Methods of manufacturing viscous liquid pharmaceutical formulations

Information

  • Patent Grant
  • 9724293
  • Patent Number
    9,724,293
  • Date Filed
    Wednesday, August 29, 2012
    12 years ago
  • Date Issued
    Tuesday, August 8, 2017
    7 years ago
Abstract
A liquid composition in an osmotic drug delivery system and a dosage form in an osmotic drug delivery system is disclosed comprising an amphiphilic molecule, a non-aqueous liquid solvent, and a pharmaceutically active agent.
Description
FIELD

The present invention relates to liquid composition in an osmotic drug delivery system and a dosage form of a liquid composition in an osmotic drug delivery device.


BACKGROUND

Development of drug suspension formulations designed for delivery from sustained delivery devices and osmotic delivery devices has resulted in a family of compositions utilizing polymer or surfactant solutions, non-aqueous liquid solvents or blends of solvents for polymer. The polymer acts as a viscosity modifier to provide acceptable stability of the suspension during self storage and performance intervals. These polymer solutions provide a stable environment for pharmaceutically active agents, e.g., small molecule drugs or therapeutic proteins or peptides.


Drug delivery devices attempt to deliver low solubility drugs by incorporating liquid drug formulations that are released at a controlled rate over time. These osmotic delivery devices are disclosed in U.S. Pat. Nos. 4,111,201; 5,324,280; 5,413,672; and 6,174,547. However, such liquid osmotic delivery systems are limited in the concentration of drug in the liquid formulation and hence, the drug loading available, leading to delivery systems that can be of an unacceptably large size, volume, or number for therapeutic purposes.


Polymers, such as polyvinyl pyrrolidone (PVP), exhibit solubility in a wide range of non-aqueous liquid solvents, but PVP is also quite soluble in water. As a result, highly viscous PVP/water gels can be produced near the formulation/water interface. These viscous gels can occlude the delivery conduit of the drug delivery device, interfering with performance of the delivery device. A need exists in the art to develop a viscous liquid compositions with improved performance characteristics in drug delivery devices, for example, osmotic drug delivery devices. There also exists a need to eliminate pluggage of discharge ports of implantable devices. Additionally, there is a need for suspending vehicles that are substantially resistant to phase separation while at the same time are sufficiently viscous to suspend pharmaceutical agents for long periods of time. Further, it is desirable to formulate polymer-free dosage forms which remain substantially homogenous for a desired dosaging time.


SUMMARY

The invention is generally related to drug delivery compositions comprising amphiphilic molecules in combination with a solvent form liquid compositions with desired properties that exhibit a desired viscosity and that exhibit a desired solvency in water. The viscous liquid compositions are useful for therapeutic drug delivery applications in which the formulation can be slowly introduced into an aqueous environment.


In one embodiment, a liquid composition in an osmotic drug delivery system comprises an amphiphilic molecule, a non-aqueous liquid solvent, and a pharmaceutically active agent. In a further embodiment, amphiphilic molecules include, but are not limited to, lipids, surfactants, amphiphilic block polymers, or amphiphilic proteins or peptides. In a further detailed embodiment, the lipid is selected from saturated lipid, unsaturated lipid, neutral lipid, anionic lipid, cationic lipid, natural lipid or synthetic lipid.


Amphiphilic molecules, for example, lipids, in combination with a solvent, form a viscous liquid formulation that exhibits a desired solvency in water for drug delivery applications in which the formulation is slowly introduced into an aqueous environment. Amphiphilic molecules, such as lipids, in non-aqueous liquid solvents can provide viscous liquid compositions for therapeutic drug or protein delivery in a system or delivery device that avoids problems of precipitation, aggregation, or formation of highly viscous gels at the lipid/solvent/water interface. The liquid composition overcomes problems which can limit delivery of a therapeutic composition from the delivery device. Amphiphilic molecules, such as lipids, in non-aqueous liquid solvents deliver therapeutic drugs or proteins from a delivery device into an aqueous environment in a continuous measured flow to the target tissue of interest.


Amphiphilic molecules, e.g., lipids in non-aqueous liquid solvents, can replace delivery systems comprising high molecular weight molecules, e.g., polymers such as PVP or PLGA, when forming therapeutic drug delivery systems in which particular rheological properties are desired. When introduced into an aqueous environment, lipids can be amphiphilic molecules capable of forming small self-assembled structures in water with forms ranging from bi-layers (multilamellar vesicles, unilamellar vesicles/liposomes) to micelles and even inverted micelles (hexagonal structure).


Drug delivery compositions comprising lipids in non-aqueous liquid solvents have been formulated into stable suspensions of a therapeutic drug suitable for dispensing from osmotic delivery devices for extended intervals. Compositions comprising lipids and non-aqueous liquid solvents can exhibit a variety of solubility properties in water and further can exhibit significant partitioning of the lipids at a water interface resulting in viscous liquid compositions e.g., viscous lipid gels.


In a further embodiment, drug delivery compositions comprising lipids in non-aqueous liquid solvents have been formulated into stable suspensions wherein the pharmaceutically active agent is suspended in a particle within said liquid composition. The particles containing the pharmaceutically active agent can be formed by a number of different processes, e.g., spray drying, lyophilization, or supercritical fluid processing.


In a detailed embodiment, non-aqueous liquid solvents useful to produce such gels include, but are not limited to, lauryl lactate (LL), lauryl alcohol (LA), benzyl alcohol (BA), benzyl benzoate (BB), 1:1 benzyl benzoate: benzyl alcohol, benzyl alcohol, ethyl hexyl lactate, glycerol formal, tetraglycol (glycofurol; GF), N-1-methyl-2-pyrrolidone (NMP), dimethyl sulfoxide, (DMSO), polyethyleneglycol (e.g., PEG 400), triglycerides (triolein, trilaurin, tricarprin, tricaprylin), ethanol, isopropanol, t-butyl alcohol, cyclohexanol, glycerin, glycerol, α-tocopherol (vitamin E) vegetable oil, sesame oil, soybean oil, cottonseed oil or peanut oil.


Various amphiphilic molecules can be used, e.g., lipids, surfactants, amphiphilic block polymers, or amphiphilic proteins or peptides. Lipid molecules include, but are not limited to, saturated and unsaturated lipids; neutral, cationic, or anionic lipids; or natural or synthetic lipids. Anionic lipids can induce formation of a helical segment of a protein, and can play an important physiological role. In a detailed embodiment, lipids can be one or a mixture of two or more from any of the following classes including, but not limited to: phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylglycerol (PG), phosphosphatidylinositol (PI), phosphatidylserine (PS), phosphatidic acid (PA) sphingomyelin (SM).


In a detailed embodiment, individual lipids include, but are not limited to: neutral lipids—dioleoyl phosphatidylcholine (DOPC), dimyristoylphosphatidylcholine (DMPC), dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), egg phosphatidylcholine (egg PC), soy phosphatidylcholine (soy PC), partially or fully hydrogenated phosphatidylcholines (PHSPC or HSPC), palmitoyl-oleoyl phosphatidylcholine (POPC), stearyloleoylphosphatidylcholine (SOPC); and anionic lipids—dioleoy phosphatidylglyserol (DOPG), dimyristoylphosphatidylglycerol (DMPG), dipalmitoylphosphatidylglyserol (DPPG), distearoylphosphatidylglyserol (DSPG). Various PEG (polyethylene glycol)-lipids can also be included for added benefit. Examples of PEG-lipids include, but are not limited to mPEG-DPPE, mPEG-DMPE, mPEG-DSPE, -mPEG-ceramide-DSPE, or mPEG-DS. Block copolymer surfactants (e.g., Pluronic® surfactants or Pluronic® surfactant F-127), or sorbitan esters structurants (e.g., Span® 80, Sigma Aldrich Chemical Co.) can be used. Moreover, when needed, increments of anti-oxidant lipid agents including, but not limited to, vitamin E, α-tocopherol, or ascorbic acid, can be added to prevent oxidation or peroxide formation. The increments of anti-oxidant lipid agents are already a part of the PHSPC formulations.


Amphiphilic molecule weight percent in the formulation can range from about 1 to about 100 weight percent, in detailed embodiments. Solvent weight percent in the formulation can range from about 0 to about 99 weight percent.


In a further embodiment, the pharmaceutically active agent is selected from a protein, peptide, small molecule drug, lipid drug or nucleic acid drug (e.g., DNA, RNA, antisense, ribozyme, DNAzyme, and the like).


The liquid composition is a viscous liquid composition, in a further detailed embodiment. The viscosity of the composition can be from about 1 to about 100,000 poise. A ratio by weight of amphiphilic molecule to non-aqueous liquid solvent can be between about 1 and about 4.


In a further embodiment, the pharmaceutically active agent is selected from biologically or pharmacologically active substance. In a detailed embodiment, the pharmaceutically active agent is ω-interferon, α-interferon, β-interferon, γ-interferon, erythropoietin, human growth hormone, granulocyte macrophage colony stimulating factor (GM-CSF), human growth hormone releasing hormone (huGHRH), insulin, desmopressin, infliximab, antibody or an agent conjugated to a targeting ligand, risperidone, paliperidone, glucagon-like peptide-1 (GLP-1), or bone morphogenic protein. The pharmaceutically active agent can be combined with sucrose, methionine and citrate in weight ratios of 1 to 2 to 1 to 2.15.


In another embodiment, a dosage form for osmotic drug delivery comprises a capsule including a first chamber containing a viscous liquid pharmaceutical composition and a second chamber containing an osmotic agent, the first chamber having an opening through which the pharmaceutical composition can be delivered from the first chamber to a location external of the first chamber; a movable separating member positioned in the capsule between the first chamber and the second chamber; a wall of the second chamber including a fluid permeable portion allowing fluid to pass from a surrounding environment into the second chamber; and an incompressible fluid additive located within the second chamber and substantially surrounding the osmotic agent.


In a detailed embodiment of the dosage form for osmotic drug delivery, the viscous liquid pharmaceutical composition comprises an amphiphilic molecule, a non-aqueous liquid solvent, and a pharmaceutically active agent. In a further detailed embodiment, the amphiphilic molecule is selected from lipids, surfactants, amphiphilic block polymers, or amphiphilic proteins or peptides.


In a detailed embodiment of the dosage form for osmotic drug delivery, the lipid is selected from saturated lipid, unsaturated lipid, neutral lipid, or anionic lipid. In a further detailed embodiment, the lipid is selected from phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylglycerol (PG), phosphosphatidylinositol (PI), phosphatidylserine (PS), phosphatidic acid (PA), or sphingomyelin (SM). In a further detailed embodiment, the neutral lipid is selected from dioleoyl phosphatidylcholine (DOPC), dimyritoylphosphatidylcholine (DMPC), dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), egg phosphatidylcholine (egg PC), soy phosphatidylcholine (soy PC), partially or fully hydrogenated phosphatidylcholins (PHSPC or HSPC), palmitoyl-oleoyl phosphatidylcholine (POPC), or stearyloleoylphosphatidylcholin (SOPC). In a further detailed embodiment, the anionic lipid is selected from dioleoy phosphatidylglycerol (DOPG), dimyritoylphosphatidylglycerol (DMPG), dipalmitoylphosphatidylglycerol (DPPG), or distearoylphosphatidylglycerol (DSPG).


In a detailed embodiment of the dosage form for osmotic drug delivery, the non-aqueous liquid solvent is selected from lauryl lactate (LL), lauryl alcohol (LA), benzyl alcohol (BA), benzyl benzoate (BB), 1:1 benzyl benzoate: benzyl alcohol, benzyl alcohol, ethyl hexyl lactate, glycerol formal, tetraglycol (glycofurol; GF), N-1-methyl-2-pyrrolidone (NMP), dimethyl sulfoxide, (DMSO), polyethyleneglycol (e.g., PEG 400), triglycerides (triolein, trilaurin, tricarprin, tricaprylin), ethanol, isopropanol, t-butyl alcohol, cyclohexanol, glycerin, glycerol, α-tocopherol (vitamin E) vegetable oil, sesame oil, soybean oil, cottonseed oil or peanut oil.


The pharmaceutically active agent is selected from a protein, peptide, small molecule drug, lipid drug or prodrug conjugated to lipid, in a further embodiment.


An amphiphilic molecule weight percent can be from about 1 to about 100 weight percent, in a detailed embodiment of the dosage form for osmotic drug delivery. In a further detailed embodiment, a non-aqueous liquid solvent weight percent can be from about 0 to about 99 weight percent. In further detailed embodiments, viscosity of the composition can be from about 1 to about 100,000 poise. A ratio by weight of amphiphilic molecule to non-aqueous liquid solvent can be between about 1 and about 4.


The osmotic agent is in a tabular form and the fluid additive surrounds the tabular osmotic agent, in a detailed embodiment of the dosage form for osmotic drug delivery. The separating member can be a slidable piston. The fluid permeable portion can be a membrane. In a further detailed embodiment, the osmotic agent is a tablet. The fluid additive can be a lubricating liquid for preventing freeze-up of the osmotic agent. The fluid additive can be a gel. In a further detailed embodiment, the fluid additive includes PEG. The osmotic agent includes NaCl.


In a detailed embodiment, the dosage form for the osmotic drug delivery system includes at least one gap between an inner surface of the capsule and the osmotic agent, the fluid additive filling the at least one gap to improve start-up time.


In a further aspect, the present invention provides suspending vehicles that comprise a amphiphilic molecule, a non-aqueous solvent, and a performance modifier. Preferably the suspending vehicles are substantially non-aqueous. Amphiphilic molecules can replace polymers in suspending vehicles in order to build viscosity. Performance modifiers in accordance with the present invention can be used, for example, to improve the stability and/or the flowability of lipid-based suspending vehicles. For example, the use of performance modifiers aids in softening formations, for example, stiff gels, made at exit ports upon contact by the suspending vehicle with aqueous media, for example, bodily fluids. As such, suspending vehicles are preferably flowable upon contact with an aqueous medium. Suspending vehicles are also substantially free of stiff gels upon contact with an aqueous medium.


Another benefit to using performance modifiers, such as co-solvents, is to speed-up preparation of suspending vehicles and suspensions due to increased solubility of the amphiphilic molecule, for example, the lipid, in co-solvents relative to another solvent.


Suspending vehicles comprising performance modifiers can exhibit improved rheological properties as compared to suspending vehicles which do not contain the performance modifier. For example, performance modifiers can be useful for reducing viscosity variations over small composition variations. In a preferred embodiment, the suspending vehicle has a change in viscosity of less than or equal to a factor of 10 in combination with a 10% by weight change in amount of the amphiphilic molecule in the suspending vehicle.


In some embodiments, the amphiphilic molecule in the suspending vehicle preferably comprises a phospholipid. Lipids include, but are not limited to, dioleoyl phosphatidylcholine (DOPC), dimyritoylphosphatidylcholine (DMPC), dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), egg phosphatidylcholine (egg PC), soy phosphatidylcholine (soy PC), partially hydrogenated phosphatidylcholins (PHSPC), fully hydrogenated phosphatidylcholins (HSPC), palmitoyl-oleoyl phosphatidylcholine (POPC), or stearyloleoylphosphatidylcholine (SOPC).


Suspending vehicles have a weight ratio of the non-aqueous solvent to the performance modifier that is, for example, preferably from about 1:3 to about 1:0.001, more preferably from about 1:1 to about 1:0.001, and even more preferably from about 1:0.33 to about 1:0.001. A weight ratio of the amphiphilic molecule to the performance modifier is preferably from about 0.18:1 to about 5.7:1, more preferably from about 0.43:1 to about 2.33:1, and even more preferably from about 0.67:1 to about 1.5:1. A weight ratio of the amphiphilic molecule to a combination of the performance modifier and the non-aqueous solvent is preferably from about 0.05:1 to about 19:1, more preferably from about 0.5:1 to about 1.5:1, even more preferably from about 0.75:1 to about 1.22:1.


In a preferred embodiment of a suspending vehicle, the lipid comprises dioleoyl phosphatidylcholine (DOPC), the non-aqueous solvent comprises α-tocopherol, and the performance modifier comprises ethanol, sesame oil, ethyl acetate, or combinations thereof.


Kits in accordance with the present invention comprise a suspending vehicle and instructions for suspending or dispersing a pharmaceutically active agent therein to create a pharmaceutical suspension. Other kits further comprise a dosage form and instructions for loading the dosage form with the pharmaceutical suspension.


Pharmaceutical suspensions can be created by adding a pharmaceutically active agent to suspending vehicles of the present invention. Pharmaceutical suspensions can also be added to desirable dosage forms, for example, dosage forms that are driven by pumps, for example, osmotic delivery devices.


Pharmaceutical suspensions in accordance with the present invention comprise a pharmaceutically active agent suspended or dispersed in a suspending vehicle, wherein the suspending vehicle comprises an amphiphilic molecule, a non-aqueous solvent, and a performance modifier. Suspending vehicles have a viscosity of from about 1 to about 1,000,000 poise, preferably the viscosity is from about 5 to about 100,000 poise.


Although any desirable pharmaceutically active agent may be stable in the suspending vehicles of the present invention, in preferred embodiments, the pharmaceutically active agent comprises ω-interferon.


Preferably, the dosage forms used in conjunction with the present invention have an osmotic pump. In other embodiments, the dosage forms comprise a wall that maintains its physical and chemical integrity during the life of the dosage form and is substantially impermeable to a pharmaceutical suspension; another wall that is partially permeable to an exterior fluid; a compartment defined by the wall; and an exit port in the wall in communication with the compartment; and wherein the pharmaceutical suspension is positioned within the compartment.


In some examples, the pharmaceutical suspension is flowable through the exit port upon contact with an aqueous medium, preferably under normal operating conditions of the osmotic pump. The pharmaceutical suspensions can also be substantially free of stiff gels upon contact with an aqueous medium. In other embodiments, the pharmaceutical suspensions are substantially homogeneous for at least 3 months.


Kits in accordance with the present invention comprise a dosage form containing a pharmaceutical suspension and instructions for administering the dosage form.


Methods in accordance with the present invention comprise identifying an amphiphilic molecule; identifying a non-aqueous solvent; identifying a performance modifier; mixing the amphiphilic molecule, the non-aqueous solvent, and the performance modifier to create a suspending vehicle. In some embodiments, the performance modifier improves rheological properties of the suspending vehicle as compared to a suspending vehicle which does not contain the performance modifier. Other methods include substantially dissolving or dispersing the amphiphilic molecule in the performance modifier to create a mixture; and combining the non-aqueous solvent with the mixture to create a suspending vehicle. Additionally, in other methods, the amphiphilic molecule dissolves more quickly in a combination of the non-aqueous solvent and the performance modifier as compared to dissolution in only the non-aqueous solvent.


In some methods, the mixture is vortexed. In other embodiments, it is preferable to heat the suspending vehicle to a temperature of from about 100° C. to about 120° C.


Methods in accordance with the present invention also include administering the dosage form containing pharmaceutical suspensions in accordance with the present invention to a mammal.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows that DOPC formulations can be shear thinning, a phenomena that can ease the manufacturing process of the vehicles.



FIG. 2 shows that the viscosity of DOPC formulations is temperature controlled.



FIG. 3 shows that PHSPC formulations can be shear thinning, a phenomena that can ease the manufacturing process of the vehicles.



FIG. 4 shows that the viscosity of PHSPC formulations is temperature controlled.



FIG. 5 shows images of DOPC formulations under polarized microscopy.



FIG. 6 shows images of PHSPC formulations under polarized microscopy.



FIG. 7 shows a shear rate effect on a DOPC:LL vehicle formulation at 37° C.



FIG. 8 shows a shear rate effect on a DOPC:NMP vehicle formulation at 37° C.



FIG. 9 shows the stability of ω-interferon in DOPC:LL suspension at 40° C.



FIG. 10 shows the effect of n-propanol on recovery of ω-interferon from DOPC:NMP or DOPC:LL suspensions.



FIG. 11 shows water ingress into model diffusion moderators for viscous liquid formulations, DOPC:LL or DOPC:NMP.



FIG. 12 shows a measure of the average protein released from a delivery device based on piston movement within the device.



FIG. 13 shows piston movement in a delivery device correlates to the average protein released from the device.



FIG. 14 shows viscosities of various DOPC:VitE formulations at various weight ratios.





DETAILED DESCRIPTION

A liquid composition in an osmotic drug delivery system of the present invention comprises amphiphilic molecules in combination with a solvent to form liquid compositions with desired properties that exhibit a desired viscosity and that exhibit a desired solvency in water. The viscous liquid compositions are useful for therapeutic drug delivery applications in which the formulation is slowly introduced into an aqueous environment. Amphiphilic molecules include, but are not limited to, lipids, surfactants, amphiphilic block polymers, or amphiphilic proteins or peptides.


Dynamic transfer mechanisms occur at the interface between non-aqueous liquid viscous drug suspensions as they pass through a conduit of a drug delivery device into an aqueous environment. Water ingress into the conduit can occur by one or all of several potential mechanisms: back diffusion, partitioning or percolation through the packed particles of the dispersed phase. Lipids exhibit solubility or dispersion in a wide range of non-aqueous liquid solvents. The wide range of solubility or dispersity lends to the use of lipids in formulation of viscous liquid drug compositions. When the viscous liquid composition contacts the aqueous environment, lipids can form small vesicles of one or all several potential forms: bilayers (multilamellar vesicles, unilameller vesicles/liposomes), micelles or inverted micelles (hexagonal structure) in water. This self-assembly characteristic allows for viscous delivery vehicles to be formed while at the same time limiting precipitation or the formation of highly viscous materials at the formulation/water interface.


Polymers, for example PVP, exhibit solubility in a wide range of non-aqueous liquid solvents as well, but they can also be quite soluble in water. As a result, highly viscous PVP/water gels can be produced in the non-aqueous liquid solvent/water interface. These viscous gels can occlude the delivery conduit, interfering with performance of the delivery device. In addition, while polymer gels form in the diffusion moderator of DUROS osmotic drug delivery system, they can precipitate and plug a conduit of the system, thus interfering with the release of the drug or therapeutic protein.


Amphiphilic molecules, e.g., lipids, in non-aqueous liquid solvents can provide liquid viscous drug and protein delivery systems in an osmotic delivery device. The composition can limit precipitation, aggregation or the formation of highly viscous gels in the solvent/lipid/water interface. Amphiphilic molecules, e.g., lipids, can also provide a less detrimental environment to incorporated therapeutic proteins due to the slower uptake of water after administration. Lipids can also be used as stabilizers for proteins to extend the therapeutic efficacy of the protein drug.


“Non-aqueous liquid solvent” refers to an organic molecule that will form a liquid viscous solution in combination with an amphiphilic molecule in the absence of water. The solvent can be, for example, a non-polar solvent, a protic solvent, such as an alcohol or acid, or an aprotic solvent, a polar solvent of moderately high dielectric constant, which does not contain an acidic hydrogen.


Lipids can be dissolved in various non-aqueous liquid solvents in different ratios to adjust the viscosity of the vehicle from 1-1,000,000 poise. These formulations can be shear thinning, a phenomena that can ease the manufacturing process of the vehicles. In addition, the solvent to be used can be optimized for maximal stability of the protein. In some cases, both lipids and the solvents to be used in the viscous liquid composition are approved for parenteral use by the Food and Drug Administration (FDA). Amphiphilic molecule weight percent in the formulation can range from about 1 to about 100 weight %. Solvent weight percent into the formulation can range from about 0 to about 99 weight %.


The viscous liquid compositions, including a pharmaceutically active agent cam be delivered by an osmotic drug delivery device, for example, U.S. Pat. Nos. 6,132,420; 6,174,547; 6,419,952; 6,551,613, each incorporated herein by reference in their entirety.


Viscous liquid compositions can exhibit the desired solvency in water and can have potential delivery applications in which the formulation is slowly introduced into an aqueous environment. Viscous liquid compositions comprising lipids in non-aqueous liquid solvents can replace the use of polymers such as polyvinyl pyrrolidone (PVP) to build viscosity in suspension vehicles. In an exemplary embodiment of viscous liquid compositions, solvents in which ω-interferon was shown to have a solubility of less than 0.1 mg/mL were combined with the lipid 1,2-Dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) to form a suspension of ω-interferon in solid particles in a pharmaceutical formulation. The exemplary non-aqueous liquid solvents included, but are not limited to, lauryl lactate (LL), lauryl alcohol (LA), benzyl alcohol (BA), benzyl benzoate (BB), 1:1 benzyl benzoate:benzyl alcohol, benzyl alcohol, ethyl hexyl lactate, glycerol formal, tetraglycol (glycofurol; GF), N-1-methyl-2-pyrrolidone (NMP), dimethyl sulfoxide, (DMSO), polyethyleneglycol (e.g., PEG 400), triglycerides (triolein, trilaurin, tricarprin, tricaprylin), ethanol, isopropanol, t-butyl alcohol, cyclohexanol, glycerin, glycerol, α-tocopherol (vitamin E) vegetable oil, sesame oil, soybean oil, cottonseed oil or peanut oil. The lipid, e.g., DOPC, was soluble over a wide range of solvent-to-lipid ratios. Lauryl lactate (LL) and N-methylpyrrolidone (NMP) as solvents in combination with lipid were chosen for further study.


Lipids can be dissolved in various non-aqueous liquid solvents in different ratios to adjust the viscosity of the vehicle from zero poise to 1,000,000 poise. The formulations can thin with increasing shear, a phenomena that can increase the ease with which the vehicles can be processed. Studies have shown that DOPC in lauryl lactate (LL) and N-methylpyrrolidone (NMP) at weight ratios of 1.5:1 and 3:1 lipid-to-solvent, respectively, have a viscosity of approximately 42,000 and 15,000 poise, respectively, at 37° C. and shear rates of ranging from 0.04-10 sec−1. These compositions have been shown to be shear thinning.




embedded image


1,2-Dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) has a molecular weight of 786.13 kdalton, and a phase transition temperature of −18° C.


The high viscosity and shear thinning properties of lipid/solvent solutions suggest that they can be suitable vehicles for DUROS® implants or OROS® osmotic delivery systems. See, for example, U.S. Pat. Nos. 6,132,420; 6,174,547; 6,419,952; 6,551,613, each incorporated herein by reference in their entirety. The study has determined that ω-interferon in DOPC/solvent compositions, e.g., DOPC/lauryl lactate or DOPC/α-tocopherol, at 40° C. is stable for at least four weeks. The DOPC/solvent compositions can be stable up to 3 months. A dissolution and release rate study has determined that the lipid gels allow the delivery of soluble ω-interferon into an aqueous environment.


“Amphiphilic molecule” refers to a molecule with distinct polar (hydrophilic) and non polar (hydrophobic) regions in the molecule. A “lipid” refers to a heterogeneous group of biological compounds that can contain a large organic cation or anion which possesses a long unbranched hydrocarbon chain, e.g., H3C(CH2)nCO2M+, H3C(CH2)nSO3M+, H3C(CH2)nN(CH3)3+X (n>7). The existence of distinct polar (hydrophilic) and non polar (hydrophobic) regions in the molecule can promote the formation of micelles in dilute aqueous solution. “Cationic lipids” have a net positive charge. “Anionic lipids” have a net negative charge. “Neutral lipids” have a net neutral charge.


“Lipid” refers to a heterogeneous group of biological compounds which are sparingly soluble in water, but very soluble in nonpolar solvents. As a class, lipids can be defined by their solubility. They include chemically diverse compounds. “Natural lipids” include, but are not limited to, triacylglycerols, waxes, and terpenes (e.g., monoterpenes, diterpenes, carotenoids or steroids). The more complex lipids include glycolipids and phospholids. Triacylglycerols and waxes are known as saponifiable lipid, whereas terpenes are called nonsaponifiable lipid. “Saturated lipids” have all single bonded C—C hydrocarbon chains. “Unsaturated lipids” have one or more doubled bonded (C═C) or triple bonded (C≡C) hydrocarbon chains.


“Amphiphilic block polymer” refers to block copolymers such as Pluronic® block copolymer surfactants, (BASF Corporation, Mount Olive, N.J.; e.g., Pluronic® surfactants or Pluronic® surfactant F-127), or sorbitan esters structurants (e.g., Span® 80, Sigma Aldrich Chemical Co.) can be used, which can function as antifoaming agents, wetting agents, dispersants, thickeners, and emulsifiers. Pluronic® block copolymer surfactants can be based on ethylene oxide and propylene oxide.


“Amphiphilic protein or peptide” refers to a polypeptide chain with distinct polar (hydrophilic) and non polar (hydrophobic) regions in the protein. The tertiary structure of protein is hydrophilic or highly charged (positively or negatively charged) at one region of the protein, and is hydrophobic (non-polar) at another region of the protein.


Amphiphilic molecules useful in the present invention include, but are not limited to, lipid, surfactant, amphiphilic block polymer, or amphiphilic proteins or peptides. Examples of amphiphilic molecules that can be suitable substances for forming a viscous liquid composition are fatty acid esters, e.g., glyceryl monoesters of fatty acids. Other substances which have ability of forming a viscous liquid composition can be found among amphiphilic substances such as polar lipids, surfactants and emulsifiers. Examples of glyceryl monoesters of fatty acids include glycerylmonooleate (monoolein) and glycerylmonolinoleate.


The “viscosity” of a fluid refers to resistance of the fluid to shear or flow, and is a measure of the fluid's adhesive/cohesive or frictional properties. This arises because of the internal molecular friction within the fluid producing the frictional drag effect. Viscosity is useful in the study of biopolymers because the addition of large molecules to a solvent increases its viscosity; the increase depends on the concentration, size, and structure of the polymer. Motion in one layer of a fluid causes motion in adjoining layers. To move layers with different relative velocities requires a force: the more viscous, the more force. Dynamic viscosity is also termed “absolute viscosity” and is the tangential force per unit area required to move one horizontal plane with respect to the other at unit velocity when maintained a unit distance apart by the fluid. The theoretical unit of viscosity is the Poise.


Fatty acid esters capable of forming a viscous liquid composition comprise a fatty acid component and a hydroxy-containing component wherein the fatty acid component of the fatty acid ester is a saturated or unsaturated fatty acid having a total number of carbon atoms of from C6 to C26. Specific examples of saturated fatty acid moieties in the fatty acid esters according to the invention include, but are not limited to, moieties of caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, and behenic acid. Specific examples of unsaturated fatty acid moieties in the fatty acid esters according to the invention include, but are not limited to, moieties of palmitoleic acid, oleic acid, linoleic acid, linolenic acid, and arachidonic acid. Further embodiments of fatty acid esters for use in compositions according to the invention include, but are not limited to, fatty acid esters of polyhydric alcohols, fatty acid esters of hydroxycarboxylic acids, fatty acid esters of monosaccharides, fatty acid esters of glycerylphosphate derivatives, fatty acid esters of glycerylsulfate derivatives, and mixtures thereof. In those cases where the hydroxy-containing component of the fatty acid ester is polyvalent, the hydroxy-containing component can be partially or totally esterified with a fatty acid component or with mixtures of fatty acid components.


The polyhydric alcohol component of the fatty acid ester for use in compositions according to the invention is preferably selected from the group consisting of glycerol, 1,2-propanediol, 1,3-propanediol, diacylgalactosyldycerol, diacyldigalactosylglycerol, erythritol, xylitol, adonitol, arabitol, mannitol, and sorbitol. The fatty acid esters formed from such polyhydric alcohols can be mono- or polyvalent such as, e.g., divalent, trivalent, etc. In particular fatty acid monoesters have proved to have bioadhesive properties and can be fatty acid esters for use in compositions according to the invention. The position of the polyvalent alcohol on which the ester bond(s) is(are) established can be any possible position. In those cases where the fatty acid ester is a diester, triester, etc. the fatty acid components of the fatty acid ester can be the same or different. In a detailed aspect of the present invention, the polyhydric alcohol component can be glycerol.


Further examples of fatty acid esters for use in compositions according to the invention and wherein the hydroxy-containing component is a polyhydric alcohol include, but are not limited to, glycerylmonooleate, glycerylmonolinoleate, glycerol monolinoleate, and mixtures thereof. These fatty acid esters have bioadhesive properties useful in the viscous liquid compositions described herein. In those cases where the fatty acid ester for use in compositions according to the present invention is formed between a hydroxycarboxylic acid (or a derivative thereof) and a fatty acid (or a derivative thereof), the hydroxycarboxylic acid component of the fatty acid ester is preferably selected from the group consisting of malic acid, tartaric acid, citric acid, lactic acid, and sorbic acid. An example of a fatty acid ester for use in compositions according to the invention is a fatty acid monoester of citric acid.


The hydroxy-containing component of a fatty acid ester for use in compositions according to the present invention can also be a saccharide, such as a monosaccharide such as, e.g., glucose, mannose, fructose, threose, gulose, arabinose, ribose, erythrose, lyxose, galactose, sorbose, altrose, tallose, idose, rhamnose, or allose. In those cases where the hydroxy-containing component is a monosaccharide, the fatty acid ester is preferably a fatty acid monoester of a monosaccharide selected from the group consisting of sorbose, galactose, ribose, and rhamnose. The hydroxy-containing component of a fatty acid ester for use in the viscous liquid compositions can also be a glycerylphosphate derivative such as, e.g., a phospholipid selected from, phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylglycerol (PG), phosphatidylinositol (PI), phosphatidylserine (PS), phosphatidic acid (PA), or sphingomyelin (SM).or diphosphatidylglycerol. Phospholipids can further include DEPE (1,2dielaidoyl-sn-glycerol-3-phosphoethanolamine) and DMPE (PEG 550) (1,2-dimyristoyl-sn-glycero-3-phosphoetiianolainine-N-(polyethylene glycol)550).


Compounds having a phospholipid moiety further include compounds wherein the fatty acid ester is a fatty acid ester of a glycerylphosphate derivative, and the fatty acid component is selected from the group consisting of lauric acid, myristic acid, palmitic acid, stearic acid, oleic acid, linoleic acid, linolenic acid, and behenic acid. Examples of such useful fatty acid esters can be dilauryl phosphatidylcholine, dioleoyl phosphatidylcholine (DOPC), dimyritoylphosphatidylcholine (DMPC), dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), egg phosphatidylcholine (egg PC), soy phosphatidylcholine (soy PC), partially or fully hydrogenated phosphatidylcholins (PHSPC or HSPC), palmitoyl-oleoyl phosphatidylcholine (POPC), stearyloleoylphosphatidylcholin (SOPC), dibehenoyl phosphatidylcholine, dimyristyl phosphatidylethanolamine, dipalmitoyl phosphatidylethanolamine, dioleoyl phosphatidylglycerol (DOPG), dimyristoylphosphatidylglycerol (DMPG), dipalmitoylphosphatidylglycerol (DPPG), or distearoylphosphatidylglycerol (DSPG), dilauryl phosphatidylglycerol, dipalmitoyl phosphatic acid and mixtures thereof.


Compounds that can be useful as amphiphilic molecules include, but are not limited polyethyleneglycol (PEG)-lipid compound selected from mPEG-DPPE, mPEG-DMPE, mPEG-DSPE, or mPEG-ceramide-DSPE, and Pluronic® block copolymer surfactants, selected from PLURONIC® 17R2 surfactant, PLURONIC® 17R4 surfactant, PLURONIC® 25R2 surfactant, PLURONIC® 25R4 surfactant, PLURONIC® 31R1 surfactant, PLURONIC® F108 cast solid surfactant, PLURONIC® F108 NF cast solid surfactant, PLURONIC® F108 NF prill surfactant, PLURONIC® F108 pastille surfactant, PLURONIC® F108 surfactant prill, PLURONIC® F127 cast solid surfactant, PLURONIC® F127 NF 500BHT surfactant prill, PLURONIC® F127 NF cast solid surfactant, PLURONIC® F127 NF prill surfactant, PLURONIC® F127 surfactant prill, PLURONIC® F127NF 500BHT cast solid surfactant, PLURONIC® F38 cast solid surfactant, PLURONIC® F38 surfactant pastille, PLURONIC® F68 LF pastille surfactant, PLURONIC® F68 LF cast solid surfactant, PLURONIC® F68 NF cast solid surfactant, PLURONIC® F68 NF prill surfactant, PLURONIC® F68 prill surfactant, PLURONIC® F68 surfactant, PLURONIC® F68 surfactant pastille, PLURONIC® F77 cast solid surfactant, PLURONIC® F77 MICRO-pastille surfactant, PLURONIC® F87 cast solid surfactant, PLURONIC® F87 NF cast solid surfactant, PLURONIC® F87 NF prill surfactant, PLURONIC® F87 prill surfactant, PLURONIC® F88 cast solid surfactant, PLURONIC® F88 prill surfactant, PLURONIC® F88 surfactant pastille, PLURONIC® F98 cast solid surfactant, PLURONIC® F98 prill surfactant, PLURONIC® L10 surfactant, PLURONIC® L101 surfactant, PLURONIC® L121 surfactant, PLURONIC® L31 surfactant, PLURONIC® L35 surfactant, PLURONIC® L43 surfactant, PLURONIC® L44 NF surfactant, PLURONIC® L44 surfactant, PLURONIC® L61 surfactant, PLURONIC® L62 LF surfactant, PLURONIC® L62 surfactant, PLURONIC® L62D surfactant, PLURONIC® L64 surfactant, PLURONIC® L81 surfactant, PLURONIC® L92 surfactant, PLURONIC® N-3 surfactant, PLURONIC® P103 surfactant, PLURONIC® P104 surfactant, PLURONIC® P105 surfactant, PLURONIC® P123 surfactant, PLURONIC® P65 surfactant, PLURONIC® P84 surfactant, PLURONIC® P85 surfactant, PLURONIC® F 127 micro pastille.


Compounds that can be useful as non-aqueous liquid solvent include, but are not limited to, lauryl lactate (LL), lauryl alcohol (LA), benzyl alcohol (BA), benzyl benzoate (BB), 1:1 benzyl benzoate:benzyl alcohol, benzyl alcohol, ethyl hexyl lactate, glycerol formal, tetraglycol (glycofurol; GF), N-1-methyl-2-pyrrolidone (NMP), dimethyl sulfoxide, (DMSO), polyethyleneglycol (e.g., PEG 400), triglycerides (triolein, trilaurin, tricarprin, tricaprylin), ethanol, isopropanol, t-butyl alcohol, cyclohexanol, glycerin, glycerol, α-tocopherol (vitamin E) vegetable oil, sesame oil, soybean oil, cottonseed oil or peanut oil.


Block copolymer surfactants (BASF Corporation, Mount Olive, N.J.; e.g., Pluronic® surfactants or Pluronic® surfactant F-127), or sorbitan esters structurants (e.g., Span® 80, Sigma Aldrich Chemical Co.) can function as antifoaming agents, wetting agents, dispersants, thickeners, and emulsifiers. The Pluronic® type can be block copolymers based on ethylene oxide and propylene oxide.


Most of the fatty acid esters for use in compositions according to the invention can be chemical compounds which are commercially available or can be prepared by means of esterification procedures involving e.g., reaction of a fatty acid derivative such as, e.g., the corresponding acid chloride with a hydroxy-containing compound (if necessary protected with suitable protection groups) and subsequently isolating the fatty acid ester, if necessary after removal of any protecting group. Many of the commercially available fatty acid esters can be employed in the food industry and in general, no steps are taken in order to obtain an approximately 100% pure fatty acid ester. For example, glycerylmonooleate (Danisco Ingredients A/S, Denmark) is a very pure product containing about 98% w/w monoesters of which more than about 80% w/w (such as about 92% w/w) is glycerylmonooleate. The remaining monoesters can be glycerylmonolinoleate, glyceryl monopalmitate and glyceryl monostearate. The fatty acid ester products for use in compositions according to the invention can thus be mixtures of fatty acid esters. Examples of fatty acid esters with bioadhesive properties as well as an excellent ability of forming a viscous liquid composition can be glyceryl monoesters of fatty acids. Specific examples include glycerylmonooleate (monoolein) and glycerylmonolinoleate.


The stability of the viscous liquid composition comprising an amphiphilic molecule and solvent is considerably enhanced, such as resulting in a storage stability of at least 2 years at 25° C., and at least 1 month at 40° C. The fatty acid ester product fulfills certain purity standards. Thus, the fatty acid ester product used for the preparation of the composition should contain at the most 4% of saturated fatty acid ester and should preferably contain at least 88% of fatty acid ester, more preferably at least 89%, such as at least 90% or at least 91%, in particular at least 92%, of fatty acid ester.


The amphiphilic molecule, e.g., lipid, is normally present in an amount of at least about 1% to about 100% by weight, calculated on the total composition, such as at least 25% or at least 50%, or at least 75%, or at least 100% by weight, calculated on the total composition, and a preferred amount is often in the range of 25-90% such as 40-90% by weight, in particular, 40-85%, 45-80% or 50-75% by weight, calculated on the total composition.


Normally, the concentration of the non-aqueous liquid solvent in a composition according to the invention is about 0% to about 99% by weight, at least about 10% by weight such as, e.g., at least 20%, 30%, 40%, 50%, 60%, or 70% by weight, or up to 99% by weight calculated on the composition.


In other terms, the concentration of the non-aqueous liquid solvent in the composition is in a range corresponding to from about 0% to about 99% such as, e.g., about 15%-85%, about 20%-80%, about 25%-75%, about 25%-70%, about 25%-65%, about 25%-60%, about 25%-55%, or about 25%-50% by weight based on the total composition.


Formation of the viscous liquid composition, in addition to comprising an amphiphilic molecule, a non-aqueous solvent, and a pharmaceutically active agent, can further comprise an amphiphilic substance having a molecular weight of, at most, 2000 daltons, or an emulsifier or a surfactant. Tenside surfactants (anionic, cationic, non-ionic like e.g., sorbitan esters, sorbitan macrogol esters (polysorbates)), polar lipids, glycolipids, lecithins, palmitoyl muramic acid (PMA), substances having surface active properties like e.g., certain cellulose derivatives, sorbitan oleate, sorbitan laurate, lanolin and derivatives thereof and ethoxylated derivatives of lanolin (Aqualose W20, Aqualose L30 and Aqualose L75) are also examples of suitable structurants for use in compositions according to the invention. Sorbitan esters can be a series of mixtures of partial esters of sorbitol and its mono- and di-anhydrides with fatty acids.


Examples of suitable sorbitan esters for use as structurants in a composition according to the invention can be: sorbitan di-isostearate, sorbitan dioleate, sorbitan monoisostearate, sorbitan monolaurate, sorbitan monooleate (e.g., Span® 80, Sigma Aldrich Chemical Co.), sorbitan monopalmitate, sorbitan monostearate, sorbitan sesqui-isostearate, sorbitan sesquioleate (e.g., Span® 83), sorbitan trioleate, sorbitan sesquistearate, sorbitan tri-isostearate, sorbitan tristearate, sorbitan tristearate.


Polyoxyethylene sorbitan fatty acid esters (polysorbates) can be a series of fatty acid esters or sorbitol and its anhydrides copolymerized with approximately 20 moles of ethylene oxide for each mole of sorbitol and its anhydrides. Examples of suitable polysorbates for use in the present context can be: polysorbate 20, polysorbate 21, polysorbate 40, polysorbate 60, polysorbate 61, polysorbate 65, polysorbate 80, polysorbate 81, polysorbate 85, polysorbate 120


In a further embodiment, suitable amphiphilic molecules for use in a viscous liquid composition include tocopherols. “Tocopherols” refers to all vitamin E or vitamin E-like substances, derivatives and analogs thereof. The term includes all tocol and tocotrienol derivatives such as e.g., methyl tocol. More specifically, in the present context, a tocopherol is selected from the group consisting of p-tocopherols, sorbitan esters of tocopherols, d-α-tocopherol, d,1-α-tocopherol, d-α-tocopherol acetate, d,1-α-tocopherol acetate, d-α-tocopherol succinate, d,1-α-tocopherol succinate, d-α-tocopherol nicotinate, d,1-α-tocopherol nicotinate, tocopherylpolyethylene glycol succinate such as d-α-tocopherylpolyethylene glycol succinate or d,1-α-tocopherylpolyethylene glycol succinate, and derivatives such as fatty acid ester derivatives and analogues thereof. Tocopherols for use in a composition according to the present invention can be d-a-30. In a further detailed embodiment, tocopherylpolyethylene glycol 1000 succinate (in the following denoted vitamin E TPGS or simply TPGS) or d,1-α-tocopherylpolyethylene glycol 1000 succinate can be suitable amphiphilic molecules.


Fatty acid esters can be capable of forming various crystalline phases upon contact with a hydrophilic medium such as water or glycerol. The fatty acid esters also show bioadhesive properties. Viscous liquid composition can be a cubic (three cubic liquid crystalline phases are well-characterized: i) the body-centered lattice, ii) the primitive diamond lattice, and iii) the gyroid), reverse cubic, hexagonal, reverse hexagonal, lamellar, micellar or reverse micellar phase. By the term “cubic liquid crystalline phase” herein is meant a thermodynamically stable, viscous and optically isotropic phase made of a suitable substance such as, e.g., a fatty acid ester and a liquid medium such as, e.g., a viscous liquid medium or an aqueous medium. The cubic liquid crystalline phase is contemplated to be build up of closed reversed micelles. The term “viscous liquid medium” includes media comprising an amphiphilic molecule and a non-aqueous non-aqueous liquid solvent. The term “aqueous medium” includes media containing water or another hydrophilic and water-miscible substance such as, e.g., glycerol. The terms “hexagonal phase” and “reverse hexagonal phase”, respectively, are used herein to describe thermodynamically stable, viscous and optically anisotropic phases characterized by long-range order in two dimensions and made of a suitable substance such as, e.g., a fatty acid ester and a liquid medium such as, e.g., a viscous liquid medium or an aqueous medium. The term “lamellar phase” is characterised by a long-range order in one dimension. The lamellar structure is the origin of liposomes having spherical shells of lipid bilayers. The various liquid crystalline phases can be detected and identified by use of polarized light or by means of X-ray diffraction pattern analysis. The cubic liquid crystalline phase is normally the preferred phase in the compositions of the invention, but also, e.g., the reverse hexagonal and the reverse cubic liquid crystalline phase can be a liquid crystalline phase in the compositions according to the invention.


Pharmaceutically Active Agents


“Pharmaceutically active agent” refers to any biologically or pharmacologically active substance or antigen-comprising material; the term includes drug substances which have utility in the treatment or prevention of diseases or disorders affecting animals or humans, or in the regulation of any animal or human physiological condition and it also includes any biologically active compound or composition which, when administered in an effective amount, has an effect on living cells or organisms. The pharmaceutically active agent includes, but is not limited to a protein, peptide, small molecule drug, lipid drug or nucleic acid drug (e.g., DNA, RNA, antisense, ribozyme, DNAzyme.


Examples of active substances of particular importance with respect to all aspects of the invention can be the so-called antiherpes virus agents which have been or are developed for the treatment of herpes virus infections [herpes simplex virus types 1 and 2 (HSV-1 and HSV-2), varicella zoster virus (VZV), cytomegalovirus (CMV), Epstein-Barr virus (EBV)]. The antiherpes virus agents include antiviral drugs and prodrugs thereof, such as nucleosides, nucleoside analogues, phosphorylated nucleosides (nucleotides), nucleotide analogues and salts, complexes and prodrugs thereof; e.g., guanosine analogues, deoxyguanosine analogues, guanine, guanine analogues, thymidine analogues, uracil analogues and adenine analogues. Antiherpes virus agent for use either alone or in combination in a composition according to the present invention can be selected from acyclovir, famciclovir, deciclovir, penciclovir, zidovudin, ganciclovir, didanosin, zalcitabin, valaciclovir, sorivudine, lobucavir, brivudine, cidofovir, n-docosanol, ISIS-2922, and prodrugs and analogues thereof. Details concerning active substances suitable for use in connection with the present invention as well as a description of other active substances are given below.


As mentioned above, an important example of an active substance is an antiviral drug, such as a nucleoside or a nucleoside analogue, e.g., selected from acyclovir, famciclovir, deciclovir, penciclovir, zidovudin, ganciclovir, didanosin, zalcitabin, valaciclovir, sorivudine, lobucavir, brivudine, cidofovir, n-docosanol, ISIS-2922 and salts and prodrugs thereof. However, also a large number of other drugs which in themselves have a low solubility as denned herein or the salts, esters, prodrugs or precursors of which have a low solubility can be important active substances in the compositions of the invention. Furthermore, there is also a large number of drugs which advantageously can be incorporated in a composition according to the invention, either as the sole active substance (provided the solubility criteria can be fulfilled) or in combination with another active substances. In the following is listed a number of active substances which either alone or in combination can be incorporated in a composition according to the present invention. In particular a combination of an anti-herpes virus agent and a glucocorticosteroid is of importance.


Examples of drugs which can be of particular importance in connection with application to skin or mucosal surfaces are: Acyclovir, famciclovir, ribavirin, zidovudin, ganciclovir, didanosin, zalcitabin, valaciclovir amantadin, rimantadin foskamet idoxuridin fluoruracil interferons and variants thereof, including α-interferon, β-interferon, γ-interferon, ω-interferon, tromantadin, lentinan, levofloxacin, stavudine, tacrine, vesnarinone, ampligen, atevirdine, delavirdine, hydroxyurea, indinavir sulfate, interleukin-2 fusion toxin, seragen, lamivudine, lidakol, nevirapine, onconase, saquinavir, topotecan, verteporfin, viraplex, CMV immunoglobulin, Efalith, epervudine, podophyllotoxin, proxigermanium, rifabutin, bromovinyldeoxyuridine, ukrain, cidofovir, imiquimod, lamivudine, sorivudine, viraplex afovirsen amonafide hypericin, provir, temoporfin, apbidicolin glycinate, ibobucavir, virend, AL-721, ampligen, arildone, brivudine, CD4, 2-deoxy-D-glucose, desciclovir, dichloroflavan, didanosine, ditiocarb Sodium edoxudine, enviroxime, fiacitabine, inosine Pranobex, peptide T stavudine tribavirin trifluridine vidarabine zalcitabine, miconazol fucidin, erythromycin macrolides, NSAID's, peptides insulin, polymycin, myperizin, antibiotics, nicotine sucralfate, sucrose octasulfate, salicylic acid, urea, benzoylperoxide minoxidil, heparinoid, methotrexate, ciclosporin.


A listing of substances of potential interest comprises substances of the following groups: sodium fluoride, anti-inflammatory drugs such as, e.g., ibuprofen, indomethacin, naproxen, diclofenac, tolfenamic acid, piroxicam, and the like; narcotic antagonists such as, e.g., naloxone, nalorphine, and the like; antiparkinsonism agents such as, e.g., bromocriptine, piperidin, benzhexyl, benztropine, and the like; antidepressants such as, e.g., imipramine, nortriptyline, pritiptylene, and the like; antibiotic agents such as, e.g., clindamycin, erythromycin, fusidic acid, gentamicin, mupirocien, amfomycin, neomycin, metronidazole, silver sulphadiazine, sulphamethizole, bacitracin, framycetin, polymycin B, acitromycin, and the like; antifungal agents such as, e.g., miconazol, ketoconazole, clotrimazole, amphotericin B, nystatin, mepyramin, econazol, fluconazol, flucytocine, griseoftdvin, bifonazole, amorolfine, mycostatin, itraconazole, terbenafine, terconazole, tolnaftate, and the like; antimicrobial agents such as, e.g., metronidazole, tetracyclines, oxytetracycline, and the like; antiemetics such as, e.g., metoclopramide, droperidol, haloperidol, promethazine, and the like; antihistamines such as, e.g., chlorpheniramine, terfenadine, triprolidine, and the like; antimigraine agents such as, e.g., dihydroergotamine, ergotamine, pizotyhne, and the like; coronary, cerebral or peripheral vasodilators such as, e.g., nifedipine, diltiazem, and the like; antianginals such as, e.g., glyceryl nitrate, isosorbide denitrate, molsidomine, verapamil, and the like; calcium channel blockers such as, e.g., verapamil, nifedipine, diltiazem, nicardipine, and the like; hormonal agents such as, e.g., estradiol, estron, estriol, polyestradiol, polyestriol, dienestrol, diethylstilbestrol, progesterone, dihydroergosterone, cyproterone, danazol, testosterone, and the like; contraceptive agents such as, e.g., ethynyl estradiol, lynestrenol, etynodiol, norethisterone, mestranol, norgestrel, levonorgestrel, desogestrel, medroxyprogesterone, and the like; antithrombotic agents such as, e.g., heparin, warfarin, and the like; diuretics such as, e.g., hydrochlorothiazide, flunarizine, minoxidil, and the like; antihypertensive agents such as, e.g., propanolol, metoprolol, clonidine, pindolol, and the like; corticosteroids such as, e.g., beclomethasone, betamethasone, betamethasone-17-valerate, betamethasone-dipropionate, clobetasol, clobetasol-17-butyrate, clobetasol-propionate, desonide, desoxymethasone, dexamethasone, diflucortolone, flumethasone, flumethasone-pivalate, fluocinolone acetonide, fluocinonide, hydrocortisone, hydrocortisone-17-butyrate, hydrocortisone-buteprate, methylprednisolone, triamcinolone acetonide, budesonide, halcinonide, fluprednide acetate, alklometasone-dipropionate, fluocortolone, fluticason-propionate, mometasone-furate, desoxymethasone, diflurason-diacetate; halquinol, cliochinol, chlorchinaldol, fluocinolone-acetonid, and the like; dermatological agents such as, e.g., nitrofurantoin, dithranol, clioquinol, hydroxyquinoline, isotretionin, methoxsalen, methotrexate, tretionin, trioxsalen, salicylic acid, penicillamine, and the like; steroids such as, e.g., estradiol, progesterone, norethindrone, levonorgestrol, ethynodiol, levenorgestrel, norgestimate, gestanin, desogestrel, 3-keton-desogestrel, demegestone, promethoestrol, testosterone, spironolactone, and esters thereof, nitro compounds such as, e.g., amyl nitrates, nitroglycerine and isosorbide nitrates, opioid compounds such as, e.g., morphine and morphine-like drugs such as buprenorphine, oxymorphone, hydromorphone, levorphanol, fentanyl and fentanyl derivatives and analogues, prostaglandins such as, e.g., a member of the PGA, PGB, PGE, or PGF series such as, e.g., misoprostol, dinoproston, carboprost or enaprostil, a benzamide such as, e.g., metoclopramide, scopolamine, a peptide such as, e.g., growth hormone releasing factors, growth factors (epidermal growth factor (EGF), nerve growth factor (NGF), TGF, PDGF, insulin growth factor (IGF), fibroblast growth factor (FGFα, FGFβ, etc.), and the like), somatostatin, calcitonin, insulin, vasopressin, interferons, interleukins, e.g., IL-2, IL-12, IL-21, urokinase, serratiopeptidase, superoxide dismutase (SOD), thyrotropin releasing hormone (TRH), luteinizing hormone releasing hormone (LH-RH), corticotrophin releasing hormone (CRF), growth hormone releasing hormone (GHRH), oxytocin, erythropoietin (EPO), colony stimulating factor (CSF), and the like, a xanthine such as, e.g., caffeine, theophylline, a catecholamine such as, e.g., ephedrine, salbutamol, terbutaline, a dihydropyridine such as, e.g., nifedipine, a thiazide such as, e.g., hydrochlorotiazide, flunarizine, others such as, e.g., propanthelin, silver nitrate, enzymes like Streptokinases, Streptodases, vitamins like vitamin A, tretionin, isotretionin, acitretin, vitamin D, calcipotriol, interferon-α-2b, selen disulfide, pyrethione. It will be understood that the compositions of the invention can also comprise combinations of active substances, e.g., an active substance together with a potentiator therefor. It will of course also be understood that in the aspects of the invention wherein there is no specific requirement to the active substance, e.g., with respect to solubility, any substance which has a therapeutic or prophylactic activity can be incorporated in the composition.


Pharmaceutically Acceptable Excipients for Use in a Viscous Liquid Composition


An aspect of the invention relates to compositions wherein at least a part of the viscous liquid composition at room temperature can be substituted by certain pharmaceutically acceptable excipients. Addition of a pharmaceutically acceptable excipient to a viscous liquid composition or a precursor composition will normally lead to a disruption in the viscous liquid composition. Therefore, such substance is generally only added in very small concentrations such as, e.g., about 1-5% by weight based on the total composition. Certain pharmaceutically acceptable excipients can be added in much larger concentrations without having any substantially negative influence on the biopharmaceutical properties of the composition. Thus, the concentration of such excipients can be at least about 5% by weight such as, e.g., at least about 8%, 9%, 10%, 15% or 20% by weight.


Examples of suitable pharmaceutically acceptable excipients can be found e.g., among inert diluents or fillers selected from the group consisting of sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, sodium phosphate, and a polysaccharide such as, e.g., carmelose, a chitosan, a pectin, xanthan gum, a carrageenan, locust bean gum, acacia gum, a gelatin, an alginate, and dextrans and salts thereof.


Examples of suitable pharmaceutically excipients which can be soluble in the second substance or in the liquid crystalline phase are e.g., sorbitan esters such as, e.g., polysorbates; and macrogols. In the present context, solvents like e.g., water, glycerol, alcohols like e.g., ethanol and isopropylalcohol are examples of a liquid medium and are not intended to be examples of soluble pharmaceutically acceptable excipients.


Pharmaceutical Compositions and Routes of Administration


The viscous liquid composition in an osmotic delivery system is intended for parenteral administration and for topical administration to skin or mucosa. Other applications can of course also be relevant such as, e.g., application on dentures, prostheses and application to body cavities such as the oral cavity. The mucosa is preferably selected from oral, nasal, aural, lung, rectal, vaginal, and gastrointestinal mucosa.


A bioadhesive composition for administration according to the invention can also be in the form of a multiple unit composition, in the form of, e.g., a powder. A multiple unit composition can be administered to skin or mucosa, preferably the mucosa is selected from oral, nasal, rectal, aural, vaginal, lung, and gastrointestinal mucosa. Most preferred is a bioadhesive composition intended for administration to the gastrointestinal tract.


Bioadhesive compositions according to the invention for application on skin can comprise a polysaccharide in a concentration of at least 15% w/w, calculated on the total weight of the composition. The polysaccharide is preferably selected from the group consisting of carmelose, chitosan, pectins, xanthan gums, carrageenans, locust bean gum, acacia gum, gelatins, alginates, and dextrans, and salts thereof. The compositions can be easy to apply on the wound and are believed to be able to extract water from the wound and thereby drying the wound. Apart from the active or protective substance and the bioadhesive fatty acid ester substance, the bioadhesive compositions for use according to the invention can comprise pharmaceutically or cosmetically acceptable excipients or additives normally used in pharmaceutical compositions.


The bioadhesive compositions can be in form of, e.g., a spray, a solution, a dispersion, a suspension, an emulsion, powders, gels including hydrogels, pastes, ointments, creams, drenches, delivery devices, suppositories, enemas, implants, aerosols, microcapsules, microspheres, nanoparticles, liposomes, dressings, bandages, plasters, tooth paste, dental care compositions, and in other suitable form. The bioadhesive compositions can be formulated according to conventional pharmaceutical practice, see, e.g., “Remington's Pharmaceutical Sciences” and “Encyclopedia of Pharmaceutical Technology”, edited by Swarbrick, J. & J. C. Boylan, Marcel Dekker, Inc., New York, 1988. Pharmaceutically acceptable excipients for use in bioadhesive compositions for use according to the invention can be, for example, inert diluents or fillers, such as sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate or sodium phosphate; and lubricating agents including glidants and antiadhesives, for example, magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils or talc. Other pharmaceutically acceptable excipients can be colorants, flavouring agents, plasticizers, humectants, buffering agents, solubilizing agents, release modulating agents, etc. For application to the rectal or vaginal mucosa suitable compositions for use according to the invention include suppositories (emulsion or suspension type), suspensions, solutions, enemas, and rectal gelatin capsules (solutions or suspensions). Appropriate pharmaceutically acceptable suppository bases include cocoa butter, esterified fatty acids, glycerinated gelatin, and various water-soluble or dispersible bases like polyethylene glycols and polyoxyethylene sorbitan fatty acid esters. Various additives like, e.g., enhancers or surfactants can be incorporated.


For application to the nasal mucosa, nasal sprays and aerosols for inhalation can be suitable compositions for use according to the invention. In a typically nasal formulation, the active ingredients can be dissolved or dispersed in a suitable vehicle. The pharmaceutically acceptable vehicles and excipients and optionally other pharmaceutically acceptable materials present in the composition such as diluents, enhancers, flavouring agents, preservatives etc. are all selected in accordance with conventional pharmaceutical practice in a manner understood by the persons skilled in the art of formulating Pharmaceuticals.


For application to the oral cavity, teeth, skin or nail, the compositions for use according to the invention can contain conventionally non-toxic pharmaceutically acceptable carriers and excipients including microspheres and liposomes. The formulations include creams, ointments, lotions, liniments, gels, hydrogels, suspensions, solutions sticks, sprays, pastes, dressings, bandages, plasters, tooth paste, dental care compositions, and the like. The pharmaceutically acceptable carriers or excipients can include emulsifying agents, stabilizing agents, antioxidants, buffering agents, preservatives, humectants, penetration enhancers, chelating agents, gel forming agents, ointment bases, perfumes and skin protective agents.


Examples of emulsifying agents can be naturally occurring gums, e.g., gum acacia or gum tragacanth, naturally occurring phosphatides, e.g., soybean lecithin and sorbitan monooleate derivatives. Examples of antioxidants can be butylated hydroxy anisole (BHA), ascorbic acid and derivatives thereof, a-tocopherol and derivatives thereof, vitamin E, salts of sulphur dioxide, cysteine, citric acid, ascorbyl palmitate, butylhydroxytoluene, complexing agents, chelating agents, sodium pyrosulfite, EDTA and gallic acid esters. Examples of preservatives can be parabens, such as methyl, ethyl, propyl p-hydroxybenzoate, butylparaben, isobutylparaben, isopropylparaben, potassium sorbate, sorbic acid, benzoic acid, methyl benzoate, phenoxyethanol, bronopol, bronidox, MDM hydantoin, iodopropynyl butylcarbamate, EDTA, propyleneglycol (increases the solubility of preservatives) benzalconium chloride, benzylalcohol, chlorhexidine diacetate, chlorhexidine digluconate, chlorbntol, phenetanol, phenols (phenol, o-cresol, p-cresol, chlorcresol, tricresol), alkanols (chlorbutanol, phenetanol), sorbic acid, and mercuri-compounds like e.g., phenylmercurinitrate. Examples of humectants can be glycerin, propylene glycol, sorbitol and urea. Examples of suitable release modulating agents for use according to the invention can be glycerol, sesame oil, soybean oil, lecithin and cholesterol. Examples of penetration enhancers can be oleic acid, propylene glycol, DMSO, triethanolamine, N,N-dimethylacetamider N,N-dimethylformamide, 2-pyrrolidone and derivatives thereof, -tetrahydrofuryl alcohol and Azone. Examples of chelating agents can be sodium EDTA, citric acid and phosphoric acid. Examples of other excipients for use in compositions for use according to the invention can be edible oils like almond oil, castor oil, cacao butter, coconut oil, corn oil, cottonseed oil, linseed oil, olive oil, palm oil, peanut oil, poppyseed oil, rapeseed oil, sesame oil, soybean oil, sunflower oil, and teaseed oil; and of polymers such as carmelose, sodium carmelose, hydroxypropylmethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, chitosane, pectin, xanthan gum, carrageenan, locust bean gum, acacia gum, gelatin, and alginates, and solvents such as, e.g., glycerol, ethanol, propylene glycol, polyethylene glycols such as PEG 200 and PEG 400, Pluronic, polysorbate, and ethylene glycol. Examples of ointment bases can be beeswax, paraffin, cetyl palmitate, vegetable oils, sorbitan esters of fatty acids (Span®), Carbopol®, polyethylene glycols, and condensation products between sorbitan esters of fatty acids and ethylene oxide, e.g., polyoxyethylene sorbitan monooleate (Tween®).


Pharmaceutically acceptable carriers can be determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions for administering a pharmaceutically active agent in a viscous liquid composition (see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa. 18th ed., 1990, incorporated herein by reference). The pharmaceutical compositions are generally formulated as sterile, substantially isotonic and in full compliance with all Good Manufacturing Practice (GMP) regulations of the U.S. Food and Drug Administration.


Reference to “performance modifiers” means those materials that help to improve the stability and/or performance of lipid-based suspending vehicles. In some embodiments, such improvements include changing the properties of a two-component vehicle, that is already a stable solution of lipid in non-aqueous solvent, by altering the viscosity or other properties of a vehicle. Other aspects of the invention include improving the solubility of one component in the other, thus stabilizing the vehicle.


Performance modifiers that reduce stiff phases that are sometimes formed on contact of a amphiphilic-based suspending vehicle with aqueous medium include, but are not limited to, ethyl acetate, sesame oil, dioctyl sulfosuccinate, cholesterol, polysorbate 20, polysorbate 80, sodium dodecyl sulfate, fatty acids like oleic, stearic, linoleic, myristic, tocopheryl acetate, and vitamin E TPGS.


Performance modifiers that are cosolvents, that may act a dissolution rate enhancer, phase modifier, viscosity modifier or processing aid include but are not limited to ethanol, tocopherols, vegetable oils (sesame, cottonseed, safflower, coconut, soybean, olive), caprylic/capric triglycerides, ethyl acetate, benzyl alcohol, glycofurol, theyl oleate, N, N dimethyl acetamide, polyoxaester liquid polymers, N-methylpyrrolidone, polysorbate 80, polysorbate 20, castor oil, isopropyl myristate, triacetin, polyethylene glycol, lauryl lactate, lauryl alcohol, and combinations thereof.


Performance modifiers in accordance with the present invention can be used, for example, to improve the stability and/or the flowability of lipid-based suspending vehicles. For example, the use of performance modifiers aids in softening formations made at exit ports upon contact by the suspending vehicle with aqueous media, for example, bodily fluids. Performance modifiers, such as co-solvents, can also speed-up preparation of suspending vehicles and suspensions due to increased solubility of the lipid in co-solvents relative to a non-aqueous solvent. Performance modifiers can also be useful for reducing viscosity variations over small composition variations.


Reference to “suspending vehicle” means that the pharmaceutically active agent is substantially insoluble therein. Materials that are substantially insoluble generally remain in their original physical form throughout the lifespan of a dosage form containing the suspension. For example, solid particulates would generally remain particles. If necessary, the suspending vehicle may have other materials dissolved in them.


Reference to “stiff gels” means the gels that form within a suspending vehicle or pharmaceutical suspension upon contact with an aqueous medium that may be pliable but are substantially hard enough that they would impede flow out of a dosage form.


Furthermore, relevant compositions and conditions to be fulfilled for the individual components in the compositions are claimed in the claims and described in the Examples.


Example 1

An exemplary embodiment of liquid viscous compositions comprising a therapeutic protein or drug is the protein ω-interferon (ω-IFN) in a liquid composition of 1,2-Dioleoy-sn-glycero-3-phosphatidylcholine (DOPC):N-methylpyrrolidone (NMP) (3:1 w/w) or in a liquid composition of DOPC:lauryl lactate (LL) (3:2 w/w). A summary of results demonstrates that: (1) Protein release occurs from the liquid viscous compositions into a sink of aqueous media; (2) Release rate of ω-IFN from a composition comprising lauryl lactate:DOPC vehicles demonstrates that release occurs at a therapeutically effective rate to confirm feasibility; (3) ω-IFN in anhydrous suspension is stable; (4) α-tocopherol in combination with DOPC or DMPC can further include compounds to modify the rheological properties of the vehicle itself.


The objectives of the study are to: (a) assess the preliminary stability of ω-interferon at 40° C. in vehicles composed of a lipid (1,2-Dioleoy-sn-glycero-3-phosphatidylcholine (DOPC)) and a solvent (lauryl lactate (LL) or N-methylpyrrolidone (NMP)), (b) investigate the release of ω-interferon from these vehicles into aqueous media, and (c) evaluate pump performance using lipid-based vehicles in a release rate study.


1.1 Plan of Study









TABLE 1







Characterization of Various materials









Material
Test
Method





NMP
Peroxide Content
EP 2000 2.5.5 Method A or




Oxis Method


Vehicles
Moisture Content
Karl Fischer


Protein Particles
Protein Content
RP-HPLC and SEC









Preparation and Evaluation of Suspension


Control particles (1:2:1 Protein:Sucrose:Methionine with 25 mM citrate buffer) was used for the study. The particle loading was approximately 10%, which is equivalent to drug loading of 1.67%. This is consistent with a unit dose of 25 μg/day.


The suspension is prepared in a dry box under nitrogen. Suspension composition is presented in Table 2 and 3.









TABLE 2





Composition of DOPC/NMP Formulation
















DOPC/NMP Formulation ID
PDP7-176-1, 3


Particle Source
Spray-dried in Palo Alto



facility


Vehicle Loading (3:1 DOPC:NMP)
  90%


Particle Loading
  10%


Protein Concentration
1.67%
















TABLE 3





Composition of DOPC/LL Formulation


















DOPC/LL Formulation ID
PDP7-176-2, 4



Particle Source
Spray-dried in Palo Alto




facility



Vehicle Loading (3:2 DOPC:LL)
  90%



Particle Loading
  10%



Protein Concentration
1.67%









Stability of the suspension was measured after storage at 40° C. under nitrogen in lyophilization vials. Stability samples was tested in triplicate at t=0, 2, and 4 weeks (5 mg co-interferon per timepoint). Analysis was performed using RP-HPLC to determine purity with respect to oxidation and deamidation and using SEC to determine purity with respect to aggregation and precipitation.


A dissolution study was run with 6 mg formulation in 2 mL phosphate buffered saline (PBS) in Vacutainers®. These samples was incubated at 40° C. and tested at t=0, 1, 3, and 7 days.


Release rate formulations was prepared after results for t=0 stability and t=1 week dissolution samples are generated (assuming reasonable protein recoveries).









TABLE 4







Tests and Observations to be made on the Dynamic Capillary Tube


Samples in Exp. PDP7-176








Test to be Performed
Frequency and Details of Test





X-Ray to determine location
Before immersing membrane in


of piston
PBS buffer and approximately



every two weeks thereafter


Quantify soluble and insoluble ω-
Samples were collected


interferon released from implant
approximately twice per week.



Quantify protein with HPLC or other



analytical method. Guanidine HCl



used to redissolve insoluble protein.


Visual observations of glass
Approximately weekly once glass


capillary tubes
tubes are immersed in PBS.


Visual measurement of water
Weekly once glass tubes are


ingress into glass capillary tubes
immersed in PBS.









Suspension Preparation


Suspensions for stability and dissolution testing was prepared at a target batch size of 2.0 grams. This batch size is determined by the quantity of protein particles required. Suspension is prepared in a dry box under nitrogen.


Lauryl Lactate/Dopc Vehicle and Suspension Preparation for Use in Release Rate, Stability, and Dissolution Testing

  • 1. Preheat a hot plate to maintain a target surface temperature of 80° C.
  • 2. Weigh 3.00 g DOPC
  • 3. Weigh 2.00 g LL
  • 4. Heat to 80° C. on hot plate and mix with stainless steel spatula by hand
  • 5. Transfer 2.025 g of Vehicle and add 0.225 g of protein particles.
  • 6. Using a stainless steel spatula, manually incorporate the protein particles into the vehicle. Continue to hand mix the suspension for 15 minutes while warming on the hot plate.
  • 7. Fill suspension into syringe, de-aerate under vacuum, seal syringe and store refrigerated.


NMP/DOPC Vehicle and Suspension Preparation for Use in Release Rate Testing

  • 1. Preheat a hot plate to maintain a target surface temperature of 80° C.
  • 2. Weigh 3.75 g DOPC
  • 3. Weigh 1.25 g NMP
  • 4. Heat to 80° C. on hot plate and mix with stainless steel spatula by hand
  • 5. Transfer 2.025 g of Vehicle and add 0.225 g of protein particles.
  • 6. Using a stainless steel spatula, manually incorporate the protein particles into the vehicle. Continue to hand mix the suspension for 15 minutes while warming on the hot plate.
  • 7. Fill suspension into syringe, de-aerate under vacuum, seal syringe and store refrigerated.


1.2 Particle Preparation


Protein-excipient solution was spray dried to result in spray-dried particles. Particle size can be from about 1 to about 10 μm in diameter. Other procedures for creating particles include, but are not limited to, lyophilization or supercritical fluid processing.


1.3 System Assembly


Systems were assembled by loading suspension of the liquid composition into a formulation compartment of the osmotic delivery device. A cap with an orifice was slowly screwed onto the shell of the osmotic delivery device. The exit orifice serves as a diffusion moderator for release of the liquid composition.


Example 2

Table 5 shows the solubility of a therapeutic protein, ω-interferon, in an non-aqueous liquid solvent. Thirteen non-aqueous liquid solvent candidates were identified to develop a solution formulation or a suspension formulation based on solubility of ω-interferon. The solvents at the top, e.g., lauryl lactate, are potential suspension solvents. The character of the solvents changes to potential solution solvents in combination with ω-interferon, e.g., glycerol formal, benzyl benzoate, DMSO, or glycerin.


Non-aqueous liquid solvents, such as glycerol formal, benzyl benzoate, DMSO, or glycerin are useful in combination with small molecule therapeutic drugs, therapeutic lipid prodrug, or therapeutic protein to form suspension formulations with drugs. Furthermore, non-aqueous liquid solvents, such as lauryl lactate, lauryl alcohol, NMP, ethanol, PEG 400, 1:1 benzyl benzoate:benzyl alcohol, benzyl alcohol, ethyl hexyl lactate are useful in combination with small molecule therapeutic drugs, therapeutic lipid prodrug, or therapeutic protein to form suspension formulations with drugs.









TABLE 5







ω-Interferon Solubility in Non-aqueous liquid solvents












Concentration

Concentration
Expected



by UV
Concentration
by SEC
Cone


Solvent
(mg/ml)
by rp-HPLC
(mg/ml)
(mg/ml)














Lauryl lactate
<0.1
N/A
N/A
10.00


Lauryl alcohol
<0.1
N/A
N/A
10.00


NMP
<0.1
0.00
0.00
25.00


Ethanol
<0.1
0.00
0.00
25.00


PEG 400
<0.1
0.00
0.00
25.00


1:1 Benzyl benzoate:
<0.1
0.00
0.00
10.00


Benzyl alcohol






Benzyl alcohol
0.14
0.00
0.00
10.00


Ethyl hexyl lactate
0.37
0.00
0.00
10.00









Example 3


FIG. 1 shows the shear rate effect on various DOPC vehicle formulations at 37° C. DOPC vehicle formulations that were tested include, DOPC/DMSO (3:1), DOPC/Gly (2.45:1), DOPC/NMP (2:1), DOPC/LL (2:1), DOPC/LA (2:1). The results demonstrate that at increasing shear rate DOPC formulations can be shear thinning. Shear thinning is an important property to facilitate the manufacturing process of the viscous liquid formulations.


Example 4


FIG. 2 shows the temperature effect on viscosity of DOPC formulations. The viscosity of various DOPC formulations was measured at various temperatures under a shear stress of 0.04 per second. The results indicate that viscosity of DOPC formulations is temperature dependent.


Example 5


FIG. 3 shows the shear rate effect on various PHSPC vehicle formulations at 37° C. PHSPC vehicle formulations that were tested include PHSPC/GF (1:1), PHSPC/LL (1:1), PHSPC/LA (2:1), PHSPC/GF (2.5:1). The results demonstrate that at increasing shear rate PHSPC formulations can be shear thinning. Shear thinning is an important property to facilitate the manufacturing process of the viscous liquid formulations.


Example 6


FIG. 4 shows the temperature effect on viscosity of PHSPC formulations. The viscosity of various PHSPC formulations was measured at various temperatures under a shear stress of 0.04 per second. The results indicate that viscosity of PHSPC formulations is temperature dependent.


Example 7


FIG. 5 displays images of DOPC formulations, e.g., DOPC/NMP (3:1), DOPC/LL (1:1), DOPC:H2O (1:9) visualized by polarized microscopy. All samples are birefringent, suggesting formation of lipid vesicles in the suspension.


Example 8


FIG. 6 displays images of PHSPC formulations, e.g., PHSPC/GF (1:1), PHSPC/LL (1:1), PHSPC:H2O (1:9), PHSPC/LA (2:1), as visualized by polarized microscopy. PHSPC/LL formulation is not birefringent suggesting dissolution of PHSPC in LL. However all other samples are birefringent, suggesting formation of lipid vesicles in the suspension.


Example 9

Table 6 summarizes the solubility and rheology of lipids in various solvents. Parameters measured were viscosity at 37° C. (in poise), shear thinning at 37° C. (sec−1), temperature effect on viscosity (poise).









TABLE 6







Rheology and Solubility of Lipids in Non-aqueous liquid solvents



















Temp






Lipid:Solvent
Viscosity at
Shear Thinning
effect
Viscosity


Lipid
Solvent
Ratio
37° C. (poise)
at 37° C. (1/s)
(° C.)
(poise)
Comments


















DOPC
BA
1.35
1




Low viscosity to the eye


DOPC
BA
1.78
1




Low viscosity to the eye


DOPC
NMP
2
1
33000-170 
 0.04-10
5-47
114000-1380


DOPC
NMP
2.8
1
12900
0.04
5-70
 12600-8200
No stress sweep data


DOPC
NMP
3.5
1
19500
0.04
5-70
200000-9000
No stress sweep data


DOPC
GF
0.5
1




Low viscosity to the eye


DOPC
GF
1
1




Low viscosity to the eye


DOPC
GF
2.45
1
10
No
No


DOPC
DMSO
3
1
35000-700 
  0.04-2.5
5-70
  5e4-5e3


DOPC
LL
2
1
420000-7QOQO
  0.04-0.25
5-70
  2e6-4.5e4


DOPC
LA
2
1
8760-870
0.04-1
10-35 
 3.7e5-4.8e4
Gel crash above 35° C.


PHSPC
GF
1
1
5220-440
0.04-1
5-70
  5e3-9.5e−1


PHSPC
GF
2.5
1
1300-700
0.04-1
10-70 
2.25e4-2.8e2


PHSPC
LL
1
1
13000-130 
  0.04-0.65
5-70
  2e4-3


PHSPC
LA
2
1
125
No
20-70 
  7e3-5









Example 10

Table 7 summarizes moisture levels in lipid vehicles. The moisture level of the lipid, e.g., DOPC, HSPC, PHSPC, or DMPC, is measured after vacuum at room temperature for 48 hours. Lipid vehicles with lower moisture level can be included in viscous liquid compositions.









TABLE 7







Lipid Moisture Level










Lipid
Avg. Moisture (%)






DOPC
0.6 ± 0.2



HSPC
1.85 ± 0.64



PHSPC
1.02 ± 0.52



DMPC
0.6









Example 11


FIG. 7 shows a shear rate effect on a DOPC:LL vehicle formulation at 37° C. The vehicle formulations tested was DOPC/LL (3:2). The results demonstrate that at increasing shear rate, the DOPC/LL formulation can be shear thinning. At 0.014 sec−1, η=412,000 poise. At 0.16 sec−1, η=60,600 poise. In a diffusion moderator, the shear rate is approximately 10−4 to 10−2 sec−1. In a mixer, the shear rate is approximately 101 to 102 sec−1. The ratio of DOPC/LL can be adjusted to achieve an appropriate viscosity.


Example 12


FIG. 8 shows a shear rate effect on a DOPC:NMP vehicle formulation at 37° C. The vehicle formulations tested was DOPC/NMP (3:1). The results demonstrate that at increasing shear rate, the DOPC/NMP formulation can be shear thinning. At 0.024 sec−1, η=310,000 poise. At 9.7 sec−1, η=1,040 poise. The ratio of DOPC/NMP can be adjusted to achieve an appropriate viscosity.


Example 13


FIG. 9 shows the stability of ω-interferon in DOPC:LL suspension at 40° C. The percentage of dimer, oxide and deamide of to ω-interferon was measured at 0, 2, and 4 weeks for ω-interferon in DOPC:LL suspension by MeCl2:Methanol (95:5) or MeCl2:n-propanol (50:50) extraction. The DOPC:LL suspension particle at 0 weeks contained 1.63% oxide, 1.45% deamide, 7.94% unknown, and 0.07% dimer. ω-interferon in DOPC:LL suspension was stable for 4 weeks at 40° C.


Example 14

Table 8 shows to ω-interferon recovery from viscous liquid suspensions, DOPC:NMP or DOPC:LL. Assessment of in vitro release rate recovery was measured after adding 6 mg of viscous liquid suspensions to 2 ml of release rate medium (phosphate-buffered 0.8% saline). Release of ω-IFN from the suspension into the medium was measured from t=0 days to t=7 days. Percent recovery is based on a protein concentration of 1.67 wt % in the viscous liquid suspension formulation. The results indicate that greater than 90% soluble ω-IFN is recovered after incubation for seven days at 37° C.









TABLE 8







ω-Interferon Recovery from Viscous Liquid Suspension












% Recovery
% Recovery



Time (days)
NMP:DOPC
LL:DOPC














0
88.96 (5.56)
Not available



7
95.09 (5.3) 
91.16 (0.45)





% Recovery data is an average of 3 samples (±SD).






Example 15

In the study in Example 14 (Table 8), the DOPC:NMP viscous liquid suspension showed a slight increase in recovery of soluble ω-interferon from t=0 days to 7 days. A further experiment was performed to determine whether a delay in release of protein to release rate medium was due to the fact that viscous liquid suspensions do not dissolve or disperse in phosphate-buffered saline (PBS). To compare release rates, ω-interferon concentration was measured in aqueous solution. n-propanol was added to dissolve lipid, and ω-interferon was measured again to determine whether addition of non-aqueous liquid solvent result in higher protein recoveries.



FIG. 10 shows the effect of n-propanol on recovery of ω-interferon from DOPC:NMP or DOPC:LL suspensions. The results show an increase in total ω-IFN recovered into an aqueous phase from DOPC:NMP or DOPC:LL suspensions following treatment with N-propanol. Increased recovery following N-propanol treatment occurred in both delivery systems, glass cap and spiral DM.


Example 16

Water ingress into model diffusion moderators was measured for viscous liquid formulations, DOPC:LL or DOPC:NMP. See FIG. 11. Measured after 38 days, the distance of water ingress into capillary tubes of diffusion moderators was less than 5 mm for viscous liquid formulations, DOPC:LL or DOPC:NMP. Less water ingress was observed with the DOPC:LL formulation than with the DOPC:NMP formulation. Minimal water ingress into the device, as shown in FIG. 11, has been shown to be advantageous to protein delivery from the DUROS® osmotic delivery device.


Example 17

Additional solvents in combination with a lipid, e.g., DOPC, are useful in suspension formulations of viscous liquid compositions for parenteral drug delivery. Solvents that are useful in a viscous liquid formulation have hydrophobic character, for example, sesame oil, vitamin E (α-tocopherol), or silicon medical fluid (SMF, polydimethylsiloxane).


Example 18


FIG. 12 shows a measure of average protein released from a delivery device based upon piston movement within the device. Based on piston movement, delivery of the DOPC:LL viscous liquid formulation is at target. Volumetric flow rate is consistent for the periods between Day 7 to 21 and Day 21 to 35. Target flow rate is 1.5 μl per day. Density of the viscous liquid formulation is assumed to be approximately 1 gm/ml.


Example 19


FIG. 13 shows that piston movement in a delivery device correlates to the average protein released from the device. Release of soluble ω-interferon from DOPC:LL (3:2) formulations is closer to target when target is adjusted based on x-ray results of the delivery device. Release rate samples were treated with n-propanol. The target is based on 1.03% soluble ω-interferon in suspension.


Example 20

ω-interferon is stable in DOPC:sesame oil (1.5:1) viscous liquid composition for up to four weeks at 40° C. See Table 9. Percent oxidation shows an increase from t=0 weeks to t=4 weeks. Consistency and viscosity of the viscous liquid composition indicates that sesame oil can be added as a co-solvent with vitamin E (α-tocopherol) in combination with a lipid, e.g., DOPC.









TABLE 9







Stability of ω-interferon in DOPC: sesame oil viscous liquid compositions













%

%
%
%



Monomer
% Dimer
Oxidation
Deamidation
Unknown





Spray dried
99.67 (0.01)
0.23 (0.01)
 1.9 (0.39)
2.02 (0.01)
7.17 (0.44)


Particle







t = 0
99.87 (0.03)
0.14 (0.03)
 2.1 (0.17)
1.27 (0.37)
8.72 (0.82)


SesOil:DOPC







t = 2 wks
99.67 (0.02)
0.02 (0.01)
3.78 (0.03)
1.56 (0.04)
7.57 (0.56)


SesOil:DOPC







t = 4 wks
99.85 (0.00)
0.14 (0.01)
4.29 (0.13)
2.55 (0.10)
5.59 (0.24)


SesOil:DOPC





Data is an average of four samples (±SD).






Example 21

DOPC has been combined with various solvents (1:1 weight ratio) to yield a viscous liquid composition as shown in Table 10. Lipid and solvent are mixed and heated between approximately 65 to 100° C. with continued mixing. Suitability of various solvents in the composition was evaluated.









TABLE 10







Analysis of properties of various solvent in combination with lipid, DOPC









Solvent
Result
Action





Vitamin E (α-tocopherol)
Produces clear gel.
Pursue


Ethyl oleate
Clear, low viscosity gel.
Pursue


Caprylic capric triglyceride
Clear, low viscosity gel.
Pursue


Vegetable oils
With DOPC, physical separation



(sesame, soy, cottonseed oil)
seen over time.



Poly oxyl 35 castor oil
With DOPC, physical separation



(PEG 35 Castor Oil)
seen over time.



Silicone medical fluid (SMF)
DOPC not soluble in solvent.



Poly ethylene glycol 400 (PEG)
DOPC not soluble in solvent.



Polysorbate 20, 80
DOPC not soluble in solvent.









Example 22

Suspending vehicles comprising DOPC and Vitamin E were prepared at various weight ratios and viscosity was measured. FIG. 14 shows that viscosity increases by two orders of magnitude with a 5% change in composition. At a ratio of 0.75:1 DOPC:VitE, samples looked thin to the eye in that they flowed fairly easily when the sample vial was tipped on its side. At a ratio of 0.9:1, samples looked thicker than 0.75:1, and there was still flow when the sample vial was tipped on its side.


Example 23

A performance modifier is added to suspending vehicles of Example 22. It is desirable to reduce large variation of viscosity due to small variations in composition. A viscosity change of less than a factor of 10 is obtained over a composition change of 10% DOPC by weight.


Example 24

Suspending vehicles comprising DOPC and Vitamin E were prepared at various weight ratios and were exposed to aqueous media. In some examples, PBS was used as an aqueous medium. In other examples, Bovine Serum (adult) Sigma product B2771, lot 29H8401 was used. Samples were observed at room temperature at various times from 0 to 24 hours from the time that the aqueous media was added to the suspending vehicle. Table 11 shows that stiff phases, for example, gels, are produced upon contact of the suspending vehicles with aqueous media. Such formation of stiff phases could lead to plugging of a diffusion moderator of a dosage form, such as a DUROS device.









TABLE 11







Analysis of properties of various suspending vehicles without a performance


modifier upon contact with aqueous media










Aqueous Media




Added (% by



Sample (Weight ratios)
weight)
Result: Physical properties of resulting phase





DOPC:VitE 0.75:1
10% PBS
stiff but pliable


DOPC:VitE 0.75:1
25% PBS
stiff but pliable, excess water


DOPC:VitE 0.75:1
50% PBS
stiff but pliable, excess water


DOPC:VitE 1:1
10% PBS
stiff but pliable


DOPC:VitE 1:1
25% PBS
stiff but pliable, excess water


DOPC:VitE 1:1
50% PBS
stiff but pliable, excess water


DOPC:VitE 0.75:1
10% serum
Stiff, pliable, but more crumbly than when




PBS is added, excess aqueous phase


DOPC:VitE 0.75:1
25% serum
Stiff, pliable, but more crumbly than when




PBS is added, excess aqueous phase


DOPC:VitE 0.75:1
50% serum
Stiff, pliable, but more crumbly than when




PBS is added, excess aqueous phase









Example 25

Suspending vehicles comprising DOPC, Vitamin E, and a performance modifier were prepared. In some examples, the performance modifier was sesame oil. In one example, the performance modifier was ethyl acetate. PBS was used as an aqueous medium. Samples were observed at room temperature at various times from 0 to 24 hours from the time that the aqueous media was added to the suspending vehicle. Table 12 shows that unlike the suspending vehicles prepared without either sesame oil or ethyl acetate, soft phases are produced upon contact of the suspending vehicles with aqueous media. As such, the addition of sesame oil or ethyl acetate will soften gels or the like that result from the addition of aqueous media to suspending vehicles that do not contain a performance modifier.









TABLE 12







Analysis of properties of various suspending vehicles having a performance


modifier upon contact with aqueous media










Aqueous Media




Added (% by



Sample (Weight ratios)
weight)
Result: Physical properties of resulting phase





DOPC:VitE 0.75:1
25% PBS, then
softer



added 10% ethyl




acetate



DOPC:solvent 1.5:1
25% PBS
soft phase, very easy to move in vial


(Solvent = 75:25




VitE:SesOil)




DOPC:solvent 1.5:1
50% PBS
soft phase, very easy to move in vial


(Solvent = 75:25




VitE:SesOil)









Example 26

A suspending vehicle comprising DOPC and Vitamin E was prepared without a performance modifier according to the following.


Weighed appropriate amount of DOPC in a glass jar. Weighed appropriate amount of Vitamin E directly onto the DOPC. Allowed mixture to incubate in 90° C. oven for 10 minutes. Transferred to Keyence Hybrid mixer. A hybrid mixer operates by applying a large, continuous, centrifugal force, produced by combination of the rotation of the material container and the revolution in a certain radius, removing air bubbles from the material while blending its contents. Mixed for 20 minutes in Keyence mixer. Incubated at 37° C. overnight. Heated on 85° C. hotplate for 10 minutes.


Mixed by hand with spatula for 5 minutes. Incubated sample in 90° C. oven for 10 minutes. Transferred to Keyence Hybrid mixer. Mixed for 20 minutes in Keyence mixer. Heated on 85° C. hotplate for 10 minutes.


Mixed by hand with spatula for 10 minutes. Incubated sample in 90° C. oven for 10 minutes. Transferred to Keyence Hybrid mixer. Mixed for 20 minutes in Keyence mixer. Incubated at 37° C. overnight. Heated on 85° C. hotplate for 15 minutes.


Mixed by hand with spatula for 10 minutes. Transferred to Keyence Hybrid mixer. Mixed for 20 minutes in Keyence mixer.


Total processing time took ˜3 days to obtain a suspending vehicle that had DOPC substantially dissolved in Vitamin E.


Example 27

A suspending vehicle comprising DOPC and Vitamin E was prepared with a performance modifier, for example, ethanol, according to the following.


Weighed equal parts ethanol and DOPC into a test tube. Vortexed mixture for one minute on highest setting. Allowed mixture to sit for 5 minutes at room temperature. Weighed appropriate amount of Vitamin E into same test tube. Heated to 100-120° C. while mixing with a spring mixing blade for 1.5-3 hr under full vacuum. A spring mixing blade is essentially a spring attached to a straight rod.


Total processing time took ˜4 hours to obtain a suspending vehicle that had DOPC substantially dissolved in a combination of ethanol and Vitamin E.


Example 28

A pharmaceutical suspension comprising DOPC, Vitamin E, sesame oil, and co-interferon is prepared. The suspension is subjected to stability testing. It is desirable to obtain pharmaceutical suspensions that are shelf-stable for extended periods of time. The pharmaceutical suspension remains a homogeneous formulation for at least 3 months at 37° C. Visual observations of suspension appearance and measurement of drug levels throughout the suspension are used to determine that the suspension is homogenous.


When ranges are used herein for physical properties, such as molecular weight, or chemical properties, such as chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included.


The disclosures of each patent, patent application and publication cited or described in this document are hereby incorporated herein by reference, in their entirety.


Those skilled in the art will appreciate that numerous changes and modifications can be made to the embodiments of the invention and that such changes and modifications can be made without departing from the spirit of the invention. It is, therefore, intended that the appended claims cover all such equivalent variations as fall within the true spirit and scope of the invention.

Claims
  • 1. A method of manufacturing a viscous, non-aqueous, liquid pharmaceutical formulation, comprising: applying a shear rate to a viscous, non-aqueous, liquid-based suspending vehicle sufficient to produce a reduced viscosity liquid-based suspending vehicle;mixing particles comprising a pharmaceutically active agent into the reduced viscosity liquid-based suspending vehicle at a temperature from 80° C. to 120° C., wherein the particles further comprise sucrose, methionine, and citrate buffer; andprocessing the reduced viscosity liquid-based suspending vehicle to manufacture the viscous, non-aqueous, liquid pharmaceutical formulation,wherein the viscous, non-aqueous, liquid-based suspending vehicle comprises:an amphiphilic molecule; anda solvent selected from the group consisting of: lauryl lactate, lauryl alcohol, benzyl alcohol, benzyl benzoate, 1:1 benzyl benzoate:benzyl alcohol, ethyl hexyl lactate, glycerol formal, tetraglycol, N-1-methyl-2-pyrrolidone, dimethyl sulfoxide, polyethyleneglycol, triglycerides, ethanol, isopropanol, t-butyl alcohol, cyclohexanol, glycerin, glycerol, alpha-tocopherol, vegetable oil, sesame oil, soybean oil, cottonseed oil, and peanut oil wherein the pharmaceutically active agent is selected from the group consisting of a protein or a peptide.
  • 2. The method of claim 1, further comprising maintaining the viscous, non-aqueous, liquid-based suspending vehicle at a target temperature while applying the shear rate.
  • 3. The method of claim 2, further comprising maintaining the reduced viscosity liquid-based suspending vehicle at the target temperature while processing.
  • 4. The method of claim 1, further comprising maintaining the reduced viscosity liquid-based suspending vehicle at a target temperature while processing.
  • 5. The method of claim 1, further comprising: maintaining the viscous, non-aqueous, liquid-based suspending vehicle at a first target temperature while applying the shear rate; andmaintaining the reduced viscosity liquid-based suspending vehicle at a second target temperature while processing.
  • 6. The method of claim 1, wherein the amphiphilic molecule is selected from the group consisting of: fatty acid esters, phospholipids, polyethyleneglycol-lipids, block copolymer surfactants, and sorbitan esters.
  • 7. The method of claim 1, wherein the protein or the peptide is selected from the group consisting of omega-interferon, alpha-interferon, beta-interferon, gamma-interferon, erythropoietin, human growth hormone, granulocyte macrophage colony stimulating factor, human growth hormone releasing hormone, insulin, infliximab, glucagon-like peptide-1, and bone morphogenic protein.
  • 8. The method of claim 1, wherein the particles comprise spray-dried particles.
  • 9. The method of claim 8, wherein the particles comprise a diameter of about 1 μm to about 10 μm.
  • 10. The method of claim 4, wherein the processing further comprises loading the reduced viscosity liquid-based suspending vehicle comprising the particles into a chamber of an osmotic drug delivery device.
  • 11. The method of claim 1, wherein the viscous, non-aqueous, liquid-based suspending vehicle has a viscosity of about 5 poise to about 100,000 poise.
  • 12. The method of claim 1, wherein the shear rate is about 0.04 per second to about 10 per second.
  • 13. The method of claim 1, wherein the viscous, non-aqueous, liquid-based suspending vehicle comprises polyvinylpyrrolidone.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 13/090,947, filed on 20 Apr. 2011, which is a continuation of U.S. patent application Ser. No. 12/655,397, filed on 29 Dec. 2009, now U.S. Pat. No. 7,964,183, which is a continuation of U.S. patent application Ser. No. 10/988,716, filed on 15 Nov. 2004, now U.S. Pat. No. 7,731,947, which claims the benefit of U.S. Provisional Application Ser. No. 60/520,605, filed on 17 Nov. 2003, which applications are herein incorporated by reference in their entireties in the present application.

US Referenced Citations (578)
Number Name Date Kind
2110208 Eggert Mar 1938 A
2168437 Buercklin Aug 1939 A
3025991 Gillon Mar 1962 A
3122162 Sands Feb 1964 A
3625214 Higuchi Dec 1971 A
3632768 Bergy et al. Jan 1972 A
3732865 Higuchi et al. May 1973 A
3797492 Place Mar 1974 A
3869549 Geller Mar 1975 A
3987790 Eckenhoff et al. Oct 1976 A
3995631 Higuchi et al. Dec 1976 A
3995632 Nakano et al. Dec 1976 A
4008719 Theeuwes et al. Feb 1977 A
4034756 Higuchi et al. Jul 1977 A
4078060 Benson et al. Mar 1978 A
4111201 Theeuwes Sep 1978 A
4111202 Theeuwes Sep 1978 A
4211771 Witkowski et al. Jul 1980 A
4243030 Lynch et al. Jan 1981 A
4305927 Theeuwes et al. Dec 1981 A
4310516 Chang et al. Jan 1982 A
4340054 Michaels Jul 1982 A
4350271 Eckenhoff Sep 1982 A
4373527 Fischell Feb 1983 A
4376118 Daher et al. Mar 1983 A
4439196 Higuchi Mar 1984 A
4455143 Theeuwes et al. Jun 1984 A
4455145 Theeuwes Jun 1984 A
4552561 Eckenhoff et al. Nov 1985 A
4588614 Lauchenauer May 1986 A
4594108 Greminger, Jr. et al. Jun 1986 A
4609374 Ayer Sep 1986 A
4639244 Rizk et al. Jan 1987 A
4655462 Balsells Apr 1987 A
4673405 Guittard et al. Jun 1987 A
4675184 Hasegawa et al. Jun 1987 A
4695623 Stabinsky Sep 1987 A
4727138 Goeddel et al. Feb 1988 A
4734284 Terada et al. Mar 1988 A
4743449 Yoshida et al. May 1988 A
4753651 Eckenhoff Jun 1988 A
4762791 Goeddel et al. Aug 1988 A
4765989 Wong et al. Aug 1988 A
4783337 Wong et al. Nov 1988 A
4826144 Balsells May 1989 A
4830344 Balsells May 1989 A
4845196 Cowling Jul 1989 A
4847079 Kwan Jul 1989 A
4851228 Zentner et al. Jul 1989 A
4865845 Eckenhoff et al. Sep 1989 A
4873080 Brickl et al. Oct 1989 A
4874388 Wong et al. Oct 1989 A
4876781 Balsells Oct 1989 A
4885166 Meyer et al. Dec 1989 A
4886668 Haslam et al. Dec 1989 A
4892778 Theeuwes et al. Jan 1990 A
4893795 Balsells Jan 1990 A
4897471 Stabinsky Jan 1990 A
4907788 Balsells Mar 1990 A
4915366 Balsells Apr 1990 A
4915949 Wong et al. Apr 1990 A
4915954 Ayer et al. Apr 1990 A
4917887 Hauptmann et al. Apr 1990 A
4917895 Lee et al. Apr 1990 A
4927687 Nuwayser May 1990 A
4929554 Goeddel et al. May 1990 A
4931285 Edgren et al. Jun 1990 A
4934666 Balsells Jun 1990 A
4940465 Theeuwes et al. Jul 1990 A
4952402 Sparks et al. Aug 1990 A
4961253 Balsells Oct 1990 A
4964204 Balsells Oct 1990 A
4969884 Yum Nov 1990 A
4974821 Balsells Dec 1990 A
4976966 Theeuwes et al. Dec 1990 A
5004689 Fiers et al. Apr 1991 A
5006346 Theeuwes et al. Apr 1991 A
5019382 Cummins, Jr. May 1991 A
5023088 Wong et al. Jun 1991 A
5024842 Edgren et al. Jun 1991 A
5030216 Theeuwes et al. Jul 1991 A
5034229 Magruder et al. Jul 1991 A
5057318 Magruder et al. Oct 1991 A
5059423 Magruder et al. Oct 1991 A
5071642 Lahr et al. Dec 1991 A
5072070 Balsells Dec 1991 A
5079388 Balsells Jan 1992 A
5091188 Haynes Feb 1992 A
5108078 Balsells Apr 1992 A
5110596 Magruder et al. May 1992 A
5112614 Magruder et al. May 1992 A
5113938 Clayton May 1992 A
5117066 Balsells May 1992 A
5118666 Habener Jun 1992 A
5120306 Gosselin Jun 1992 A
5120712 Habener Jun 1992 A
5120832 Goeddel et al. Jun 1992 A
5122128 Cardinal et al. Jun 1992 A
5126142 Ayer et al. Jun 1992 A
5134244 Balsells Jul 1992 A
5137727 Eckenhoff Aug 1992 A
5151093 Theeuwes et al. Sep 1992 A
5160122 Balsells Nov 1992 A
5160743 Edgren et al. Nov 1992 A
5161806 Balsells Nov 1992 A
5180591 Margruder et al. Jan 1993 A
5190765 Jao et al. Mar 1993 A
5203849 Balsells Apr 1993 A
5207752 Sorensen et al. May 1993 A
5209746 Balaban et al. May 1993 A
5213809 Wright et al. May 1993 A
5219572 Sivaramakrishnan Jun 1993 A
5221278 Linkwitz et al. Jun 1993 A
5223265 Wong Jun 1993 A
5231176 Goeddel et al. Jul 1993 A
5234424 Yum et al. Aug 1993 A
5234692 Magruder et al. Aug 1993 A
5234693 Magruder et al. Aug 1993 A
5234695 Hobbs et al. Aug 1993 A
5252338 Jao et al. Oct 1993 A
5260069 Chen Nov 1993 A
5278151 Korb et al. Jan 1994 A
5279608 Cherif Cheikh Jan 1994 A
5284655 Bogdansky et al. Feb 1994 A
5290271 Jernberg Mar 1994 A
5300302 Tachon et al. Apr 1994 A
5308348 Balaban et al. May 1994 A
5312389 Theeuwes et al. May 1994 A
5312390 Wong May 1994 A
5318558 Linkwitz et al. Jun 1994 A
5318780 Viegas et al. Jun 1994 A
5320616 Magndu et al. Jun 1994 A
5324280 Wong et al. Jun 1994 A
5336057 Fukuda et al. Aug 1994 A
5352662 Brooks et al. Oct 1994 A
5368588 Bettinger Nov 1994 A
5371089 Rattan Dec 1994 A
5374620 Clark et al. Dec 1994 A
5385738 Yamahira et al. Jan 1995 A
5411951 Mitchell May 1995 A
5413572 Wong et al. May 1995 A
5413672 Hirotsuji et al. May 1995 A
5424286 Eng Jun 1995 A
5429602 Hauser Jul 1995 A
5443459 Wong et al. Aug 1995 A
5445829 Paradissis et al. Aug 1995 A
5456679 Balaban et al. Oct 1995 A
5458888 Chen Oct 1995 A
5464929 Bezwada et al. Nov 1995 A
5472708 Chen Dec 1995 A
5486365 Takado et al. Jan 1996 A
5498255 Wong et al. Mar 1996 A
5511355 Dingler Apr 1996 A
5512293 Landrau et al. Apr 1996 A
5512549 Chen et al. Apr 1996 A
5514110 Teh May 1996 A
5529914 Hubbell et al. Jun 1996 A
5531736 Wong et al. Jul 1996 A
5540665 Mercado et al. Jul 1996 A
5540912 Roorda et al. Jul 1996 A
5543156 Roorda et al. Aug 1996 A
5545618 Buckley et al. Aug 1996 A
5557318 Gabriel Sep 1996 A
5571525 Roorda et al. Nov 1996 A
5574008 Johnson et al. Nov 1996 A
5574137 Gray et al. Nov 1996 A
5580578 Oshlack et al. Dec 1996 A
5595751 Bezwada Jan 1997 A
5595759 Wright et al. Jan 1997 A
5597579 Bezwada et al. Jan 1997 A
5602010 Hauptmann et al. Feb 1997 A
5605688 Himmler et al. Feb 1997 A
5607687 Bezwada et al. Mar 1997 A
5609885 Rivera et al. Mar 1997 A
5614221 Fjellstrom Mar 1997 A
5614492 Habener Mar 1997 A
5618552 Bezwada et al. Apr 1997 A
5620698 Bezwada et al. Apr 1997 A
5620705 Dong et al. Apr 1997 A
5633011 Dong et al. May 1997 A
5635213 Nystrom et al. Jun 1997 A
5639477 Maruyama et al. Jun 1997 A
5645850 Bezwada et al. Jul 1997 A
5648088 Bezwada et al. Jul 1997 A
5660847 Magruder et al. Aug 1997 A
5660858 Parikh et al. Aug 1997 A
5668170 Gyory Sep 1997 A
5676942 Testa et al. Oct 1997 A
5686097 Taskovich et al. Nov 1997 A
5690925 Gray et al. Nov 1997 A
5690952 Magruder et al. Nov 1997 A
5698213 Jamiolkowski et al. Dec 1997 A
5700486 Canal et al. Dec 1997 A
5700583 Jamiolkowski et al. Dec 1997 A
5703200 Bezwada et al. Dec 1997 A
5711967 Juch Jan 1998 A
5713847 Howard, III et al. Feb 1998 A
5728088 Margruder et al. Mar 1998 A
5728396 Peery et al. Mar 1998 A
5733572 Unger et al. Mar 1998 A
5736159 Chen et al. Apr 1998 A
5738845 Imakawa Apr 1998 A
5747058 Tipton et al. May 1998 A
5756450 Hahn et al. May 1998 A
5782396 Mastri et al. Jul 1998 A
5795591 Lee et al. Aug 1998 A
5795779 McCormick et al. Aug 1998 A
5807876 Armistead et al. Sep 1998 A
5814323 Lyle Sep 1998 A
5817129 Labrecque et al. Oct 1998 A
5830501 Dong et al. Nov 1998 A
5836935 Ashton et al. Nov 1998 A
5843891 Sherman Dec 1998 A
5844017 Jamiolkowski et al. Dec 1998 A
5858746 Hubbell et al. Jan 1999 A
5859150 Jamiolkowski et al. Jan 1999 A
5861166 Eckenhoff Jan 1999 A
5871770 Margruder et al. Feb 1999 A
5874388 Hsu Feb 1999 A
5876746 Jona et al. Mar 1999 A
5882676 Lee et al. Mar 1999 A
5904935 Eckenhoff et al. May 1999 A
5906830 Farinas et al. May 1999 A
5908621 Glue et al. Jun 1999 A
5928666 Farinas et al. Jul 1999 A
5932547 Stevenson et al. Aug 1999 A
5938654 Wong et al. Aug 1999 A
5939286 Johnson et al. Aug 1999 A
5948430 Zerbe et al. Sep 1999 A
5958909 Habener Sep 1999 A
5962023 Jamiolkowski et al. Oct 1999 A
5972370 Eckenhoff et al. Oct 1999 A
5972373 Yajima et al. Oct 1999 A
5976109 Heruth Nov 1999 A
5985305 Peery et al. Nov 1999 A
5997527 Gumucio et al. Dec 1999 A
5997902 Maruyama et al. Dec 1999 A
6007805 Foster et al. Dec 1999 A
6017545 Modi Jan 2000 A
6022561 Carlsson et al. Feb 2000 A
6029361 Newman Feb 2000 A
6060450 Soos et al. May 2000 A
6069133 Carlo et al. May 2000 A
6074660 Jamiolkowski et al. Jun 2000 A
6074673 Guillen Jun 2000 A
6100346 Jamiolkowski et al. Aug 2000 A
6113938 Chen et al. Sep 2000 A
6124261 Stevenson et al. Sep 2000 A
6124281 Lewis et al. Sep 2000 A
6127520 Ueda et al. Oct 2000 A
6129761 Hubbell Oct 2000 A
6130200 Brodbeck et al. Oct 2000 A
6132420 Dionne et al. Oct 2000 A
6133249 Hills Oct 2000 A
6133429 Davis et al. Oct 2000 A
6147168 Jamiolkowski et al. Nov 2000 A
6156331 Peery et al. Dec 2000 A
6174547 Dong et al. Jan 2001 B1
6177096 Zerbe et al. Jan 2001 B1
6183461 Matsuura et al. Feb 2001 B1
6190350 Davis et al. Feb 2001 B1
6191102 DiMarchi et al. Feb 2001 B1
6204022 Johnson et al. Mar 2001 B1
6217906 Gumucio et al. Apr 2001 B1
6217908 Mathiowitz et al. Apr 2001 B1
6218431 Schoen et al. Apr 2001 B1
6224894 Jamiolkowski et al. May 2001 B1
6235712 Stevenson et al. May 2001 B1
6245357 Edgren et al. Jun 2001 B1
6248112 Gambale et al. Jun 2001 B1
6258377 New et al. Jul 2001 B1
6261584 Peery et al. Jul 2001 B1
6268343 Knudsen et al. Jul 2001 B1
6270787 Ayer Aug 2001 B1
6283949 Roorda Sep 2001 B1
6284264 Zerbe et al. Sep 2001 B1
6284725 Coolidge et al. Sep 2001 B1
6284727 Kim et al. Sep 2001 B1
6287295 Chen et al. Sep 2001 B1
6329336 Bridon et al. Dec 2001 B1
6331311 Brodbeck et al. Dec 2001 B1
6372218 Cummins, Jr. Apr 2002 B1
6372256 Jamiolkowski et al. Apr 2002 B2
6375978 Kleiner et al. Apr 2002 B1
6395292 Peery et al. May 2002 B2
6403655 Bezwada et al. Jun 2002 B1
6419952 Wong et al. Jul 2002 B2
6433144 Morris et al. Aug 2002 B1
6436091 Harper et al. Aug 2002 B1
6447522 Gambale et al. Sep 2002 B2
6451974 Hansen Sep 2002 B1
6458385 Jamiolkowski et al. Oct 2002 B2
6458924 Knudsen et al. Oct 2002 B2
6461605 Cutler et al. Oct 2002 B1
6464688 Harper et al. Oct 2002 B1
6468961 Brodbeck et al. Oct 2002 B1
6471688 Harper et al. Oct 2002 B1
6472512 LaFleur et al. Oct 2002 B1
6485706 McCoy et al. Nov 2002 B1
6506724 Hiles et al. Jan 2003 B1
6508808 Carr et al. Jan 2003 B1
6514500 Bridon et al. Feb 2003 B1
6514517 Jamilolkowski et al. Feb 2003 B2
6524305 Peterson et al. Feb 2003 B1
6528486 Larsen et al. Mar 2003 B1
6541021 Johnson et al. Apr 2003 B1
6544252 Theeuwes et al. Apr 2003 B1
6551613 Dong et al. Apr 2003 B1
6569420 Chen et al. May 2003 B2
6572890 Faour et al. Jun 2003 B2
6579851 Goeke et al. Jun 2003 B2
6592887 Zerbe et al. Jul 2003 B2
6593295 Bridon et al. Jul 2003 B2
6670368 Breault et al. Dec 2003 B1
6673767 Brodbeck et al. Jan 2004 B1
6682522 Carr et al. Jan 2004 B2
6703225 Kojima et al. Mar 2004 B1
6703359 Young et al. Mar 2004 B1
6706689 Coolidge et al. Mar 2004 B2
6709671 Zerbe et al. Mar 2004 B2
6720407 Hughes et al. Apr 2004 B1
6730328 Maskiwicz et al. May 2004 B2
6767887 Hoffmann et al. Jul 2004 B1
6821949 Bridon et al. Nov 2004 B2
6833256 Pontzer et al. Dec 2004 B1
6835194 Johnson et al. Dec 2004 B2
6840931 Peterson et al. Jan 2005 B2
6849708 Habener Feb 2005 B1
6849714 Bridon et al. Feb 2005 B1
6858576 Young et al. Feb 2005 B1
6872700 Young et al. Mar 2005 B1
6875748 Manthorpe et al. Apr 2005 B2
6887470 Bridon et al. May 2005 B1
6887849 Bridon et al. May 2005 B2
6899887 Ayer May 2005 B2
6902744 Kolterman et al. Jun 2005 B1
6903186 Dong Jun 2005 B1
6924264 Prickett et al. Aug 2005 B1
6939556 Lautenbach Sep 2005 B2
6956026 Beeley et al. Oct 2005 B2
6969702 Bertilsson et al. Nov 2005 B2
6976981 Ayer Dec 2005 B2
6989366 Beeley et al. Jan 2006 B2
6992065 Okumu Jan 2006 B2
6997922 Theeuwes et al. Feb 2006 B2
7014636 Gilbert Mar 2006 B2
7022674 DeFelippis et al. Apr 2006 B2
7041646 Pan et al. May 2006 B2
7074423 Fereira et al. Jul 2006 B2
7084243 Glaesner et al. Aug 2006 B2
7101567 Sano et al. Sep 2006 B1
7101843 Glaesner et al. Sep 2006 B2
7112335 Lautenbach Sep 2006 B2
7115569 Beeley et al. Oct 2006 B2
7138375 Beeley et al. Nov 2006 B2
7138486 Habener et al. Nov 2006 B2
7141547 Rosen et al. Nov 2006 B2
7144863 DeFelippis et al. Dec 2006 B2
7153825 Young et al. Dec 2006 B2
7157555 Beeley et al. Jan 2007 B1
7163688 Peery et al. Jan 2007 B2
7199217 DiMarchi et al. Apr 2007 B2
7205409 Pei et al. Apr 2007 B2
7207982 Dionne et al. Apr 2007 B2
7241457 Chen et al. Jul 2007 B2
7258869 Berry et al. Aug 2007 B1
7297761 Beeley et al. Nov 2007 B2
7316680 Gilbert Jan 2008 B2
7393827 Nadler Jul 2008 B2
7407499 Trautman Aug 2008 B2
7442682 Kitaura et al. Oct 2008 B2
7456254 Wright et al. Nov 2008 B2
7459432 Cowley et al. Dec 2008 B2
7521423 Young et al. Apr 2009 B2
7563871 Wright et al. Jul 2009 B2
7612176 Wright et al. Nov 2009 B2
7655254 Dennis et al. Feb 2010 B2
7655257 Peery et al. Feb 2010 B2
7666835 Bloom et al. Feb 2010 B2
7682356 Alessi et al. Mar 2010 B2
7727519 Moran Jun 2010 B2
7731947 Eliaz et al. Jun 2010 B2
7741269 Young et al. Jun 2010 B2
7825091 Bloom et al. Nov 2010 B2
7829109 Chen et al. Nov 2010 B2
7833543 Gilson et al. Nov 2010 B2
7879028 Alessi et al. Feb 2011 B2
7919109 Berry et al. Apr 2011 B2
7959938 Rohloff et al. Jun 2011 B2
7964183 Eliaz et al. Jun 2011 B2
8048438 Berry et al. Nov 2011 B2
8052996 Lautenbach et al. Nov 2011 B2
8058233 Cowley et al. Nov 2011 B2
8101576 Bloom Jan 2012 B2
8114430 Rohloff et al. Feb 2012 B2
8114437 Rohloff et al. Feb 2012 B2
8158150 Lautenbach et al. Apr 2012 B2
8173150 Berry et al. May 2012 B2
8206745 Rohloff et al. Jun 2012 B2
8211467 Rohloff et al. Jul 2012 B2
8217001 Cowley et al. Jul 2012 B2
8257691 Eliaz et al. Sep 2012 B2
8263545 Levy et al. Sep 2012 B2
8263736 Bloom Sep 2012 B2
8268341 Berry Sep 2012 B2
8273365 Lautenbach et al. Sep 2012 B2
8273713 Pittner et al. Sep 2012 B2
8278267 Weyer et al. Oct 2012 B2
8298561 Alessi et al. Oct 2012 B2
8299025 Alessi et al. Oct 2012 B2
8343140 Alessi et al. Jan 2013 B2
8367095 Lautenbach et al. Feb 2013 B2
8372424 Berry et al. Feb 2013 B2
8398967 Eliaz et al. Mar 2013 B2
8440226 Rohloff et al. May 2013 B2
8460694 Rohloff et al. Jun 2013 B2
8470353 Lautenbach et al. Jun 2013 B2
8801700 Alessi et al. Aug 2014 B2
8865202 Zerbe et al. Oct 2014 B2
8926595 Alessi et al. Jan 2015 B2
8940316 Alessi et al. Jan 2015 B2
8992962 Lautenbach et al. Mar 2015 B2
9095553 Rohloff et al. Aug 2015 B2
20010012511 Bezwada et al. Aug 2001 A1
20010021377 Jamiolkowski et al. Sep 2001 A1
20010021822 Ayer Sep 2001 A1
20010022974 Ayer Sep 2001 A1
20010026793 Jamiolkowski et al. Oct 2001 A1
20010027311 Chen et al. Oct 2001 A1
20010036472 Wong et al. Nov 2001 A1
20010037190 Beisswenger Nov 2001 A1
20020001631 Okumu Jan 2002 A1
20020004481 Cleland et al. Jan 2002 A1
20020012818 Ruppi et al. Jan 2002 A1
20020034532 Brodbeck et al. Mar 2002 A1
20020037309 Jaworowicz et al. Mar 2002 A1
20020048600 Bhatt et al. Apr 2002 A1
20020136848 Yoshii et al. Sep 2002 A1
20020137666 Beeley et al. Sep 2002 A1
20020141985 Pittner et al. Oct 2002 A1
20020197185 Jamiolkowski et al. Dec 2002 A1
20030032947 Harper et al. Feb 2003 A1
20030045454 Okumu et al. Mar 2003 A1
20030059376 Libbey, III et al. Mar 2003 A1
20030060425 Ahlem et al. Mar 2003 A1
20030104063 Babcock et al. Jun 2003 A1
20030108608 Laridon et al. Jun 2003 A1
20030108609 Berry et al. Jun 2003 A1
20030113380 Ramstack et al. Jun 2003 A1
20030114837 Peterson et al. Jun 2003 A1
20030118660 Rickey et al. Jun 2003 A1
20030138403 Drustrup Jul 2003 A1
20030138491 Tracy et al. Jul 2003 A1
20030157178 Chen et al. Aug 2003 A1
20030170289 Chen et al. Sep 2003 A1
20030180364 Chen et al. Sep 2003 A1
20030186858 Arentsen Oct 2003 A1
20030191099 Bohlmann et al. Oct 2003 A1
20030211974 Brodbeck et al. Nov 2003 A1
20030215515 Truong-Le et al. Nov 2003 A1
20040001689 Goldsmith et al. Jan 2004 A1
20040001889 Chen et al. Jan 2004 A1
20040002442 Pan et al. Jan 2004 A1
20040022859 Chen et al. Feb 2004 A1
20040024068 Levy et al. Feb 2004 A1
20040024069 Chen et al. Feb 2004 A1
20040029784 Hathaway Feb 2004 A1
20040039376 Peery et al. Feb 2004 A1
20040157951 Glaesner et al. Mar 2004 A1
20040097906 Fereira et al. May 2004 A1
20040101557 Gibson et al. May 2004 A1
20040102762 Gilbert May 2004 A1
20040115236 Chan et al. Jun 2004 A1
20040142867 Oi et al. Jul 2004 A1
20040142902 Struijker-Boudier Jul 2004 A1
20040151753 Chen et al. Aug 2004 A1
20040198654 Glaesner et al. Oct 2004 A1
20040209801 Brand et al. Oct 2004 A1
20040224903 Berry et al. Nov 2004 A1
20040225113 LaFleur et al. Nov 2004 A1
20040243106 Ayer Dec 2004 A1
20040265273 Li et al. Dec 2004 A1
20040266683 Hathaway et al. Dec 2004 A1
20040266692 Young et al. Dec 2004 A1
20050004557 Russell Jan 2005 A1
20050008661 Fereira et al. Jan 2005 A1
20050009742 Bertilsson et al. Jan 2005 A1
20050010196 Fereira et al. Jan 2005 A1
20050070883 Brown et al. Mar 2005 A1
20050079200 Rathenow et al. Apr 2005 A1
20050079202 Chen et al. Apr 2005 A1
20050095284 Trautman May 2005 A1
20050101943 Ayer et al. May 2005 A1
20050106214 Chen May 2005 A1
20050112188 Eliaz et al. May 2005 A1
20050118206 Luk et al. Jun 2005 A1
20050118221 Blakely et al. Jun 2005 A1
20050131386 Freeman et al. Jun 2005 A1
20050131389 Peterson et al. Jun 2005 A1
20050175701 Pan et al. Aug 2005 A1
20050201980 Moran Sep 2005 A1
20050215475 Ong et al. Sep 2005 A1
20050266087 Junnarkar et al. Dec 2005 A1
20050271702 Wright et al. Dec 2005 A1
20050276856 Fereira et al. Dec 2005 A1
20050281879 Chen et al. Dec 2005 A1
20060013879 Brodbeck et al. Jan 2006 A9
20060014678 Cowley et al. Jan 2006 A1
20060030526 Liu et al. Feb 2006 A1
20060069029 Kolterman et al. Mar 2006 A1
20060084604 Kitaura et al. Apr 2006 A1
20060094652 Levy et al. May 2006 A1
20060141040 Chen et al. Jun 2006 A1
20060142234 Chen et al. Jun 2006 A1
20060160736 Nadler Jul 2006 A1
20060178304 Juul-Mortensen et al. Aug 2006 A1
20060193918 Rohloff et al. Aug 2006 A1
20060216242 Rohloff et al. Sep 2006 A1
20060233841 Brodbeck et al. Oct 2006 A1
20060246138 Rohloff et al. Nov 2006 A1
20060251618 Dennis et al. Nov 2006 A1
20060263433 Ayer et al. Nov 2006 A1
20060264890 Moberg et al. Nov 2006 A1
20060280795 Penhasi et al. Dec 2006 A1
20060293232 Levy et al. Dec 2006 A1
20070027105 Junnarkar et al. Feb 2007 A1
20070149011 Kent et al. Jun 2007 A1
20070166352 Wright et al. Jul 2007 A1
20070248572 Moran et al. Oct 2007 A1
20070281024 Lautenbach et al. Dec 2007 A1
20080020016 Li et al. Jan 2008 A1
20080038316 Wong et al. Feb 2008 A1
20080064636 Bloom et al. Mar 2008 A1
20080091176 Alessi et al. Apr 2008 A1
20080110515 Angelosanto et al. May 2008 A1
20080112994 Junnarkar et al. May 2008 A1
20080200383 Jennings et al. Aug 2008 A1
20080207512 Roth et al. Aug 2008 A1
20080226625 Berry et al. Sep 2008 A1
20080226689 Berry et al. Sep 2008 A1
20080260838 Hokenson et al. Oct 2008 A1
20080260840 Alessi et al. Oct 2008 A1
20080269725 Deem et al. Oct 2008 A1
20080312157 Levy et al. Dec 2008 A1
20090022727 Houston et al. Jan 2009 A1
20090036364 Levy et al. Feb 2009 A1
20090042781 Petersen et al. Feb 2009 A1
20090074734 Rottiers Mar 2009 A1
20090087408 Berry et al. Apr 2009 A1
20090156474 Roth et al. Jun 2009 A1
20090163447 Maggio Jun 2009 A1
20090186817 Ghosh et al. Jul 2009 A1
20090202481 Li et al. Aug 2009 A1
20090202608 Alessi et al. Aug 2009 A1
20090209460 Young et al. Aug 2009 A1
20090210019 Kim et al. Aug 2009 A1
20090215694 Kolterman et al. Aug 2009 A1
20090247463 Wright et al. Oct 2009 A1
20090286723 Levy et al. Nov 2009 A1
20090312246 Baron et al. Dec 2009 A1
20100092566 Alessi et al. Apr 2010 A1
20100105627 Salama et al. Apr 2010 A1
20100144621 Kim et al. Jun 2010 A1
20100185184 Alessi et al. Jul 2010 A1
20100297209 Rohloff et al. Nov 2010 A1
20110076317 Alessi et al. Mar 2011 A1
20110104111 Rohloff et al. May 2011 A1
20110152181 Alsina-Fernandez et al. Jun 2011 A1
20110152182 Alsina-Fernandez et al. Jun 2011 A1
20110160708 Berry et al. Jun 2011 A1
20110166554 Alessi et al. Jul 2011 A1
20110264077 Rohloff et al. Oct 2011 A1
20110306549 Tatarkiewicz et al. Dec 2011 A1
20120208755 Leung Aug 2012 A1
20130034210 Rohloff et al. Feb 2013 A1
20150111818 Alessi et al. Apr 2015 A1
20150231062 Lautenbach et al. Aug 2015 A1
20150231256 Berry et al. Aug 2015 A1
Foreign Referenced Citations (135)
Number Date Country
0079405 May 1983 EP
0254394 Jan 1988 EP
0295411 Dec 1988 EP
0368339 May 1990 EP
0373867 Jun 1990 EP
0431942 Jun 1991 EP
0379147 Sep 1994 EP
0627231 Dec 1994 EP
0 729 747 May 1997 EP
0 771 817 May 1997 EP
0841359 May 1998 EP
0767689 Jun 1999 EP
1046399 Oct 2000 EP
1084703 Mar 2001 EP
1600187 Jan 2009 EP
2020990 Sep 2010 EP
640907 Jul 1928 FR
1049104 Nov 1966 GB
1518683 Jul 1978 GB
9-241153 Sep 1997 JP
11-100353 Apr 1999 JP
2006213727 Aug 2006 JP
592113 Aug 2012 NZ
200634060 Oct 2006 TW
WO 9107160 May 1991 WO
WO 9306819 Apr 1993 WO
WO 9306821 Apr 1993 WO
WO 9308832 May 1993 WO
WO 9309763 May 1993 WO
WO 9323083 Nov 1993 WO
WO 9409743 May 1994 WO
WO 9421262 Sep 1994 WO
WO 9501167 Jan 1995 WO
WO 9509006 Apr 1995 WO
WO 9509007 Apr 1995 WO
WO 9534285 Dec 1995 WO
WO 9601134 Jan 1996 WO
WO 9603116 Feb 1996 WO
WO 9639142 Dec 1996 WO
WO 9640049 Dec 1996 WO
WO 9640139 Dec 1996 WO
WO 9640355 Dec 1996 WO
WO 9715289 May 1997 WO
WO 9715296 May 1997 WO
WO 9728181 Aug 1997 WO
WO 9746204 Dec 1997 WO
WO 9747339 Dec 1997 WO
WO 9800152 Jan 1998 WO
WO 9800157 Jan 1998 WO
WO 9800158 Jan 1998 WO
WO 9802169 Jan 1998 WO
WO 9816250 Apr 1998 WO
WO 9817315 Apr 1998 WO
WO 9820930 May 1998 WO
WO 9827960 Jul 1998 WO
WO 98027962 Jul 1998 WO
WO 9830231 Jul 1998 WO
WO 9832463 Jul 1998 WO
WO-9827963 Jul 1998 WO
WO 9842317 Oct 1998 WO
WO 9847487 Oct 1998 WO
WO 9851282 Nov 1998 WO
WO 9903453 Jan 1999 WO
WO 9904767 Feb 1999 WO
WO 9904768 Feb 1999 WO
WO 9916419 Apr 1999 WO
WO 9925728 May 1999 WO
WO 9929306 Jun 1999 WO
WO 9933446 Jul 1999 WO
WO 9939700 Aug 1999 WO
WO 9940788 Aug 1999 WO
WO 9944659 Sep 1999 WO
WO 9962501 Dec 1999 WO
WO 9964061 Dec 1999 WO
WO-0013663 Mar 2000 WO
WO 0029206 May 2000 WO
WO 0038652 Jul 2000 WO
WO 0039280 Jul 2000 WO
WO 0040273 Jul 2000 WO
WO 0041548 Jul 2000 WO
WO 0045790 Aug 2000 WO
WO0045790 Aug 2000 WO
WO 0045790 Aug 2000 WO
WO 0054745 Sep 2000 WO
WO 0066138 Nov 2000 WO
WO 0067728 Nov 2000 WO
WO 0143528 Jun 2001 WO
WO 0151041 Jul 2001 WO
WO-0178683 Oct 2001 WO
WO-0228366 Apr 2002 WO
WO 0236072 May 2002 WO
WO 0243800 Jun 2002 WO
WO 0245752 Jun 2002 WO
WO 0247716 Jun 2002 WO
WO 02069983 Sep 2002 WO
WO-02067895 Sep 2002 WO
WO 02076344 Oct 2002 WO
WO-02076344 Oct 2002 WO
WO 03000230 Jan 2003 WO
WO 03007981 Jan 2003 WO
WO 03011892 Feb 2003 WO
WO 03024357 Mar 2003 WO
WO 03024503 Mar 2003 WO
WO 03030923 Apr 2003 WO
WO 03041684 May 2003 WO
WO 03041757 May 2003 WO
WO 03053400 Jul 2003 WO
WO 03072113 Sep 2003 WO
WO 03072133 Sep 2003 WO
WO 2004002565 Jan 2004 WO
WO 2004052336 Jun 2004 WO
WO 2004056338 Jul 2004 WO
WO 2004089335 Oct 2004 WO
WO 2005048930 Jun 2005 WO
WO 2005048952 Jun 2005 WO
WO 2005102293 Nov 2005 WO
WO 2006017772 Feb 2006 WO
WO 2006023526 Mar 2006 WO
WO 2006081279 Aug 2006 WO
WO 2006083761 Aug 2006 WO
WO 2006084139 Aug 2006 WO
WO 2006086727 Aug 2006 WO
WO 2006101815 Sep 2006 WO
WO 2006111169 Oct 2006 WO
WO 2007024700 Mar 2007 WO
WO 2007056681 May 2007 WO
WO 2007075534 Jul 2007 WO
WO 2007084460 Jul 2007 WO
WO 2007133778 Nov 2007 WO
WO 2007140416 Dec 2007 WO
WO 2008021133 Feb 2008 WO
WO 2008061355 May 2008 WO
WO 2008133908 Nov 2008 WO
WO 2008134425 Nov 2008 WO
WO 2009109927 Sep 2009 WO
Non-Patent Literature Citations (218)
Entry
Bakan, D. et al., “Physicochemical Characterization of a Synthetic Lipid Emulsion for Hepatocyte-Selective Delivery of Lipophilic Compounds: Application to Polyiodinated triglycerides as Contrast Agents for Computed Tomography”, Journal of Pharmaceutical Science, 1996, 85(9), 908-914.
Cantor, R.S., “Theory of Lipid Monolayers Comprised of Mixtures of Flexible and Stiff Amphiphiles in Anthermal Solvents: Fluid Phase Coexistence”, The Journal of Chemical Physics, 1996, 104(20), 8082-8095.
Chapman, D. et al., “Physical Studies of Phospholipids. VI. Thermotropic and Lyotropic Mesornorphism of Some 1,2-Diacylphosphatidylcholines (lecithins)”, Chemistry and Physics of Lipids, 1967, 1(5), 445-475.
Felker, T.E. et al., “The Rate of Transfer of Unesterified Cholesterol from Rat Erythrocytes to Emulsions Modeling Nascent Triglyceride-Rich Lipoproteins and Chylomicrons Depends on the Degree of Fluidity of the Surface”, The Journal of Nutritional Biochemistry, 1993, 4(1), 630-634.
Kabalnov, A. et al, “Phospholipids as Emulsion Stabilizers.2. Phase Behavior Versus Emulsion Stability”, Journal of Colloid and Interface Science, 1996, 184(1), 227-235.
Kabalnov, A. et al., “Macroemulsion Type and Stability of Alkane-Water-Phospholipid Systems”, Book of Abstracts, 210th ACS National Meeting, Chicago, Il, Aug. 20-24, 1995, Issue Pt 1.
Massey, John B., “Interaction of Vitamin E with Saturated Phospholipid Bilayers”, Biochemical and Biophysical Research Communications, 1982, 106(3), 842-847.
Wang, X. et al., “Preferential Interaction of α-tocopherol with Phosphatidylcholines in Mixed Aqueous Dispersions of Phosphatidylcholine and Phosphatidylethanolamine”, European Journal of Biochemistry, 2000, 267(21), 6362-6368.
Wang, X. et al., “Ripple Phases Induced by α-Tocopherol in Saturated Diacylphosphatidylcholines”, Archives of Biochemistry and Biophysics, 2000, 377(2), 304-314.
Wang, X.Y. et al., “The Distribution of α-Tocopherol in Mixed Aqueous Dispersions of Phosphatidylcholine and Phosphattidylethanolamine”, Biochimica et Biophysica Acta-Biomembranes, 2000, 1509(1-2), 361-372.
Jain, R.A. et al., “Controlled Delivery of Drugs from a Novel Injectable in Situ Formed Biodegradable PLGA Microsphere System”, Journal of Microencapsulation, 2000, 17(3), 343-362.
Fujii, M. et al., “Effect of phosphatidylcholine on Skin Permeation of Indomethacin from gel prepared with Liquid Paraffin and Hydrogenated Phospholipid”, International Journal of Pharmaceutics, 2001, 222(1), 57-64.
Fujii, M. et al., Enhancement of Skin Permeation of Miconazole by Phospholipid and Dodecyl 2-(N, N-dimethylamino) Propionate (DDAIP), International Journal of Pharmaceutics, 2002, 234(1-2), 121-128.
Schmalfuβ, U. et al., “Modification of Drug Penetration into Human Skin Using Microemulsions”, Journal of Controlled Release, 1997, 46(3), 279-285.
Erõs, I. et al., “Multiple Phase Emulsions as Controlled Drug Delivery Therapeutic Systems”, Proc.-Conf. Colloid Chem., 1993, 6th Meeting Date, 193-196.
Nordén, T.P. et al., “Physicochemical Characterization of a Drug-Containing Phospholipid-Stabilized o/w Emulsion for Intravenous Administration”, European Journal of Pharmaceutical Sciences, 2001, 13(4), 393-401.
Bakhtiar, C. et al, Taking Delivery. (Ingredients: Liposomes). (liposomes in cosmetic delivery systems), Soap Perfumery & Cosmetics, 2003, 76(3), 59(4).
Das, S. et al., “Reviewing Antisense Oligonucleotide Therapy: Part 2, Delivery Issues”, BioPharm, Nov. 1999, 12(11), 44.
Larsson, K., “Stability of Emulsions Formed by Polar Lipids”, Progress in the Chemistry of Fats and Other Lipids, 1978, 16, 163-169.
Ortiz, A. et al., A Differential Scanning Calorimetry Study of the Interaction of α-Tocopherol with Mixtures of Phospholipids, Biochim et Biophys Acta, 1987, 898(2), 214-222.
Lundberg, B.., “A Submicron Lipid Emulsion Coated with Amphipathic Polyethylene Glycol for Parenteral Administration of Paclitaxel (Taxol)”, The Journal of Pharmacy and Pharmacology, 1997, 49(1), 16-21.
Hodoshima, N. et al., “Lipid Nanoparticles for Delivering Antitumor Drugs”, International Journal of Pharmaceutics, 1997, 146(1), 81-92.
Quintanar Guerrero D. et al., “Applications of the Ion-Pair Concept to Hydrophilic Substances with Special Emphasis on Peptides”, Pharmaceutical Research, 1997, 14(2), 119-127.
Bauer, K.H. et al., “Non-Aqueous Emulsions as Vehicles for Capsule Fillings”, Drug Development and Industrial Pharmacy, 1984, 10(5), 699-712.
Bohlinder, K. et al., “Use and Characteristics of a Novel Lipid Particle-Forming Matrix as a Drug-Carrier System”, European Journal of Pharmaceutical Sciences, 1994, 2(4), 271-279.
Supplementary European Search Report, Application No. EP 04 81 1199, Jun. 2, 2009 (EP Application No. 04 81 1199 is a foreign application related to the present U.S. application).
Chaumeil, J.C., “Micronization: A Method of Improving the Bioavailability of Poorly Soluble Drugs,” Methods and Findings in Experimental and Clinical Pharmacology, Apr. 1998, 20(3), 211-215.
Eppstein et al. “Biological Activity of Liposome-Encapsulated Murine Interferon γ is Mediated by a Cell Membrane Receptor.” PNAS. 82(1985):3688-3692.
Knepp et al. “Stability of Nonaqueous Suspension Formulations of Plasma Derived Factor IX and Recombinant Human Alpha Interferon at Elevated Temperatures.” Pharma. Res. 15.7(1998):1090-1095.
Office Action issued for Argentine Published Patent Application No. AR 046844 A1 mailed Apr. 9, 2015 (English translation) 4 pages.
Adolf, “Human interferon omega—a review,” Mult. Sclr. 1:S44-47 (1995).
Henry et al., “Comparing ITCA 650, continuous subcutaneous delivery of exenatide via DUROS® device, vs. twice daily exenatide injections in metformin-treated type 2 diabetes,” oral presentation at the 46th Annual Meeting of the European Association for the Study of Diabetes in Stockholm, Sweden , 21 pages (Sep. 20-24, 2010).
Ishiwata et al., “Clinical effects of the recombinant feline interferon-omega on experimental parvovirus infection in beagle dogs,” J. Vet. Med. Sci. 60(8):911-917 (1998).
Johnson et al., “How interferons fight disease,” Sci. Am. 270(5):68-75 (May 1994).
Nielsen, “Incretin mimetics and DPP-IV inhibitors for the treatment of type 2 diabetes,” Drug Discovery Today 10(10):703-710 (May 15, 2005).
Palmeri et al., “5-Fluorouracil and recombinant α-interferon-2a in the treatment of advanced colorectal carcinoma: a dose optimization study,” J. Chemotherapy 2(5):327-330 (Oct. 1990).
PCT International Search Report for PCT/US2009/000916, 4 pages (Aug. 12, 2009).
“Intarcia Therapeutics Announces Final Results from a Phase 2 Study of Injectable Omega Interferon plus Ribavirin for the Treatment of Hepatitis C Genotype-1 ,” NLV Partners Press Coverage Portfolio News (Apr. 12, 2007) (Press Release).
Roberts et al., “The Evolution of the Type I Interferons1,” J. Interferon Cytokine Res. 18(10):805-816 (Oct. 1998).
Rohloff et al., “DUROS Technology Delivers Peptides and Proteins at Consistent Rate Continuously for 3 to 12 Months,” J. Diabetes Sci. & Tech., 2(3):461-467 (May 1, 2008).
Smith, “Peripheral Neuro-hormones as a Strategy to Treat Obesity,” oral presentation at the 2007 Cardiometabolic Health Congress in Boston, MA, pp. 1-35 (Sep. 26-29, 2007).
Written Opinion for International Patent Application No. PCT/US2009/005629 (corresponding to U.S. Appl. No. 12/587,946), 5 pages (Apr. 15, 2011).
Zhang et al., “Efficacy observations of different dosages of interferon to treat 150 Hepatitis B carriers,” Current Physician 2(12):45-46 (1997).
Costantino et al., “Protein Spray Freeze Drying. 2. Effect of Formulation Variables on particle Size and Stability,” J. Pharm. Sci. 91:388-395 (2002).
Huggins et al., “Synergistic antiviral effects of ribavirin and the C-nucleoside analogs tiazofurin and selenazofurin against togaviruses, bunyaviruses, and arenaviruses,” Antimicrobial Agents & Chemotherapy, 26(4):476-480 (1984).
Lublin et al., “Defining the clinical course of multiple sclerosis: results of an internatinal survey,” Neurology. 46:907-911 (1996).
Madsbad, “Exenatide and liraglutide: different approaches to develop GLP-1 receptor agonists (incretin mimetics)—preclinical and clinical results,” Best Practice & Research Clinical Endocrinology & Metabolism 23:463-77 (2009).
Patti et al., “Natural interferon-b treatment of relapsing-remitting and secondary-progressive multiple sclerosis patients: two-year study,” Acta. Neurol. Scand. 100:283-289 (1990).
Paty et al., “Interferon beta-1 b is effective in relapsing-remitting multple sclerosis,” Neurology 43:662-667 (1993).
Quianzon et al., “Lixisenatide—Once-daily Glucagon-like Peptide-1 Diabetes,” US Endocrinology 7(2):104-109 (2011).
Ratner et al., “Dose-dependent effects of the one-daily GLP-1 receptor agonist lixisenatide in patients with Type 2 diabetes inadequately controlled with metfmmin: a randomized, double-blind, placebo-controlled trial,” Diabetic Medicine 27(9):1024-1032 (Sep. 2010).
“Sequence Listings for International Patent Application Publication No. W02009109927, WIPO Patentscope”, http://patentscope.wipo.int/search/docservcepdf—pct!d00000008776887, 1 page (last visited Nov. 14, 2012).
Shire et al., “Challenges in the Development of High Protein Concentration Formulations,” J. Pharm. Sci. 93:1390-1402 (2004).
“Abstracts 2007,” Diabetologia Clinical & Experimental Diabetes & Metabolism, Springer, Berlin, Germany, vol. 50 S243 (Aug. 21, 2007) (paragraph [0586]) (XP002538652).
Jetschmann et al., “Open-label rising-dose study of omega interferon in IFN-naive patients with chronic hepatitis C,” Gastroenterology 122:A278-A347 (Apr. 1, 2002) (Abstract M1454).
Bray, “Gut Signals and Energy Balance: Ghrelin, Peptide YY, Leptin, and Amylin,” (Dec. 19, 2007) (slides and transcript for presentation at Medscape CME).
“Implantable infusion pumps: technology poised for takeoff,” BBI Newsletter 17(12):209-211 (Dec. 1994).
Adamson et al., “Phase I trial and pharmacokinetic study of all-trans-retinoic acid administered on an intermittent schedule in combination with interferon-alpha2a in pediatric patients with refractory cancer,” J. Clin. Oncol. 15(11):3330-3337 (Nov. 1997).
Adolf et al., “Monoclonal antibodies and enzyme immunoassays specific for human interferon (IFN) ω1: evidence that IFN-ω1 is a component of human leukocyte IFN,” Virology 175(2):410-471 (Apr. 1990).
Adolf et al., “Antigenic structure of human interferon ω1 (Interferon αII1): comparison with other human interferons,” J. Gen. Virol. 68(6):1669-1676 (Jun. 1987).
Adolf et al., “Purification and characterization of natural human interferon ω1,” J. Bio. Chem. 265(16):9290-9295 (Jun. 1990).
Adolf et al., “Human interferon ω1: isolation of the gene, expression in Chinese hamster ovary cells and characterization of the recombinant protein,” Biochim. Biophys. Acta 108(9):167-174 (Jun. 1991).
Andrx Pharmaceuticals, LLC, ANDA for Concerta® Extended-Release Tablets, 6 pages (correspondence dated Sep. 6, 2005).
ASTM International, Annual Book of ASTM Standards, 8.02:208-211, 584-587 (1984).
Ansel et al., “Dosage Form Design: Pharmaceutical and Formulation Considerations,” Pharmaceutical Dosage Forms and Drug Delivery Systems, Ch. 3 at 87-92 (7th ed. Lippincott Williams & Wilkins 1999).
Ansel et al., “Modified-Release Dosage Forms and Drug Delivery Systems,” Pharmaceutical Dosage Forms and Drug Delivery Systems, Ch. 8 at 229-243 (7th ed. Lippincott Williams & Wilkins 1999).
Aulitzky, “Successful Treatment of Metastatic Renal Cell Carcinoma With a Biologically Active Dose of Recombinant Interferon-Gama,” Journal of Clinical Oncology 7(12):1875-1884 (1989).
Hauck, “Engineer's Guide to Plastics,” Materials Engineering 5(72):38-45 (Jul. 17, 1972).
Bailon et al., “Rational Design of a Potent, Long-lasting Form of Interferon: A 40 kDa Branched Polyethylene Glycol-conjugated Interferon Alpha-2a for the Treatment of Hepatitis C,” Bioconjugate Chemistry 12(2):195-202 (2001).
Balkwill,F., “Interferons,” Lancet 1(8646):1060-1063 (May 1989).
Bekkering et al., “Estimation of early hepatitis C viral clearance in patients receiving daily interferon and ribavirin therapy using a mathematical model,” Hepatology 33(2):419-423 (Feb. 2001).
Bell et al., “Hamster preproglucagon contains the sequence of glucagon and two related peptides,” Nature 302:716-718 (1983).
Bell et al, “Impact of moisture on thermally induced denaturation and decomposition of lyophilized bovine somatotropin,” Drug Delivery Research & Dev. Biopolymers, (35):201-209 (1995).
Bertoncello et al., “Haematopoietic radioprotection by Cremophor EL: a polyethoxylated castor oil,” Int. J. Radiat. Biol. 67(1):57-64 (1995).
Bolinger et al., “Recombinant interferon γ for treatment of chronic granulomatous disease and other disorders,” Clin. Pharm. 11(10):834-850 (Oct. 1992).
Bonkovsky et al., “Outcomes research in chronic viral hepatitis C: effects of interferon therapy,” Can. J. Gastroenterol. 14(Supp. B):21B-29B (Jul.-Aug. 2000).
Borden et al., “Second-generation interferons for cancer: clinical targets,” Semin. Cancer Biol. 10(2):125-144 (Apr. 2000).
Boué et al., “Antiviral and antiluteolytic activity of recombinant bovine IFN-ω1 obtained from Pichia pastoris,” J. Interferon & Cytokine Res. 20:677-683 (2000).
Buckwold et. al. “Antiviral activity of CHO-SS cell-derived human omega interferon and other human interferons against HCV RNA replicons and related viruses,” Antiviral Res. 73(2):118-25 (Feb. 2007) (Epub Sep. 11, 2006).
CAS No. 56-81-5 (Nov. 16, 1984).
Chang et al., “Biodegradeable polyester implants and suspension injection for sustained release of a cognitive enhancer,” Pharm. Tech. 20(1):80-84 (1996).
Clark et al., “The diabetic Zucker fatty rat,” Proc. Soc. Exp. Biol. 173(1):68-75 (1983).
Condino-Neto, “Interferon-γ improves splicing efficiency of CYBB gene transcripts in an interferon responsive variant of chronic granulomatous disease due to a splice site consensus region mutation,” Blood 95(11):3548-3554 (Jun. 2000).
Darney, “Subdermal progestin implant contraception,” Current Opinion in Obstetrics & Gynecology 3:470-476 (1991).
Dash, A. K. et al., “Therapeutic applications of implantable drug delivery systems,” Journal of Pharmacological and Toxicological Methods, 40(1):1-12 (1998).
Davis et al., “Durability of viral response to interferon alone or in combination with oral ribavirin in patients with chronic hepatitis C,” Prog. Abstr. 50th Annu. Mtg. Postgrad. Courses Am. Assn. Study Liver Dis., Dallas, TX (Nov. 5-9, 1999)(Abstract 570 ).
Deacon, C. F. et al., “GLP-1-(9-36) amide reduces blood glucose in anesthetized pigs by a mechanism that does not involve insulin secretion,” Am. J. Physiol. Endocrinol. Metab., 282:E873-E879 (2002).
Desai et al., “Protein structure in the lyophilized state: a hydrogen isotope exchange/NMR study with bovine pancreatic trypsin inhibitor,” J. Am. Chem. Soc. 116(21):9420-9422 (1994).
Di Marco et al., “Combined treatment of relapse of chronic hepatitis C with high-dose α-2B interferon plus ribavirin for 6 or 12 months,” Prog. Abstr. 50th Annu. Mtg. Postgrad. Courses Am. Assn. Study Liver Dis., Dallas, TX (Nov. 5-9, 1999)(Abstract 569).
Dorr et al., “Phase I-II trial of interferon-alpha 2b by continuous subcutaneous infusion over 28 days,” J. Interferon Res. 8:717-725 (1988).
Uhlig et al., “The electro-smotic acutation of implantable insulin micropumps,” J. Biomed. Materials Res. 17:931-943 (1983).
Efendic, S. et al., et al., “Overview of incretin hormones,” Horm. Metab. Res., 36(11-12):742-746 (2004).
Eissele, R. et al., “Rat gastric somatostatin and gastrin release: interactions of exendin-4 and truncated glucagon-like peptide-1 (GLP-1) amide,” Life Sci., 55(8):629-634 (1994).
Elias et al., “Infusional Interleukin-2 and 5-fluorouracil with subcutaneous interferon-α for the treatment of patients with advanced renal cell carcinoma: a southwest oncology group Phase II study,” Cancer 89(3):597-603 (Aug. 2000).
Eng, J. et al., “Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom. Further evidence for an exendin receptor on dispersed acini from guinea pig pancreas,” J. Biol. Chem., 267(11):7402-7405 (1992).
Eng, J. et al., “Purification and structure of exendin-3, a new pancreatic secretagogue isolated from Heloderma horridum venom,” J. Biol. Chem., 265(33):20259-20262 (1990).
Fang et al., “The impact of baseline liver histology on virologic response to interferon α-2b±ρ ribavirin therapy in patients with chronic hepatitis C,” Prog. Abstr. 50th Annu. Mtg. Postgrad. Courses Am. Assn. Study Liver Dis., Dallas, TX (Nov. 5-9, 1999)(Abstract 572).
Ferenci et al, “Combination of interferon (IFN) induction therapy and ribavirin in chronic hepatitis C,” Prog. Abstr. Dig. Dis. Week 2000, San Diego, CA (May 21-24, 2000) (Abstract 977).
Fontaine et al., “Recovery from chronic hepatitis C in long-term responders to ribarivin plus interferon α,” Lancet 356(9223):41 (Jul. 2000).
Franchetti et al., “Furanfurin and Thiophenfurin: Two Novel TiazofurinAnalogues. Synthesis, Structure, Antitumor Activity, and Interactions with Inosine Monophosphate Dehydrogenase,” J. Medicinal Chem. 38(19):3829-3837 (1995).
Luft et al., “Electro-osmotic valve for the controlled administration of drugs,” Med. & Biological Engineering & Computing 45-50 (Jan. 1978) (non-English with English abstract).
Gan To Kagaku Ryoho, “Phase II study of recombinant leukocyte A interferon (Ro22-8181) in malignant brain tumors,” Cancer & Chemotherapy 12(4):913-920 (Apr. 1985) (non-English with English abtract).
Gappa et al., “Juvenile laryngeal papillomatosis—a case report,” Pneumologie 45(11):936-938 (Nov. 1991) (XP009079028) (non-English with English abstract).
Gause et al., “Phase I study of subcutaneously administered interleukin-2 in combination with interferon alfa-2a in patients with advanced cancer,” J. Clin. Oncol. 14(8):2234-2241 (Aug. 1996).
Ghiglione, M., et al., “How glucagon-like is glucagon-like peptide-1?” Diabetologia 27:599-600 (1984).
Glue et al., “A dose-ranging study of Peg-intron and ribavirin in chronic hepatitis C—safety, efficacy, and virological rationale,” Prog. Abstr. 50th Annu. Mtg. Postgrad. Courses Am. Assn. Study Liver Dis., Dallas, TX(Nov. 5-9, 1999)(Abstract 571).
Goke, R. et al., “Exendin-4 is a high potency agonist and truncated exendin-(9-39)-amide an antagonist at the glucagon-like peptide 1-(7-36)-amide receptor of insulin-secreting beta-cells,” J. Biol. Chem., 268(26):19650-19655 (1993).
Gonzales et al., “Randomized controlled trial including an initial 4-week ‘induction’ period during one year of high-dose interferon α-2B treatment for chronic hepatitis C,” Prog. Abstr. Dig. Dis. Week 2000, San Diego, CA (May 21-24, 2000) (Abstract 975).
Gosland et al., “A phase I trial of 5-day continuous infusion cisplatin and interferon alpha,” Cancer Chemother. Pharmacol. 37(1-2):39-46 (1995).
Grant et al., “Combination therapy with interferon-α plus N-acetyl cystein for chronic hepatitis C: a placebo controlled double-blind multicentre study,” J. Med. Virol. 61(4):439-442 (Aug. 2000).
Gutniak, M. et al., “Antidiabetogenic effect of glucagon-like peptide-1 (7-36)amide in normal subjects and patients with diabetes mellitus,” N. Engl. J. Med., 326(20):1316-1322 (1992).
Hageman, “The Role of Moisture in Protein Stability,” Drug Dev. & Ind. Pharm. 14(14):2047-2070 (1988).
Heathcote et al., “Peginterferon alfa-2a in Patients With Chronic Hepatitis C and Cirrhosis,” New England J. Med. 343(23):1673-1680 (2000).
Heim et al., “Intracellular signaling and antiviral effects of interferons,” Dig. Liver Dis. 32(3):257-263 (Apr. 2000).
Heinrich, G. et al., “Pre-proglucagon messenger ribonucleic acid: nucleotide and encoded amino acid sequences of the rat pancreatic complementary deoxyribonucleic acid,” Endocrinol., 115:2176-2181 (1984).
Hellstrand et al., “Histamine and cytokine therapy,” Acta Oncol. 37(4):347-353 (1998).
Hellstrand et al., “Histamine and the response to IFN-α in chronic hepatitis C,” Interferon Cytokine Res. 18(1):21-22 (Jan. 1998).
Hellstrand et al., “Histamine in immunotherapy of advanced melanoma: a pilot study,” Cancer Immunol Immunother. 39(6):416-419 (Dec. 1994).
Hisatomi et al., “Toxicity of polyoxyethylene hydrogenated castor oil 60 (HCO-60) in experimental animals,” J. Toxicol. Sci., 18(3):1-9 (1993).
Hoffmann-La Roche Inc., Pegasys® (peginterferon alfa-2a), 15 pages (2002).
Horton et al., “Antitumor effects of interferon-omega: in vivo therapy of human tumor xenografts in nude mice” Cancer Res 59(16):4064-4068 (Aug. 1999).
Hubel et al., “A phase I/II study of idarubicin, dexamethasone and interferon-alpha (1-Dexa) in patients with relapsed or refractory multiple myeloma” Leukemia 11 Suppl 5:S47-S51 (Dec 1997).
Iacobelli et al., “A phase I study of recombinant interferon-alpha administered as a seven-day continuous venous infusion at circadian-rhythm modulated rate in patients with cancer,” Am. J. Clin. Oncol. 18(1):27-31 (1995).
IFNB Multiple Sclerosis Study Group, “Interferonβ-1b is effective in relapsing-remitting multiple sclerosis,” Neurology 43(4):655-667 (Apr. 1993).
Intermune® Inc., Infergen® (Interferon alfacon-1), 5 pages (2002).
“Introduction to Antibodies”, http://www.chemicon.com/resource/ANT101/a1.asp, 8 pages (retrieved May 2, 2007).
Isaacs et al., “Virus interference. I. The interferon,” Pro. R. Soc. Lond. B. Biol. Sci. 147:258-267 (1957).
Khalili et al., “Interferon and ribavirin versus interferon and amantadine in interferon nonresponders with chronic hepatitis C,” Am. J. Gastroenterol. 95(5):1284-1289 (May 2000).
Kildsig et al., “Theoretical Justification of Reciprocal Rate Plots in Studies of Water Vapor Transmission through Films,” J. Pharma. Sci. 29(11):1634-01637 (Nov. 17, 1970).
Kirkwood et al., “Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: The Eastern Cooperative Oncology Group Trial EST 1684,” J. Clin. Oncol. 14(1):7-17 (1996).
Kita et al., “Characterization of a polyethylene glycol conjugate of recombinant human interferon-γ,” Drug Des. Deliv. 6(3):157-0167 (Sep. 1990).
Knepp et al, “Identification of antioxidants for prevention of peroxide-mediated oxidation of recombinant human ciliary neurotrophic factor and recombinant human nerve growth factor,” J. Pharm. Sci. Tech. 50(3):163-171 (1996).
Knobler et al., “Systemic α-interferon therapy of multiple sclerosis,” Neurology 34(10):1273-1279 (Oct. 1984).
Kovacevic et al., “Treatment of chronic viral hepatitis B in secondary membranoproliferative glomerulonephritis using recombinant α-2 interferon,” Maksic Dj Vojnosanit. Pregl. 57(2):235-240 (Mar.-Apr. 2000) (non-English with English abstract).
Kracke et al., “Mx proteins in blood leukocytes for monitoring interferon β-1b therapy in patients with MS,” Neurology 54(1):193-199 (Jan. 2000).
Kronenberger et al., “Influence of interferon-α on CD82-expression in HCV-positive patients,” Prog. Abstr. Dig. Dis. Week 2000, San Diego, CA (May 21-24, 2000) (Abstract 976).
Krown et al., “Interferons and interferon inducers in cancer treatment,” Semin. Oncol. 13(2):207-217 (1986).
Kubes et al., “Cross-species antiviral and antiproliferative activity of human interferon-ω,” J. Interferon Res. 14:57-59 (1994).
Kunzi et al., “Role of interferon-stimulated gene ISG-15 in the interferon-ω-mediated inhibition of human immunodeficiency virus replication,” J. Interferon Cytokine Res. 16(11):919-927 (Nov. 1996).
Lee et al., “Dynamics of hepatitis C virus quasispecies turnover during interferon-A treatment,” Prog. Abstr. Dig. Dis. Week 2000, San Diego, CA (May 21-24, 2000) (Abstract 974).
Lee, “Therapy of hepatits C: interferon alfa-2A trials,” Hepatology 26: 89S-95S (Sep. 1997) (XP000981288).
Lopez, L. C. et al., “Mammalian pancreatic preproglucagon contains three glucagon-related peptides,” Proc. Natl. Acad. Sci. USA, 80(18):5485-5489 (1983).
Lukaszewski et al., “Pegylated α interferon is an effective treatment for virulent Venezuelan equine encephalitis virus and has profound effects on host immune response to infection,” J. Virol. 74(11):5006-5015 (Jun. 2000).
Lund, P. K. et al., “Pancreatic preproglucagon cDNA contains two glucagon-related coding sequences arranged in tandem,” Proc. Natl. Acad. Sci. USA, 79(2):345-349 (1982).
Magnuson et al. “Enhanced recovery of a secreted mammalian protein from suspension culture of genetically modified tobacco cells,” Protein Expression & Purification 7:220-228 (1996).
Malley et al., “Chronic Toxicity and Oncogenicity of N-Methylpyrrolidone (Nmp) in Rats and Mice by Dietary Administration,” Drug Chem Toxicol. 24(4):315-38 (Nov. 2001).
Manning et al, “Stability of protein pharmaceuticals,” Pharm. Res. 6(11):903-918 (1989).
Marincola et al., “Combination therapy with interferon alfa-2a and interleukin-2 for the treatment of metastatic cancer,” J. Clinical Oncol. 13(5):1110-1122 (1995) (XP009078965).
McHutchison et al., “Interferon α-2b alone or in combination with ribavirin as initial treatment for chronic hepatitis C,” N. Engl. J. Med. 339(21):1485-1492 (Nov. 1998).
McHutchison, et al., “Open-label phase 1B study of hepatitis C viral dynamics with omega interferon treatment,” Hepatology 34(4):A333 (Oct. 1, 2001) (XP004716177) (Abstract Only).
Meier, J. J. et al., “The glucagon-like peptide-1 metabolite GLP-1-(9-36) amide reduces postprandial glycemia independently of gastric emptying and insulin secretion in humans,” Am. J. Physiol. Endocrinol. Metab., 290(6):E1118-E1123 (2006).
Merad et al., “Generation of monocyte-derived dendritic cells from patients with renal cell cancer: modulation of their functional properties after therapy with biological response modifiers (IFN-α plus IL-2 and IL-12),” J. Immunother. 23(3):369-378 (May-Jun. 2000).
Milella et al., “Neutralizing antibodies to recombinant α-interferon and response to therapy in chronic hepatitis C infection,” Liver 13(3):146-150 (Jun. 1993).
Mohler, “Primer on electrodeposited coatings,” Materials Engineering 5:38-45 (1972).
Mojsov, S., “Structural requirements for biological activity of glucagon-like peptide-I,” Int. J. Peptide Protein Research, 40:333-343 (1992).
Morgan, “Structure and Moisture Permeability of Film-Forming Poloyers,” Ind. Eng. Chem. 45(10):2296-2306 (1953).
Motzer et al., “Phase I trial of 40-kd branched pegylated interferon alfa-2a for patients with advanced renal cell carcinoma,” J. Clinical Oncol. 19(5):1312-1319 (2001).
Nauck, M. A. et al., “Normalization of fasting glycaemia by intravenous GLP-1 ([7-36 amide] or [7-37]) in type 2 diabetic patients,” Diabet. Med., 15(11):937-945(1998).
Neumann et al., “Hepatitis C Viral Dynamics In Vivo and the Antiviral Efficacy of Interferon-alpha Therapy,” Science 282:103-107 (Dec. 1998).
Nieforth et al., “Use of an indirect pharmacodynamic stimulation model of MX protein induction to compare in vivo activity of interferon-α-2a and a polyethylene glycol-modified derivative in healthy subjects,” Clin. Pharmacol. Ther. 59(6):636-646 (Jun. 1996).
Olaso et al., “Early prediction of lack of response to treatment with interferon and interferon plus ribavirin using biochemical and virological criteria in patients with chronic hepatitis C,” Esp. Quimioter. 12(3):220-228 (Sep. 1999) (non-English with English abstract).
Panitch, “Interferons in multiple sclerosis,” Drugs 44(6):946-962 (Dec. 1992).
Patzelt, C. et al., “Identification and processing of proglucagon in pancreatic islets,” Nature, 282:260-266 (1979).
Peterson, R. G. et al., “Zucker Diabetic Fatty Rat as a Model for Non-insulin-dependent Diabetes Mellitus,” ILAR Journal, 32(3):16-19 (1990).
Peterson, R. G. et al., “Neuropathic complications in the Zucker diabetic fatty rat (ZDF/Drt-fa),” Frontiers in diabetes research. Lessons from Animal Diabetes III, Shafrir, E. (ed.), pp. 456-458, Smith-Gordon, London (1990).
Pimstone et al., “High dose (780 MIU/52 weeks) interferon monotherapy is highly effective treatment for hepatitis C,” Prog. Abstr. Dig. Dis. Week 2000, San Diego, CA (May 21-24, 2000) (Abstract 973).
Plauth et al, “Open-label phase II study of omega interferon in previously untreated HCV infected patients,” Hepatology 34(4):A331 (Oct. 1, 2001) (XP004716169) (Abstract Only).
Plauth et al, “Open-label study of omega interferon in previously untreated HCV-infected patients,” J. Hepatology 36(Supp. 1):125 (Apr. 2002) (XP002511882) (Abstract Only).
Pohl, M. et al., “Molecular cloning of the helodermin and exendin-4 cDNAs in the lizard. Relationship to vasoactive intestinal polypeptide/pituitary adenylate cyclase activating polypeptide and glucagon-like peptide 1 and evidence against the existence of mammalian homologues,” J. Biol. Chem., 273(16):9778-9784 (1998).
Poynard et al., “Is an ‘a la carte’ combined interferon α 2b plus ribavirin possible for the first line treatment in patients with chronic hepatitis C,” Hepatology 31(1):211-218 (Jan. 2000).
Poynard et al., “Randomized trial of interferon α 2b plus ribavirin for 48 weeks or for 24 weeks versus interferon α 2b plus placebo for 48 weeks for the treatment of chronic infection with hepatitis C virus,” Lancet 352(9138):1426-1432 (Oct. 1998).
“Intarcia Presents Positive ITCA 650 Phase 2 Study Results for Type 2 Diabetes at EASD,” Intarcia Therapeutics, Inc. (Sep. 22, 2010) (Press Release).
Quesada et al., “Interferons in Hematological Malignancies”, eds. Baron et al., U. Tex. 487-495 (1987).
Rajkumar et al., “Phase I evaluation of radiation combined with recombinant interferon alpha-2a and BCNU for patients with high-grade glioma,” Int'l J. Radiat. Oncol. Biol. Phys. 40(2):297-302 (Jan. 15, 1998).
Roche Pharmaceuticals, Roferon®—A (Interferon alfa-2a, recombinant), 22 pages (2003).
Roff et al., “Handbook of Common Polymers”, Cleveland Rubber Co. 72 pages (1971).
Rogers et al., “Permeability Valves,” Ind. & Eng. Chem. 49(11):1933-1936 (Nov. 17, 1957).
Roman et al., “Cholestasis in the rat by means of intravenous administration of cyclosporine vehicle, Cremophor EL,” Transplantation 48(4);554-558 (1989).
Roth et al., “High Dose Etretinate and Interferon-alpha—A Phase I Study in Squamous Cell Carcinomas and Transitional Cell Carcinomas,” Acta Oncol. 38(5):613-617 (1999).
Roth et al., “Combination therapy with amylin and peptide YY[3-36] in obese rodents: anorexigenic synergy and weight loss additivity,” Endocrinol. 148(12):6054-61 (Dec. 2007).
Schepp, W. et al., “Exendin-4 and exendin-(9-39)NH2: agonist and antagonist, respectively, at the rat parietal cell receptor for glucagon-like peptide-1-(7-36)NH2,” Eur. J. Pharmacol., 269(2):183-191 (1994).
Schering Corp., Intron® A for Injection, 6 pages (2001).
Schering Corp., PGE-Intron™ (Peginterferon alfa-2b) Powder for Injection, 29 pages (2003).
Sen et al., “The interferon system: a bird's eye view of its biochemistry,” J. Biol. Chem. 267(8):5017-5020 (Mar. 1992).
Shiffman et al., “A decline in HCV-RNA level during interferon or ihterferon/ribavirin therapy in patients with virologic nonresponse is associated with an improvement in hepatic histology,” Prog. Abstr. 50th Annu. Mtg. Postgrad. Courses Am. Assn. Study Liver Dis., Dallas, TX (Nov. 5-9, 1999) (Abstract 567).
Shima et al., “Serum total bile acid level as a sensitive indicator of hepatic histological improvement in chronic hepatitis C patients responding to interferon treatment,” J. Gastroenterol. Hepatol. 15(3):294-299 (Mar. 2000).
Shiratori et al., “Histologic improvement of fibrosis in patients with hepatitis C who have sustained response to interferon therapy,” Ann. Int. Med. 132(7):517-524 (Apr. 2000).
Simon et al., “A longitudinal study of T1 hypointense lesions in relapsing MS: MSCRG trial of interferon β1a,” Neurology 55(2):185-192 (Jul. 2000).
Sparks, J. D. et al., “Lipoprotein alterations in 10- and 20-week-old Zucker diabetic fatty rats: hyperinsulinemic versus insulinopenic hyperglycemia,” Metabolism, 47(11):1315-1324 (1998).
Sulkowskli et al., “Pegylated Interferon Alfa-2A (Pegasys™) and Ribavirin Combination Therapy for Chronic Hepatitis C: A Phase II Open-Label Study,” Gastroenterology 118(4, Supp. 2) (2000) (Abstract 236).
Sulkowski, M., et al., “Peginterferon-α-2a (40kD) and ribavirin in patients with chronic hepatitis C: a phase II open label study,” Biodrugs 16(2):105-109 (2002).
Talpaz et al., “Phase I study of polyethylene glycol formulation of interferon alpha-2B (Schering 54031) in Philadelphia chromosome-positive chronic myelogenous leukemia,” Blood 98(6):1708-1713 (2001).
Talsania, T., et al., “Peripheral exendin-4 and peptide YY(3-36) synergistically reduce food intake through different mechanisms in mice,” Endocrinology 146(9):3748-56 ( Sep. 2005).
Tanaka, H., et al., “Effect of interferon therapy on the incidence of hepatocellular carcinoma and mortality of patients with chronic hepatitis C: a retrospective cohort study of 738 patients,” Int. J. Cancer 87(5):741-749 (Sep. 2000).
Tong et al., “Prediction of response during interferon α 2b therapy in chronic hepatitis C patients using viral and biochemical characteristics: a comparison,” Hepatology 26(6):1640-01645 (Dec. 1997).
Touza Rey et al., “The clinical response to interferon-γ in a patient with chronic granulomatous disease and brain abscesses due to Aspergillus fumigatus,” Ann. Med. Int. 17(2):86-87 (Feb. 2000).
Trudeau et al., “A phase I study of recombinant human interferon alpha-2b combined with 5-fluorouracil and cisplatin in patients with advanced cancer,” Cancer Chemother. Pharmacol. 35(6):496-500 (1995).
Tseng, C. C. et al., “Glucose-dependent insulinotropic peptide: structure of the precursor and tissue-specific expression in rat,” PNAS USA, 90(5):1992-1996 (1993).
Tsung et al., “Preparation and Stabilization of Heparin/Gelatin Complex Coacervate Microcapsules,” J. Pharm. Sci. 86(5):603-7 (May 1997).
Unniappan et al., “Effects of dipeptidyl peptidase IV on the satiety actions of peptide YY,” Diabetologia; Clinical and Experimental Diabetes and Metabolism 49(8):1915-1923 (Jun. 27, 2006).
Vokes et al., “A phase I trial of concomitant chemoradiotherapy with cisplatin dose intensification and granulocyte-colony stimulating factor support for advanced malignancies of the chest,” Cancer Chemother. Pharmacol. 35(4):304-312 (1995).
Vrabec, J. T., “Tympanic membrane perforations in the diabetic rat: a model of impaired wound healing,” Otolaryngol. Head Neck Surg., 118(3 Pt. 1):304-308 (1998).
Wang et al, “Parenteral formulations of proteins and peptides: stability and stabilizers,” J. Parenter. Sci. Technol. 42(2S):S4-S26 (1988).
Weinstock-Guttman et al., “What is new in the treatment of multiple sclerosis?” Drugs 59(3):401-410 (Mar. 2000).
Weissmann et al., “The interferon genes,” Prog. Nucleic Acid Res. Mol. Biol. 33:251-300 (1986).
Wright et al., “Preliminary experience with α-2b-interferon therapy of viral hepatitis in liver allograft recipients,” Transplantation 53(1):121-123 (Jan. 1992).
Young et al., “Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta),” Diabetes, 48(5):1026-1034 (1999).
Younossi et al., “The role of amantadine, rimantadine, ursodeoxycholic acid, and NSAIDs, alone or in combination with α interferons, in the treatment of chronic hepatitis C,” Semin. Liver Dis. 19(Supp. 1):95-102 (1999).
Yu et al., “Preparation, characterization, and in vivo evaluation of an oil suspension of a bovine growth hormone releasing factor analog,” J. Pharm. Sci. 85(4):396-401 (1996).
Zeidner et al., “Treatment of FeLV-induced immunodeficiency syndrome (feLV-FAIDS) with controlled release capsular implantation of 2′,3′-dideoxycytidine,” Antivir. Res. 11(3):147-0160 (Apr. 1989).
Zein, “Interferons in the management of viral hepatitis,” Cytokines Cell Mol. Ther. 4(4):229-241 (Dec. 1998).
Zeuzem et al., “Peginterferon Alfa-2a in Patients with Chronic Hepatitis C,” New Engl. J. Med. 343(23):1666-1672 (2000).
Zeuzem et al., “Hepatitis C virus dynamics in vivo: effect of ribavirin and interferon α on viral turnover,” Hepatology 28(1):245-252 (Jul. 1998).
Zhang et al., “Report on Large Dosage Interferon to Treat 30 Cases of Viral Encephalitis,” J. Clinical Pediatrics 14(2):83-84 (1996).
Zhang et al, “A new strategy for enhancing the stability of lyophilized protein: the effect of the reconstitution medium on keratinocyte growth factor,” Pharm. Res. 12(10):1447-1452 (1995).
Zheng et al. “Therapeutic Effect of Interferon Varied Dose in Treating Virus Encephalitis,” Beijing Med. J. 13(2):80-81 (1998).
Ziesche et al., “A preliminary study of long-term treatment with interferon γ-1b and low-dose prednisolone in patients with idiopathic pulmonary fibrosis,” New Engl. J. Med. 341(17):1264-1269 (Oct. 1999).
Jordan et al., “Guidelines for Antiemetic Treatment of Chemotherapy-Induced Nauseaa and Vomiting: Past, Present and Future Recommendations,” The Oncologist 12(9):1143-1150 (2007).
Related Publications (1)
Number Date Country
20130052237 A1 Feb 2013 US
Provisional Applications (1)
Number Date Country
60520605 Nov 2003 US
Continuations (3)
Number Date Country
Parent 13090947 Apr 2011 US
Child 13598405 US
Parent 12655397 Dec 2009 US
Child 13090947 US
Parent 10988716 Nov 2004 US
Child 12655397 US