the solution was cooled immediately to 65° and poured onto a mixture of 75 mg (0.1 mmoles) o distearylphosphatidyl-ethanolamine (Cal. Biochem, La Jolla) and 41 mg (0.2 mmoles) dicyclohexylcarbodiimide (DCCI) . The 28 ml (0.2 mmoles) of triethylamine was added, the air swep out of the tube with nitrogen gas, the tube capped, and heate at 65°C for 48 hours.
After this time, the tube was cooled to room temperature, and 6 ml of chloroform added. The chloroform solution wa washed with three successive 6 ml volumes of water, centrifuged after each wash, and the phases separated with Pasteur pipette. The remaining chloroform phase was filtere with suction to remove suspended distearolyphosphatidyl ethanola ine. The filtrate was dried under vacuum to obtai 212 mg of semi-crystalline solid.
This solid was dissolved in 15 ml of a mixture of volumes ethanol with 1 volume water and passed through a 50 m deep and 21 mm diameter bed of H+ Dowex 50 cation exchang resin, and washed with 100 ml of the same solvent.
The filtrate was evaporated to dryness to obtain 131 m colorless wax. 291 mg of such wax was dissolved in 2.5 m chloroform and transferred to the top of a 21 mm x 280 m column of silica gel wetted with chloroform. The chromatogra was developed by passing through the column, in sequence, 10 ml each of: 100% chloroform, 0% (1% NH40H in methanol) ;
90% chloroform, 10% (1% NH4OH in methanol) ;
85% chloroform, 15% (1% NH4OH in methanol) ;
80% chloroform, 20% (1% NH4OH in methanol); 70% chloroform, 30% (1% NH4OH in methanol) ;
Individual 25 ml portions of effluent were saved and assayed by TLC on SF02-coated plates, using CHC13, CH3OH, H20, con. NH4OH, 130, 70, 8, 0.5 v/v as developer and I2 vapor absorption for visualization. The 275-325 ml portions of column effluent contained a single material, P04 +, of R£ = 0.89. When combined and evaporated to dryness, these afforded 319 mg colorless wax.
Phosphate analysis agrees with a molecular weight of possibly 115,000. Apparently, the polymerization of the poly (lactic acid) occurred at a rate comparable to that at which it reacted with phosphatidylethanolamine.
This side-reaction could probably be minimized by working with more dilute solutions of the reactants. B. Preparation of poly (glycolic acid) amide of DSPE
A mixture of 266 mg. (3.50 mmoles) glycolic acid, 745 mg
(3.60 mmoles) dicyclohexyl carbodiimide, 75 mg. (0.10 mmoles) distearoyl phosphatidyl ethanolamine, 32 microliters (0.23 mmoles triethyl amine, and 5.0 ml dry dimethyl sulfoxide was heated at 75° C, under a nitrogen atmosphere, cooled to room temperature, then diluted with an equal volume of chloroform, and then washed with three successive equal volumes of water to remove dimethyl sulfoxide. Centrifuge and separate phases with a Pasteur pipette each time. Filter the chloroform phase with suction to remove a small amount of suspended material and vacuum evaporate the filtrate to dryness to obtain 572 mg. pale amber wax.
Re-dissolve this material in 2.5 ml chloroform and transfer to the top of a 21 mm X 270 mm column of silica gel (Merck Hieselgel 60) which has been wetted with chloroform.
Develop the chromatogram by passing through the column, in sequence, 100 ml each of: 100% chloroform, 0 % (1% NH4OH in methanol) ;
90% chloroform, 10% (1% NH4OH in methanol) ;
85% chloroform, 15% (1% NH4OH in methanol) ;
80% chloroform, 20% (1% NH4OH in methanol) ;
70% chloroform, 30% (1% NH4OH in methanol) . Collect individual 25 ml portions of effluent and assay each by TLC on Si)2-coated plates, using CH Cl3, CH3 OH, H20, con-NH4OH; 130, 70, 8, 0.5 v/v as developer.
Almost all the P04 + material will be in the 275-300 ml portion of effluent. Evaporation of this to dryness under vacuum, followed by high-vacuum drying, affords 281 mg of colorless wax.
Phosphate analysis suggests a molecular weight of 924,000.
Manipulation of solvent volume during reaction and molar ratios of glycolic acid and dicyclohexyl carbodiimide would probably result in other sized molecules.
Example 5 Preparation of REVs and MLVs A. Sized REVs A total of 15 μmoles of the selected lipid components, in the mole ratios indicated in the examples below, were dis¬ solved in chloroform and dried as a thin film by rotary evapo¬ ration. This lipid film was dissolved in 1 ml of diethyl ether washed with distilled water. To this lipid solution was added 0.34 ml of an aqueous buffer solution containing 5 πi Tris, 100 mM NaCl, 0.1 mM EDTA, pH 7.4, and the mixture was emulsified by sonication for 1 minute, maintaining the tempe rature of the solution at or below room temperature. Wher the liposomes were prepared to contain encapsulated [1251] tyraminyl-inulin, such was included in the phosphate buffer at a concentration of about 4 μCi/ml buffer.
The ether solvent was removed under reduced pressure at room temperature, and the resulting gel was taken up in 0.1 ml of the above buffer, and shaken vigorously. The resulting REV suspension had particle sizes, as determined by microscopic examination, of between about 0.1 to 20 microns, and was com¬ posed predominantly of relatively large (greater than 1 micron) vesicles having one or only a few bilayer lamellae.
The liposomes were extruded twice through a polycarbonate filter (Szoka, 1978), having a selected pore size of 0.4 microns or 0.2 microns. Liposomes extruded through the 0.4 micron filter averaged 0.17+ (0.05) micron diameters, and through the 0.2 micron filter, 0.16 (0.05) micron diameters. Non-encapsulated [1251] tyraminyl-inulin was removed by passing the extruded liposomes through Sephadex G-50 (Phar¬ macia) .
B. Sized MLVs
Multilamellar vesicle (MLV) liposomes were prepared according to standard procedures by dissolving a mixture of lipids in an organic solvent containing primarily CHC13 and drying the lipids as a thin film by rotation under reduced pressure. In some cases a radioactive label for the lipid phase was added to the lipid solution before drying. The lipid film was hydrated by addition of the desired aqueous phase and 3 mm glass beads followed by agitation with a vortex and shaking above the phase transition temperature of the phospholipid component for at least 1 hour. In some cases a radioactive label for the aqueous phase was included in the buffer. In some cases the hydrated lipid was repeatedly frozen and thawed three times to provide for ease of the following extrusion step.
The size of the liposome samples was controlled by extru¬ sion through defined pore polycarbonate filters using pres¬ surized nitrogen gas. In one procedure, the' liposomes were extruded one time through a filter with pores of 0.4 μm and then ten times through a filter with pores of 0.1 μm. In another procedure, the liposomes were extruded three times through a filter with 0.2 μm pores followed by repeated extrusion with 0.05 μm pores until the mean diameter of the particles was below 100 nm as determined by DLS. Unencapsu- lated aqueous components were removed by passing the extruded sample through a gel permeation column separating the lipo¬ somes in the void volume from the small molecules in the included volume.
C. Loading 67Ga Into DF-Containing Liposomes
The protocol for preparation of Ga67-DF labeled liposomes as adapted from known procedures (Gabizon) . Briefly, lipo¬ somes were prepared with the ion chelator desferal mesylate encapsulated in the internal aqueous phase to bind irrever¬ sibly Ga transported through the bilayer by hydroxyquinoline (oxine) .
D. Dynamic Light Scattering Liposome particle size distribution measurements were obtained by DLS using a NICOMP Model 200 with a Brookhaven Instruments BI-2030AT autocorrelator attached. The instru¬ ments were operated according to the manufacturer's instruc¬ tions. The NICOMP results were expressed as the mean diameter and standard deviation of a Gaussian distribution of vesicles by relative volume. Example 6 Liposome Blood Lifetime Measurements
A. Measuring Blood Circulation Time and Blood/RES Ratios
In vivo studies of liposomes were performed in two dif- ferent animal models: Swiss-Webster mice at 25 g each and laboratory rats at 200-300 g each. The studies in mice involved tail vein injection of liposome samples at 1 μM phospholipid/mouse followed by animal sacrifice after a defined time and tissue removal for label quantitation by gamma counting. The weight and percent of the injected dose in each tissue were determined. The studies in rats involved establishment of a chronic catheter in a femoral vein for removal of blood samples at defined times after injection of liposome samples in a catheter in the other femoral artery at 3-4 μM phospholipid/rat. The percent of the injected dose remaining in the blood at several time points up to 24 hours was determined.
B. Time Course of Liposome Retention in the Bloodstream PEG-PE composed of methoxy PEG, molecular weight 1900 and l-palmitoyl-2-oleyl-PE (POPE) was prepared as in Example 2. The PEG-POPE lipid was combined with and partially hydrogen- ated egg PC (PHEPC) in a lipid:lipid mole ratio of about 0.1:2, and the lipid mixture was hydrated and extruded through a 0.1 micron polycarbonate membrane, as described in Example 4, to produce MLVs with average size about 0.1 micron. The MLV lipids included a small amount of radiolabeled lipid marker 14C-cholesteryl oleate, and the encapsulated marker 3H- inulin. The liposome composition was injected and the percent initial injected dose in mice was determined as described in Example 4, at 1, 2, 3, 4, and 24 after injection. The time course of loss of radiolabeled material is seen in Figure which is a plot of percent injected dose for encapsulate inulin (solid circles), inulin marker corrected to the initia injection point of 100% (open circles) , and lipid marke (closed triangles), over a 24-hour period post injection. A seen, both lipid and encapsulated markers showed greater tha 10% of original injected dose after 24 hours.
C. 24 Hour Blood Liposome Levels Studies to determine percent injected dose in the blood, and blood/RES ratios of a liposomal marker, 24 hours afte intravenous liposome injection, were carried out as describe above. Liposome formulations having the compositions shown a the left in Table 3 below were prepared as described above. Unless otherwise noted, the lipid-derivatized PEG was PEG 1900, and the liposome size was 0.1 micron. The percent dos remaining in the blood 24 hours after intravenous administra tion, and 24-hour blood/RES ratios which were measured ar shown in the center and right columns in the table, respectively.
Table 3
Lipid Composition* 24 Hours After IV Dose
% Injected Dose in Blood B/RES
PG:PC:Chol (.75:9.25:5) 0.2 0.01 PC: Choi (10:5) 0.8 0.03 PBG-DSPE:PC:Chol 23.0 3.0 PEG-DSPE:PC:Chol (250 nm) 9.0 0.5 PEG5o0o-DSPE : PC : Choi 21.0 2.2 PEG120-DSPE : PC : Choi 5.0 2.0 PEG-DSPE:PC (0.75:9.25) 22.0 0.2 PEG-DSPE : PG : PC : Choi 40.0 4.0 (0.75:2.25:7:5) PEG-DSPE :NaCholSO« : PC : Choi 25.0 2.5 (0.75:0.75:9.25:4.25)
*A11 formulations contain 33% cholesterol and 7.5% charged component an were 100 nm mean diameter except as noted. PEG-DSPE consisted of PEG1 except as noted. As seen, percent dose remaining in the blood 24 hours after injection ranged between 5-40% for liposomes containing PEG-derivatized lipids. By contrast, in both' liposome formu¬ lations lacking PEG-derivatized lipids, less than 1% of lipo¬ some marker remained after 24 hours. Also as seen in Table 3, blood/RES ratios increased from 0.01-0.03 in control liposomes to at least 0.2, and as high as 4.0 in liposomes containing PEG-derivatized liposomes.
Example 7 Effect of Phospholipid Acyl-Chain Saturation on Blood/RES Ratios in PEG-PE Liposomes
PEG-PE composed of methoxy PEG, molecular weight 1900 and distearylPE (DSPE) was prepared as in Example 2. The PEG-PE lipids were formulated with selected lipids from among sphin- gomyelin (SM) , fully hydrogenated soy PC (PC) , cholesterol (Choi) , partially hydrogenated soy PC (PHSPC) , and partially hydrogenated PC lipids identified as PC IV1, IV10, IV20, IV30, and IV40 in Table 4. The lipid components were mixed in the molar ratios shown at the left in Table 5, and used to form MLVs sized to 0.1 micron as described in Example 4.
Table 4
Figure imgf000050_0001
49
Table 5
Blood RES B/RES% Remaining
PEG-PE:SM:PC:Choi
0.2:1:1:1 19.23 6.58 2.92
PEG-PE:PHSPC:Choi
0.15:1.85:1 20.54 7.17 2.86
PEG-PE:PC IVl:Chol
0.15:1.85:1 17.24 13.71 1.26
Figure imgf000052_0001
PEG-PE:PC IVl:Chol (two animals) 0.15:1.85:1 19.16 10.07
PEG-PE:PC IVl0:Chol (two animals) 0.15:1.85:1 12.19 7.31 PEG-PE:PC IVl0:Chol
0.15:1.85:1 2.4 3.5
Figure imgf000052_0002
PEG-PE:PC IV20:Chol
0.15:1.85:1 24.56 7.52 3.27 62.75
PEG-PE:PC IV20:Chol
0.15:1.85:1 5.2 5.7 0.91 22.1
PEG-PE:PC IV40:Chol 0.15:1.85:1 19.44 8.87 2.19 53.88
PEG-PE:PC IV:Choi
0.15:1.85:0.5 20.3 8.8 2.31 45.5 PEG-PE:EPC:Choi
0.15:1.85:1 15.3 9.6 1.59 45.9
24 hours after injection, the percent material injected
(as measured by percent of 67Ga-desferal) remaining the blood and in the liver (L) and spleen (S) were determined, and these values are shown in the two data columns at the left in Table
4. The blood and L+S (RES) values were used to calculate a blood/RES value for each composition. The column at the right in Table 4 shows total amount of radioactivity recovered. The two low total recovery values in the table indicate anomalous clearance behavior. The results from the table demonstrate that the blood/RES ratios are largely independent of the fluidity, or degree of saturation of the phospholipid components forming the lipo¬ somes. In particular, there was no systematic change in blood/RES ratio observed among liposomes containing largely saturated PC components (e.g., IV1 and IV10 PC's), largely unsaturated PC components (IV40) , and intermediate-saturation components (e.g., IV20) .
In addition, a comparison of blood/RES ratios obtained using the relatively saturated PEG-DSPE compound and the rela- tively unsaturated PEG-POPE compound (Example 5) indicates that the degree of saturation of the derivatized lipid is itself not critical to the ability of the liposomes to evade uptake by the RES.
Example 8
Effect of Cholesterol and Ethoxylated Cholesterol on Blood/RES Ratios in PEG-PE Liposomes
A. Effect of added cholesterol PEG-PE composed of methoxy PEG, molecular weight 1900 and was derivatized DSPE as described in Example 6. The PEG-PE lipids were formulated with selected lipids from among sphin- gomyelin (SM) , fully hydrogenated soy PC (PC) , and cholesterol
(Choi) , as indicated in the column at the left in Table 5 below. The three formulations shown in the table contain about 30, 15, and 0 mole percent cholesterol. Both REVs (0.3 micron size) and MLVs (0.1 micron size) were prepared, sub¬ stantially as in Example 4, with encapsulated tritium-labeled inulin.
The percent encapsulated inulin remaining in the blood 2 and 24 hours after administration, given at the right in Table 6 below, show no measurable effect of cholesterol, in the range 0-30 mole percent.
Table 6
% Injected Dose
3H-Inulin In Blood
2 HR. 24 HR. 2 HR. 24 HR. JH Aqueous Label *C - Lipid Label (Leakage)
1) SM:PC:Choi:PEG-DSPE 1: 1: 1: 0.2
100 nm MLV 19 5 48 24 300 nm REV 23 15 67 20
2) SM:PC:Choi:PEG-DSPE 1: 1: 0.5: 0.2
300 nm REV 23 15 71 17
3) SM:PC:PEG-DSPE 1: 1: 0.2
100 nm MLV 19 6 58 24 300 nm REV 32 23 76 43 B. Effect of ethoxylated cholesterol
Methoxy-ethyoxy-cholesterol was prepared by coupling methoxy ethanol to cholesterol via the trifluorosulfonate coupling method described in Section I. PEG-PE composed of methoxy PEG, molecular weight 1900 and was derivatized DSPE as described in Example 6. The PEG-PE lipids were formulated with selected lipids from among distearylPC (DSPC) , partially hydrogenated soy PC (PHSPC) , cholesterol, and ethoxylated cholesterol, as indicated at the right in Table 7. The data show that (a) ethoxylated cholesterol, in combination with PEG-PE, gives about the same degree of enhancement of liposome lifetime in the blood as PEG-PE alone. By itself, the ethoxy¬ lated cholesterol provides a moderate degree of enhancement of liposome lifetime, but substantially less than that provided by PEG-PE.
Table 7
Formulation % Injected Dose In Blood r,C-Chol-01eate 2 HR. 24 HR.
HSPC:Choi:PEG-DSPE 55 9
1.85: 1: 0.15 HSPC:Choi:PEG-DSPE:PEG5-Chol 57 9 1.85: 0.85: 0.15: 0.15
HSPC:Choi:HPC:PEG5-Chol 15 2 1.85: 0.85: 0.15: 0.15
HSPC:Choi:HPG 4 1
1.85: 1: 0.15 Example 9 Effect of Charged Lipid Components on Blood/RES Ratios in PEG-PE Liposomes PEG-PE composed of methoxy PEG, molecular weight 1900 and was derivatized DSPE as described in Example 6. The PEG- PE lipids were formulated with lipids selected from among egg PG (PG) , partially hydrogenated egg PC (PHEPC) , and choleste¬ rol (Choi), as indicated in the Figure 6. The two formula¬ tions shown in the figure contained about 4.7 mole percent (triangles) or 14 mole percent (circles) PG. The lipids were prepared as MLVs, sized to 0.1 micron as in Example 4.
The percent of injected liposome dose present 0.25, 1, 2, 4, and 24 hours after injection are plotted for both formula¬ tions in Figure 6. As seen, the percent PG in the composition had little or no effect on liposome retention in the bloodstr¬ eam. The rate of loss of encapsulated marker seen is also similar to that observed for similarly prepared liposomes containing no PG.
Example 10
Effect of Liposome Size on Blood Lifetime PEG-DSPE, prepared as above with PEG-1900, was formulated with partially hydrogenated egg PC (PHEPC) , and cholesterol (Choi), at a mole ratio of 0.15: 1.85: 1. The liposomes were sized by extrusion through 0.25, 0.15 or 0.1 micron polycarbo¬ nate filters, to produce liposome sizes of about 0.4, 0.2, and 0.1 microns, respectively. Non-encapsulated 3H-inulin was removed by gel filtration.
Each of the three liposome were injected intravenously, and the percent of injected liposome marker in the blood was measured at 1, 2, 3, 4, and 24 hours, with the results shown in Figure 10. All three formulations show long blood half- lives, as evidence by at least about 10% liposome marker remaining after 24 hours. The 0.1 micron formulation (solid squares) is longer lived than the 0.2 micron formulation (solid circles) which is in turn longer lived than the 0.4 micron formulation.
Example 11 Effect of Other Hydrophilic Polymers on Blood Lifetime Polylactic acid-DSPE or polyglycolic acid-DSPE, prepared as above, was formulated with lipids selected from among hydrogenated soy PC (HSPC) , and cholesterol (Choi) , at a weight ratio of either 2: 3.5: 1 or 1: 3.5: 1. The liposomes were sized by extrusion through a 0.1 micron polycarbonate filter. The liposomes were labeled by Ga-DF as described in Example 5. These liposome formulations were injected intravenously, and the percent of injected liposome marker in the blood was measured at 1, 2, 3, 4, and 24 hours, with the results shown in Figure 11. When normalized at 15 minutes, about 3.9% of the liposome marker was present after 24 hours with polylactic acid-DSPE (solid squares) . An average value of about 6% of the liposome marker was present after 24 hours after with polyglycolic acid-DSPE (open triangles) .
Example 12 Plasma Kinetics of PEG-Coated and Uncoated Liposomes
PEG-PE composed of methoxy PEG, molecular weight 1900 and distearyl PE (DSPE) was prepared as in Example 2. The PEG-PE lipids were formulated with PHEPC, and cholesterol, in a mole ratio of 0.15:1.85:1. A second lipid mixture contained the same lipids, but without PEG-PE. Liposomes were prepared from the two lipid mixtures as described in Example 5, by lipid hydration in the presence of DF followed by sizing to 0.1 micron, removal of non-entrapped DF by gel filtration, and Ga labeling as described in Example 5. Both compositions contained 10 μM lipid/ml in 0.15 M NaCl, 0.5 mM desferal.
The two liposome compositions (0.4 ml) were injected IV in animals, as described in Example 6. At time 0.25, 1, 3 or 5, and 24 hours after injection, blood samples were removed and assayed for amount inulin remaining in the blood, expressed as a percentage of the amount measured immediately after injection. The results are shown in Figure 9. As seen, the PEG-coated liposomes have a blood halflife of about 11 hours, and nearly 30% of the injected material is present in the blood after 24 hours. By contrast, uncoated liposomes showed a halflife in the blood of less than 1 hour. At 24 hours, the amount of injected material was undetectable.
Example 13
Treatment with Free Vasopressin Male adult rats of the Brattleboro strain, congenitally deficient for VP (Valtin and Schroeder, 1964) , were acclimated to metabolic cages for at least three days before treatment. In most experiments, anesthesia was induced using a mixture of Nitrous oxide 2100 cc/ in, oxygen 400 cc/min, and 5% Iso- flurane (Aerrane) . Anesthesia was maintained throughout sur¬ gery with the same gas mixture but with a reduced percentage of isoflurane (2%) . In other experiments, inhaled ethyl ether was used for the anesthetic.
A. Surgery
The neck and ventral sides of both hindlimbs were shaved. A small incision was made at the midpoint between the ears at the neck to allow the cannula to be externalized out the neck. The rat was placed on its back and stabilized. An incision was made on the ventral hindlimb at the inguinal area, and fascia was tweezed apart to expose the femoral vascular complex. Further blunt dissection isolated the femoral vein or artery. For the vein, blood flow was occluded by passing a 3-0 silk tie (Davis-Geek, Danbury, CT) beneath the vessel distally and applying tension to the vein while clamping the vessel proximally with a bulldog clamp. This allowed the vein to become distended and easier to cut. Using small scissors, the vein was nicked and a bevelled 6-inch section of poly¬ ethylene catheter tubing (PE_50 Clay-Adams) filled with normal saline (0.9%) or 5% dextrose was inserted into the vein. Correct placement was verified by venous drawback and flush using a 1 cc syringe. The catheter was anchored loosely to the vein by passing a small tie under the vessel and knotting it around the cannula.
B. Dose Administration
Once animals had been surgically prepared, 150-1000 μl of test solution was administered intravenously. The dose volume administered was adjusted to give the desired total amount of vasopressin, for example 0.2, 0.8, and 3μg of aqueous vaso- pressin. The venous catheter was then removed, the femoral vein was tied off, and both leg incisions were closed. The animals were allowed to regain consciousness.
C. Urine Production Studies After a control period in which the rats showed diabetes insipidus and the urine volume was well established, the animals were weighed and anesthetized with inhaled anesthesia. The femoral vein was cannulated and the dose administered by the surgical procedures described. The animals were main- tained in metabolic cages and the urine was collected over the course of several days after dose administration. The urine volume was determined hourly immediately after dose administration and thereafter at least twice each day.
Figure 12 shows the percent predosage urine flow in animals treated with saline (open circles) or 0.2 μg (solid squares) 0.8 μg (solid triangles), and 2 μg (solid circles) vasopressin solution. At day 1 after drug administration, urine production showed a dose-dependent drop. At day 2 and thereafter, urine production was substantially back to control levels.
Example 14 Treatment with PEG-Liposomal Vasopressin
Large unilamellar liposomes were prepared according to the reverse phase evaporation procedure (Szoka and Papahad- jopoulos, 1978) . The lipid composition used in preparing the liposomes was PEG-DSPE, PC, sphingomyelin, and cholesterol, in a mole ratio 0.2: 1: 1: 1. Lipid mixtures for a 1 ml final volume of liposomes with a phospholipid concentration of 10 μmol/ml were dissolved in chloroform and evaporated to dryness under reduced pressure. After residual solvent was removed by high vacuum for 1 hour the mixture was dissolved in ethyl ether freshly washed with phosphate buffer at pH 7. Then, 0.34 ml of aqueous buffer (5 mM Tris, 100 mM NaCl, 0.1 mM EDTA) containing sufficient arginine vasopressin to give a final drug concentration of 510 μg/ml was added to the lipids in ether. A trace amount of 3H-labeled vasopressin was added to the vasopressin solution for determination of protein concentrations. Then the ether was removed by controlled rotoevaporation, and additional drug-free buffer added to give a 1 ml solution immediately after gel dispersion. The vasopressin content of the resulting liposomes was determined by separating free from liposome-bound by gel filtration on Bio-Gel A15. The liposomes were extruded through 0.4 μm Nuclepore filters (Olsen et al., 1979) and particle size distribution measured by dynamic light scattering with a Nicomp model 200. Mean diameter ranged between 0.2 μm and 0.5 μm and showed a low polydispersity. Unbound vasopressin was removed by dialysis and monitored by gel chromatography as above.
PEG-liposome preparations from above were administered intravenously to rats prepared as in Example 13, at liposomal doses 2 μg (solid squares) 8 μg (solid triangles) , and 24 μg (closed circles) vasopressin solution. Percent predosage urine flow was measured as above, with the results shown in Figure 14. The data show the substantially the same dose- dependent depression in urine production in the first day after drug administration, presumably resulting predominantly from non-entrapped vasopressin in the lipsome formulations. In contrast to free peptide administration, however, all three formulation produced a significant inhibition in urine production with respect to control (open circles) over a 2-8 day period after liposome administration.
Example 15 Treatment with PEG-Liposomal Vasopressin: Effect of Liposome Cholesterol Concentration Large unilamellar liposomes were prepared as in Example 14, with liposomes containing either 33, 16, or 0 mole percent cholesterol. Vasopressin encapsulation, lipsome sizing, and free peptide removal was carried out as in Example 14.
The three PEG-liposome preparations were administered intravenously to rats prepared as in Example 13, at liposomal doses giving 8 μg vasopressin, and the percent predosage urine production was measured over a 9 day period following liposome administration. The results are shown in Figure 15, for saline control (open circles) and PEG-liposomes with 0 (solid squares) , 16 (solid triangles) , and 33 (solid circles) mole percent cholesterol.
The day-1 response shows a marked dependence on percent cholesterol, with the greatest effect on urine flow being produced in liposomes with the lowest mole ratio of choleste¬ rol. This result is consistent with in vitro stability studies of vasopressin release from PEG-liposomes in serum: In the presence of serum, little release of vasopressin was seen in liposomes containing greater than 30 mole percent cholesterol. By contrast, formulations containing reduced amounts of cholesterol showed increasingly higher release rates of encapsulated peptide. Thus, it would appear that the significantly higher diuretic effect seen after 1 day with low and no cholesterol formulations is due to the presence of free peptide released from the liposomes in serum.
Interestingly, all three formulations produced a marked, and substantially similar diuretic effect over a 2-8 day period following liposome drug administration, as was seen in the method described in Example 14.
Example 16 Blood Clearance Kinetics of M-CSF from PEG-Liposomes A lipid film containing PEG-DSPE, PHEPC IV-40, cholesterol, and α-tocopherol, in a mole ratio 5:61:33:1, was hydrated with distilled water, and the resulting MLVs were sonicated for 30 minutes to form SUVs. M-CSF was concentrated and a portion of the protein was labeled with U125I-iodine.
Equal volumes (2 ml) of the protein solution and SUVs were mixed, and the mixture was frozen in an acetone/dry bath, and lyophilized overnight. The dried material was rehydrated in 0.8 ml of distilled water, and the resulting liposome suspension was extruded 1 time through a 0.4 μ polycarbonate filter, and 3 times through a 0.2 μ filter. The sized liposomes were diluted to 10 ml in distilled water, washed washed two times with high speed centrifugation, and the washed pellet was resuspended in 0.9 μl of sterile buffer, to a final concentration of 40 μ ol lipid/ml and between 0.5 and 1.25 mg protein/ml.
PEG-liposome preparations from above were administered intravenously to animals as in Example 6, and the blood levels of M-CSF were measured at 1, 2, 4, and 24 hours after rats prepared as in Example 14, at liposomal doses 2 μg (solid squares) 8 μg (solid triangles) , and 24 μg (closed circles) vasopressin solution. Similar measurements were made for an equivalent amount of M-CSF administered in solution form. The plasma kinetics for the PEG-liposome formulation containing 30 mole percent cholesterol are shown in Figure 15. The data show rapid clearance of free protein (solid triangles) with less than 1% protein remaining in the blood at 24 hours, compared with about 8% for liposome-associated protein (solid circles) . Percent liposomes remaining in the bloodstream, as judged by percent lipid marker, was slightly greater than 10%. A comparison of the clearance of rates of the lipid and pro¬ tein markers indicates that about 20% of the protein marker was released from the liposomes by 24 hours post injection. The plasma kinetics obtained with cholesterol-free PEG- liposomes is shown in Figure 16. Percent liposomes remaining after 24 hours was about 8.5 (solid triangles) compared with about 4.5% for liposome-associated M-CSF. The results indica¬ ted that about 40-50% of the originally encapsulated protein leaked from the liposomes in the 24-hour period post injection. The radioactive counts in the liposome lipid marker and in the encapsulated protein were normalized to 100% initial values, and the percent injected dose released into the blood¬ stream over time was then determined from the'difference be- tween the normalized protein and normalized liposome marker radioactivity levels. A plot of the calculated values of percent protein released at 1, 2, 4, and 24 hours post injec¬ tion is shown in Figure 17.
The plot for the cholesterol-free formulation (solid triangles) shows a protein release peak at 2 hours, with a gradual decline in amount released in the 2-24 hour period in the no-cholesterol formulation (solid triangles) . The amount of protein released from the liposomes at 24 hours was between 3-4 percent of the total administered. The plot for the formulation containing 30 mole percent cholesterol (solid circles) shows a gradual increase in release protein release rate over 24 hours. The amount of protein released from the liposomes at 24 hours was about 3 percent of the total administered. Thus, both formulations showed relatively high levels of protein release (3% or greater) at 24 hours.
Example 17 Subcutaneously Administered Liposomes MLVs were prepared by thin-film hydration as described in Example 5. The lipid composition of the thin film was PEG- DSPE, HEPC, and cholesterol, in a mole ratio 0.15:1.85:1. The thin film was hydrated with an aqueous buffer (5 mM Tris, 100 mM NaCl, 0.1 mM EDTA) containing arginine vasopressin at 7.5 mgs/ml. The MLVs were sized by repeated extrusion through a 0.1 micron polycarbonate membrane, and free (non-encapsulated) peptide was removed by gel filtration, as in Example 15. The final concentration of PEG-liposomes in the suspension was 100 μM/ml.
Vasopressin in free form was administered subcutaneously (1 ml) to Brattleboro rats, as in Example 13. The site of subcutaneous injection was the dorsal neck region. The doses administered were 2 μg (solid triangles) , 25 μg (solid circles) , 50 μg (solid squares) , and 100 μg (solid diamonds) . The percent of predosage urine flow observed is plotted in Figure 18A.
Vasopressin encapsulated in PEG-liposomes, prepared as above, was administered subcutaneously (1 ml) to animals as above. The doses administered were 25 μg (solid triangles), 100 μg (solid circles) , and 400 μg (solid squares) . The percent of predosage urine flow observed is plotted in Figure 18B.
Although the invention has been described and illustrated with respect to specific liposome formulations, liposome- entrapped compounds, and treatment methods, it will be apparent that a variety of related compositions, compounds and treatment methods without departing from the invention.

Claims

IT IS CLAIMED:
1. A liposome composition effective to extend to at leas 24 hours, the period of effective activity of an therapeuti compound which can be administered intravenously in a therapeu tically effective amount and which cleared in free form in th bloodstream with a halflife of less than about 4 hours comprising liposomes (i) composed of vesicle-forming lipids and betwee 1-20 mole percent of a vesicle-forming lipid derivatized with hydrophilic polymer, and (ii) having a selected mean particl diameter in the size range between about 0.1 to 0.4 microns, an the compound in liposome-entrapped form, for intravenous administration at a dose of the compositio which contains an amount of the liposome-entrapped compound whic is at least three times such therapeutically effective amount.
2. The composition of claim 1, wherein the hydrophili polymer is polyethyleneglycol having a molecular weight betwee about 1,000-5,000 daltons.
3. The composition of claim 2, wherein the polymer i derivatized to a phospholipid.
4. The composition of claim 1, wherein the polymer i selected from the group consisting of polylactic acid an polyglyclic acid.
5. A liposome composition effective to extend to at leas 48 hours, the period of therapeutic activity of an intravenousl injected polypeptide which can be administered intravenously in therapeutically effective amount, which is cleared in th bloodstream with a halflife of less than about 4 hours, and whos therapeutically active blood concentration is in the picogra nanogram/ml concentration range, comprising liposomes (i) composed of vesicle-forming lipids and betwe
1-20 mole percent of a vesicle-forming lipid derivatized with hydrophilic polymer, and (ii) having a selected mean partic diameter in the size range between about 0.1 to 0.4 microns, a the polypeptide in liposome-entrapped form, for intravenous administration at a dose of the compositi which contains an amount of the liposome-entrapped compound whi is at least three times such therapeutically effective amount.
6. The composition of claim 5, wherein the hydrophil polymer is polyethyleneglycol having a molecular weight betwe about 1,000-5,000 daltons.
7. The composition of claim 5, wherein the polypeptide is peptide hormone which is therapeutically active at a plas concentration in the picogram/ml range, and the liposo composition is effective to release the hormone in therapeutically effective dose for a period of at least five da after intravenous administration of the composition.
8. The composition of claim 7, wherein the peptide hormo is vasopressin.
9. The composition of claim 1, wherein the compound is protein selected from the group consisting of superoxi dismutase, glucocerebrosidase, asparaginase, adenosine deaminas interferons (alpha, beta, and gamma) , interleuk (1,2,3,4,5,6,7), tissue necrosis factor (TNF - alpha, beta colony stimulating factors (M-CSF (macrophage) , G-C
(granulocyte) , GM-CSF (granulocyte, macrophage) , TP prourokinase, and urokinase, HIV-1 vaccine, hepatitis B vaccin malaria vaccine, and melanoma vaccine, erythropoietin (EPO) factor VIII, bone growth factor, fibroblast growth factor insulin-like growth factor, nerve growth factor, platelet-derive growth factor, tumor growth factors (alpha, beta) , somatomedin (IGF-1) , and a ribosome inhibitor protein.
10. The composition of claim 9, wherein the protein i macrophage colony stimulating factor.
11. A method for extending to at least 24 hours, the perio of effective activity of an therapeutic compound which can b administered intravenously in a therapeutically effective amount and which has a halflife in the bloodstream in free form of les than about 4 hours, comprising providing a liposome composition containing liposomes (i composed of vesicle-forming lipids and between 1-20 mole percen of a vesicle-forming lipid derivatized with a hydrophili polymer, and (ii) having a selected mean particle diameter in th size range between about 0.1 to 0.4 microns, and the compound a least about 70% in liposome-entrapped form, and administering the composition intravenously to a subject a a dose which contains an amount of the compound which is at leas three times such therapeutically effective amount.
12. The method of claim 11, wherein the hydrophilic polyme is polyethyleneglycol having a molecular weight between abou 1,000-5,000 daltons.
13. The method of claim 11, wherein the polymer is selecte from the group consisting of polylactic acid and polyglycoli acid.
14. The method of claim 11, wherein the compound ii peptide hormone which is therapeutically active at a pl concentration in the picogram-to-nanogram/ml range, and s administering is effective to release the hormone in therapeutically effective dose for a period 'of at least days.
15. The method of claim 14, wherein the peptide hormone vasopressin.
16. The method of claim 11, wherein the compound i protein selected from the group consisting of superox dismutase, glucocerebrosidase, asparaginase, adenosine deamina interferons (alpha, beta, and gamma) , interleu (1,2,3,4,5,6,7), tissue necrosis factor (TNF - alpha, bet colony stimulating factors (M-CSF (macrophage) , G-
(granulocyte) , GM-CSF (granulocyte, macrophage) , T prourokinase, and urokinase, HIV-1 vaccine, hepatitis B vacc malaria vaccine, and melanoma vaccine, erythropoietin (EP factor VIII, bone growth factor, fibroblast growth fact insulin-like growth factor, nerve growth factor, platelet-der growth factor, tumor growth factors (alpha, beta) , somatomedi (IGF-1), and a ribosome inhibitor protein.
17. The method of claim 16, wherein the protein macrophage colony stimulating factor.
18. A liposome composition effective to extend to at l one week, the period of effective activity of a therapeutic pound which can be administered in a therapeutically effec amount, comprising liposomes (i) composed of vesicle-forming lipids and bet 1-20 mole percent of a vesicle-forming lipid derivatized wi hydrophilic polymer, and (ii) having a selected mean particl diameter in the size range between about 0.07-.15 microns, the compound in liposome-entrapped form, for subcutaneous administration at a dose "of the compositio which contains an amount of the liposome-entrapped compound whic is at least ten times such therapeutically effectiv intravenously administered amount.
19. The composition of claim 18, wherein the compound is polypeptide selected from the group consisting of superoxide dis utase, glucocerebrosidase, asparaginase, adenosine deaminase interferons (alpha, beta, and gamma) , interleuki (1,2,3,4,5,6,7), tissue necrosis factor (TNF - alpha, beta) colony stimulating factors (M-CSF (macrophage) , G-CSF (granulo cyte) , GM-CSF (granulocyte, macrophage) , TPA, prourokinase, an urokinase, HIV-1 vaccine, hepatitis B vaccine, malaria vaccine and melanoma vaccine, erythropoietin (EPO) , factor VIII, bon growth factor, fibroblast growth factor, insulin-like growt factor, nerve growth factor, platelet-derived growth factor tumor growth factors (alpha, beta) , somatomedin C (IGF-1) , and ribosome inhibitor protein.
20. The composition of claim 19, wherein the polypeptide i vasopressin.
21. A method of extending to at least one week, the perio of effective activity of an therapeutic compound which can b administered intravenously in a therapeutically effective amount comprising providing a liposome composition containing liposomes (i composed of vesicle-forming lipids and between 1-20 mole percen of a vesicle-forming lipid derivatized with a hydrophili polymer, and (ii) having a selected mean particle diameter in th size range between about 0.07 to 0.15 microns, and the compo at least about 70% in liposome-entrapped form, and administering the composition subcutaneously to a subject a dose which contains an amount of the compound" which is at le ten times such therapeutically effective intravenous administered amount.
22. The method of claim 21, wherein the compound is peptide hormone selected from the group consisting of superoxi dismutase, glucocerebrosidase, asparaginase, adenosine deaminas interferons (alpha, beta, and gamma) , interleuk (1,2,3,4,5,6,7), tissue necrosis factor (TNF - alpha, beta colony stimulating factors (M-CSF (macrophage) , G-CSF (granul cyte) , GM-CSF (granulocyte, macrophage) , TPA, prourokinase, a urokinase, HIV-1 vaccine, hepatitis B vaccine, malaria vaccin and melanoma vaccine, erythropoietin (EPO), factor VIII, bo growth factor, fibroblast growth factor, insulin-like gro factor, nerve growth factor, platelet-derived growth facto tumor growth factors (alpha, beta), somatomedin C (IGF-1), an ribosome inhibitor protein.
23. The method of claim 22, wherein the polypeptide vasopressin.
24. A liposome composition composed of vesicle-form lipids and a vesicle-forming lipid derivatized with a hydrophi polymer selected from the group consisting of polylactic acid polyglycolic acid.
25. A lipid composition composed of a vesicle-forming li having a polar head group, and a polylactic acid moi derivatized to said head group.
26. A lipid composition composed of a vesicle-forming lip having a polar head group, and a polyglycolic acid moie derivatized to said head group.
PCT/US1990/006034 1989-10-20 1990-10-19 Liposome microreservoir composition and method WO1991005545A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
KR1019920700918A KR0134982B1 (en) 1989-10-20 1990-10-19 Lipsome microreservoir composition and method
EP90916409A EP0496813B1 (en) 1989-10-20 1990-10-19 Liposome microreservoir composition and method
KR1019920700919A KR920703013A (en) 1989-10-20 1990-10-19 Liposomal Microretent Compositions and Methods
DE69015207T DE69015207T2 (en) 1989-10-20 1990-10-19 LIPOSOME MICRORESERVOIR COMPOSITION AND METHOD.
NO92921213A NO921213L (en) 1989-10-20 1992-03-27 LIPOSOM MICROR RESERVE COMPOSITION AND PROCEDURE
FI921763A FI921763A0 (en) 1989-10-20 1992-04-21 MIKRORESERVOARKOMPOSITION OCH FOERFARANDE FOER LIPOSOM.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US07/425,224 US5013556A (en) 1989-10-20 1989-10-20 Liposomes with enhanced circulation time
US425,224 1989-10-20

Publications (1)

Publication Number Publication Date
WO1991005545A1 true WO1991005545A1 (en) 1991-05-02

Family

ID=23685679

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US1990/006211 WO1991005546A1 (en) 1989-10-20 1990-10-19 Solid tumor treatment method and composition
PCT/US1990/006034 WO1991005545A1 (en) 1989-10-20 1990-10-19 Liposome microreservoir composition and method

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US1990/006211 WO1991005546A1 (en) 1989-10-20 1990-10-19 Solid tumor treatment method and composition

Country Status (18)

Country Link
US (2) US5013556A (en)
EP (2) EP0496813B1 (en)
JP (4) JP2667051B2 (en)
KR (2) KR920703013A (en)
AT (2) ATE115401T1 (en)
AU (2) AU654120B2 (en)
CA (2) CA2067133C (en)
DE (3) DE69019366T2 (en)
DK (1) DK0496835T3 (en)
ES (1) ES2071976T3 (en)
FI (2) FI921763A0 (en)
GR (1) GR3017060T3 (en)
HK (1) HK14097A (en)
IL (2) IL96069A (en)
LU (1) LU88854I2 (en)
NL (1) NL960031I2 (en)
NO (3) NO921213L (en)
WO (2) WO1991005546A1 (en)

Cited By (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994007466A1 (en) * 1992-10-07 1994-04-14 Liposome Technology, Inc. Compositions for treatmewnt of inflamed tissues
WO1994007537A1 (en) * 1992-10-05 1994-04-14 Stichting Voor De Technische Wetenschappen Pharmaceutical composition for site-specific release of a clot-dissolving protein
WO1994020073A1 (en) * 1993-03-03 1994-09-15 Liposome Technology, Inc. Lipid-polymer conjugates and liposomes
WO1994026251A1 (en) * 1993-05-07 1994-11-24 Sequus Pharmaceuticals, Inc. Subcutaneous liposome delivery method
WO1996010391A1 (en) * 1994-09-30 1996-04-11 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
US5514670A (en) * 1993-08-13 1996-05-07 Pharmos Corporation Submicron emulsions for delivery of peptides
WO1997021429A1 (en) * 1995-12-11 1997-06-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem Radiation-protective phospholipid
WO1998008489A1 (en) * 1996-08-26 1998-03-05 Transgene S.A. Cationic lipid-nucleic acid complexes
WO1998016202A2 (en) * 1996-10-11 1998-04-23 Sequus Pharmaceuticals, Inc. Fusogenic liposome composition and method
US5744155A (en) * 1993-08-13 1998-04-28 Friedman; Doron Bioadhesive emulsion preparations for enhanced drug delivery
WO1999027908A1 (en) * 1997-12-04 1999-06-10 Yissum Research Development Company Of The Hebrew University Of Jerusalem Combined chemo-immunotherapy with liposomal drugs and cytokines
US6083923A (en) * 1997-10-31 2000-07-04 Isis Pharmaceuticals Inc. Liposomal oligonucleotide compositions for modulating RAS gene expression
US6096720A (en) * 1995-08-01 2000-08-01 Novartis Ag Liposomal oligonucleotide compositions
WO2000066126A2 (en) * 1999-04-29 2000-11-09 Alza Corporation Liposome compositions for improved drug retention
WO2001011069A1 (en) 1999-08-06 2001-02-15 Celltech R&D Limited Freeze/thawed lipid complexes and their preparation
US6224903B1 (en) 1996-10-11 2001-05-01 Sequus Pharmaceuticals, Inc. Polymer-lipid conjugate for fusion of target membranes
US6316024B1 (en) 1996-10-11 2001-11-13 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US6673364B1 (en) 1995-06-07 2004-01-06 The University Of British Columbia Liposome having an exchangeable component
US6734171B1 (en) 1997-10-10 2004-05-11 Inex Pharmaceuticals Corp. Methods for encapsulating nucleic acids in lipid bilayers
EP1651187A2 (en) * 2003-07-31 2006-05-03 The Board Of Regents, The University Of Texas System Sterile preparations of phospholipids and anti-inflammatory pharmaceuticals and methods for making and using same
WO2007115168A2 (en) 2006-03-31 2007-10-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of eg5 gene
US7358068B2 (en) 2002-09-13 2008-04-15 Replicor, Inc. Antiviral oligonucleotides
EP2003207A2 (en) 2000-01-18 2008-12-17 Isis Pharmaceuticals, Inc. Antisense inhibition of PTP1B expression
EP2140872A2 (en) 2008-07-02 2010-01-06 Kenneth Wileford Composition for the treatment of skin conditions comprising sea water with additional magnesium
US7691824B2 (en) 2006-04-28 2010-04-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the JC virus
EP2174945A1 (en) 2001-08-01 2010-04-14 Genzyme Corporation Antisense modulation of apolipoprotein B expression
WO2010042281A2 (en) 2008-08-25 2010-04-15 Excaliard Pharmaceuticals Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
US7718632B2 (en) 2007-06-15 2010-05-18 Novartis Ag RNAi inhibition of alpha-ENaC expression
WO2010068816A1 (en) 2008-12-10 2010-06-17 Alnylam Pharmaceuticals, Inc. Gnaq targeted dsrna compositions and methods for inhibiting expression
EP2202239A1 (en) 2005-11-01 2010-06-30 Alnylam Pharmaceuticals Inc. RNAI inhibition of influenza virus replication
EP2221376A2 (en) 2001-06-21 2010-08-25 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
WO2010099341A1 (en) 2009-02-26 2010-09-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mig-12 gene
WO2010105209A1 (en) 2009-03-12 2010-09-16 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
EP2246443A1 (en) 2001-05-14 2010-11-03 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
EP2270231A2 (en) 2001-10-09 2011-01-05 ISIS Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
EP2270230A2 (en) 2001-10-09 2011-01-05 ISIS Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
EP2272985A1 (en) 2001-08-07 2011-01-12 ISIS Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (A) expression
EP2272958A1 (en) 2002-09-26 2011-01-12 ISIS Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
EP2280019A1 (en) 2001-07-25 2011-02-02 ISIS Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US7888498B2 (en) 2006-05-22 2011-02-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of IKK-B gene
EP2284267A2 (en) 2003-08-18 2011-02-16 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US7960350B2 (en) 2003-10-24 2011-06-14 Ader Enterprises, Inc. Composition and method for the treatment of eye disease
EP2332955A2 (en) 2001-07-30 2011-06-15 ISIS Pharmaceuticals, Inc. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
EP2336318A1 (en) 2002-11-13 2011-06-22 Genzyme Corporation Antisense modulation of apolipoprotein B expression
EP2336319A1 (en) 2002-11-13 2011-06-22 Genzyme Corporation Antisense modulation of apolipoprotein B expression
WO2011123468A1 (en) 2010-03-29 2011-10-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
US8048998B2 (en) 2007-01-19 2011-11-01 Exiqon A/S Mediated cellular delivery of LNA oligonucleotides
EP2388318A1 (en) 2001-12-10 2011-11-23 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
EP2392583A1 (en) 2006-05-19 2011-12-07 Alnylam Europe AG. RNAi modulation of Aha and therapeutic uses thereof
US8148344B2 (en) 2008-03-27 2012-04-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for mediating RNAi in vivo
WO2012064824A1 (en) 2010-11-09 2012-05-18 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of eg5 and vegf genes
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
EP2468865A1 (en) 2004-02-06 2012-06-27 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of stat3 expression
WO2012106508A1 (en) 2011-02-02 2012-08-09 Pfizer Inc. Method of treating keloids or hypertrophic scars using antisense compounds targeting connective tissue growth factor (ctgf)
US8293719B2 (en) 2004-03-12 2012-10-23 Alnylam Pharmaceuticals, Inc. iRNA agents targeting VEGF
US8324366B2 (en) 2008-04-29 2012-12-04 Alnylam Pharmaceuticals, Inc. Compositions and methods for delivering RNAI using lipoproteins
WO2012166903A1 (en) 2011-06-02 2012-12-06 President And Fellows Of Harvard College Methods and uses for ex vivo tissue culture systems
WO2012177949A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of protein c (proc) genes
WO2012177906A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
WO2012177947A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
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
WO2013019857A2 (en) 2011-08-01 2013-02-07 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
EP2617828A1 (en) 2007-12-10 2013-07-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of factor VII gene
US20130224121A1 (en) * 2012-02-23 2013-08-29 Canon Kabushiki Kaisha Particle containing hydrophobic dye having cyanine structure, and contrast agent containing the particle
WO2013155204A2 (en) 2012-04-10 2013-10-17 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
US8598333B2 (en) 2006-05-26 2013-12-03 Alnylam Pharmaceuticals, Inc. SiRNA silencing of genes expressed in cancer
EP2690175A2 (en) 2008-09-02 2014-01-29 Alnylam Pharmaceuticals Compositions and methods for combined inhibition of mutant EGFR gene and IL-6 expression
EP2712926A2 (en) 2008-03-05 2014-04-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 and VEGF genes
WO2014107731A1 (en) 2013-01-07 2014-07-10 Biomedical Research Models, Inc. Therapeutic vaccines for treating herpes simplex virus type 2 infections
WO2014160871A2 (en) 2013-03-27 2014-10-02 The General Hospital Corporation Methods and agents for treating alzheimer's disease
WO2014182661A2 (en) 2013-05-06 2014-11-13 Alnylam Pharmaceuticals, Inc Dosages and methods for delivering lipid formulated nucleic acid molecules
EP2810643A2 (en) 2009-08-14 2014-12-10 Alnylam Pharmaceuticals Inc. Lipid formulated compositions and mehods for inhibiting expression of a gene from the ebola virus
US8926955B2 (en) 2008-12-22 2015-01-06 Creabilis S.A. Synthesis of polymer conjugates of indolocarbazole compounds
WO2015050990A1 (en) 2013-10-02 2015-04-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
EP2905336A1 (en) 2007-03-29 2015-08-12 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of a gene from the ebola
EP2937418A1 (en) 2008-10-20 2015-10-28 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
EP2944646A1 (en) 2005-09-29 2015-11-18 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
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
WO2016094897A1 (en) 2014-12-12 2016-06-16 The Jackson Laboratory Compositions and methods relating to the treatment of cancer, autoimmune disease, and neurodegenerative disease
WO2016100716A1 (en) 2014-12-18 2016-06-23 Vasant Jadhav Reversirtm compounds
EP3109321A1 (en) 2008-09-25 2016-12-28 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
WO2017015109A1 (en) 2015-07-17 2017-01-26 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
WO2017048843A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
EP3252068A2 (en) 2009-10-12 2017-12-06 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
EP3456827A2 (en) 2010-06-02 2019-03-20 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
US10485764B2 (en) 2015-07-02 2019-11-26 Otsuka Pharmaceutical Co., Ltd. Lyophilized pharmaceutical compositions
US10519190B2 (en) 2017-08-03 2019-12-31 Otsuka Pharmaceutical Co., Ltd. Drug compound and purification methods thereof
WO2020037125A1 (en) 2018-08-16 2020-02-20 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the lect2 gene
US10590416B2 (en) 2017-07-06 2020-03-17 Arrowhead Pharmaceuticals, Inc. RNAi agents for inhibiting expression of alpha-ENaC and methods of use
WO2020113135A1 (en) 2018-11-29 2020-06-04 Flagship Pioneering Innovations V, Inc. Methods of modulating rna
EP3674409A1 (en) 2011-03-29 2020-07-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of tmprss6 gene
EP3693463A1 (en) 2013-10-04 2020-08-12 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
WO2021046122A1 (en) 2019-09-03 2021-03-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
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
WO2021067747A1 (en) 2019-10-04 2021-04-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
WO2021087325A1 (en) 2019-11-01 2021-05-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
WO2021163066A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
WO2021207167A1 (en) 2020-04-06 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
WO2021207189A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing scn9a expression
WO2021237097A1 (en) 2020-05-21 2021-11-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
WO2023283403A2 (en) 2021-07-09 2023-01-12 Alnylam Pharmaceuticals, Inc. Bis-rnai compounds for cns delivery

Families Citing this family (1490)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5882330A (en) * 1984-05-25 1999-03-16 Lemelson; Jerome H. Drugs and methods for treating diseases
US5456663A (en) * 1984-05-25 1995-10-10 Lemelson; Jerome H. Drugs and methods for treating diseases
US5925375A (en) * 1987-05-22 1999-07-20 The Liposome Company, Inc. Therapeutic use of multilamellar liposomal prostaglandin formulations
JPH0720857B2 (en) * 1988-08-11 1995-03-08 テルモ株式会社 Liposome and its manufacturing method
GB8824593D0 (en) * 1988-10-20 1988-11-23 Royal Free Hosp School Med Liposomes
US6132763A (en) * 1988-10-20 2000-10-17 Polymasc Pharmaceuticals Plc Liposomes
US5153000A (en) * 1988-11-22 1992-10-06 Kao Corporation Phosphate, liposome comprising the phosphate as membrane constituent, and cosmetic and liposome preparation comprising the liposome
US5527528A (en) * 1989-10-20 1996-06-18 Sequus Pharmaceuticals, Inc. Solid-tumor treatment method
US20010051183A1 (en) * 1989-10-20 2001-12-13 Alza Corporation Liposomes with enhanced circulation time and method of treatment
US5843473A (en) * 1989-10-20 1998-12-01 Sequus Pharmaceuticals, Inc. Method of treatment of infected tissues
US5620689A (en) * 1989-10-20 1997-04-15 Sequus Pharmaceuuticals, Inc. Liposomes for treatment of B-cell and T-cell disorders
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5305757A (en) * 1989-12-22 1994-04-26 Unger Evan C Gas filled liposomes and their use as ultrasonic contrast agents
US5585112A (en) * 1989-12-22 1996-12-17 Imarx Pharmaceutical Corp. Method of preparing gas and gaseous precursor-filled microspheres
US5705187A (en) * 1989-12-22 1998-01-06 Imarx Pharmaceutical Corp. Compositions of lipids and stabilizing materials
US5733572A (en) * 1989-12-22 1998-03-31 Imarx Pharmaceutical Corp. Gas and gaseous precursor filled microspheres as topical and subcutaneous delivery vehicles
US5469854A (en) * 1989-12-22 1995-11-28 Imarx Pharmaceutical Corp. Methods of preparing gas-filled liposomes
US6551576B1 (en) 1989-12-22 2003-04-22 Bristol-Myers Squibb Medical Imaging, Inc. Container with multi-phase composition for use in diagnostic and therapeutic applications
US5776429A (en) * 1989-12-22 1998-07-07 Imarx Pharmaceutical Corp. Method of preparing gas-filled microspheres using a lyophilized lipids
US6146657A (en) 1989-12-22 2000-11-14 Imarx Pharmaceutical Corp. Gas-filled lipid spheres for use in diagnostic and therapeutic applications
US6001335A (en) * 1989-12-22 1999-12-14 Imarx Pharmaceutical Corp. Contrasting agents for ultrasonic imaging and methods for preparing the same
US5542935A (en) 1989-12-22 1996-08-06 Imarx Pharmaceutical Corp. Therapeutic delivery systems related applications
US6088613A (en) * 1989-12-22 2000-07-11 Imarx Pharmaceutical Corp. Method of magnetic resonance focused surgical and therapeutic ultrasound
US5773024A (en) * 1989-12-22 1998-06-30 Imarx Pharmaceutical Corp. Container with multi-phase composition for use in diagnostic and therapeutic applications
US5580575A (en) * 1989-12-22 1996-12-03 Imarx Pharmaceutical Corp. Therapeutic drug delivery systems
US5922304A (en) 1989-12-22 1999-07-13 Imarx Pharmaceutical Corp. Gaseous precursor filled microspheres as magnetic resonance imaging contrast agents
US5352435A (en) * 1989-12-22 1994-10-04 Unger Evan C Ionophore containing liposomes for ultrasound imaging
US5441746A (en) * 1989-12-22 1995-08-15 Molecular Bioquest, Inc. Electromagnetic wave absorbing, surface modified magnetic particles for use in medical applications, and their method of production
US5656211A (en) * 1989-12-22 1997-08-12 Imarx Pharmaceutical Corp. Apparatus and method for making gas-filled vesicles of optimal size
US5882678A (en) * 1990-01-12 1999-03-16 The Liposome Co, Inc. Interdigitation-fusion liposomes containing arachidonic acid metabolites
US5162361A (en) * 1990-04-10 1992-11-10 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Method of treating diseases associated with elevated levels of interleukin 1
US20060084797A1 (en) * 1990-06-11 2006-04-20 Gilead Sciences, Inc. High affinity TGFbeta nucleic acid ligands and inhibitors
US6465188B1 (en) * 1990-06-11 2002-10-15 Gilead Sciences, Inc. Nucleic acid ligand complexes
US20030113369A1 (en) * 1991-01-16 2003-06-19 Martin Francis J. Liposomes with enhanced circulation time and method of treatment
US5205290A (en) 1991-04-05 1993-04-27 Unger Evan C Low density microspheres and their use as contrast agents for computed tomography
US5874062A (en) * 1991-04-05 1999-02-23 Imarx Pharmaceutical Corp. Methods of computed tomography using perfluorocarbon gaseous filled microspheres as contrast agents
JP3220180B2 (en) * 1991-05-23 2001-10-22 三菱化学株式会社 Drug-containing protein-bound liposomes
GB9111611D0 (en) * 1991-05-30 1991-07-24 Sandoz Ltd Liposomes
EP0636028B1 (en) 1992-04-03 2004-03-03 The Regents Of The University Of California Self-assembling polynucleotide delivery system comprising an amphiphatic cationic peptide
ZA933926B (en) * 1992-06-17 1994-01-03 Amgen Inc Polyoxymethylene-oxyethylene copolymers in conjuction with blomolecules
DE69324591T3 (en) * 1992-08-05 2004-02-12 Meito Sangyo K.K., Nagoya COMPOSITE MATERIAL WITH A SMALL DIAMETER THAT CONTAINS A WATER-SOLUBLE CARBOXYLPOLYSACCHARIDE AND MAGNETIC IRON OXIDE
DE4236237A1 (en) * 1992-10-27 1994-04-28 Behringwerke Ag Prodrugs, their preparation and use as medicines
US6764693B1 (en) * 1992-12-11 2004-07-20 Amaox, Ltd. Free radical quenching composition and a method to increase intracellular and/or extracellular antioxidants
JP3351476B2 (en) * 1993-01-22 2002-11-25 三菱化学株式会社 Phospholipid derivatives and liposomes containing the same
US6180134B1 (en) 1993-03-23 2001-01-30 Sequus Pharmaceuticals, Inc. Enhanced ciruclation effector composition and method
US6326353B1 (en) * 1993-03-23 2001-12-04 Sequus Pharmaceuticals, Inc. Enhanced circulation effector composition and method
WO1994021281A1 (en) * 1993-03-23 1994-09-29 Liposome Technology, Inc. Polymer-polypeptide composition and method
US5359030A (en) * 1993-05-10 1994-10-25 Protein Delivery, Inc. Conjugation-stabilized polypeptide compositions, therapeutic delivery and diagnostic formulations comprising same, and method of making and using the same
US6191105B1 (en) 1993-05-10 2001-02-20 Protein Delivery, Inc. Hydrophilic and lipophilic balanced microemulsion formulations of free-form and/or conjugation-stabilized therapeutic agents such as insulin
US5681811A (en) * 1993-05-10 1997-10-28 Protein Delivery, Inc. Conjugation-stabilized therapeutic agent compositions, delivery and diagnostic formulations comprising same, and method of making and using the same
US20050181976A1 (en) * 1993-05-10 2005-08-18 Ekwuribe Nnochiri N. Amphiphilic oligomers
ES2165875T3 (en) * 1993-05-21 2002-04-01 Liposome Co Inc REDUCTION OF ADVERSE PHYSIOLOGICAL REACTIONS INDUCED BY LIPOSOMES.
JP4076026B2 (en) * 1993-10-25 2008-04-16 トランセイヴ,インコーポレイテッド Liposome defensin
US5415869A (en) * 1993-11-12 1995-05-16 The Research Foundation Of State University Of New York Taxol formulation
US5468499A (en) * 1993-11-15 1995-11-21 Ohio State University Liposomes containing the salt of phosphoramide mustard and related compounds
US7083572B2 (en) * 1993-11-30 2006-08-01 Bristol-Myers Squibb Medical Imaging, Inc. Therapeutic delivery systems
JPH09506866A (en) * 1993-12-14 1997-07-08 ジョーンズ ホプキンス ユニバーシティー スクール オブ メディシン Controlled release of pharmaceutically active substances for immunotherapy
US6312719B1 (en) * 1994-03-04 2001-11-06 The University Of British Columbia Liposome compositions and methods for the treatment of atherosclerosis
US6773719B2 (en) * 1994-03-04 2004-08-10 Esperion Luv Development, Inc. Liposomal compositions, and methods of using liposomal compositions to treat dislipidemias
US20010055581A1 (en) * 1994-03-18 2001-12-27 Lawrence Tamarkin Composition and method for delivery of biologically-active factors
US5736121A (en) * 1994-05-23 1998-04-07 Imarx Pharmaceutical Corp. Stabilized homogenous suspensions as computed tomography contrast agents
DE4423131A1 (en) * 1994-07-01 1996-01-04 Bayer Ag New hIL-4 mutant proteins as antagonists or partial agonists of human interleukin 4
US5626862A (en) 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors
US5512294A (en) * 1994-08-05 1996-04-30 Li; King C. Targeted polymerized liposome contrast agents
US6132764A (en) * 1994-08-05 2000-10-17 Targesome, Inc. Targeted polymerized liposome diagnostic and treatment agents
US6001968A (en) 1994-08-17 1999-12-14 The Rockefeller University OB polypeptides, modified forms and compositions
US6350730B1 (en) 1994-08-17 2002-02-26 The Rockefeller University OB polypeptides and modified forms as modulators of body weight
US6124439A (en) * 1994-08-17 2000-09-26 The Rockefeller University OB polypeptide antibodies and method of making
US6048837A (en) * 1994-08-17 2000-04-11 The Rockefeller University OB polypeptides as modulators of body weight
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
US6471956B1 (en) 1994-08-17 2002-10-29 The Rockefeller University Ob polypeptides, modified forms and compositions thereto
US6124448A (en) * 1994-08-17 2000-09-26 The Rockfeller University Nucleic acid primers and probes for the mammalian OB gene
US5885613A (en) * 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
ATE219660T1 (en) * 1994-09-30 2002-07-15 Inex Pharmaceuticals Corp AGENT FOR INTRODUCING POLYANIONIC MATERIALS INTO CELLS
DE69519802T2 (en) * 1994-10-14 2001-04-19 Liposome Co Inc ETHERLIPID LIPOSOMES AND THEIR THERAPEUTIC USE
CN1080575C (en) * 1994-10-14 2002-03-13 蛋白质传送股份有限公司 Conjugation-stabilized polypeptide compositions, therapeutic delivery and diagnostic formulations comprising same, and method of making and using the same
US6214388B1 (en) 1994-11-09 2001-04-10 The Regents Of The University Of California Immunoliposomes that optimize internalization into target cells
US6743779B1 (en) 1994-11-29 2004-06-01 Imarx Pharmaceutical Corp. Methods for delivering compounds into a cell
US5827531A (en) * 1994-12-02 1998-10-27 The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration Microcapsules and methods for making
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US6008202A (en) * 1995-01-23 1999-12-28 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5830430A (en) * 1995-02-21 1998-11-03 Imarx Pharmaceutical Corp. Cationic lipids and the use thereof
IL112834A (en) * 1995-03-01 2000-12-06 Yeda Res & Dev Pharmaceutical compositions for controlled release of soluble receptors
US5658588A (en) * 1995-03-31 1997-08-19 University Of Cincinnati Fibrinogen-coated liposomes
GB9509016D0 (en) * 1995-05-03 1995-06-21 Royal Free Hosp School Med Tissue entrapment
US8071737B2 (en) * 1995-05-04 2011-12-06 Glead Sciences, Inc. Nucleic acid ligand complexes
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
US5997898A (en) * 1995-06-06 1999-12-07 Imarx Pharmaceutical Corp. Stabilized compositions of fluorinated amphiphiles for methods of therapeutic delivery
US6033645A (en) * 1996-06-19 2000-03-07 Unger; Evan C. Methods for diagnostic imaging by regulating the administration rate of a contrast agent
US6231834B1 (en) 1995-06-07 2001-05-15 Imarx Pharmaceutical Corp. Methods for ultrasound imaging involving the use of a contrast agent and multiple images and processing of same
US6139819A (en) 1995-06-07 2000-10-31 Imarx Pharmaceutical Corp. Targeted contrast agents for diagnostic and therapeutic use
US5981501A (en) * 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
EP1489184A1 (en) * 1995-06-07 2004-12-22 Inex Pharmaceutical Corp. Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US7422902B1 (en) 1995-06-07 2008-09-09 The University Of British Columbia Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US6521211B1 (en) 1995-06-07 2003-02-18 Bristol-Myers Squibb Medical Imaging, Inc. Methods of imaging and treatment with targeted compositions
US6051600A (en) * 1995-09-12 2000-04-18 Mayhew; Eric Liposomal hydrolysis-promoting hydrophobic taxane derivatives
US6107332A (en) 1995-09-12 2000-08-22 The Liposome Company, Inc. Hydrolysis-promoting hydrophobic taxane derivatives
US5834025A (en) 1995-09-29 1998-11-10 Nanosystems L.L.C. Reduction of intravenously administered nanoparticulate-formulation-induced adverse physiological reactions
US5858397A (en) * 1995-10-11 1999-01-12 University Of British Columbia Liposomal formulations of mitoxantrone
CN1119145C (en) * 1995-10-11 2003-08-27 埃斯佩里安Luv发展公司 Liposomal compositions and method of using them
US20030040467A1 (en) * 1998-06-15 2003-02-27 Mary Ann Pelleymounter Ig/ob fusions and uses thereof.
US6936439B2 (en) * 1995-11-22 2005-08-30 Amgen Inc. OB fusion protein compositions and methods
US5626654A (en) 1995-12-05 1997-05-06 Xerox Corporation Ink compositions containing liposomes
JP2000503204A (en) 1996-01-08 2000-03-21 ジェネンテック インコーポレーテッド WSX receptor and ligands
US6392069B2 (en) 1996-01-08 2002-05-21 Canji, Inc. Compositions for enhancing delivery of nucleic acids to cells
US20040014709A1 (en) * 1996-01-08 2004-01-22 Canji, Inc. Methods and compositions for interferon therapy
US5789244A (en) * 1996-01-08 1998-08-04 Canji, Inc. Compositions and methods for the treatment of cancer using recombinant viral vector delivery systems
US7002027B1 (en) 1996-01-08 2006-02-21 Canji, Inc. Compositions and methods for therapeutic use
US6096336A (en) * 1996-01-30 2000-08-01 The Stehlin Foundation For Cancer Research Liposomal prodrugs comprising derivatives of camptothecin and methods of treating cancer using these prodrugs
US5908624A (en) * 1996-06-27 1999-06-01 Albany Medical College Antigenic modulation of cells
US6667053B1 (en) 1996-02-16 2003-12-23 Elan Pharmaceuticals, Inc. D and L etherlipid stereoisomers and liposomes
US5942246A (en) * 1996-02-16 1999-08-24 The Liposome Company, Inc. Etherlipid containing multiple lipid liposomes
USRE39042E1 (en) * 1996-02-16 2006-03-28 The Liposome Company, Inc. Etherlipid-containing multiple lipid liposomes
US6007839A (en) * 1996-02-16 1999-12-28 The Liposome Company, Inc. Preparation of pharmaceutical compositions containing etherlipid-containing multiple lipid liposomes
US5965159A (en) * 1996-02-16 1999-10-12 The Liposome Company, Inc. Etherlipid-containing multiple lipid liposomes
AU2549297A (en) 1996-03-28 1997-10-17 Board Of Trustees Of The University Of Illinois, The Materials and methods for making improved echogenic liposome compositions
AU736301B2 (en) 1996-05-01 2001-07-26 Imarx Therapeutics, Inc. Methods for delivering compounds into a cell
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
US20070275921A1 (en) * 1996-06-06 2007-11-29 Isis Pharmaceuticals, Inc. Oligomeric Compounds That Facilitate Risc Loading
US20050042647A1 (en) * 1996-06-06 2005-02-24 Baker Brenda F. Phosphorous-linked oligomeric compounds and their use in gene modulation
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
WO2005121371A2 (en) * 2004-06-03 2005-12-22 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
US5919480A (en) 1996-06-24 1999-07-06 Yissum Research Development Company Of The Hebrew University Of Jerusalem Liposomal influenza vaccine composition and method
US8007784B1 (en) 1996-06-27 2011-08-30 Albany Medical College Antigenic modulation of cells
US6284267B1 (en) 1996-08-14 2001-09-04 Nutrimed Biotech Amphiphilic materials and liposome formulations thereof
US7368129B1 (en) 1996-08-14 2008-05-06 Nutrimed Biotech Amphiphilic materials and liposome formulations thereof
US5851984A (en) * 1996-08-16 1998-12-22 Genentech, Inc. Method of enhancing proliferation or differentiation of hematopoietic stem cells using Wnt polypeptides
US6159462A (en) * 1996-08-16 2000-12-12 Genentech, Inc. Uses of Wnt polypeptides
ATE252372T1 (en) * 1996-08-23 2003-11-15 Sequus Pharm Inc LIPOSOMES CONTAINING CISPLATIN
US6414139B1 (en) 1996-09-03 2002-07-02 Imarx Therapeutics, Inc. Silicon amphiphilic compounds and the use thereof
US5846517A (en) * 1996-09-11 1998-12-08 Imarx Pharmaceutical Corp. Methods for diagnostic imaging using a renal contrast agent and a vasodilator
CA2263568C (en) * 1996-09-11 2008-12-02 Imarx Pharmaceutical Corp. Methods for diagnostic imaging using a contrast agent and a renal vasodilator
JP2001503396A (en) 1996-10-11 2001-03-13 アルザ コーポレイション Therapeutic liposome compositions and methods
DK0964690T3 (en) * 1996-10-15 2003-10-20 Liposome Co Inc Peptide-lipid conjugates, liposomes and liposomal drug administration
US6339069B1 (en) * 1996-10-15 2002-01-15 Elan Pharmaceuticalstechnologies, Inc. Peptide-lipid conjugates, liposomes and lipsomal drug delivery
US20070036722A1 (en) * 1996-10-28 2007-02-15 Pal Rongved Separation processes
US6261537B1 (en) 1996-10-28 2001-07-17 Nycomed Imaging As Diagnostic/therapeutic agents having microbubbles coupled to one or more vectors
US6331289B1 (en) 1996-10-28 2001-12-18 Nycomed Imaging As Targeted diagnostic/therapeutic agents having more than one different vectors
ATE318618T1 (en) * 1996-10-28 2006-03-15 Amersham Health As IMPROVEMENTS IN OR RELATED TO DIAGNOSTIC/THERAPEUTIC COMPOUNDS
US5827533A (en) * 1997-02-06 1998-10-27 Duke University Liposomes containing active agents aggregated with lipid surfactants
US6207186B1 (en) * 1997-02-14 2001-03-27 The Regents Of The University Of California Lamellar gels and methods for making and using
US6537246B1 (en) 1997-06-18 2003-03-25 Imarx Therapeutics, Inc. Oxygen delivery agents and uses for the same
US6090800A (en) 1997-05-06 2000-07-18 Imarx Pharmaceutical Corp. Lipid soluble steroid prodrugs
US6143276A (en) * 1997-03-21 2000-11-07 Imarx Pharmaceutical Corp. Methods for delivering bioactive agents to regions of elevated temperatures
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
IL132120A0 (en) 1997-04-03 2001-03-19 Guilford Pharm Inc Biodegradable terephthalate polyester-poly (phosphate) polymers compositions articles and methods for making and using the same
AU743758B2 (en) 1997-04-07 2002-02-07 Genentech Inc. Anti-VEGF antibodies
ES2361267T3 (en) 1997-04-07 2011-06-15 Genentech Inc. PROCEDURE FOR THE PRODUCTION OF HUMANIZED ANTIBODIES THROUGH RANDOM MUTAGENESIS.
AU7104598A (en) 1997-04-09 1998-10-30 Philipp Lang New technique to monitor drug delivery noninvasively (in vivo)
US20020039594A1 (en) * 1997-05-13 2002-04-04 Evan C. Unger Solid porous matrices and methods of making and using the same
US6416740B1 (en) 1997-05-13 2002-07-09 Bristol-Myers Squibb Medical Imaging, Inc. Acoustically active drug delivery systems
DE69841002D1 (en) * 1997-05-14 2009-09-03 Univ British Columbia Highly effective encapsulation of nucleic acids in lipid vesicles
DE19724796A1 (en) * 1997-06-06 1998-12-10 Max Delbrueck Centrum Antitumor therapy agents
US6113947A (en) * 1997-06-13 2000-09-05 Genentech, Inc. Controlled release microencapsulated NGF formulation
US6663899B2 (en) 1997-06-13 2003-12-16 Genentech, Inc. Controlled release microencapsulated NGF formulation
US6217886B1 (en) 1997-07-14 2001-04-17 The Board Of Trustees Of The University Of Illinois Materials and methods for making improved micelle compositions
US6548047B1 (en) 1997-09-15 2003-04-15 Bristol-Myers Squibb Medical Imaging, Inc. Thermal preactivation of gaseous precursor filled compositions
IL135060A (en) 1997-09-18 2005-12-18 Skyepharma Inc Sustained release liposomal anesthetic compositions and methods for the preparation thereof
US7229841B2 (en) * 2001-04-30 2007-06-12 Cytimmune Sciences, Inc. Colloidal metal compositions and methods
EP1030652B1 (en) 1997-11-14 2012-04-25 Pacira Pharmaceuticals, Inc. Production of multivesicular liposomes
US7192589B2 (en) 1998-09-16 2007-03-20 Genentech, Inc. Treatment of inflammatory disorders with STIgMA immunoadhesins
CA2309358A1 (en) 1997-11-21 1999-06-03 Genentech, Inc. A-33 related antigens and their pharmacological uses
US6787132B1 (en) * 1997-12-04 2004-09-07 Yissum Research Development Company Of The Hebrew University Of Jerusalem Combined chemo-immunotherapy with liposomal drugs and cytokines
EP1947119A3 (en) 1997-12-12 2012-12-19 Genentech, Inc. Treatment of cancer with anti-erb2 antibodies in combination with a chemotherapeutic agent
US6123923A (en) 1997-12-18 2000-09-26 Imarx Pharmaceutical Corp. Optoacoustic contrast agents and methods for their use
US20010003580A1 (en) 1998-01-14 2001-06-14 Poh K. Hui Preparation of a lipid blend and a phospholipid suspension containing the lipid blend
EP1510579B1 (en) 1998-02-04 2009-08-05 Genentech, Inc. Use of heregulin as an epithelial cell growth factor
DE69925461T2 (en) * 1998-02-09 2006-04-27 Bracco International B.V. TARGETED DISTRIBUTION OF BIOLOGICAL-ACTIVE MEDIA
US6942859B2 (en) 1998-03-13 2005-09-13 University Of Southern California Red blood cells covalently bound with polymers
US6312685B1 (en) 1998-03-13 2001-11-06 Timothy C. Fisher Red blood cells covalently bound with two different polyethylene glycol derivatives
DK2016951T3 (en) 1998-03-17 2012-09-24 Genentech Inc VEGF and BMP1 homologous polypeptides
US6433012B1 (en) * 1998-03-25 2002-08-13 Large Scale Biology Corp. Method for inhibiting inflammatory disease
KR20010042168A (en) * 1998-03-25 2001-05-25 바이오소스 테크놀로지스 인코포레이티드 Benzoates derivatives for inhibiting angiogenesis
US6858706B2 (en) * 1998-04-07 2005-02-22 St. Jude Children's Research Hospital Polypeptide comprising the amino acid of an N-terminal choline binding protein a truncate, vaccine derived therefrom and uses thereof
WO1999055306A1 (en) * 1998-04-27 1999-11-04 Opperbas Holding B.V. Pharmaceutical composition comprising factor viii and neutral liposomes
US6986902B1 (en) * 1998-04-28 2006-01-17 Inex Pharmaceuticals Corporation Polyanionic polymers which enhance fusogenicity
US6448224B1 (en) 1998-05-06 2002-09-10 St. Jude Children's Research Hospital Antibiotics and methods of using the same
US6331407B1 (en) 1998-05-06 2001-12-18 St. Jude Children's Research Hospital Antibiotics and methods of using the same
US6169078B1 (en) * 1998-05-12 2001-01-02 University Of Florida Materials and methods for the intracellular delivery of substances
EP3112468A1 (en) 1998-05-15 2017-01-04 Genentech, Inc. Il-17 homologous polypeptides and therapeutic uses thereof
DE69936382T3 (en) 1998-05-15 2011-07-07 Genentech, Inc., Calif. THERAPEUTIC USES OF IL-17 HOMOLOGOUS POLYPEPTIDE
EP1865061A3 (en) 1998-05-15 2007-12-19 Genentech, Inc. IL-17 homologous polypeptides and therapeutic uses thereof
EP1086138B1 (en) 1998-06-12 2009-09-23 Genentech, Inc. Monoclonal antibodies, cross-reactive antibodies and method for producing the same
US6726925B1 (en) * 1998-06-18 2004-04-27 Duke University Temperature-sensitive liposomal formulation
US6200598B1 (en) * 1998-06-18 2001-03-13 Duke University Temperature-sensitive liposomal formulation
US20040229363A1 (en) * 1998-06-24 2004-11-18 Ed Nolan High efficiency transfection based on low electric field strength, long pulse length
US20030022854A1 (en) * 1998-06-25 2003-01-30 Dow Steven W. Vaccines using nucleic acid-lipid complexes
US6693086B1 (en) * 1998-06-25 2004-02-17 National Jewish Medical And Research Center Systemic immune activation method using nucleic acid-lipid complexes
US20040247662A1 (en) * 1998-06-25 2004-12-09 Dow Steven W. Systemic immune activation method using nucleic acid-lipid complexes
NZ509186A (en) 1998-07-17 2005-01-28 Skyepharma Inc Biodegradable compositions for the controlled release of encapsulated substances
US20020172678A1 (en) 2000-06-23 2002-11-21 Napoleone Ferrara EG-VEGF nucleic acids and polypeptides and methods of use
US6703381B1 (en) 1998-08-14 2004-03-09 Nobex Corporation Methods for delivery therapeutic compounds across the blood-brain barrier
CN1205923C (en) * 1998-09-16 2005-06-15 阿尔萨公司 Liposome-entrapped topoisomerase inhibitors
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
US6419709B1 (en) 1998-10-02 2002-07-16 Guilford Pharmaceuticals, Inc. Biodegradable terephthalate polyester-poly(Phosphite) compositions, articles, and methods of using the same
US6153212A (en) * 1998-10-02 2000-11-28 Guilford Pharmaceuticals Inc. Biodegradable terephthalate polyester-poly (phosphonate) compositions, articles, and methods of using the same
US6660843B1 (en) * 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
DK1124961T3 (en) 1998-10-23 2007-04-10 Amgen Inc Thrombopoietic compounds
EP1950300A3 (en) 1998-11-18 2011-03-23 Genentech, Inc. Antibody variants with higher binding affinity compared to parent antibodies
EP1143953A3 (en) * 1998-11-20 2002-02-06 Genentech, Inc. Method of inhibiting angiogenesis
US6773911B1 (en) 1998-11-23 2004-08-10 Amgen Canada Inc. Apoptosis-inducing factor
EP1820859B9 (en) 1998-12-22 2009-10-28 Genentech, Inc. Methods and compositions for inhibiting neoplastic cell growth
DE69926536T3 (en) 1998-12-22 2013-09-12 Genentech, Inc. ANTAGONISTS OF VASCULAR ENDOTHELIAL CELL GROWTH FACTORS AND ITS APPLICATION
US6350464B1 (en) 1999-01-11 2002-02-26 Guilford Pharmaceuticals, Inc. Methods for treating ovarian cancer, poly (phosphoester) compositions, and biodegradable articles for same
US6818227B1 (en) * 1999-02-08 2004-11-16 Alza Corporation Liposome composition and method for administration of a radiosensitizer
US6537585B1 (en) 1999-03-26 2003-03-25 Guilford Pharmaceuticals, Inc. Methods and compositions for treating solid tumors
US6379698B1 (en) * 1999-04-06 2002-04-30 Isis Pharmaceuticals, Inc. Fusogenic lipids and vesicles
US7112337B2 (en) 1999-04-23 2006-09-26 Alza Corporation Liposome composition for delivery of nucleic acid
CA2270600A1 (en) * 1999-05-03 2000-11-03 Infectio Recherche Inc. Method and formulations for the treatment of diseases, particularly those caused by human immunodeficiency virus and leishmania
EP1645289A1 (en) 1999-05-07 2006-04-12 Genentech, Inc. Treatment of autoimmune diseases with antagonists which bind to B cell surface markers
AU4430200A (en) * 1999-05-11 2000-11-21 Sankyo Company Limited Liposome preparation of fat-soluble antitumor drug
US20040229779A1 (en) * 1999-05-14 2004-11-18 Ramesh Kekuda Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20040023874A1 (en) * 2002-03-15 2004-02-05 Burgess Catherine E. Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US6974684B2 (en) * 2001-08-08 2005-12-13 Curagen Corporation Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20040067490A1 (en) * 2001-09-07 2004-04-08 Mei Zhong Therapeutic polypeptides, nucleic acids encoding same, and methods of use
AU5152700A (en) 1999-06-15 2001-01-02 Genentech Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7169889B1 (en) 1999-06-19 2007-01-30 Biocon Limited Insulin prodrugs hydrolyzable in vivo to yield peglylated insulin
US6309633B1 (en) 1999-06-19 2001-10-30 Nobex Corporation Amphiphilic drug-oligomer conjugates with hydroyzable lipophile components and methods for making and using the same
CA2376849C (en) * 1999-06-24 2008-10-14 Kyowa Hakko Kogyo Co., Ltd. Method of inhibiting leakage of drug encapsulated in liposomes
IL146954A0 (en) 1999-06-25 2002-08-14 Genentech Inc HUMANIZED ANTI-ErbB2 ANTIBODIES AND TREATMENT WITH ANTI-ErbB2 ANTIBODIES
US6352996B1 (en) 1999-08-03 2002-03-05 The Stehlin Foundation For Cancer Research Liposomal prodrugs comprising derivatives of camptothecin and methods of treating cancer using these prodrugs
KR20090126330A (en) 1999-08-27 2009-12-08 제넨테크, 인크. Dosages for treatment with anti-erbb2 antibodies
KR100510795B1 (en) * 1999-08-31 2005-08-30 Compositions and Methods for the Treatment of Tumor
KR20010085996A (en) * 1999-09-03 2001-09-07 스티븐 엠. 오드레 Compositions and methods for the prevention or treatment of cancer and bone loss associated with cancer
WO2001026625A2 (en) 1999-10-08 2001-04-19 Alza Corp Neutral-cationic lipid for nucleic acid and drug delivery
IT1315253B1 (en) * 1999-10-22 2003-02-03 Novuspharma Spa LIPOSOMIAL PREPARATION OF 6,9-BIS- (2-AMINOXYL) AMINO | BENZOG | ISOCHINOLIN-5,10-DIONE DIMALEATO
US7067111B1 (en) * 1999-10-25 2006-06-27 Board Of Regents, University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US6511676B1 (en) * 1999-11-05 2003-01-28 Teni Boulikas Therapy for human cancers using cisplatin and other drugs or genes encapsulated into liposomes
ES2629683T3 (en) 1999-11-30 2017-08-14 Mayo Foundation For Medical Education And Research B7-H1, a new immunoregulatory molecule
EP1690872A3 (en) 1999-12-01 2006-08-23 Genentech, Inc. Composition and methods for the diagnosis of tumours
US6593308B2 (en) 1999-12-03 2003-07-15 The Regents Of The University Of California Targeted drug delivery with a hyaluronan ligand
AU2136501A (en) * 1999-12-10 2001-06-18 Celator Technologies Inc. Lipid carrier compositions with protected surface reactive functions
EP2290081A3 (en) 1999-12-23 2012-08-01 Genentech, Inc. IL-17 homologous polypeptide and therapeutic uses thereof
US20040009229A1 (en) * 2000-01-05 2004-01-15 Unger Evan Charles Stabilized nanoparticle formulations of camptotheca derivatives
JP2003520210A (en) * 2000-01-05 2003-07-02 イマレックス セラピューティクス, インコーポレイテッド Pharmaceutical formulations for delivery of drugs with low water solubility
CN1425066B (en) * 2000-01-13 2010-05-26 杰南技术公司 Novel stra6 polypeptides
WO2001054666A1 (en) * 2000-01-28 2001-08-02 Alza Corporation Liposomes containing an entrapped compound in supersaturated solution
US20030176385A1 (en) * 2000-02-15 2003-09-18 Jingfang Ju Antisense modulation of protein expression
JP4922824B2 (en) * 2000-04-03 2012-04-25 参天製薬株式会社 Delivery substance and drug delivery system using the same
CA2403425C (en) 2000-04-11 2013-08-27 Genentech, Inc. Multivalent antibodies and uses therefor
EP1272160B1 (en) * 2000-04-12 2007-01-17 Liplasome Pharma A/S Lipid-based drug delivery systems for topical application
EP1290013B1 (en) 2000-04-21 2006-03-08 Amgen Inc. Apo-ai/aii peptide derivatives
US6667300B2 (en) * 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US7030219B2 (en) * 2000-04-28 2006-04-18 Johns Hopkins University B7-DC, Dendritic cell co-stimulatory molecules
US6677136B2 (en) 2000-05-03 2004-01-13 Amgen Inc. Glucagon antagonists
WO2001085131A2 (en) * 2000-05-11 2001-11-15 Celator Technologies Inc. Lipid carrier compositions for improved drug retention
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
AU6531101A (en) 2000-06-02 2001-12-17 Genentech Inc Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2001091807A2 (en) 2000-06-02 2001-12-06 Board Of Regents Ethylenedicysteine (ec)-drug conjugates
US20030072794A1 (en) * 2000-06-09 2003-04-17 Teni Boulikas Encapsulation of plasmid DNA (lipogenes™) and therapeutic agents with nuclear localization signal/fusogenic peptide conjugates into targeted liposome complexes
EP2052742A1 (en) 2000-06-20 2009-04-29 Biogen Idec Inc. Treatment of B-cell associated diseases such as malignancies and autoimmune diseases using a cold anti-CD20 antibody/radiolabeled anti-CD22 antibody combination
EP2792747A1 (en) 2000-06-23 2014-10-22 Genentech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
EP2075253A1 (en) 2000-06-23 2009-07-01 Genentech, Inc. Compositions and methds for the diagnosis and treatment of disorders involving angiogensis
US20040023259A1 (en) * 2000-07-26 2004-02-05 Luca Rastelli Therapeutic polypeptides, nucleic acids encoding same, and methods of use
ATE412009T1 (en) 2000-08-24 2008-11-15 Genentech Inc METHOD FOR INHIBITING IL-22 INDUCED PAP1
EP1944317A3 (en) 2000-09-01 2008-09-17 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030133972A1 (en) * 2000-10-11 2003-07-17 Targesome, Inc. Targeted multivalent macromolecules
US7875585B2 (en) 2000-10-18 2011-01-25 Robert Sackstein Hematopoietic cell E-selectin / L-selectin ligand glycosylated CD44 polypeptide
US7045283B2 (en) 2000-10-18 2006-05-16 The Regents Of The University Of California Methods of high-throughput screening for internalizing antibodies
JP2004512345A (en) * 2000-11-02 2004-04-22 スミスクライン・ビーチャム・コーポレイション Receptor antagonist-lipid conjugates and delivery vehicles containing the same
DE10056136A1 (en) * 2000-11-07 2002-05-16 Nemod New Modalities Inhibiting leukocyte or tumor cell adhesion to vascular endothelial cells e.g. for combating inflammation or metastasis, using e.g. pregnancy proteins or selectin binding liposomes containing calcium-binding compound
AU2002239607A1 (en) * 2000-11-30 2002-06-11 Baxter Healthcare Corporation Ahf associated dispersion system and method for preparation
US7625584B2 (en) * 2000-11-30 2009-12-01 The Research Foundation Of State University Of New York Method of complexing a protein by the use of a dispersed system and proteins thereof
US20020127635A1 (en) * 2000-11-30 2002-09-12 Balasubramanian Sathyamangalam V. Method of complexing a protein by the use of a dispersed system and proteins thereof
US8110218B2 (en) * 2000-11-30 2012-02-07 The Research Foundation Of State University Of New York Compositions and methods for less immunogenic protein-lipid complexes
US20060014674A1 (en) 2000-12-18 2006-01-19 Dennis Keith Methods for preparing purified lipopeptides
CN102070703B (en) * 2000-12-18 2014-08-06 卡比斯特制药公司 Methods for preparing purified daptomycin
WO2002059145A1 (en) * 2000-12-18 2002-08-01 Cubist Pharmaceuticals, Inc. Methods for preparing purified lipopeptides
US20040077604A1 (en) 2001-12-19 2004-04-22 Lenard Lichtenberger Method and compositions employing formulations of lecithin oils and nsaids for protecting the gastrointestinal tract and providingenhanced therapeutic activity
US6964849B2 (en) 2001-01-11 2005-11-15 Curagen Corporation Proteins and nucleic acids encoding same
US20040043928A1 (en) * 2001-08-02 2004-03-04 Ramesh Kekuda Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20020159996A1 (en) 2001-01-31 2002-10-31 Kandasamy Hariharan Use of CD23 antagonists for the treatment of neoplastic disorders
AR035779A1 (en) 2001-02-06 2004-07-14 Genetics Inst Llc FUSION POLYPEPTIDES DERIVED FROM GLICOPROTEIN IB PLATE ALFA AND METHODS OF USE OF THE SAME
US6867183B2 (en) * 2001-02-15 2005-03-15 Nobex Corporation Pharmaceutical compositions of insulin drug-oligomer conjugates and methods of treating diseases therewith
US7060675B2 (en) 2001-02-15 2006-06-13 Nobex Corporation Methods of treating diabetes mellitus
US7087726B2 (en) 2001-02-22 2006-08-08 Genentech, Inc. Anti-interferon-α antibodies
WO2002072011A2 (en) * 2001-03-08 2002-09-19 Targesome, Inc. Stabilized therapeutic and imaging agents
HUP0303616A3 (en) * 2001-03-26 2006-07-28 Alza Corp Mountain View Liposome composition for improved intracellular delivery of a therapeutic agent
US20040126900A1 (en) * 2001-04-13 2004-07-01 Barry Stephen E High affinity peptide- containing nanoparticles
AU2002256398A2 (en) * 2001-04-30 2002-11-11 Targeted Genetics Corporation Lipid-comprising drug delivery complexes and methods for their production
WO2002087541A1 (en) * 2001-04-30 2002-11-07 Protiva Biotherapeutics Inc. Lipid-based formulations for gene transfer
GB0111279D0 (en) * 2001-05-10 2001-06-27 Nycomed Imaging As Radiolabelled liposomes
NZ529267A (en) 2001-05-11 2006-05-26 Amgen Inc Peptides and related molecules that bind to tall-1
US20050261214A1 (en) * 2001-05-15 2005-11-24 Serge Braun Complexes for transferring substances of interest into a cell
US20020197253A1 (en) * 2001-05-22 2002-12-26 Cheek Dennis J. Compositions and methods for promoting or inhibiting NDPK
AU2002310046A1 (en) * 2001-05-22 2002-12-03 Duke University Compositions and methods for inhibiting metastasis
CA2448956C (en) 2001-05-30 2017-10-03 Genentech, Inc. Anti-ngf antibodies for the treatment of various disorders
EP1395243A2 (en) * 2001-05-31 2004-03-10 SkyePharma Inc. Encapsulation of nanosuspensions in liposomes and microspheres
US6713452B2 (en) 2001-06-04 2004-03-30 Nobex Corporation Mixtures of calcitonin drug-oligomer conjugates comprising polyalkylene glycol, uses thereof, and methods of making same
US6828305B2 (en) * 2001-06-04 2004-12-07 Nobex Corporation Mixtures of growth hormone drug-oligomer conjugates comprising polyalkylene glycol, uses thereof, and methods of making same
US6835802B2 (en) * 2001-06-04 2004-12-28 Nobex Corporation Methods of synthesizing substantially monodispersed mixtures of polymers having polyethylene glycol moieties
US6858580B2 (en) * 2001-06-04 2005-02-22 Nobex Corporation Mixtures of drug-oligomer conjugates comprising polyalkylene glycol, uses thereof, and methods of making same
US7713932B2 (en) 2001-06-04 2010-05-11 Biocon Limited Calcitonin drug-oligomer conjugates, and uses thereof
US6828297B2 (en) * 2001-06-04 2004-12-07 Nobex Corporation Mixtures of insulin drug-oligomer conjugates comprising polyalkylene glycol, uses thereof, and methods of making same
US20070160576A1 (en) 2001-06-05 2007-07-12 Genentech, Inc. IL-17A/F heterologous polypeptides and therapeutic uses thereof
AU2002312410A1 (en) * 2001-06-08 2002-12-23 Target Protein Technologies, Inc. Tissue-specific endothelial membrane proteins
ATE522623T1 (en) 2001-06-20 2011-09-15 Genentech Inc COMPOSITIONS AND METHODS FOR THE TREATMENT AND DIAGNOSIS OF LUNG TUMORS
US20050107595A1 (en) * 2001-06-20 2005-05-19 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7803915B2 (en) * 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
CN1827766B (en) 2001-06-28 2010-08-25 徐荣祥 In vitro cell cultivation method
US20030087274A1 (en) * 2001-07-05 2003-05-08 Anderson David W. Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20040029790A1 (en) * 2001-07-05 2004-02-12 Meera Patturajan Novel human proteins, polynucleotides encoding them and methods of using the same
WO2003007915A2 (en) * 2001-07-19 2003-01-30 Guilford Pharmaceuticals, Inc. Compositions for treatment of head and neck cancers, and methods of making and using the same
WO2003007914A2 (en) * 2001-07-19 2003-01-30 Guilford Pharmaceuticals, Inc. Biocompatible polymer containing composition for treatment of prostate cancers
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
CZ2004300A3 (en) * 2001-07-29 2005-03-16 Yissum Research Development Company Of The Hebrew Osteogenic growth oligopeptides as stimulants of hematopoiesis
US20040030096A1 (en) * 2001-08-02 2004-02-12 Linda Gorman Novel human proteins, polynucleotides encoding them and methods of using the same
US20030059375A1 (en) * 2001-08-20 2003-03-27 Transave, Inc. Method for treating lung cancers
EP1575916B1 (en) 2001-08-31 2013-05-22 The Rockefeller University Phosphodiesterase activity and regulation of phosphodiesterase 1-b-mediated signaling in brain
US20040235068A1 (en) * 2001-09-05 2004-11-25 Levinson Arthur D. Methods for the identification of polypeptide antigens associated with disorders involving aberrant cell proliferation and compositions useful for the treatment of such disorders
US7312192B2 (en) * 2001-09-07 2007-12-25 Biocon Limited Insulin polypeptide-oligomer conjugates, proinsulin polypeptide-oligomer conjugates and methods of synthesizing same
US6913903B2 (en) * 2001-09-07 2005-07-05 Nobex Corporation Methods of synthesizing insulin polypeptide-oligomer conjugates, and proinsulin polypeptide-oligomer conjugates and methods of synthesizing same
US7196059B2 (en) * 2001-09-07 2007-03-27 Biocon Limited Pharmaceutical compositions of insulin drug-oligomer conjugates and methods of treating diseases therewith
US7166571B2 (en) * 2001-09-07 2007-01-23 Biocon Limited Insulin polypeptide-oligomer conjugates, proinsulin polypeptide-oligomer conjugates and methods of synthesizing same
US7547673B2 (en) * 2001-09-13 2009-06-16 The Johns Hopkins University Therapeutics for cancer using 3-bromopyruvate and other selective inhibitors of ATP production
EP2153843B1 (en) 2001-09-18 2012-08-15 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7981863B2 (en) 2001-09-19 2011-07-19 Neuronova Ab Treatment of Parkinson's disease with PDGF
EP1438021A2 (en) * 2001-09-28 2004-07-21 Esperion Therapeutics Inc. Methods and apparatus for extrusion of vesicles at high pressure
DE10148065A1 (en) * 2001-09-28 2003-04-17 Max Planck Gesellschaft (Ester) -lysolecithins in liposomes
US20030199442A1 (en) * 2001-10-09 2003-10-23 Alsobrook John P. Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US7332474B2 (en) * 2001-10-11 2008-02-19 Amgen Inc. Peptides and related compounds having thrombopoietic activity
WO2005049075A2 (en) 2003-11-17 2005-06-02 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
CA2465860A1 (en) 2001-11-02 2004-04-22 Insert Therapeutics, Inc. Methods and compositions for therapeutic use of rna interference
WO2003039595A2 (en) * 2001-11-07 2003-05-15 Inex Pharmaceuticals Corporation Mucosal adjuvants comprising an oligonucleotide and a cationic lipid
US20040219201A1 (en) * 2001-12-06 2004-11-04 Yechezkel Barenholz Tempamine compositions and methods of use
JP2005525095A (en) 2002-01-02 2005-08-25 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
IL162838A0 (en) 2002-01-04 2005-11-20 Univ Rockefeller Compositions and methods for prevention and treatment peptide-of amyloid- related disorders
US20040052928A1 (en) * 2002-09-06 2004-03-18 Ehud Gazit Peptides and methods using same for diagnosing and treating amyloid-associated diseases
US7781396B2 (en) * 2002-01-31 2010-08-24 Tel Aviv University Future Technology Development L.P. Peptides directed for diagnosis and treatment of amyloid-associated disease
EP1487273B1 (en) 2002-02-13 2009-01-21 Immunology Laboratories, Inc. Compositions and methods for treatment of microbial infections
WO2003072035A2 (en) 2002-02-22 2003-09-04 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US20100311954A1 (en) * 2002-03-01 2010-12-09 Xencor, Inc. Optimized Proteins that Target Ep-CAM
US20070122406A1 (en) 2005-07-08 2007-05-31 Xencor, Inc. Optimized proteins that target Ep-CAM
US20090042291A1 (en) * 2002-03-01 2009-02-12 Xencor, Inc. Optimized Fc variants
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US20030180712A1 (en) 2002-03-20 2003-09-25 Biostratum Ab Inhibition of the beta3 subunit of L-type Ca2+ channels
US20040009216A1 (en) * 2002-04-05 2004-01-15 Rodrigueza Wendi V. Compositions and methods for dosing liposomes of certain sizes to treat or prevent disease
EP2011886A3 (en) 2002-04-16 2009-02-11 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
CA2483476A1 (en) * 2002-04-22 2003-10-30 Absorber, Ab Fusion polypeptides and methods for inhibiting microbial adhesion
US20040009944A1 (en) * 2002-05-10 2004-01-15 Inex Pharmaceuticals Corporation Methylated immunostimulatory oligonucleotides and methods of using the same
US7199107B2 (en) 2002-05-23 2007-04-03 Isis Pharmaceuticals, Inc. Antisense modulation of kinesin-like 1 expression
EP2305710A3 (en) 2002-06-03 2013-05-29 Genentech, Inc. Synthetic antibody phage libraries
AU2003236521A1 (en) * 2002-06-13 2003-12-31 Nobex Corporation Methods of reducing hypoglycemic episodes in the treatment of diabetes mellitus
US7901708B2 (en) 2002-06-28 2011-03-08 Protiva Biotherapeutics, Inc. Liposomal apparatus and manufacturing methods
AU2003241598B2 (en) * 2002-07-02 2009-11-05 Nanotx Corp. Radiolabeled compounds and liposomes and their methods of making and using the same
EP1528915A2 (en) * 2002-07-03 2005-05-11 Aphton Corporation Liposomal vaccine
US20050169979A1 (en) * 2002-07-03 2005-08-04 Dov Michaeli Liposomal vaccine
US20040247661A1 (en) * 2002-07-03 2004-12-09 Dov Michaeli Liposomal vaccine
JP2006517384A (en) 2002-07-08 2006-07-27 ジェネンテック・インコーポレーテッド Compositions and methods for the treatment of immune related diseases
US7625563B2 (en) * 2002-07-15 2009-12-01 Board Of Regents, The University Of Texas System Cancer treatment methods using selected immunoconjugates for binding to aminophospholipids
EP1585966B8 (en) 2002-07-15 2012-03-07 F. Hoffmann-La Roche AG Treatment of cancer with the anti-ErbB2 antibody rhuMAb 2C4
US7615223B2 (en) * 2002-07-15 2009-11-10 Board Of Regents, The University Of Texas System Selected immunoconjugates for binding to aminophospholipids
CA2894009C (en) * 2002-07-15 2016-12-06 Board Of Regents, The University Of Texas System Selected antibodies binding to anionic phospholipids and aminophospholipids and their use in treatment
US7714109B2 (en) * 2002-07-15 2010-05-11 Board Of Regents, The University Of Texas System Combinations and kits for cancer treatment using selected antibodies to aminophospholipids
US7622118B2 (en) * 2002-07-15 2009-11-24 Board Of Regents, The University Of Texas System Cancer treatment methods using selected antibodies to aminophospholipids
US7678386B2 (en) * 2002-07-15 2010-03-16 Board Of Regents The University Of Texas Liposomes coated with selected antibodies that bind to aminophospholipids
US7572448B2 (en) * 2002-07-15 2009-08-11 Board Of Regents, The University Of Texas System Combined cancer treatment methods using selected antibodies to aminophospholipids
AU2003252075A1 (en) * 2002-07-16 2004-02-02 University Of South Florida Human immunosuppressive protein
US20040161423A1 (en) * 2002-07-18 2004-08-19 Sanjeev Kumar (Mendiratta) Polymer modified anti-angiogenic serpins
US9186322B2 (en) * 2002-08-02 2015-11-17 Insmed Incorporated Platinum aggregates and process for producing the same
AU2003302314A1 (en) * 2002-08-02 2004-07-09 Transave, Inc. Platinum aggregates and process for producing the same
PT1534335E (en) 2002-08-14 2012-02-28 Macrogenics Inc Fcgammariib-specific antibodies and methods of use thereof
EP1393720A1 (en) * 2002-08-27 2004-03-03 Universiteit Utrecht Vesicle-encapsulated corticosteroids for treatment of cancer
WO2004019866A2 (en) 2002-08-28 2004-03-11 Immunex Corporation Compositions and methods for treating cardiovascular disease
US20070231379A1 (en) * 2002-08-29 2007-10-04 Slater James L Liposome-entrapped topoisomerase inhibitors
AR036316A1 (en) * 2002-08-29 2004-08-25 Monte Verde S A A PHARMACEUTICAL COMPOSITION OF SMALL SIZE LIPOSOMES AND PREPARATION METHOD
WO2004022003A2 (en) 2002-09-06 2004-03-18 University Of South Florida Materials and methods for treatment of allergic diseases
US20080214437A1 (en) * 2002-09-06 2008-09-04 Mohapatra Shyam S Methods and compositions for reducing activity of the atrial natriuretic peptide receptor and for treatment of diseases
CA2498008C (en) 2002-09-11 2014-02-04 Genentech, Inc. Novel composition and methods for the treatment of immune related diseases
EP1578373A4 (en) 2002-09-11 2007-10-24 Genentech Inc Novel compositions and methods for the treatment of immune related diseases
US20050196382A1 (en) * 2002-09-13 2005-09-08 Replicor, Inc. Antiviral oligonucleotides targeting viral families
EP2444409A2 (en) 2002-09-16 2012-04-25 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US6919426B2 (en) 2002-09-19 2005-07-19 Amgen Inc. Peptides and related molecules that modulate nerve growth factor activity
AU2003279084A1 (en) 2002-09-25 2004-04-19 Genentech, Inc. Novel compositions and methods for the treatment of psoriasis
EP2042517B1 (en) 2002-09-27 2012-11-14 Xencor, Inc. Optimized FC variants and methods for their generation
US8129330B2 (en) * 2002-09-30 2012-03-06 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
US20040062748A1 (en) * 2002-09-30 2004-04-01 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
US7432351B1 (en) 2002-10-04 2008-10-07 Mayo Foundation For Medical Education And Research B7-H1 variants
US20060147450A1 (en) * 2002-10-04 2006-07-06 Shelton David L Methods for treating cardiac arrhythmia and preventing death due to cardiac arrhythmia using ngf antagonists
AU2003304238A1 (en) 2002-10-08 2005-01-13 Rinat Neuroscience Corp. Methods for treating post-surgical pain by administering an anti-nerve growth factor antagonist antibody and compositions containing the same
UA80447C2 (en) * 2002-10-08 2007-09-25 Methods for treating pain by administering nerve growth factor antagonist and opioid analgesic
ZA200502612B (en) * 2002-10-08 2007-07-25 Rinat Neuroscience Corp Methods for treating post-surgical pain by administering a nerve crowth factor antagonist and compositions containing the same
BR0315157A (en) * 2002-10-09 2005-08-09 Rinat Neuroscience Corp Methods of treating alzheimer's disease by employing antibodies directed against amyloid beta peptide and compositions thereof
EP2322203A3 (en) 2002-10-29 2011-07-27 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
JP2006515318A (en) * 2002-10-29 2006-05-25 ファルマシア・コーポレーション Specifically associated cancer-related gene, polypeptide encoded thereby and method of use thereof
WO2004044139A2 (en) 2002-11-05 2004-05-27 Isis Parmaceuticals, Inc. Modified oligonucleotides for use in rna interference
CA2504720C (en) 2002-11-05 2013-12-24 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
JP2006516094A (en) 2002-11-08 2006-06-22 ジェネンテック・インコーポレーテッド Compositions and methods for treatment of natural killer cell related diseases
US20040093198A1 (en) * 2002-11-08 2004-05-13 Carbon Design Systems Hardware simulation with access restrictions
AU2003285200A1 (en) * 2002-11-09 2004-06-03 Nobex Corporation Modified carbamate-containing prodrugs and methods of synthesizing same
US20050158375A1 (en) * 2002-11-15 2005-07-21 Toshikiro Kimura Pharmaceutical composition containing liposomes for treating cancer
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
EP2314676A1 (en) 2002-11-26 2011-04-27 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
CN100558398C (en) 2002-11-26 2009-11-11 拜奥康有限公司 Natriuresis chemical compound, conjugate and the application thereof of modifying
US7648962B2 (en) * 2002-11-26 2010-01-19 Biocon Limited Natriuretic compounds, conjugates, and uses thereof
US7491699B2 (en) * 2002-12-09 2009-02-17 Ramot At Tel Aviv University Ltd. Peptide nanostructures and methods of generating and using the same
US20040219204A1 (en) * 2002-12-19 2004-11-04 Huang Ken Shi Kun Method of treating angiogenic tissue growth
US9498530B2 (en) 2002-12-24 2016-11-22 Rinat Neuroscience Corp. Methods for treating osteoarthritis pain by administering a nerve growth factor antagonist and compositions containing the same
US7569364B2 (en) * 2002-12-24 2009-08-04 Pfizer Inc. Anti-NGF antibodies and methods using same
DK2270048T3 (en) 2002-12-24 2016-01-18 Rinat Neuroscience Corp Anti-NGF antibodies and methods for their use
US8980310B2 (en) * 2002-12-31 2015-03-17 Bharat Serums and Vaccines, Ltd. Non-pegylated long-circulating liposomes
CN1756533B (en) 2002-12-31 2010-06-16 Bsv研究及开发私人有限公司 Non-pegylated long-circulating liposomes
ATE426575T1 (en) 2003-01-07 2009-04-15 Univ Ramot PEPTIDE ANOSTRUCTURES CONTAINING FOREIGN MATERIAL AND METHOD FOR PRODUCING THE SAME
WO2004063216A1 (en) * 2003-01-10 2004-07-29 Yamanouchi Pharmaceutical Co., Ltd. Conjugate for retention in blood and cancer tissue-specific drug delivery
US7169892B2 (en) 2003-01-10 2007-01-30 Astellas Pharma Inc. Lipid-peptide-polymer conjugates for long blood circulation and tumor specific drug delivery systems
WO2004072284A1 (en) 2003-02-11 2004-08-26 Antisense Therapeutics Ltd Modulation of insulin like growth factor i receptor expression
EP1594441B1 (en) 2003-02-19 2010-12-15 Rinat Neuroscience Corp. Method for treating pain by administering a nerve growth factor antagonist and an nsaid and composition containing the same
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US7968115B2 (en) 2004-03-05 2011-06-28 Board Of Regents, The University Of Texas System Liposomal curcumin for treatment of cancer
US20060198882A1 (en) * 2003-03-21 2006-09-07 Yechezkel Barenholz Stable liposomes or micelles comprising a sphinolipid and a peg-lipopolymer
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
AU2004227847A1 (en) 2003-03-31 2004-10-21 Alza Corporation Lipid particles having asymmetric lipid coating and method of preparing same
ZA200507757B (en) 2003-04-04 2007-01-31 Genentech Inc High concentration antibody and protein formulations
RU2358762C9 (en) 2003-04-09 2016-10-10 Джинентех, Инк. Treatment of autoimmune diseases in patient suffering from inadequate response to tnf-alpha inhibitor
ATE395101T1 (en) * 2003-04-15 2008-05-15 Opperbas Holding Bv PHARMACEUTICAL COMPOSITION CONTAINING PROTEINS AND/OR POLYPEPTIDES AND COLLOID PARTICLES
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
WO2004094457A2 (en) * 2003-04-16 2004-11-04 Arizona Board Of Regents, Acting For And On Behalf Of, Arizona State University Stable rgd peptidomimetic composition
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
NZ563042A (en) 2003-05-12 2008-09-26 Affymax Inc Novel spacer moiety for poly(ethylene glycol)-modified peptide-based compounds
CN1820024B (en) 2003-05-12 2011-06-22 阿费麦克斯公司 Novel poly(ethylene glycol) modified compounds and uses thereof
WO2005000896A2 (en) * 2003-05-30 2005-01-06 Genentech, Inc. Polypeptides that bind an anti-tissue factor antibody and uses thereof
CN1829741A (en) 2003-05-30 2006-09-06 健泰科生物技术公司 Treatment with anti-VEGF antibodies
BRPI0410886A (en) 2003-06-03 2006-07-04 Isis Pharmaceuticals Inc double stranded compound, pharmaceutical composition, pharmaceutically acceptable salt, methods of modifying human survivin-encoding nucleic acid, inhibiting suvivin expression in cells or tissues, and treating a condition associated with suvivin expression or overexpression, and single stranded RNA oligonucleotide
EP1628624A2 (en) * 2003-06-04 2006-03-01 CANJI, Inc. Methods and compositions for interferon therapy
CA2528343A1 (en) 2003-06-06 2005-01-06 Genentech, Inc. Modulating the interaction between hgf beta chain and c-met
WO2004111089A2 (en) 2003-06-11 2004-12-23 WYETH A Corporaton of the State of Delaware Platelet glycoprotein ib alpha variant fusion polypeptides and methods of use thereof
EP1649061B1 (en) 2003-07-03 2010-08-25 University Of Medicine And Dentistry Of New Jersey Genes as diagnostic tools for autism
US7939058B2 (en) 2003-07-03 2011-05-10 University Of Southern California Uses of IL-12 in hematopoiesis
SI1641822T1 (en) 2003-07-08 2013-08-30 Genentech, Inc. Il-17 a/f heterologous polypeptides and therapeutic uses thereof
CA2532228C (en) * 2003-07-16 2017-02-14 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
CN100431525C (en) * 2003-07-17 2008-11-12 台湾东洋药品工业股份有限公司 Production method of liposome suspended liquid and products thereof
EP1648914A4 (en) 2003-07-31 2009-12-16 Regulus Therapeutics Inc Oligomeric compounds and compositions for use in modulation of small non-coding rnas
WO2005019258A2 (en) 2003-08-11 2005-03-03 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US20050054614A1 (en) * 2003-08-14 2005-03-10 Diacovo Thomas G. Methods of inhibiting leukocyte accumulation
WO2005016348A1 (en) * 2003-08-14 2005-02-24 Icos Corporation Method of inhibiting immune responses stimulated by an endogenous factor
DE602004019546D1 (en) * 2003-08-26 2009-04-02 Smithkline Beecham Corp HETEROFUNCTIONAL COPOLYMERS OF GLYCEROL AND POLYETHYLENE GLYCOL, ITS CONJUGATES AND COMPOSITIONS
US8986731B2 (en) * 2003-08-26 2015-03-24 Biolitec Pharma Marketing Ltd Pegylated liposomal formulations of hydrophobic photosensitizers for photodynamic therapy
US11324698B2 (en) 2003-08-28 2022-05-10 Vgsk Technologies, Inc. Sterically stabilized carrier for aerosol therapeutics, compositions and methods for treating the respiratory tract of a mammal
US8846079B1 (en) 2004-12-01 2014-09-30 Vgsk Technologies, Inc. Sterically stabilized carrier for aerosol therapeutics, compositions and methods for treating the respiratory tract of a mammal
US20060115523A1 (en) * 2004-12-01 2006-06-01 Konduri Kameswari S Sterically stabilized liposome and triamcinolone composition for treating the respiratory tract of a mammal
WO2005023844A1 (en) * 2003-09-03 2005-03-17 Kyowa Hakko Kogyo Co., Ltd. Compound modified with glycerol derivative
US20050233435A1 (en) * 2003-09-04 2005-10-20 Nyu Medical Center Plasmodium axenic liver stages as a noninfectious whole organism malaria vaccine
US20070123480A1 (en) * 2003-09-11 2007-05-31 Replicor Inc. Oligonucleotides targeting prion diseases
CA2551022C (en) * 2003-09-15 2013-06-04 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
KR100750788B1 (en) 2003-09-18 2007-08-20 아이시스 파마수티컬즈 인코포레이티드 4 MODULATION OF eIF4E EXPRESSION
WO2005027901A1 (en) * 2003-09-25 2005-03-31 Tel Aviv University Future Technology Development L.P. Compositions and methods using same for treating amyloid-associated diseases
US8101720B2 (en) * 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US7625707B2 (en) * 2003-10-02 2009-12-01 Ramot At Tel Aviv University Ltd. Antibacterial agents and methods of identifying and utilizing same
DK1678194T3 (en) 2003-10-10 2013-10-07 Alchemia Oncology Pty Ltd MODULATING SYNTHESIS AND DEGRADING HYALURONANE IN THE TREATMENT OF DISEASE
WO2005034979A2 (en) * 2003-10-11 2005-04-21 Inex Pharmaceuticals Corporation Methods and compositions for enhancing innate immunity and antibody dependent cellular cytotoxicity
TW200517114A (en) 2003-10-15 2005-06-01 Combinatorx Inc Methods and reagents for the treatment of immunoinflammatory disorders
KR20060123170A (en) * 2003-10-24 2006-12-01 알자 코포레이션 Preparation of lipid particles
WO2005040163A1 (en) * 2003-10-28 2005-05-06 Dr. Reddy's Laboratories Ltd Heterocyclic compounds that block the effects of advanced glycation end products (age)
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
US7915227B2 (en) * 2003-11-14 2011-03-29 Het Nederlands Kanker Instituut Pharmaceutical formulations employing short-chain sphingolipids and their use
US20050129753A1 (en) * 2003-11-14 2005-06-16 Gabizon Alberto A. Method for drug loading in liposomes
CN1914226B (en) * 2003-11-25 2012-02-01 达纳-法伯癌症研究院有限公司 Antibodies against SARS-COV and methods of use thereof
EP1701979A2 (en) * 2003-12-03 2006-09-20 Xencor, Inc. Optimized antibodies that target the epidermal growth factor receptor
US9050378B2 (en) * 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
US7312320B2 (en) 2003-12-10 2007-12-25 Novimmune Sa Neutralizing antibodies and methods of use thereof
CA2548282A1 (en) 2003-12-11 2005-06-30 Genentech, Inc. Methods and compositions for inhibiting c-met dimerization and activation
JP4503617B2 (en) 2003-12-23 2010-07-14 ライナット ニューロサイエンス コーポレイション Agonist anti-trkC antibody and method using the antibody
ES2543833T3 (en) 2004-01-07 2015-08-24 Novartis Vaccines And Diagnostics, Inc. M-CSF-specific monoclonal antibody and uses thereof
WO2005070466A2 (en) * 2004-01-15 2005-08-04 Alza Corporation Liposome composition for delivery of therapeutic agents
EP2363480A3 (en) 2004-01-20 2015-10-07 Isis Pharmaceuticals, Inc. Modulation of glucocorticoid receptor expression
US7468431B2 (en) 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
CA2559853A1 (en) * 2004-02-17 2005-10-13 University Of South Florida Materials and methods for treatment of inflammatory and cell proliferation disorders
US20050181035A1 (en) * 2004-02-17 2005-08-18 Dow Steven W. Systemic immune activation method using non CpG nucleic acids
ATE452147T1 (en) 2004-02-19 2010-01-15 Genentech Inc ANTIBODIES WITH CORRECTED CDR
US8784881B2 (en) 2004-03-05 2014-07-22 Board Of Regents, The University Of Texas System Liposomal curcumin for treatment of diseases
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
CN1679957B (en) * 2004-03-15 2010-06-09 尼普洛株式会社 A pharmaceutical composition containing liposomes for treating a cancer
WO2005089268A2 (en) 2004-03-15 2005-09-29 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of rna by rnase h
WO2005089448A2 (en) * 2004-03-18 2005-09-29 Transave, Inc. Administration of cisplatin by inhalation
US20070065522A1 (en) * 2004-03-18 2007-03-22 Transave, Inc. Administration of high potency platinum compound formulations by inhalation
EP1730280B1 (en) * 2004-03-26 2018-10-24 Curis, Inc. Rna interference modulators of hedgehog signaling and uses thereof
WO2005092388A1 (en) * 2004-03-26 2005-10-06 Terumo Kabushiki Kaisha Liposome preparation
US20050244869A1 (en) * 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
ATE456580T1 (en) * 2004-04-07 2010-02-15 Rinat Neuroscience Corp METHOD FOR PAIN TREATMENT IN BONE CANCER BY ADMINISTRATION OF A NGF ANTAGONIST
US7794713B2 (en) 2004-04-07 2010-09-14 Lpath, Inc. Compositions and methods for the treatment and prevention of hyperproliferative diseases
BRPI0509412A (en) * 2004-04-16 2007-09-04 Genentech Inc polychondritis or multiplex mononeuritis treatment method in mammals and industrialized article
US20150017671A1 (en) 2004-04-16 2015-01-15 Yaping Shou Methods for detecting lp-pla2 activity and inhibition of lp-pla2 activity
US8147867B2 (en) 2004-05-03 2012-04-03 Hermes Biosciences, Inc. Liposomes useful for drug delivery
US8658203B2 (en) 2004-05-03 2014-02-25 Merrimack Pharmaceuticals, Inc. Liposomes useful for drug delivery to the brain
ES2605792T3 (en) 2004-05-13 2017-03-16 Icos Corporation Quinazolinone used as a human phosphatidylinositol 3-kinase delta inhibitor
EP1750673B1 (en) * 2004-05-17 2009-12-02 Tekmira Pharmaceuticals Corporation Liposomal formulations comprising dihydrosphingomyelin and methods of use thereof
US20060292554A1 (en) * 2004-05-18 2006-12-28 Genentech, Inc. Major coat protein variants for C-terminal and bi-terminal display
US20050260260A1 (en) * 2004-05-19 2005-11-24 Edward Kisak Liposome compositions for the delivery of macromolecules
CA2566174A1 (en) * 2004-05-21 2005-12-01 Transave, Inc. Treatment of lung diseases and pre-lung disease conditions
WO2005115384A2 (en) * 2004-05-25 2005-12-08 Metabolex, Inc. Bicyclic, substituted triazoles as modulators of ppar and methods of their preparation
RU2006145872A (en) * 2004-05-25 2008-06-27 Метаболекс, Инк. (Us) SUBSTITUTED TRIAZOLES AS PPAR MODULATORS AND METHODS FOR PRODUCING THEM
CA2567883A1 (en) * 2004-05-25 2005-12-15 Icos Corporation Methods for treating and/or preventing aberrant proliferation of hematopoietic cells
ES2623362T3 (en) * 2004-05-28 2017-07-11 Human Biomolecular Research Institute Metabolically stable pain relievers and pain medications
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
EP1766052A4 (en) * 2004-06-03 2009-12-16 Isis Pharmaceuticals Inc Chimeric gapped oligomeric compositions
US20090048192A1 (en) * 2004-06-03 2009-02-19 Isis Pharmaceuticals, Inc. Double Strand Compositions Comprising Differentially Modified Strands for Use in Gene Modulation
BRPI0510915A (en) 2004-06-04 2007-11-13 Genentech Inc Method for treating multiple sclerosis and manufactured article
JP4796062B2 (en) * 2004-06-07 2011-10-19 プロチバ バイオセラピューティクス インコーポレイティッド Lipid-encapsulating interfering RNA
WO2005120152A2 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US8168635B2 (en) * 2004-06-19 2012-05-01 Human Biomolecular Research Institute Modulators of central nervous system neurotransmitters
US8415298B2 (en) 2004-06-21 2013-04-09 The Board Of Trustees Of The Leland Stanford Junior University Of Stanford Administration of FGF2 for treamtent of anxiety
WO2006014253A2 (en) * 2004-07-02 2006-02-09 Genentech, Inc. Factor viia variants
CA2572439A1 (en) 2004-07-02 2006-01-12 Protiva Biotherapeutics, Inc. Immunostimulatory sirna molecules and uses therefor
EP1773400A2 (en) * 2004-07-08 2007-04-18 Amgen Inc. Therapeutic peptides
WO2006006172A2 (en) * 2004-07-15 2006-01-19 Ramot At Tel Aviv University Ltd. Use of anti-amyloid agents for treating and typing pathogen infections
CA2565961A1 (en) 2004-07-15 2006-02-23 Xencor, Inc. Optimized fc variants
WO2006007712A1 (en) * 2004-07-19 2006-01-26 Protiva Biotherapeutics, Inc. Methods comprising polyethylene glycol-lipid conjugates for delivery of therapeutic agents
DK2626368T3 (en) * 2004-07-19 2017-02-27 Biocon Ltd INSULIN OLIGOMS CONJUGATES, FORMULATIONS AND APPLICATIONS THEREOF
DE602005019167D1 (en) 2004-07-20 2010-03-18 Genentech Inc ANGIOPOIETIN-LIKE 4 PROTEIN HEMMER COMBINATIONS AND THEIR USE
CA2570323C (en) 2004-07-26 2014-08-26 Genentech, Inc. Methods and compositions for modulating hepatocyte growth factor activation
KR20070040824A (en) * 2004-07-30 2007-04-17 리나트 뉴로사이언스 코퍼레이션 Antibodies directed against amyloid-beta peptide and methods using same
US9132116B2 (en) * 2004-08-02 2015-09-15 Willowcroft Pharm Inc. Mast cell stabilizers to prevent or treat laminitis
US8568637B2 (en) * 2004-08-02 2013-10-29 Ramot At Tel-Aviv University Ltd. Method of forming a fiber made of peptide nanostructures
AU2005277591B2 (en) 2004-08-17 2011-07-14 Covidien Lp Anti-adhesion barrier
JP2006056807A (en) * 2004-08-18 2006-03-02 Konica Minolta Medical & Graphic Inc Preparation for use in photodynamic therapy
ATE539745T1 (en) 2004-08-19 2012-01-15 Univ Tel Aviv Future Tech Dev COMPOSITIONS FOR TREATING AMYLOID-ASSOCIATED DISEASES
JP4715133B2 (en) * 2004-08-26 2011-07-06 コニカミノルタエムジー株式会社 Anti-tumor liposome preparation and production method thereof
JP2006069929A (en) * 2004-08-31 2006-03-16 Konica Minolta Medical & Graphic Inc Preparation for treating mycosis and method for producing the same
WO2006027780A2 (en) 2004-09-08 2006-03-16 Ramot At Tel Aviv University Ltd. Peptide nanostructures containing end-capping modified peptides and methods of generating and using the same
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
JP2008512444A (en) * 2004-09-09 2008-04-24 イッスム・リサーチ・ディベロップメント・カンパニー・オブ・ザ・ヘブルー・ユニバーシティ・オブ・エルサレム Liposome formulation containing amphiphilic weak base-like tempamine for the treatment of neurodegenerative conditions
AU2005281352B2 (en) * 2004-09-09 2011-01-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem Liposomal compositions of glucocorticoid and glucocorticoid derivatives
US6998028B1 (en) * 2004-09-24 2006-02-14 Superpower, Inc. Methods for forming superconducting conductors
EA011879B1 (en) * 2004-09-24 2009-06-30 Эмджин Инк. MODIFIED Fc MOLECULES
US8435948B2 (en) 2004-09-29 2013-05-07 Mount Sinai School Of Medicine Of New York University Methods for inhibiting osteoclastic bone resorption and bone loss comprising administration of an anti-FSH or anti-FSHR antibody
US20060110387A1 (en) 2004-10-05 2006-05-25 Genentech, Inc. Method for treating vasculitis
US9315862B2 (en) 2004-10-05 2016-04-19 California Institute Of Technology Aptamer regulated nucleic acids and uses thereof
WO2006042237A2 (en) * 2004-10-06 2006-04-20 Mayo Foundation For Medical Education And Research B7-h1 and methods of diagnosis, prognosis, and treatment of cancer
DE602005008270D1 (en) * 2004-10-08 2008-08-28 Alza Corp METHOD FOR INTRODUCING A LIPID-LINKED PART INTO A PRE-FORMED LIPID ASSEMBLY WITH MICROWAVES
TW200612993A (en) * 2004-10-08 2006-05-01 Alza Corp Lipopolymer conjugates
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
KR101287918B1 (en) * 2004-10-26 2013-07-19 오르토 바이오테크 프로덕츠 엘.피. Pegylated liposomal doxorubicin in combination with ecteinescidin 743
WO2006050327A2 (en) * 2004-10-28 2006-05-11 Alza Corporation Lyophilized liposome formulations and method
RS50510B (en) 2004-10-29 2010-03-02 Pharma Mar S.A., Sociedad Unipersonal FORMULATIONS CONTAINING ECTEINASCIDINE AND DISAHARID
TW200618820A (en) * 2004-11-05 2006-06-16 Alza Corp Liposome formulations of boronic acid compounds
AU2005304914B2 (en) 2004-11-05 2012-02-16 Tekmira Pharmaceuticals Corporation Compositions and methods for stabilizing liposomal camptothecin formulations
AU2005306802A1 (en) * 2004-11-08 2006-05-26 Transave, Inc. Methods of treating cancer with lipid-based platinum compound formulations administered intraperitoneally
EP1814910A4 (en) * 2004-11-11 2009-04-29 Affymax Inc Novel peptides that bind to the erythropoietin receptor
CA2587470A1 (en) * 2004-11-15 2006-05-18 Yissum Research Development Company Of The Hebrew University Of Jerusale M Combination therapy associating preferably a ceramide with a cytotoxic drug
CA2587411A1 (en) * 2004-11-17 2006-05-26 Protiva Biotherapeutics, Inc. Sirna silencing of apolipoprotein b
EP1813288B1 (en) * 2004-11-18 2018-07-18 Terumo Kabushiki Kaisha Medicinal composition, medicinal preparation, and combination preparation
WO2006066158A2 (en) * 2004-12-14 2006-06-22 Alnylam Pharmaceuticals, Inc. Rnai modulation of mll-af4 and uses thereof
EP1841793B1 (en) 2005-01-07 2010-03-31 Diadexus, Inc. Ovr110 antibody compositions and methods of use
US20080207505A1 (en) * 2005-01-12 2008-08-28 James Kenneth D Bna Conjugates and Methods of Use
EP1846030B1 (en) 2005-01-21 2018-11-21 Genentech, Inc. Fixed dosing of her antibodies
US9937127B2 (en) * 2005-01-28 2018-04-10 Kyowa Hakko Kirin Co., Ltd. Method of producing fine particles surface-modified with water-soluble substance
CA2596079A1 (en) 2005-01-31 2006-08-10 Vaxinnate Corporation Novel polypeptide ligands for toll-like receptor 2 (tlr2)
US8029783B2 (en) * 2005-02-02 2011-10-04 Genentech, Inc. DR5 antibodies and articles of manufacture containing same
CA2596506C (en) 2005-02-09 2021-04-06 Avi Biopharma, Inc. Antisense composition and method for treating muscle atrophy
JP5651285B2 (en) 2005-02-15 2015-01-07 デューク ユニバーシティ Anti-CD19 antibodies and use in oncology
WO2006089106A2 (en) * 2005-02-17 2006-08-24 Icos Corporation Phosphoinositide 3-kinase inhibitors for inhibiting leukocyte accumulation
WO2006088245A1 (en) * 2005-02-18 2006-08-24 The University Of Tokushima Polyoxyalkylene chain-containing lipid derivative and lipid film structure containing such derivative
US8329178B2 (en) 2005-02-18 2012-12-11 Dana-Farber Cancer Institute, Inc. Antibodies against CXCR4 and methods of use thereof
NZ590431A (en) 2005-02-23 2012-08-31 Genentech Inc Extending time to disease progression or survival in cancer patients using a HER dimerization inhibitor
TW200714289A (en) * 2005-02-28 2007-04-16 Genentech Inc Treatment of bone disorders
US20060204505A1 (en) * 2005-03-08 2006-09-14 Sliwkowski Mark X Methods for identifying tumors responsive to treatment with HER dimerization inhibitors (HDIs)
CN101175769A (en) 2005-03-10 2008-05-07 健泰科生物技术公司 Methods and compositions for modulating vascular integrity
JP2006248978A (en) * 2005-03-10 2006-09-21 Mebiopharm Co Ltd New liposome preparation
WO2006099445A2 (en) * 2005-03-14 2006-09-21 Massachusetts Institute Of Technology Nanocells for diagnosis and treatment of diseases and disorders
EP1865778A4 (en) 2005-03-21 2010-12-08 Metabolex Inc Methods for avoiding edema in the treatment of metabolic, inflammatory, and cardiovascular disorders
CN1840193B (en) * 2005-03-29 2010-05-12 中国科学院生物物理研究所 Nanometer capsule of anthracene nucleus anticancer antibiotic with polyethylene glycol-phospholipid
ATE541928T1 (en) 2005-03-31 2012-02-15 Calando Pharmaceuticals Inc RIBONUCLEOTIDE REDUCTASE SUBUNITY 2 INHIBITORS AND USES THEREOF
UY29504A1 (en) 2005-04-29 2006-10-31 Rinat Neuroscience Corp DIRECTED ANTIBODIES AGAINST BETA AMYLOID PEPTIDE AND METHODS USING THE SAME.
EP2221316A1 (en) 2005-05-05 2010-08-25 Duke University Anti-CD19 antibody therapy for autoimmune disease
WO2006122079A1 (en) 2005-05-06 2006-11-16 Zymogenetics, Inc. Il-31 monoclonal antibodies and methods of use
US8124084B2 (en) 2005-05-17 2012-02-28 University Of Connecticut Compositions and methods for immunomodulation in an organism using IL-15 and soluble IL-15Ra
US7919461B2 (en) * 2005-06-03 2011-04-05 Affymax, Inc. Erythropoietin receptor peptide formulations and uses
US7550433B2 (en) 2005-06-03 2009-06-23 Affymax, Inc. Erythropoietin receptor peptide formulations and uses
US8324159B2 (en) * 2005-06-03 2012-12-04 Affymax, Inc. Erythropoietin receptor peptide formulations and uses
NZ563341A (en) 2005-06-06 2009-10-30 Genentech Inc Methods for identifying agents that modulate a gene that encodes for a PRO1568 polypeptide
TW200726485A (en) * 2005-07-01 2007-07-16 Alza Corp Liposomal delivery vehicle for hydrophobic drugs
CN101282994B (en) * 2005-07-22 2013-09-18 Y's治疗有限公司 Anti-CD26 antibodies and methods of use thereof
JP5657862B2 (en) * 2005-07-28 2015-01-21 ノバルティス アーゲー Use of antibodies against M-CSF
WO2007016285A2 (en) * 2005-07-28 2007-02-08 Novartis Ag M-csf specific monoclonal antibody and uses thereof
US20070055200A1 (en) 2005-08-10 2007-03-08 Gilbert Scott J Needle-free jet injection drug delivery device
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
EP1995321A2 (en) 2005-08-15 2008-11-26 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
US20070054873A1 (en) * 2005-08-26 2007-03-08 Protiva Biotherapeutics, Inc. Glucocorticoid modulation of nucleic acid-mediated immune stimulation
WO2007043048A2 (en) * 2005-10-11 2007-04-19 Ramot At Tel Aviv University Ltd. Self-assembled fmoc-ff hydrogels
TW200732350A (en) 2005-10-21 2007-09-01 Amgen Inc Methods for generating monovalent IgG
US7875602B2 (en) * 2005-10-21 2011-01-25 Sutter West Bay Hospitals Camptothecin derivatives as chemoradiosensitizing agents
JP2009513708A (en) 2005-10-31 2009-04-02 オンコメッド ファーマシューティカルズ インコーポレイテッド Compositions and methods for diagnosis and treatment of cancer
GB0522082D0 (en) 2005-10-31 2005-12-07 Pharma Mar Sa Formulations
EP2395012B8 (en) 2005-11-02 2018-06-06 Arbutus Biopharma Corporation Modified siRNA molecules and uses thereof
US7879212B2 (en) * 2005-11-03 2011-02-01 Ramot At Tel-Aviv University Ltd. Peptide nanostructure-coated electrodes
DE102005053066A1 (en) * 2005-11-04 2007-05-10 Basf Ag Use of copolymers as solubilizers for sparingly water-soluble compounds
US9107824B2 (en) 2005-11-08 2015-08-18 Insmed Incorporated Methods of treating cancer with high potency lipid-based platinum compound formulations administered intraperitoneally
US20070190180A1 (en) * 2005-11-08 2007-08-16 Pilkiewicz Frank G Methods of treating cancer with high potency lipid-based platinum compound formulations administered intravenously
US20070190182A1 (en) * 2005-11-08 2007-08-16 Pilkiewicz Frank G Methods of treating cancer with high potency lipid-based platinum compound formulations administered intraperitoneally
US20070190181A1 (en) * 2005-11-08 2007-08-16 Pilkiewicz Frank G Methods of treating cancer with lipid-based platinum compound forumulations administered intravenously
EP2564864B8 (en) 2005-11-12 2015-05-13 The Board of Trustees of The Leland Stanford Junior University FGF2-related methods for diagnosing and treating depression
KR20080068062A (en) 2005-11-14 2008-07-22 리나트 뉴로사이언스 코퍼레이션 Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
AR056806A1 (en) 2005-11-14 2007-10-24 Amgen Inc RANKL- PTH / PTHRP ANTIBODY CHEMICAL MOLECULES
MY149159A (en) 2005-11-15 2013-07-31 Hoffmann La Roche Method for treating joint damage
AP2809A (en) 2005-11-18 2013-12-31 Glenmark Pharmaceuticals Sa Anti-Alpha2 Integrin antibodies and their uses
EP1966377A2 (en) 2005-11-21 2008-09-10 Isis Pharmaceuticals, Inc. Modulation of eif4e-bp2 expression
US20090293137A1 (en) 2005-11-21 2009-11-26 Genentech, Inc. Novel Gene Disruptions, Compositions and Methods Relating Thereto
US20070122350A1 (en) * 2005-11-30 2007-05-31 Transave, Inc. Safe and effective methods of administering therapeutic agents
WO2007126439A2 (en) 2005-12-02 2007-11-08 Genentech, Inc. Compositions and methods for the treatment of diseases and disorders associated with cytokine signaling involving antibodies that bind to il-22 and il-22r
US8466263B2 (en) 2005-12-02 2013-06-18 Dana-Farber Cancer Institute, Inc. Carbonic anhydrase IX (G250) anitbodies
CN101365424A (en) * 2005-12-08 2009-02-11 耶路撒冷希伯来大学伊森姆研究发展公司 Methods for affecting liposome composition by ultrasound irradiation
US20080300320A1 (en) * 2005-12-09 2008-12-04 Basf Se Use of Polyvinyl Lactam-Polyalkylene Block Copolymers as Solubilisers for Poorly Water-Soluble Compounds
US9149543B2 (en) 2005-12-15 2015-10-06 The Trustees Of The University Of Pennsylvania Methods and models for rapid, widespread delivery of genetic material to the CNS using non-viral, cationic lipid-mediated vectors
WO2007082144A2 (en) * 2006-01-05 2007-07-19 Mayo Foundation For Medical Education And Research B7-h1 and survivin in cancer
WO2007082154A2 (en) * 2006-01-05 2007-07-19 Mayo Foundation For Medical Education And Research B7-h1 and b7-h4 in cancer
CA2636149A1 (en) 2006-01-05 2007-07-19 Novartis Ag Methods for preventing and treating cancer metastasis and bone loss associated with cancer metastasis
PT2161038E (en) 2006-01-26 2014-03-10 Isis Pharmaceuticals Inc Compositions and their uses directed to huntingtin
CA2635011A1 (en) 2006-01-26 2007-11-22 Recopharma Ab Use of sialylated fusion polypeptides for inhibition of oculotropic viruses
US7829097B2 (en) * 2006-02-06 2010-11-09 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Use of HMGB1 for protection against ischemia reperfusion injury
ZA200807714B (en) 2006-02-17 2010-01-27 Genentech Inc Gene disruptions, compositions and methods relating thereto
WO2007103469A2 (en) 2006-03-06 2007-09-13 Aeres Biomedical Ltd. Humanized anti-cd22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
US20070218116A1 (en) * 2006-03-14 2007-09-20 Schwendener Reto A Compositions and methods for the treatment of tumors and tumor metastases
US9119782B2 (en) * 2006-03-20 2015-09-01 Mary P. McCourt Drug delivery means
UA100969C2 (en) 2006-03-21 2013-02-25 Дженентек, Інк. Combinatorial therapy involving alpha5beta1 antagonists
EP2389946A1 (en) 2006-03-23 2011-11-30 Novartis AG Anti-tumor cell antigen antibody therapeutics
CA2647277A1 (en) 2006-04-05 2007-11-08 Genentech, Inc. Method for using boc/cdo to modulate hedgehog signaling
US8758723B2 (en) 2006-04-19 2014-06-24 The Board Of Regents Of The University Of Texas System Compositions and methods for cellular imaging and therapy
EP2082645A1 (en) 2006-04-19 2009-07-29 Genentech, Inc. Novel gene disruptions, compositions and methods relating thereto
WO2007124361A2 (en) * 2006-04-20 2007-11-01 Mayo Foundation For Medical Education And Research Soluble b7-h1
EP2881468B1 (en) 2006-04-21 2017-04-12 Intervet International B.V. Pestivirus species
CA2656990A1 (en) * 2006-04-28 2007-11-08 University Of South Florida Materials and methods for reducing inflammation by inhibition of the atrial natriuretic peptide receptor
WO2007127428A2 (en) * 2006-04-28 2007-11-08 University Of Florida Research Foundation, Inc. Double-stranded/self-complementary vectors with a truncated cba promoter and methods of gene delivery
US8697120B2 (en) * 2006-05-01 2014-04-15 Johns Hopkins University Method and use of nano-scale devices for reduction of tissue injury in ischemic and reperfusion injury
AU2007245599B2 (en) * 2006-05-03 2012-05-10 Baltic Technology Development, Ltd. Antisense agents combining strongly bound base - modified oligonucleotide and artificial nuclease
US20070264322A1 (en) * 2006-05-10 2007-11-15 Huang Ken S Method for making liposomes conjugated with temperature-sensitive ligands
JP5282031B2 (en) * 2006-05-15 2013-09-04 ドミトリィ ビー カーポティン Magnetic fine particles containing organic matter
EP2024393A2 (en) 2006-05-15 2009-02-18 Sea Lane Biotechnologies,llc. Neutralizing antibodies to influenza viruses
US8106013B2 (en) * 2006-05-19 2012-01-31 Georgia Tech Research Corporation ABC transporter ligand GATX1
WO2007137301A2 (en) * 2006-05-23 2007-11-29 Isis Pharmaceuticals, Inc. Modulation of chrebp expression
US7862812B2 (en) 2006-05-31 2011-01-04 Lpath, Inc. Methods for decreasing immune response and treating immune conditions
US7915399B2 (en) * 2006-06-09 2011-03-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
KR101376634B1 (en) 2006-06-19 2014-03-27 더 존스 홉킨스 유니버시티 Tumor-specific delivery of therapeutic agents via liposomase
US7981425B2 (en) 2006-06-19 2011-07-19 Amgen Inc. Thrombopoietic compounds
EP2505209A1 (en) 2006-06-26 2012-10-03 MacroGenics, Inc. Fcgamma-RIIB-specific antibodies and methods of the use thereof
CA2654661C (en) 2006-06-26 2012-10-23 Amgen Inc. Compositions comprising modified lcat and use thereof for treating atherosclerosis
JP5072275B2 (en) * 2006-07-03 2012-11-14 テルモ株式会社 Method for separating closed vesicles, method for producing preparation and evaluation method
EP2037737B1 (en) 2006-07-11 2014-04-02 University Of Medicine And Dentistry Of New Jersey Cell membrane repair proteins, nucleic acids encoding the same and associated methods of use
US8343539B2 (en) 2006-07-14 2013-01-01 Regents Of The University Of Minnesota Compounds that bind α5β1 integrin and methods of use
WO2008008873A2 (en) * 2006-07-14 2008-01-17 Georgia Tech Research Corporation Clc channel ligand
DK2046809T3 (en) 2006-07-19 2017-03-13 Univ Pennsylvania WSX-1 / IL-27 AS A TARGET OBJECTIVE FOR ANTI-INFLAMMATORY REACTIONS
US8198253B2 (en) 2006-07-19 2012-06-12 Isis Pharmaceuticals, Inc. Compositions and their uses directed to HBXIP
JP4936312B2 (en) * 2006-07-20 2012-05-23 株式会社島津製作所 Novel amphiphile, drug delivery system and molecular imaging system using the same
WO2008013918A2 (en) * 2006-07-26 2008-01-31 Myelin Repair Foundation, Inc. Cell cycle regulation and differentiation
EP2420252A1 (en) 2006-08-04 2012-02-22 Novartis AG EPHB3-specific antibody and uses thereof
EP2057465A4 (en) 2006-08-09 2010-04-21 Homestead Clinical Corp Organ-specific proteins and methods of their use
ME01786B (en) 2006-08-14 2014-09-20 Xencor Inc Optimized antibodies that target cd19
PE20081214A1 (en) 2006-08-18 2008-09-24 Novartis Ag PRLR SPECIFIC ANTIBODY AND ITS USES
JP2010501172A (en) 2006-08-25 2010-01-21 オンコセラピー・サイエンス株式会社 Prognostic markers and therapeutic targets for lung cancer
MY162024A (en) 2006-08-28 2017-05-31 La Jolla Inst Allergy & Immunology Antagonistic human light-specific human monoclonal antibodies
AU2007290277B2 (en) 2006-08-29 2013-04-11 Genentech, Inc. Use of tenecteplase for treating acute ischemic stroke
SI2594586T1 (en) 2006-09-01 2014-11-28 Zymogenetics, Inc. IL-31 monoclonal antibodies and methods of use
CA2600220C (en) * 2006-09-07 2014-12-09 Canadian Blood Services Surface cross-linked lipidic particles, methods of production and uses therefor
DK2066694T3 (en) 2006-09-29 2016-02-08 Oncomed Pharm Inc Compositions and Methods for Diagnosing and Treating Cancer
CN101557818A (en) 2006-10-03 2009-10-14 新泽西医科和牙科大学 ATAP peptides, nucleic acids encoding the same and associated methods of use
US10925977B2 (en) 2006-10-05 2021-02-23 Ceil>Point, LLC Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
AR063384A1 (en) 2006-10-25 2009-01-28 Amgen Inc THERAPEUTIC AGENTS BASED ON PEPTIDES DERIVED FROM TOXINS
CN101679506B (en) * 2006-10-27 2015-02-25 勒帕斯公司 Compositions and methods for binding sphingosine-1-phosphate
WO2008055072A2 (en) 2006-10-27 2008-05-08 Lpath, Inc. Compositions and methods for treating ocular diseases and conditions
US8158595B2 (en) * 2006-11-09 2012-04-17 California Institute Of Technology Modular aptamer-regulated ribozymes
WO2008055314A1 (en) 2006-11-10 2008-05-15 Dimerix Bioscience Pty Ltd Thyrotropin releasing hormone receptor-orexin receptor hetero-dimers/-oligomers
WO2008064018A1 (en) 2006-11-13 2008-05-29 Eli Lilly & Co. Thienopyrimidinones for treatment of inflammatory disorders and cancers
JP5391073B2 (en) 2006-11-27 2014-01-15 ディアデクサス インコーポレーテッド Ovr110 antibody compositions and methods of use
ES2435437T3 (en) 2006-12-07 2013-12-19 Novartis Ag Antagonist Antibodies Against Ephb3
EP1932517A3 (en) * 2006-12-11 2008-07-16 Universiteit Utrecht Holding B.V. Liposomes containing a polyphenol derivative such as caffeic acid and a method of post-loading thereof
EP2114413B1 (en) * 2006-12-18 2014-10-22 The Johns Hopkins University Therapeutics for treating cancer using 3-bromopyruvate
US8536113B2 (en) * 2006-12-21 2013-09-17 Janssen Biotech, Inc. EGFR binding peptides and uses thereof
WO2008079982A2 (en) 2006-12-21 2008-07-03 Centocor, Inc. Liposome composition for targeting egfr receptor
WO2008079976A2 (en) 2006-12-21 2008-07-03 Centocor, Inc. Dimeric high affinity egfr constructs and uses thereof
EP2913341A1 (en) 2006-12-22 2015-09-02 University of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
WO2008083169A2 (en) * 2006-12-26 2008-07-10 The Johns Hopkins University Compositions and methods for the treatment of immunologic disorders
AU2007339762B2 (en) * 2006-12-27 2011-03-31 The Johns Hopkins University Compositions and methods for treating inflammation and auto-immune diseases
US7989173B2 (en) 2006-12-27 2011-08-02 The Johns Hopkins University Detection and diagnosis of inflammatory disorders
US20090142342A1 (en) * 2006-12-27 2009-06-04 Johns Hopkins University B7-h4 receptor agonist compositions and methods for treating inflammation and auto-immune diseases
US7638541B2 (en) 2006-12-28 2009-12-29 Metabolex Inc. 5-ethyl-2-{4-[4-(4-tetrazol-1-yl-phenoxymethyl)-thiazol-2-yl]-piperidin-1-yl}-pyrimidine
WO2008091655A2 (en) * 2007-01-23 2008-07-31 The Regents Of The University Of California Methods, compositions and device for directed and controlled heating and release of agents
EP2641971A1 (en) 2007-01-29 2013-09-25 Isis Pharmaceuticals, Inc. Compounds and methods for modulating protein expression
WO2008094275A1 (en) 2007-01-30 2008-08-07 New York University Peptides for treatment of conditions associated with nitric oxide
KR101508397B1 (en) 2007-02-22 2015-04-08 제넨테크, 인크. Methods for detecting inflammatory bowel disease
PE20090681A1 (en) 2007-03-02 2009-06-10 Genentech Inc PREDICTION OF RESPONSE TO A HER INHIBITOR
CA2681302C (en) * 2007-03-19 2013-07-23 Dhiraj Khattar Proliposomal and liposomal compositions of poorly water-soluble compounds
JP4510842B2 (en) * 2007-03-26 2010-07-28 キヤノン株式会社 Polyhydroxyalkanoate-coated liposome
SG194368A1 (en) 2007-05-04 2013-11-29 Technophage Investigacao E Desenvolvimento Em Biotecnologia Sa Engineered rabbit antibody variable domains and uses thereof
EP2703011A3 (en) 2007-05-07 2014-03-26 MedImmune, LLC Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
US20090175784A1 (en) * 2007-05-11 2009-07-09 Joshua Goldstein Anti-Alpha V Immunoliposome Composition, Methods, and Uses
US20090196913A1 (en) * 2007-05-11 2009-08-06 Ken Shi Kun Huang Anti-Alpha-V Immunoliposome Composition, Methods, and Uses
US20090163698A1 (en) * 2007-05-11 2009-06-25 John Joseph Grigsby Method for Preparing Antibody Conjugates
JP2010526868A (en) 2007-05-14 2010-08-05 ノビミューン エスアー Fc receptor binding polypeptide having altered effector function
EP2160200B1 (en) 2007-05-14 2013-07-10 The University of Chicago Antibody-LIGHT fusion products as cancer therapeutics
MX2009012609A (en) 2007-05-22 2009-12-07 Amgen Inc Compositions and methods for producing bioactive fusion proteins.
WO2008148023A2 (en) * 2007-05-23 2008-12-04 Medical College Of Georgia Research Institute, Inc. Compositions and methods for treating neurological disorders
EP2164875A4 (en) 2007-05-24 2011-11-30 Us Dept Veterans Affairs Intranuclear protein transduction through a nucleoside salvage pathway
EP3392273A1 (en) 2007-05-30 2018-10-24 Xencor, Inc. Methods and compositions for inhibiting cd32b expressing cells
CA2724432A1 (en) * 2007-05-30 2008-12-11 Lpath, Inc. Compositions and methods for binding lysophosphatidic acid
US9163091B2 (en) * 2007-05-30 2015-10-20 Lpath, Inc. Compositions and methods for binding lysophosphatidic acid
TW200916094A (en) * 2007-06-27 2009-04-16 Poniard Pharmaceuticals Inc Stabilized picoplatin dosage form
US8153595B2 (en) * 2007-07-13 2012-04-10 The Johns Hopkins University B7-DC variants immunogenic compositions and methods of use thereof
NZ602024A (en) 2007-07-16 2014-04-30 Genentech Inc Anti-cd79b antibodies and immunoconjugates and methods of use
WO2009011855A2 (en) * 2007-07-16 2009-01-22 California Institute Of Technology Selection of nucleic acid-based sensor domains within nucleic acid switch platform
JP2010533495A (en) 2007-07-16 2010-10-28 ジェネンテック, インコーポレイテッド Humanized CD79b antibodies and immunoconjugates and their use
CN101754962B (en) * 2007-07-19 2013-12-25 赛马拜制药公司 N-azacyclic substituted pyrrole, imidazole, triazole and tetrazole derivatives as agonists of RUP3 or GPR119 for treatment of diabetes and metabolic disorders
EA201070187A1 (en) 2007-07-26 2010-08-30 Эмджен Инк. MODIFIED ENZYMES LECITHIN-CHOLESTEROL ACYLTRANSFERASE
JP2010534730A (en) * 2007-07-26 2010-11-11 ビーエーエスエフ ソシエタス・ヨーロピア Process for the preparation of solid-form copolymers based on polyethers obtained by graft polymerization in solution
US7666973B2 (en) 2007-07-30 2010-02-23 Tyco Healthcare Group Lp Carbonate copolymers
WO2009017833A2 (en) 2007-08-02 2009-02-05 Arresto Biosciences Methods and compositions for treatment and diagnosis of fibrosis, tumor invasion, angiogenesis, and metastasis
WO2009020093A1 (en) * 2007-08-09 2009-02-12 Daiichi Sankyo Company, Limited Immunoliposome inducing apoptosis into cell expressing death domain-containing receptor
US20090048423A1 (en) * 2007-08-15 2009-02-19 Tyco Healthcare Group Lp Phospholipid Copolymers
US8268958B2 (en) 2007-08-15 2012-09-18 Tyco Healthcare Group Ip Phospholipid copolymers
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
US20100179106A1 (en) * 2007-09-07 2010-07-15 Gencia Corporation Mitochondrial compositions and uses thereof
US8865667B2 (en) * 2007-09-12 2014-10-21 California Institute Of Technology Higher-order cellular information processing devices
US20110014118A1 (en) * 2007-09-21 2011-01-20 Lawrence Tamarkin Nanotherapeutic colloidal metal compositions and methods
US20090104114A1 (en) * 2007-09-21 2009-04-23 Cytimmune Sciences, Inc. Nanotherapeutic Colloidal Metal Compositions and Methods
CN101827613A (en) 2007-09-27 2010-09-08 免疫疫苗技术有限公司 Use of liposomes in a carrier comprising a continuous hydrophobic phase for delivery of polynucleotides in vivo
US20100239652A1 (en) * 2007-09-28 2010-09-23 Universitatsspital Basel Immunoliposomes for treatment of cancer
CA2700953A1 (en) 2007-10-02 2009-04-09 Amgen Inc. Increasing erythropoietin using nucleic acids hybridizable to micro-rna and precursors thereof
US20100209452A1 (en) * 2007-10-03 2010-08-19 Immunovaccine Technologies, Inc Compositions comprising an antigen, an amphipathic compound and a hydrophobic carrier, and uses thereof
JP5868594B2 (en) * 2007-10-16 2016-02-24 バイオコン・リミテッドBiocon Limited Orally administrable solid pharmaceutical composition and process thereof
US8361465B2 (en) * 2007-10-26 2013-01-29 Lpath, Inc. Use of anti-sphingosine-1-phosphate antibodies in combination with chemotherapeutic agents
RU2530561C2 (en) 2007-11-05 2014-10-10 Медиммун, Ллк Method of treating scleroderma
US20110152346A1 (en) * 2007-11-05 2011-06-23 Baltic Technology Development Ltd. Use of Oligonucleotides with Modified Bases in Hybridization of Nucleic Acids
RU2503460C2 (en) 2007-11-07 2014-01-10 Дженентек Инк. Using antimicrobial polypeptide for treating microbial disorders
US20110033476A1 (en) * 2007-11-12 2011-02-10 Theraclone Sciences Inc. Compositions and methods for the therapy and diagnosis of influenza
CN104059143A (en) * 2007-11-12 2014-09-24 特罗科隆科学有限公司 Compositions and methods for the therapy and diagnosis of influenza
US8598165B2 (en) 2007-11-26 2013-12-03 University Of Kansas Morpholines as selective inhibitors of cytochrome P450 2A13
MX2010006148A (en) 2007-12-06 2011-02-23 Dana Farber Cancer Inst Inc Antibodies against influenza virus and methods of use thereof.
HUE036548T2 (en) 2007-12-07 2018-08-28 Zymogenetics Inc Humanized antibody molecules specific for IL-31
US9029524B2 (en) * 2007-12-10 2015-05-12 California Institute Of Technology Signal activated RNA interference
RU2570559C2 (en) 2007-12-17 2015-12-10 Пфайзер Лимитед Treatment of interstitial cystitis
WO2009086514A1 (en) 2007-12-28 2009-07-09 Dana-Farber Cancer Institute, Inc. Humanized monoclonal antibodies and methods of use
EP3100718B1 (en) 2008-01-02 2019-11-27 Arbutus Biopharma Corporation Improved compositions and methods for the delivery of nucleic acids
US20090181094A1 (en) * 2008-01-15 2009-07-16 Eric Yueh-Lang Sheu Molecular Cage for Sustained Release Control of Pharmaceutical and Cosmetic Agents
AR070141A1 (en) * 2008-01-23 2010-03-17 Glenmark Pharmaceuticals Sa SPECIFIC HUMANIZED ANTIBODIES FOR VON WILLEBRAND FACTOR
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
NZ587132A (en) 2008-01-31 2012-10-26 Genentech Inc Anti-cd79b antibodies and immunoconjugates and methods of use
US20110020325A1 (en) * 2008-02-29 2011-01-27 Kwon Eugene D Methods for reducing granulomatous inflammation
JP5544309B2 (en) * 2008-03-10 2014-07-09 セラクローン サイエンシーズ, インコーポレイテッド Compositions and methods for treatment and diagnosis of cytomegalovirus infection
WO2009121004A2 (en) 2008-03-28 2009-10-01 Sea Lane Biotechnologies, Llc Neutralizing molecules to viral antigens
JP5711655B2 (en) * 2008-03-31 2015-05-07 シマベイ セラピューティクス, インコーポレーテッド Oxymethylene aryl compounds and uses thereof
EP2283133A2 (en) 2008-04-04 2011-02-16 Calando Pharmaceuticals, Inc. Compositions and use of epas1 inhibitors
MX364200B (en) 2008-04-09 2019-04-16 Genentech Inc Novel compositions and methods for the treatment of immune related diseases.
CA2718975A1 (en) 2008-04-10 2009-10-15 Cell Signaling Technology, Inc. Compositions and methods for detecting egfr mutations in cancer
WO2009127060A1 (en) 2008-04-15 2009-10-22 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
WO2009134917A2 (en) * 2008-04-29 2009-11-05 Wyeth Methods for treating inflammation
WO2009136298A2 (en) * 2008-05-09 2009-11-12 Recopharma Ab Compositions and methods for inhibiting shiga toxin and shiga-like toxin
US20090280134A1 (en) * 2008-05-09 2009-11-12 Recopharma Ab Compositions and methods for inhibiting toxin a from clostridium difficile
SI2279004T1 (en) 2008-05-16 2015-05-29 F. Hoffmann-La Roche Ag Use of biomarkers for assessing treatment of gastrointestinal inflammatory disorders with beta7integrin antagonists
US8093018B2 (en) 2008-05-20 2012-01-10 Otsuka Pharmaceutical Co., Ltd. Antibody identifying an antigen-bound antibody and an antigen-unbound antibody, and method for preparing the same
WO2009140853A1 (en) * 2008-05-23 2009-11-26 The University Of Hong Kong Combination therapy for the treatment of influenza
EP2305214B1 (en) 2008-06-05 2018-01-03 Shimadzu Corporation Novel molecular assembly, molecular probe for molecular imaging and molecular probe for drug delivery system using the same, and molecular imaging system and drug delivery system
WO2009146523A1 (en) 2008-06-05 2009-12-10 Immunovaccine Technologies Inc. Compositions comprising liposomes, an antigen, a polynucleotide and a carrier comprising a continuous phase of a hydrophobic substance
US8173621B2 (en) 2008-06-11 2012-05-08 Gilead Pharmasset Llc Nucleoside cyclicphosphates
WO2009150623A1 (en) 2008-06-13 2009-12-17 Pfizer Inc Treatment of chronic prostatitis
US20110104074A1 (en) * 2008-06-18 2011-05-05 University Of Louisville Research Foundation, Inc. Methods for targeted cancer treatment and detection
JP5560270B2 (en) 2008-07-08 2014-07-23 オンコメッド ファーマシューティカルズ インコーポレイテッド NOTCH binding agents and antagonists and methods of use thereof
US8883211B2 (en) 2008-07-10 2014-11-11 Serina Therapeutics, Inc. Polyoxazolines with inert terminating groups, polyoxazolines prepared from protected initiating groups and related compounds
US20100008900A1 (en) * 2008-07-14 2010-01-14 The University Of Hong Kong Annexin ii compositions for treating or monitoring inflammation or immune-mediated disorders
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
EP2323667A4 (en) * 2008-08-07 2012-07-25 Isis Pharmaceuticals Inc Modulation of transthyretin expression for the treatment of cns related disorders
US8652843B2 (en) 2008-08-12 2014-02-18 Oncomed Pharmaceuticals, Inc. DDR1-binding agents and methods of use thereof
CN102186820B (en) 2008-08-15 2013-08-28 乔治城大学 Fluorescent regulators of rassf1a expression and human cancer cell proliferation
ES2464729T3 (en) 2008-08-21 2014-06-03 The Johns Hopkins University Methods and compositions for the administration of 3-halopyruvate and related compounds for the treatment of cancer
MY150599A (en) 2008-08-22 2014-01-30 Sanofi Aventis [4-(5-aminomethyl-2-fluoro-phenyl)-piperidin-1-yl]-[7-fluoro-1-(2-methoxy-ethyl)-4-trifluoromethoxy-1h-indol-3-yl]-methanone as an inhibitor of mast cell tryptase
WO2010098788A2 (en) * 2008-08-25 2010-09-02 Amplimmune, Inc. Pd-i antagonists and methods for treating infectious disease
NZ591130A (en) 2008-08-25 2012-09-28 Amplimmune Inc Compositions comprising a PD-1 antagonists and cyclophosphamide and methods of use thereof
EP3301116A1 (en) 2008-08-25 2018-04-04 Dana Farber Cancer Institute, Inc. Conserved influenza hemagglutinin epitope and antibodies thereto
BRPI0913476A2 (en) 2008-09-10 2015-12-01 Genentech Inc "Uses of a tspan12 antagonist and Uses of a norrin antagonist"
TWI445716B (en) 2008-09-12 2014-07-21 Rinat Neuroscience Corp Pcsk9 antagonists
TW201438738A (en) 2008-09-16 2014-10-16 Genentech Inc Methods for treating progressive multiple sclerosis
CN102405286A (en) 2008-09-22 2012-04-04 阿克赛医药公司 Reduced size self-delivering rnai compounds
CA2740125A1 (en) 2008-10-09 2010-04-15 Northeastern University Multifunctional self-assembling polymeric nanosystems
EP2350043B9 (en) 2008-10-09 2014-08-20 TEKMIRA Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
AU2009307656B2 (en) * 2008-10-21 2015-07-02 Cymabay Therapeutics, Inc. Aryl GPR120 receptor agonists and uses thereof
WO2010046778A2 (en) * 2008-10-21 2010-04-29 International Vaccine Institute Novel shigella frotein antigens and methods
US8871202B2 (en) 2008-10-24 2014-10-28 Lpath, Inc. Prevention and treatment of pain using antibodies to sphingosine-1-phosphate
JP5775458B2 (en) * 2008-11-06 2015-09-09 グレンマーク ファーマシューティカルズ, エセ.アー. Treatment using anti-α2 integrin antibody
KR20230147211A (en) 2008-11-10 2023-10-20 알닐람 파마슈티칼스 인코포레이티드 Novel lipids and compositions for the delivery of therapeutics
US9492449B2 (en) 2008-11-13 2016-11-15 Gilead Calistoga Llc Therapies for hematologic malignancies
CN104042618B (en) 2008-11-13 2018-02-16 吉利德卡利斯托加公司 The treatment of malignant hematologic disease
DK2752189T3 (en) 2008-11-22 2017-01-16 Hoffmann La Roche APPLICATION OF ANTI-VEGF ANTIBODY IN COMBINATION WITH CHEMOTHERY TO TREAT CANCER CANCER
US20110230399A1 (en) * 2008-11-25 2011-09-22 Bowen Richard L Methods for Treating Obesity Related Disease
US20110160222A1 (en) * 2008-11-26 2011-06-30 Metabolex, Inc. Modulators of glucose homeostasis for the treatment of diabetes and metabolic disorders
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
MX358603B (en) 2008-12-04 2018-08-28 Curna Inc Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene.
US20110237649A1 (en) 2008-12-04 2011-09-29 Opko Curna, Llc Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirtuin 1
US8628762B2 (en) 2008-12-10 2014-01-14 Icahn School Of Medicine At Mount Sinai T-helper cell type 17 lineage-specific adjuvants, compositions and methods
AR074776A1 (en) 2008-12-18 2011-02-09 Sanofi Aventis METHOD TO TREAT MACULAR DEGENERATION; MODULATING THE PATIENT'S IMMUNE SYSTEM
AR074760A1 (en) * 2008-12-18 2011-02-09 Metabolex Inc GPR120 RECEIVER AGONISTS AND USES OF THE SAME IN MEDICINES FOR THE TREATMENT OF DIABETES AND METABOLIC SYNDROME.
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
NZ593648A (en) * 2008-12-23 2013-09-27 Gilead Pharmasset Llc Nucleoside phosphoramidates
AU2009329872B2 (en) 2008-12-23 2016-07-07 Gilead Pharmasset Llc Synthesis of purine nucleosides
PA8855701A1 (en) 2008-12-23 2010-07-27 NUCLEOSID ANALOGS
WO2010078536A1 (en) 2009-01-05 2010-07-08 Rxi Pharmaceuticals Corporation Inhibition of pcsk9 through rnai
CA3036963A1 (en) 2009-01-29 2010-08-05 Arbutus Biopharma Corporation Lipid formulations comprising cationic lipid and a targeting lipid comprising n-acetyl galactosamine for delivery of nucleic acid
WO2010086828A2 (en) 2009-02-02 2010-08-05 Rinat Neuroscience Corporation Agonist anti-trkb monoclonal antibodies
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
CN102439149B (en) 2009-02-12 2018-01-02 库尔纳公司 By suppressing to treat the related diseases of GDNF for the natural antisense transcript of the glial derived neurotrophic factor (GDNF)
PL2396038T3 (en) 2009-02-12 2016-05-31 Curna Inc Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
US8329882B2 (en) 2009-02-18 2012-12-11 California Institute Of Technology Genetic control of mammalian cells with synthetic RNA regulatory systems
US8975389B2 (en) 2009-03-02 2015-03-10 Alnylam Pharmaceuticals, Inc. Nucleic acid chemical modifications
EP2403946A4 (en) 2009-03-04 2012-11-14 Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirt 1
EP2228059A1 (en) 2009-03-12 2010-09-15 Universitätsspital Basel Chemotherapeutic composition for the treatment of cancer
EP2408919B1 (en) 2009-03-16 2017-10-18 CuRNA, Inc. Treatment of nuclear factor (erythroid-derived 2)-like 2 (nrf2) related diseases by inhibition of natural antisense transcript to nrf2
SI3260136T1 (en) 2009-03-17 2021-05-31 Theraclone Sciences, Inc. Human immunodeficiency virus (hiv) -neutralizing antibodies
US9708604B2 (en) 2009-03-17 2017-07-18 Curna, Inc. Treatment of delta-like 1 homolog (DLK1) related diseases by inhibition of natural antisense transcript to DLK1
MX2011009955A (en) 2009-03-24 2011-11-18 Gilead Calistoga Llc Atropisomers of2-purinyl-3-tolyl-quinazolinone derivatives and methods of use.
KR101523127B1 (en) 2009-03-25 2015-05-26 제넨테크, 인크. Novel anti-alpha5beta1 antibodies and uses thereof
EP2414399A1 (en) 2009-04-01 2012-02-08 F. Hoffmann-La Roche AG Anti-fcrh5 antibodies and immunoconjugates and methods of use
KR101766927B1 (en) 2009-04-01 2017-08-09 제넨테크, 인크. Treatment of insulin-resistant disorders
US9145555B2 (en) 2009-04-02 2015-09-29 California Institute Of Technology Integrated—ligand-responsive microRNAs
AU2010239312A1 (en) * 2009-04-20 2011-11-10 Gilead Calistoga Llc Methods of treatment for solid tumors
EP3524275A1 (en) 2009-04-22 2019-08-14 Massachusetts Institute Of Technology Innate immune supression enables repeated delivery of long rna molecules
US8609101B2 (en) 2009-04-23 2013-12-17 Theraclone Sciences, Inc. Granulocyte-macrophage colony-stimulating factor (GM-CSF) neutralizing antibodies
US9067986B2 (en) 2009-04-27 2015-06-30 Oncomed Pharmaceuticals, Inc. Method for making heteromultimeric molecules
EP2248903A1 (en) 2009-04-29 2010-11-10 Universitat Autònoma De Barcelona Methods and reagents for efficient and targeted gene transfer to monocytes and macrophages
US8524784B2 (en) * 2009-04-30 2013-09-03 Intezyne Technologies, Incorporated Polymer micelles containing anthracylines for the treatment of cancer
EP2424359A4 (en) 2009-04-30 2014-01-15 Intezyne Technologies Inc Polymer micelles containing anthracylines for the treatment of cancer
WO2010127195A2 (en) 2009-05-01 2010-11-04 Curna, Inc. Antisense oligonucleotides of hemoglobins
SG175276A1 (en) 2009-05-05 2011-11-28 Novimmune Sa Anti-il-17f antibodies and methods of use thereof
WO2010129687A1 (en) 2009-05-05 2010-11-11 Alnylam Pharmaceuticals, Inc Methods of delivering oligonucleotides to immune cells
CA3151387A1 (en) 2009-05-05 2010-11-11 Arbutus Biopharma Corporation Lipid compositions for the delivery of therapeutic agents
US20120046236A1 (en) 2009-05-06 2012-02-23 Opko Curna Llc Treatment of tristetraproline (ttp) related diseases by inhibition of natural antisense transcript to ttp
CN106237345A (en) 2009-05-06 2016-12-21 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
JP5922017B2 (en) 2009-05-18 2016-05-24 クルナ・インコーポレーテッド Treatment of reprogramming factor-related diseases by suppression of natural antisense transcripts against the reprogramming factor
US8858948B2 (en) 2009-05-20 2014-10-14 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
WO2011019423A2 (en) 2009-05-20 2011-02-17 Schering Corporation Modulation of pilr receptors to treat microbial infections
TWI598358B (en) 2009-05-20 2017-09-11 基利法瑪席特有限責任公司 Nucleoside phosphoramidates
JP2012527248A (en) 2009-05-22 2012-11-08 クルナ・インコーポレーテッド Treatment of TFE3 and insulin receptor substrate 2 (IRS2) related diseases by inhibition of natural antisense transcripts against transcription factor E3 (TFE3)
JP5960049B2 (en) 2009-05-28 2016-08-02 クルナ・インコーポレーテッド Treatment of antiviral gene-related diseases by suppression of natural antisense transcripts against antiviral genes
NZ622843A (en) 2009-06-10 2015-10-30 Tekmira Pharmaceuticals Corp Improved lipid formulation
WO2010143193A1 (en) 2009-06-11 2010-12-16 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Targeted liposomes comprising n-containing bisphosphonates and uses thereof
CN102612560B (en) 2009-06-16 2017-10-17 库尔纳公司 By suppressing to treat the related diseases of PON1 for the natural antisense transcript of PON1 (PON1)
US20120171170A1 (en) 2009-06-16 2012-07-05 Opko Curna, Llc Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
WO2010146511A1 (en) 2009-06-17 2010-12-23 Pfizer Limited Treatment of overactive bladder
CA2765889A1 (en) 2009-06-24 2010-12-29 Opko Curna, Llc Treatment of tumor necrosis factor receptor 2 (tnfr2) related diseases by inhibition of natural antisense transcript to tnfr2
KR101829073B1 (en) 2009-06-26 2018-02-13 아이2 파마슈티컬스, 인크. Expression of surrogate light chains
CA2765815A1 (en) 2009-06-26 2010-12-29 Opko Curna, Llc Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
EP2450055B1 (en) 2009-06-30 2018-01-03 Obshestvo S OgranichennoyOtvetstvennostiu"OncoMax" Method for suppressing renal tumor growth by blocking fibroblast growth factor receptor
US9018187B2 (en) 2009-07-01 2015-04-28 Protiva Biotherapeutics, Inc. Cationic lipids and methods for the delivery of therapeutic agents
CA2767127A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
US8569256B2 (en) 2009-07-01 2013-10-29 Protiva Biotherapeutics, Inc. Cationic lipids and methods for the delivery of therapeutic agents
KR20120051002A (en) 2009-07-07 2012-05-21 제넨테크, 인크. Diagnosis and treatment of autoimmune demyelinating diseases
JP5872466B2 (en) 2009-07-17 2016-03-01 テクニカル ユニバーシティ オブ デンマークTechnical University Of Denmark Encapsulation method for producing radionuclide-containing nanoparticles
AU2010276160A1 (en) 2009-07-21 2012-02-09 Gilead Calistoga Llc Treatment of liver disorders with PI3K inhibitors
US9937128B2 (en) 2009-08-03 2018-04-10 The University Of North Carolina At Chapel Hill Liposomes comprising a calcium phosphate-containing precipitate
CN102762731B (en) 2009-08-05 2018-06-22 库尔纳公司 By inhibiting to treat insulin gene (INS) relevant disease for the natural antisense transcript of insulin gene (INS)
CN102573909A (en) 2009-08-15 2012-07-11 霍夫曼-拉罗奇有限公司 Anti-angiogenesis therapy for the treatment of previously treated breast cancer
FR2955324A1 (en) 2010-01-15 2011-07-22 Sanofi Aventis DISUBSTITUTED 4- (5-AMINOMETHYL-PHENYL) -PIPERIDIN-1-YL] -1H-INDOL-3-YL] -METHANONES
EP2470657B1 (en) 2009-08-25 2019-10-23 CuRNA, Inc. Treatment of 'iq motif containing gtpase activating protein' (iqgap) related diseases by inhibition of natural antisense transcript to iqgap
JP2013503204A (en) 2009-08-31 2013-01-31 アンプリミューン, インコーポレイテッド B7-H4 fusion protein and methods of use thereof
US20110059111A1 (en) 2009-09-01 2011-03-10 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Mammalian receptors as targets for antibody and active vaccination therapy against mold infections
ES2599076T3 (en) 2009-09-02 2017-01-31 Genentech, Inc. Smoothened mutant and methods of use thereof
EP2483281B1 (en) * 2009-10-01 2014-06-04 Cymabay Therapeutics, Inc. Substituted tetrazol-1-yl-phenoxymethyl-thiazol-2-yl-piperidinyl-pyrimidine salts
US8535912B2 (en) * 2009-10-15 2013-09-17 Genentech, Inc. Chimeric fibroblast growth factors with altered receptor specificity
DK2488204T3 (en) 2009-10-16 2016-06-06 Oncomed Pharm Inc Therapeutic combination and use of DLL4 antagonist antibodies and blood pressure lowering agents
WO2011049625A1 (en) 2009-10-20 2011-04-28 Mansour Samadpour Method for aflatoxin screening of products
CA2778442A1 (en) 2009-10-22 2011-04-28 Genentech, Inc. Methods and compositions for modulating hepsin activation of macrophage-stimulating protein
EP2493468A1 (en) 2009-10-30 2012-09-05 Sanofi Amino-benzoic acid derivatives for use in the treatment of dihydrogenase-related disorders
KR101821768B1 (en) 2009-11-05 2018-01-24 리젠 파마슈티컬스 소시에떼 아노님 Novel benzopyran kinase modulators
WO2011056234A1 (en) 2009-11-06 2011-05-12 Fibrogen, Inc. Treatment for radiation-induced disorders
WO2011062997A2 (en) 2009-11-17 2011-05-26 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin
MX2012006072A (en) 2009-11-30 2012-07-23 Genentech Inc Antibodies for treating and diagnosing tumors expressing slc34a2 (tat211 = seqid2 ).
WO2011071957A1 (en) 2009-12-07 2011-06-16 Sea Lane Biotechnologies, Llc Conjugates comprising an antibody surrogate scaffold with improved pharmacokinetic properties
WO2011071860A2 (en) 2009-12-07 2011-06-16 Alnylam Pharmaceuticals, Inc. Compositions for nucleic acid delivery
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
WO2011081904A1 (en) 2009-12-14 2011-07-07 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Delivery of transthyretin across the blood-brain barrier as a treatment for alzheimer's disease
KR101823702B1 (en) 2009-12-16 2018-01-30 큐알엔에이, 인크. Treatment of membrane bound transcription factor peptidase, site 1 (mbtps1) related diseases by inhibition of natural antisense transcript to mbtps1
EP2513308B1 (en) 2009-12-17 2017-01-18 Merck Sharp & Dohme Corp. Modulation of pilr to treat immune disorders
US20130017223A1 (en) 2009-12-18 2013-01-17 The University Of British Columbia Methods and compositions for delivery of nucleic acids
JP6031356B2 (en) 2009-12-23 2016-11-24 カッパーアールエヌエー,インコーポレイテッド Treatment of uncoupling protein 2 (UCP2) -related diseases by inhibition of natural antisense transcripts against UCP2.
US9023996B2 (en) 2009-12-23 2015-05-05 Synimmune Gmbh Anti-FLT3 antibodies
JP2013515725A (en) 2009-12-23 2013-05-09 サノフイ [4 [4- (5-Aminomethyl-2-fluoro-phenyl) -piperidin-1-yl]-(1H-pyrrolo-pyridin-yl) -methanones and their synthesis
DK2516648T3 (en) 2009-12-23 2018-02-12 Curna Inc TREATMENT OF HEPATOCYTE GROWTH FACTOR (HGF) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT AGAINST HGF
AR079661A1 (en) 2009-12-23 2012-02-08 Sanofi Aventis DERIVATIVES OF N- (4F-3- {1- [INDOL-3-CARBON]] PIPERIDIN-4-IL} BENCIL) ACETAMIDS
JP5728718B2 (en) 2009-12-28 2015-06-03 オンコセラピー・サイエンス株式会社 Anti-CDH3 antibody and use thereof
CN102770540B (en) 2009-12-29 2017-06-23 库尔纳公司 P63 relevant diseases are treated by suppressing the natural antisense transcript of oncoprotein 63 (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
US20110159588A1 (en) 2009-12-30 2011-06-30 Kui Lin Methods for Modulating a PDGF-AA Mediated Biological Response
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
WO2011083845A1 (en) * 2010-01-08 2011-07-14 富士フイルム株式会社 Targeting agent for tumor site
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
TWI535445B (en) 2010-01-12 2016-06-01 安可美德藥物股份有限公司 Wnt antagonists and methods of treatment and screening
CN102958534B (en) 2010-01-13 2014-11-05 昂考梅德药品有限公司 Notch1 binding agents and methods of use thereof
WO2011090971A2 (en) 2010-01-19 2011-07-28 The Trustees Of Columbia University In The City Of New York Osteocalcin as a treatment for male reproductive disorders
WO2011089211A1 (en) 2010-01-22 2011-07-28 Synimmune Gmbh Anti-cd133 antibodies and methods of using the same
KR101853510B1 (en) 2010-01-25 2018-06-20 큐알엔에이, 인크. Treatment of rnase h1 related diseases by inhibition of natural antisense transcript to rnase h1
US8198337B2 (en) * 2010-01-27 2012-06-12 Momentive Performance Materials Inc. Demulsifier compositions and methods for separating emulsions using the same
WO2011092708A2 (en) 2010-02-01 2011-08-04 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Liposomes comprising amphipathic drugs and method for their preparation
EP2539452B1 (en) 2010-02-22 2016-07-27 CuRNA, Inc. Treatment of pyrroline-5-carboxylate reductase 1 (pycr1) related diseases by inhibition of natural antisense transcript to pycr1
KR101839161B1 (en) 2010-02-23 2018-03-16 제넨테크, 인크. Anti-angiogenesis therapy for the treatment of ovarian cancer
BR112012020373A8 (en) 2010-02-23 2018-01-02 Genentech Inc isolated antibody, cell, isolated nucleic acid, method of identification, cell proliferation inhibition, therapeutic treatment, determination of the presence of a tat419 protein, diagnosis of the presence of cancer and distribution of cytotoxic agent
US8975376B2 (en) 2010-02-23 2015-03-10 Sanofi Anti-alpha2 integrin antibodies and their uses
SA114360064B1 (en) 2010-02-24 2016-01-05 رينات نيوروساينس كوربوريشن Antagonist anti-il-7 receptor antibodies and methods
CA2791278C (en) 2010-02-25 2015-11-24 The Johns Hopkins University Sustained delivery of therapeutic agents to an eye compartment
AU2011225716A1 (en) 2010-03-11 2012-09-27 Pfizer Inc. Antibodies with pH dependent antigen binding
JP5937059B2 (en) 2010-03-22 2016-06-22 ジェネンテック, インコーポレイテッド Compositions and methods useful for stabilizing protein-containing formulations
RU2615143C2 (en) 2010-03-24 2017-04-04 Адвирна Self-delivered rnai compounds of reduced size
CA2793959C (en) 2010-03-25 2019-06-04 Oregon Health & Science University Cmv glycoproteins and recombinant vectors
EP2552957A4 (en) 2010-03-29 2013-11-20 Zymeworks Inc Antibodies with enhanced or suppressed effector function
EP2552933A1 (en) 2010-03-31 2013-02-06 Gilead Pharmasset LLC Purine nucleoside phosphoramidate
EP2552965A2 (en) 2010-03-31 2013-02-06 Boehringer Ingelheim International GmbH Anti-cd40 antibodies
EP2552931B1 (en) 2010-03-31 2014-07-23 Gilead Pharmasset LLC Stereoselective synthesis of phosphorus containing actives
PL3290428T3 (en) 2010-03-31 2022-02-07 Gilead Pharmasset Llc Tablet comprising crystalline (s)-isopropyl 2-(((s)-(((2r,3r,4r,5r)-5-(2,4-dioxo-3,4-dihydropyrimidin-1 (2h)-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)amino)propanoate
US9102938B2 (en) 2010-04-01 2015-08-11 Alnylam Pharmaceuticals, Inc. 2′ and 5′ modified monomers and oligonucleotides
EP2556085A2 (en) 2010-04-05 2013-02-13 Bar-Ilan University Protease-activatable pore-forming polypeptides
EP3517613A1 (en) 2010-04-09 2019-07-31 CuRNA, Inc. Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
WO2011133868A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Conformationally restricted dinucleotide monomers and oligonucleotides
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
US10913767B2 (en) 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
WO2011137114A1 (en) 2010-04-26 2011-11-03 Abraxis Bioscience, Llc Sparc binding antibodies and uses thereof
US20130195962A1 (en) * 2010-04-28 2013-08-01 National University Corporation Hokkaido University Lipid membrane structure
JP5896175B2 (en) 2010-04-29 2016-03-30 アイオーニス ファーマシューティカルズ, インコーポレーテッドIonis Pharmaceuticals,Inc. Regulation of transthyretin expression
US20130156845A1 (en) 2010-04-29 2013-06-20 Alnylam Pharmaceuticals, Inc. Lipid formulated single stranded rna
ES2552954T3 (en) 2010-04-30 2015-12-03 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies and methods for the use of antibodies
CN102958941A (en) 2010-05-03 2013-03-06 霍夫曼-拉罗奇有限公司 Compositions and methods for the diagnosis and treatment of tumor
EP2566510A1 (en) 2010-05-03 2013-03-13 F. Hoffmann-La Roche AG Compositions and methods useful for reducing the viscosity of protein-containing formulations
KR20130101442A (en) 2010-05-03 2013-09-13 큐알엔에이, 인크. Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
CN109593814A (en) 2010-05-12 2019-04-09 哥伦比亚大学纽约管理委员会 Screening analysis, the method and pharmaceutical composition for identifying medicament
TWI531370B (en) 2010-05-14 2016-05-01 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
CN107674073B (en) 2010-05-17 2021-09-10 印蔻真治疗公司 3, 5-disubstituted-3H-imidazo [4,5-B ] pyridine compounds as modulators of protein kinases
JP6267510B2 (en) 2010-05-18 2018-01-24 ニューメディシンズ,インコーポレーテッド IL-12 formulation for enhancing hematopoiesis
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
DK2576783T3 (en) 2010-05-26 2018-03-12 Curna Inc TREATMENT OF ATONAL HOMOLOGY 1- (ATOH1) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPTS AT ATOH1
WO2011153380A2 (en) 2010-06-02 2011-12-08 Dana-Farber Cancer Institute, Inc. Humanized monoclonal antibodies and methods of use
WO2011153243A2 (en) 2010-06-02 2011-12-08 Genentech, Inc. Anti-angiogenesis therapy for treating gastric cancer
JP5893611B2 (en) 2010-06-03 2016-03-23 アルニラム・ファーマシューティカルズ・インコーポレーテッド Biodegradable lipids for delivery of active agents
US8299117B2 (en) 2010-06-16 2012-10-30 Metabolex Inc. GPR120 receptor agonists and uses thereof
ES2526124T3 (en) 2010-06-16 2015-01-07 Cymabay Therapeutics, Inc. GPR120 receptor agonists and their uses
CA2803317A1 (en) * 2010-06-19 2011-12-22 Western University Of Health Sciences Novel formulation of pegylated-liposome encapsulated glycopeptide antibiotics
CN103037843A (en) 2010-06-23 2013-04-10 麦它波莱克斯股份有限公司 Compositions of 5-ethyl-2-{4-[4-(4-tetrazol-1-Yl-phenoxymethyl)-thiazol-2-Yl]-piperidin-1-Yl}-pyrimidine
NZ605147A (en) 2010-06-24 2014-12-24 Genentech Inc Compositions and methods containing alkylglycosides for stabilizing protein-containing formulations
CN102309448B (en) * 2010-06-29 2014-07-09 中国人民解放军军事医学科学院毒物药物研究所 Pulmonary delivery ciprofloxacin pharmaceutical composition and preparation method thereof
US9006417B2 (en) 2010-06-30 2015-04-14 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
EP2591114B1 (en) 2010-07-06 2016-06-08 GlaxoSmithKline Biologicals SA Immunisation of large mammals with low doses of rna
WO2012006378A1 (en) 2010-07-06 2012-01-12 Novartis Ag Liposomes with lipids having an advantageous pka- value for rna delivery
PT3243526T (en) 2010-07-06 2020-03-04 Glaxosmithkline Biologicals Sa Delivery of rna to trigger multiple immune pathways
HRP20221522T1 (en) 2010-07-06 2023-02-17 Glaxosmithkline Biologicals S.A. Virion-like delivery particles for self-replicating rna molecules
KR20130050966A (en) 2010-07-12 2013-05-16 씨오브이엑스 테크놀로지스 아일랜드 리미티드 Multifunctional antibody conjugates
CN103068982B (en) 2010-07-14 2017-06-09 库尔纳公司 DLG relevant diseases are treated by suppressing the natural antisense transcript of the big homologue of plate-like (DLG)
WO2012012750A1 (en) 2010-07-23 2012-01-26 Trustees Of Boston University ANTI-DEsupR INHIBITORS AS THERAPEUTICS FOR INHIBITION OF PATHOLOGICAL ANGIOGENESIS AND TUMOR CELL INVASIVENESS AND FOR MOLECULAR IMAGING AND TARGETED DELIVERY
WO2012016188A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
WO2012016184A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
US8906943B2 (en) 2010-08-05 2014-12-09 John R. Cashman Synthetic compounds and methods to decrease nicotine self-administration
EP2420250A1 (en) 2010-08-13 2012-02-22 Universitätsklinikum Münster Anti-Syndecan-4 antibodies
EP2603237A4 (en) 2010-08-12 2014-05-21 Theraclone Sciences Inc Anti-hemagglutinin antibody compositions and methods of use thereof
CA2809837C (en) 2010-08-31 2019-12-31 Theraclone Sciences, Inc. Human immunodeficiency virus (hiv)-neutralizing antibodies
DK2611467T3 (en) 2010-08-31 2022-08-01 Glaxosmithkline Biologicals Sa Small liposomes for delivery of immunogen-encoding RNA
EP4119155A1 (en) 2010-08-31 2023-01-18 GlaxoSmithKline Biologicals S.A. Pegylated liposomes for delivery of immunogen-encoding rna
US8551479B2 (en) 2010-09-10 2013-10-08 Oncomed Pharmaceuticals, Inc. Methods for treating melanoma
WO2012047968A2 (en) 2010-10-05 2012-04-12 Genentech, Inc. Mutant smoothened and methods of using the same
CA2813901C (en) 2010-10-06 2019-11-12 Curna, Inc. Treatment of sialidase 4 (neu4) related diseases by inhibition of natural antisense transcript to neu4
EP4098325A1 (en) 2010-10-11 2022-12-07 GlaxoSmithKline Biologicals S.A. Antigen delivery platforms
WO2012054723A2 (en) 2010-10-22 2012-04-26 Opko Curna Llc Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
US20120201821A1 (en) 2010-10-25 2012-08-09 Gonzalez Jr Lino Treatment of Gastrointestinal Inflammation and Psoriasis and Asthma
EP2633052B1 (en) 2010-10-27 2018-04-11 CuRNA, Inc. Treatment of interferon-related developmental regulator 1 (ifrd1) related diseases by inhibition of natural antisense transcript to ifrd1
SG190006A1 (en) 2010-11-04 2013-06-28 Boehringer Ingelheim Int Anti-il-23 antibodies
WO2012061811A2 (en) 2010-11-05 2012-05-10 Fibrogen, Inc. Treatment method for lung remodeling diseases
WO2012065044A2 (en) 2010-11-12 2012-05-18 Purdue Research Foundation Treating bladder tumor cells using fibronectin attachment protein as a target
EP2640831A1 (en) 2010-11-17 2013-09-25 Sea Lane Biotechnologies,llc. Influenza virus neutralizing agents that mimic the binding site of an influenza neutralizing antibody
JP2014505666A (en) 2010-11-18 2014-03-06 ザ ジェネラル ホスピタル コーポレイション Antihypertensive composition and use for cancer treatment
US8987225B2 (en) 2010-11-23 2015-03-24 Curna, Inc. Treatment of NANOG related diseases by inhibition of natural antisense transcript to NANOG
WO2012075140A1 (en) 2010-11-30 2012-06-07 Pharmasset, Inc. Compounds
JP5947807B2 (en) 2010-12-14 2016-07-06 テクニカル ユニバーシティ オブ デンマークTechnical University Of Denmark Encapsulation of radionuclides in nanoparticle compositions
EP2468259A1 (en) * 2010-12-23 2012-06-27 Traslational Cancer Drugs Pharma, S.L. Pharmaceutical compositions of pyridinium and quinolinium derivatives
CA2834968C (en) 2011-01-05 2018-01-09 Livon Laboratories Methods of making liposomes, liposome compositions made by the methods, and methods of using the same
EP3202760B1 (en) 2011-01-11 2019-08-21 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
CA2821985C (en) 2011-01-11 2019-07-09 Dimerix Bioscience Pty Ltd Combination therapy
RU2440142C1 (en) 2011-02-07 2012-01-20 Общество С Ограниченной Ответственностью "Онкомакс" Antibody, stopping or retarding tumour growth (versions), method of suppressing tumour growth, method of diagnosing malignant lesions
WO2012109495A1 (en) 2011-02-09 2012-08-16 Metabolic Solutions Development Company, Llc Cellular targets of thiazolidinediones
EP2675478A4 (en) 2011-02-14 2015-06-10 Theraclone Sciences Inc Compositions and methods for the therapy and diagnosis of influenza
PL226015B1 (en) * 2011-03-03 2017-06-30 Wrocławskie Centrum Badań Eit + Spółka Z Ograniczoną Liposome preparation containing anticancer active substance, process for the preparation thereof and pharmaceutical compositions containing thereof
AU2012225310B2 (en) 2011-03-09 2017-08-03 Cell Signaling Technology, Inc. Methods and reagents for creating monoclonal antibodies
AU2012229188A1 (en) 2011-03-15 2013-09-26 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
SG10201601789TA (en) 2011-03-16 2016-04-28 Amgen Inc Potent And Selective Inhibitors Of Nav1.3 And Nav1.7
US10184942B2 (en) 2011-03-17 2019-01-22 University Of South Florida Natriuretic peptide receptor as a biomarker for diagnosis and prognosis of cancer
WO2012134975A1 (en) 2011-03-28 2012-10-04 St. Jude Children's Research Hospital Methods and compositions employing immunogenic fusion proteins
CA2831732C (en) 2011-03-31 2019-12-31 Genentech, Inc. Use of a monoclonal anti-beta7 antibody for treating gastrointestinal inflammatory disorders
EP2508176A1 (en) 2011-04-08 2012-10-10 Lipotarg Gmbh Novel combination treatment of cancer
ES2634450T3 (en) 2011-04-13 2017-09-27 Ionis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
KR101589135B1 (en) 2011-04-20 2016-01-29 주식회사 셀앤바이오 Humanized anti-EMAPII antibodies and uses thereof
EP2699264B1 (en) 2011-04-20 2018-03-14 Medlmmune, LLC Antibodies and other molecules that bind b7-h1 and pd-1
US9775841B2 (en) 2011-05-04 2017-10-03 Rhizen Pharmaceuticals Sa Compounds as modulators of protein kinases
JP6400470B2 (en) 2011-05-16 2018-10-03 ジェネロン(シャンハイ)コーポレイション リミテッド Multispecific Fab fusion proteins and methods of use
US9593330B2 (en) 2011-06-09 2017-03-14 Curna, Inc. Treatment of frataxin (FXN) related diseases by inhibition of natural antisense transcript to FXN
AU2012267786B2 (en) 2011-06-10 2017-08-03 Oregon Health & Science University CMV glycoproteins and recombinant vectors
AU2012271357A1 (en) 2011-06-16 2013-05-02 Ionis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
EP2537532A1 (en) 2011-06-22 2012-12-26 J. Stefan Institute Cathepsin-binding compounds bound to a nanodevice and their diagnostic and therapeutic use
WO2013003652A1 (en) 2011-06-28 2013-01-03 Sea Lane Biotechnologies, Llc Multispecific stacked variable domain binding proteins
JP2014522842A (en) 2011-07-06 2014-09-08 ノバルティス アーゲー Immunogenic combination compositions and uses thereof
RS61235B1 (en) 2011-07-13 2021-01-29 Yissum Res Dev Co Of Hebrew Univ Jerusalem Ltd Liposomes co-encapsulating a bisphosphonate and an amphipathic agent
MX2014000578A (en) 2011-07-14 2014-04-30 Pfizer Treatment with anti-pcsk9 antibodies.
US9120858B2 (en) 2011-07-22 2015-09-01 The Research Foundation Of State University Of New York Antibodies to the B12-transcobalamin receptor
US20130022551A1 (en) 2011-07-22 2013-01-24 Trustees Of Boston University DEspR ANTAGONISTS AND AGONISTS AS THERAPEUTICS
WO2013015821A1 (en) 2011-07-22 2013-01-31 The Research Foundation Of State University Of New York Antibodies to the b12-transcobalamin receptor
CA2845536A1 (en) 2011-08-15 2013-02-21 Amplimmune, Inc. Anti-b7-h4 antibodies and their uses
FR2979239A1 (en) * 2011-08-25 2013-03-01 Trophos LIPOSOME COMPRISING AT LEAST ONE CHOLESTEROL DERIVATIVE
US8822651B2 (en) 2011-08-30 2014-09-02 Theraclone Sciences, Inc. Human rhinovirus (HRV) antibodies
CA2847057C (en) 2011-08-31 2019-06-11 St. Jude Children's Research Hospital Methods and compositions to detect the level of lysosomal exocytosis activity and methods of use
SG11201400100SA (en) 2011-09-09 2014-06-27 Univ Osaka Dengue-virus serotype neutralizing antibodies
EP2568289A3 (en) 2011-09-12 2013-04-03 International AIDS Vaccine Initiative Immunoselection of recombinant vesicular stomatitis virus expressing hiv-1 proteins by broadly neutralizing antibodies
CA2849073A1 (en) 2011-09-19 2013-03-28 Gencia Corporation Modified creatine compounds
MX353322B (en) 2011-09-20 2018-01-05 Ionis Pharmaceuticals Inc Antisense modulation of gcgr expression.
AU2012310328A1 (en) 2011-09-23 2014-04-10 Technophage, Investigação E Desenvolvimento Em Biotecnologia, S.A. Anti-Tumor Necrosis Factor-alpha agents and uses thereof
PL3485903T3 (en) 2011-09-23 2023-06-12 Mereo Biopharma 5, Inc. Vegf/dll4 binding agents and uses thereof
MX2014003313A (en) 2011-09-23 2014-07-09 Amgen Res Munich Gmbh Bispecific binding molecules for 5t4 and cd3.
US9701623B2 (en) 2011-09-27 2017-07-11 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
KR102162901B1 (en) 2011-09-29 2020-10-08 피엘엑스 옵코 인코포레이티드 Ph dependent carriers for targeted release of pharmaceuticals along the gastrointestinal tract, compositions therefrom, and making and using same
US20130085139A1 (en) 2011-10-04 2013-04-04 Royal Holloway And Bedford New College Oligomers
CA2850857C (en) 2011-10-06 2022-07-26 Immunovaccine Technologies Inc. Liposome compositions comprising an adjuvant that activates or increases the activity of tlr2 and uses thereof
MX2014004022A (en) 2011-10-14 2014-04-30 Genentech Inc ANTI-HtrA1 ANTIBODIES AND METHODS OF USE.
US8883989B2 (en) 2011-10-18 2014-11-11 Regents Of The University Of Minnesota Fractalkine binding polynucleotides and methods of use
MX350944B (en) 2011-10-25 2017-09-26 Ionis Pharmaceuticals Inc Antisense modulation of gccr expression.
EP2586461A1 (en) 2011-10-27 2013-05-01 Christopher L. Parks Viral particles derived from an enveloped virus
BR112014009953A2 (en) 2011-10-28 2017-12-05 Genentech Inc tumor growth inhibition method, melanoma treatment, industrialized article and use
MY187874A (en) 2011-10-31 2021-10-26 Genentech Inc Antibody formulations
KR101688898B1 (en) * 2011-11-03 2016-12-22 타이완 리포솜 캄퍼니 리미티드 Pharmaceutical compositions of hydrophobic camptothecin derivatives
US10980798B2 (en) 2011-11-03 2021-04-20 Taiwan Liposome Company, Ltd. Pharmaceutical compositions of hydrophobic camptothecin derivatives
JP5936702B2 (en) 2011-11-11 2016-06-22 ライナット ニューロサイエンス コーポレイション Antibodies specific for TROP-2 and uses thereof
TWI679212B (en) 2011-11-15 2019-12-11 美商安進股份有限公司 Binding molecules for e3 of bcma and cd3
ES2755732T3 (en) 2011-11-16 2020-04-23 Boehringer Ingelheim Int Anti IL-36R antibodies
CA2858876A1 (en) 2011-12-15 2013-06-20 The University Of Chicago Methods and compositions for cancer therapy using mutant light molecules with increased affinity to receptors
WO2013093693A1 (en) 2011-12-22 2013-06-27 Rinat Neuroscience Corp. Staphylococcus aureus specific antibodies and uses thereof
EP2794659A1 (en) 2011-12-22 2014-10-29 Rinat Neuroscience Corp. Human growth hormone receptor antagonist antibodies and methods of use thereof
EP3539982A3 (en) 2011-12-23 2020-01-15 Pfizer Inc Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
WO2013101771A2 (en) 2011-12-30 2013-07-04 Genentech, Inc. Compositions and method for treating autoimmune diseases
AR090410A1 (en) 2012-01-09 2014-11-12 Covx Technologies Ireland Ltd MUTANT ANTIBODIES AND CONJUGATION OF THE SAME
US9636381B2 (en) 2012-01-18 2017-05-02 Neumedicines, Inc. Methods for radiation protection by administering IL-12
CA2862292C (en) 2012-01-20 2019-10-08 Sea Lane Biotechnologies, Llc Binding molecule conjugates
US20150004219A1 (en) 2012-02-02 2015-01-01 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Stable liposomes for drug delivery
MY169341A (en) 2012-02-06 2019-03-21 Inhibrx Inc Cd47 antibodies and methods of use thereof
ES2773299T3 (en) * 2012-02-17 2020-07-10 Celsion Corp Thermosensitive nanoparticle formulations and their preparation method
US20150037334A1 (en) 2012-03-01 2015-02-05 Amgen Research (Munich) Gmbh Long life polypeptide binding molecules
PL2834241T3 (en) 2012-03-05 2021-07-12 Gilead Calistoga Llc Polymorphic forms of (s)-2-(1-(9h-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3h)-one
US20150031750A1 (en) 2012-03-15 2015-01-29 The Scripps Research Institute Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
PL2831117T3 (en) 2012-03-29 2018-03-30 Novimmune Sa Anti-tlr4 antibodies and uses thereof
CA2865719C (en) 2012-03-30 2020-09-22 Rhizen Pharmaceuticals Sa Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of c-met protein kinases
WO2013151649A1 (en) 2012-04-04 2013-10-10 Sialix Inc Glycan-interacting compounds
US20160054299A9 (en) 2012-04-24 2016-02-25 University Of Miami Perforin 2 defense against invasive and multidrug resistant pathogens
CA2871985C (en) 2012-05-03 2023-10-10 Boehringer Ingelheim International Gmbh Anti-il-23p19 antibodies
US9441045B2 (en) 2012-05-04 2016-09-13 Dana-Farber Cancer Institute, Inc. Affinity matured anti-CCR4 humanized monoclonal antibodies and methods of use
CA2872519C (en) 2012-05-04 2017-09-05 The Johns Hopkins University Lipid-based drug carriers for rapid penetration through mucus linings
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
US9789193B2 (en) 2012-06-15 2017-10-17 The Brigham And Women's Hospital, Inc. Compositions for treating cancer and methods for making the same
WO2013192546A1 (en) 2012-06-22 2013-12-27 Cytomx Therapeutics, Inc. Activatable antibodies having non-binding steric moieties and mehtods of using the same
ES2631608T3 (en) 2012-06-27 2017-09-01 International Aids Vaccine Initiative Env-glycoprotein variant of HIV-1
WO2014015016A1 (en) * 2012-07-18 2014-01-23 Onyx Therapeutics, Inc. Liposomal compositions of epoxyketone-based proteasome inhibitors
US8603470B1 (en) 2012-08-07 2013-12-10 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases
CN103565745A (en) 2012-08-10 2014-02-12 德克萨斯州大学系统董事会 Neuroprotective liposome compositions and methods for treatment of stroke
SG10201701165TA (en) 2012-08-14 2017-03-30 Univ Nanyang Tech Angiopoietin-like 4 antibody and a method of its use in cancer treatment
WO2014039533A2 (en) 2012-09-04 2014-03-13 Eleison Pharmaceuticals, Llc Preventing pulmonary recurrence of cancer with lipid-complexed cisplatin
EP2943194A1 (en) 2012-09-17 2015-11-18 Chemedest Ltd. Treatment of peripheral neuropathy using gfr(alpha)3 type receptor agonists
WO2014055897A2 (en) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Human monoclonal anti-pd-l1 antibodies and methods of use
PL2903691T3 (en) 2012-10-05 2019-10-31 Hoffmann La Roche Methods for diagnosing and treating inflammatory bowel disease
JP2015536933A (en) 2012-10-23 2015-12-24 オンコメッド ファーマシューティカルズ インコーポレイテッド Methods of treating neuroendocrine tumors using Wnt pathway binding agents
WO2014071018A1 (en) 2012-10-31 2014-05-08 Oncomed Pharmaceuticals, Inc. Methods and monitoring of treatment with a dll4 antagonist
DK2925782T3 (en) 2012-12-03 2020-04-06 Novimmune Sa ANTI-CD47 ANTIBODIES AND PROCEDURES FOR USE THEREOF
US10188728B2 (en) 2012-12-12 2019-01-29 Temple University—Of the Commonwealth System of Higher Education Compositions and methods for treatment of cancer
WO2014100483A1 (en) 2012-12-19 2014-06-26 Amplimmune, Inc. Anti-human b7-h4 antibodies and their uses
WO2014100379A1 (en) 2012-12-19 2014-06-26 The Research Foundation For The State University Of New York Compositions and method for light triggered release of materials from nanovesicles
GB201223053D0 (en) 2012-12-20 2013-02-06 Medical Res Council Receptor
EP2935311B1 (en) 2012-12-20 2021-03-31 Amgen Inc. Apj receptor agonists and uses thereof
RU2691428C2 (en) 2012-12-21 2019-06-13 МЕДИММЬЮН, ЭлЭлСи Antibodies to h7cr
EP2948478B1 (en) 2013-01-25 2019-04-03 Amgen Inc. Antibodies targeting cdh19 for melanoma
JO3519B1 (en) 2013-01-25 2020-07-05 Amgen Inc Antibody constructs for CDH19 and CD3
WO2014124006A1 (en) 2013-02-05 2014-08-14 The Johns Hopkins University Nanoparticles for magnetic resonance imaging tracking and methods of making and using thereof
WO2014123580A1 (en) 2013-02-06 2014-08-14 Inhibrx Llc Non-platelet depleting and non-red blood cell depleting cd47 antibodies and methods of use thereof
EP2956169B1 (en) 2013-02-12 2018-04-11 THE UNITED STATES OF AMERICA, represented by the S Monoclonal antibodies that neutralize norovirus
WO2014165277A2 (en) 2013-03-12 2014-10-09 Amgen Inc. POTENT AND SELECTIVE INHIBITORS OF Nav1.7
BR112015022936A2 (en) 2013-03-14 2017-07-18 J Schentag Jerome cholestosome vesicles for incorporation of molecules in chylomicrons
US9302005B2 (en) 2013-03-14 2016-04-05 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
EP2970449B1 (en) 2013-03-15 2019-09-25 Amgen Research (Munich) GmbH Single chain binding molecules comprising n-terminal abp
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
WO2014144061A2 (en) 2013-03-15 2014-09-18 Dana-Farber Cancer Institute, Inc. Flavivirus neutralizing antibodies and methods of use thereof
WO2014140368A1 (en) 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Antibody constructs for influenza m2 and cd3
US9707244B2 (en) 2013-03-15 2017-07-18 Gencia Corporation Compositions and methods for treating conditions that affect epidermis
WO2014152232A2 (en) 2013-03-15 2014-09-25 Dyax Corp. Anti-plasma kallikrein antibodies
US10159688B2 (en) * 2013-03-15 2018-12-25 M. Alphabet 3, L.L.C. Methods and compositions for enhancing oxygen levels in tissues
US11156608B2 (en) 2013-03-15 2021-10-26 The Trustees Of The University Of Pennsylvania Method for the site-specific covalent cross-linking of antibodies to surfaces
US20140283157A1 (en) 2013-03-15 2014-09-18 Diadexus, Inc. Lipoprotein-associated phospholipase a2 antibody compositions and methods of use
AR095374A1 (en) 2013-03-15 2015-10-14 Amgen Res (Munich) Gmbh UNION MOLECULES FOR BCMA AND CD3
BR112015024856A2 (en) 2013-03-27 2018-04-17 Genentech Inc determination or monitoring methods
CA2909357C (en) 2013-04-12 2022-02-22 Icahn School Of Medicine At Mount Sinai Use of ketamine to treat post-traumatic stress disorder
CN105377867B (en) 2013-05-03 2019-11-12 加利福尼亚大学董事会 The cyclic annular dinucleotides of I type interferon induces
SG10201913751RA (en) 2013-05-06 2020-03-30 Scholar Rock Inc Compositions and methods for growth factor modulation
RU2015147721A (en) 2013-05-07 2017-06-15 Ринат Нейросаенз Корпорэйшн ANTIBODIES AGAINST GLUCAGON RECEPTOR AND METHODS OF USING THEM
EP2994163B1 (en) 2013-05-09 2019-08-28 The United States of America, as represented by The Secretary, Department of Health and Human Services Single-domain vhh antibodies directed to norovirus gi.1 and gii.4 and their use
BR112015029395A2 (en) 2013-05-24 2017-09-19 Medimmune Llc ANTI-B7-H5 ANTIBODIES AND THEIR USES
SG11201509839TA (en) 2013-05-31 2016-01-28 Genentech Inc Anti-wall teichoic antibodies and conjugates
MX369758B (en) 2013-05-31 2019-11-20 Genentech Inc Anti-wall teichoic antibodies and conjugates.
ES2891755T3 (en) 2013-06-06 2022-01-31 Pf Medicament Anti-C10orf54 antibodies and uses thereof
DK3007704T3 (en) 2013-06-13 2021-03-29 Antisense Therapeutics Ltd Combination therapy for acromegaly
US10208125B2 (en) 2013-07-15 2019-02-19 University of Pittsburgh—of the Commonwealth System of Higher Education Anti-mucin 1 binding agents and uses thereof
PT3406633T (en) 2013-07-25 2022-05-04 Cytomx Therapeutics Inc Multispecific antibodies, multispecific activatable antibodies and methods of using the same
US20160193148A1 (en) * 2013-08-01 2016-07-07 University Of Georgia Research Foundation, Inc. Liposomal formulations for the treatment of bacterial infections
KR101809072B1 (en) 2013-08-02 2017-12-14 화이자 인코포레이티드 Anti-cxcr4 antibodies and antibody-drug conjugates
BR112016002845A2 (en) 2013-08-12 2017-09-12 Genentech Inc compositions and methods for treating complement-associated conditions
EP3705494A3 (en) 2013-08-14 2020-12-09 Sachdev Sidhu Antibodies against frizzled proteins and methods of use thereof
EP3038600B1 (en) * 2013-08-27 2020-06-03 Northeastern University Nanoparticle drug delivery system and method of treating cancer and neurotrauma
US10647983B2 (en) 2013-09-04 2020-05-12 Cold Spring Harbor Laboratory Reducing nonsense-mediated mRNA decay
US20150065381A1 (en) 2013-09-05 2015-03-05 International Aids Vaccine Initiative Methods of identifying novel hiv-1 immunogens
US11236338B2 (en) 2013-09-05 2022-02-01 Sarepta Therapeutics, Inc. Antisense-induced exon2 inclusion in acid alpha-glucosidase
AR097648A1 (en) 2013-09-13 2016-04-06 Amgen Inc COMBINATION OF EPIGENETIC FACTORS AND BIESPECTIVE COMPOUNDS THAT HAVE LIKE DIANA CD33 AND CD3 IN THE TREATMENT OF MYELOID LEUKEMIA
PL3046537T3 (en) * 2013-09-16 2022-01-31 Glycomine, Inc. Pharmaceutical preparation of carbohydrates for therapeutic use
DE102013015334A1 (en) * 2013-09-17 2015-03-19 Fresenius Medical Care Deutschland Gmbh Magnesium-liposome complexes
WO2015042466A2 (en) 2013-09-19 2015-03-26 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Compositions and methods for inhibiting jc virus (jcv)
MX2016003957A (en) 2013-09-25 2017-02-02 Cytomx Therapeutics Inc Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof.
EP3052131B1 (en) 2013-10-01 2018-12-05 Mayo Foundation for Medical Education and Research Methods for treating cancer in patients with elevated levels of bim
US10058604B2 (en) 2013-10-07 2018-08-28 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
EP3070167A4 (en) 2013-11-06 2017-06-07 Osaka University Antibody having broad neutralizing activity in group 1 influenza a virus
WO2015087187A1 (en) 2013-12-10 2015-06-18 Rinat Neuroscience Corp. Anti-sclerostin antibodies
EP3080611B1 (en) 2013-12-13 2018-11-14 The General Hospital Corporation Soluble high molecular weight (hmw) tau species and applications thereof
JP2017500319A (en) 2013-12-20 2017-01-05 ギリアード カリストガ エルエルシー Polymorphic form of the hydrochloride salt of (S) -2- (1- (9H-purin-6-ylamino) propyl) -5-fluoro-3-phenylquinazolin-4 (3H) -one
WO2015095601A1 (en) 2013-12-20 2015-06-25 Gilead Calistoga Llc Process methods for phosphatidylinositol 3-kinase inhibitors
CA2936940C (en) 2014-01-14 2023-07-04 The Johns Hopkins University Cyclodextrin compositions encapsulating a selective atp inhibitor and uses thereof
DK3628328T3 (en) 2014-01-31 2022-12-05 Cytomx Therapeutics Inc MATRIPTASE AND U PLASMINOGEN ACTIVATOR SUBSTRATES AND OTHER CLEAVABLE PARTS AND METHODS OF USING THEREOF
WO2015116902A1 (en) 2014-01-31 2015-08-06 Genentech, Inc. G-protein coupled receptors in hedgehog signaling
AU2015315834B2 (en) 2014-01-31 2019-12-12 Boehringer Ingelheim International Gmbh Novel anti-BAFF antibodies
US20170044232A1 (en) 2014-02-04 2017-02-16 Genentech, Inc. Mutant smoothened and methods of using the same
EP3450571B1 (en) 2014-02-24 2023-04-05 Celgene Corporation Methods of using an activator of cereblon for neural cell expansion and the treatment of central nervous system disorders
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
NZ711451A (en) 2014-03-07 2016-05-27 Alexion Pharma Inc Anti-c5 antibodies having improved pharmacokinetics
WO2015143194A2 (en) 2014-03-19 2015-09-24 Dana-Farber Cancer Institute, Inc. Immunogenetic restriction on elicitation of antibodies
EP4324481A2 (en) 2014-03-21 2024-02-21 Teva Pharmaceuticals International GmbH Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
RU2016136716A (en) 2014-03-27 2018-04-28 Дженентек, Инк. METHODS FOR DIAGNOSTIC AND TREATMENT OF INFLAMMATORY DISEASES OF THE GUT
JP2017513931A (en) 2014-04-03 2017-06-01 インビクタス オンコロジー ピーヴィティー.リミテッド Supramolecular combinatorial treatment
KR20160145813A (en) 2014-04-25 2016-12-20 다나-파버 캔서 인스티튜트 인크. Middle east respiratory syndrome coronavirus neutralizing antibodies and methods of use thereof
US10308697B2 (en) 2014-04-30 2019-06-04 President And Fellows Of Harvard College Fusion proteins for treating cancer and related methods
SG11201608953WA (en) 2014-04-30 2016-11-29 Pfizer Anti-ptk7 antibody-drug conjugates
WO2015171918A2 (en) 2014-05-07 2015-11-12 Louisiana State University And Agricultural And Mechanical College Compositions and uses for treatment thereof
JP6868394B2 (en) 2014-05-16 2021-05-12 ファイザー・インク Bispecific antibody
EA028614B1 (en) 2014-05-22 2017-12-29 Общество С Ограниченной Ответственностью "Русские Фармацевтические Технологии" Selective inhibitors interfering with fibroblast growth factor receptor and frs2 interaction for the prevention and treatment of cancer
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
CN106714830B (en) 2014-05-30 2020-08-25 上海复宏汉霖生物技术股份有限公司 anti-Epidermal Growth Factor Receptor (EGFR) antibodies
WO2015191781A2 (en) 2014-06-10 2015-12-17 Amgen Inc. Apelin polypeptides
JP6353082B2 (en) 2014-06-11 2018-07-04 ギリアード サイエンシーズ, インコーポレイテッド Methods for treating cardiovascular disease
BR112016028642A2 (en) 2014-06-13 2017-08-22 Gilead Sciences Inc compound, pharmaceutical composition, method of treating a disease or condition, method of inhibiting the activity of a phosphatidylinositol 3-kinase polypeptide, method of inhibiting immune reactions or overgrowth or destructive cancer cell proliferation, kit, and, use of a compound, salt, isomer or a pharmaceutically acceptable mixture thereof.
TWI695011B (en) 2014-06-18 2020-06-01 美商梅爾莎納醫療公司 Monoclonal antibodies against her2 epitope and methods of use thereof
WO2015196070A1 (en) 2014-06-20 2015-12-23 Genentech, Inc. Chagasin-based scaffold compositions, methods, and uses
WO2015200901A1 (en) 2014-06-26 2015-12-30 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
EP3171896A4 (en) 2014-07-23 2018-03-21 Mayo Foundation for Medical Education and Research Targeting dna-pkcs and b7-h1 to treat cancer
EP3172235A2 (en) 2014-07-25 2017-05-31 Cytomx Therapeutics Inc. Anti-cd3 antibodies, activatable anti-cd3 antibodies, multispecific anti-cd3 antibodies, multispecific activatable anti-cd3 antibodies, and methods of using the same
AR101669A1 (en) 2014-07-31 2017-01-04 Amgen Res (Munich) Gmbh ANTIBODY CONSTRUCTS FOR CDH19 AND CD3
MX2017001401A (en) 2014-07-31 2017-08-07 Amgen Res (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody constructs.
WO2016020799A1 (en) 2014-08-06 2016-02-11 Rinat Neuroscience Corp. Methods for reducing ldl-cholesterol
CA2956991A1 (en) 2014-08-06 2016-02-11 Rinat Neuroscience Corp. Methods for reducing ldl-cholesterol
WO2016034968A1 (en) 2014-09-02 2016-03-10 Pfizer Inc. Therapeutic antibody
US10472424B2 (en) 2014-09-23 2019-11-12 Pfizer Inc. Treatment with anti-PCSK9 antibodies
BR112017006825A2 (en) 2014-10-03 2017-12-12 Dana Farber Cancer Inst Inc antibodies to glucocorticoid-induced tumor necrosis factor receptor (gitr) and their methods of use
CA2962949C (en) 2014-10-06 2024-03-05 Dana-Farber Cancer Institute, Inc. Humanized cc chemokine receptor 4 (ccr4) antibodies and methods of use thereof
GB201417589D0 (en) 2014-10-06 2014-11-19 Cantab Biopharmaceuticals Patents Ltd Pharmaceutical Formulations
EP3212233B1 (en) 2014-10-31 2020-06-24 Oncomed Pharmaceuticals, Inc. Combination therapy for treatment of disease
US9879087B2 (en) 2014-11-12 2018-01-30 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
WO2016077566A1 (en) 2014-11-12 2016-05-19 Research Institute At Nationwide Children's Hospital Modulation of alternative mdm2 splicing
US20170306046A1 (en) 2014-11-12 2017-10-26 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
JP7175608B2 (en) 2014-11-19 2022-11-21 ザ トラスティーズ オブ コロンビア ユニバーシティ イン ザ シティ オブ ニューヨーク Osteocalcin as a treatment for age-related frailty
MX2017007055A (en) 2014-12-03 2017-11-08 Genentech Inc Anti-staphylococcus aureus antibody rifamycin conjugates and uses thereof.
BR112017011478A2 (en) 2014-12-03 2018-02-27 Genentech, Inc. antibody-antibiotic conjugate compound, antibody-antibiotic conjugate, pharmaceutical composition, methods of treating a bacterial infection and exterminating staph aureus, process for compound preparation, kit and antibiotic-binder intermediate
US10072070B2 (en) 2014-12-05 2018-09-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Potent anti-influenza A neuraminidase subtype N1 antibody
TWI595006B (en) 2014-12-09 2017-08-11 禮納特神經系統科學公司 Anti-pd-1 antibodies and methods of use thereof
ES2934940T3 (en) 2014-12-11 2023-02-28 Pf Medicament Anti-C10orf54 antibodies and uses thereof
BR112017012706A2 (en) 2014-12-15 2018-03-13 The Johns Hopkins University sunitinib formulations and methods for their use in the treatment of eye disorders
MA41374A (en) 2015-01-20 2017-11-28 Cytomx Therapeutics Inc MATRIX METALLOPROTEASE CLIVABLE AND SERINE PROTEASE CLIVABLE SUBSTRATES AND METHODS OF USE THEREOF
AU2016211696B2 (en) 2015-01-27 2018-05-10 The Johns Hopkins University Hypotonic hydrogel formulations for enhanced transport of active agents at mucosal surfaces
EP3250927B1 (en) 2015-01-28 2020-02-19 H. Hoffnabb-La Roche Ag Gene expression markers and treatment of multiple sclerosis
US20180215801A1 (en) 2015-01-29 2018-08-02 Board Of Trustees Of Michigan State University Cryptic polypeptides and uses thereof
US20160250177A1 (en) * 2015-02-17 2016-09-01 Mallinckrodt Llc Modified docetaxel liposome formulations and uses thereof
US10550173B2 (en) 2015-02-19 2020-02-04 Compugen, Ltd. PVRIG polypeptides and methods of treatment
SI3258951T1 (en) 2015-02-19 2020-07-31 Compugen Ltd. Anti-pvrig antibodies and methods of use
JP2018507868A (en) 2015-02-26 2018-03-22 ジェネンテック, インコーポレイテッド Integrin beta 7 antagonist and method of treating Crohn's disease
HUE061070T2 (en) 2015-03-03 2023-05-28 Kymab Ltd Antibodies, uses & methods
EP3265063A4 (en) * 2015-03-03 2018-11-07 Cureport, Inc. Dual loaded liposomal pharmaceutical formulations
WO2016141167A1 (en) 2015-03-03 2016-09-09 Cureport, Inc. Combination liposomal pharmaceutical formulations
JP6826055B2 (en) 2015-03-13 2021-02-03 サイトメックス セラピューティクス インコーポレイテッド Anti-PDL1 antibody, activating anti-PDL1 antibody, and how to use it
JP6859275B2 (en) 2015-03-17 2021-04-14 リポメディックス・ファーマシューティカルズ・リミテッドLipomedix Pharmaceuticals Ltd. How to treat bladder cancer
MA41795A (en) 2015-03-18 2018-01-23 Sarepta Therapeutics Inc EXCLUSION OF AN EXON INDUCED BY ANTISENSE COMPOUNDS IN MYOSTATIN
US10174292B2 (en) 2015-03-20 2019-01-08 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
US9931394B2 (en) 2015-03-23 2018-04-03 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
US9758575B2 (en) 2015-04-06 2017-09-12 Yung Shin Pharmaceutical Industrial Co. Ltd. Antibodies which specifically bind to canine vascular endothelial growth factor and uses thereof
WO2016164835A1 (en) 2015-04-08 2016-10-13 Dana-Farber Cancer Institute, Inc. Humanized influenza monoclonal antibodies and methods of use thereof
CN108136002B (en) 2015-04-13 2022-05-31 辉瑞公司 Therapeutic antibodies and their use
JP6907124B2 (en) 2015-04-17 2021-07-21 アムゲン リサーチ (ミュンヘン) ゲーエムベーハーAMGEN Research(Munich)GmbH Bispecific antibody construct against CDH3 and CD3
US10556956B2 (en) 2015-05-01 2020-02-11 Dana-Farber Cancer Institute, Inc. Pharmaceutical compositions comprising humanized anti-CCR4 IgG4 antibody
CN107922490A (en) 2015-05-04 2018-04-17 西托姆克斯治疗公司 Anti- ITGa3 antibody, can activate anti-ITGa3 antibody and its application method
EP3292150B1 (en) 2015-05-04 2020-02-05 Cytomx Therapeutics Inc. Activatable anti-cd166 antibodies, and methods of use thereof
MY194184A (en) 2015-05-04 2022-11-17 Cytomx Therapeutics Inc Anti-cd71 antibodies, activatable anti-cd71 antibodies, and methods of use thereof
EP3736287A1 (en) 2015-05-11 2020-11-11 The Johns Hopkins University Autoimmune antibodies for use in inhibiting cancer cell growth
WO2016180986A1 (en) 2015-05-14 2016-11-17 Fundación Centro Nacional De Investigaciones Cardiovasculares Carlos Iii (Cnic) Mirna compositions for the treatment of mature b-cell neoplasms
US11318131B2 (en) 2015-05-18 2022-05-03 Ipsen Biopharm Ltd. Nanoliposomal irinotecan for use in treating small cell lung cancer
AR105618A1 (en) 2015-05-29 2017-10-25 Genentech Inc METHODATION OF THE PROMOTER OF THE BINDING TO THE PROGRAMMED DEATH RECEIVER (PD-L1) IN CANCER
EP3302497A4 (en) 2015-06-01 2019-01-16 Sarepta Therapeutics, Inc. Antisense-induced exon exclusion in type vii collagen
EP3307754B1 (en) 2015-06-10 2022-09-14 Elysium Health, Inc. Nicotinamide riboside and pterostilbene compositions and methods for treatment of skin disorders
JP7014706B2 (en) 2015-07-13 2022-02-01 サイトメックス セラピューティクス インコーポレイテッド Anti-PD-1 antibody, activating anti-PD-1 antibody, and how to use it
CA2991980A1 (en) 2015-07-13 2017-01-19 Compugen Ltd. Hide1 compositions and methods
WO2017015431A1 (en) 2015-07-21 2017-01-26 Dyax Corp. A monoclonal antibody inhibitor of factor xiia
JP6916776B2 (en) 2015-07-29 2021-08-11 アラーガン、インコーポレイテッドAllergan,Incorporated Heavy chain only antibody against ANG-2
TWI717375B (en) 2015-07-31 2021-02-01 德商安美基研究(慕尼黑)公司 Antibody constructs for cd70 and cd3
TWI796283B (en) 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Antibody constructs for msln and cd3
TWI793062B (en) 2015-07-31 2023-02-21 德商安美基研究(慕尼黑)公司 Antibody constructs for dll3 and cd3
JO3792B1 (en) 2015-07-31 2021-01-31 Amgen Res Munich Gmbh Antibody constructs for flt3 and cd3
TWI744242B (en) 2015-07-31 2021-11-01 德商安美基研究(慕尼黑)公司 Antibody constructs for egfrviii and cd3
US10308711B2 (en) 2015-08-14 2019-06-04 Allergan, Inc. Heavy chain only antibodies to PDGF
MX2018001659A (en) 2015-08-20 2018-05-28 Ipsen Biopharm Ltd Combination therapy using liposomal irinotecan and a parp inhibitor for cancer treatment.
EP3337478B1 (en) 2015-08-21 2020-08-12 Ipsen Biopharm Ltd. Drug combination comprising liposomal irinotecan, oxaliplatin, 5-fluorouracil and leucovorin for treating metastatic pancreatic cancer
PL3344655T3 (en) 2015-09-01 2024-02-19 Boehringer Ingelheim International Gmbh Use of anti-cd40 antibodies for treatment of lupus nephritis
TWI799366B (en) 2015-09-15 2023-04-21 美商建南德克公司 Cystine knot scaffold platform
MX2018003196A (en) 2015-09-15 2019-05-16 Scholar Rock Inc Anti-pro/latent-myostatin antibodies and uses thereof.
AU2016326609B2 (en) 2015-09-23 2023-03-09 Mereo Biopharma 5, Inc. Methods and compositions for treatment of cancer
WO2017058881A1 (en) 2015-09-28 2017-04-06 The Trustees Of Columbia University In The City Of New York Use of pentoxifylline with immune checkpoint-blockade therapies for the treatment of melanoma
WO2017055966A1 (en) 2015-10-01 2017-04-06 Pfizer Inc. Low viscosity antibody compositions
JP2018529747A (en) 2015-10-06 2018-10-11 ジェネンテック, インコーポレイテッド Methods for treating multiple sclerosis
EP3858993A1 (en) 2015-10-09 2021-08-04 Sarepta Therapeutics, Inc. Compositions and methods for treating duchenne muscular dystrophy and related disorders
GB201518170D0 (en) 2015-10-14 2015-11-25 Cantab Biopharmaceuticals Patents Ltd Colloidal particles for subcutaneous administration with intravenous administration of therapeutic agent
GB201518172D0 (en) 2015-10-14 2015-11-25 Cantab Biopharmaceuticals Patents Ltd Colloidal particles for use in medicine
GB201518171D0 (en) 2015-10-14 2015-11-25 Cantab Biopharmaceuticals Patents Ltd Colloidal particles for topical administration with therapeutic agent
WO2017066714A1 (en) 2015-10-16 2017-04-20 Compugen Ltd. Anti-vsig1 antibodies and drug conjugates
BR122021024957B1 (en) 2015-10-16 2023-12-12 Ipsen Biopharm Ltd Processes for producing a storage-stabilized liposomal irinotecan composition
WO2017075037A1 (en) 2015-10-27 2017-05-04 Scholar Rock, Inc. Primed growth factors and uses thereof
AU2016344399B2 (en) 2015-10-30 2019-03-28 Genentech, Inc. Anti-HtrA1 antibodies and methods of use thereof
WO2017075045A2 (en) 2015-10-30 2017-05-04 Mayo Foundation For Medical Education And Research Antibodies to b7-h1
WO2017075670A1 (en) 2015-11-05 2017-05-11 Children's Hospital Los Angeles "mobilizing leukemia cells"
WO2017079563A1 (en) 2015-11-06 2017-05-11 The Johns Hopkins University Methods of treating liver fibrosis by administering 3-bromopyruvate
WO2017083403A1 (en) 2015-11-10 2017-05-18 Children's Research Institute, Children's National Medical Center Echinomycin formulation, method of making and method of use thereof
IL302822A (en) 2015-11-12 2023-07-01 Seagen Inc Glycan-interacting compounds and methods of use
EP3373978A4 (en) 2015-11-12 2019-06-26 Graybug Vision, Inc. Aggregating microparticles for therapy
TWI812873B (en) 2015-11-30 2023-08-21 美商輝瑞股份有限公司 Antibodies and antibody fragments for site-specific conjugation
TWI778491B (en) 2015-11-30 2022-09-21 美商輝瑞股份有限公司 Site specific her2 antibody drug conjugates
MX2018006613A (en) 2015-12-02 2019-01-30 Stcube & Co Inc Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof.
KR20180100122A (en) 2015-12-02 2018-09-07 주식회사 에스티사이언스 Antibodies specific for glycated BTLA (B- and T-lymphocyte weakening factor)
MA43415A (en) 2015-12-09 2018-10-17 Modernatx Inc MODIFIED RNA CODING FOR A URIDINE DIPHOSPHATE GLUCURONOSYLTRANSFERASE AND ASSOCIATED USES
WO2017105990A1 (en) 2015-12-14 2017-06-22 Massachusetts Institute Of Technology Ph-responsive mucoadhesive polymeric encapsulated microorganisms
EP3389720A1 (en) 2015-12-18 2018-10-24 The General Hospital Corporation Polyacetal polymers, conjugates, particles and uses thereof
WO2017110772A1 (en) 2015-12-21 2017-06-29 富士フイルム株式会社 Liposome and liposome composition
EP4310503A3 (en) 2015-12-30 2024-03-20 Momenta Pharmaceuticals, Inc. Methods related to biologics
SG11201805709RA (en) 2016-01-08 2018-07-30 Scholar Rock Inc Anti-pro/latent myostatin antibodies and methods of use thereof
EP3405579A1 (en) 2016-01-22 2018-11-28 Modernatx, Inc. Messenger ribonucleic acids for the production of intracellular binding polypeptides and methods of use thereof
JOP20170017B1 (en) 2016-01-25 2021-08-17 Amgen Res Munich Gmbh Pharmaceutical composition comprising bispecific antibody constructs
HRP20220081T1 (en) 2016-02-03 2022-04-15 Amgen Research (Munich) Gmbh Bcma and cd3 bispecific t cell engaging antibody constructs
ES2935419T3 (en) 2016-02-03 2023-03-06 Amgen Res Munich Gmbh Bispecific antibody constructs for PSMA and CD3 that bind to T cells
EA039859B1 (en) 2016-02-03 2022-03-21 Эмджен Рисерч (Мюник) Гмбх Bispecific antibody constructs binding egfrviii and cd3
JP2019509721A (en) 2016-02-04 2019-04-11 キュリス,インコーポレイテッド Mutant smoothened and method of using the same
CA3015663A1 (en) * 2016-03-07 2017-09-14 Memorial Sloan Kettering Cancer Center Bone marrow-, reticuloendothelial system-, and/or lymph node-targeted radiolabeled liposomes and methods of their diagnostic and therapeutic use
US11357849B2 (en) 2016-03-07 2022-06-14 Musc Foundation For Research Development Anti-nucleolin antibodies
JP2019509737A (en) 2016-03-11 2019-04-11 スカラー ロック インコーポレイテッドScholar Rock,Inc. TGFβ1-binding immunoglobulin and use thereof
EP3430058A4 (en) 2016-03-15 2019-10-23 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
EA201892366A1 (en) 2016-04-18 2019-03-29 Сарепта Терапьютикс, Инк. ANTISMINAL OLIGOMERS AND METHODS OF THEIR APPLICATION FOR THE TREATMENT OF DISEASES ASSOCIATED WITH THE GENE OF THE ACID ALPHA-GLUCOSIDASE
US10973748B2 (en) 2016-04-21 2021-04-13 Versitech Limited Compositions and methods for lightening skin and reducing hyperpigmentation
CA3026880A1 (en) 2016-06-08 2017-12-14 Paul Foster Treatment of igg4-related diseases with anti-cd19 antibodies crossbinding to cd32b
ITUA20164630A1 (en) 2016-06-23 2017-12-23 Paolo Blasi PHARMACOLOGICAL ADIUVANTS FOR TUMOR THERMAL WELDING
EP3474886B1 (en) 2016-06-27 2021-08-04 Alexion Pharmaceuticals, Inc. Methods for treating hypophosphatasia in children and adolescents
WO2018005657A1 (en) 2016-06-28 2018-01-04 Verily Life Sciences Llc Serial filtration to generate small cholesterol-containing liposomes
JP7082967B2 (en) 2016-07-22 2022-06-09 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Glucocorticoid-induced tumor necrosis factor receptor (GITR) antibody and its usage
CN109862896A (en) 2016-08-03 2019-06-07 西玛贝医药公司 For treating the Oxymethylene aryl compound of inflammatory gastrointestinal disease or gastrointestinal disorder
TWI754659B (en) 2016-08-08 2022-02-11 台灣微脂體股份有限公司 Delivery vehicle, method and kit for preparing a liposomal composition containing mild acidic agent
WO2018045379A1 (en) 2016-09-02 2018-03-08 Dana-Farber Cancer Institute, Inc. Composition and methods of treating b cell disorders
BR112019004214A2 (en) 2016-09-06 2019-05-28 Dana Farber Cancer Inst Inc Methods to Treat or Prevent Zika Virus Infection
JP2019534858A (en) 2016-09-09 2019-12-05 ジェネンテック, インコーポレイテッド Selective peptide inhibitor of FRIZZLED
TW201825674A (en) 2016-09-09 2018-07-16 美商艾斯合顧問有限公司 Oncolytic virus expressing bispecific engager molecules
US11236177B2 (en) 2016-09-14 2022-02-01 The Trustees Of The University Of Pennsylvania Proximity-based sortase-mediated protein purification and ligation
KR102391338B1 (en) 2016-09-16 2022-04-26 상하이 헨리우스 바이오테크, 인크. anti-PD-1 antibody
WO2018055574A1 (en) 2016-09-23 2018-03-29 Teva Pharmaceuticals International Gmbh Treating refractory migraine
AU2017331592A1 (en) 2016-09-23 2019-04-11 Teva Pharmaceuticals International Gmbh Treating cluster headache
SG11201903674YA (en) 2016-10-26 2019-05-30 Modernatx Inc Messenger ribonucleic acids for enhancing immune responses and methods of use thereof
BR112019007844A2 (en) 2016-11-02 2019-07-16 Ipsen Biopharm Ltd treatment of gastric cancer using combination therapies comprising liposomal irinotecan, oxaliplatin, 5-fluoroacyl (and leucovorin)
WO2018083248A1 (en) 2016-11-03 2018-05-11 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses & methods
JP2020503256A (en) 2016-11-04 2020-01-30 ノビミューン エスアー Anti-CD19 antibodies and methods of using them
US11339209B2 (en) 2016-11-14 2022-05-24 Novartis Ag Compositions, methods, and therapeutic uses related to fusogenic protein minion
WO2018094143A1 (en) 2016-11-17 2018-05-24 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
WO2018106738A1 (en) 2016-12-05 2018-06-14 Massachusetts Institute Of Technology Brush-arm star polymers, conjugates and particles, and uses thereof
US10772926B2 (en) 2016-12-16 2020-09-15 Nutragen Health Innovations, Inc. Natural drugs for the treatment of inflammation and melanoma
RS63849B9 (en) 2016-12-16 2023-06-30 Pfizer Glp-1 receptor agonists and uses thereof
CN110382530A (en) 2017-01-06 2019-10-25 供石公司 Hypospecificity, 1 inhibitor of background permissive TGF β and application thereof
JP7198757B2 (en) 2017-01-06 2023-01-04 スカラー ロック インコーポレイテッド Methods for treating metabolic disorders by inhibiting myostatin activation
WO2018129395A1 (en) 2017-01-06 2018-07-12 Scholar Rock, Inc. Methods for treating metabolic diseases by inhibiting myostatin activation
CN110430894A (en) 2017-02-01 2019-11-08 莫得纳特斯公司 The immune modulating treatment MRNA composition of encoding activating oncogenic mutation peptide
JOP20190189A1 (en) 2017-02-02 2019-08-01 Amgen Res Munich Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
WO2018152496A1 (en) 2017-02-17 2018-08-23 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Compositions and methods for the diagnosis and treatment of zika virus infection
US11198735B2 (en) 2017-03-03 2021-12-14 Rinat Neuroscience Corp. Anti-GITR antibodies and methods of use thereof
JP2020510671A (en) 2017-03-03 2020-04-09 シアトル ジェネティックス, インコーポレイテッド Glycan interacting compounds and methods of use
CA3055574A1 (en) 2017-03-09 2018-09-13 Cytomx Therapeutics, Inc. Cd147 antibodies, activatable cd147 antibodies, and methods of making and use thereof
WO2018183173A1 (en) 2017-03-27 2018-10-04 Boehringer Ingelheim International Gmbh Anti il-36r antibodies combination therapy
US11359200B2 (en) 2017-04-09 2022-06-14 The Cleveland Clinic Foundation Cancer treatment by MALAT1 inhibition
CA3059542A1 (en) 2017-04-12 2018-10-18 Pfizer Inc. Antibodies having conditional affinity and methods of use thereof
AU2018250695A1 (en) 2017-04-14 2019-11-07 Kodiak Sciences Inc. Complement factor D antagonist antibodies and conjugates thereof
US11866506B2 (en) 2017-04-21 2024-01-09 Mellitus, Llc Anti-CD59 antibodies
US11236151B2 (en) 2017-04-25 2022-02-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibodies and methods for the diagnosis and treatment of Epstein Barr virus infection
KR20190141243A (en) 2017-05-05 2019-12-23 암젠 인크 Pharmaceutical compositions comprising bispecific antibody constructs for improved storage and administration
US11318190B2 (en) 2017-05-05 2022-05-03 United States Government As Represented By The Department Of Veterans Affairs Methods and compositions for treating liver disease
RU2019139817A (en) 2017-05-10 2021-06-10 Грейбуг Вижн, Инк. DELAYED RELEASE MICROPARTICLES AND THEIR SUSPENSIONS FOR DRUG THERAPY
KR20200006985A (en) 2017-05-12 2020-01-21 오거스타 유니버시티 리서치 인스티튜트, 인크. Human alpha fetal protein-specific T cell receptors and uses thereof
US10994025B2 (en) 2017-05-12 2021-05-04 Massachusetts Institute Of Technology Argonaute protein-double stranded RNA complexes and uses related thereto
CA3065008A1 (en) 2017-05-26 2018-11-29 Novimmune Sa Anti-cd47 x anti-mesothelin antibodies and methods of use thereof
EP3630835A1 (en) 2017-05-31 2020-04-08 STCube & Co., Inc. Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof
KR20200024158A (en) 2017-05-31 2020-03-06 주식회사 에스티큐브앤컴퍼니 How to treat cancer using antibodies and molecules that immunospecifically bind to BTN1A1
US11168144B2 (en) 2017-06-01 2021-11-09 Cytomx Therapeutics, Inc. Activatable anti-PDL1 antibodies, and methods of use thereof
KR20200026209A (en) 2017-06-06 2020-03-10 주식회사 에스티큐브앤컴퍼니 How to treat cancer using antibodies and molecules that bind BTN1A1 or BTN1A1-ligand
GB201710838D0 (en) 2017-07-05 2017-08-16 Ucl Business Plc Bispecific antibodies
AU2018301442A1 (en) 2017-07-13 2020-01-30 Massachusetts Institute Of Technology Targeting the HDAC2-Sp3 complex to enhance synaptic function
WO2019014586A1 (en) 2017-07-14 2019-01-17 Cytomx Therapeutics, Inc. Anti-cd166 antibodies and uses thereof
WO2019018629A1 (en) 2017-07-19 2019-01-24 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Antibodies and methods for the diagnosis and treatment of hepatitis b virus infection
EP3655779A1 (en) 2017-07-20 2020-05-27 CytomX Therapeutics, Inc. Methods of qualitatively and/or quantitatively analyzing properties of activatable antibodies and uses thereof
MA49634A (en) 2017-07-21 2020-05-27 Modernatx Inc MODIFIED RNA CODING FOR A PROPIONYL-COA-CARBOXYLASE AND ASSOCIATED USES
WO2019016784A1 (en) 2017-07-21 2019-01-24 Universidade De Coimbra Anti-nucleolin antibody
US20200179494A1 (en) 2017-07-24 2020-06-11 Modernatx, Inc. Modified mRNA Encoding a Glucose 6 Phosphatase and Uses Thereof
KR20230066126A (en) 2017-07-27 2023-05-12 알렉시온 파마슈티칼스, 인코포레이티드 High concentration anti-c5 antibody formulations
EP3658583A1 (en) 2017-07-28 2020-06-03 Scholar Rock, Inc. Ltbp complex-specific inhibitors of tgf-beta 1 and uses thereof
US11135187B2 (en) 2017-08-22 2021-10-05 National Institutes Of Health (Nih) Compositions and methods for treating diabetic retinopathy
BR112020004231A2 (en) 2017-08-30 2020-09-08 Cytomx Therapeutics, Inc. activable anti-cd166 antibodies and methods of using them
AU2018330180A1 (en) 2017-09-07 2020-03-19 Augusta University Research Institute, Inc. Antibodies to programmed cell death protein 1
US11364303B2 (en) 2017-09-29 2022-06-21 Pfizer Inc. Cysteine engineered antibody drug conjugates
CA3078974A1 (en) 2017-10-12 2019-04-18 Immunowake Inc. Vegfr-antibody light chain fusion protein
WO2019075405A1 (en) 2017-10-14 2019-04-18 Cytomx Therapeutics, Inc. Antibodies, activatable antibodies, bispecific antibodies, and bispecific activatable antibodies and methods of use thereof
JP2020537655A (en) 2017-10-17 2020-12-24 ルヒゼン ファーマスティカルズ エスエー CRAC channel modulator for the treatment of esophageal cancer
US11555189B2 (en) 2017-10-18 2023-01-17 Sarepta Therapeutics, Inc. Antisense oligomer compounds
WO2019082124A1 (en) 2017-10-26 2019-05-02 Rhizen Pharmaceuticals Sa Composition and method for treating diffuse large b-cell lymphoma
PE20201255A1 (en) 2017-10-27 2020-11-16 Pfizer CD123 ANTIBODIES AND ANTIBODY-DRUG CONJUGATES SPECIFIC TO CD123 AND USES THEREOF
BR112020008219A2 (en) 2017-10-30 2020-10-27 Rhizen Pharmaceuticals Sa calcium channel modulators activated by calcium release to treat hematological and solid cancers
WO2019086626A1 (en) 2017-11-03 2019-05-09 Centro Nacional De Investigaciones Cardiovasculares Carlos Iii (F.S.P.) MIRNAs AND COMBINATIONS THEREOF FOR USE IN THE TREATMENT OF HUMAN B CELL NEOPLASIAS
US20200289520A1 (en) 2017-12-06 2020-09-17 Rhizen Pharmaceuticals Sa Composition and method for treating peripheral t-cell lymphoma and cutaneous t-cell lymphoma
JP7344206B2 (en) 2017-12-11 2023-09-13 アムジェン インコーポレイテッド Continuous manufacturing process for bispecific antibody products
UY38041A (en) 2017-12-29 2019-06-28 Amgen Inc CONSTRUCTION OF BIESPECFIC ANTIBODY DIRECTED TO MUC17 AND CD3
WO2019165101A1 (en) 2018-02-22 2019-08-29 Verily Life Sciences Llc Combining orthogonal chemistries for preparation of multiplexed nanoparticles
CA3034912A1 (en) 2018-02-28 2019-08-28 Pfizer Inc. Il-15 variants and uses thereof
EP3762420A1 (en) 2018-03-09 2021-01-13 CytomX Therapeutics, Inc. Activatable cd147 antibodies and methods of making and use thereof
AU2019235523A1 (en) 2018-03-14 2020-10-29 Novimmune Sa Anti-CD3 epsilon antibodies and methods of use thereof
SG11202006217YA (en) 2018-03-14 2020-07-29 Beijing Xuanyi Pharmasciences Co Ltd Anti-claudin 18.2 antibodies
TW202003020A (en) 2018-03-21 2020-01-16 美國科羅拉多州立大學研究基金會 Cancer vaccine compositions and methods of use thereof
US10722528B2 (en) 2018-03-28 2020-07-28 Augusta University Research Institute, Inc. Compositions and methods for inhibiting metastasis
WO2019195561A2 (en) 2018-04-06 2019-10-10 BioLegend, Inc. Anti-tetraspanin 33 agents and compositions and methods for making and using the same
KR20200141470A (en) 2018-04-06 2020-12-18 칠드런'즈 메디컬 센터 코포레이션 Composition and method for adjusting somatic cell reprogramming and imprinting
WO2019197607A1 (en) 2018-04-13 2019-10-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Fc-engineered anti-human ige antibodies and methods of use
EP3788071A1 (en) 2018-05-02 2021-03-10 The United States Of America, As Represented By The Secretary, Department of Health and Human Services Antibodies and methods for the diagnosis, prevention, and treatment of epstein barr virus infection
US11434292B2 (en) 2018-05-23 2022-09-06 Pfizer Inc. Antibodies specific for CD3 and uses thereof
MX2020012607A (en) 2018-05-23 2021-01-29 Pfizer Antibodies specific for gucy2c and uses thereof.
US20210196823A1 (en) 2018-06-08 2021-07-01 Pfizer Inc. Methods of Treating Metabolic Disease
CA3045644C (en) 2018-06-13 2024-01-16 Pfizer Inc. Glp-1 receptor agonists and uses thereof
ES2943510T3 (en) 2018-06-15 2023-06-13 Pfizer GLP-1 receptor agonists and uses thereof
CN112533629A (en) 2018-06-19 2021-03-19 阿尔莫生物科技股份有限公司 Compositions and methods for combined use of IL-10 agents with chimeric antigen receptor cell therapy
KR20210027389A (en) 2018-06-28 2021-03-10 크리스퍼 테라퓨틱스 아게 Compositions and methods for genome editing by insertion of donor polynucleotides
AU2019291890A1 (en) 2018-06-29 2020-12-17 Boehringer Ingelheim International Gmbh Anti-CD40 antibodies for use in treating autoimmune disease
CA3105161C (en) 2018-07-06 2022-11-15 Pfizer Inc. Manufacturing process and intermediates for a pyrrolo[2,3- d]pyrimidine compound and use thereof
WO2020014460A1 (en) 2018-07-11 2020-01-16 Scholar Rock, Inc. HIGH-AFFINITY, ISOFORM-SELECTIVE TGFβ1 INHIBITORS AND USE THEREOF
WO2020014473A1 (en) 2018-07-11 2020-01-16 Scholar Rock, Inc. TGFβ1 INHIBITORS AND USE THEREOF
MA52165A (en) 2018-07-11 2021-06-02 Scholar Rock Inc SELECTIVE TGFBETA1 ISOFORM INHIBITORS AND ASSOCIATED USE
JP2021531007A (en) 2018-07-20 2021-11-18 ピエール、ファーブル、メディカマン Receptor for VISTA
US20220023450A1 (en) * 2018-09-11 2022-01-27 Memorial Sloan Kettering Cancer Center Bone marrow-, reticuloendothelial system-, and/or lymph node-targeted radiolabeled liposomes and methods of their diagnostic and therapeutic use
EP3856343A1 (en) 2018-09-25 2021-08-04 Biolegend, Inc. Anti-tlr9 agents and compositions and methods for making and using the same
WO2020065584A1 (en) 2018-09-27 2020-04-02 Phosphogam, Inc. Methods and compositions for the expansion and use of allogeneic gamma/delta-t cells
CA3114567A1 (en) 2018-09-28 2020-04-02 Lyvgen Biopharma Co., Ltd. Anti-cd40 binding molecules having engineered fc domains and therapeutic uses thereof
WO2020076776A1 (en) 2018-10-10 2020-04-16 Boehringer Ingelheim International Gmbh Method for membrane gas transfer in high density bioreactor culture
CN112789058A (en) 2018-10-11 2021-05-11 安进公司 Downstream processing of bispecific antibody constructs
WO2020079652A1 (en) 2018-10-17 2020-04-23 Insilico Medicine Hong Kong Limited Kinase inhibitors
MX2021004598A (en) 2018-10-23 2021-06-15 Scholar Rock Inc Rgmc-selective inhibitors and use thereof.
EP3870352A2 (en) * 2018-10-26 2021-09-01 University of Connecticut A continuous processing system and methods for internal and external modifications to nanoparticles
CA3118445A1 (en) 2018-11-02 2020-05-07 Cytomx Therapeutics, Inc. Activatable anti-cd166 antibodies and methods of use thereof
EP3883928A4 (en) 2018-11-22 2022-06-29 Qilu Regor Therapeutics Inc. Glp-1r agonists and uses thereof
KR20210102318A (en) 2018-12-06 2021-08-19 싸이톰스 테라퓨틱스, 인크. Matrix metalloprotease-cleavable substrates and serine or cysteine protease-cleavable substrates and methods of use thereof
CA3177829A1 (en) 2018-12-12 2020-06-18 Kite Pharma, Inc. Chimeric antigen and t cell receptors and methods of use
CA3127452A1 (en) 2019-01-22 2020-07-30 Massachusetts Institute Of Technology In vitro human blood brain barrier
JP2020117502A (en) 2019-01-28 2020-08-06 ファイザー・インク Method of treating signs and symptoms of osteoarthritis
AU2020216177A1 (en) 2019-01-30 2021-09-23 Scholar Rock, Inc. LTBP complex-specific inhibitors of TGFβ and uses thereof
TWI820301B (en) 2019-02-15 2023-11-01 美商輝瑞股份有限公司 Crystalline pyrimidinyl-3,8-diazabicyclo[3.2.1] octanylmethanone compound and use thereof
US20220135663A1 (en) 2019-02-18 2022-05-05 Pfizer Inc. Method of treatment of Chronic Low Back Pain
CN113874394B (en) 2019-02-20 2024-01-19 和铂抗体有限公司 Antibodies to
US11795160B2 (en) 2019-02-22 2023-10-24 Insilico Medicine Ip Limited Kinase inhibitors
CN113677372A (en) 2019-02-26 2021-11-19 西托姆克斯治疗公司 Combination therapy of activatable immune checkpoint inhibitors and conjugated activatable antibodies
US20200283796A1 (en) 2019-03-05 2020-09-10 Massachusetts Institute Of Technology Dna launched rna replicon system (drep) and uses thereof
BR112021017637A8 (en) 2019-03-08 2022-08-16 Massachusetts Inst Technology SYNTHETIC ONCOLYTIC VIRUS, PHARMACEUTICAL COMPOSITION, AND METHOD TO TREAT CANCER
AU2020248645A1 (en) 2019-03-27 2021-10-28 Tigatx, Inc. Engineered IgA antibodies and methods of use
AR118536A1 (en) 2019-04-01 2021-10-20 Genentech Inc COMPOSITIONS AND METHODS TO STABILIZE FORMULATIONS CONTAINING PROTEIN
KR20210146389A (en) 2019-04-02 2021-12-03 어레이 바이오파마 인크. protein tyrosine phosphatase inhibitor
EP3990452A1 (en) 2019-06-28 2022-05-04 Pfizer Inc. 5-(thiophen-2-yl)-1h-tetrazole derivatives as bckdk inhibitors useful for treating various diseases
TW202115086A (en) 2019-06-28 2021-04-16 美商輝瑞大藥廠 Bckdk inhibitors
LT6699B (en) 2019-07-15 2020-02-10 UAB "Valentis" Method for production of proliposomes by using up to 5% ethanol and the use thereof for encapsulation of lipophilic substances
CN112300279A (en) 2019-07-26 2021-02-02 上海复宏汉霖生物技术股份有限公司 Methods and compositions directed to anti-CD 73 antibodies and variants
US10758329B1 (en) 2019-08-20 2020-09-01 Raymond L. Wright, III Hydrating mouth guard
JP2022548310A (en) 2019-09-23 2022-11-17 シートムエックス セラピューティクス,インコーポレイテッド Anti-CD47 antibodies, activatable anti-CD47 antibodies, and methods of use thereof
WO2021062096A1 (en) 2019-09-26 2021-04-01 Massachusetts Institute Of Technology Microrna-based logic gates and uses thereof
US20220411511A1 (en) 2019-09-26 2022-12-29 Stcube & Co. Antibodies specific to glycosylated ctla-4 and methods of use thereof
US11834659B2 (en) 2019-09-26 2023-12-05 Massachusetts Institute Of Technology Trans-activated functional RNA by strand displacement and uses thereof
US20220356234A1 (en) 2019-10-02 2022-11-10 Alexion Pharmaceuticals, Inc. Complement inhibitors for treating drug-induced complement-mediated response
TWI771766B (en) 2019-10-04 2022-07-21 美商輝瑞股份有限公司 Diacylglycerol acyltransferase 2 inhibitor
AU2020363372A1 (en) 2019-10-07 2022-05-19 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
US20220356248A1 (en) 2019-10-09 2022-11-10 Stcube & Co Antibodies specific to glycosylated lag3 and methods of use thereof
CA3157509A1 (en) 2019-10-10 2021-04-15 Kodiak Sciences Inc. Methods of treating an eye disorder
EP4041773A1 (en) 2019-10-11 2022-08-17 Beth Israel Deaconess Medical Center, Inc. Anti-tn antibodies and uses thereof
WO2021079273A1 (en) 2019-10-21 2021-04-29 Rhizen Pharmaceuticals Ag Compositions comprising a dhodh inhibitor for the treatment of acute myeloid leukemia
BR112022010599A2 (en) 2019-12-10 2022-08-16 Pfizer SOLID ACID FORMS 2-((4-((S)-2-(5-CHLOROPYRIDIN-2-IL)-2-METHYLBENZO[D][1,3]DIOXOL-4-YL)PIPERIDIN-1-IL) METHYL)-1-(((S)-OXETAN-2-YL)METHYL)-1H-BENZO[D]IMIDAZOL-6-CARBOXYLIC, 1,3-DIHYDROXY-2-(HYDROXYMETHYL)PROPAN-2-AMINE SALT
EP4087659A2 (en) 2020-01-11 2022-11-16 Scholar Rock, Inc. Tgf-beta inhibitors and use thereof
US20230057012A1 (en) 2020-01-11 2023-02-23 Scholar Rock, Inc. Tgfb inhibitors and use thereof
JP2023511274A (en) 2020-01-14 2023-03-17 シンセカイン インコーポレイテッド IL2 orthologs and usage
WO2021146383A1 (en) 2020-01-17 2021-07-22 BioLegend, Inc. Anti-tlr7 agents and compositions and methods for making and using the same
US20230067811A1 (en) 2020-01-24 2023-03-02 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
JP2022058085A (en) 2020-02-24 2022-04-11 ファイザー・インク Combination of inhibitors of diacylglycerol acyltransferase 2 and inhibitors of acetyl-coa carboxylase
AU2021232062A1 (en) 2020-03-06 2022-09-29 Colorado State University Research Foundation Production of vaccines comprising inactivated SARS-CoV-2 viral particles
EP4118105A2 (en) 2020-03-09 2023-01-18 Pfizer Inc. Cd80-fc fusion protein and uses thereof
GB202003632D0 (en) 2020-03-12 2020-04-29 Harbour Antibodies Bv SARS-Cov-2 (SARS2, COVID-19) antibodies
US20230095383A1 (en) 2020-03-17 2023-03-30 Dst Pharma, Inc. Methods and compositions for treating viral infections
KR20220158031A (en) 2020-03-26 2022-11-29 피엘엑스 옵코 인코포레이티드 Pharmaceutical carrier capable of pH dependent reconstitution and method of manufacturing and using the same
EP4125896A1 (en) 2020-03-27 2023-02-08 Pfizer Inc. Treatment of type 2 diabetes or obesity or overweight with 2-[(4-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl} piperidin-1-yl)methyl]-1-[(2s)-oxetan-2-ylmethyl]-1h-benzimidazole-6-carboxylic acid or a pharmaceutically salt thereof
WO2021205322A1 (en) 2020-04-08 2021-10-14 Pfizer Inc. Crystalline forms of 3-cyano-1-[4-[6-(1-methyl-1h-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-4-yl]-1h-pyrazol-1-yl]cyclobutaneacetonitrile, and use thereof
AU2021259628A1 (en) 2020-04-24 2023-01-05 Millennium Pharmaceuticals, Inc. Anti-CD19 antibodies and uses thereof
US20230181750A1 (en) 2020-05-06 2023-06-15 Crispr Therapeutics Ag Mask peptides and masked anti-ptk7 antibodies comprising such
US20210355463A1 (en) 2020-05-15 2021-11-18 Crispr Therapeutics Ag Messenger rna encoding cas9 for use in genome-editing systems
TW202210473A (en) 2020-05-27 2022-03-16 大陸商上海齊魯銳格醫藥研發有限公司 Salt and crystal forms of glp-1r agonists and uses thereof
AU2021289169B2 (en) 2020-06-09 2024-01-04 Pfizer Inc. Spiro compounds as melanocortin 4 receptor antagonists and uses thereof
WO2021260577A2 (en) 2020-06-26 2021-12-30 Pfizer Inc. Methods of treating inflammatory bowel disease with tl1a antibodies
WO2022006562A1 (en) 2020-07-03 2022-01-06 Dana-Farber Cancer Institute, Inc. Multispecific coronavirus antibodies
CN116323668A (en) 2020-07-17 2023-06-23 辉瑞公司 Therapeutic antibodies and uses thereof
JP2023535225A (en) 2020-07-24 2023-08-16 ストランド セラピューティクス インコーポレイテッド Lipid nanoparticles containing modified nucleotides
CA3187514A1 (en) 2020-07-28 2022-02-03 Andrew BELFIELD Fused bicyclic raf inhibitors and methods for use thereof
US20220041595A1 (en) 2020-07-28 2022-02-10 Jazz Pharmaceuticals Ireland Limited Chiral synthesis of fused bicyclic raf inhibitors
US20230272091A1 (en) 2020-08-05 2023-08-31 Synthekine, Inc. Il10ra binding molecules and methods of use
WO2022032040A1 (en) 2020-08-05 2022-02-10 Synthekine, Inc. Il2rb/il2rg synthetic cytokines
EP4192489A2 (en) 2020-08-05 2023-06-14 Synthekine, Inc. Il2rb binding molecules and methods of use
KR20230061390A (en) 2020-08-05 2023-05-08 신테카인, 인크. Compositions and methods related to IL27 receptor binding
AR123156A1 (en) 2020-08-06 2022-11-02 Qilu Regor Therapeutics Inc GLP-1R AGONISTS AND THEIR USES
WO2022036041A1 (en) 2020-08-14 2022-02-17 Kite Pharma, Inc Improving immune cell function
AU2021340793A1 (en) 2020-09-14 2023-05-11 Vor Biopharma Inc. Single domain antibodies against cd33
JP2023542678A (en) 2020-09-21 2023-10-11 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Use of anti-CD40 antibodies for the treatment of inflammatory conditions.
WO2022076573A1 (en) 2020-10-06 2022-04-14 Xencor, Inc. Biomarkers, methods, and compositions for treating autoimmune disease including systemic lupus erythematous (sle)
TW202227421A (en) 2020-10-14 2022-07-16 大陸商上海齊魯銳格醫藥研發有限公司 Crystal forms of glp-1r agonists and uses thereof
WO2022093640A1 (en) 2020-10-30 2022-05-05 BioLegend, Inc. Anti-nkg2c agents and compositions and methods for making and using the same
WO2022093641A1 (en) 2020-10-30 2022-05-05 BioLegend, Inc. Anti-nkg2a agents and compositions and methods for making and using the same
US11058637B1 (en) * 2020-11-25 2021-07-13 King Abdulaziz University Surface-modified emulsomes for intranasal delivery of drugs
IL303195A (en) 2020-11-25 2023-07-01 Akagera Medicines Inc Lipid nanoparticles for delivery of nucleic acids, and related methods of use
IL303741A (en) 2020-12-18 2023-08-01 Momenta Pharmaceuticals Inc Antibodies against integrin alpha 11 beta 1
AR124599A1 (en) 2021-01-08 2023-04-12 Strand Therapeutics Inc CONSTRUCTS OF EXPRESSION AND THEIR USES
US11357727B1 (en) 2021-01-22 2022-06-14 Pacira Pharmaceuticals, Inc. Manufacturing of bupivacaine multivesicular liposomes
US11033495B1 (en) 2021-01-22 2021-06-15 Pacira Pharmaceuticals, Inc. Manufacturing of bupivacaine multivesicular liposomes
US11278494B1 (en) 2021-01-22 2022-03-22 Pacira Pharmaceuticals, Inc. Manufacturing of bupivacaine multivesicular liposomes
WO2022170008A2 (en) 2021-02-05 2022-08-11 Boehringer Ingelheim International Gmbh Anti-il1rap antibodies
CN117222641A (en) 2021-02-24 2023-12-12 英矽智能科技知识产权有限公司 Analogues for the treatment of disease
JP2024510162A (en) 2021-03-11 2024-03-06 カイト ファーマ インコーポレイテッド Improving immune cell function
WO2022195504A1 (en) 2021-03-19 2022-09-22 Pfizer Inc. Method of treating osteoarthritis pain with an anti ngf antibody
AU2022245196A1 (en) 2021-03-22 2023-09-21 Novimmune S.A. Bispecific antibodies targeting cd47 and pd-l1 and methods of use thereof
WO2022200387A1 (en) 2021-03-22 2022-09-29 Novimmune S.A. Bispecific antibodies targeting cd47 and pd-l1 and methods of use thereof
WO2022204581A2 (en) 2021-03-26 2022-09-29 Scholar Rock, Inc. Tgf-beta inhibitors and use thereof
WO2022215054A1 (en) 2021-04-09 2022-10-13 Takeda Pharmaceutical Company Limited Antibodies targeting complement factor d and uses therof
WO2022226291A1 (en) 2021-04-22 2022-10-27 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating cancer
IL307939A (en) 2021-04-26 2023-12-01 Millennium Pharm Inc Anti-clec12a antibodies and uses thereof
CA3214355A1 (en) 2021-04-26 2022-11-03 Antara Banerjee Anti-adgre2 antibodies and uses thereof
WO2022235940A1 (en) 2021-05-06 2022-11-10 Dana-Farber Cancer Institute, Inc. Antibodies against alk and methods of use thereof
WO2022245877A1 (en) 2021-05-17 2022-11-24 Curia Ip Holdings, Llc Sars-cov-2 spike protein antibodies
US20220372114A1 (en) 2021-05-17 2022-11-24 Curia Ip Holdings, Llc Sars-cov-2 spike protein antibodies
WO2022256723A2 (en) 2021-06-03 2022-12-08 Scholar Rock, Inc. Tgf-beta inhibitors and therapeutic use thereof
WO2022271867A1 (en) 2021-06-23 2022-12-29 Scholar Rock, Inc. A myostatin pathway inhibitor in combination with a glp-1 pathway activator for use in treating metabolic disorders
TW202306985A (en) 2021-07-12 2023-02-16 美商建南德克公司 Structures for reducing antibody-lipase binding
WO2023288277A1 (en) 2021-07-14 2023-01-19 Scholar Rock, Inc. Ltbp complex-specific inhibitors of tgfb1 and uses thereof
WO2023007374A1 (en) 2021-07-27 2023-02-02 Pfizer Inc. Method of treatment of cancer pain with tanezumab
GB202111758D0 (en) 2021-08-17 2021-09-29 Cantab Biopharmaceuticals Patents Ltd Modified colloidal particles for use in the treatment of haemophilia A
GB202111759D0 (en) 2021-08-17 2021-09-29 Cantab Biopharmaceuticals Patents Ltd Modified colloidal particles
GB202111757D0 (en) 2021-08-17 2021-09-29 Cantab Biopharmaceuticals Patents Ltd Modified colloidal particles for use in the treatment of haemophilia A
WO2023031741A1 (en) 2021-08-31 2023-03-09 Pfizer Inc. Solid forms of 2-[(4-{6-[(4-cyano-2-fluorobenzyl)oxy]pyridin-2-yl}piperidin-1-yl)methyl]-1-[(2s)-oxetan-2-ylmethyl]-1h-benzimidazole-6-carboxylic acid, 1,3-dihydroxy-2-(hydroxymethyl)propan-2-amine salt
WO2023034901A1 (en) 2021-09-01 2023-03-09 The Broad Institute, Inc. Tumor avatar vaccine compositions and uses thereof
WO2023036982A1 (en) 2021-09-10 2023-03-16 Harbour Antibodies Bv Anti-sars2-s antibodies
GB202112935D0 (en) 2021-09-10 2021-10-27 Harbour Antibodies Bv Sars-cov-2 (sars2, covid-19) heavy chain only antibodies
WO2023068382A2 (en) 2021-10-20 2023-04-27 Takeda Pharmaceutical Company Limited Compositions targeting bcma and methods of use thereof
WO2023077148A1 (en) 2021-11-01 2023-05-04 Tome Biosciences, Inc. Single construct platform for simultaneous delivery of gene editing machinery and nucleic acid cargo
WO2023081471A1 (en) 2021-11-05 2023-05-11 Dana-Farber Cancer Institute, Inc. Human broadly crossreactive influenza monoclonal antibodies and methods of use thereof
WO2023097024A1 (en) 2021-11-24 2023-06-01 Dana-Farber Cancer Institute, Inc. Antibodies against ctla-4 and methods of use thereof
TW202337429A (en) 2021-12-01 2023-10-01 美商輝瑞股份有限公司 Bckdk inhibitors and/or degraders
WO2023105387A1 (en) 2021-12-06 2023-06-15 Pfizer Inc. Melanocortin 4 receptor antagonists and uses thereof
WO2023105371A1 (en) 2021-12-08 2023-06-15 Array Biopharma Inc. Crystalline form of n-(2-chloro-3-((5-chloro-3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4-fluorophenyl)-3-fluoroazetidine-1-sulfonamide
WO2023114544A1 (en) 2021-12-17 2023-06-22 Dana-Farber Cancer Institute, Inc. Antibodies and uses thereof
WO2023111817A1 (en) 2021-12-17 2023-06-22 Pfizer Inc. Crystalline forms of [(1r,5s,6r)-3-{2-[(2s)-2-methylazetidin-1-yl]-6-(trifluoromethyl) pyrimidin-4-yl}-3-azabicyclo[3.1.0]hex-6-yl]acetic acid
WO2023114543A2 (en) 2021-12-17 2023-06-22 Dana-Farber Cancer Institute, Inc. Platform for antibody discovery
WO2023122764A1 (en) 2021-12-22 2023-06-29 Tome Biosciences, Inc. Co-delivery of a gene editor construct and a donor template
WO2023178357A1 (en) 2022-03-18 2023-09-21 Evolveimmune Therapeutics, Inc. Bispecific antibody fusion molecules and methods of use thereof
WO2023201238A1 (en) 2022-04-11 2023-10-19 Vor Biopharma Inc. Binding agents and methods of use thereof
WO2023205744A1 (en) 2022-04-20 2023-10-26 Tome Biosciences, Inc. Programmable gene insertion compositions
WO2023212618A1 (en) 2022-04-26 2023-11-02 Strand Therapeutics Inc. Lipid nanoparticles comprising venezuelan equine encephalitis (vee) replicon and uses thereof
WO2023215831A1 (en) 2022-05-04 2023-11-09 Tome Biosciences, Inc. Guide rna compositions for programmable gene insertion
WO2023220641A2 (en) 2022-05-11 2023-11-16 Juno Therapeutics, Inc. Methods and uses related to t cell therapy and production of same
WO2023225670A2 (en) 2022-05-20 2023-11-23 Tome Biosciences, Inc. Ex vivo programmable gene insertion
WO2023228023A1 (en) 2022-05-23 2023-11-30 Pfizer Inc. Treatment of type 2 diabetes or weight management control with 2-((4-((s)-2-(5-chloropyridin-2-yl)-2-methylbenzo[d][1,3]dioxol-4-yl)piperidin-1-yl)methyl)-1-(((s)-oxetan-2-yl)methyl)-1h-benzo[d]imidazole-6-carboxylic acid or a pharmaceutically salt thereof
WO2023235852A1 (en) 2022-06-03 2023-12-07 Zenas Biopharma, Inc. Methods and compositions for treating igg4- related diseases
WO2024020051A1 (en) 2022-07-19 2024-01-25 BioLegend, Inc. Anti-cd157 antibodies, antigen-binding fragments thereof and compositions and methods for making and using the same
WO2024030829A1 (en) 2022-08-01 2024-02-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies that bind to the underside of influenza viral neuraminidase
WO2024030847A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable moieties and methods of use thereof
WO2024030858A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable substrates and methods of use thereof
WO2024030845A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable moieties and methods of use thereof
WO2024030850A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable substrates and methods of use thereof
WO2024030843A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable moieties and methods of use thereof
WO2024039672A2 (en) 2022-08-15 2024-02-22 Dana-Farber Cancer Institute, Inc. Antibodies against msln and methods of use thereof
WO2024039670A1 (en) 2022-08-15 2024-02-22 Dana-Farber Cancer Institute, Inc. Antibodies against cldn4 and methods of use thereof
WO2024040114A2 (en) 2022-08-18 2024-02-22 BioLegend, Inc. Anti-axl antibodies, antigen-binding fragments thereof and methods for making and using the same

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0072111A1 (en) * 1981-07-20 1983-02-16 Lipid Specialties, Inc. Synthetic phospholipid compounds and their preparation
EP0118316A2 (en) * 1983-03-07 1984-09-12 Lipid Specialties, Inc. Synthetic phospholipid compounds, their preparation and use
GB2185397A (en) * 1986-01-17 1987-07-22 Cosmas Damian Ltd Drug delivery system
WO1988004924A1 (en) * 1986-12-24 1988-07-14 Liposome Technology, Inc. Liposomes with enhanced circulation time
EP0354855A2 (en) * 1988-08-11 1990-02-14 Terumo Kabushiki Kaisha Liposomes on which adsorption of proteins is inhibited
WO1990004384A1 (en) * 1988-10-20 1990-05-03 Royal Free Hospital School Of Medicine Liposomes

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3993754A (en) * 1974-10-09 1976-11-23 The United States Of America As Represented By The United States Energy Research And Development Administration Liposome-encapsulated actinomycin for cancer chemotherapy
US4501728A (en) * 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
JPS60100516A (en) * 1983-11-04 1985-06-04 Takeda Chem Ind Ltd Preparation of sustained release microcapsule
US4885172A (en) * 1985-06-26 1989-12-05 The Liposome Company, Inc. Composition for targeting, storing and loading of liposomes
IE58981B1 (en) * 1985-10-15 1993-12-15 Vestar Inc Anthracycline antineoplastic agents encapsulated in phospholipid micellular particles
US4797285A (en) * 1985-12-06 1989-01-10 Yissum Research And Development Company Of The Hebrew University Of Jerusalem Lipsome/anthraquinone drug composition and method
US4837028A (en) * 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US4863739A (en) * 1987-05-19 1989-09-05 Board Of Regents, The University Of Texas System Liposome compositions of anthracycline derivatives
JPH0696636B2 (en) * 1988-03-30 1994-11-30 富士写真フイルム株式会社 2,4-bis (o-methoxypolyethylene glycol) -6-cholesteryl-S-triazine compound
AU616040B2 (en) * 1988-08-11 1991-10-17 Terumo Kabushiki Kaisha Agents for inhibiting adsorption of proteins on the liposome surface
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0072111A1 (en) * 1981-07-20 1983-02-16 Lipid Specialties, Inc. Synthetic phospholipid compounds and their preparation
EP0118316A2 (en) * 1983-03-07 1984-09-12 Lipid Specialties, Inc. Synthetic phospholipid compounds, their preparation and use
GB2185397A (en) * 1986-01-17 1987-07-22 Cosmas Damian Ltd Drug delivery system
WO1988004924A1 (en) * 1986-12-24 1988-07-14 Liposome Technology, Inc. Liposomes with enhanced circulation time
EP0354855A2 (en) * 1988-08-11 1990-02-14 Terumo Kabushiki Kaisha Liposomes on which adsorption of proteins is inhibited
WO1990004384A1 (en) * 1988-10-20 1990-05-03 Royal Free Hospital School Of Medicine Liposomes

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Derwent File Supplier WPIL, 1989, AN 89-335922 (46), Derwent Publications, Ltd. (London, GB), & JP-A-1249798 (FUJI PHOTO FILM CO., LTD), 5 October 1989 *
Patent Abstracts of Japan, volume 13, no. 592 (C-671)(3940), 26 December 1989; & JP-A-1249717 (FUJI PHOTO FILM CO., LTD.) 5 October 1989 *

Cited By (159)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5356633A (en) * 1989-10-20 1994-10-18 Liposome Technology, Inc. Method of treatment of inflamed tissues
WO1994007537A1 (en) * 1992-10-05 1994-04-14 Stichting Voor De Technische Wetenschappen Pharmaceutical composition for site-specific release of a clot-dissolving protein
WO1994007466A1 (en) * 1992-10-07 1994-04-14 Liposome Technology, Inc. Compositions for treatmewnt of inflamed tissues
WO1994020073A1 (en) * 1993-03-03 1994-09-15 Liposome Technology, Inc. Lipid-polymer conjugates and liposomes
WO1994026251A1 (en) * 1993-05-07 1994-11-24 Sequus Pharmaceuticals, Inc. Subcutaneous liposome delivery method
US5514670A (en) * 1993-08-13 1996-05-07 Pharmos Corporation Submicron emulsions for delivery of peptides
US5993846A (en) * 1993-08-13 1999-11-30 Pharmos Corporation Bioadhesive emulsion preparations for enhanced drug delivery
US5744155A (en) * 1993-08-13 1998-04-28 Friedman; Doron Bioadhesive emulsion preparations for enhanced drug delivery
WO1996010391A1 (en) * 1994-09-30 1996-04-11 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
US5820873A (en) * 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
US6673364B1 (en) 1995-06-07 2004-01-06 The University Of British Columbia Liposome having an exchangeable component
US6096720A (en) * 1995-08-01 2000-08-01 Novartis Ag Liposomal oligonucleotide compositions
US5817856A (en) * 1995-12-11 1998-10-06 Yissum Research Development Company Of The Hebrew University Of Jerusalem Radiation-protective phospholipid and method
WO1997021429A1 (en) * 1995-12-11 1997-06-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem Radiation-protective phospholipid
US6165501A (en) * 1995-12-11 2000-12-26 Yissum Research Development Company Of The Hebrew University Of Jerusalem Radiation-protective phospholipid and method
WO1998008489A1 (en) * 1996-08-26 1998-03-05 Transgene S.A. Cationic lipid-nucleic acid complexes
US6271208B1 (en) 1996-08-26 2001-08-07 Transgene S.A. Process of making cationic lipid-nucleic acid complexes
US6316024B1 (en) 1996-10-11 2001-11-13 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
WO1998016202A3 (en) * 1996-10-11 1998-07-16 Sequus Pharm Inc Fusogenic liposome composition and method
AU715063B2 (en) * 1996-10-11 2000-01-13 Alza Corporation Fusogenic liposome composition and method
WO1998016202A2 (en) * 1996-10-11 1998-04-23 Sequus Pharmaceuticals, Inc. Fusogenic liposome composition and method
US6224903B1 (en) 1996-10-11 2001-05-01 Sequus Pharmaceuticals, Inc. Polymer-lipid conjugate for fusion of target membranes
US6734171B1 (en) 1997-10-10 2004-05-11 Inex Pharmaceuticals Corp. Methods for encapsulating nucleic acids in lipid bilayers
US6083923A (en) * 1997-10-31 2000-07-04 Isis Pharmaceuticals Inc. Liposomal oligonucleotide compositions for modulating RAS gene expression
WO1999027908A1 (en) * 1997-12-04 1999-06-10 Yissum Research Development Company Of The Hebrew University Of Jerusalem Combined chemo-immunotherapy with liposomal drugs and cytokines
WO2000066126A3 (en) * 1999-04-29 2001-04-05 Alza Corp Liposome compositions for improved drug retention
WO2000066126A2 (en) * 1999-04-29 2000-11-09 Alza Corporation Liposome compositions for improved drug retention
WO2001011069A1 (en) 1999-08-06 2001-02-15 Celltech R&D Limited Freeze/thawed lipid complexes and their preparation
EP2329830A2 (en) 2000-01-18 2011-06-08 ISIS Pharmaceuticals, Inc. Antisense inhibition of PTP1B expression
EP2003207A2 (en) 2000-01-18 2008-12-17 Isis Pharmaceuticals, Inc. Antisense inhibition of PTP1B expression
EP2213291A1 (en) 2000-01-18 2010-08-04 Isis Pharmaceuticals, Inc. Antisense inhibition of PTP1B expression
EP2365094A1 (en) 2001-05-14 2011-09-14 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
EP2246443A1 (en) 2001-05-14 2010-11-03 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
EP2221376A2 (en) 2001-06-21 2010-08-25 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
EP2270024A1 (en) 2001-06-21 2011-01-05 ISIS Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
EP2280019A1 (en) 2001-07-25 2011-02-02 ISIS Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
EP2332955A2 (en) 2001-07-30 2011-06-15 ISIS Pharmaceuticals, Inc. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
EP2174945A1 (en) 2001-08-01 2010-04-14 Genzyme Corporation Antisense modulation of apolipoprotein B expression
EP2336145A1 (en) 2001-08-01 2011-06-22 Genzyme Corporation Antisense modulation of apolipoprotein B expression
EP2316968A1 (en) 2001-08-07 2011-05-04 ISIS Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (A) expression
EP2272985A1 (en) 2001-08-07 2011-01-12 ISIS Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (A) expression
EP2270231A2 (en) 2001-10-09 2011-01-05 ISIS Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
EP2270230A2 (en) 2001-10-09 2011-01-05 ISIS Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
EP2388318A1 (en) 2001-12-10 2011-11-23 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
US8008270B2 (en) 2002-09-13 2011-08-30 Replicor Inc. Antiviral oligonucleotides targeting viral families
US8008269B2 (en) 2002-09-13 2011-08-30 Replicor Inc. Antiviral oligonucleotides
US7358068B2 (en) 2002-09-13 2008-04-15 Replicor, Inc. Antiviral oligonucleotides
EP2330194A2 (en) 2002-09-13 2011-06-08 Replicor, Inc. Non-sequence complementary antiviral oligonucleotides
US8067385B2 (en) 2002-09-13 2011-11-29 Replicor, Inc. Antiviral oligonucleotides targeting HBV
EP2272958A1 (en) 2002-09-26 2011-01-12 ISIS Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
EP2336318A1 (en) 2002-11-13 2011-06-22 Genzyme Corporation Antisense modulation of apolipoprotein B expression
EP2336319A1 (en) 2002-11-13 2011-06-22 Genzyme Corporation Antisense modulation of apolipoprotein B expression
EP1651187A2 (en) * 2003-07-31 2006-05-03 The Board Of Regents, The University Of Texas System Sterile preparations of phospholipids and anti-inflammatory pharmaceuticals and methods for making and using same
EP1651187A4 (en) * 2003-07-31 2009-03-11 Univ Texas Sterile preparations of phospholipids and anti-inflammatory pharmaceuticals and methods for making and using same
EP2284267A2 (en) 2003-08-18 2011-02-16 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US7960350B2 (en) 2003-10-24 2011-06-14 Ader Enterprises, Inc. Composition and method for the treatment of eye disease
US8338381B2 (en) 2003-10-24 2012-12-25 ADER Enterprise, Inc. Composition and method for the treatment of eye disease
EP2468865A1 (en) 2004-02-06 2012-06-27 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of stat3 expression
EP2527444A2 (en) 2004-02-06 2012-11-28 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of stat3 expression
US8293719B2 (en) 2004-03-12 2012-10-23 Alnylam Pharmaceuticals, Inc. iRNA agents targeting VEGF
EP2944646A1 (en) 2005-09-29 2015-11-18 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
EP2202239A1 (en) 2005-11-01 2010-06-30 Alnylam Pharmaceuticals Inc. RNAI inhibition of influenza virus replication
US7718629B2 (en) 2006-03-31 2010-05-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 gene
WO2007115168A2 (en) 2006-03-31 2007-10-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of eg5 gene
US7691824B2 (en) 2006-04-28 2010-04-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the JC virus
EP2392583A1 (en) 2006-05-19 2011-12-07 Alnylam Europe AG. RNAi modulation of Aha and therapeutic uses thereof
EP2584051A1 (en) 2006-05-22 2013-04-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expressions of IKK-B gene
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
US8048998B2 (en) 2007-01-19 2011-11-01 Exiqon A/S Mediated cellular delivery of LNA oligonucleotides
EP2905336A1 (en) 2007-03-29 2015-08-12 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of a gene from the ebola
US9074212B2 (en) 2007-06-15 2015-07-07 Arrowhead Research Corporation RNAi inhibition of alpha-ENaC expression
US8119612B2 (en) 2007-06-15 2012-02-21 Novartis Ag RNAi inhibition of alpha-ENaC expression
US7943592B2 (en) 2007-06-15 2011-05-17 Novartis Ag RNAi inhibition of alpha-ENaC expression
US8168606B2 (en) 2007-06-15 2012-05-01 Novartis Ag RNAi inhibition of alpha-ENaC expression
US7939508B2 (en) 2007-06-15 2011-05-10 Novartis Ag RNAi inhibition of alpha-ENaC expression
US10544418B2 (en) 2007-06-15 2020-01-28 Arrowhead Pharmaceuticals, Inc. RNAi inhibition of alpha-ENaC expression
US9476052B2 (en) 2007-06-15 2016-10-25 Arrowhead Pharmaceuticals, Inc. RNAi inhibition of alpha-ENaC expression
US11208662B2 (en) 2007-06-15 2021-12-28 Arrowhead Pharmaceuticals, Inc. RNAi inhibition of alpha-ENaC expression
US7718632B2 (en) 2007-06-15 2010-05-18 Novartis Ag RNAi inhibition of alpha-ENaC expression
US9914927B2 (en) 2007-06-15 2018-03-13 Arrowhead Pharmaceuticals, Inc. RNAi inhibition of alpha-ENaC expression
EP2848688A1 (en) 2007-12-10 2015-03-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor VII gene
EP2617828A1 (en) 2007-12-10 2013-07-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of factor VII gene
EP2712926A2 (en) 2008-03-05 2014-04-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 and VEGF genes
US8148344B2 (en) 2008-03-27 2012-04-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for mediating RNAi in vivo
US8324366B2 (en) 2008-04-29 2012-12-04 Alnylam Pharmaceuticals, Inc. Compositions and methods for delivering RNAI using lipoproteins
EP2140872A2 (en) 2008-07-02 2010-01-06 Kenneth Wileford Composition for the treatment of skin conditions comprising sea water with additional magnesium
EP3081648A1 (en) 2008-08-25 2016-10-19 Excaliard Pharmaceuticals, Inc. Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
WO2010042281A2 (en) 2008-08-25 2010-04-15 Excaliard Pharmaceuticals Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
EP2690175A2 (en) 2008-09-02 2014-01-29 Alnylam Pharmaceuticals Compositions and methods for combined inhibition of mutant EGFR gene and IL-6 expression
EP3208337A1 (en) 2008-09-02 2017-08-23 Alnylam Pharmaceuticals, Inc. Compositions for combined inhibition of mutant egfr and il-6 expression
EP3584320A1 (en) 2008-09-25 2019-12-25 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
EP3109321A1 (en) 2008-09-25 2016-12-28 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
EP3354733A1 (en) 2008-10-20 2018-08-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
EP3848461A1 (en) 2008-10-20 2021-07-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
EP2937418A1 (en) 2008-10-20 2015-10-28 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
EP3225281A1 (en) 2008-12-10 2017-10-04 Alnylam Pharmaceuticals, Inc. Gnaq targeted dsrna compositions and methods for inhibiting expression
WO2010068816A1 (en) 2008-12-10 2010-06-17 Alnylam Pharmaceuticals, Inc. Gnaq targeted dsrna compositions and methods for inhibiting expression
US8926955B2 (en) 2008-12-22 2015-01-06 Creabilis S.A. Synthesis of polymer conjugates of indolocarbazole compounds
WO2010099341A1 (en) 2009-02-26 2010-09-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mig-12 gene
WO2010105209A1 (en) 2009-03-12 2010-09-16 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
EP2810643A2 (en) 2009-08-14 2014-12-10 Alnylam Pharmaceuticals Inc. Lipid formulated compositions and mehods for inhibiting expression of a gene from the ebola virus
EP3252068A2 (en) 2009-10-12 2017-12-06 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
EP4089169A1 (en) 2009-10-12 2022-11-16 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
EP3329924A1 (en) 2010-03-29 2018-06-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
WO2011123468A1 (en) 2010-03-29 2011-10-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
EP3456827A2 (en) 2010-06-02 2019-03-20 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
WO2012064824A1 (en) 2010-11-09 2012-05-18 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of eg5 and vegf genes
WO2012078967A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for increasing erythropoietin (epo) production
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
WO2012106508A1 (en) 2011-02-02 2012-08-09 Pfizer Inc. Method of treating keloids or hypertrophic scars using antisense compounds targeting connective tissue growth factor (ctgf)
EP3674409A1 (en) 2011-03-29 2020-07-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of tmprss6 gene
EP3354343A1 (en) 2011-06-02 2018-08-01 President and Fellows of Harvard College Methods and uses for ex vivo tissue culture systems
WO2012166903A1 (en) 2011-06-02 2012-12-06 President And Fellows Of Harvard College Methods and uses for ex vivo tissue culture systems
EP3388068A1 (en) 2011-06-21 2018-10-17 Alnylam Pharmaceuticals, Inc. Composition and methods for inhibition of expression of protein c (proc) genes
EP3693464A2 (en) 2011-06-21 2020-08-12 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
WO2012177949A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of protein c (proc) genes
WO2012177906A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
EP3564393A1 (en) 2011-06-21 2019-11-06 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
WO2012177947A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
EP3597750A1 (en) 2011-06-23 2020-01-22 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
EP3366312A1 (en) 2011-06-23 2018-08-29 Alnylam Pharmaceuticals, Inc. Serpina 1 sirnas: compositions of matter and methods of treatment
EP4134433A1 (en) 2011-06-23 2023-02-15 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
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
WO2013019857A2 (en) 2011-08-01 2013-02-07 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
US20130224121A1 (en) * 2012-02-23 2013-08-29 Canon Kabushiki Kaisha Particle containing hydrophobic dye having cyanine structure, and contrast agent containing the particle
US9138492B2 (en) * 2012-02-23 2015-09-22 Canon Kabushiki Kaisha Particle containing hydrophobic dye having cyanine structure, and contrast agent containing the particle
WO2013155204A2 (en) 2012-04-10 2013-10-17 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
EP3868883A1 (en) 2012-04-10 2021-08-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
EP3284824A1 (en) 2012-04-10 2018-02-21 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
WO2014107731A1 (en) 2013-01-07 2014-07-10 Biomedical Research Models, Inc. Therapeutic vaccines for treating herpes simplex virus type 2 infections
WO2014160871A2 (en) 2013-03-27 2014-10-02 The General Hospital Corporation Methods and agents for treating alzheimer's disease
EP3708184A1 (en) 2013-03-27 2020-09-16 The General Hospital Corporation Methods and agents for treating alzheimer s disease
WO2014182661A2 (en) 2013-05-06 2014-11-13 Alnylam Pharmaceuticals, Inc Dosages and methods for delivering lipid formulated nucleic acid molecules
WO2015050990A1 (en) 2013-10-02 2015-04-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
EP3693463A1 (en) 2013-10-04 2020-08-12 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
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
WO2016094897A1 (en) 2014-12-12 2016-06-16 The Jackson Laboratory Compositions and methods relating to the treatment of cancer, autoimmune disease, and neurodegenerative disease
WO2016100716A1 (en) 2014-12-18 2016-06-23 Vasant Jadhav Reversirtm 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
US10485764B2 (en) 2015-07-02 2019-11-26 Otsuka Pharmaceutical Co., Ltd. Lyophilized pharmaceutical compositions
WO2017015109A1 (en) 2015-07-17 2017-01-26 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
EP3919619A1 (en) 2015-07-17 2021-12-08 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
WO2017048843A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
US10590416B2 (en) 2017-07-06 2020-03-17 Arrowhead Pharmaceuticals, Inc. RNAi agents for inhibiting expression of alpha-ENaC and methods of use
US11214802B2 (en) 2017-07-06 2022-01-04 Arrowhead Pharmaceuticals, Inc. RNAi agents for inhibiting expression of alpha-ENaC and methods of use
US10519190B2 (en) 2017-08-03 2019-12-31 Otsuka Pharmaceutical Co., Ltd. Drug compound and purification methods thereof
WO2020037125A1 (en) 2018-08-16 2020-02-20 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the lect2 gene
WO2020113135A1 (en) 2018-11-29 2020-06-04 Flagship Pioneering Innovations V, Inc. Methods of modulating rna
WO2021046122A1 (en) 2019-09-03 2021-03-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
WO2021067747A1 (en) 2019-10-04 2021-04-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
WO2021087325A1 (en) 2019-11-01 2021-05-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
WO2021163066A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
WO2021207167A1 (en) 2020-04-06 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
WO2021207189A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing scn9a expression
WO2021237097A1 (en) 2020-05-21 2021-11-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
WO2023283403A2 (en) 2021-07-09 2023-01-12 Alnylam Pharmaceuticals, Inc. Bis-rnai compounds for cns delivery

Also Published As

Publication number Publication date
JP2889549B2 (en) 1999-05-10
ATE122229T1 (en) 1995-05-15
ATE115401T1 (en) 1994-12-15
AU6637490A (en) 1991-05-16
FI921764A0 (en) 1992-04-21
AU6898291A (en) 1991-05-16
CA2067178A1 (en) 1991-04-21
AU642679B2 (en) 1993-10-28
LU88854I2 (en) 1997-03-11
JP2667051B2 (en) 1997-10-22
KR920703013A (en) 1992-12-17
WO1991005546A1 (en) 1991-05-02
DE69015207T2 (en) 1995-05-04
IL96069A (en) 1995-12-08
NO921213D0 (en) 1992-03-27
FI921763A (en) 1992-04-21
CA2067133A1 (en) 1991-04-21
EP0496835A1 (en) 1992-08-05
DE69019366D1 (en) 1995-06-14
HK14097A (en) 1997-02-14
EP0496813B1 (en) 1994-12-14
KR920703012A (en) 1992-12-17
JP3571335B2 (en) 2004-09-29
DE19675048I2 (en) 2009-05-28
DE69015207D1 (en) 1995-01-26
FI921763A0 (en) 1992-04-21
US5013556A (en) 1991-05-07
DE69019366T2 (en) 1995-10-05
JP2001181214A (en) 2001-07-03
NO921214L (en) 1992-06-01
JP3921050B2 (en) 2007-05-30
NO304637B1 (en) 1999-01-25
CA2067178C (en) 1997-03-25
IL96070A0 (en) 1991-07-18
IL96069A0 (en) 1991-07-18
NO1999003I1 (en) 1999-02-23
EP0496813A1 (en) 1992-08-05
FI921764A (en) 1992-04-21
CA2067133C (en) 1998-07-21
NO921213L (en) 1992-06-04
JPH101431A (en) 1998-01-06
IL96070A (en) 1995-03-30
NL960031I1 (en) 1997-03-03
JPH05501264A (en) 1993-03-11
ES2071976T3 (en) 1995-07-01
EP0496835B1 (en) 1995-05-10
NO921214D0 (en) 1992-03-27
DK0496835T3 (en) 1995-07-17
FI105151B (en) 2000-06-30
GR3017060T3 (en) 1995-11-30
JPH05505173A (en) 1993-08-05
US5213804A (en) 1993-05-25
KR0134982B1 (en) 1998-04-22
NL960031I2 (en) 2005-04-01
AU654120B2 (en) 1994-10-27

Similar Documents

Publication Publication Date Title
AU642679B2 (en) Liposome microreservoir composition and method
US5225212A (en) Microreservoir liposome composition and method
EP0632719B1 (en) Method of treatment of infected tissues
EP0662820B1 (en) Compositions for treatmewnt of inflamed tissues
US4844904A (en) Liposome composition
JP2001510451A (en) Ion carrier carrying weakly basic drug-liposome in the middle
US20030113369A1 (en) Liposomes with enhanced circulation time and method of treatment
AU2004218489A1 (en) Liposome composition for reduction of liposome-induced complement activation
US20010051183A1 (en) Liposomes with enhanced circulation time and method of treatment
WO2001041738A2 (en) Lipid carrier compositions with protected surface reactive functions
WO2000009071A2 (en) A novel liposomal formulation useful in treatment of cancer and other proliferation diseases
WO2005021012A1 (en) Drug carrier having gemcitabine enclosed therein
JPH04244017A (en) Liposome preparation
WO2003022250A2 (en) Unilamellar vesicles stabilized with short chain hydrophilic polymers
ZA200507816B (en) Liposome composition for reduction of liposome-induced complement activation
KR100768265B1 (en) Heparin coated liposomes to prolong circulation time in bloodstream and preparation method thereof
Scherphof et al. In Vivo Uptake and Processing of Liposomes by Parenchymal and Non-Parenchymal Liver Cells; Application to Immunotherapeutic Treatment of Hepatic Metastases
WO1999065481A1 (en) Liposomal formulations of busulphan

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA FI JP KR NO

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LU NL SE

WWE Wipo information: entry into national phase

Ref document number: 2067133

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 921763

Country of ref document: FI

WWE Wipo information: entry into national phase

Ref document number: 1990916409

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1990916409

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1990916409

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1990916409

Country of ref document: EP