The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Apr. 4, 2022, is named 135975-11104_SL.txt and is 4,031 bytes in size.
The invention relates to the manufacture, composition, and use of an extended release protein therapeutic. Specifically, the invention relates to the manufacture, composition, and use of a plurality of polymer coated protein microspheres for the extended and uniform release of protein in an aqueous-based or physiological environment over time.
The extended release of a therapeutic protein administered toward a biological target, such as e.g., the retina or a tumor, or administered parenterally is desirable for the treatment of many different conditions, including cancers, cardiovascular diseases, vascular conditions, orthopedic disorders, dental disorders, wounds, autoimmune diseases, gastrointestinal disorders, and ocular diseases. Biocompatible and biodegradable polymers for the controlled and extended delivery of drugs have been in use for decades. As the polymer degrades over time, the therapeutic drug is slowly released.
In the case of intraocular therapeutics, there is a significant unmet medical need for extended release formulations to deliver protein therapeutics effectively over time with as few intraocular injections as possible. In the case of other diseases, such as cancer, diseases of inflammation, and other diseases, there is a need for improved implantable extended release formulations containing protein therapeutics.
Applicants have discovered and herein disclose and claim methods of manufacturing and using microparticles containing a biodegradable polymer and a therapeutic protein, which is capable of releasing a therapeutically effective amount of the therapeutic protein uniformly over an extended period of time.
In one aspect, the invention provides a microparticle comprising a protein coated with a polymer. In one embodiment, the microparticle has a diameter of from about 2 microns to about 70 microns. In one embodiment, the microparticle has a diameter of about 15 microns.
In one embodiment, the protein is an antigen-binding protein. In one embodiment, the protein comprises an Fc domain. In one embodiment, the protein comprises a receptor domain. In one embodiment, the protein is an antibody. In another embodiment, the protein is a receptor-Fc-fusion protein. In another embodiment, the protein is a trap-type protein, which comprises a cognate-receptor fragment and an Fc domain. In one particular embodiment, the protein is a VEGF-Trap protein. In one embodiment, the VEGF-Trap protein comprises an amino acid sequence set forth in SEQ ID NO:1.
In one embodiment, the polymer is a biodegradable polymer. In some embodiments, the polymer is selected from the group consisting of polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-
In one embodiment, the microparticle comprises a micronized protein core of less that ten microns and a polymer cortex. In one embodiment, the micronized protein core is at least 50% coated with polymer, which means that no more than 50% of the surface of the micronized protein core is exposed. In one embodiment, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99%, or 100% of the surface of the micronized protein core is coated with polymer.
In one embodiment, the microparticle of greater than 10 microns in size comprises (a) a micronized protein core of less that 10 microns, wherein the protein is any one or more of an antibody or antibody fragment, a receptor or soluble fragment thereof, a soluble T-cell receptor fragment, a soluble MHC fragment, a receptor-Fc-fusion protein, a trap-type protein, and a VEGF-Trap protein; and (b) a polymer coat, wherein the polymer is any one or more of a biocompatible polymer, a biodegradable polymer, a bio-erodible polymer, polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-
In one embodiment, the microparticle of an average diameter of about 15 microns to about 30 microns comprises (a) a micronized protein core of about 10 to about 12 microns, wherein the protein is a VEGF-Trap protein; and (b) a polymer coat, wherein the polymer is any one or more of PCL, PLGA, ethyl cellulose and polyorthoester, and copolymers or derivatives thereof.
In one aspect, the invention provides a plurality of microparticles, which range in size from about two microns to about 70 microns, and which comprise a micronized protein core of about two microns to about 30 microns, and a polymer cortex.
In one embodiment, the protein is an antigen-binding protein. In some embodiments, the antigen-binding protein is any one or more of an antibody or antibody fragment, a receptor or soluble fragment thereof, a soluble T-cell receptor fragment, a soluble MHC fragment, a receptor-Fc-fusion protein, a trap-type protein, and a VEGF-Trap protein. In one embodiment, the protein comprises an Fc domain. In one embodiment, the protein is an antibody. In another embodiment, the protein is a receptor-Fc-fusion protein. In another embodiment, the protein is a trap-type protein, which comprises a cognate-receptor fragment and an Fc domain. In one particular embodiment, the protein is a VEGF-Trap protein. In a specific embodiment, the VEGF-Trap protein comprises the amino acid sequence set forth in SEQ ID NO:1.
In one embodiment, the polymer is a biocompatible polymer. In one embodiment, the polymer is a bioerodible polymer. In one embodiment, the polymer is a biodegradable polymer. In some embodiments, the polymer is selected from the group consisting of polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-
In one embodiment, the micronized protein core of most microparticles of the plurality of microparticles is at least 50% coated with polymer, which means that no more than 50% of the surface of the micronized protein core is exposed. In one embodiment, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99%, or 100% of the surface of the micronized protein core is coated with polymer.
In one embodiment, the plurality of microparticles, which range in size from about two microns to about 70 microns, comprise (a) a micronized protein core of from about two microns to about 30 microns, wherein the protein is any one or more of an antibody or antibody fragment, a receptor or soluble fragment thereof, a soluble T-cell receptor fragment, a soluble MHC fragment, a receptor-Fc-fusion protein, a trap-type protein, and a VEGF-Trap protein; and (b) a polymer cortex, wherein the polymer is any one or more of a biocompatible polymer, a biodegradable polymer, a bio-erodible polymer, polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-
In one embodiment, the plurality of microparticles, which range in size from about two microns to about 70 microns, with a median size of from about 15 microns to about 30 microns, comprise (a) a micronized protein core of from about two microns to about 30 microns, with a median size of about 10 microns to about 12 microns, wherein the protein is a VEGF-Trap protein; and (b) a polymer cortex, wherein the polymer is any one or more of PLA, PCL, PLGA, ethyl cellulose and polyorthoester, and copolymers or derivatives thereof.
In one aspect, the invention provides a method of manufacturing a microparticle, which comprises a protein core and a polymer cortex. In one embodiment, the manufactured microparticle has a diameter of about two microns to about 70 microns, or a median diameter of about 15 microns to about 30 microns. In one embodiment, the method of manufacturing the microparticle comprises (1) obtaining a protein particle; (2) suspending the protein particle in a solution comprising the polymer and a solvent; and (3) removing the solvent, wherein a microparticle is formed comprising the protein core coated with the polymer cortex.
In one embodiment, the protein particle of step (1) is a micronized protein particle, which is obtained by spray drying a solution comprising the protein. In some embodiments, the protein solution is spray dried via dual-nozzle sonication, single-nozzle sonication, or electrospray. In some embodiments, the resultant micronized protein particle, which forms the core of the manufactured microparticle, has a diameter of from about two microns to about 30 microns, with a median diameter of about 10 microns to about 12 microns.
In some embodiments, the protein which forms the core is an antigen-binding protein. In some embodiments, the antigen-binding protein is any one or more of an antibody (e.g., IgG) or antibody fragment, a receptor or soluble fragment thereof, a soluble T-cell receptor fragment, a soluble MHC fragment, a receptor-Fc-fusion protein, a trap-type protein, and a VEGF-Trap protein. In a specific embodiment, the protein is a VEGF-Trap comprising the amino acid sequence set forth in SEQ ID NO:1.
In one embodiment, the solvent is removed at step (3) by creating a dispersion of the protein-polymer-solvent mixture of step (2) and allowing the solvent to evaporate from the droplets created by the dispersion. In one embodiment, the dispersion is created by spray-drying, which may be performed by dual-nozzle sonication, single-nozzle sonication, or electrospray. In one embodiment, the solvent is removed from the droplets by applying heat or air, or by chemical extraction.
In one embodiment, the polymer is biodegradable, bioerodible, and/or biocompatible. In some embodiments, the polymer is any one or more of polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-
In one aspect, the invention provides a method of manufacturing a microparticle comprising the steps of (1) forming a micronized protein particle having a diameter of from about two microns to about 30 microns, with a median diameter of from about 10 microns to 12 microns, by spray-drying a solution containing a protein, wherein the protein is an antigen-binding protein. In some embodiments, the antigen-binding protein is any one or more of an antibody or antibody fragment, a receptor or soluble fragment thereof, a soluble T-cell receptor fragment, a soluble MHC fragment, a receptor-Fc-fusion protein, a trap-type protein, and a VEGF-Trap protein (e.g., one having the sequence of SEQ ID NO:1); (2) suspending the micronized protein particle in a solution comprising the polymer and a solvent, wherein the polymer is any one or more of a biodegradable polymer, a bioerodible polymer, a biocompatible polymer, polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-
In some embodiments, the spray-drying of step (1) or step (3) is performed via dual-nozzle sonication, single-nozzle sonication, or electrospray.
In one embodiment, the method of manufacturing the microparticle comprises the steps of (1) forming a micronized VEGF-Trap particle having a diameter of from about 10 microns to 12 microns by spray-drying a solution containing a VEGF Trap protein; (2) suspending the micronized VEGF Trap particle in a solution comprising polyorthoester incorporating a latent acid and a compatible solvent, or ethylcellulose and a compatible solvent; and (3) removing the solvent by (a) spray-drying the micronized VEGF Trap particle-polyorthoester-latent acid-solvent suspension or the micronized VEGF Trap particle-ethyl cellulose-solvent suspension and (b) driving off the solvent by applying heat or air, or by extracting the solvent, wherein a microparticle is formed having a diameter of about 15 microns to about 30 microns, and comprising a VEGF-Trap core and a polymer cortex of polyorthoester, and copolymers or derivatives thereof.
In one aspect, the invention provides an extended release formulation of a therapeutic protein for the release or delivery of a steady level of the therapeutic protein over time. The extended release formulation comprises a plurality of microparticles, which range in size from about two microns to about 70 microns, each of which comprises a micronized protein core of about two microns to about 30 microns, and a polymer cortex.
In one embodiment, the therapeutic protein is an antigen-binding protein. In some embodiments, the antigen-binding protein is any one or more of an antibody (e.g., IgG) or antibody fragment, a receptor or soluble fragment thereof, a soluble T-cell receptor fragment, a soluble MHC fragment, a receptor-Fc-fusion protein, a trap-type protein, and a VEGF-Trap protein (e.g., one of which has a primary structure of SEQ ID NO:1). In one embodiment, the therapeutic protein comprises an Fc domain. In one embodiment, the protein is an antibody. In another embodiment, the protein is an IgG. In another embodiment, the therapeutic protein is a receptor-Fc-fusion protein. In another embodiment, the therapeutic protein is a trap-type protein, which comprises a cognate-receptor fragment and an Fc domain. In one particular embodiment, the therapeutic protein is a VEGF-Trap protein. In yet another embodiment, the VEGF-Trap comprises the amino acid sequence set forth in SEQ ID NO:1.
In one embodiment, the polymer cortex comprises a biocompatible polymer. In one embodiment, the polymer cortex comprises a bioerodible polymer. In one embodiment, the polymer cortex comprises a biodegradable polymer. In some embodiments, the polymer cortex comprises a polymer selected from the group consisting of polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-
In one embodiment, plurality of microparticles comprises a collection of microparticles having a range of thicknesses of the polymer cortex, such that individual microparticles of the collection of microparticles degrades at a different rate, which allows for a uniform rate of release of the therapeutic protein.
In one embodiment, the plurality of microparticles comprises a mixture of uncoated micronized protein particles and microparticles having a range of thicknesses of the polymer cortex, which allows for the release of therapeutic protein at periodic intervals based on cortex thickness.
In one embodiment, the plurality of microparticles comprises a mixture of microparticles having polymer cortices of varying levels of hydrophobicity to control the timing or duration of degradation and subsequent release. In one embodiment, the microparticles each comprise an inner polymer layer and an outer polymer layer, wherein the outer polymer layer limits the hydration of the inner polymer layer to control release of the therapeutic protein.
In one embodiment, the therapeutic protein is released from the plurality of microparticles at a rate of from about 0.01 mg/week to about 0.30 mg/week for a duration of at least 60 days, when the microparticles are in an aqueous environment. In one embodiment, the aqueous environment is in vitro buffer. In one embodiment, the aqueous environment is in vivo. In one embodiment, the aqueous environment is ex vivo. In one embodiment, the aqueous environment is a vitreous humor.
In one embodiment, the extended release formulation comprises a plurality of microparticles, which range in size from about two microns to about 70 microns and which comprise (a) a core of micronized therapeutic protein of from about two microns to about 30 microns, wherein the therapeutic protein is an antigen-binding protein, which in some cases can be any one or more of an antibody or antibody fragment, a receptor or soluble fragment thereof, a soluble T-cell receptor fragment, a soluble MHC fragment, a receptor-Fc-fusion protein, a trap-type protein, and a VEGF-Trap protein; and (b) a polymer cortex of a range of thicknesses, wherein the polymer is any one or more of a biocompatible polymer, a biodegradable polymer, a bio-erodible polymer, polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-
In one embodiment, the extended release formulation comprises a plurality of microparticles, which range in size from about two microns to about 70 microns, with a median size of from about 15 microns to about 30 microns, and which comprise (a) a micronized protein core of from about two microns to about 30 microns, with a median size of about 10 microns to about 12 microns, wherein the protein is a VEGF-Trap protein; and (b) a polymer cortex of a range of thicknesses, wherein the polymer is any one or more of PLGA, ethyl cellulose, and polyorthoester, and copolymers or derivatives thereof, such that in an aqueous environment the microparticles release or deliver a steady level of VEGF Trap at a rate of about 0.06±0.02 mg/week for at least 60 days.
In one aspect, the invention provides a method for modulating the release of a protein. In one embodiment, the method comprises the step of making a plurality of microparticles as described in the previous aspect, followed by the step of placing the microparticles into a solvent. The solvent in some embodiments is aqueous. The solvent can be in vitro, such as in a phosphate buffered solution. The solvent can be in vivo, such as e.g. vitreous humor.
The micro particle and protein core particle of the subject invention are roughly spherical in shape. Some microparticles and protein cores will approach sphericity, while others will be more irregular in shape. Thus, as used herein, the term “diameter” means each and any of the following: (a) the diameter of a sphere which circumscribes the microparticle or protein core, (b) the diameter of the largest sphere that fits within the confines of the microparticle or the protein core, (c) any measure between the circumscribed sphere of (a) and the confined sphere of (b), including the mean between the two, (d) the length of the longest axis of the microparticle or protein core, (e) the length of the shortest axis of the microparticle or protein core, (f) any measure between the length of the long axis (d) and the length of the short axis (e), including the mean between the two, and/or (g) equivalent circular diameter (“ECD”), as determined by micro-flow imaging (MFI), nanoparticle tracking analysis (NTA), or light obscuration methods such as dynamic light scattering (DLS). See generally Sharma et al., Micro-flow imaging: flow microscopy applied to subvisible particulate analysis in protein formulations, AAPS J. 2010 September; 12(3): 455-64. Diameter is generally expressed in micrometers (μm or micron). Diameter can be determined by optical measurement
“Micronized protein particle” or “protein particle” means a particle containing multiple molecules of protein with low, very low, or close to zero amounts of water (e.g., <3% water by weight). As used herein, the micronized protein particle is generally spherical in shape and has an ECD ranging from 2 microns to about 35 microns. The micronized protein particle is not limited to any particular protein entity, and is suited to the preparation and delivery of a therapeutic protein. Common therapeutic proteins include inter alia antigen-binding proteins, such as e.g., soluble receptor fragments, antibodies (including IgGs) and derivatives or fragments of antibodies, other Fc containing proteins, including Fc fusion proteins, and receptor-Fc fusion proteins, including the trap-type proteins (Huang, C., Curr. Opin. Biotechnol. 20: 692-99 (2009)) such as e.g. VEGF-Trap.
The micronized protein particle of the invention can be made by any method known in the art for making micron-sized protein particles. For example, the protein particle may be made by inter alia spray-drying (infra), lyophilization, jet milling, hanging drop crystallization (Ruth et al., Acta Crystallographica D56: 524-28 (2000)), gradual precipitation (U.S. Pat. No. 7,998,477 (2011)), lyophilization of a protein-PEG (polyethylene glycol) aqueous mixture (Morita et al., Pharma. Res. 17: 1367-73 (2000)), supercritical fluid precipitation (U.S. Pat. No. 6,063,910 (2000)), or high pressure carbon dioxide induced particle formation (Bustami et al., Pharma. Res. 17: 1360-66 (2000)).
As used herein, the term “protein” refers to a molecule comprising two or more amino acid residues joined to each other by peptide bonds. Peptides, polypeptides and proteins are also inclusive of modifications including, but not limited to, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation. Polypeptides can be of scientific or commercial interest, including protein-based drugs. Polypeptides include, among other things, antibodies and chimeric or fusion proteins. Polypeptides are produced by recombinant animal cell lines using cell culture methods.
An “antibody” is intended to refer to immunoglobulin molecules consisting of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain has a heavy chain variable region (HCVR or VH) and a heavy chain constant region. The heavy chain constant region contains three domains, CH1, CH2 and CH3. Each light chain has of a light chain variable region and a light chain constant region. The light chain constant region consists of one domain (CL). The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The term “antibody” includes reference to both glycosylated and non-glycosylated immunoglobulins of any isotype or subclass. The term “antibody” is inclusive of, but not limited to, those that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell transfected to express the antibody. An IgG comprises a subset of antibodies.
“Fc fusion proteins” comprise part or all of two or more proteins, one of which is an Fc portion of an immunoglobulin molecule, that are not fused in their natural state. Preparation of fusion proteins comprising certain heterologous polypeptides fused to various portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al., Proc. Natl. Acad. Sci. USA 88: 10535, 1991; Byrn et al., Nature 344:677, 1990; and Hollenbaugh et al., “Construction of Immunoglobulin Fusion Proteins”, in Current Protocols in Immunology, Suppl. 4, pages 10.19.1-10.19.11, 1992. “Receptor Fc fusion proteins” comprise one or more of one or more extracellular domain(s) of a receptor coupled to an Fc moiety, which in some embodiments comprises a hinge region followed by a CH2 and CH3 domain of an immunoglobulin. In some embodiments, the Fc-fusion protein contains two or more distinct receptor chains that bind to a single or more than one ligand(s). For example, an Fc-fusion protein is a trap, such as for example an IL-1 trap (e.g., Rilonacept, which contains the IL-1RAcP ligand binding region fused to the IL-1R1 extracellular region fused to Fc of hIgG1; see U.S. Pat. No. 6,927,004, which is herein incorporated by reference in its entirety), or a VEGF Trap (e.g., Aflibercept, which contains the Ig domain 2 of the VEGF receptor Flt1 fused to the Ig domain 3 of the VEGF receptor Flk1 fused to Fc of hIgG1; e.g., SEQ ID NO:1; see U.S. Pat. Nos. 7,087,411 and 7,279,159, which are herein incorporated by reference in their entirety).
As used herein, the term “polymer” refers to a macromolecule comprising repeating monomers connected by covalent chemical bonds. Polymers used in the practice of this invention are biocompatible and biodegradable. A biocompatible and biodegradable polymer can be natural or synthetic. Natural polymers include polynucleotides, polypeptides, such as naturally occurring proteins, recombinant proteins, gelatin, collagens, fibrins, fibroin, polyaspartates, polyglutamates, polylysine, leucine-glutamate co-polymers; and polysaccharides, such as cellulose alginates, dextran and dextran hydrogel polymers, amylose, inulin, pectin and guar gum, chitosan, chitin, heparin, and hyaluronic acid. Synthetic biocompatible or biodegradable polymers include polylactic acid (PLA), polyglycolic acid (PGA), polylactic-polyglycolic copolymer (PLGA), poly-
Ethyl cellulose (EC) is a well-known and readily available biomaterial used in the pharmaceutical and food sciences. It is a cellulose derivative in which some of the glucose hydroxyl groups are replaced with ethyl ether. See Martinac et al., J. Microencapsulation, 22(5): 549-561 (2005) and references therein, which describe methods of using ethyl cellulose as biocompatible polymers in the manufacture of microspheres. See also U.S. Pat. No. 4,210,529 (1980) and references therein for a detailed description of ethyl cellulose and methods of making derivatives of ethyl cellulose.
Poly-
Poly-ε-caprolactone (PCL) is another biocompatible and biodegradable polymer approved by the FDA for use in humans as a drug delivery device. PCL is a polyester of ε-caprolactone, which hydrolyses rapidly in the body to form a non-toxic or low toxicity hydroxycarboxylic acid. For a description of the manufacture of PCL, see Labet and Thielemans, Chemical Society Reviews 38: 3484-3504 (2009) and references therein. For a description of the manufacture and use of PCL-based microspheres and nanospheres as delivery systems, see Sinha et al., Int. J. Pharm., 278(1): 1-23 (2004) and references therein.
Polyorthoester (POE) is a bioerodible polymer designed for drug delivery. It is generally a polymer of a ketene acetal, preferably a cyclic diketene acetal, such as e.g., 3,9-dimethylene-2,4,8,10-tetraoxa spiro[5.5]-undecane, which is polymerized via glycol condensation to form the orthoester linkages. A description of polyorthoester synthesis and various types can be found e.g. in U.S. Pat. No. 4,304,767. Polyorthoesters can be modified to control their drug release profile and degradation rates by swapping in or out various hydrophobic diols and polyols, such as e.g., replacing a hexanetriol with a decanetriol; as well as adding latent acids, such as e.g., octanedioic acid or the like, to the backbone to increase pH sensitivity. Other modifications to the polyorthoester include the integration of an amine to increase functionality. The formation, description, and use of polyorthoesters are described in U.S. Pat. Nos. 5,968,543; 4,764,364; Heller and Barr, Biomacromolecules, 5(5): 1625-32 (2004); and Heller, Adv. Drug. Deliv. Rev., 57: 2053-62 (2005).
As used herein, the phrase “spray-dry” means a method of producing a dry powder comprising micron-sized particles from a slurry or suspension by using a spray-dryer. Spray dryers employ an atomizer or spray nozzle to disperse the suspension or slurry into a controlled drop size spray. Drop sizes from 10 to 500 μm can be generated by spray-drying. As the solvent (water or organic solvent) dries, the protein substance dries into a micron-sized particle, forming a powder-like substance; or in the case of a protein-polymer suspension, during drying, the polymer hardened shell around the protein load.
The microparticles of the invention comprise a protein core surrounded by a polymer cortex or coat. Briefly, a micronized protein particle is formed, which is then dispersed in a polymer solution (polymer dissolved in solvent) to form a protein-polymer suspension. The protein-polymer suspension is then dispersed into micronized (atomized) droplets, and the solvent is driven-off to form the microparticle.
In one embodiment, the micronized protein particle is formed by making a solution of the protein and then subjecting that protein solution to dispersion and heat to form a dry powder comprising the protein. One method to form the micronized protein particles is by spray-drying. In one embodiment, the protein is a therapeutic protein that is formulated to include buffers, stabilizers and other pharmaceutically acceptable excipients to make a pharmaceutical formulation of the therapeutic protein. Exemplary pharmaceutical formulations are described in U.S. Pat. Nos. 7,365,165, 7,572,893, 7,608,261, 7,655,758, 7,807,164, US 2010-0279933, US 2011-0171241, and PCT/US11/54856.
The amount of therapeutic protein contained within the pharmaceutical formulations of the present invention may vary depending on the specific properties desired of the formulations, as well as the particular circumstances and purposes for which the formulations are intended to be used. In certain embodiments, the pharmaceutical formulations may contain about 1 mg/mL to about 500 mg/mL of protein; about 5 mg/mL to about 400 mg/mL of protein; about 5 mg/mL to about 200 mg/mL of protein; about 25 mg/mL to about 180 mg/mL of protein; about 25 mg/mL to about 150 mg/mL of protein; or about 50 mg/mL to about 180 mg/mL of protein. For example, the formulations of the present invention may comprise about 1 mg/mL; about 2 mg/mL; about 5 mg/mL; about 10 mg/mL; about 15 mg/mL; about 20 mg/mL; about 25 mg/mL; about 30 mg/mL; about 35 mg/mL; about 40 mg/mL; about 45 mg/mL; about 50 mg/mL; about 55 mg/mL; about 60 mg/mL; about 65 mg/mL; about 70 mg/mL; about 75 mg/mL; about 80 mg/mL; about 85 mg/mL; about 86 mg/mL; about 87 mg/mL; about 88 mg/mL; about 89 mg/mL; about 90 mg/mL; about 95 mg/mL; about 100 mg/mL; about 105 mg/mL; about 110 mg/mL; about 115 mg/mL; about 120 mg/mL; about 125 mg/mL; about 130 mg/mL; about 131 mg/mL; about 132 mg/mL; about 133 mg/mL; about 134 mg/mL; about 135 mg/mL; about 140 mg/mL; about 145 mg/mL; about 150 mg/mL; about 155 mg/mL; about 160 mg/mL; about 165 mg/mL; about 170 mg/mL; about 175 mg/mL; about 180 mg/mL; about 185 mg/mL; about 190 mg/mL; about 195 mg/mL; about 200 mg/mL; about 205 mg/mL; about 210 mg/mL; about 215 mg/mL; about 220 mg/mL; about 225 mg/mL; about 230 mg/mL; about 235 mg/mL; about 240 mg/mL; about 245 mg/mL; about 250 mg/mL; about 255 mg/mL; about 260 mg/mL; about 265 mg/mL; about 270 mg/mL; about 275 mg/mL; about 280 mg/mL; about 285 mg/mL; about 200 mg/mL; about 200 mg/mL; or about 300 mg/mL of therapeutic protein.
The pharmaceutical formulations of the present invention comprise one or more excipients. The term “excipient,” as used herein, means any non-therapeutic agent added to the formulation to provide a desired consistency, viscosity or stabilizing effect.
The pharmaceutical formulations of the present invention may also comprise one or more carbohydrate, e.g., one or more sugar. The sugar can be a reducing sugar or a non-reducing sugar. “Reducing sugars” include, e.g., sugars with a ketone or aldehyde group and contain a reactive hemiacetal group, which allows the sugar to act as a reducing agent. Specific examples of reducing sugars include fructose, glucose, glyceraldehyde, lactose, arabinose, mannose, xylose, ribose, rhamnose, galactose and maltose. Non-reducing sugars can comprise an anomeric carbon that is an acetal and is not substantially reactive with amino acids or polypeptides to initiate a Maillard reaction. Specific examples of non-reducing sugars include sucrose, trehalose, sorbose, sucralose, melezitose and raffinose. Sugar acids include, for example, saccharic acids, gluconate and other polyhydroxy sugars and salts thereof.
The amount of sugar contained within the pharmaceutical formulations of the present invention will vary depending on the specific circumstances and intended purposes for which the formulations are used. In certain embodiments, the formulations may contain about 0.1% to about 20% sugar; about 0.5% to about 20% sugar; about 1% to about 20% sugar; about 2% to about 15% sugar; about 3% to about 10% sugar; about 4% to about 10% sugar; or about 5% to about 10% sugar. For example, the pharmaceutical formulations of the present invention may comprise about 0.5%; about 1.0%; about 1.5%; about 2.0%; about 2.5%; about 3.0%; about 3.5%; about 4.0%; about 4.5%; about 5.0%; about 5.5%; about 6.0%; 6.5%; about 7.0%; about 7.5%; about 8.0%; about 8.5%; about 9.0%; about 9.5%; about 10.0%; about 10.5%; about 11.0%; about 11.5%; about 12.0%; about 12.5%; about 13.0%; about 13.5%; about 14.0%; about 14.5%; about 15.0%; about 15.5%; about 16.0%; 16.5%; about 17.0%; about 17.5%; about 18.0%; about 18.5%; about 19.0%; about 19.5%; or about 20.0% sugar (e.g., sucrose).
The pharmaceutical formulations of the present invention may also comprise one or more surfactant. As used herein, the term “surfactant” means a substance which reduces the surface tension of a fluid in which it is dissolved and/or reduces the interfacial tension between oil and water. Surfactants can be ionic or non-ionic. Exemplary non-ionic surfactants that can be included in the formulations of the present invention include, e.g., alkyl poly(ethylene oxide), alkyl polyglucosides (e.g., octyl glucoside and decyl maltoside), fatty alcohols such as cetyl alcohol and oleyl alcohol, cocamide MEA, cocamide DEA, and cocamide TEA. Specific non-ionic surfactants that can be included in the formulations of the present invention include, e.g., polysorbates such as polysorbate 20, polysorbate 28, polysorbate 40, polysorbate 60, polysorbate 65, polysorbate 80, polysorbate 81, and polysorbate 85; poloxamers such as poloxamer 188, poloxamer 407; polyethylene-polypropylene glycol; or polyethylene glycol (PEG). Polysorbate 20 is also known as TWEEN 20, sorbitan monolaurate and polyoxyethylenesorbitan monolaurate.
The amount of surfactant contained within the pharmaceutical formulations of the present invention may vary depending on the specific properties desired of the formulations, as well as the particular circumstances and purposes for which the formulations are intended to be used. In certain embodiments, the formulations may contain about 0.05% to about 5% surfactant; or about 0.1% to about 0.2% surfactant. For example, the formulations of the present invention may comprise about 0.05%; about 0.06%; about 0.07%; about 0.08%; about 0.09%; about 0.10%; about 0.11%; about 0.12%; about 0.13%; about 0.14%; about 0.15%; about 0.16%; about 0.17%; about 0.18%; about 0.19%; about 0.20%; about 0.21%; about 0.22%; about 0.23%; about 0.24%; about 0.25%; about 0.26%; about 0.27%; about 0.28%; about 0.29%; or about 0.30% surfactant (e.g., polysorbate 20).
The pharmaceutical formulations of the present invention may also comprise one or more buffers. In some embodiments, the buffer has a buffering range that overlaps fully or in part the range of pH 5.5-7.4. In one embodiment, the buffer has a pKa of about 6.0±0.5. In certain embodiments, the buffer comprises a phosphate buffer. In certain embodiments, the phosphate is present at a concentration of 5 mM±0.75 mM to 15 mM±2.25 mM; 6 mM±0.9 mM to 14 mM±2.1 mM; 7 mM±1.05 mM to 13 mM±1.95 mM; 8 mM±1.2 mM to 12 mM±1.8 mM; 9 mM±1.35 mM to 11 mM±1.65 mM; 10 mM±1.5 mM; or about 10 mM. In certain embodiments, the buffer system comprises histidine at 10 mM±1.5 mM, at a pH of 6.0±0.5.
The pharmaceutical formulations of the present invention may have a pH of from about 5.0 to about 8.0. For example, the formulations of the present invention may have a pH of about 5.0; about 5.2; about 5.4; about 5.6; about 5.8; about 6.0; about 6.2; about 6.4; about 6.6; about 6.8; about 7.0; about 7.2; about 7.4; about 7.6; about 7.8; or about 8.0.
In one particular embodiment, the therapeutic protein is a VEGF Trap protein. Pharmaceutical formulations for the formation of micronized VEGF Trap protein particles may contain from about 10 mg/mL to about 100 mg/mL VEGF Trap protein, about 10 mg/mL, about 15 mg/mL, about 20 mg/mL, about 25 mg/mL, about 30 mg/mL, about 35 mg/mL, about 40 mg/mL, about 45 mg/mL, about 50 mg/mL, about 55 mg/mL, about 60 mg/mL, about 65 mg/mL, about 70 mg/mL, about 75 mg/mL, about 80 mg/mL, about 85 mg/mL, about 90 mg/mL, about 95 mg/mL, or about 100 mg/mL VEGF Trap protein. Solutions may contain one or more buffers of from about 5 mM to about 50 mM. In one embodiment, the buffer is about 10 mM phosphate at a pH of about 6±0.5. Solutions may also contain sucrose at a concentration of from about 1% to about 10%. In one embodiment, the solution contains sucrose at about 2% w/w.
In some embodiments, the therapeutic protein solution contains VEGF Trap protein at about 25 mg/mL or about 50 mg/mL in 10 mM phosphate, pH 6.2, 2% sucrose, and optionally 0.1% polysorbate.
The therapeutic protein formulation is then subjected to dispersion and drying to form micronized protein particles. One method of making the micronized protein particles is to subject the protein solution to spray-drying. Spray-drying is generally known in the art and may be performed on equipment such as e.g., a BÜCHI Mini Spray Dryer B-290 (Büchi Labortechnik AG, Flawil, CH). In one particular embodiment, the protein solution (e.g., but not limited to any one of the VEGF Trap formulations described above) is pumped into the spray dryer at a rate of about 2 mL/min to about 15 mL/min, or about 7 mL/min. The inlet temperature of the spray dryer is set at a temperature above the boiling point of water, such as e.g., at about 130° C. The outlet temperature at a temperature below the boiling point of water and above ambient temperature, such as e.g., 55° C. In one specific embodiment, a protein solution (e.g., VEGF Trap solution or IgG solution) is pumped into a BÜCHI Mini Spray Dryer B-290 at about 7 mL/min, with an inlet temperature of about 130° C. and an outlet temperature of about 55° C., with the aspirator set at 33 m3/h and the spray gas at 530 L/h.
The resulting micronized protein particles range in size from about 1 μm to about 100 μm in diameter, depending upon the particular formulation and concentration of protein and excipients. In some embodiments, the micronized protein particles have a diameter of from about 1 μm to about 100 μm, from about 1 μm to about 40 μm, from about 2 μm to about 15 μm, from about 2.5 μm to about 13 μm, from about 3 μm to about 10 μm, about 5 μm, about 6 μm, about 7 μm, about 8 μm, about 9 μm, about 10 μm, about 11 μm, or about 12 μm.
The micronized protein particles are then coated with a biocompatible and biodegradable polymer. This is can be accomplished by suspending the micronized protein particles in a polymer solution. A polymer solution is essentially a polymer dissolved in a solvent. For example, the biocompatible and biodegradable polymer may be dissolved in inter alia methylene chloride, tetrahydrofuran, ethyl acetate, or some other useful solvent. Ethyl acetate is widely known as a safe solvent and is often used in the preparation of drugs, implants and foodstuffs.
In some embodiments, the polymer can be ethyl cellulose (“EC”), poly(lactic acid) (“PLA”), polyorthoester (“POE”), poly-
The micronized protein particles are added to the polymer solution at about 10 mg/mL to about 100 mg/mL, about 15 mg/mL to about 95 mg/mL, about 20 mg/mL to about 90 mg/mL, about 25 mg/mL to about 85 mg/mL, about 30 mg/mL to about 80 mg/mL, about 35 mg/mL to about 75 mg/mL, about 40 mg/mL to about 70 mg/mL, about 45 mg/mL to about 65 mg/mL, about 50 mg/mL to about 60 mg/mL, at about 25 mg/mL, at about 30 mg/mL, at about 35 mg/mL, at about 40 mg/mL, at about 45 mg/mL, or at about 50 mg/mL. The particles are mixed to form a slurry or suspension, which is then subjected to dispersion and drying to form the polymer coated protein particle (i.e., microparticle).
In one embodiment, the protein particle-polymer solution suspension is subjected the spray-drying, which is performed in a manner similar to the method for manufacturing the micronized protein particles, but with a reduced intake temperature to protect against igniting the organic solvent or polymer. Briefly, the protein particle-polymer solution suspension is pumped into the spray dryer at a rate of about 5 mL/min to about 20 mL/min, or about 12.5 mL/min. The suspension was pumped at 12.5 mL/min into the spray dryer with an aspirator air and spray gas flow rate of about 530 L/h and 35 m3/h (mm), respectively. The inlet temperature was set at 90° and the outlet temperature was set at about 54° C. The inlet temperature of the spray dryer is set at a temperature above the flash point of the solvent, such as e.g., at about 90° C. The outlet temperature at a temperature below the intake temperature and above ambient temperature, such as e.g., about 54° C. In one particular embodiment, a suspension containing about 50 mg/mL of protein particle (e.g., VEGF Trap) in about 50 mg/mL to about 250 mg/mL polymer/ethyl acetate solution is pumped into a BÜCHI Mini Spray Dryer B-290 at about 12.5 mL/min, with an inlet temperature of about 90° C. and an outlet temperature of about 54° C., with the aspirator set at about 35 m3/h and the spray gas at about 530 L/h.
The resulting microparticles, which contain a protein particle core within a polymer cortex, have a range of diameters of from about 2 μm to about 70 μm, about 5 μm to about 65 μm, about 10 μm to about 60 μm, about 15 μm to about 55 μm, about 20 μm to about 50 μm, about 15 μm, about 20 μm, about 25 μm, or about 30 μm. The size variation in large part reflects the thickness of the polymer cortex, although the diameter of the protein core could contribute to size variation to some extent. Manipulating the starting concentration of the polymer solution, and/or the polymer itself can control the diameter of the microparticle. For example, those microparticles which were manufactured using 50 mg/mL polymer have a median size of about 15 μm to 20 μm, whereas those microparticles which were manufactured using 250 mg/mL polymer had a median size of about 30 μm.
The microparticles of the instant invention are useful in the time-release or extended release of protein therapeutics. For example, it is envisioned that the VEGF Trap microparticles are useful in the extended release of VEGF Trap therapeutic protein in, for example, the vitreous for the treatment of vascular eye disorders, or subcutaneous implantation for the extended release of VEGF Trap to treat cancer or other disorders.
The microparticles of the instant invention release protein in a physiological aqueous environment at about 37° C. at a relatively constant rate over an extended period of time, to at least 60 days. In general, those microparticles manufactured with a higher concentration of polymer (e.g., 250 mg/mL) tended to show a relatively linear protein release profile; whereas those microparticles manufactured with a lower concentration of polymer (e.g., 50 mg/mL) tended to show an initial burst followed by an onset of a delayed burst release. Furthermore, microparticles formed from a higher concentration of polymer showed a slower rate of release of protein than those formed from a lower concentration of particles. The quality of protein released from the microparticles over time was consistent with the quality of the stating protein material. Little to no protein degradation occurred.
The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the methods and compositions of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, sizes, etc.) but some experimental errors and deviations should be accounted for.
In the following examples, VEGF-Trap protein (“VGT”), which is a dimer of the polypeptide comprising the amino acid sequence SEQ ID NO:1, serves as an exemplar receptor-Fc-fusion protein.
Solutions containing 25 mg/mL VEGF Trap protein (“VGT”), 25 mg/mL VGT plus 0.1% polysorbate 80, and 50 mg/mL VGT in 10 mM phosphate, 2% sucrose, pH 6.2 were each independently atomized in a spray dry micronizer (BÜCHI Mini Spray Dryer B-290, Büchi Labortechnik AG, Flawil, CH) to form droplets containing VEGF Trap. Heat was applied to evaporate the water from the droplets, resulting in a powder containing VEGF Trap. The inlet temperature was set at 130° C. and outlet temperature at about 55° C. The aspirator was set at 33 m3/h and spray gas at 530 L/h. The VGT solution was pumped at about 7 mL/min.
The size of the resultant VGT particles was measured by micro-flow imaging (MFI) and dynamic light imaging (DLS).
VGT particles were reconstituted in water for injection and examined via size exclusion, i.e., size exclusion-ultra performance liquid chromatography (SE-UPLC) to determine protein purity. No change in purity was noted after micronization relative to starting material (see Table 3).
Various polymers were used or are contemplated for use in the manufacture of the polymer cortex of the microparticles. Those polymers include inter alia ethyl cellulose (“EC”), polyorthoester (“POE”), poly-
Ethyl Cellulose Coating
Micronized VEGF Trap particles were suspended in a solution of 50 mg/mL ethyl cellulose in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-50-EC suspension”.
Micronized VEGF Trap particles were suspended in a solution of 100 mg/mL ethyl cellulose in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-100-EC suspension”.
Micronized VEGF Trap particles are suspended in a solution of 250 mg/mL ethyl cellulose in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-250-EC suspension”.
Polyorthoester Coating
Micronized VEGF Trap particles were suspended in a solution of 50 mg/mL polyorthoester containing about 5% latent acid in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-50-POE suspension”.
Micronized VEGF Trap particles were suspended in a solution of 250 mg/mL polyorthoester containing about 5% latent acid in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-250-POE suspension”.
Poly-
Micronized VEGF Trap particles were suspended in a solution of 50 mg/mL PLGA in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-50-PLGA suspension”.
Micronized VEGF Trap particles were suspended in a solution of 200 mg/mL PLGA in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-200-PLGA suspension”.
Micronized VEGF Trap particles were suspended in a solution of 250 mg/mL PLGA in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-250-PLGA suspension”.
Poly-ε-Caprolactone Coating
Micronized VEGF Trap particles are suspended in a solution of 50 mg/mL PCL in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-50-PCL suspension”.
Micronized VEGF Trap particles are suspended in a solution of 250 mg/mL PCL in ethyl acetate at a concentration of about 50 mg/mL VGT; herein designated “VGT-250-PCL suspension”.
PCL has a low Tg and may not be suitable for heat-drying as described below, but can be used for solvent extraction in an aqueous bath with polyvinyl alcohol (PVA), for example.
Each VGT polymer suspension, which was made according to Example 2 (supra), was subjected to spray drying using a BÜCHI Mini Spray Dryer B-290 (Büchi Labortechnik AG, Flawil, CH). Briefly, each suspension was atomized to form microdroplets, which were subsequently heat dried to remove the solvent and form the polymer-coated protein microparticles. The suspension was pumped at 12.5 mL/min into the spray dryer with an aspirator air and spray gas flow rate of about 530 L/h and 35 m3/h, respectively. The inlet temperature was set at 90° and the outlet temperature was set at about 54° C.
Spray dried polymer coated protein particles manufactured according to the exemplified process generate a plurality of microparticles having a range of equivalent circular diameters of from about 2.5 μm to about 65 μm (
The diameter of the microparticle correlates with the starting concentration of the polymer solution (Table 2,
The stability of the VEGF-Trap protein was assessed using quantitative size exclusion chromatography (SE-UPLC), which allows for the quantification of smaller degradation products and larger aggregation products relative to the intact monomer. The results are described in Table 3. Essentially, the protein remained stable throughout the spray drying and spray coating processes.
The average ratio of protein to polymer by weight was also determined for the manufactured microparticles. A collection of microparticles manufactured with varying polymers and polymer concentration was extracted and subjected to quantitative reverse phase chromatography (RP-HPLC). The results are presented in Table 3. The data may be interpreted to support the theory that a higher starting concentration of polymer yields a thicker polymer cortex on the microparticle.
1Based on extracted VEGF Trap after 1 hour reconstitution to remove uncoated VEGF Trap.
2Average of percent native by SE-UPLC (n = 4).
3Average of percent weight to weight loading of VGT to polymer by RP-HPLC (n = 4).
The release of protein from microparticles was determined by suspending various batches of microparticles in buffer (10 mM phosphate, 0.03% polysorbate 20, pH 7.0) and measuring the amount and quality of protein released into solution over time while incubated at 37° C. At 1-2 week intervals, the microparticles were pelleted by mild centrifugation and 80% of the supernatant containing released protein was collected for subsequent analysis. An equivalent amount of fresh buffer was replaced and the microparticles were resuspended by mild vortexing and returned to the 37° C. incubation chamber. Protein amount and quality in the supernatant was assessed by size exclusion chromatography.
In general, those microparticles manufactured with a higher concentration of polymer (e.g., 250 mg/mL) tended to show a relatively linear protein release profile; whereas those microparticles manufactured with a lower concentration of polymer (e.g., 50 mg/mL) tended to show an initial burst followed by an onset of a delayed burst release. The data showing the extended release of protein, which remained stable, for up to about 60 days is depicted in
Particle size distributions were controlled by polymer concentration and atomization spray gas flow. Increased polymer concentration shifted the distribution towards larger particles (200 mg/mL PLGA at 45 mm spray gas flow v. 100 mg/mL PLGA at 45 mm spray gas flow; see Table 5). Similarly, a lower atomization spray gas flow resulted in larger droplets and thus, larger particles (100 mg/mL PLGA at 25 mm spray gas flow v. 100 mg/mL PLGA at 45 mm spray gas flow; see Table 5).
VEGF Trap or IgG was spray coated with low molecular weight (202S) poly(lactic acid) (PLA-LMW), high molecular weight (203S) poly(lactic acid) (PLA-HMW), polyanhydride poly[1,6-bis(p-carboxyphenoxy)hexane] (pCPH), poly(hydroxybutyric acid-cohydroxyvaleric acid) (PHB-PVA), PEG-poly(lactic acid) block copolymer (PEG-PLA), and poly-
VEGF Trap and IgG were extracted from their respective polymer coats and measured for purity by SE-UPLC. The results are summarized in Table 7. The proteins generally were compatible with the spray coating process for the polymers tested. Protein remained stable for at least 14 days for those polymers that continued to release protein.
This application is a continuation application of U.S. patent application Ser. No. 15/450,335, filed on Mar. 6, 2017, which is a divisional of U.S. patent application Ser. No. 13/680,069, filed Nov. 18, 2012, which claims priority to U.S. Provisional Patent Application No. 61/561,525, filed Nov. 18, 2011, all of which are herein specifically incorporated by reference in their entirety.
Number | Name | Date | Kind |
---|---|---|---|
4210529 | Petersen | Jul 1980 | A |
4304767 | Heller et al. | Dec 1981 | A |
4764364 | Heller et al. | Aug 1988 | A |
5104221 | Bott et al. | Apr 1992 | A |
5968543 | Heller et al. | Oct 1999 | A |
5985309 | Edwards et al. | Nov 1999 | A |
6004549 | Reichert et al. | Dec 1999 | A |
6011011 | Hageman | Jan 2000 | A |
6019968 | Platz et al. | Feb 2000 | A |
6063910 | Debenedetti et al. | May 2000 | A |
6284282 | Maa et al. | Sep 2001 | B1 |
6308434 | Chickering, III et al. | Oct 2001 | B1 |
6927004 | Eurlings et al. | Aug 2005 | B2 |
6927044 | Stahl et al. | Aug 2005 | B2 |
6956021 | Edwards et al. | Oct 2005 | B1 |
7087411 | Daly et al. | Aug 2006 | B2 |
7279158 | Wang et al. | Oct 2007 | B2 |
7279159 | Daly et al. | Oct 2007 | B2 |
7303746 | Wiegand et al. | Dec 2007 | B2 |
7303747 | Wiegand et al. | Dec 2007 | B2 |
7365165 | Dix et al. | Apr 2008 | B2 |
7374758 | Papadopoulos et al. | May 2008 | B2 |
7572893 | Dix et al. | Aug 2009 | B2 |
7608261 | Furfine et al. | Oct 2009 | B2 |
7655758 | Dix et al. | Feb 2010 | B2 |
7807164 | Furfine et al. | Oct 2010 | B2 |
7927583 | Stahl et al. | Apr 2011 | B2 |
7998477 | Yakovlevsky et al. | Aug 2011 | B2 |
8063182 | Brockhaus et al. | Nov 2011 | B1 |
8173599 | Moon et al. | May 2012 | B2 |
8586713 | Davis et al. | Nov 2013 | B2 |
8623395 | de Juan, Jr. et al. | Jan 2014 | B2 |
9033911 | de Juan, Jr. et al. | May 2015 | B2 |
9155702 | Lee et al. | Oct 2015 | B2 |
9376479 | Govardhan et al. | Jun 2016 | B2 |
9610301 | Lassner et al. | Apr 2017 | B2 |
10611850 | Christian et al. | Apr 2020 | B2 |
20040043076 | Dulieu et al. | Mar 2004 | A1 |
20040142043 | Maeda et al. | Jul 2004 | A1 |
20040208928 | Liao et al. | Oct 2004 | A1 |
20040209804 | Govardhan et al. | Oct 2004 | A1 |
20040219224 | Yakovlevsky et al. | Nov 2004 | A1 |
20050175708 | Carrasquillo et al. | Aug 2005 | A1 |
20050186143 | Stevenson et al. | Aug 2005 | A1 |
20050250704 | Bassarab et al. | Nov 2005 | A1 |
20050281861 | Hughes et al. | Dec 2005 | A1 |
20060210641 | Shalaby | Sep 2006 | A1 |
20070059336 | Hughes et al. | Mar 2007 | A1 |
20070071825 | Curdy et al. | Mar 2007 | A1 |
20070264341 | Lee et al. | Nov 2007 | A1 |
20070292475 | Campbell et al. | Dec 2007 | A1 |
20070298116 | Bechtold-Peters et al. | Dec 2007 | A1 |
20080071064 | Schultz-Fademrecht et al. | Mar 2008 | A1 |
20080305115 | Tice et al. | Dec 2008 | A1 |
20090226530 | Lassner et al. | Sep 2009 | A1 |
20090269414 | Lee et al. | Oct 2009 | A1 |
20100279933 | Dix et al. | Nov 2010 | A1 |
20110046870 | Ross et al. | Feb 2011 | A1 |
20110104151 | Nettles et al. | May 2011 | A1 |
20110171241 | Dix et al. | Jul 2011 | A1 |
20110212180 | Ramtoola | Sep 2011 | A1 |
20120095439 | de Juan, Jr. et al. | Apr 2012 | A1 |
20130129830 | Chen et al. | May 2013 | A1 |
20130324942 | de Juan, Jr. et al. | Dec 2013 | A1 |
20140170204 | Desai et al. | Jun 2014 | A1 |
20140363482 | Guo et al. | Dec 2014 | A1 |
20150119807 | Desai et al. | Apr 2015 | A1 |
20150216829 | Conte et al. | Aug 2015 | A1 |
20150250647 | de Juan, Jr. et al. | Sep 2015 | A1 |
20160030553 | Legon | Feb 2016 | A1 |
20160176986 | Armstrong et al. | Jun 2016 | A1 |
20170181978 | Chen et al. | Jun 2017 | A1 |
20170198058 | Christian et al. | Jul 2017 | A1 |
20180289623 | Chen et al. | Oct 2018 | A1 |
20200022917 | Brudnicki et al. | Jan 2020 | A1 |
Number | Date | Country |
---|---|---|
2012340107 | Jun 2014 | AU |
2016340072 | May 2018 | AU |
2016369557 | Jun 2018 | AU |
112014011915 | May 2017 | BR |
112018007507 | Oct 2018 | BR |
112018010743 | Nov 2018 | BR |
2710062 | Jul 2009 | CA |
2711984 | Jul 2009 | CA |
2855749 | May 2013 | CA |
3076725 | May 2013 | CA |
3001346 | Apr 2017 | CA |
3003654 | Jun 2017 | CA |
2018001623 | Jul 2018 | CL |
1222403 | Jul 1999 | CN |
101132800 | Feb 2008 | CN |
101559041 | Oct 2009 | CN |
102365109 | Feb 2012 | CN |
103108656 | May 2013 | CN |
103209664 | Jul 2013 | CN |
103717237 | Apr 2014 | CN |
103957898 | Jul 2014 | CN |
105688188 | Jun 2016 | CN |
108348462 | Jul 2018 | CN |
108366968 | Aug 2018 | CN |
2018005920 | Jun 2018 | CO |
102006030164 | Jan 2008 | DE |
2790681 | Apr 2020 | DK |
3384903 | Jul 2020 | DK |
201890979 | Oct 2018 | EA |
201891164 | Nov 2018 | EA |
2790681 | Oct 2014 | EP |
3362041 | Aug 2018 | EP |
3384903 | Oct 2018 | EP |
3389636 | Oct 2018 | EP |
3574897 | Dec 2019 | EP |
2775104 | Jul 2020 | ES |
1251461 | Feb 2019 | HK |
258570 | May 2018 | IL |
259204 | Jul 2018 | IL |
232328 | Nov 2018 | IL |
262651 | Nov 2020 | IL |
H08-501305 | Feb 1996 | JP |
H11-228389 | Aug 1999 | JP |
2004-285079 | Oct 2004 | JP |
2010-522743 | Jul 2010 | JP |
2014-533698 | Dec 2014 | JP |
2017-525679 | Sep 2017 | JP |
6267649 | Jan 2018 | JP |
2018-141004 | Sep 2018 | JP |
2018-538243 | Dec 2018 | JP |
2019-504825 | Feb 2019 | JP |
6549653 | Jul 2019 | JP |
6727372 | Jul 2020 | JP |
10-2003-0011858 | Feb 2003 | KR |
10-2009-0025373 | Mar 2009 | KR |
10-2018-0063311 | Jun 2018 | KR |
10-2018-0088836 | Aug 2018 | KR |
2020-0018712 | Feb 2020 | KR |
10-2133352 | Jul 2020 | KR |
2018005382 | Aug 2018 | MX |
2018005383 | Aug 2018 | MX |
2018005384 | Aug 2018 | MX |
2018005387 | Aug 2018 | MX |
2018005388 | Aug 2018 | MX |
2018005390 | Aug 2018 | MX |
362286 | Jan 2019 | MX |
2018004695 | Mar 2019 | MX |
12018500830 | Oct 2018 | PH |
3384903 | Jun 2020 | PT |
2177785 | Jan 2002 | RU |
2237471 | Oct 2004 | RU |
2014124682 | Dec 2015 | RU |
2018101248 | Feb 2019 | RU |
11201402152 | Jun 2014 | SG |
11201803197 | May 2018 | SG |
WO-1992018164 | Oct 1992 | WO |
WO-199618417 | Jun 1996 | WO |
WO-1999038536 | Aug 1999 | WO |
WO-2001030320 | May 2001 | WO |
WO-2003092665 | Nov 2003 | WO |
WO-2007084765 | Jul 2007 | WO |
WO-2008109886 | Sep 2008 | WO |
WO-2008115883 | Sep 2008 | WO |
WO-2008153997 | Dec 2008 | WO |
WO-2009090189 | Jul 2009 | WO |
WO-2010111132 | Sep 2010 | WO |
WO-2011041642 | Apr 2011 | WO |
2012047954 | Apr 2012 | WO |
WO-2012142318 | Oct 2012 | WO |
WO-2013075068 | May 2013 | WO |
WO-2017066554 | Apr 2017 | WO |
WO-2017106716 | Jun 2017 | WO |
201800078 | May 2020 | ZA |
Entry |
---|
U.S. Appl. No. 13/680,069, filed Nov. 18, 2012, Abandoned. |
U.S. Appl. No. 15/450,335, filed Mar. 6, 2017, Issued. |
Hollenbaugh et al., Construction of Immunoglobulin Fusion Proteins. Current Protocols in Immunology. 2002;48:10.19A.1-10.19A.11. |
Sharma et al., Micro-flow imaging: flow microscopy applied to sub-visible particulate analysis in protein formulations. AAPS J. Sep. 2010;12(3):455-64. |
European Office Action for Application No. 22150172.9, dated May 27, 2022, 17 pages. |
Abdul-Fattah et al., The Challenge of Drying Method Selection for Protein Pharmaceuticals: Product Quality Implications. J Pharm Sci. Aug. 2007;96(8):1886-916. |
Ajmera et al., Stabilisation of proteins via mixtures of amino acids during spray drying. Int J Pharm. Mar. 10, 2014;463(1):98-107. |
Ajmera, Stable spray dried protein formulation and implementation in vaccine development. Doctoral thesis, Christian Albrecht University, Kiel, Germany, 140 pages. (2014). |
Albin et al., Theoretical and experimental studies of glucose sensitive membranes. J Control Rel. 1987;6:267-91. |
Andrieu-Soler et al., Intravitreous injection of PLGA microspheres encapsulating GDNF promotes the survival of photoreceptors in the rd1/rd1 mouse. Mol Vis. Nov. 17, 2005;11:1002-11. |
Arpagaus et al., Spray dried biodegradable polymers as target material for controlled drug delivery. Best@buchi. vol. 46, 8 pages, (2007). |
Arthur et al., Detection of protein aggregates by sedimentation velocity analytical ultracentrifugation (SV-AUC): sources of variability and their relative importance. J Pharm Sci. Oct. 2009;98(10):3522-39. |
Ashkenazi et al., Protection against endotoxic shock by a tumor necrosis factor receptor immunoadhesin. Proc Natl Acad Sci U S A. Dec. 1, 1991;88(23):10535-9. |
Astete et al., Synthesis and characterization of PLGA nanoparticles. J Biomater Sci Polym Ed. 2006;17(3):247-89. |
Atha et al., Mechanism of precipitation of proteins by polyethylene glycols. Analysis in terms of excluded volume. J Biol Chem. Dec. 10, 1981;256(23):12108-17. |
Bauman et al., Antagonism of platelet-derived growth factor receptor in non small cell lung cancer: rationale and investigations. Clin Cancer Res. Aug. 1, 2007;13(15 Pt 2):s4632-6. |
Bernards et al., Nanostructured thin film polymer devices for constant-rate protein delivery. Nano Lett. Oct. 10, 2012;12(10):5355-61. |
Bernards et al., Ocular biocompatibility and structural integrity of micro- and nanostructured poly(caprolactone) films. J Ocul Pharmacol Ther. Mar. 2013;29(2):249-57. |
Bhambhani et al., Formulation design and high-throughput excipient selection based on structural integrity and conformational stability of dilute and highly concentrated IgG1 monoclonal antibody solutions. J Pharm Sci. Mar. 2012;101(3):1120-35. (Abstract only). |
Blixt et al., Arraying the post-translational glycoproteome (PTG). Curr Opin Chem Biol. Feb. 2014;18:62-9. |
Broadhead et al., The effect of process and formulation variables on the properties of spray-dried beta-galactosidase. J Pharm Pharmacol. Jun. 1994;46(6):458-67. |
Brown et al., Primary endpoint results of a phase II study of vascular endothelial growth factor trap-eye in wet age-related macular degeneration. Ophthalmology. Jun. 2011;118(6):1089-97. |
Burke et al., Poly(lactide-co-glycolide) microsphere formulations of darbepoetin alfa: spray drying is an alternative to encapsulation by spray-freeze drying. Pharm Res. Mar. 2004;21(3):500-6. |
Bustami et al., Generation of micro-particles of proteins for aerosol delivery using high pressure modified carbon dioxide. Pharm Res. Nov. 2000;17(11):1360-6. |
Butler et al., The choice of mammalian cell host and possibilities for glycosylation engineering. Curr Opin Biotechnol. Dec. 2014;30:107-12. |
Byrn et al., Biological properties of a CD4 immunoadhesin. Nature. Apr. 12, 1990;344(6267):667-70. |
Chen et al., Profiling glycoprotein n-linked oligosaccharide by capillary electrophoresis. Electrophoresis. Nov. 1998;19(15):2639-44. (Abstract only). |
Cleland et al., A specific molar ratio of stabilizer to protein is required for storage stability of a lyophilized monoclonal antibody. J Pharm Sci. Mar. 2001;90(3):310-21. |
Costantino et al., Effect of mannitol crystallization on the stability and aerosol performance of a spray-dried pharmaceutical protein, recombinant humanized anti-IgE monoclonal antibody. J Pharm Sci. Nov. 1998;87(11):1406-11. |
De Boer et al., Characterization of inhalation aerosols: a critical evaluation of cascade impactor analysis and laser diffraction technique. Int J Pharm. Dec. 5, 2002;249(1-2):219-31. |
Do et al., The Da Vinci Study: phase 2 primary results of VEGF Trap-Eye in patients with diabetic macular edema. Ophthalmology. Sep. 2011;118(9):1819-26. |
Doat et al., Intravitreous injection of PLGA microspheres encapsulating GDNF promotes the survival of photoreceptors in the rd1/rd1 mouse. Mol Vis. 2005;11:1002-11. |
Elversson et al., Aqueous two-phase systems as a formulation concept for spray-dried protein. Int J Pharm. Apr. 27, 2005;294(1-2):73-87. |
Erdinc, Encapsulation of proteins by spray drying. Queen's University, Canada. 2007;1-89. |
Fischer et al., Average protein density is a molecular-weight-dependent function. Protein Science. 2004;13:2825-8. |
Frisken, Revisiting the method of cumulants for the analysis of dynamic light-scattering data. Appl Opt. Aug. 20, 2001;40(24):4087-91. |
Gavini et al., PLGA microspheres for the occular delivery of a peptide drug, vancomycin using emulsification/spray-drying as the preparation method: in vitro/in vivo studies. Eur J Pharm Biopharm. 2004;57(2):207-12. |
Gerald et al., Angiopoietin-2: an attractive target for improved antiangiogenic tumor therapy. Cancer Res. Mar. 15, 2013;73(6):1649-57. |
Ghaderi et al., Production platforms for biotherapeutic glycoproteins. Occurrence, impact, and challenges of non-human sialylation. Biotechnol Genet Eng Rev. 2012;28:147-75. |
Gibas et al., Review: synthetic polymer hydrogels for biomedical applications. Chemistry and Chemical Technology. 2010;4:297-304. |
Gikanga et al., Manufacturing of High-Concentration Monoclonal Antibody Formulations via Spray Drying—the Road to Manufacturing Scale. PDA J Pharm Sci Technol. Jan.-Feb. 2015;69(1):59-73. |
Gin et al., Chemistry. Designing the next generation of chemical separation membranes. Science. May 6, 2011;332(6030):674-6. |
Goldberg et al., Formulation development of therapeutic monoclonal antibodies using high-throughput fluorescence and static light scattering techniques: role of conformational and colloidal stability. J Pharm Sci. Apr. 2011;100(4):1306-15. |
He et al., Use of a nanoporous biodegradable miniature device to regulate cytokine release for cancer treatment. J Control Release. May 10, 2011;151(3):239-45. |
Heier et al., The 1-year Results of CLEAR-IT 2, a Phase 2 Study of Vascular Endothelial Growth Factor Trap-Eye Dosed As-needed After 12-week Fixed Dosing. Ophthalmology. 2011;118(6):1098-106. |
Heier, VEGF Trap-Eye for Exudative AMD. Retinal Physician. Apr. 2009;1-7. |
Heller et al., Poly(ortho esters)—from concept to reality. Biomacromolecules. Sep.-Oct. 2004;5(5):1625-32. |
Heller et al., Poly(ortho esters): synthesis, characterization, properties and uses. Adv Drug Deliv Rev. Oct. 16, 2002;54(7):1015-39. |
Heller, Ocular delivery using poly(ortho esters), Adv Drug Deliv Rev. Dec. 13, 2005;57(14):2053-62. |
Herrmann et al., New insight into the role of polyethylene glycol acting as protein release modifier in lipidic implants. Pharm Res. Aug. 2007;24(8):1527-37. |
Hoe et al., Manufacturing and device options for the delivery of biotherapeutics. J Aerosol Med Pulm Drug Deliv. Oct. 2014;27(5):315-28. |
Holash et al., VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc Natl Acad Sci U S A. Aug. 20, 2002;99(17):11393-8. |
Huang, Receptor-Fc fusion therapeutics, traps, and MIMETIBODY technology. Curr Opin Biotechnol. Dec. 2009;20(6):692-9. |
Ishihara et al., Glucose Induced Permeation Control of Insulin through a Complex Membrane Consisting of Immobilized Glucose Oxidase and a Poly(amine). Polymer J. 1984;16(8):625-31. |
Jain et al., Controlled drug delivery by biodegradable poly(ester) devices: different preparative approaches. Drug Dev Ind Pharm. Aug. 1998;24(8):703-27. |
Jakubke et al., Amino acids, peptides, proteins. M: Mir. 1985:92-4. |
Jezek et al., Viscosity of concentrated therapeutic protein compositions. Adv Drug Deliv Rev. Oct. 2011;63(13):1107-17. |
Johnson et al., A month-long effect from a single injection of microencapsulated human growth hormone. Nat Med. Jul. 1996;2(7):795-9. |
Joubert et al., Classification and characterization of therapeutic antibody aggregates. J Biol Chem. Jul. 15, 2011;286(28):25118-33. |
Kim et al., BSA-FITC-loaded microcapsules for in vivo delivery. Biomaterials. Feb. 2009;30(5):902-9. |
Kono et al., Adding to the Mix: Fibroblast Growth Ffactor and Platelet-Derived Growth Factor Receptor Pathways as Targets in Non-Small Cell Lung Cancer. Curr Cancer Drug Targets. Feb. 2012;12(2):107-23. |
Kost et al., Glucose-sensitive membranes containing glucose oxidase: activity, swelling, and permeability studies. J Biomed Mater Res. Nov.-Dec. 1985;19(9):1117-33. |
Labet et al., Synthesis of polycaprolactone: a review. Chem Soc Rev. Dec. 2009;38(12):3484-504. |
Lance et al., In vivo and in vitro sustained release of ranibizumab from a nanoporous thin-film device. Drug Deliv Transl Res. Dec. 2016;6(6):771-780. |
Lechuga-Ballesteros et al., Trileucine improves aerosol performance and stability of spray-dried powders for inhalation. J Pharm Sci. Jan. 2008;97(1):287-302. |
Lee et al., In vivo characterization of sustained-release formulations of human growth hormone. J Pharmacol Exp Ther. Jun. 1997;281(3):1431-9. |
Lee et al., Nano spray drying: A novel method for preparing protein nanoparticles for protein therapy. Int J Pharm. Jan. 17, 2011;403(1-2):192-200. |
Li et al., Preparation and characterization of chitosan nanopores membranes for the transport of drugs. Int J Pharm. Nov. 28, 2011;420(2):371-7. |
Mahadevan et al., Statistical-mechanical model of protein precipitation by nonionic polymer. AlChE Journal. 1990;36(10):1517-28. |
Mahadevan et al., Theory of precipitation of protein mixtures by nonionic polymer. AIChE Journal. 1992;38(4):573-91. |
Martinac et al., Spray-dried chitosan/ethylcellulose microspheres for nasal drug delivery: swelling study and evaluation of in vitro drug release properties. J Microencapsul. Aug. 2005;22(5):549-61. |
Mercier et al., Organic polymers and ceramics. Introduction to Materials Science, Chapter 5. 2002;89-119. |
Mordenti et al., Intraocular pharmacokinetics and safety of a humanized monoclonal antibody in rabbits after intravitreal administration of a solution or a PLGA microsphere formulation. Toxicol Sci. Nov. 1999;52(1):101-6. |
Morita et al., Formation and isolation of spherical fine protein microparticles through lyophilization of protein-poly(ethylene glycol) aqueous mixture. Pharm Res. Nov. 2000;17(11):1367-73. |
Mowen et al., Unconventional post-translational modifications in immunological signaling. Nat Immunol. Jun. 2014;15(6):512-20. |
Nguyen et al., Protein powders for encapsulation: a comparison of spray-freeze drying and spray drying of darbepoetin alfa. Pharm Res. 2004;21:507-14. |
Odaka et al., Ligand-binding enhances the affinity of dimerization of the extracellular domain of the epidermal growth factor receptor1. J Biochem. 1997;122:116-121. (Abstract Only). |
Oka et al., Forming disulfides in the endoplasmic reticulum. Biochim Biophys Acta. Nov. 2013;1833(11):2425-9. |
Pan et al., Comparison of long-acting bevacizumab formulations in the treatment of choroidal neovascularization in a rat model. J Ocul Pharmacol Ther. Jun. 2011;27(3):219-24. |
Papadopoulos et al., Binding and neutralization of vascular endothelial growth factor (VEGF) and related ligands by VEGF Trap, ranibizumab and bevacizumab. Angiogenesis. Jun. 2012;15(2):171-85. |
Philo, Is any measurement method optimal for all aggregate sizes and types? AAPS J. Sep. 8, 2006;8(3):E564-71. |
Plomp et al., Recent advances in clinical glycoproteomics of immunoglobulins (lgs). Mol Cell Proteomics. 2016;15:2217-28. (Abstract Only). |
Polson et al., The franctionation of protein mixtures by linear polymers of high molecular weight. Biochim Biophys Acta. 1964;82:463-75. |
Regeneron, Eyelea Prescribing information. Regeneron Pharmaceuticals, Inc. pp. 1-15. (2011). |
Ruth et al., alpha-L-iduronidase forms semi-crystalline spherulites with amyloid-like properties. Acta Crystallogr D Biol Crystallogr. Apr. 2000;56(Pt 4):524-8. |
Safety data sheet (SDS) Methylene Chloride, Sankyo Chemical Co., Ltd. 2017, 22 pages. |
Savage et al., Determination of adequate moisture content for efficient dry-heat viral inactivation in lyophilized factor VIII by loss on drying and by near infrared spectroscopy. Biologicals. Jun. 1998;26(2):119-24. (Abstract only). |
Schlesinger et al., A Tunable, Biodegradable, Thin-Film Polymer Device as a Long-Acting Implant Delivering Tenofovir Alafenamide Fumarate for HIV Pre-exposure Prophylaxis. Pharm Res. Jul. 2016;33(7):1649-56. |
Schule et al., Conformational analysis of protein secondary structure during spray-drying of antibody/mannitol formulations. Eur J Pharm Biopharm. 2007;65:1-9. |
Schule et al., Stabilization of IgG1 in spray-dried powders for inhalation. Eur J Pharm Biopharm. 2008;69:793-807. |
Sharma et al., Polyethylene glycol-induced precipitation of interferon Alpha-2a followed by vacuum drying: development of a novel process for obtaining a dry, stable powder. AAPS Pharm Sci. Jan. 26, 2004;6(1):E4. |
Shire et al., Challenges in the development of high protein concentration formulations. J Pharm Sci. 2004;93:1390-402. |
Sim et al., Branched polyethylene glycol for protein precipitation. Biotechnol Bioeng. Mar. 2012;109(3):736-46. |
Sinha et al., Comparison of LC and LC/MS Methods for Quantifying N-Glycosylation in Recombinant IgGs. J Am Soc Mass Apectrum. 2008;19:1643-54. |
Sinha et al., Poly-epsilon-caprolactone microspheres and nanospheres: an overview. Int J Pharm. Jun. 18, 2004;278(1):1-23. |
Stock et al., Platelet-derived growth factor receptor-a: a novel therapeutic target in human hepatocellular cancer. Mol Cancer Ther. 2007;6:1932-41. |
Sutyagin et al., Chemistry and Physics of Polymers: schoolbook. A.V. Publishing House TPU. 2003, p. 208. |
Thurston et al., The complex role of angiopoietin-2 in the angiopoietin-tie signaling pathway. Cold Spring Harb Perspect Med. Sep. 1, 2012;2(9):a006550. |
Towns, Moisture content in proteins: its effects and measurement. Chromatography A. 1995;705:115-27. (Abstract Only). |
Treuheit et al., Inverse relationship of protein concentration and aggregation. Pharm Res. Apr. 2002;19(4):511-6. |
Vehring, Pharmaceutical engineering via spray drying. Pharm Res. 2007;25:999-1022. |
Vroman et al., Biodegradable Polymers. Materials 2009;2:307-44. |
Wirtz et al., Transport properties of template synthesized gold and carbon nanotube membranes. Int J Nanosci. 2002;1(3,4):255. |
Wu et al., A systems biology perspective on sVEGFR1: its biological function, pathogenic role and therapeutic use. J Cell Mol Med. Mar. 2010;14(3):528-52. |
Wyati, Light scattering and the absolute characterization of macromolecules. Anal Chim Acta. 1993;272:1-40. |
Yadav et al., Tumour Angiogenesis and Angiogenic Inhibitors: A Review. J Clin Diagn Res. Jun. 2015;9(6):XE01-XE05. |
Yang et al., Reservoir-based polymer drug delivery systems. J Lab Autom. Feb. 2012;17(1):50-8. |
Ye et al., Issues in long-term protein delivery using biodegradable microparticles. J Control Release. Sep. 1, 2010;146(2):241-60. |
Yeo et al., A new microencapsulation method using an ultrasonic atomizer based on interfacial solvent exchange. J Control Release. Dec. 10, 2004;100(3):379-88. |
Zachary, VEGF signalling: integration and multi-tasking in endothelial cell biology. Biochem Soc Trans. Dec. 2003;31(Pt 6):1171-7. |
Brazilian Search Report and Written Opinion for Application No. 112018010743, dated Aug. 10, 2020, 6 pages. |
Canadian Office Action dated Mar. 22, 2019, 5 pages. |
Canadian Office Action for Application No. 3076725, dated Apr. 9, 2021, 4 pages. |
Chilean Office Action for Application No. 2018001623, dated Aug. 18, 2020, 34 pages. |
Chilean Office Action for corresponding Chilean application, dated Feb. 7, 2020, 3 pages. |
Chinese Notification to Grant for Application No. 201280056324, dated Nov. 5, 2015, 3 pages. |
Chinese Office Action for Application No. 201280056324, dated Jun. 2, 2015, 5 pages. |
Chinese Office Action for Application No. 201610035281, dated Apr. 4, 2019, 11 pages. |
Chinese Office Action for Application No. 201610035281, dated Jul. 4, 2018, 13 pages. |
Chinese Office Action for Application No. 201680067420, dated Jun. 28, 2020, 20 pages. |
Chinese Office Action for Application No. 201680067420, dated Nov. 18, 2020, 13 pages. |
Chinese Office Action for Application No. 201680072951, dated Jun. 22, 2020, 18 pages. |
Chinese Office Action for Application No. 201680072951, dated Mar. 26, 2021, 7 pages. |
Chinese Office Action for Application No. 201680072951, dated Sep. 11, 2020, 8 pages. |
Chinese Search Report for Application No. 201280056324, dated May 25, 2015, 2 pages. |
Chinese Search Report for Application No. 201610035281, dated Jun. 26, 2018, 2 pages. |
Chinese Search Report for Application No. 201680067420, dated Jun. 19, 2020, 1 page. |
Chinese Search Report for Application No. 201680072951, dated Jun. 16, 2020, 2 pages. |
Colombian Office Action for Application No. NC20180005920, dated May 2, 2020, 5 pages. |
European Decision to Grant for Application No. 12795967, dated Jan. 8, 2020, 2 pages. |
European Decision to Grant for Application No. 18173299, dated May 4, 2020, 2 pages. |
European Notice of Allowance for Application No. 19180351, dated Mar. 5, 2021, 6 pages. |
European Notice of Opposition for Application No. 12795967, dated Oct. 21, 2020, 19 pages. |
European Office Action dated Jan. 10, 2018, 4 pages. |
European Office Action dated Nov. 28, 2018. |
European Office Action for Application No. 12795967, dated Apr. 12, 2019, 3 pages. |
European Office Action for Application No. 12795967, dated Aug. 1, 2018, 4 pages. |
European Office Action for Application No. 12795967, dated Jan. 10, 2018, 4 pages. |
European Office Action for Application No. 12795967, dated Nov. 28, 2018, 4 pages. |
European Office Action for Application No. 12795967, dated Nov. 3, 2020, 1 page. |
European Office Action for Application No. 12795967, dated Sep. 23, 2019, 6 pages. |
European Office Action for Application No. 16788338, dated Sep. 29, 2020, 5 pages. |
European Office Action for Application No. 16799339.8, dated Sep. 30, 2020, 5 pages. |
European Office Action for Application No. 16820541, dated Feb. 4, 2021, 4 pages. |
European Office Action for Application No. 16820541, dated Jun. 16, 2020, 6 pages. |
European Office Action for Application No. 16820541, dated Nov. 6, 2019, 6 pages. |
European Office Action for Application No. 18173299, dated Feb. 27, 2020, 6 pages. |
European Office Action for Application No. 18173299, dated Jul. 11, 2019, 5 pages. |
European Office Action for Application No. 19180351, dated Sep. 25, 2020, 6 pages. |
European Search Report and Search Opinion for Application No. 18173299, dated Aug. 8, 2018, 8 pages. |
European Search Report and Search Opinion for Application No. 19180351, dated Aug. 21, 2019, 8 pages. |
Indian Office Action for Application No. 201817015063, dated Jul. 8, 2020, 7 pages. |
International Preliminary Report on Patentability for Application No. PCT/US2012/065735, dated May 30, 2014, 9 pages. |
International Preliminary Report on Patentability for Application No. PCT/US2016/057019, dated Apr. 26, 2018, 8 pages. |
International Preliminary Report on Patentability for Application No. PCT/US2016/067280, dated Jun. 28, 2018, 9 pages. |
International Search Report and Written Opinion dated Jun. 18, 2018, 10 pages. |
International Search Report and Written Opinion for Application No. PCT/US2012/065735, dated Feb. 15, 2013, 11 pages. |
International Search Report and Written Opinion for Application No. PCT/US2016/067280, dated Mar. 10, 2017. |
Israeli Office Action for the corresponding Israel application, dated Jun. 28, 2020, 4 pages. |
Japanese Decision to Grant for Application No. 2014-542536, dated Dec. 4, 2017, 5 pages. |
Japanese Decision to Grant for Application No. 2017-149732, dated Jun. 13, 2019, 6 pages. |
Japanese Decision to Grant for Application No. 2019-079952, dated Jun. 3, 2020, 6 pages. |
Japanese Office Action dated May 9, 2018, 11 pages. |
Japanese Office Action for Application No. 2014-542536, dated Aug. 29, 2016, 12 pages. |
Japanese Office Action for Application No. 2014-542536, dated Mar. 28, 2017, 12 pages. |
Japanese Office Action for Application No. 2014-542536, dated Nov. 14, 2017, 6 pages. |
Japanese Office Action for Application No. 2017-149732, Apr. 20, 2018, 22 pages. |
Japanese Office Action for Application No. 2017-149732, dated Dec. 26, 2018, 24 pages. |
Japanese Office Action for Application No. 2018-519460 dated Jun. 22, 2020, 2 pages. |
Japanese Office Action for Application No. 2018-531166, dated Oct. 12, 2020, 13 pages. |
Japanese Search Report for Application No. 2018-519460, dated Apr. 20, 2020, 48 pages. |
Korean Grant of Patent for Application 10-2014-7014549, dated May 28, 2020, 2 pages. |
Korean Office Action for Application No. 10-2014-7014549, dated Dec. 19, 2019, 6 pages. |
Korean Office Action for Application No. 10-2014-7014549, dated Jun. 12, 2019, 11 pages. |
Korean Office Action for Application No. 10-2020-7003704, dated May 6, 2020, 6 pages. |
Korean Office Action for Application No. 10-2020-7003704, dated Nov. 20, 2020, 4 pages. |
Korean Office Action for the corresponding Korean application, dated Dec. 19, 2019, 4 pages. |
Mexican Office Action dated Nov. 1, 2017, 12 pages. |
Mexican Office Action for Application No. 2018004695, dated Mar. 2, 2021, 7 pages. |
Non-Final Office Action received for U.S. Appl. No. 15/766,586, dated Mar. 9, 2021, 19 pages. |
Notice of Grounds for Rejection issued by the Korean Patent Office dated May 6, 2020 for the corresponding Korean application, 4 pages. |
Report of opposition to European Patent No. 2790681, dated Nov. 10, 2020, 5 pages. |
Russian Decision to Grant for Application No. 2014124682, dated Nov. 17, 2017, 13 pages. |
Russian Office Action for Application No. 2018101248, dated Mar. 2, 2021, 16 pages. |
Russian Search Report for Application No. 2018101248, dated Mar. 2, 2021, 4 pages. |
Singapore Written Opinion dated Sep. 11, 2019, 9 pages. |
Singapore Written Opinion for Application No. 11201402152Y, dated Nov. 13, 2017, 7 pages. |
Singapore Written Opinion for Application No. 11201802867U, dated Jul. 17, 2020, 6 pages. |
Singapore Written Opinion for Application No. 11201802867U, dated Mar. 28, 2019, 6 pages. |
U.S. Advisory Action (PTOL-303) for U.S. Appl. No. 13/680,069, dated Mar. 9, 2016. |
U.S. Final Office Action for U.S. Appl. No. 15/766,586, dated Nov. 18, 2020, 23 pages. |
U.S. Final Office Action for U.S. Appl. No. 15/775,479, dated Nov. 16, 2020, 20 pages. |
U.S. Final Rejection for U.S. Appl. No. 13/680,069, dated Jun. 20, 2014. |
U.S. Final Rejection for U.S. Appl. No. 13/680,069, dated Nov. 6, 2015. |
U.S. Non-Final Office Action for U.S. Appl. No. 15/766,586, dated Mar. 9, 2020, 19 pages. |
U.S. Non-Final Office Action for U.S. Appl. No. 15/766,586, dated May 21, 2020, 19 pages. |
U.S. Non-Final Rejection for U.S. Appl. No. 13/680,069, dated Dec. 17, 2013. |
U.S. Non-Final Rejection for U.S. Appl. No. 13/680,069, dated Dec. 5, 2016. |
U.S. Non-Final Rejection for U.S. Appl. No. 13/680,069, dated Mar. 31, 2015. |
U.S. Non-Final Rejection for U.S. Appl. No. 15/775,479, dated May 11, 2020. |
US Patent Assignment No. 61561525. |
U.S. Requirement for Restriction/Election for U.S. Appl. No. 13/680,069, dated Aug. 21, 2013. |
U.S. Requirement for Restriction/Election for U.S. Appl. No. 15/775,479, dated Mar. 5, 2020. |
Number | Date | Country | |
---|---|---|---|
20220249387 A1 | Aug 2022 | US |
Number | Date | Country | |
---|---|---|---|
61561525 | Nov 2011 | US |
Number | Date | Country | |
---|---|---|---|
Parent | 13680069 | Nov 2012 | US |
Child | 15450335 | US |
Number | Date | Country | |
---|---|---|---|
Parent | 15450335 | Mar 2017 | US |
Child | 17677282 | US |