CA2255564C - Aerodynamically light particles for pulmonary drug delivery - Google Patents

Aerodynamically light particles for pulmonary drug delivery Download PDF

Info

Publication number
CA2255564C
CA2255564C CA002255564A CA2255564A CA2255564C CA 2255564 C CA2255564 C CA 2255564C CA 002255564 A CA002255564 A CA 002255564A CA 2255564 A CA2255564 A CA 2255564A CA 2255564 C CA2255564 C CA 2255564C
Authority
CA
Canada
Prior art keywords
particles
amino acid
acid
poly
lysine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA002255564A
Other languages
French (fr)
Other versions
CA2255564A1 (en
Inventor
David A. Edwards
Giovanni Caponetti
Jeffrey S. Hrkach
Noah Lotan
Justin Hanes
Abdell Aziz Ben-Jebria
Robert S. Langer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Penn State Research Foundation
Massachusetts Institute of Technology
Original Assignee
Penn State Research Foundation
Massachusetts Institute of Technology
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27096992&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2255564(C) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Penn State Research Foundation, Massachusetts Institute of Technology filed Critical Penn State Research Foundation
Publication of CA2255564A1 publication Critical patent/CA2255564A1/en
Application granted granted Critical
Publication of CA2255564C publication Critical patent/CA2255564C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0075Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a dry powder inhaler [DPI], e.g. comprising micronized drug mixed with lactose carrier particles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1641Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poloxamers
    • A61K9/1647Polyesters, e.g. poly(lactide-co-glycolide)

Abstract

Improved aerodynamically light particles for delivery to the pulmonary syste m, and methods for their preparation and administration are provided. In a preferred embodiment, the aerodynamically light particles are made of a biodegradable material and have a tap density less than 0.4 g/cm3 and a mass mean diameter between 5 .mu.m and 30 .mu.m. T he particles may be formed of biodegradable materials such as biodegradable polymers. For example, the particles may be formed of a functionalized polyester graft copolymer consisting of a linear .alpha.-hydroxy-acid polyester backbone having at least one amino acid group incorporated therein and at least one poly(amino acid) side chain extending from an amino acid group in the polyester backbone. In one embodiment, aerodynamically light particles having a large mean diameter for example greater than 5 .mu.m, can be used for enhanced delivery of a therapeutic or diagnostic agent to the alveolar region of the lung. The aerodynamically light particles optionally can incorporate a therapeutic or diagnostic agent, and may be effectively aerosolized for administration to the respiratory tract to permit systemic or local delivery of wide variety of incrorporated agents.

Description

AERODYNAMICALLY LIGHT PARTICLES FOR
PULMONARY DRUG DELIVERY
Background of the Invention The present application relates generally to biodegradable particles of low density and large size fox delivery to the pulmonary system.
Biodegradable particles have been developed for the controlled-release and delivery of protein and peptide drugs. Langer, R., Scierzce>
249: 1527-1533 (1990). Examples include the use of biodegradable particles for gene therapy (Mulligan, R. C. Science, 260: 926-932 ( 1993)) and for 'single-shot' immunization by vaccine delivery (Eldridge et al., Mol. Ifzzfzzunol., 28: 287-294 (1991)).
Aerosols for the delivery of therapeutic agents to the respiratory tract have been developed. Adjei, A. and Garren, J. Phan~t. Res. 7, 565-569 (1990); and Zanen, P. and Lamm, J.-W.J. Int. J. Pharnz. 114, 111-115 (1995). The respiratory tract encompasses the upper airways, including the oropharynx and larynx, followed by the lower airways, which include the trachea followed by bifurcations into the bronchi and bronchioli. The upper and lower airways are called the conducting airways. The terminal bronchioli then divide into respiratory bronchioli which then Lead to the ultimate respiratory zone, the alveoli, or deep lung.
Gouda, I. "Aerosols for delivery of therapeutic and diagnostic agents to the respiratory tract," in Critical Reviews in Therapeutic Drug Carrier Systems 6:273-313, 1990. The deep lung, or alveoli, are the nrimarv target of inhaled therapeutic aerosols for systemic drug delivery.
Inhaled aerosols have been used for the treatment of local lung disorders including asthma and cystic fibrosis (Anderson et al., Alll. Rev.
Respir. Dis., 140: 1317-1324 (1989)) and have potential for the systemic delivery of peptides and proteins as well (Patton and Platz, Advanced Drug Delivery Revieit~s, 8:179-196 (1992)). However, pulmonary drug delivery strategies present many difficulties for the delivery of _2_ macromolecules; these include protein denaturation during aerosolization, excessive loss of inhaled drug in the oropharyngeal cavity {often exceeding 80%), poor control over the site of deposition, irreproducibility of therapeutic results owing to variations in breathing patterns, the often too-rapid absorption of drug potentially resulting in local toxic effects, ' and phagocytosis by lung macrophages.
Considerable attention has been devoted to the design of therapeutic aerosol inhalers to improve the efficiency of inhalation therapies. Timsina et. al. , Int. J. Pharnz. 101, 1-13 (1995); and Tansey, I.P., Sprm~ Technol. Market 4, 26-29 (1994). Attention has also been given to the design of dry powder aerosol surface texture, regarding particularly the need to avoid particle aggregation, a phenomenon which considerably diminishes the efficiency of inhalation therapies. French, D. L. , Edwards, D. A. and Niven, R. W . , J. Aerosol Sci. 27, 769-783 (1996). Attention has not been given to the possibility of using large particle size (greater than5 hem) as a means to improve aerosolization efficiency, despite the fact that intraparticle adhesion diminishes with increasing particle size. French, D.L., Edwards, D.A. and Niven, R.W.
J. Aerosol Sci. 27, 769-783 (1996). - This is because particles°of standard mass density (mass density near 1 g/cm3) and mean diameters greater than 5 lcm are known to deposit excessively in the upper airways or in the inhaler device. Heyder, J. et al. , J. Aerosol Sci. , 17: 811-825 ( 1986).
For this reason, dry powder aerosols for inhalation therapy are generally produced with mean diameters primarily in the range of less than5 ~cm.
Ganderton. D., J. Bioplzarrnaceutical Sciences 3:101-105 (1992): and Gonda, I. "Physico-Chemical Principles in Aerosol Delivery," in Topics in Pharmaceutical Sciences 1991, Crommelin, D.J. and K.K. Midha, Eds., Medphartn Scientific Publishers, Stuttgart, pp. 95-115, 1992.
Large "carrier" particles (containing no drug) have been co-delivered with therapeutic aerosols to aid in achieving efficient aerosolization among other possible benefits. French, D.L., Edwards, D.A. and Niven, R.W.
J. Aerosol Sci. 27, 769-783 (1996).

Local and systemic inhalation therapies can often benefit from a relatively slow controlled release of the therapeutic agent. Gonda, L, "Physico-chemical principles in aerosol delivery," in: Topics in Pharmaceutical Sciences 1991, D.J.A. Crommelin and K.K. Midha, ' S Eds., Stuttgart: Medpharm Scientific Publishers, pp. 95-1I7, (I992).
Slow release from a therapeutic aerosol can prolong the residence of an administered drug in the airways or acini, and diminish the rate of drug appearance in the bloodstream. Also, patient compliance is increased by reducing the frequency of dosing. Langer, R., Science, 249:1527-1533 (1990); and Gonda, I. "Aerosols for delivery of therapeutic and diagnostic agents to the respiratory tract," in Critical Reviews in Therapeutic Drug Carrier Systems 6:273-313, (1990).
The human lungs can remove or rapidly degrade hydrolytically cleavable deposited aerosols over periods ranging from minutes to hours.
In the upper airways, ciliated epithelia contribute to the "mucociliary escalator" by which particles are swept from the airways toward the mouth. Pavia, D. "Lung Mucociliary Clearance," in Aerosols and the Lung: Clinical and Experimental Aspects, Clarke, S.W. and Pavia, D., Eds. , Butterworths, London, 1984. Anderson et al. , Alfa. Rev. Respir.
Dis. , 140: 1317-1324 (1989). In the deep lungs, alveolar macrophages are capable of phagocytosing particles soon after their deposition.
Warheit, M.B. and Hartsky, M.A., Microscopy Res. Tech. 26: 412-422 (1993); Brain, J.D., "Physiology and Pathophysiology of Pulmonary Macrophages," in The Reticuloendotlzelial System, S.M. Reichard and J.
Filkins, Eds., Plenum, New York, pp. 315-327, 1985; Dorries. A.M. and Valberg, P.A., AnZ. Rev. Resp. Disease 146, 831-837 (1991); and Gehr, P. et al. Microscopy Res. afad TeclZ., 26, 423-436 (1993). As the diameter of particles exceeds 3 p,m, there is increasingly less phagocytosis . by macrophages. Kawaguchi, H. et al. , Biomaterials 7: 61-66 ( 1986);
Krenis, L.J. and Strauss, B., Proc. Soc. Exp. Med., 107:748-750 (1961);
and Rudt, S. and Muller, R.H., J. Contr. Rel., 22: 263-272 (1992).
However, increasing the particle size also minimizes the probability of particles (possessing standard mass density) entering the airways and acini due to excessive deposition in the oropharyngeal or nasal regions.
Heyder, J. et al. , J. Aerosol Sci. , 17: 811-825 (1986). An effective dry- .
powder inhalation therapy for both short and long term release of therapeutics, either for local or systemic delivery, requires a powder that ' displays minimum aggregation and is capable of avoiding or suspending the lung's natural clearance mechanisms until drugs have been effectively delivered.
There is a need for improved inhaled aerosols for pulmonary delivery of therapeutic agents which axe capable of delivering the drug in an effective amount into the airways or the alveolar zone of the lung.
There further is a need fox the development of drug carriers for use as inhaled aerosols which are biodegradable and are capable of controlled release of drug within the airways or in the alveolar zone of the lung.
IS It is therefore an object of the present invention to provide improved carriers for the pulmonary delivery of therapeutic and diagnostic agents. It is a further object of the invention to provide inhaled aerosols which are effective carriers for delivery of therapeutic or diagnostic agents to the deep lung. It is another object of the invention to provide carriers for pulmonary delivery which avoid phagocytosis in the deep lung. It is a further object of the invention to provide carriers for pulmonary delivery which are capable of biodegrading and optionally releasing incorporated agents at a controlled rate.
Summary of the Invention Improved aerodynamically light particles for delivery to the pulmonary system, and methods for their preparation and administration are provided. In a preferred embodiment, the particles are made of a .
biodegradable material, have a tap density less than 0.4 g/cm3 and a mean diameter between 5 ~sm and 30 ~.rn. In one embodiment, for example, at least 90 % of the particles have a mean diameter between 5 ~cm and 30 ~cm. The particles may be formed of biodegradable materials such as biodegradable synthetic polymers, proteins, or other water-soluble v materials such as certain polysaccharides. For example, the particles may be formed of a functionalized polyester graft copolymer with a linear a-S hydroxy-acid polyester backbone with at least one amino acid residue incorporated per molecule therein and at least one poly(amino acid) side chain extending from an amino acid group in the polyester backbone.
Other examples include particles formed of water-soluble excipients, such as trehalose or lactose, or proteins, such as lysozyme or insulin. The particles can be used for delivery of a therapeutic or diagnostic agent to the airways or the alveolar region of the lung. The particles may be effectively aerosolized for administration to the respiratory tract and can be used to systemically or locally deliver a wide variety of incorporated agents. The particles incorporating an agent can optionally be co-delivered with larger carrier particles, not carrying an incorporated agent, which have, for example, a mean diameter ranging between about 50 hem and 100 ~,rn.
Brief Description of the Drawings Figure 1 is a graph comparing total particle mass of aerodynamically light and non-light, control particles deposited on the nonrespirable and respirable stages of a cascade impactor following aerosolization.
Figure 2 is a graph comparing total particle mass deposited in the trachea and after the carina (lungs) in rat lungs and upper airways following intratracheal aerosolization during forced ventilation of aerodynamically light poly(lactic acid-co-lysine-graft-lysine) (PLAL-Lys) particles and control, non-light poly(L-lactic acid) (PLA) particles.
Figure 3 is a graph comparing total particle recovery of aerodynamically light PLAL-Lys particles and control PLA particles in rat lungs following bronchoalveolar lavage.

-s-Figure 4 is a graph representing serum insulin levels (ng/ml) over time (hours) following administration via inhalation or subcutaneous injection of porous PLGA particles.
Figure 5 is a graph representing serum insulin levels (ng/ml) over time (hours) following administration via inhalation or subcutaneous °
injection of non-porous PLGA particles. Darkened circles represent administration via inhalation. Darkened triangles represent administration via subcutaneous injection. Empty diamonds represent untreated controls.
Figure 6 is a graph representing serum glucose concentrations (mg/dl) following administration of porous PLGA particles via inhalation.
Darkened circles represent administration via inhalation. Darkened triangles represent untreated controls.
Figure 7 is a graph representing serum testosterone levels (ng/ml) over time (hours} following administration via inhalation or subcutaneous I5 injection of porous PLGA particles with a diameter of 20.4 Vim.
Darkened circles represent administration via inhalation. Darkened triangles represent administration via subcutaneous injection.
Figure 8 is a graph representing serum testosterone levels (ng/ml) over time (hours) following administration via inhalation or subcutaneous injection of porous PLGA particles with a diameter of 10.1 hem.
Darkened circles represent administration via inhalation. Darkened triangles represent administration via subcutaneous injection.
Detailed Description of the Invention Aerodynamically light, biodegradable particles for improved delivery to the respiratory tract are provided. The particles can incorporate a therapeutic or diagnostic agent, and can be used for controlled systemic or local delivery of the agent to the respiratory tract via aerosolization. In a preferred embodiment, the particles have a tap density less than about 0.4 g/cm3. Features of the particle which can contribute to low tap density include irregular surface texture and porous structure. Administration of the low density particles to the lung by aerosolization permits deep lung delivery of relatively large diameter therapeutic aerosols, for example, greater than S ,um in mean diameter.
A rough surface texture also can reduce particle agglomeration and ° S provide a highly flowabIe powder, which is ideal for aerosolization via dry powder inhaler devices, leading to lower deposition in the mouth, throat and inhaler device.
Density and Size of Aerodynamically Light Particles Particle Size The mass mean diameter of the particles can be measured using a Coulter Counter. The aerodynamically light particles are preferably at least about S microns in diameter. The diameter of particles in a sample will range depending upon depending on factors such as particle composition and methods of synthesis. The distribution of size of particles in a sample can be selected to permit optimal deposition within targeted sites within the respiratory tract.
The particles may be fabricated or separated, for example, by filtration, to provide a particle sample with a preselected size distribution.
For example, greater than 30 % , SO % , 70 % , or 80 % of the particles in a sample can have a diameter within a selected range of at least S ~cm.
The selected range within which a certain percentage of the particles must fall may be, for example, between about S and 30 ~.m, or optionally between S and 15 ~cm. In one preferred embodiment, at least a portion of the particles have a diameter between about 9 and l I ~,m. Optionally, the 2S particle sample also can be fabricated wherein at least 90 % , or optionally 9S % or 99 % , have a diameter within the selected range. The presence of the higher proportion of the aerodynamically light, larger diameter (at least about S ~cm) particles in the particle sample enhances the delivery of therapeutic or diagnostic agents incorporated therein to the deep lung.
In one embodiment, in the particle sample, the interquartile range may be 2 ~.m, with a mean diameter for example of 7.5, 8.0, 8.5, 9.0, 9.5, 10.0, 10.5, I1.0, 11.5, 12.0, 12.5, 13.0 or 13.5 ~.m. Thus, for _g_ example, at least 30 % , 40 % , 50 % or 60 % of the particles may have diameters within the selected ranges of 5.5-7.5 Vim, 6.0-8.0 ,um, 6.5-8.5 ~cm, 7.0 - 9.0 p,m, 7.5 - 9.5 ~cm, 8.0 - 10.0 ~cm, 8.5-10.5 ~cm, 9.0-11.0 .
Vim, 9.5-1I.5 ~cm, 10.0-12.0 Vim, 10.5-12.S~,m, 11.0-13.0 p,m, 11.5-13.5 p.m, I2.0-14.0 ,um, 12.5-14.5 ~m or I3.0-15.0 ~,m. Preferably the ' above-listed percentages of particles have diameters within a 1 E.cm range, for example, 6.0-7.0 hem, 10.0-11.0 ,um or 13.0-14.0 ,um.
Particles having a tap density less than about 0.4 g/cm3 and a mean diameter of at least about 5 ~,m are more capable of escaping inertial and gravitational deposition in the oropharyngeal region than smaller or more dense particles, and are targeted to the airways of the deep lung. The use of larger particles (mean diameter greater than 5 ~.m) is advantageous since they are able to aerosolize more efficiently than smaller, denser particles such as those currently used for inhalation I5 therapies.
Tn comparison to smaller, denser particles, the larger (greater than 5 ~,m) aerodynamically light particles also can potentially more successfully avoid phagocytic engulfment by alveolar macrophages and clearance from the lungs, due to size exclusion of tile particles from the phagocytes' cytosolic space. For particles of statistically isotropic shape (on average, particles of the powder possess no distinguishable orientation), such as spheres with rough surfaces, the particle envelope volume is approximately equivalent to the volume of cytosolic space required within a macrophage for complete particle phagocytosis.
Aerodynamically light particles thus are capable of a longer term release of an incorporated diagnostic or therapeutic agent than smaller, denser particles. Following inhalation, aerodynamically light biodegradable particles can deposit in the lungs (due to their relatively low tap density), and subsequently undergo slow degradation and drug release, without the majority of the particles being phagocytosed by alveolar macrophages. The agent can be delivered relatively slowly into the alveolar fluid, and at a controlled rate into the blood stream, minimizing possible toxic responses of exposed cells to an excessively high concentration of the agent. The aerodynamically light particles thus are highly suitable for inhalation therapies, particularly in controlled release applications.
- 5 The particles may be fabricated with the appropriate material, surface roughness, diameter and tap density for localized delivery to selected regions of the respiratory tract such as the deep lung or upper airways. For example, higher density or larger particles may be used for upper airway delivery, or a mixture of different sized particles in a sample, provided with the same or different incorporated agent may be administered to target different regions of the lung in one administration.
Particle Density and Deposition The particles have a diameter of at least about 5 ~cm and optionally incorporate a therapeutic or diagnostic agent. The particles are preferably aerodynamically light. As used herein, the phrase "aerodynamically light particles" refers to particles having a tap density less than about 0.4 g/cm3. The tap density of particles of a dry powder may be obtained using a GeoPyc'"' (Micrometrics Instrument Corp., Norcross, GA 30093).
Tap density is a standard measure of the envelope mass density. The envelope mass density of an isotropic particle is defined as the mass of the particle divided by the minimum sphere envelope volume within which it can be enclosed.
Inertial impaction and gravitational settling of aerosols are predominant deposition mechanisms in the airways and acini of the lungs during normal breathing conditions. Edwards, D.A., J. Aerosol Sci.
26:293-317 (1995). The importance of both deposition mechanisms increases in proportion to the »zass of aerosols and not to particle (or envelope) volume. Since the site of aerosol deposition in the Lungs is . determined by the mass of the aerosol (at least for particles of mean aerodynamic diameter greater than approximately I tcm), diminishing the tap density by increasing particle surface irregularities and particle porosity permits the delivery of larger particle envelope volumes into the lungs, all other physical parameters being equal.
The low tap density particles have a small aerodynamic diameter in comparison to the actual envelope sphere diameter. The aerodynamic diameter, daer, is related to the envelope sphere diameter, d (Gonda, I. , ' "Physico-chemical principles in aerosol delivery, " in Topics in Pharmaceutical Sciences 1991 (eds. D.J.A. CrommeIin and K.K.
Midha), pp. 95-II7, Stuttgart: Medpharm Scientific Publishers, 1992) by the formula:
daer = d'~p where the envelope mass p is in units of g/cm'. Maximal deposition of monodisperse aerosol particles in the alveolar region of the human lung (approximately 60%) occurs for an aerodynamic diameter of approximately daer=3 I~m. I-Ieyder, J. et al. , J. Aerosol Sci. , 17: 811-825 (1986). Due to their small envelope mass density, the actual diameter d of aerodynamically light particles comprising a monodisperse inhaled powder that will exhibit maximum deep-lung deposition is:
d = 3/~p ~,m (where p < 1 g/cm3);
where d is always greater than 3 ~cm. Fox example, aerodynamically light particles that display an envelope mass density, p = O.I g/cm3, will exhibit a maximum deposition for particles having envelope diameters -as large as 9.~ ~,m. The increased particle size diminishes interparticle adhesion forces. Visser, J., Powder Techfzology, 58:1-10. Thus, large particle size increases efficiency of aerosolization to the deep lung for particles of low envelope mass density, in addition to contributing to lower phaaocytic losses.
q WO 97!44013 PCTlUS97/08895 Particle Materials The aerodynamically light particles preferably are biodegradable . and biocompatible, and optionally are capable of biodegrading at a controlled rate for release of an incorporated thereapeutic or diagnostic agent. The particles can be made of any material which is capable of forming a particle having a tap density less than about 0.4 g/cm~. Both inorganic and organic materials can be used. Other non-polymeric materials (e. g. fatty acids) may be used which are capable of forming aerodynamically light particles as defined herein. Different properties of the particle can contribute to the aerodynamic lightness including the composition forming the particle, and the presence of irregular surface structure or pores or cavities within the particle.
Polymeric Particles The particles may be formed from any biocompatible, and preferably biodegradable polymer, copolymer, or blend, which is capable of forming particles having a tap density less than about 0.4 g/cm3.
Surface eroding polymers such as polyanhydrides may be used to form the aerodynamically light particles. For example, polyanhydrides such as poly[(p-carboxyphenoxy)-hexane anhydride] (PCPH) may be used.
Biodegradable polyanhydrides are described, for example, in U.S. Patent No. 4,857,311.
In another embodiment, bulk eroding polymers such as those based on polyesters, including poly(hydroxy acids), can be used. Preferred poly(hydroxy acids) are polyglycolic acid (PGA), polylactic acid (PLA) and copolymers and coblends thereof. In one embodiment, the polyester has incorporated therein a charged or functionalizable group such as an amino acid.
Other polymers include polyamides, polycarbonates, polyalkylenes such as polyethylene, polypropylene, polyethylene glycol), polyethylene oxide), poly{ethylene terephthalate), poly vinyl compounds such as polyvinyl alcohols, polyvinyl ethers, and polyvinyl esters, polymers of acrylic and methacrylic acids, celluloses and other polysaccharides, and peptides or proteins, or copolymers or blends thereof. Polymers may be selected with or modified to have the appropriate stability and degradation rates in vivo for different controlled drug delivery applications.
Polyester Graft Copolymers In one preferred embodiment, the aerodynamically light particles ' are formed from functionalized polyester graft copolymers, as described in I-Irkach et al., Macromolecules, 28:4736-4739 (1995}; and Hrkach et al. , "Poly(L-Lactic acid-co-amino acid) Graft Copolymers: A Class of Functional Biodegradable Biomaterials" in Hydrogels and Biodegradable Polymers for Bioapplications, ACS Symposium Series No. 627, Raphael M. Ottenbrite et al., Eds., American Chemical Society, Chapter 8, pp.
93-101, 1996. The functionalized graft copolymers are copolymers of polyesters, such as poly(glycolic acid) or poly(lactic acid), and another polymer including functionalizable or ionizable groups, such as a poly(amino acid). In a preferred embodiment, comb-like graft copolymers are used which include a linear polyester backbone having amino acids incorporated therein, and poly(amino acid) side chains which extend from the amino acid residues in the polyester backbone. The polyesters may be polymers of «-hydroxy acids such as lactic acid, glycolic acid, hydroxybutyric acid and hydroxyvaleric acid, or derivatives or combinations thereof. The polymers can include ionizable side chains, such as polylysine and polyaniline. Other ionizable groups, such as amino or carboxyl groups, may be incorporated into the polymer, covalently or noncovalently, to enhance surface roughness and porosity.
An exemplary polyester graft copolymer is poIy(lactic acid-co-lysine-graft-lysine) (PLAL-Lys), which has a polyester backbone consisting of poly(L-lactic acid-co- L-lysine) (PLAL), and grafted poly-lysine chains. PLAL-Lys is a comb-like graft copolymer having a backbone composition, for example, of 98 mot % lactic acid and 2 mol % .
lysine and poly(lysine) side chains extending from the lysine sites of the backbone.

PLAL-Lys may be synthesized as follows. First, the PLAL
copolymer consisting of L-lactic acid units and approximately 1-2 % N a carbobenzoxy-L-lysine (Z-L-lysine) units is synthesized as described in Barrera et al. , J. Arzz. Chem. Soc. , 1 I5:11010 (1993). Removal of the Z
S protecting groups of the randomly incorporated lysine groups in the polymer chain of PLAL yields the free e-amine which can undergo further chemical modification. The use of the poly(lactic acid) copolymer is advantageous since it biodegrades into lactic acid and lysine, which can be processed by the body. The existing backbone lysine groups are used as initiating sites for the growth of poIy(amino acid) side chains.
The lysine E-amino groups of linear poly(L-lactic acid-co-L-lysine) copolymers initiate the ring opening polymerization of an amino acid N-E
carboxyanhydride (NCA) to produce poly(L-lactic acid-co-amino acid) comb-like graft copolymers. In a preferred embodiment, NCAs are synthesized by reacting the appropriate amino acid with triphosgene.
Daly et al. , Tetrahedron Lett. , 29:5859 (1988). The advantage of using triphosgene over phosgene gas is that it is a solid material, and therefore, safer and easier to handle. It also is soluble in THF and hexane so any excess is efficiently separated from the NCAs.
The ring opening polymerization of amino acid N-carboxyanhydrides (NCAs) is initiated by nucleophilic initiators such as amines, alcohols, and water. The primary amine initiated ring opening polymerization of NCAs allows efficient control over the degree of polymerization when the monomer to initiator ratio (M/I) is Iess than 150.
Kricheldorf, H. R. in Models of Biopolymers by Ring-Opening Polymerization, Penczek, S., Ed., CRC Press, Boca Raton, 1990. Chapter 1; Kricheldorf, H. R. a Aminoacid-N Carboxy-Anhydrides and Related Heterocycles, Springer-Verlag, Berlin, 1987; and Imanishi, Y. in Ring-Opening Polytzterization, Ivin, K. 3. and Saegusa, T., Eds., Elsevier, London, 1984, Volume 2, Chapter 8. Methods for using lysine e-amino groups as polymeric initiators for NCA polymerizations are described in the art. Sela, M. et al. , J. Arzz. Chem. Soc.. , 78: 746 (1956).

In the reaction of an amino acid NCA with PLAL, the nucIeophilic primary e-amino group of the lysine side chain attacks C-5 of the NCA. This leads to ring opening to form an amide linkage, accompanied by evolution of a molecule of CO~. The amino group formed by the evolution of COZ propogates the polymerization by ' attacking subsequent NCA molecules. The degree of polymerization of the poly(amino acid) side chains, the amino acid content in the resulting graft copolymers and the physical and chemical characteristics of the resulting copolymers can be controlled by adjusting the ratio of NCA to lysine e-amino groups in the PLAL polymer, for example, by adjusting the length of the poly(amino acid) side chains and the total amino acid content .
The poly(amino acid) side chains grafted onto or incorporated into the polyester backbone can include any amino acid, such as aspartic acid, IS alanine or lysine, or mixtures thereof. The functional groups present in the amino acid side chains, which can be chemically modified, include amino, carboxylic acid, thiol, guanido, imidazole and hydroxyl groups.
As used herein, the term "amino acid" includes natural and synthetic amino -acids and derivatives thereof. The polymers can be c~-enared with a range of side chain lengths. The side chains preferably include between 10 and 100 amino acids, and have an overall amino acid content between 7 and 72 % . However, the side chains can include more than 100 amino acids and can have an overall amino acid content greater than 72%, depending on the reaction conditions. Poly(amino acids) can be grafted to the PLAL backbone in any suitable solvent. Suitable solvents include polar organic solvents such as dioxane, DMF, CH,CI2, and mixtures thereof. In a preferred embodiment, the reaction is conducted in dioxane at room temperature for a period of time between about 2 and 4 days.
Alternatively, the particles may be formed from polymers or blends of polymers with different polyester/amino acid backbones and grafted amino acid side chains. For example, poly(lactic acid-co-lysine-graft-alanine-lysine) (PLAL-Ala-Lys), or a blend of PLAL-Lys with poly(lactic acid-co-glycolic acid-block-ethylene oxide) (PLGA-PEG) (PLAL-Lys-PLGA-PEG) may be used.
- In the synthesis, the graft copolymers may be tailored to optimize different characteristics of the aerodynamically light particle including: i) ' S interactions between the agent to be delivered and the copolymer to provide stabilization of the agent and retention of activity upon delivery;
ii} rate of polymer degradation and, thereby, rate of drug release profiles;
iii) surface characteristics and targeting capabilities via chemical modification; and iv) particle porosity.
Therapeutic Agents Any of a variety of therapeutic agents can be incorporated within the particles, which can locally or systemically deliver the incorporated agents following administration to the lungs of an animal. Examples include synthetic inorganic and organic compounds or molecules, proteins and peptides, polysaccharides and other sugars, lipids, and nucleic acid molecules having therapeutic, prophylactic or diagnostic activities.
Nucleic acid molecules include genes, antisense molecules which bind to complementary DNA to inhibit transcription, ribozymes and ribozyme guide sequences. The agents to be incorporated can have a variety of biological activities, such as vasoactive agents, neuroactive agents, hormones, anticoagulants, immunomodulating agents, cytotoxic agents, prophylactic agents, antibiotics, antivirals, antisense, antigens, and antibodies. In some instances, the proteins rnay be antibodies or antigens which otherwise would have to be administered by injection to elicit an appropriate response. Compounds with a wide range of molecular weight, for example, between i00 and 500,000 grams per mole, can be encapsulated.
Proteins are defined as consisting of I00 amino acid residues or more; peptides are less than 100 amino acid residues. Unless otherwise stated, the term protein refers to both proteins and peptides. Examples include insulin and other hormones. Polysaccharides, such as heparin, can also be administered.

WO 97/44013 PCTlUS97/08895 Aerosols including the aerodynamically light particles are useful for a variety of inhalation therapies. The particles can incorporate small and large drugs, release the incorporated drugs over time periods ranging from hours to months, and withstand extreme conditions during aerosolization or following deposition in the lungs that might otherwise ' harm the encapsulated agents.
The agents can be locally delivered within the lung or can be systemically administered. For example, genes for the treatment of diseases such as cystic fibrosis can be administered, as can beta agonists for asthma. Other specific therapeutic agents include insulin, calcitonin, leuprolide (or L,hIRH), G-CSF, parathyroid hormone-related peptide, somatostatin, testosterone, progesterone, estradiol, nicotine, fentanyl, norethisterone, clonidine, scopolomine, salicylate, cromolyn sodium, salmeterol, formeterol, albeterol, and valium.
Diagnostic Agents Any of a variety of diagnostic agents can be incorporated within the particles, which can locally or systemically deliver the incorporated agents following administration to the lungs of an animal, including gases and other imaging agents.
Gases Any biocompatible or pharmacologically acceptable gas can be incorporated into the particles or trapped in the pores of the particles.
The term gas refers to any compound which is a gas or capable of forming a gas at the temperature at which imaging is being performed.
The gas may be composed of a single compound such as oxygen, nitrogen, xenon, argon, nitrogen or a mixture of compounds such as air.
Examples of fluorinated gases include CF,~, C~F~, C3F~, C4F~, SF,;, CZF4, and C3F6.
Other Imaging Agents Other imaging agents which may be utilized include commercially available agents used in positron emission tomography (PET), computer assisted tomography (CAT), single photon emission computerized tomography, x-ray, fluoroscopy, and magnetic resonance imaging (MRI).
Examples of suitable materials for use as contrast agents in MRI
include the gatalinium chelates currently available, such as diethylene triamine pentacetic acid (DTPA} and gatopentotate dimeglumine, as well as iron, magnesium, manganese, copper and chromium.
Examples of materials useful for CAT and x-rays include iodine based materials for intravenous administration, such as ionic monomers typified by diatrizoate and iothalamate, non-ionic monomers such as iopamidol, isohexol, and ioversol, non-ionic dimers, such as iotrol and iodixanoi, and ionic dimers, for example, ioxagalte.
Particles incorporating these agents can be detected using standard techniques available in the art and commercially available equipment.
Formation of Aerodynamically Light Polymeric Particles Aerodynamically light polymeric particles may be prepared using single and double emulsion solvent evaporation, spray drying, solvent extraction or other methods well known to those of ordinary skill in the art. The particles may be made, for example, using methods for making microspheres or microcapsules known in the art.
Methods for making microspheres are described in the literature, for example, in Mathiowitz and Langer, J. Controlled Release 5,13-22 (1987); Mathiowitz et al. , Reactive Polymers 6, 275-283 (1987); and Mathiowitz et al., J. Appl. Polymer Sci. 35, 755-774 (1988). The selection of the method depends on the polymer selection, the size, external morphology, and crystallinity that is desired, as described, for example, by Mathiowitz et al. , Scanning Microscopy 4, 329-340 ( 1990);
Mathiowitz et al., J. Appl. Polymer Sci. 45, 125-134 (1992); and Benita et al., J. Pharm. Sci. 73, 1721-1724 (1984}.
In solvent evaporation, described for example, in Mathiowitz, et al., (1990), Benita, and U.S. Patent No. 4,272,398 to Jaffe, a polymer is dissolved in a volatile organic solvent, such as methylene chloride.
Several different polymer concentrations can be used, for example, WO 97!44013 PCT/US97/08895 between 0.05 and 0.20 g/ml. An agent to be incorporated, either in soluble form or dispersed as fine particles, is optionally added to the polymer solution, and the mixture is suspended in an aqueous phase that contains a surface active agent such as polyvinyl alcohol). The resulting emulsion is stirred until most of the organic solvent evaporates, leaving solid microspheres, which may be washed with water and dried overnight in a lyophilizer.
Microspheres with different sizes (typically between 1 and 1000 microns) and morphologies can be obtained. This method is especially useful for relatively stable polymers such as polyesters and polystyrene.
However, labile polymers such as polyanhydrides may degrade due to exposure to water. Solvent removal may be a preferred method for preparing microspheres from these polymers.
Solvent removal was primarily designed for use with polyanhydrides. In this method, a therapeutic or diagnostic agent can be dispersed or dissolved in a solution of a selected polymer in a volatile organic solvent like methylene chloride. The mixture can then be suspended in oil, such as silicon oil, by stirring, to form an emulsion. As the solvent diffuses into the oil phase, the emulsion droplets harden into solid polymer microspheres. Unlike solvent evaporation, this method can be used to make microspheres from polymers with high melting points arid a wide range of molecular weights. Microspheres having a diameter between one and 300 microns can be obtained using this procedure.

WO 97/44013 PCT/LlS97/08895 Targeting of Particles Targeting molecules can be attached to the particles via reactive functional groups on the particles. For example, targeting molecules can be attached to the amino acid groups of functionalized polyester graft S copolymer particles, such as PLAL-Lys particles. Targeting molecules permit binding interactions of the particle with specific receptor sites, such as those within the lungs. The particles can be targeted by attaching Iigands which specifically or non-specifically bind to particular targets.
Exemplary targeting molecules include antibodies and fragments thereof including the variable regions, lectins, and hormones or other organic molecules capable of specific binding to receptors on the surfaces of the target cells.
Administration The particles can be administered to the respiratory system alone IS or in any appropriate pharmaceutically acceptable carrier, such as a liquid, for example saline, or a powder. In one embodiment, particles incorporating a prophylactic, therapeutic or diagnostic agent are co-delivered with larger carrier particles that do not include an incorporated agent. Preferably, the larger particles have a mass mean diameter between about SO and 100 lcm.
Aerosol dosage, formulations and delivery systems may be selected for a particular therapeutic application, as described, for example, in Gonda, I. "Aerosols for delivery of therapeutic and diagnostic agents to the respiratory tract," in Critical Reviews in Therapeutic Drug Carrier Systems, 6:273-313, 1990; and in Moren, "Aerosol dosage forms and formulations," in: Aerosols ih Medicine. Principles, Diagnosis arid Therapy, Moren, et al. , Eds, Elsevier, Amsterdam, 1985.
The greater efficiency of aerosolization by aerodynamically Iight . particles of relatively large size permits more of an incorporated agent to be delivered than is possible with the same mass of relatively dense aerosols. The relatively large particle size also minimizes potential drug losses caused by particle phagocytosis. When the particles are formed from biocompatible polymers, the system can provide controlled release in the lungs and long-time local action or systemic bioavailability of the incorporated agent. Denaturation of macromolecular drugs can be minimized during aerosolization since macromolecules are contained and protected within a polymeric shell. The enzymatic degradation of proteins or peptides can be minimized by co-incorporating peptidase-inhibitors.
Diagnostic Applications The particles can be combined with a pharmaceutically acceptable carrier, then an effective amount for detection administered to a patient via inhalation. Particles containing an incorporated imaging agent may be used for a variety of diagnostic applications, including detecting and characterizing tumor masses and tissues.
The present invention will be further understood by reference to the following non-limiting examples.
Example 1: Synthesis of Aerodynamically Light Poly[(p-carboxyphenoxy)-hexane anhydride] ("PCPH") Particles Aerodynamically light poly[(p-carboxyphenoxy)-hexane anhydride) ("PCPH") particles were synthesized as follows. 100 mg PCPH (MW
approximately 25,000) was dissolved in 3.0 mL methylene chloride. To this clear solution was added 5.0 mL 1 % w/v aqueous polyvinyl alcohol (PVA, MW approximately 25,000, 88 mole% hydrolyzed) saturated with methylene chloride, and the mixture was vortexed (Vortex Genie 2, Fisher Scientific) at maximum speed for one minute. The resulting milky-white emulsion was poured into a beaker containing 95 mL 1 PVA and homogenized (Siiverson Homogenizers) at 6000 RPM for one minute using a 0.75 inch tip. After homogenization, the mixture was stirred with a magnetic stirring bar and the methylene chloride quickly extracted from the polymer particles by adding 2 mL isopropyl alcohol.
The mixture was stirred for 35 minutes to allow complete hardening of the microparticles. The hardened particles were collected by centrifugation and washed several times with double distilled water. The particles were freeze dried to obtain a free-flowing powder void of clumps. Yield, 85-90 % .
The mean diameter of this batch was 6.0 ,um, however, particles with mean diameters ranging from a few hundred nanometers to several millimeters may be made with only slight modifications. Scanning electron rnicrograph photos of a typical batch of PCPH particles showed the particles to be highly porous with irregular surface shape. The particles had a tap density Less than 0.4 g/cm3.
Example 2: Synthesis of PLAL-Lys and PLAL-Lys-Ala Polymeric and Copolymeric Particles Aerodynamically Light PLAL-Lys Particles PLAL-Lys particles were prepared by dissolving 50 mg of the graft copolymer in 0.5 ml dimethylsulfoxide, then adding 1.5 m1 dichloromethane dropwise. The polymer solution is emulsified in 100 ml IS of 5 % w/v polyvinyl alcohol solution {average molecular weight 25KDa, 88 % hydrolyzed) using a homogenizes (Silverson) at a speed of approximately 7500 rpm. The resulting dispersion was stirred using a magnetic stirrer for 1 hour. Following this period, the pH was brought to between 7.0 and 7.2 by addition of a 0.1 N NaOH solution. Stirring was continued for an additional 2 hours until the dichloromethane was completely evaporated and the particles hardened. The particles were then isolated by centrifugation at 4000 rpm (1600 g) for 10 minutes (Sorvall RC-SB). The supernatant was discarded and the precipitate washed three times with distilled water followed by centrifugation for 10 minutes at 4000 rpm each time. Finally, the particles were resuspended in 5 ml of distilled water, the dispersion frozen in liquid nitrogen, and lyophilized (Labconco freeze dryer 8) for at Least 48 hours. Particle sizing was performed using a Coulter counter. Average particle mean diameters ranged from between 100 nm and I4 p,m, depending upon processing parameters such as homogenization speed and time. AlI
particles exhibited tap densities less than 0.4 g/em3. Scanning electron micrograph photos of the particles showed them to be highly porous with irregular surfaces.
Aerodynamically Light PLAL Ala-Lys Particles 100 mg of PLAL-Ala-Lys was completely dissolved in 0.4 ml triftuoroethanol, then 1.0 ml methylene chloride was added dropwise.
The polymer solution was emulsified in 100 ml of 1 % w/v polyvinyl alcohol solution (average molecular weight 25 KDa, 80 % hydrolyzed) using a sonicator (Sonic&Material VC-250) for 15 seconds at an output of 40 W. 2 ml of 1 % PVA solution was added to the mixture and it was vortexed at the highest speed for 30 seconds. The mixture was quickly poured into a beaker containing 100 ml 0.3 % PVA solution, and stirred for three hours allowing evaporation of the methylene chloride. Scanning electron micrograph photos of the particles showed them to possess highly irregular surfaces.
Aerodynamically Light Copolymer Particles Polymeric aerodynamically light particles consisting of a blend of PLAL-Lys and PLGA-PEG were made. 50 mg of the PLGA-PEG
polymer (molecular weight of PEG: 20 KDa, 1:2 weight ratio of PEG:PLGA, 75:25 lactide:glycolide) was completely dissolved in 1 ml dichloromethane. 3 mg of poly(lactide-co-lysine)-polylysine graft copolymer was dissolved in 0.1 ml dimethylsulfoxide and mixed with the first polymer solution. 0.2 ml of TE buffer, pH 7.6, was emulsified in the polymer solution by probe sonication (Sonic&Material VC-250) for 10 seconds at an output of 40W. To this first emulsion, 2 ml of distilled water was added and mixed using a vortex mixer at 4000 rpm for 60 secands. The resulting dispersion was agitated by using a magnetic stirrer for 3 hours until methylene chloride was completely evaporated and microspheres formed. The spheres were then isolated by centrifugation at 5000 rpm for 30 min. The supernatant was discarded, the precipitate washed three times with distilled water and resuspended in 5 ml of water.
The dispersion was frozen in liquid nitrogen and lyophilized for 48 hours.

By scanning electron microscopy (SEM), the PLAL-Lys-PLGA-PEG particles were highly surface rough and porous. The particles had a mean particle diameter of 7 um. The blend of PLAL-Lys with poly(lactic acid) (PLA) and/or PLGA-PEG copolymers can be adjusted to adjust panicle porosity and size.
Variables which may be manipulated to alter the size distribution of the particles include: polymer concentration, polymer molecular weight, surfactant type (e. g. , PVA, PEG, etc. ), surfactant concentration, and mixing intensity. Variables which may be manipulated to alter the surface shape and porosity of the particles include: polymer concentration, polymer molecular weight, rate of methylene chloride extraction by isopropyl alcohol (or another miscible solvent), volume of isopropyl alcohol added, inclusion of an inner water phase, volume of inner water phase, inclusion of salts or other highly water-soluble molecules in the inner water phase which leak out of the hardening sphere by osmotic pressure, causing the formation of channels, or pores, in proportion to their concentration, and surfactant type and concentration.
Additionally, processing parameters such as homogenization speed and time can be adjusted. Neither PLAL, PLA nor PLGA-PEG alone yields an aerodynamically light structure when prepared by these techniques.
Example 3: Synthesis of Spray-Dried Particles Aerodynamically Light Particles Containing Polymer and Drug Soluble in Common Solvent Aerodynamically light 50:50 PLGA particles were prepared by spray drying with testosterone encapsulated within the particles according to the following procedures. 2.0 g poly (D,L-lactic-co-glycolic acid) with a molar ratio of 50:50 (PLGA 50:50, ResomerTM RG503, B.I. Chemicals, Montvale, NJ) and 0.50 g testosterone (Sigma Chemical Co., St. Louis, MO) were completely dissolved in 100 mL dichloromethane at room temperature. The mixture was subsequently spray-dried through a 0.5 mm nozzle at a flow rate of 5 mLlmin using a Buchi laboratory spray-drier (model 190, Buchi, Germany). The flow rate of compressed air was 700 nl/h. The inlet temperature was set to 30°C and the outlet temperature to 25°C. The aspirator was set to achieve a vacuum of -20 to -25 bar. The yield was 51 % and the mean particle size was approximately 5 ,um. The particles were aerodynamically light, as determined by a tap density less than or equal to 0.4 g/cm'.
Larger particle size can be achieved by lowering the inlet compressed air flow rate, as well as by changing other variables.
Porosity and surface roughness can be increased by varying the inlet and outlet temperatures, among other factors.
Aerodynamically Light Particles Containing Polymer and Drug in Different Solvents Aerodynamically light PLA particles with a model hydrophilic drug {dextran) were prepared by spray drying using the following procedure. 2.0 mL of an aqueous 10% w/v FITC-dextran (MW 70,000, Sigma Chemical Co.) solution was emulsified into 100 mL of a 2%a w/v solution of poly (D,L-lactic acid) (PLA, Resomer 8206, B.I. Chemicals) in dichloromethane by probe sonication (Vibracell Sonicator, Branson).
The emulsion was subsequently spray-dried at a flow rate of 5 mL/min with an air flow rate of 700 nl/h (inlet temperature =30°C, outlet temperature = 21°C, -20 mbar vacuum). The yield is 56%. The particles were aerodynamically light {tap density less than 0.4 glcm~).
Aerodynamically Light Protein Particles Aerodynamically light lysozyrne particles were prepared by spray drying using the following procedure. 4.75 g lysozyme (Sigma) was dissolved in 95 mL double distilled water (5 % w/v solution) and spray-dried using a 0.5 mm nozzle and a Buchi laboratory spray-drier. The flow rate of compressed air was 725 ni/h. The flow rate of the Iysozyme solution was set such that, at a set inlet temperature of 97-100°C, the outlet temperature is between 55 and 57°C. The aspirator was set to achieve a vacuum of -30 mbar. The enzymatic activity of lysozyme was found to be unaffected by this process and the yield of the aerodynamically light particles (tap density less than 0.4 g/cm3) was 66 % .
Aerodynamically Light High-Molecular Weight Water-Soluble Particles Aerodynamically light dextran particles were prepared by spray drying using the following procedure. 6.04 g DEAE dextran {Sigma) was dissolved in 242 mL double distilled water (2.5 % w/v solution) and spray-dried using a 0.5 mm nozzle and a Buchi laboratory spray-drier.
The flow rate of compressed air was 750 nI/h. The flow rate of the DEAE-dextran solution was set such that, at a set inlet temperature of 155°C, the outlet temperature was 80°C. The aspirator was set to achieve a vacuum of -20 mbar. The yield of the aerodynamically light particles (tap density less than 0.4 g/cm~) was 66 % and the size range ranged between 1 and 15 tcm.
Aerodynamically Light Low-Molecular Weight Water-Soluble Particles Aerodynamically light trehalose particles were prepared by spray drying using the following procedure. 4.9 g trehaIose (Sigma) was dissolved in 192 mL double distilled water (2.5 % w/v solution) and spray-dried using a 0.5 mm nozzle arid a Buchi laboratory spray-drier.
The flow rate of compressed air 650 nl/h. The flow rate of the trehalose solution was set such that, at a set inlet temperature of I00°C, the outlet temperature was 60°C. The aspirator was set to achieve a vacuum of -30 mbar. The yield of the aerodynamically light particles (tap density less than 0.4 g/cm3) was 36 % and the size range ranged between 1 and 15 Vim.
Aerodynamically Light Low-Molecular Weight Water-Soluble Particles Polyethylene glycol (PEG) is a water-soluble macromolecule, however, it cannot be spray dried from an aqueous solution since it melts at room temperatures below that needed to evaporate water. PEG was spray-dried at low temperatures from a solution in dichloromethane, a low boiling organic solvent. Aerodynamically light PEG particles were prepared by spray drying using the following procedure. 5.0 g PEG
(MW 15,000-20,000, Sigma) was dissolved in 100 mL double distilled water (5.0 % w/v soiution) and spray-dried using a 0.5 mm nozzle and a Buchi laboratory spray-drier. The flow rate of compressed air was 750 ' nl/h. The flow rate of the PEG solution was set such that, at a set inlet temperature of 45 °C, the outlet temperature was 34-35 °C. The aspirator was set to achieve a vacuum of -22 mbar. The yield of the aerodynamically light particles (tap density less than 0.4 g/cm3) was 67 and the size range ranged between 1 and 15 ~,m.
Example 4: IZtlodamine Isothiocyanate Labeling of PLAL and PLAL-Lys Particles Aerodynamically light particles were compared with control particles, referred to herein as "non-light" particles. Lysine amine groups on the surface of aerodynamically light (PLAL-Lys) and control, non-light (PLAL) particles, with similar mean diameters (between 6 and 7 ~cm) and size distributions (standard deviations between 3 and 4 ~,m) were labeled with lRhodamine isothiocyanate. The tap density of the porous PLAL-Lys particles was 0.1 g/cm3 and that of the denser PLAL particles was 0.8 g/cm3.
The rhodamine-labeled particles were characterized by confocal microscopy. A limited number of lysine functionalities on the surface of the solid particle were able to react with rhodamine isothiocyanate, as evidenced by the fluorescent image. In the aerodynamically light particle, the higher lysine content in the graft copolymer and the porous particle structure result in a higher level of rhodamine attachment, with rhodamine attachment dispersed throughout the interstices of the porous structure.
This also demonstrates that targeting molecules can be attached to the aerodynamically light particles for interaction with specific receptor sites within the lungs via chemical attachment of appropriate targeting agents to the particle surface.

Example 5: Aerosolization of PLAL and PLAL-Lys Particles To determine whether large aerodynamically light particles can escape {mouth, throat and inhaler) deposition and more efficiently enter the airways and acini than nonporous particles of similar size (referred to S herein as non-light or control particles), the aerosolization and deposition of aerodynamically light PLAL-Lys (mean diameter 6.3 ~,m) arid control, non-light PLAL (mean diameter 6.9 ,um) particles was compared in vitro using a cascade impactor system.
20 mg of the aerodynamically light or non-light microparticles IO were placed in gelatine capsules (Eli Lilly), the capsules loaded into a Spinhaler dry powder inhaler (DPI) (Fisons), and the DPI activated.
Particles were aerosolized into a Mark I Andersen Impactor (Andersen Samplers, GA) from the DPI for 30 seconds at 28.3 1/min flow rate.
Each plate of the Andersen Impactor was previously coated with Tween 15 80 by immersing the plates in an acetone solution (5 % w/vol) and subsequently evaporating the acetone in a oven at 60°C for 5 min. After aerosolization and deposition, particles were collected from each stage of the impactor system in separate volumetric flasks by rinsing each stage with a NaOH solution (0.2 N) in order to completely degrade the 20 polymers. After incubation at 37°C fox I2 h, the fluorescence of each solution was measured (wavelengths of 554 nm excitation, 574 nm emission) .
Particles were determined as nonrespirable (mean aerodynamic diameter exceeding 4.7 pm: impactor estimate) if they deposited on the 25 first three stages of the impactor, and respirable (mean aerodynamic diameter 4.7 ~cm or less) if they deposited on subsequent stages. Figure 1 shows that less than 10% of the non-light (PLAL) particles that exit the DPI are respirable. This is consistent with the large size of the microparticles and their standard mass density. On the other hand, 30 greater than 55 % of the aerodynamically light (PLAL-Lys) particles are respirable, even though the geometrical dimensions of the two particle types are almost identical. The lower tap density of the aerodynamically light (PLAL-Lys) microparticles is responsible for this improvement in particle penetration, as discussed further below.
The non-light (PLAL) particles also inefficiently aerosolize from the DPI; typically, less than 40 % of the non-light particles exited the Spinhaler DPI for the protocol used. The aerodynamically light (PLAL- ' Lys) particles exhibited much more efficient aerosolization (approximately 80% of the aerodynamically light microparticles typically exited the DPI
during aerosolization}.
The combined effects of efficient aerosolization and high respirable fraction of aerosolized particle mass means that a far greater fraction of an aerodynamically light particle powder is likely to deposit in the lungs than of a non-light particle powder.
Example 6: In Vivo Aerosolization of PLAL and PLAL-Lys Particles The penetration of aerodynamically light and non-light polymeric PLAL-Lys and PLAL microparticles into the lungs was evaluated in an in vivo experiment involving the aerosolization of the microparticles into the airways of live rats.
Male Sprague Dawley rats (150-200 g) were anesthetized using ketamine (90mg/kg)/xylazine (lOmg/kg). The anesthetized rat was placed ventral side up on a surgical table provided with a temperature controlled pad to maintain physiological temperature. The animal was cannulated above the carina with an endotracheal tube connected to a Harvard ventilator. The animal was force ventilated for 20 minutes at 300 ml/min. 50 mg of aerodynamically light (PLAL-Lys) or non-light (PLA) microparticles were introduced into the endotracheal tube.
Following the period of forced ventilation, the animal was euthanized and the lungs and trachea were separately washed using bronchoalveolar lavage. A tracheal cannula was inserted, tied into place, and the airways were washed with 10 mI aliquots of HBSS. The lavage procedure was repeated until a total volume of 30 ml was collected. The lavage fluid was centrifuged (400 g) and the pellets collected and resuspended in 2 ml of phenol red-free Hanks balanced salt solution (Gibco, Grand Island, NY) without Ca2 and Mg2~' {HBSS). 100 ml were removed for particle counting using a hemacytometer. The remaining solution was mixed with 10 ml of 0.4 N NaOH. After incubation at 37 °C
for 12 h, the fluorescence of each solution was measured (wavelengths of ~ S 554 nm excitation, 574 nm emission). Figure 2 is a bar graph showing total particle mass deposited in the trachea and after the carina (lungs} in rat lungs and upper airways following intratracheal aerosolization during forced ventilation. The PLAL-Lys aerodynamically light particles had a mean diameter of 6.9 ~cm. The non-light PLAL
particles had a mean diameter of 6.7 ,um. Percent tracheal aerodynamically light particle deposition was 54.5, and non-light deposition was 77Ø Percent aerodynamically light particle deposition in the lungs was 46.8 and non-light deposition was 23Ø
The non-light (PLAL) particles deposited primarily in the trachea {approximately 79 % of all particle mass that entered the trachea) . This result is similar to the in vitro performance of the non-light microparticles and is consistent with the relatively large size of the nonlight particles.
Approximately 54% of the aerodynamically light (PLAL-Lys) particle mass deposited in the trachea. Therefore, about half of the aerodynamically light particle mass that enters the trachea traverses through the trachea and into the airways and acini of the rat lungs, demonstrating the effective penetration of the aerodynamically light particles into the lungs.
Following bronchoalveolar Iavage, particles remaining in the rat lungs were obtained by careful dissection of the individual lobes of the lungs. The lobes were placed in separate petri dishes containing 5 ml of HBSS. Each lobe was teased through 60 mesh screen to dissociate the tissue and was then filtered through cotton gauze to remove tissue debris and connective tissue. The petri dish and gauze were washed with an additional 15 ml of HBSS to maximize microparticle collection. Each tissue preparation was centrifuged and resuspended in 2 m1 of HBSS and the number of particles counted in a hemacytometer. The particle numbers remaining in the lungs following the bronchoalveolar lavage are shown in Figure 3. Lobe numbers correspond to: 1) left lung, 2) anterior, 3) median, 4) posterior, 5) postcaval. A considerably greater , number of aerodynamically light PLAL-Lys particles enters every lobe of the lungs than the nonlight PLAL particles, even though the geometrical "
dimensions of the two types of particles are essentially the same. These results reflect both the efficiency of aerodynamically light particle aerosolization and the propensity of the aerodynamically light particles to escape deposition prior to the caring or first bifurcation.
Rxample 7: In Vivo Aerosoiization of PLGA Porous and Non-Porous Particles Including Insulin Insulin was encapsulated into porous and nonporous polymeric particles to test whether large particle size can increase systemic bioavailability. The mass densities and mean diameters of the two particles were designed such that they each possessed an aerodynamic diameter (approximately 2 ~,m) suitable for deep lung deposition, with the mean diameter of the porous particles >5 ~,m and that of the nonporous particles less than 5 ~,m (see Figures 4-6). Identical masses of the porous or nonporous particles were administered to rats as an inhalation aerosol or injected subcutaneously (controls).
Rats were anesthetized and cannulated as previously described.
The animal was force ventilated for between 10 and 20 minutes at 300 ml/min. Two types of aerosols were delivered to the animal via the endotracheal tube. Following the period of forced ventilation, the neck of the animal was sutured and the animal revived within one to two hours.
Blood samples (300 ~.I) were periodically withdrawn from the tail vein over a period of two to six days. These samples were mixed with assay buffer, centrifuged, and the supernatant examined for the presence of (endogenous and exogenous) insulin or testosterone using radioimmunoassays (ICN Pharmaceuticals, Costa Mesa, CA). Glucose was measured using a colorimetric assay (Sigma). Control studies involved subcutaneous injection of the same amount of powder as was inhaled. The particles were injected into the scruff of the neck.
Serum insulin concentrations were monitored as a function of time following inhalation or injection. For both porous {Figure 4) and nonporous (Figure 5) particles, blood levels of insulin reach high values within the first hour following inhalation. Only in the case of the large porous particles do blood levels of insulin remain elevated (p < 0.05) beyond 4 h, with a relatively constant insulin release continuing to at least 96 h (0.04<p<0.2).
IO These results are confirmed by serum glucose values which show falling glucose levels for the first 10 h after inhalation of the porous insulin particles, followed by relatively constant low glucose levels for the remainder of the 96 h period, as shown in Figure 6. In the case of small nonporous insulin particles, initially suppressed glucose values rose after 24 h.
Similar biphasic release profiles of macromolecules from PLGA
polymers have been reported in the literature (S. Cohen et al. Plaarm.
Res. 8, 713 (1991)). For the large porous particles, insulin bioavailability relative to subcutaneous injection is 87.5 % , whereas the small nonporous particles yield a relative bioavailability of 12% following inhalation. By comparison, bioavaiIability (relative to subcutaneous injection) of insulin administered to rats as an inhalation liquid aerosol using a similar endotracheal method has been reported as 37.3 % (P. Colthorpe et al.
Phar-rn. Res. 9, 764 (1992)). Absolute bioavailability of insulin inhaled into rat lungs in the form of a Iactose/insulin powder via a dry powder inhaler connected to an endotracheal tube has been reported as 6.5 % (F.
Komada et al. J. Phanrz. Sci. 83, 863 (1994)).
Given the short systemic half life of insulin (11 minutes), and the 12-24 h time scale of particle clearance from the central and upper airways, the appearance of exogenous insulin in the bloodstream several days following inhalation appears to indicate that large porous particles achieve long, non-phagocytosed life-times when administered to the deep Lung. To test this hypothesis, the lungs of rats were savaged both immediately following inhalation of the porous and nonporous insulin particles, and 48 h after inhalation. .
In the case of nonporous particles, 30 % ~ 3 % of phagocytic cells contained particles immediately following inhalation, and 39 % ~5 % ' contained particles 48 h after inhalation. By contrast only 8 % ~ 2 % of phagocytic cells contained large porous particles right after inhalation, and 12.5 % t 3.5 % contained particles 48 h after inhalation. In the small nonporous particle case, 17.5 % t 1.5 % of the phagocytic cell population contained 3 or more particles 48 h after inhalation, compared to 4 % t 1 in the case of the large nonporous particles. Inflammatory response was also elevated in the small nonporous particle case; neutrophils represented 34 % t 12 % of the phagocytic cell population 48 h following inhalation of the small nonporous particles, compared to 8.S % t3.5 % in the large porous particle case (alveolar macrophages represented 100 % of phagocytic cells immediately following inhalation). These results support in vitro experimental data appearing elsewhere that show phagocytosis of particles diminishes precipitously as particle diameter increases beyond 3,um (H. Kawaguchi, et al. Biomaterials 7, 61 (1986). L.J. ~renis, and B. Strauss, Proc. Soc. Exp. Med. 107, 748 (1961). S. Rudt, and R.Ii.
Muller, J. Contr. Rel. 22, 263 (1992)).
Example 8: In Vivo AerosoIization of PLGA Porous Particles Including Testosterone A second model drug, testosterone, was encapsulated in porous particles of two different mean geometric diameters (10.1 ~.m and 20.4 ~cm) to further determine whether increased bioavailability correlates with increasing size of porous particles. An identical mass of powder was administered to rats as an inhalation aerosol or as a subcutaneous injection (controls). Serum testosterone concentrations were monitored as a function of time following inhalation or injection (Figures 7 and 8).
Blood levels of testosterone remain well above background levels (p < O.OS) for between 12 and 24 h, even though the systemic half-life of testosterone is between 10 and 20 minutes. Testosterone bioavailability relative to subcutaneous injection is 177 % for the 20.4 lcm diameter , particles (Figure 7) and 53 % for the 10.1 tcm diameter porous particles (Figure 8).
S The increase in testosterone bioavailability with increasing size of porous particles is especially notable given that the mean diameter of the 20.4 tcm particles is approximately ten times larger than that of nonporous conventional therapeutic particles (D. Ganderton, J. Biopharmaceutical Sciences 3, 101 (1992). The relatively short time scale of testosterone release observed both for the inhalation and subcutaneous controls is near the several hour in vitro time scale of release reported elsewhere for 50:50 PLGA microparticles of similar size encapsulating a therapeutic substance (bupivacaine) of similar molecular weight and Iipophilicity (P.
Le Corre et al. Int. J. Pharm. 107, 41 (1994)).
By making particles with high porosity, relatively large particles {i.e., those possessing the same aerodynamic diameter as smaller, nonporous particles) can enter the lungs, since it is particle mass that dictates location of aerosol deposition in the lungs. The increased aerosolization efficiency of large, light particles lowers the probability of deposition losses prior to particle entry into the airways, thereby increasing the systemic bioavailability of an inhaled drug.

Claims (19)

We claim:
1. A particulate system for delivery to the pulmonary system comprising: particles comprising (a) a therapeutic or diagnostic agent and a biodegradable and biocompatible excipient and (b) characterized by a tap density of less than 0.4 g/cm3 and wherein at least 50% of the particles have a mass mean diameter between 5µm and 30µm.
2. The system of claim 1 wherein at least 90 % of the particles have a mass mean diameter between 5 µm and 30 µm.
3. The system of claim 2 wherein at least 50% of the particles have a mass mean diameter between 5 µm and 15 µm.
4. The system of claim 1 further comprising a pharmaceutically acceptable carrier for administration to the lungs.
5. The system of claim 1 wherein the particles comprise a functionalized polyester graft copolymer comprising:
a linear .alpha.-hydroxy-acid polyester backbone having at least one amino acid group incorporated therein; and at least one poly(amino acid) side chain extending from an amino acid group in the polyester backbone.
6. The system of claim 5 wherein the polyester is a polymer of an .alpha.-hydroxy acid selected from the group consisting of lactic acid, glycolic acid, hydroxybutyric acid and valeric acid, and copolymers thereof.
7. The system of claim 5 wherein the linear .alpha.-hydroxy-acid polyester backbone comprises a poly(lactic acid) polymer having at least one lysine group incorporated therein; and wherein the poly(amino acid) extends from at least one lysine group in the polyester backbone.
8. The system of claim 7 wherein the poly(amino acid) is a polymer of an amino acid selected from the group consisting of aspartic acid, lysine and alanine, and copolymers thereof.
9. The system of claim 7 wherein the polymer is selected from the group consisting of poly(lactic acid-co-lysine-graft-lysine) and poly(lactic acid-co-lysine-graft-alanine-lysine).
10. The system of claim 5 wherein the particles comprise a blend of a polyester and the functionalized polyester graft copolymer.
11. The system of claim 10 wherein the particles comprise a blend of poly(lactic acid-co-lysine-graft-lysine) and poly(lactic acid-co-glycolic acid-block-ethylene oxide).
12. The system of claim 5 wherein the poly(amino acid) side chain includes between about 10 and 100 amino acids.
13. The system of claim 5 wherein the polymer has an amino acid content of between about 7 and 70%.
14. The system of claim 1, wherein the particles incorporate a therapeutic or diagnostic agent.
15. The system of claim 14 wherein the agent is selected from the group consisting of proteins, polysaccharides, lipids, nucleic acid molecules, synthetic organic or inorganic molecules and combinations thereof.
16. The system of claim 1 wherein a targeting molecule is attached to the particle.
17. The system of claim 1 combined with large biodegradable carrier particles, containing no therapeutic, the mass mean diameter of which is in the range of 50 µm and 100 µm.
18. A method for making aerodynamically light particles comprising a therapeutic or diagnostic agent, a biodegradable and biocompatible excipient and having a tap density of less than 0.4 g/cm3 and wherein at least 50% of the particles have a mass mean diameter between 5 µm and 30 µm, for administration to the respiratory tract by aerosolization, the method comprising forming the particles from a functionalized polyester graft copolymer comprising:
a linear .alpha.-hydroxy-acid polyester backbone having at least one amino acid group incorporated therein; and at least one poly(amino acid) side chain extending from an amino acid group in the polyester backbone.
19. The method of claim 18 wherein the aerodynamically light particles are made by a solvent evaporation process.
CA002255564A 1996-05-24 1997-05-23 Aerodynamically light particles for pulmonary drug delivery Expired - Lifetime CA2255564C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US65557096A 1996-05-24 1996-05-24
US655570 1996-05-24
US08/739,308 US5874064A (en) 1996-05-24 1996-10-29 Aerodynamically light particles for pulmonary drug delivery
PCT/US1997/008895 WO1997044013A1 (en) 1996-05-24 1997-05-23 Aerodynamically light particles for pulmonary drug delivery
US739308 2007-04-24

Publications (2)

Publication Number Publication Date
CA2255564A1 CA2255564A1 (en) 1997-11-27
CA2255564C true CA2255564C (en) 2006-08-29

Family

ID=27096992

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002255564A Expired - Lifetime CA2255564C (en) 1996-05-24 1997-05-23 Aerodynamically light particles for pulmonary drug delivery

Country Status (10)

Country Link
US (6) US5874064A (en)
EP (1) EP0907356B1 (en)
JP (2) JP4315468B2 (en)
AT (1) ATE215357T1 (en)
CA (1) CA2255564C (en)
DE (1) DE69711626T2 (en)
DK (1) DK0907356T3 (en)
ES (1) ES2175420T3 (en)
PT (1) PT907356E (en)
WO (1) WO1997044013A1 (en)

Families Citing this family (581)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ244439A (en) * 1991-09-25 1994-01-26 Fisons Plc Pressurised aerosol compositions comprising hydrofluoroalkane, dispersed
US6582728B1 (en) * 1992-07-08 2003-06-24 Inhale Therapeutic Systems, Inc. Spray drying of macromolecules to produce inhaleable dry powders
US6051256A (en) * 1994-03-07 2000-04-18 Inhale Therapeutic Systems Dispersible macromolecule compositions and methods for their preparation and use
US6428771B1 (en) * 1995-05-15 2002-08-06 Pharmaceutical Discovery Corporation Method for drug delivery to the pulmonary system
US5874064A (en) * 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US6254854B1 (en) * 1996-05-24 2001-07-03 The Penn Research Foundation Porous particles for deep lung delivery
US6503480B1 (en) * 1997-05-23 2003-01-07 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US6652837B1 (en) * 1996-05-24 2003-11-25 Massachusetts Institute Of Technology Preparation of novel particles for inhalation
US20020052310A1 (en) * 1997-09-15 2002-05-02 Massachusetts Institute Of Technology The Penn State Research Foundation Particles for inhalation having sustained release properties
US8022095B2 (en) * 1996-08-16 2011-09-20 Pozen, Inc. Methods of treating headaches using 5-HT agonists in combination with long-acting NSAIDs
EP0946169B1 (en) * 1996-12-20 2003-02-26 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
JP2001507702A (en) 1996-12-31 2001-06-12 インヘイル・セラピューティックス・システムズ・インコーポレテッド Method for spray drying an aqueous suspension of a hydrophobic drug having a hydrophilic excipient and a composition made by the method
US20030203036A1 (en) * 2000-03-17 2003-10-30 Gordon Marc S. Systems and processes for spray drying hydrophobic drugs with hydrophilic excipients
US6126919A (en) * 1997-02-07 2000-10-03 3M Innovative Properties Company Biocompatible compounds for pharmaceutical drug delivery systems
US7779020B2 (en) * 2002-03-01 2010-08-17 International Business Machines Corporation Small-footprint applicative query interpreter method, system and program product
CN1271277A (en) * 1997-07-18 2000-10-25 因菲米德有限公司 Biodegradable macromers fro the controlled release of biologically active substances
US7052678B2 (en) 1997-09-15 2006-05-30 Massachusetts Institute Of Technology Particles for inhalation having sustained release properties
US20020017295A1 (en) * 2000-07-07 2002-02-14 Weers Jeffry G. Phospholipid-based powders for inhalation
US6946117B1 (en) * 1997-09-29 2005-09-20 Nektar Therapeutics Stabilized preparations for use in nebulizers
US6433040B1 (en) * 1997-09-29 2002-08-13 Inhale Therapeutic Systems, Inc. Stabilized bioactive preparations and methods of use
US6309623B1 (en) 1997-09-29 2001-10-30 Inhale Therapeutic Systems, Inc. Stabilized preparations for use in metered dose inhalers
US20060165606A1 (en) * 1997-09-29 2006-07-27 Nektar Therapeutics Pulmonary delivery particles comprising water insoluble or crystalline active agents
US6565885B1 (en) * 1997-09-29 2003-05-20 Inhale Therapeutic Systems, Inc. Methods of spray drying pharmaceutical compositions
ZA9811377B (en) * 1997-12-12 1999-08-27 Expression Genetics Inc Positively charged poly[alpha-(omega-aminoalkyl) glycolic acid[ for the delivery of a bioactive agent via tissue and cellular uptake.
GB9727102D0 (en) * 1997-12-22 1998-02-25 Andaris Ltd Microparticles and their therapeutic use
US6958148B1 (en) * 1998-01-20 2005-10-25 Pericor Science, Inc. Linkage of agents to body tissue using microparticles and transglutaminase
CA2231968A1 (en) * 1998-03-11 1999-09-11 Smoke-Stop, A Partnership Consisting Of Art Slutsky Method of producing a nicotine medicament
US6284282B1 (en) 1998-04-29 2001-09-04 Genentech, Inc. Method of spray freeze drying proteins for pharmaceutical administration
US6423345B2 (en) * 1998-04-30 2002-07-23 Acusphere, Inc. Matrices formed of polymer and hydrophobic compounds for use in drug delivery
EP1273290A1 (en) * 1998-06-24 2003-01-08 Advanced Inhalation Research, Inc. Large porous particles emitted from an inhaler
CA2336139C (en) * 1998-06-24 2008-10-14 Advanced Inhalation Research, Inc. Large porous particles emitted from an inhaler
US6956021B1 (en) 1998-08-25 2005-10-18 Advanced Inhalation Research, Inc. Stable spray-dried protein formulations
US7056504B1 (en) 1998-08-27 2006-06-06 Massachusetts Institute Of Technology Rationally designed heparinases derived from heparinase I and II
MA25590A1 (en) 1998-09-14 2002-12-31 Inhale Therapeutic Syst ACTIVE AGENT FOR DRY POWDER DELIVERY
US8933032B2 (en) 1998-10-20 2015-01-13 Children's Hospital Medical Center Surfactant protein D for the treatment of disorders associated with lung injury
SK286981B6 (en) * 1999-03-05 2009-08-06 Chiesi Farmaceutici S.P.A. Powder for use in a dry powder inhaler, carrier for the powder and method for the production of powder
US20050214227A1 (en) * 1999-03-08 2005-09-29 Powderject Research Limited Microparticle formulations for sustained-release of bioactive compounds
EP1190364A2 (en) 1999-04-23 2002-03-27 Massachusetts Institute Of Technology System and method for polymer notation
US6428769B1 (en) * 1999-05-04 2002-08-06 Aradigm Corporation Acute testosterone administration
JP2003501404A (en) * 1999-06-04 2003-01-14 デルルックス ファーマシューティカル コーポレイション Preparation comprising dehydrated particles of drug and method of preparing same
US6858199B1 (en) 2000-06-09 2005-02-22 Advanced Inhalation Research, Inc. High efficient delivery of a large therapeutic mass aerosol
US9006175B2 (en) 1999-06-29 2015-04-14 Mannkind Corporation Potentiation of glucose elimination
AU779986B2 (en) 1999-06-29 2005-02-24 Mannkind Corporation Purification and stabilization of peptide and protein pharmaceutical agents
US7252840B1 (en) 1999-08-25 2007-08-07 Advanced Inhalation Research, Inc. Use of simple amino acids to form porous particles
US20010036481A1 (en) * 1999-08-25 2001-11-01 Advanced Inhalation Research, Inc. Modulation of release from dry powder formulations
US6586008B1 (en) * 1999-08-25 2003-07-01 Advanced Inhalation Research, Inc. Use of simple amino acids to form porous particles during spray drying
WO2001013891A2 (en) * 1999-08-25 2001-03-01 Advanced Inhalation Research, Inc. Modulation of release from dry powder formulations
US7678364B2 (en) 1999-08-25 2010-03-16 Alkermes, Inc. Particles for inhalation having sustained release properties
US6749835B1 (en) 1999-08-25 2004-06-15 Advanced Inhalation Research, Inc. Formulation for spray-drying large porous particles
US6706892B1 (en) * 1999-09-07 2004-03-16 Conjuchem, Inc. Pulmonary delivery for bioconjugation
US6676930B2 (en) 1999-11-28 2004-01-13 Scientific Development And Research, Inc. Composition and method for treatment of otitis media
US6156294A (en) 1999-11-28 2000-12-05 Scientific Development And Research, Inc. Composition and method for treatment of otitis media
WO2001045731A1 (en) 1999-12-21 2001-06-28 Rxkinetix, Inc. Particulate drug-containing products and method of manufacture
US6761909B1 (en) * 1999-12-21 2004-07-13 Rxkinetix, Inc. Particulate insulin-containing products and method of manufacture
CN100333790C (en) * 1999-12-30 2007-08-29 希龙公司 Methods for pulmonary delivery of interleukin-2
WO2001051030A1 (en) * 2000-01-10 2001-07-19 Dura Pharmaceuticals, Inc. Pharmaceutical formulation and method for pulmonary and oral delivery
US7029700B2 (en) * 2000-01-14 2006-04-18 Brown University Research Foundation Micronized freeze-dried particles
AU2001231000A1 (en) 2000-01-19 2001-07-31 Pharmaceutical Discovery Corporation Dry powder formulations of antihistamine for nasal administration
AU2002219944B2 (en) 2000-11-28 2008-02-21 Medimmune, Llc Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
US6656467B2 (en) * 2000-01-27 2003-12-02 Medimmune, Inc. Ultra high affinity neutralizing antibodies
US7229619B1 (en) 2000-11-28 2007-06-12 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
AU4724401A (en) 2000-02-28 2001-09-12 Genesegues Inc Nanocapsule encapsulation system and method
ES2390761T3 (en) * 2000-03-01 2012-11-16 Medimmune, Llc Recombinant high-potency antibodies and method for their production
EP1266013B1 (en) * 2000-03-08 2014-10-15 Massachusetts Institute Of Technology Heparinase iii and uses thereof
US20010046955A1 (en) * 2000-03-14 2001-11-29 Gelfand Erwin W. Method for reducing allergen-induced airway hyperresponsiveness
US8404217B2 (en) * 2000-05-10 2013-03-26 Novartis Ag Formulation for pulmonary administration of antifungal agents, and associated methods of manufacture and use
AU6124601A (en) * 2000-05-10 2001-11-20 Alliance Pharmaceutical Corporation Phospholipid-based powders for drug delivery
US7871598B1 (en) 2000-05-10 2011-01-18 Novartis Ag Stable metal ion-lipid powdered pharmaceutical compositions for drug delivery and methods of use
US7575761B2 (en) * 2000-06-30 2009-08-18 Novartis Pharma Ag Spray drying process control of drying kinetics
US20020106368A1 (en) * 2000-07-28 2002-08-08 Adrian Bot Novel methods and compositions to upregulate, redirect or limit immune responses to peptides, proteins and other bioactive compounds and vectors expressing the same
AU2001277230A1 (en) * 2000-08-01 2002-02-13 Inhale Therapeutic Systems, Inc. Apparatus and process to produce particles having a narrow size distribution andparticles made thereby
US20060177416A1 (en) 2003-10-14 2006-08-10 Medivas, Llc Polymer particle delivery compositions and methods of use
WO2002023190A2 (en) 2000-09-12 2002-03-21 Massachusetts Institute Of Technology Methods and products related to low molecular weight heparin
US6613308B2 (en) 2000-09-19 2003-09-02 Advanced Inhalation Research, Inc. Pulmonary delivery in treating disorders of the central nervous system
US6514482B1 (en) 2000-09-19 2003-02-04 Advanced Inhalation Research, Inc. Pulmonary delivery in treating disorders of the central nervous system
CA2423469A1 (en) 2000-10-18 2002-04-25 Massachusetts Institute Of Technology Methods and products related to pulmonary delivery of polysaccharides
GB0028268D0 (en) * 2000-11-20 2001-01-03 Norske Stats Oljeselskap Well treatment
US6818216B2 (en) 2000-11-28 2004-11-16 Medimmune, Inc. Anti-RSV antibodies
US7179900B2 (en) * 2000-11-28 2007-02-20 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
US6855493B2 (en) * 2000-11-28 2005-02-15 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
EP2168571B1 (en) * 2000-11-30 2018-08-22 Vectura Limited Particles for use in a Pharmaceutical Composition
US20020128179A1 (en) * 2000-12-01 2002-09-12 Tacon William C. Shaped microparticles for pulmonary drug delivery
WO2002043705A2 (en) * 2000-12-01 2002-06-06 University Of Florida Aerodynamically light vaccine for active pulmonary immunization
EP2341060B1 (en) 2000-12-12 2019-02-20 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
US7247704B2 (en) 2000-12-18 2007-07-24 Arriva Pharmaceuticals, Inc. Multifunctional protease inhibitors and their use in treatment of disease
ES2390425T3 (en) 2000-12-22 2012-11-12 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Use of repulsive targeting molecules (RGM) and their modulators
KR20030074693A (en) * 2000-12-28 2003-09-19 알투스 바이올로직스 인코포레이티드 Crystals of whole antibodies and fragments thereof and methods for making and using them
US20020141946A1 (en) * 2000-12-29 2002-10-03 Advanced Inhalation Research, Inc. Particles for inhalation having rapid release properties
EP1345629A2 (en) 2000-12-29 2003-09-24 Advanced Inhalation Research, Inc. Particles for inhalation having sustained release properties
US6749845B2 (en) * 2001-02-15 2004-06-15 Aeropharm Technology, Inc. Modulated release particles for lung delivery
US6848197B2 (en) * 2001-04-18 2005-02-01 Advanced Inhalation Research, Inc. Control of process humidity to produce large, porous particles
CN1314445C (en) * 2001-05-21 2007-05-09 耐科塔医药公司 Pulmonary administration of chemically modified insulin
JP2005503425A (en) 2001-05-24 2005-02-03 アレックザ モレキュラー デリヴァリー コーポレイション Delivery of drug ester by the prescribed inhalation route
US20070122353A1 (en) * 2001-05-24 2007-05-31 Hale Ron L Drug condensation aerosols and kits
US20030051728A1 (en) * 2001-06-05 2003-03-20 Lloyd Peter M. Method and device for delivering a physiologically active compound
US7645442B2 (en) 2001-05-24 2010-01-12 Alexza Pharmaceuticals, Inc. Rapid-heating drug delivery article and method of use
US6805853B2 (en) * 2001-11-09 2004-10-19 Alexza Molecular Delivery Corporation Delivery of diazepam through an inhalation route
US7458374B2 (en) * 2002-05-13 2008-12-02 Alexza Pharmaceuticals, Inc. Method and apparatus for vaporizing a compound
US6759029B2 (en) * 2001-05-24 2004-07-06 Alexza Molecular Delivery Corporation Delivery of rizatriptan and zolmitriptan through an inhalation route
EP1392258B1 (en) 2001-05-24 2008-11-26 Alexza Pharmaceuticals, Inc. Delivery of alprazolam, estazolam, midazolam or triazolam through an inhalation route
US7498019B2 (en) 2001-05-24 2009-03-03 Alexza Pharmaceuticals, Inc. Delivery of compounds for the treatment of headache through an inhalation route
US7090830B2 (en) * 2001-05-24 2006-08-15 Alexza Pharmaceuticals, Inc. Drug condensation aerosols and kits
WO2002100357A1 (en) * 2001-06-12 2002-12-19 Trial Corporation Cosmetic
AU2002322295C1 (en) * 2001-06-21 2008-12-18 Altus Pharmaceuticals Inc. Spherical protein particles and methods of making and using them
US6730772B2 (en) * 2001-06-22 2004-05-04 Venkatram P. Shastri Degradable polymers from derivatized ring-opened epoxides
GB0208742D0 (en) 2002-04-17 2002-05-29 Bradford Particle Design Ltd Particulate materials
US6921390B2 (en) * 2001-07-23 2005-07-26 Boston Scientific Scimed, Inc. Long-term indwelling medical devices containing slow-releasing antimicrobial agents and having a surfactant surface
DE10137102A1 (en) * 2001-07-30 2003-02-27 Deutsches Krebsforsch Polyvalent vaccine against diseases caused by papillomaviruses, processes for their production and their use
GB0118689D0 (en) * 2001-08-01 2001-09-19 Psimedica Ltd Pharmaceutical formulation
AU2002333644A1 (en) 2001-09-17 2003-04-01 Glaxo Group Limited Dry powder medicament formulations
EP1446104B2 (en) * 2001-11-01 2011-08-03 Novartis AG Spray drying methods
AU2002366267B2 (en) * 2001-11-19 2007-05-10 Becton, Dickinson And Company Pharmaceutical compositions in particulate form
PT1455755E (en) * 2001-11-20 2013-06-18 Civitas Therapeutics Inc Improved particulate compositions for pulmonary delivery
US20030129250A1 (en) * 2001-11-20 2003-07-10 Advanced Inhalation Research Inc. Particulate compositions for improving solubility of poorly soluble agents
WO2003043586A2 (en) * 2001-11-20 2003-05-30 Advanced Inhalation Research, Inc. Compositions for sustained action product delivery
US20030168057A1 (en) * 2001-12-14 2003-09-11 Inhale Therapeutic Systems, Inc. Electronically controllable aerosol delivery
JP2005514393A (en) * 2001-12-19 2005-05-19 ネクター セラピューティクス Supplying aminoglycosides to the lung
EP1480691A4 (en) * 2002-03-05 2007-11-28 Univ State Cleveland Agglomerated particles for aerosol drug delivery
AU2002316574B2 (en) * 2002-03-15 2008-05-01 Brandeis University Central airway administration for systemic delivery of therapeutics
US20040063912A1 (en) * 2002-03-15 2004-04-01 The Brigham And Women's Hospital, Inc. Central airway administration for systemic delivery of therapeutics
US7008644B2 (en) * 2002-03-20 2006-03-07 Advanced Inhalation Research, Inc. Method and apparatus for producing dry particles
US20050163725A1 (en) * 2002-03-20 2005-07-28 Blizzard Charles D. Method for administration of growth hormone via pulmonary delivery
JP2005521695A (en) * 2002-03-20 2005-07-21 アドバンスト インハレーション リサーチ,インコーポレイテッド Method of administering growth hormone by pulmonary delivery
ES2300568T3 (en) 2002-03-20 2008-06-16 Mannkind Corporation INHALATION APPARATUS
WO2003079992A2 (en) 2002-03-20 2003-10-02 Advanced Inhalation Research, Inc. Pulmonary delivery for levodopa
WO2003087189A1 (en) * 2002-04-05 2003-10-23 Valorisation - Recherche, Societe En Commandite Functionalized polymers and their biomedical and pharmaceutical uses
US20040038303A1 (en) * 2002-04-08 2004-02-26 Unger Gretchen M. Biologic modulations with nanoparticles
US7582284B2 (en) * 2002-04-17 2009-09-01 Nektar Therapeutics Particulate materials
GB0216562D0 (en) * 2002-04-25 2002-08-28 Bradford Particle Design Ltd Particulate materials
AU2003243191B2 (en) 2002-05-02 2007-09-20 President And Fellows Of Harvard College Formulations limiting spread of pulmonary infections
US9339459B2 (en) 2003-04-24 2016-05-17 Nektar Therapeutics Particulate materials
WO2003094900A1 (en) 2002-05-13 2003-11-20 Alexza Molecular Delivery Corporation Delivery of drug amines through an inhalation route
US6930137B2 (en) * 2002-05-31 2005-08-16 Fina Technology, Inc. Method of improving blown film processing performance and physical properties
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
US7425618B2 (en) * 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
CA2494485A1 (en) * 2002-07-25 2004-02-05 Medimmune, Inc. Methods of treating and preventing rsv, hmpv, and piv using anti-rsv, anti-hmpv, and anti-piv antibodies
DE10234165B4 (en) * 2002-07-26 2008-01-03 Advanced Micro Devices, Inc., Sunnyvale A method of filling a trench formed in a substrate with an insulating material
EP1534335B9 (en) 2002-08-14 2016-01-13 Macrogenics, Inc. Fcgammariib-specific antibodies and methods of use thereof
WO2004030659A1 (en) * 2002-09-30 2004-04-15 Acusphere, Inc. Sustained release porous microparticles for inhalation
EP1551876B1 (en) 2002-10-16 2011-03-16 Purdue Pharma L.P. Antibodies that bind cell-associated ca 125/0722p and methods of use thereof
US6800663B2 (en) * 2002-10-18 2004-10-05 Alkermes Controlled Therapeutics Inc. Ii, Crosslinked hydrogel copolymers
AU2003294470B2 (en) 2002-11-26 2009-09-17 Alexza Pharmaceuticals, Inc. Use of loxapine and amoxapine for the manufacture of a medicament for the treatment of pain
US20040105818A1 (en) * 2002-11-26 2004-06-03 Alexza Molecular Delivery Corporation Diuretic aerosols and methods of making and using them
US7913688B2 (en) * 2002-11-27 2011-03-29 Alexza Pharmaceuticals, Inc. Inhalation device for producing a drug aerosol
US20040206350A1 (en) * 2002-12-19 2004-10-21 Nektar Therapeutics Aerosolization apparatus with non-circular aerosolization chamber
MXPA05007154A (en) 2002-12-30 2005-09-21 Nektar Therapeutics Prefilming atomizer.
WO2004060903A2 (en) * 2002-12-31 2004-07-22 Nektar Therapeutics Aerosolizable pharmaceutical formulation for fungal infection therapy
TR200502522T2 (en) * 2002-12-31 2005-09-21 Nektar Therapeutics Pharmaceutical formulation containing an insoluble active ingredient
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP2272533A1 (en) 2003-01-13 2011-01-12 MacroGenics, Inc. Soluble FcyR fusion proteins and methods of use thereof
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
WO2004073729A1 (en) * 2003-02-21 2004-09-02 Translational Research Ltd. Compositions for nasal administration of drug
AU2003220808B2 (en) 2003-03-27 2008-08-21 Bioactis Limited Powder medicine applicator for nasal cavity
PL1610850T3 (en) * 2003-04-09 2012-11-30 Novartis Ag Aerosolization apparatus with air inlet shield
US8869794B1 (en) 2003-04-09 2014-10-28 Novartis Pharma Ag Aerosolization apparatus with capsule puncturing member
KR101224235B1 (en) 2003-04-11 2013-01-25 메디뮨 엘엘씨 Recombinant IL-9 Antibodies and Uses Thereof
WO2004104490A1 (en) 2003-05-21 2004-12-02 Alexza Pharmaceuticals, Inc. Self-contained heating unit and drug-supply unit employing same
GB0313604D0 (en) * 2003-06-12 2003-07-16 Britannia Pharmaceuticals Ltd Delivery device for powdered medicament
ES2872350T3 (en) * 2003-06-13 2021-11-02 Civitas Therapeutics Inc Low Dose Pharmaceutical Powders For Inhalation
WO2004112799A1 (en) * 2003-06-13 2004-12-29 Chrysalis Technologies Incorporated Methods and apparatus for producing nanoscale particles
US20050032173A1 (en) * 2003-08-05 2005-02-10 Mauricio Rojas Fusion proteins with a membrane translocating sequence and methods of using same to inhibit an immune response
ATE420660T1 (en) 2003-08-08 2009-01-15 Amgen Fremont Inc ANTIBODIES TO PARATH HORMONE (PTH) AND THEIR USES
US7318925B2 (en) * 2003-08-08 2008-01-15 Amgen Fremont, Inc. Methods of use for antibodies against parathyroid hormone
EP2272566A3 (en) 2003-08-18 2013-01-02 MedImmune, LLC Humanisation of antibodies
US20060228350A1 (en) * 2003-08-18 2006-10-12 Medimmune, Inc. Framework-shuffling of antibodies
EP1663159A4 (en) * 2003-09-10 2010-06-09 Map Pharmaceuticals Inc Aerosol formulations for delivery of dihydroergotamine to the systemic circulation via pulmonary inhalation
GB0327723D0 (en) * 2003-09-15 2003-12-31 Vectura Ltd Pharmaceutical compositions
US7943179B2 (en) * 2003-09-23 2011-05-17 Massachusetts Institute Of Technology pH triggerable polymeric particles
CA2533887A1 (en) * 2003-09-30 2005-04-14 Acusphere, Inc. Injectable, oral, or topical sustained release pharmaceutical formulations
US20050181059A1 (en) * 2003-09-30 2005-08-18 Spherics, Inc. Nanoparticulate therapeutic biologically active agents
US7338171B2 (en) * 2003-10-27 2008-03-04 Jen-Chuen Hsieh Method and apparatus for visual drive control
US20050214224A1 (en) * 2003-11-04 2005-09-29 Nektar Therapeutics Lipid formulations for spontaneous drug encapsulation
JP2007517892A (en) * 2004-01-12 2007-07-05 マンカインド コーポレイション Methods for reducing serum proinsulin levels in type 2 diabetes
WO2005078848A2 (en) * 2004-02-11 2005-08-25 University Of Tennessee Research Foundation Inhibition of tumor growth and invasion by anti-matrix metalloproteinase dnazymes
US20050207983A1 (en) 2004-03-05 2005-09-22 Pulmatrix, Inc. Formulations decreasing particle exhalation
US20080090753A1 (en) * 2004-03-12 2008-04-17 Biodel, Inc. Rapid Acting Injectable Insulin Compositions
US7279457B2 (en) * 2004-03-12 2007-10-09 Biodel, Inc. Rapid acting drug delivery compositions
WO2005110379A2 (en) * 2004-05-07 2005-11-24 President And Fellows Of Harvard College Pulmonary malarial vaccine
US7723306B2 (en) 2004-05-10 2010-05-25 Boehringer Ingelheim Pharma Gmbh & Co. Kg Spray-dried powder comprising at least one 1,4 O-linked saccharose-derivative and methods for their preparation
US7611709B2 (en) 2004-05-10 2009-11-03 Boehringer Ingelheim Pharma Gmbh And Co. Kg 1,4 O-linked saccharose derivatives for stabilization of antibodies or antibody derivatives
US7727962B2 (en) 2004-05-10 2010-06-01 Boehringer Ingelheim Pharma Gmbh & Co. Kg Powder comprising new compositions of oligosaccharides and methods for their preparation
JP2007537833A (en) * 2004-05-20 2007-12-27 ディスカバリー ラボラトリーズ,インコーポレイテッド Method, system and apparatus for non-invasive lung inhalation
US7540286B2 (en) * 2004-06-03 2009-06-02 Alexza Pharmaceuticals, Inc. Multiple dose condensation aerosol devices and methods of forming condensation aerosols
WO2006002140A2 (en) * 2004-06-21 2006-01-05 Nektar Therapeutics Compositions comprising amphotericin b
US8513204B2 (en) * 2004-06-21 2013-08-20 Novartis Ag Compositions comprising amphotericin B, mehods and systems
KR20070054644A (en) * 2004-07-26 2007-05-29 액테리온 파마슈티칼 리미티드 Treatment of pulmonary hypertension by inhaled iloprost with a microparticle formulation
US8673360B2 (en) * 2004-08-10 2014-03-18 Shin Nippon Biomedical Laboratories, Ltd. Compositions that enable rapid-acting and highly absorptive intranasal administration
US20100006092A1 (en) * 2004-08-12 2010-01-14 Alexza Pharmaceuticals, Inc. Aerosol Drug Delivery Device Incorporating Percussively Activated Heat Packages
US7581540B2 (en) * 2004-08-12 2009-09-01 Alexza Pharmaceuticals, Inc. Aerosol drug delivery device incorporating percussively activated heat packages
MX2007001903A (en) 2004-08-20 2007-08-02 Mannkind Corp Catalysis of diketopiperazine synthesis.
KR101306384B1 (en) * 2004-08-23 2013-09-09 맨카인드 코포레이션 Diketopiperazine salts, diketomorpholine salts or diketodioxane salts for drug delivery
EP3073267A1 (en) 2004-09-21 2016-09-28 Medimmune, Inc. Antibodies against and methods for producing vaccines for respiratory syncytial virus
EP1812068A4 (en) * 2004-10-29 2010-06-09 Medimmune Inc Methods of preventing and treating rsv infections and related conditions
DE102004053373A1 (en) * 2004-11-02 2006-05-04 Justus-Liebig-Universität Giessen Invention relating to nano / meso-fiber anisometric particles in the form of nano / meso-fiber tubes, cables and their curved or branched modifications
US8337815B2 (en) * 2004-12-23 2012-12-25 Discovery Laboratories, Inc. Pulmonary surfactant formulations
US8627821B2 (en) 2005-01-10 2014-01-14 Pulmatrix, Inc. Method and device for decreasing contamination
EP1848410A4 (en) * 2005-02-17 2011-11-09 Medivas Llc Polymer particle delivery compositions and methods of use
JP5153613B2 (en) 2005-03-18 2013-02-27 メディミューン,エルエルシー Antibody framework shuffle
WO2008157379A2 (en) 2007-06-21 2008-12-24 Macrogenics, Inc. Covalent diabodies and uses thereof
CA2605024C (en) 2005-04-15 2018-05-22 Macrogenics, Inc. Covalent diabodies and uses thereof
US8028697B2 (en) 2005-04-28 2011-10-04 Trudell Medical International Ventilator circuit and method for the use thereof
EP2402001A1 (en) * 2005-05-18 2012-01-04 Pulmatrix, Inc. Formulations for alteration of biophysical properties of mucosal lining
US20110177996A1 (en) * 2005-05-23 2011-07-21 Children's Hospital Medical Center Regulatory proteins in lung repair and treatment of lung disease
US20070031342A1 (en) * 2005-06-22 2007-02-08 Nektar Therapeutics Sustained release microparticles for pulmonary delivery
KR20080025174A (en) 2005-06-23 2008-03-19 메디뮨 인코포레이티드 Antibody formulations having optimized aggregation and fragmentation profiles
US7700739B2 (en) * 2005-06-30 2010-04-20 Abbott Laboratories IL-12/p40 binding proteins
US7919583B2 (en) * 2005-08-08 2011-04-05 Discovery Genomics, Inc. Integration-site directed vector systems
SI2573114T1 (en) 2005-08-10 2016-08-31 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
RU2008108510A (en) * 2005-08-11 2009-09-20 Президент Энд Феллоуз Гарвард Колледж (Us) METHODS AND COMPOSITIONS OF DRIED CELL FORMS
EP2500356A3 (en) 2005-08-19 2012-10-24 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
RU2515108C2 (en) 2005-08-19 2014-05-10 Эббви Инк Immunoglobulin with double variable domains and its applications
US7612181B2 (en) * 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20090215992A1 (en) * 2005-08-19 2009-08-27 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
JP5465878B2 (en) 2005-09-14 2014-04-09 マンカインド コーポレイション Method of drug formulation based on increasing the affinity of crystalline microparticle surfaces for active agents
WO2007038246A2 (en) * 2005-09-22 2007-04-05 Medivas, Llc Solid polymer delivery compositions and methods for use thereof
WO2007035938A2 (en) 2005-09-22 2007-03-29 Medivas, Llc BIS-(α-AMINO)-DIOL-DIESTER-CONTAINING POLY(ESTER AMIDE) AND POLY(ESTER URETHANE) COMPOSITIONS AND METHODS OF USE
WO2007036501A2 (en) * 2005-09-26 2007-04-05 Basf Aktiengesellschaft Graft copolymer and use of the same
AU2006297394B9 (en) * 2005-09-29 2013-09-19 Novartis Ag Receptacles and kits, such as for dry powder packaging
US20070086952A1 (en) * 2005-09-29 2007-04-19 Biodel, Inc. Rapid Acting and Prolonged Acting Inhalable Insulin Preparations
US7713929B2 (en) 2006-04-12 2010-05-11 Biodel Inc. Rapid acting and long acting insulin combination formulations
US8084420B2 (en) * 2005-09-29 2011-12-27 Biodel Inc. Rapid acting and long acting insulin combination formulations
US8906864B2 (en) * 2005-09-30 2014-12-09 AbbVie Deutschland GmbH & Co. KG Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
WO2007053923A2 (en) * 2005-11-11 2007-05-18 Biolab Sanus Farmacêutica Ltda. Solid pharmaceutical composition comprising agglomerate nanoparticles and a process for producing the same
EP1957113A4 (en) * 2005-11-21 2011-11-09 Medivas Llc Polymer particles for delivery of macromolecules and methods of use
AU2006319358B2 (en) * 2005-11-30 2012-01-19 AbbVie Deutschland GmbH & Co. KG Anti-Abeta globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
KR101439828B1 (en) 2005-11-30 2014-09-17 애브비 인코포레이티드 Monoclonal antibodies against amyloid beta protein and uses thereof
JP2009524584A (en) * 2005-12-07 2009-07-02 メディバス エルエルシー Method for building a polymer-biologic delivery composition
CA2631492A1 (en) * 2005-12-15 2007-06-21 Acusphere, Inc. Processes for making particle-based pharmaceutical formulations for oral administration
CA2631493A1 (en) * 2005-12-15 2007-06-21 Acusphere, Inc. Processes for making particle-based pharmaceutical formulations for pulmonary or nasal administration
CN104383546B (en) 2006-02-22 2021-03-02 曼金德公司 Method for improving the pharmaceutical properties of microparticles comprising diketopiperazines and an active agent
US8968702B2 (en) * 2006-03-30 2015-03-03 Duke University Inhibition of HIF-1 activation for anti-tumor and anti-inflammatory responses
WO2007123793A2 (en) * 2006-04-04 2007-11-01 Stc.Unm Swellable particles for drug delivery
WO2007121256A2 (en) * 2006-04-12 2007-10-25 Biodel, Inc. Rapid acting and long acting insulin combination formulations
US20090317335A1 (en) * 2006-04-20 2009-12-24 Wenbin Lin Hybrid Nanomaterials as Multimodal Imaging Contrast Agents
JP5445130B2 (en) * 2006-05-02 2014-03-19 メディバス エルエルシー Delivery of ophthalmic drugs to the exterior or interior of the eye
EP2021141A4 (en) * 2006-05-09 2013-07-03 Medivas Llc Biodegradable water soluble polymers
LT2029173T (en) 2006-06-26 2016-11-10 Macrogenics, Inc. Fc riib-specific antibodies and methods of use thereof
EP2064243A2 (en) 2006-08-28 2009-06-03 Kyowa Hakko Kirin Co., Ltd. Antagonistic human light-specific human monoclonal antibodies
ES2902063T3 (en) 2006-09-08 2022-03-24 Abbvie Bahamas Ltd Interleukin-13 binding proteins
JP2010506842A (en) 2006-10-16 2010-03-04 メディミューン,エルエルシー Molecules with reduced half-life, compositions thereof and uses
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
ES2652415T3 (en) * 2006-12-26 2018-02-02 Shin Nippon Biomedical Laboratories, Ltd. Preparation for transnasal application
PL2148691T3 (en) 2007-02-05 2015-12-31 Apellis Pharmaceuticals Inc Compstatin analogues for use in the treatment of inflammatory conditions of the respiratory system
KR20090129998A (en) 2007-02-11 2009-12-17 맵 파마슈티컬스, 인코포레이티드 Method of therapeutic administration of dhe to enable rapid relief of migraine while minimizing side effect profile
WO2008104386A2 (en) * 2007-02-27 2008-09-04 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
EP2121088B1 (en) 2007-03-09 2016-07-13 Alexza Pharmaceuticals, Inc. Heating unit for use in a drug delivery device
WO2008124522A2 (en) * 2007-04-04 2008-10-16 Biodel, Inc. Amylin formulations
WO2008137747A1 (en) 2007-05-02 2008-11-13 The Regents Of The University Of Michigan Nanoemulsion therapeutic compositions and methods of using the same
EP2164868B1 (en) 2007-05-04 2015-03-25 Technophage, Investigação E Desenvolvimento Em Biotecnologia, SA Engineered rabbit antibody variable domains and uses thereof
PE20090329A1 (en) * 2007-05-30 2009-03-27 Abbott Lab HUMANIZED ANTIBODIES AGAINST GLOBULOMER AB (20-42) AND ITS USES
US20090232801A1 (en) * 2007-05-30 2009-09-17 Abbot Laboratories Humanized Antibodies Which Bind To AB (1-42) Globulomer And Uses Thereof
EP1997830A1 (en) * 2007-06-01 2008-12-03 AIMM Therapeutics B.V. RSV specific binding molecules and means for producing them
WO2009015143A1 (en) * 2007-07-24 2009-01-29 Medivas, Llc Biodegradable cationic polymer gene transfer compositions and methods of use
JP2010535032A (en) 2007-07-31 2010-11-18 メディミューン,エルエルシー Multispecific epitope binding proteins and uses thereof
GB0715376D0 (en) * 2007-08-07 2007-09-19 Smith & Nephew Coating
JP5270884B2 (en) * 2007-09-03 2013-08-21 大阪瓦斯株式会社 Dispersant and polylactic acid based composite resin particles containing the dispersant
EP2033971A1 (en) * 2007-09-06 2009-03-11 Abbott GmbH & Co. KG Bone Morphogenetic Protein (BMP) binding domains of proteins of the Repulsive Guidance Molecule (RGM) protein family and functional fragments thereof and their application
US8785396B2 (en) 2007-10-24 2014-07-22 Mannkind Corporation Method and composition for treating migraines
AU2009204309B2 (en) * 2008-01-04 2012-11-22 Biodel, Inc. Insulin formulations for insulin release as a function of tissue glucose levels
US20110105995A1 (en) * 2008-01-16 2011-05-05 Zhu Ting F Uniform-sized, multi-drug carrying, and photosensitive liposomes for advanced drug delivery
AU2009205995B2 (en) 2008-01-18 2014-04-03 Medimmune, Llc Cysteine engineered antibodies for site-specific conjugation
WO2009139939A2 (en) * 2008-02-22 2009-11-19 The University Of North Carolina At Chapel Hill Hybrid nanoparticles as anti-cancer therapeutic agents and dual therapeutic/imaging contrast agents
US8962803B2 (en) * 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
HUE028653T2 (en) 2008-03-17 2016-12-28 Discovery Lab Inc Ventilation circuit adaptor and proximal aerosol delivery system
SG190572A1 (en) 2008-04-29 2013-06-28 Abbott Lab Dual variable domain immunoglobulins and uses thereof
US20100260668A1 (en) * 2008-04-29 2010-10-14 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
MY159667A (en) 2008-05-09 2017-01-13 Abbvie Inc Antibodies to receptor of advanced glycation end products (rage) and uses thereof
US8834930B2 (en) * 2008-05-15 2014-09-16 Novartis Ag Pulmonary delivery of a fluoroquinolone
RU2010153578A (en) 2008-06-03 2012-07-20 Эбботт Лэборетриз (Us) IMMUNOGLOBULINS WITH DOUBLE VARIABLE DOMAINS AND THEIR APPLICATION
CA2726087A1 (en) * 2008-06-03 2009-12-10 Tariq Ghayur Dual variable domain immunoglobulins and uses thereof
US8485180B2 (en) 2008-06-13 2013-07-16 Mannkind Corporation Dry powder drug delivery system
CN104689432B (en) 2008-06-13 2018-07-06 曼金德公司 Diskus and the system for drug conveying
US8530611B2 (en) 2008-06-20 2013-09-10 Board Of Regents, The University Of Texas System Biodegradable photoluminescent polymers
JP5479465B2 (en) 2008-06-20 2014-04-23 マンカインド コーポレイション Interactive device and method for profiling inhalation efforts in real time
TW201014602A (en) 2008-07-08 2010-04-16 Abbott Lab Prostaglandin E2 binding proteins and uses thereof
RU2011104348A (en) * 2008-07-08 2012-08-20 Эбботт Лэборетриз (Us) IMMUNOGLOBULINS WITH DOUBLE VARIABLE DOMAIN AGAINST PROSTAGLANDINE E2 AND THEIR APPLICATION
WO2010008564A2 (en) * 2008-07-16 2010-01-21 Recombinetics Plaice dna transposon system
TWI494123B (en) 2008-08-11 2015-08-01 Mannkind Corp Use of ultrarapid acting insulin
EP2323671A4 (en) * 2008-08-13 2012-09-26 Medivas Llc Aabb-poly(depsipeptide) biodegradable polymers and methods of use
WO2010036945A2 (en) * 2008-09-26 2010-04-01 The Regents Of The University Of Michigan Nanoemulsion therapeutic compositions and methods of using the same
EP2626098B1 (en) 2008-10-22 2020-08-19 Trudell Medical International Modular aerosol delivery system
WO2010057197A1 (en) 2008-11-17 2010-05-20 The Regents Of The University Of Michigan Cancer vaccine compositions and methods of using the same
BRPI0922807A2 (en) * 2008-12-04 2015-12-22 Abbott Lab double variable domain imonuglobulins and their uses
SI2786762T1 (en) 2008-12-19 2019-07-31 Macrogenics, Inc. Covalent diabodies and uses thereof
US8314106B2 (en) 2008-12-29 2012-11-20 Mannkind Corporation Substituted diketopiperazine analogs for use as drug delivery agents
KR20110110349A (en) * 2009-01-29 2011-10-06 아보트 러보러터리즈 Il-1 binding proteins
US20110165063A1 (en) * 2009-01-29 2011-07-07 Abbott Laboratories Il-1 binding proteins
WO2010087927A2 (en) 2009-02-02 2010-08-05 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
CA2752805A1 (en) * 2009-02-18 2010-08-26 Bayer Pharma Aktiengesellschaft Formulation comprising drospirenone for subcutaneous or intramuscular administration
US8030026B2 (en) * 2009-02-24 2011-10-04 Abbott Laboratories Antibodies to troponin I and methods of use thereof
US9060927B2 (en) * 2009-03-03 2015-06-23 Biodel Inc. Insulin formulations for rapid uptake
EP2810652A3 (en) 2009-03-05 2015-03-11 AbbVie Inc. IL-17 binding proteins
US8283162B2 (en) * 2009-03-10 2012-10-09 Abbott Laboratories Antibodies relating to PIVKAII and uses thereof
PL2405963T3 (en) 2009-03-11 2014-04-30 Mannkind Corp Apparatus, system and method for measuring resistance of an inhaler
DK2413902T3 (en) 2009-03-18 2019-10-07 Incarda Therapeutics Inc Unit doses, aerosols, kits and methods for treating cardiac conditions by pulmonary administration
JP5671001B2 (en) 2009-03-26 2015-02-18 パルマトリックス,インコーポレイテッド Dry powder formulation and method for treating lung disease
WO2010111132A2 (en) 2009-03-27 2010-09-30 Bend Research, Inc. Spray-drying process
DE102009016119A1 (en) * 2009-04-03 2010-10-14 Evonik Degussa Gmbh Nutritional supplement containing alpha-keto acids to support diabetes therapy
GB0908129D0 (en) * 2009-05-12 2009-06-24 Innovata Ltd Composition
WO2010131486A1 (en) * 2009-05-15 2010-11-18 Shin Nippon Biomedical Laboratories, Ltd. Intranasal pharmaceutical compositions with improved pharmacokinetics
AU2010254550B2 (en) 2009-05-27 2015-10-15 Selecta Biosciences, Inc. Targeted synthetic nanocarriers with pH sensitive release of immunomodulatory agents
WO2010141329A1 (en) 2009-06-01 2010-12-09 Medimmune, Llc Molecules with extended half-lives and uses thereof
MY172858A (en) * 2009-06-12 2019-12-12 Mannkind Corp Diketopiperazine microparticles with defined isomer contents
EP2440184B1 (en) 2009-06-12 2023-04-05 MannKind Corporation Diketopiperazine microparticles with defined specific surface areas
WO2011005481A1 (en) 2009-06-22 2011-01-13 Medimmune, Llc ENGINEERED Fc REGIONS FOR SITE-SPECIFIC CONJUGATION
WO2010151804A1 (en) * 2009-06-26 2010-12-29 Map Pharmaceuticals, Inc. Administration of dihydroergotamine mesylate particles using a metered dose inhaler
UY32808A (en) * 2009-07-29 2011-02-28 Abbott Lab IMMUNOGLOBULINS AS A DUAL VARIABLE DOMAIN AND USES OF THE SAME
GB2472327B (en) * 2009-07-31 2013-03-13 Shin Nippon Biomedical Lab Ltd Intranasal granisetron and nasal applicator
US8568719B2 (en) 2009-08-13 2013-10-29 Crucell Holland B.V. Antibodies against human respiratory syncytial virus (RSV) and methods of use
EP3029070A1 (en) 2009-08-29 2016-06-08 AbbVie Inc. Therapeutic dll4 binding proteins
KR20120060877A (en) 2009-09-01 2012-06-12 아보트 러보러터리즈 Dual variable domain immunoglobulins and uses thereof
US20110189183A1 (en) 2009-09-18 2011-08-04 Robert Anthony Williamson Antibodies against candida, collections thereof and methods of use
US8568726B2 (en) 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
US9096877B2 (en) 2009-10-07 2015-08-04 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
AU2010306677B2 (en) 2009-10-15 2013-05-23 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
GB0918450D0 (en) * 2009-10-21 2009-12-09 Innovata Ltd Composition
UY32979A (en) * 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
WO2011053707A1 (en) 2009-10-31 2011-05-05 Abbott Laboratories Antibodies to receptor for advanced glycation end products (rage) and uses thereof
WO2011056889A1 (en) 2009-11-03 2011-05-12 Mannkind Corporation An apparatus and method for simulating inhalation efforts
US8449916B1 (en) 2009-11-06 2013-05-28 Iowa State University Research Foundation, Inc. Antimicrobial compositions and methods
SG181563A1 (en) 2009-12-08 2012-07-30 Abbott Gmbh & Co Kg Monoclonal antibodies against the rgm a protein for use in the treatment of retinal nerve fiber layer degeneration
EP2542582A4 (en) 2010-03-02 2013-12-04 Abbvie Inc Therapeutic dll4 binding proteins
US20110256135A1 (en) 2010-03-17 2011-10-20 Wolfgang Fraunhofer Anti-nerve growth factor (ngf) antibody compositions
CA2796339C (en) 2010-04-15 2020-03-31 Abbott Laboratories Amyloid-beta binding proteins
KR20130066631A (en) 2010-05-06 2013-06-20 노파르티스 아게 Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
ES2949159T3 (en) 2010-05-06 2023-09-26 Novartis Ag Compositions and methods of use for low-density lipoprotein-related protein 6 (LRP6) therapeutic antibodies
SI2571532T1 (en) 2010-05-14 2017-10-30 Abbvie Inc. Il-1 binding proteins
AU2011258156B2 (en) 2010-05-26 2016-11-24 Selecta Biosciences, Inc. Multivalent synthetic nanocarrier vaccines
RU2571331C1 (en) 2010-06-21 2015-12-20 Маннкайнд Корпорейшн Systems and methods for dry powder drug delivery
US20130150286A1 (en) 2010-06-25 2013-06-13 Jean-Claude Sirard Methods and pharmaceutical compositions for the treatment of respiratory tract infections
WO2012006500A2 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
EA027835B1 (en) 2010-07-09 2017-09-29 Круселл Холланд Б.В. Anti-human respiratory syncytial virus (rsv) antibodies and methods of use thereof
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
US9120862B2 (en) 2010-07-26 2015-09-01 Abbott Laboratories Antibodies relating to PIVKA-II and uses thereof
CA2807127C (en) 2010-08-02 2019-02-12 Leslie S. Johnson Covalent diabodies and uses thereof
KR20130100118A (en) 2010-08-03 2013-09-09 아비에 인코포레이티드 Dual variable domain immunoglobulins and uses therof
EP3533803B1 (en) 2010-08-14 2021-10-27 AbbVie Inc. Anti-amyloid-beta antibodies
HUE058226T2 (en) 2010-08-19 2022-07-28 Zoetis Belgium S A Anti-ngf antibodies and their use
GB201013989D0 (en) 2010-08-20 2010-10-06 Univ Southampton Biological materials and methods of using the same
CU24094B1 (en) 2010-08-20 2015-04-29 Novartis Ag ANTIBODIES FOR THE RECEIVER 3 OF THE EPIDERMAL GROWTH FACTOR (HER3)
CA2809433A1 (en) 2010-08-26 2012-03-01 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
EP2448571B1 (en) 2010-08-30 2013-06-12 Pulmatrix, Inc. Respirably dry powder comprising calcium lactate, sodium chloride and leucine
EP2611438B1 (en) 2010-08-30 2020-04-01 Pulmatrix Operating Company, Inc. Dry powder formulations and methods for treating pulmonary diseases
PT2611529T (en) 2010-09-03 2019-05-09 Bend Res Inc Spray-drying method
US9084944B2 (en) 2010-09-03 2015-07-21 Bend Research, Inc. Spray-drying apparatus and methods of using the same
US8815294B2 (en) 2010-09-03 2014-08-26 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives and a carrier material
WO2012040502A1 (en) 2010-09-24 2012-03-29 Bend Research, Inc. High-temperature spray drying process and apparatus
MX354828B (en) 2010-09-29 2018-03-22 Pulmatrix Operating Co Inc Monovalent metal cation dry powders for inhalation.
DK3470057T3 (en) 2010-09-29 2021-11-22 Pulmatrix Operating Co Inc CATIONIC DRY POWDER INCLUDING MAGNESIUM SALT
EP2643353A1 (en) 2010-11-24 2013-10-02 Novartis AG Multispecific molecules
KR20130133247A (en) 2010-12-21 2013-12-06 애브비 인코포레이티드 Il-1-alpha and -beta bispecific dual variable domain immunoglobulins and their use
US20120275996A1 (en) 2010-12-21 2012-11-01 Abbott Laboratories IL-1 Binding Proteins
DK2694402T3 (en) 2011-04-01 2017-07-03 Mannkind Corp BLISTER PACKAGE FOR PHARMACEUTICAL CYLINDER AMPULS
LT2699264T (en) 2011-04-20 2018-07-10 Medimmune, Llc Antibodies and other molecules that bind b7-h1 and pd-1
KR102030531B1 (en) 2011-05-21 2019-10-10 마크로제닉스, 인크. Deimmunized serum-binding domains and their use for extending serum half-life
WO2012174472A1 (en) 2011-06-17 2012-12-20 Mannkind Corporation High capacity diketopiperazine microparticles
MX363606B (en) 2011-06-22 2019-03-28 Apellis Pharmaceuticals Inc Methods of treating chronic disorders with complement inhibitors.
ES2558357T3 (en) 2011-06-23 2016-02-03 Dsm Ip Assets B.V. Micro- or nanoparticles comprising a biodegradable polyesteramide copolymer for use in the delivery of bioactive agents
US9873765B2 (en) 2011-06-23 2018-01-23 Dsm Ip Assets, B.V. Biodegradable polyesteramide copolymers for drug delivery
JP6049712B2 (en) 2011-07-08 2016-12-21 ザ ユニバーシティ オブ ノース カロライナ アット チャペル ヒルThe University Of North Carolina At Chapel Hill Metal bisphosphonate nanoparticles for anti-cancer treatment and imaging and bone disorder treatment
EP2734236A4 (en) 2011-07-13 2015-04-15 Abbvie Inc Methods and compositions for treating asthma using anti-il-13 antibodies
WO2013010955A1 (en) 2011-07-15 2013-01-24 Morphosys Ag Antibodies that are cross-reactive for macrophage migration inhibitory factor (mif) and d-dopachrome tautomerase (d-dt)
AU2012290306B2 (en) 2011-07-29 2017-08-17 Selecta Biosciences, Inc. Synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte (CTL) immune responses
AU2012296613B2 (en) 2011-08-15 2016-05-12 Amplimmune, Inc. Anti-B7-H4 antibodies and their uses
AU2012328885B2 (en) 2011-10-24 2017-08-31 Mannkind Corporation Methods and compositions for treating pain
MX2014004977A (en) 2011-10-24 2014-09-11 Abbvie Inc Immunobinders directed against sclerostin.
AU2012345645B2 (en) 2011-12-02 2017-09-07 Abvacc, Inc. Immunogenic composition comprising panton-valentine leukocidin (PVL) derived polypeptides
EP2788382A2 (en) 2011-12-05 2014-10-15 Novartis AG Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
CN104159924B (en) 2011-12-05 2018-03-16 诺华股份有限公司 The antibody of EGF-R ELISA 3 (HER3)
CA2855570A1 (en) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
US10118958B2 (en) 2011-12-14 2018-11-06 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
AU2012351947B2 (en) 2011-12-16 2017-08-17 Novartis Ag Aerosolization apparatus for inhalation profile-independent drug delivery
JP2015502397A (en) 2011-12-23 2015-01-22 ファイザー・インク Engineered antibody constant regions for site-specific conjugation, and methods and uses therefor
TW201333035A (en) 2011-12-30 2013-08-16 Abbvie Inc Dual specific binding proteins directed against IL-13 and/or IL-17
MY176695A (en) 2012-01-27 2020-08-19 Abbvie Inc Composition and method for the diagnosis and treatment of diseases associated with neurite degeneration
CN107596518B (en) 2012-02-29 2021-04-23 普马特里克斯营业公司 Inhalable dry powder
SI2723323T1 (en) 2012-03-28 2016-04-29 Discovery Laboratories, Inc. Lyophilization of synthetic liposomal pulmonary surfactant
JP6267685B2 (en) 2012-04-13 2018-01-24 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッドGlaxosmithkline Intellectual Property Development Limited Aggregated particles
EP2844227B1 (en) 2012-05-03 2020-11-18 Kala Pharmaceuticals, Inc. Pharmaceutical nanoparticles showing improved mucosal transport
US9827191B2 (en) 2012-05-03 2017-11-28 The Johns Hopkins University Compositions and methods for ophthalmic and/or other applications
JP6360039B2 (en) 2012-05-03 2018-07-18 カラ ファーマシューティカルズ インコーポレイテッド Composition comprising a plurality of coated particles, pharmaceutical composition, pharmaceutical formulation and method of forming the particles
US11596599B2 (en) 2012-05-03 2023-03-07 The Johns Hopkins University Compositions and methods for ophthalmic and/or other applications
ES2703540T3 (en) 2012-06-04 2019-03-11 Novartis Ag Site-specific marking methods and molecules produced in this way
EP2859018B1 (en) 2012-06-06 2021-09-22 Zoetis Services LLC Caninized anti-ngf antibodies and methods thereof
AU2013289957B2 (en) 2012-07-12 2017-02-23 Mannkind Corporation Dry powder drug delivery systems and methods
WO2014011955A2 (en) 2012-07-12 2014-01-16 Abbvie, Inc. Il-1 binding proteins
WO2014066856A1 (en) 2012-10-26 2014-05-01 Mannkind Corporation Inhalable influenza vaccine compositions and methods
CA2890263C (en) 2012-11-01 2020-03-10 Abbvie Inc. Anti-vegf/dll4 dual variable domain immunoglobulins and uses thereof
WO2014074797A1 (en) 2012-11-09 2014-05-15 Civitas Therapeutics, Inc. Ultra low density pulmonary powders
MX2015005874A (en) 2012-11-09 2015-09-10 Pfizer Platelet-derived growth factor b specific antibodies and compositions and uses thereof.
US10034988B2 (en) 2012-11-28 2018-07-31 Fontem Holdings I B.V. Methods and devices for compound delivery
WO2014100439A2 (en) 2012-12-19 2014-06-26 Amplimmune, Inc. B7-h4 specific antibodies, and compositions and methods of use thereof
US9757529B2 (en) 2012-12-20 2017-09-12 Otitopic Inc. Dry powder inhaler and methods of use
US9757395B2 (en) 2012-12-20 2017-09-12 Otitopic Inc. Dry powder inhaler and methods of use
JP6359031B2 (en) 2012-12-21 2018-07-18 メディミューン,エルエルシー Anti-H7CR antibody
US9597339B2 (en) 2013-02-01 2017-03-21 Glialogix, Inc. Compositions and methods for the treatment of neurodegenerative and other diseases
WO2014124258A2 (en) 2013-02-08 2014-08-14 Irm Llc Specific sites for modifying antibodies to make immunoconjugates
CN105143271A (en) 2013-02-08 2015-12-09 Irm责任有限公司 Specific sites for modifying antibodies to make immunoconjugates
WO2014127214A1 (en) 2013-02-15 2014-08-21 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US9688688B2 (en) 2013-02-20 2017-06-27 Kala Pharmaceuticals, Inc. Crystalline forms of 4-((4-((4-fluoro-2-methyl-1H-indol-5-yl)oxy)-6-methoxyquinazolin-7-yl)oxy)-1-(2-oxa-7-azaspiro[3.5]nonan-7-yl)butan-1-one and uses thereof
ES2831625T3 (en) 2013-02-20 2021-06-09 Kala Pharmaceuticals Inc Therapeutic compounds and their uses
US9498532B2 (en) 2013-03-13 2016-11-22 Novartis Ag Antibody drug conjugates
JP2016512241A (en) 2013-03-14 2016-04-25 アボット・ラボラトリーズAbbott Laboratories HCVNS3 recombinant antigen for improved antibody detection and mutants thereof
EP2968526A4 (en) 2013-03-14 2016-11-09 Abbott Lab Hcv antigen-antibody combination assay and methods and compositions for use therein
JP6739329B2 (en) 2013-03-14 2020-08-12 アボット・ラボラトリーズAbbott Laboratories HCV core lipid binding domain monoclonal antibody
EP2970479B1 (en) 2013-03-14 2019-04-24 Novartis AG Antibodies against notch 3
US9469686B2 (en) 2013-03-15 2016-10-18 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
CN105007950B (en) 2013-03-15 2019-01-15 诺华股份有限公司 Antibody drug conjugate
EP3587404B1 (en) 2013-03-15 2022-07-13 MannKind Corporation Microcrystalline diketopiperazine compositions, methods for preparation and use thereof
WO2014144280A2 (en) 2013-03-15 2014-09-18 Abbvie Inc. DUAL SPECIFIC BINDING PROTEINS DIRECTED AGAINST IL-1β AND / OR IL-17
WO2014165303A1 (en) 2013-04-01 2014-10-09 Pulmatrix, Inc. Tiotropium dry powders
CN105473133A (en) 2013-04-30 2016-04-06 欧缇托匹克公司 Dry powder formulations and methods of use
AU2014268298B2 (en) 2013-05-24 2019-01-17 Medlmmune, Llc Anti-B7-H5 antibodies and their uses
WO2014194293A1 (en) 2013-05-30 2014-12-04 Amplimmune, Inc. Improved methods for the selection of patients for pd-1 or b7-h4 targeted therapies, and combination therapies thereof
SG11201509982UA (en) 2013-06-06 2016-04-28 Igenica Biotherapeutics Inc
ES2753419T3 (en) 2013-06-07 2020-04-08 Univ Duke Complement factor H inhibitors
PT3010535T (en) 2013-06-19 2020-07-03 Integrated Biotherapeutics Inc Toxoid peptides derived from phenol soluble modulin, delta toxin, superantigens, and fusions thereof
BR112016000937A8 (en) 2013-07-18 2021-06-22 Mannkind Corp dry powder pharmaceutical formulations, method for making a dry powder formulation and use of a dry powder pharmaceutical formulation
JP2016530930A (en) 2013-08-05 2016-10-06 マンカインド コーポレイション Ventilation device and method
US11384149B2 (en) 2013-08-09 2022-07-12 Macrogenics, Inc. Bi-specific monovalent Fc diabodies that are capable of binding CD32B and CD79b and uses thereof
UA116479C2 (en) 2013-08-09 2018-03-26 Макродженікс, Інк. Bi-specific monovalent fc diabodies that are capable of binding cd32b and cd79b and uses thereof
EP2840091A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific diabodies that are capable of binding gpA33 and CD3 and uses thereof
EP2839842A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific monovalent diabodies that are capable of binding CD123 and CD3 and uses thereof
KR20160055275A (en) 2013-09-17 2016-05-17 유니버시티 헬스 네트워크 Agents directed against a cis rgma/neogenin interaction or lipid rafts and use of the same in the methods of treatment
WO2015042412A1 (en) 2013-09-20 2015-03-26 E-Nicotine Technology. Inc. Devices and methods for modifying delivery devices
US9890173B2 (en) 2013-11-01 2018-02-13 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10287572B2 (en) 2013-11-01 2019-05-14 Regents Of The University Of Minnesota Protein scaffolds and methods of use
JP6426194B2 (en) 2013-11-01 2018-11-21 カラ ファーマシューティカルズ インコーポレイテッド Crystalline forms of therapeutic compounds and uses thereof
JP6590802B2 (en) 2013-11-06 2019-10-16 ザ ユニバーシティ オブ シカゴThe University Of Chicago Nanoscale transporter for delivery or co-delivery of chemotherapeutic drugs, nucleic acids and photosensitizers
SG10201810298VA (en) 2013-11-13 2018-12-28 Pfizer Tumor necrosis factor-like ligand 1a specific antibodies and compositions and uses thereof
CA2931978A1 (en) 2013-12-02 2015-06-11 Abbvie Inc. Compositions and methods for treating osteoarthritis
JP2017504325A (en) 2014-01-14 2017-02-09 インテグレイテッド バイオセラピューティクス,インコーポレイテッド Method for targeting immunological function to bacterial infection site using cell wall targeting domain of bacteriocin
WO2015109212A1 (en) 2014-01-17 2015-07-23 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
WO2015110930A1 (en) 2014-01-24 2015-07-30 Pfizer Inc. Modified interleukin 21 receptor proteins
EP3104880B1 (en) 2014-02-14 2020-03-25 MacroGenics, Inc. Improved methods for the treatment of vascularizing cancers
CA2977083A1 (en) 2014-02-20 2015-08-27 Kambiz Yadidi Dry powder formulations for inhalation
JP6870988B2 (en) 2014-02-24 2021-05-19 セルジーン コーポレイション How to use cereblon activators for neuronal amplification and treatment of central nervous system disorders
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
US20150291689A1 (en) 2014-03-09 2015-10-15 Abbvie, Inc. Compositions and Methods for Treating Rheumatoid Arthritis
KR20160125515A (en) 2014-03-12 2016-10-31 노파르티스 아게 Specific sites for modifying antibodies to make immunoconjugates
TW202214691A (en) 2014-03-21 2022-04-16 美商艾伯維有限公司 Anti-egfr antibodies and antibody drug conjugates
WO2015148905A1 (en) 2014-03-28 2015-10-01 Mannkind Corporation Use of ultrarapid acting insulin
CN106413750B (en) 2014-05-16 2022-04-29 免疫医疗有限责任公司 Molecules with altered neonatal Fc receptor binding with enhanced therapeutic and diagnostic properties
CN113549153A (en) 2014-05-29 2021-10-26 宏观基因有限公司 Trispecific binding molecules and methods of use thereof
TWI693232B (en) 2014-06-26 2020-05-11 美商宏觀基因股份有限公司 Covalently bonded diabodies having immunoreactivity with pd-1 and lag-3, and methods of use thereof
ES2729202T3 (en) 2014-07-16 2019-10-30 Dana Farber Cancer Inst Inc Et Al Inhibition of HER3 in low grade serous ovarian cancers
AU2015296142A1 (en) 2014-07-31 2017-02-23 Otitopic Inc. Dry powder formulations for inhalation
WO2016020791A1 (en) 2014-08-05 2016-02-11 Novartis Ag Ckit antibody drug conjugates
TN2016000577A1 (en) 2014-08-12 2018-04-04 Novartis Ag Anti-cdh6 antibody drug conjugates
SG10201902499VA (en) 2014-09-03 2019-04-29 Genesegues Inc Therapeutic nanoparticles and related compositions, methods and systems
NZ730348A (en) 2014-09-26 2024-03-22 Macrogenics Inc Bi-specific monovalent diabodies that are capable of binding cd19 and cd3, and uses thereof
US10561806B2 (en) 2014-10-02 2020-02-18 Mannkind Corporation Mouthpiece cover for an inhaler
EP3204039B1 (en) 2014-10-10 2022-06-08 The Regents Of The University Of Michigan Nanoemulsion compositions for preventing, suppressing or eliminating allergic and inflammatory disease
US10806694B2 (en) 2014-10-14 2020-10-20 The University Of Chicago Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
JP6731404B2 (en) 2014-10-14 2020-07-29 ザ ユニバーシティ オブ シカゴThe University Of Chicago Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiation therapy, chemotherapy, immunotherapy, and any combination thereof
MX2017005692A (en) 2014-10-31 2017-08-07 Glaxosmithkline Ip Dev Ltd Powder formulation.
CA2962719A1 (en) 2014-10-31 2016-05-06 Bend Research Inc. Process for forming active domains dispersed in a matrix
MA40910A (en) 2014-11-07 2017-09-12 Civitas Therapeutics Inc RAPAMYCIN POWDERS FOR PULMONARY ADMINISTRATION
EP3229838B1 (en) 2014-12-11 2020-09-09 Pierre Fabre Medicament Anti-c10orf54 antibodies and uses thereof
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
JP6847835B2 (en) 2014-12-12 2021-03-24 ザ ジェネラル ホスピタル コーポレイション Treatment of breast cancer brain metastases
WO2016097297A1 (en) 2014-12-18 2016-06-23 Dsm Ip Assets B.V. Drug delivery system for delivery of acid sensitive drugs
TW201639596A (en) 2015-01-24 2016-11-16 艾伯維有限公司 Compositions and methods for treating psoriatic arthritis
AU2015380455A1 (en) 2015-01-26 2017-08-03 Macrogenics, Inc. Multivalent molecules comprising DR5-binding domains
TW201632202A (en) 2015-01-30 2016-09-16 諾華公司 Treatment of breast cancer
WO2016139482A1 (en) 2015-03-03 2016-09-09 Kymab Limited Antibodies, uses & methods
HUE048284T2 (en) 2015-05-29 2020-07-28 Abbvie Inc Anti-cd40 antibodies and uses thereof
TWI773646B (en) 2015-06-08 2022-08-11 美商宏觀基因股份有限公司 Lag-3-binding molecules and methods of use thereof
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
WO2016203432A1 (en) 2015-06-17 2016-12-22 Novartis Ag Antibody drug conjugates
JP2018524346A (en) 2015-07-02 2018-08-30 サイヴィタス セラピューティックス,インコーポレイテッド Triptan powder for pulmonary delivery
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
CA3025896A1 (en) 2015-07-23 2017-01-26 The Regents Of The University Of California Antibodies to coagulation factor xia and uses thereof
KR20180034588A (en) 2015-07-30 2018-04-04 마크로제닉스, 인크. PD-1-binding molecules and methods for their use
US20170073420A1 (en) 2015-09-11 2017-03-16 Abbvie Inc. Methods for treating relapsing forms of multiple sclerosis
US9862760B2 (en) 2015-09-16 2018-01-09 Novartis Ag Polyomavirus neutralizing antibodies
CN117503905A (en) 2015-10-07 2024-02-06 阿佩利斯制药有限公司 dosing regimen
WO2017062619A2 (en) 2015-10-08 2017-04-13 Macrogenics, Inc. Combination therapy for the treatment of cancer
KR20180064541A (en) 2015-10-23 2018-06-14 화이자 인코포레이티드 Anti-IL-2 antibodies and compositions and uses thereof
MA44334A (en) 2015-10-29 2018-09-05 Novartis Ag ANTIBODY CONJUGATES INCLUDING A TOLL-TYPE RECEPTOR AGONIST
KR20180100122A (en) 2015-12-02 2018-09-07 주식회사 에스티사이언스 Antibodies specific for glycated BTLA (B- and T-lymphocyte weakening factor)
AU2016365318B2 (en) 2015-12-02 2024-04-18 Board Of Regents, The University Of Texas System Antibodies and molecules that immunospecifically bind to BTN1A1 and the therapeutic uses thereof
CR20180318A (en) 2015-12-14 2018-09-19 Macrogenics Inc BISPECIFIC MOLECULES THAT HAVE IMMUNORREACTIVITY WITH PD-1 AND CTLA-4, AND METHODS OF USE OF THE SAME
EP3851457A1 (en) 2016-01-21 2021-07-21 Novartis AG Multispecific molecules targeting cll-1
MX2018009248A (en) 2016-02-01 2019-01-21 Incarda Therapeutics Inc Combining electronic monitoring with inhaled pharmacological therapy to manage cardiac arrhythmias including atrial fibrillation.
UY37127A (en) 2016-02-17 2017-08-31 Macrogenics Inc MOLECULES OF UNION TO ROR1, AND METHODS OF USE OF THE SAME
LT3423105T (en) * 2016-03-02 2021-09-10 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
BR112018070199A2 (en) 2016-04-01 2019-01-29 Therapeuticsmd Inc pharmaceutical composition of steroid hormone
US10286077B2 (en) 2016-04-01 2019-05-14 Therapeuticsmd, Inc. Steroid hormone compositions in medium chain oils
MY198114A (en) 2016-04-15 2023-08-04 Macrogenics Inc Novel b7-h3-binding molecules, antibody drug conjugates thereof and methods of use thereof
AU2017261317A1 (en) 2016-05-05 2018-12-13 Liquidia Technologies, Inc. Dry powder treprostinil for the treatment of pulmonary hypertension
JP7090034B2 (en) 2016-05-20 2022-06-23 ザ ユニバーシティ オブ シカゴ Nanoparticles for chemotherapy, targeted therapy, photodynamic therapy, immunotherapy and any combination thereof
JP2019517480A (en) 2016-06-01 2019-06-24 アッヴィ・インコーポレイテッド Anti-RGMa (Repulsive Guidance Molecule A) antagonist antibody for treating spinal cord injury and pain
CA3026041A1 (en) 2016-06-03 2017-12-07 M et P Pharma AG Nasal pharmaceutical compositions with a porous excipient
WO2017214456A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-cd98 antibodies and antibody drug conjugates
CN109563167A (en) 2016-06-08 2019-04-02 艾伯维公司 Anti- B7-H3 antibody and antibody drug conjugates
WO2017214335A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
TWI784957B (en) 2016-06-20 2022-12-01 英商克馬伯有限公司 Immunocytokines
US10392399B2 (en) 2016-09-08 2019-08-27 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
EP3509421A4 (en) 2016-09-08 2020-05-20 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10253036B2 (en) 2016-09-08 2019-04-09 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
TW201818933A (en) 2016-10-21 2018-06-01 美商吉利亞洛吉克斯公司 Compositions and methods for the treatment of neurodegenerative and other diseases
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
WO2018098370A1 (en) 2016-11-23 2018-05-31 Immunoah Therapeutics, Inc. 4-1bb binding proteins and uses thereof
US10898544B2 (en) 2016-12-13 2021-01-26 Emory University Polypeptides for managing viral infections
JOP20190155A1 (en) 2016-12-21 2019-06-23 Novartis Ag Antibody drug conjugates for ablating hematopoietic stem cells
EP3558368A4 (en) 2016-12-23 2020-12-30 MacroGenics, Inc. Adam9-binding molecules, and methods of use thereof
HUE063346T2 (en) 2017-01-20 2024-01-28 M et P Pharma AG Nasal pharmaceutical compositions for reducing the risks of exposure to air pollutants
US11680101B2 (en) 2017-01-27 2023-06-20 Kymab Limited Anti-OPG antibodies
JOP20190187A1 (en) 2017-02-03 2019-08-01 Novartis Ag Anti-ccr7 antibody drug conjugates
WO2018185618A1 (en) 2017-04-03 2018-10-11 Novartis Ag Anti-cdh6 antibody drug conjugates and anti-gitr antibody combinations and methods of treatment
US11932694B2 (en) 2017-04-19 2024-03-19 Bluefin Biomedicine, Inc. Anti-VTCN1 antibodies and antibody drug conjugates
AR111651A1 (en) 2017-04-28 2019-08-07 Novartis Ag CONJUGATES OF ANTIBODIES THAT INCLUDE TOLL TYPE RECEIVER AGONISTS AND COMBINATION THERAPIES
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
CN110869018A (en) 2017-05-10 2020-03-06 英凯达治疗公司 Unit dose, aerosol, kit and method for treating a cardiac condition by pulmonary administration
JP7210476B2 (en) 2017-05-22 2023-01-23 インスメッド インコーポレイテッド Lipo-glycopeptide cleavable derivatives and uses thereof
EP3630834A1 (en) 2017-05-31 2020-04-08 STCube & Co., Inc. Methods of treating cancer using antibodies and molecules that immunospecifically bind to btn1a1
EP3630835A1 (en) 2017-05-31 2020-04-08 STCube & Co., Inc. Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof
CN110997724A (en) 2017-06-06 2020-04-10 斯特库伯株式会社 Methods of treating cancer using antibodies and molecules that bind BTN1A1 or BTN1A 1-ligands
WO2018232014A1 (en) 2017-06-13 2018-12-20 Integrated Biotherapeutics, Inc. Immunogenic compositions comprising staphylococcus aureus leukocidin luka and lukb derived polypeptides
WO2019028250A1 (en) 2017-08-02 2019-02-07 The University Of Chicago Nanoscale metal-organic layers and metal-organic nanoplates for x-ray induced photodynamic therapy, radiotherapy, rodiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
WO2019049862A1 (en) * 2017-09-05 2019-03-14 味の素株式会社 Polylysine derivative
US11744967B2 (en) 2017-09-26 2023-09-05 Shin Nippon Biomedical Laboratories, Ltd. Intranasal delivery devices
CA3083363A1 (en) 2017-12-01 2019-06-06 Novartis Ag Polyomavirus neutralizing antibodies
KR20200092328A (en) 2017-12-01 2020-08-03 화이자 인코포레이티드 Anti-CXCR5 antibodies and compositions and uses thereof
WO2019156565A1 (en) 2018-02-12 2019-08-15 Fast Forward Pharmaceuticals B.V. Improved antagonistic anti-human cd40 monoclonal antibodies
US20210001062A1 (en) * 2018-03-08 2021-01-07 University Of Washington Devices, systems, and methods for delivery of a reperfusion-injury-modifying drug to a patient
WO2019177690A1 (en) 2018-03-12 2019-09-19 Zoetis Services Llc Anti-ngf antibodies and methods thereof
US10744087B2 (en) 2018-03-22 2020-08-18 Incarda Therapeutics, Inc. Method to slow ventricular rate
AU2019242451A1 (en) 2018-03-29 2020-11-12 Pfizer Inc. LFA3 variants and compositions and uses thereof
US10633458B2 (en) 2018-04-10 2020-04-28 Y-Biologics Inc. Cell engaging binding molecules
US20190314324A1 (en) 2018-04-13 2019-10-17 The University Of Chicago Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
EP3801766A1 (en) 2018-05-31 2021-04-14 Novartis AG Hepatitis b antibodies
AU2019277029C1 (en) 2018-06-01 2024-01-04 Novartis Ag Binding molecules against BCMA and uses thereof
IL305431A (en) 2018-06-01 2023-10-01 Eisai R&D Man Co Ltd Splicing modulator antibody-drug conjugates and methods of use
AR115571A1 (en) 2018-06-20 2021-02-03 Novartis Ag DRUG ANTIBODY CONJUGATES FOR HEMATOPOIETIC STEM CELL ABLATION
CA3111339A1 (en) 2018-07-10 2020-01-16 Rush University Medical Center Use of immunomodulators to control infection and stimulate healing in normal and diabetic wounds
BR112021000934A2 (en) 2018-07-20 2021-04-27 Pierre Fabre Medicament receiver for sight
WO2020023486A1 (en) 2018-07-23 2020-01-30 Trevi Therapeutics, Inc. Treatment of chronic cough, breathlessness and dyspnea
CN112823167A (en) 2018-09-07 2021-05-18 辉瑞大药厂 Anti-alphavbeta 8 antibodies and compositions and uses thereof
WO2020053742A2 (en) 2018-09-10 2020-03-19 Novartis Ag Anti-hla-hbv peptide antibodies
WO2020102454A1 (en) 2018-11-13 2020-05-22 Regents Of The University Of Minnesota Cd40 targeted peptides and uses thereof
WO2020106358A1 (en) 2018-11-20 2020-05-28 Takeda Vaccines, Inc. Novel anti-zika virus antibodies and uses thereof
JP2022512401A (en) 2018-12-13 2022-02-03 エーザイ・アール・アンド・ディー・マネジメント株式会社 Harboxy Diene Splicing Regulatory Antibodies-Drug Conjugates and Their Usage
WO2020128863A1 (en) 2018-12-19 2020-06-25 Novartis Ag Anti-tnf-alpha antibodies
BR112021011900A2 (en) 2018-12-21 2021-09-08 Novartis Ag ANTIBODIES TO PMEL17 AND CONJUGATES THEREOF
EP3911739A1 (en) 2019-01-16 2021-11-24 University Of Rochester Improvement of epithelial or endothelial barrier function
CN113557228A (en) 2019-03-08 2021-10-26 阿比蒂斯有限公司 Site-specific antibody conjugates and antibody-drug conjugates as specific examples thereof
CA3140063A1 (en) 2019-05-20 2020-11-26 Novartis Ag Antibody drug conjugates having linkers comprising hydrophilic groups
US11020384B2 (en) 2019-08-01 2021-06-01 Incarda Therapeutics, Inc. Antiarrhythmic formulation
JP2022549504A (en) 2019-09-26 2022-11-25 エスティーキューブ アンド カンパニー Antibodies specific for glycosylated CTLA-4 and methods of use thereof
WO2021072277A1 (en) 2019-10-09 2021-04-15 Stcube & Co. Antibodies specific to glycosylated lag3 and methods of use thereof
US11572414B2 (en) 2019-11-07 2023-02-07 Eisai R&D Management Co., Ltd. Eribulin antibody-drug conjugates and methods of use
CA3168704A1 (en) 2020-01-24 2021-07-29 Pfizer Inc. Anti-e-selectin antibodies, compositions and methods of use
US20230203191A1 (en) 2020-03-30 2023-06-29 Danisco Us Inc Engineered antibodies
WO2021202463A1 (en) 2020-03-30 2021-10-07 Danisco Us Inc Anti-rsv antibodies
EP4138884A1 (en) 2020-04-20 2023-03-01 Sorrento Therapeutics, Inc. Pulmonary administration of ace2 polypeptides
US20230181756A1 (en) 2020-04-30 2023-06-15 Novartis Ag Ccr7 antibody drug conjugates for treating cancer
CN115461363A (en) 2020-05-01 2022-12-09 诺华股份有限公司 Immunoglobulin variants
EP4143236A1 (en) 2020-05-01 2023-03-08 Novartis AG Engineered immunoglobulins
TW202210504A (en) 2020-05-29 2022-03-16 德國科隆大學 Neutralizing antibodies against sars-related coronavirus
KR20230020441A (en) 2020-06-05 2023-02-10 에자이 알앤드디 매니지먼트 가부시키가이샤 Anti-BCMA antibody-drug conjugates and methods of use
AR123728A1 (en) 2020-10-09 2023-01-04 Pfizer CD1A ANTIBODIES AND THEIR USE
GB202017058D0 (en) 2020-10-27 2020-12-09 Kymab Ltd Antibodies and uses thereof
EP4240494A1 (en) 2020-11-06 2023-09-13 Novartis AG Anti-cd19 agent and b cell targeting agent combination therapy for treating b cell malignancies
EP4240765A2 (en) 2020-11-06 2023-09-13 Novartis AG Antibody fc variants
WO2022097060A1 (en) 2020-11-06 2022-05-12 Novartis Ag Cd19 binding molecules and uses thereof
EP4251648A2 (en) 2020-11-24 2023-10-04 Novartis AG Anti-cd48 antibodies, antibody drug conjugates, and uses thereof
EP4259200A1 (en) 2020-12-11 2023-10-18 Boehringer Ingelheim International GmbH Formulation for multi-purpose application
UY39610A (en) 2021-01-20 2022-08-31 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES
GB202101125D0 (en) 2021-01-27 2021-03-10 Kymab Ltd Antibodies and uses thereof
WO2022221720A1 (en) 2021-04-16 2022-10-20 Novartis Ag Antibody drug conjugates and methods for making thereof
WO2023073599A1 (en) 2021-10-28 2023-05-04 Novartis Ag Engineered fc variants
GB202117111D0 (en) 2021-11-26 2022-01-12 Kymab Ltd Antibodies for use as therapeutics against bacterial infections
EP4190810A1 (en) 2021-12-01 2023-06-07 Universität zu Köln Neutralizing antibodies against sars-related coronavirus
WO2023099688A1 (en) 2021-12-01 2023-06-08 Universität Zu Köln Neutralizing antibodies against sars-related coronavirus
WO2023114375A2 (en) * 2021-12-16 2023-06-22 Alveolus Bio, Inc. Inhalable or ingestible lactic acid compositions for the treatment of chronic lung disease
WO2023209568A1 (en) 2022-04-26 2023-11-02 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2024013727A1 (en) 2022-07-15 2024-01-18 Janssen Biotech, Inc. Material and methods for improved bioengineered pairing of antigen-binding variable regions
WO2024028731A1 (en) 2022-08-05 2024-02-08 Janssen Biotech, Inc. Transferrin receptor binding proteins for treating brain tumors
WO2024028732A1 (en) 2022-08-05 2024-02-08 Janssen Biotech, Inc. Cd98 binding constructs for treating brain tumors

Family Cites Families (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2533065A (en) * 1947-03-08 1950-12-05 George V Taplin Micropulverized therapeutic agents
US2470296A (en) * 1948-04-30 1949-05-17 Abbott Lab Inhalator
US2992645A (en) * 1958-05-06 1961-07-18 Benger Lab Ltd Disperser for powders
US3957965A (en) * 1967-08-08 1976-05-18 Fisons Limited Sodium chromoglycate inhalation medicament
AU439432B2 (en) * 1968-11-28 1972-08-15 Dulux Australia Ltd Polymer and coating composition
US4173488A (en) * 1968-12-23 1979-11-06 Champion International Corporation Oil-in-water emulsions containing hydropholeic starch
US3781230A (en) * 1968-12-23 1973-12-25 Champion Int Corp Microcapsular opacifier system
GB1410588A (en) * 1971-08-10 1975-10-22 Fisons Ltd Composition
US4089800A (en) * 1975-04-04 1978-05-16 Ppg Industries, Inc. Method of preparing microcapsules
US4161516A (en) * 1975-07-25 1979-07-17 Fisons Limited Composition for treating airway disease
US4272398A (en) * 1978-08-17 1981-06-09 The United States Of America As Represented By The Secretary Of Agriculture Microencapsulation process
US4743545A (en) * 1984-08-09 1988-05-10 Torobin Leonard B Hollow porous microspheres containing biocatalyst
US4352883A (en) * 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US4391909A (en) * 1979-03-28 1983-07-05 Damon Corporation Microcapsules containing viable tissue cells
US4276885A (en) * 1979-05-04 1981-07-07 Rasor Associates, Inc Ultrasonic image enhancement
CY1492A (en) * 1981-07-08 1990-02-16 Draco Ab Powder inhalator
EP0072046B1 (en) * 1981-07-24 1986-01-15 FISONS plc Inhalation drugs, methods for their production and pharmaceutical formulations containing them
US5260306A (en) * 1981-07-24 1993-11-09 Fisons Plc Inhalation pharmaceuticals
DE3141641A1 (en) * 1981-10-16 1983-04-28 Schering Ag, 1000 Berlin Und 4619 Bergkamen ULTRASONIC CONTRAST AGENTS AND THEIR PRODUCTION
US4718433A (en) * 1983-01-27 1988-01-12 Feinstein Steven B Contrast agents for ultrasonic imaging
US4572203A (en) * 1983-01-27 1986-02-25 Feinstein Steven B Contact agents for ultrasonic imaging
AU565354B2 (en) * 1983-11-14 1987-09-10 Bio-Mimetics Inc. Bioadhesive compositions and methods of treatment therewith
US4865789A (en) * 1983-11-14 1989-09-12 Akzo Nv Method for making porous structures
US4818542A (en) * 1983-11-14 1989-04-04 The University Of Kentucky Research Foundation Porous microspheres for drug delivery and methods for making same
CA1215922A (en) * 1984-05-25 1986-12-30 Connaught Laboratories Limited Microencapsulation of living tissue and cells
US4679555A (en) * 1984-08-07 1987-07-14 Key Pharmaceuticals, Inc. Method and apparatus for intrapulmonary delivery of heparin
SE459005B (en) * 1985-07-12 1989-05-29 Aake Rikard Lindahl SET TO MANUFACTURE SPHERICAL POLYMER PARTICLES
WO1987003197A1 (en) * 1985-11-29 1987-06-04 Fisons Plc Pharmaceutical composition including sodium cromoglycate
GB8601100D0 (en) * 1986-01-17 1986-02-19 Cosmas Damian Ltd Drug delivery system
US4741872A (en) * 1986-05-16 1988-05-03 The University Of Kentucky Research Foundation Preparation of biodegradable microspheres useful as carriers for macromolecules
US5160745A (en) * 1986-05-16 1992-11-03 The University Of Kentucky Research Foundation Biodegradable microspheres as a carrier for macromolecules
ES2053549T3 (en) * 1986-08-11 1994-08-01 Innovata Biomed Ltd A PROCESS FOR THE PREPARATION OF AN APPROPRIATE PHARMACEUTICAL FORMULATION FOR INHALATION.
ATE76311T1 (en) 1986-08-19 1992-06-15 Genentech Inc DEVICE AND DISPERSION FOR INTRAPULMONARY DELIVERY OF POLYPEPTIDE GROWTH SUBSTANCES AND CYTOKINES.
US5075109A (en) * 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
DE3637926C1 (en) * 1986-11-05 1987-11-26 Schering Ag Ultrasonic manometry in a liquid using microbubbles
JPS63122620A (en) * 1986-11-12 1988-05-26 Sanraku Inc Polylactic acid microsphere and production thereof
US4897256A (en) * 1986-11-25 1990-01-30 Abbott Laboratories LHRH analog formulations
NZ222907A (en) * 1986-12-16 1990-08-28 Novo Industri As Preparation for intranasal administration containing a phospholipid absorption enhancing system
US5204113A (en) * 1987-04-09 1993-04-20 Fisons Plc Pharmaceutical compositions containing pentamidine
US4861627A (en) * 1987-05-01 1989-08-29 Massachusetts Institute Of Technology Preparation of multiwall polymeric microcapsules
GB8712176D0 (en) * 1987-05-22 1987-06-24 Cosmas Damian Ltd Drug delivery system
US5690954A (en) * 1987-05-22 1997-11-25 Danbiosyst Uk Limited Enhanced uptake drug delivery system having microspheres containing an active drug and a bioavailability improving material
US5069936A (en) * 1987-06-25 1991-12-03 Yen Richard C K Manufacturing protein microspheres
US4857311A (en) * 1987-07-31 1989-08-15 Massachusetts Institute Of Technology Polyanhydrides with improved hydrolytic degradation properties
GB8723846D0 (en) * 1987-10-10 1987-11-11 Danbiosyst Ltd Bioadhesive microsphere drug delivery system
US4855144A (en) * 1987-10-23 1989-08-08 Advanced Polymer Systems Synthetic melanin aggregates
IE61591B1 (en) * 1987-12-29 1994-11-16 Molecular Biosystems Inc Concentrated stabilized microbubble-type ultrasonic imaging agent and method of production
JP2670680B2 (en) * 1988-02-24 1997-10-29 株式会社ビーエムジー Polylactic acid microspheres containing physiologically active substance and method for producing the same
AU3073789A (en) * 1988-03-31 1989-10-05 Abbott Laboratories Drug delivery using pulmonary surfactant to facilitate absorption
US5064650A (en) * 1988-04-19 1991-11-12 Southwest Research Institute Controlled-release salt sensitive capsule for oral use and adhesive system
US4917119A (en) * 1988-11-30 1990-04-17 R. J. Reynolds Tobacco Company Drug delivery article
GB8900267D0 (en) 1989-01-06 1989-03-08 Riker Laboratories Inc Narcotic analgesic formulations and apparatus containing same
US4976968A (en) * 1989-02-24 1990-12-11 Clinical Technologies Associates, Inc. Anhydrous delivery systems for pharmacological agents
EP0471036B2 (en) * 1989-05-04 2004-06-23 Southern Research Institute Encapsulation process
US5176132A (en) * 1989-05-31 1993-01-05 Fisons Plc Medicament inhalation device and formulation
US5174988A (en) * 1989-07-27 1992-12-29 Scientific Development & Research, Inc. Phospholipid delivery system
CA2066594A1 (en) * 1989-10-03 1991-04-04 Robert P. Eury Erodible macroporous hydrogel particles and preparation thereof
IT1237118B (en) * 1989-10-27 1993-05-18 Miat Spa MULTI-DOSE INHALER FOR POWDER DRUGS.
US5707644A (en) * 1989-11-04 1998-01-13 Danbiosyst Uk Limited Small particle compositions for intranasal drug delivery
US5271961A (en) * 1989-11-06 1993-12-21 Alkermes Controlled Therapeutics, Inc. Method for producing protein microspheres
JP2571874B2 (en) * 1989-11-06 1997-01-16 アルカーメス コントロールド セラピューティクス,インコーポレイテッド Protein microsphere composition
AU634606B2 (en) * 1989-11-06 1993-02-25 Alkermes Controlled Therapeutics, Inc. Method for producing protein microspheres
US5123414A (en) * 1989-12-22 1992-06-23 Unger Evan C Liposomes as contrast agents for ultrasonic imaging and methods for preparing the same
US5352435A (en) * 1989-12-22 1994-10-04 Unger Evan C Ionophore containing liposomes for ultrasound imaging
US5334381A (en) * 1989-12-22 1994-08-02 Unger Evan C Liposomes as contrast agents for ultrasonic imaging and methods for preparing the same
GB9003821D0 (en) * 1990-02-20 1990-04-18 Danbiosyst Uk Diagnostic aid
AU636481B2 (en) * 1990-05-18 1993-04-29 Bracco International B.V. Polymeric gas or air filled microballoons usable as suspensions in liquid carriers for ultrasonic echography
SE9002017D0 (en) * 1990-06-06 1990-06-06 Kabivitrum Ab PROCESS FOR MANUFACTURE OF MATRICES
EP0536235B1 (en) 1990-06-29 1997-01-22 FISONS plc Pressurised aerosol compositions
NZ239802A (en) * 1990-09-21 1993-09-27 Merrell Dow Pharma A superior tasting pharmaceutical composition having porous particles produced through in situ gas generation and a process for its production
US5145684A (en) * 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
GB9107628D0 (en) * 1991-04-10 1991-05-29 Moonbrook Limited Preparation of diagnostic agents
SE9302777D0 (en) 1993-08-27 1993-08-27 Astra Ab Process for conditioning substances
US5327883A (en) * 1991-05-20 1994-07-12 Dura Pharmaceuticals, Inc. Apparatus for aerosolizing powdered medicine and process and using
JP3319754B2 (en) * 1991-06-03 2002-09-03 ニユコメド・イメージング・アクシエセルカペト Improvements in or on contrast agents
ES2158911T5 (en) * 1991-06-10 2009-12-09 Schering Corporation FORMULATIONS IN AEROSOL WITHOUT CHLOROFLUOROCARBONOS.
ATE359842T1 (en) 1991-07-02 2007-05-15 Nektar Therapeutics DISPENSING DEVICE FOR MIST-FORMED MEDICATIONS
US5409688A (en) * 1991-09-17 1995-04-25 Sonus Pharmaceuticals, Inc. Gaseous ultrasound contrast media
US5674471A (en) * 1991-12-12 1997-10-07 Glaxo Group Limited Aerosol formulations containing P134a and salbutamol
NZ246421A (en) 1991-12-18 1996-05-28 Minnesota Mining & Mfg Aerosol formulation containing a drug and a propellant and which is substantially free of surfactant
US5169871A (en) * 1991-12-19 1992-12-08 Hoechst Celanese Corp. Highly porous compressible polymeric powders
ES2181691T5 (en) * 1992-06-11 2007-10-01 Alkermes Controlled Therapeutics, Inc. ERYTHROPOYETINE PROTEIN DISTRIBUTION SYSTEM.
US6582728B1 (en) * 1992-07-08 2003-06-24 Inhale Therapeutic Systems, Inc. Spray drying of macromolecules to produce inhaleable dry powders
GB2269992A (en) * 1992-08-14 1994-03-02 Rh Ne Poulenc Rorer Limited Powder inhalation formulations
DE69332105T2 (en) * 1992-09-29 2003-03-06 Inhale Therapeutic Systems San PULMONAL DELIVERY OF ACTIVE FRAGMENT OF PARATHORMON
GB9221329D0 (en) * 1992-10-10 1992-11-25 Delta Biotechnology Ltd Preparation of further diagnostic agents
WO1994016739A1 (en) * 1993-01-25 1994-08-04 Sonus Pharmaceuticals, Inc. Phase shift colloids as ultrasound contrast agents
US5456917A (en) * 1993-04-12 1995-10-10 Cambridge Scientific, Inc. Method for making a bioerodible material for the sustained release of a medicament and the material made from the method
US5830853A (en) * 1994-06-23 1998-11-03 Astra Aktiebolag Systemic administration of a therapeutic preparation
TW402506B (en) * 1993-06-24 2000-08-21 Astra Ab Therapeutic preparation for inhalation
GB9313650D0 (en) 1993-07-01 1993-08-18 Glaxo Group Ltd Method and apparatus for the formation of particles
GB9313642D0 (en) 1993-07-01 1993-08-18 Glaxo Group Ltd Method and apparatus for the formation of particles
DE4323636A1 (en) * 1993-07-15 1995-01-19 Hoechst Ag Pharmaceutical preparations from coated, poorly water-soluble pharmaceutical substances for inhalation pharmaceutical forms and processes for their preparation
WO1995007072A2 (en) * 1993-09-09 1995-03-16 Schering Aktiengesellschaft Active principles and gas containing microparticles
BR9407686A (en) * 1993-10-01 1997-02-04 Astra Ab Process and apparatus for the treatment of a finely divided powder medicine using the apparatus and agglomerates
MX9603936A (en) * 1994-03-07 1997-05-31 Inhale Therapeutic Syst Methods and compositions for pulmonary delivery of insulin.
GB9412273D0 (en) * 1994-06-18 1994-08-10 Univ Nottingham Administration means
GB9413202D0 (en) 1994-06-30 1994-08-24 Univ Bradford Method and apparatus for the formation of particles
MX9702357A (en) * 1994-09-29 1997-06-28 Andaris Ltd Spray-dried microparticles as therapeutic vehicles.
GB9423419D0 (en) * 1994-11-19 1995-01-11 Andaris Ltd Preparation of hollow microcapsules
GB9501841D0 (en) * 1995-01-31 1995-03-22 Co Ordinated Drug Dev Improvements in and relating to carrier particles for use in dry powder inhalers
US5612053A (en) * 1995-04-07 1997-03-18 Edward Mendell Co., Inc. Controlled release insufflation carrier for medicaments
US5780014A (en) 1995-04-14 1998-07-14 Inhale Therapeutic Systems Method and apparatus for pulmonary administration of dry powder alpha 1-antitrypsin
AU6402096A (en) * 1995-06-29 1997-01-30 Pharmacopeia, Inc. Combinatorial 1,4-benzodiazepin-2,5-dione library
GB9606677D0 (en) * 1996-03-29 1996-06-05 Glaxo Wellcome Inc Process and device
US5855913A (en) 1997-01-16 1999-01-05 Massachusetts Instite Of Technology Particles incorporating surfactants for pulmonary drug delivery
US5874064A (en) * 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US5985309A (en) 1996-05-24 1999-11-16 Massachusetts Institute Of Technology Preparation of particles for inhalation
US6254854B1 (en) 1996-05-24 2001-07-03 The Penn Research Foundation Porous particles for deep lung delivery
USRE37053E1 (en) 1996-05-24 2001-02-13 Massachusetts Institute Of Technology Particles incorporating surfactants for pulmonary drug delivery

Also Published As

Publication number Publication date
US6136295A (en) 2000-10-24
JP2006160767A (en) 2006-06-22
EP0907356A1 (en) 1999-04-14
DE69711626D1 (en) 2002-05-08
DE69711626T2 (en) 2002-10-24
US6942868B2 (en) 2005-09-13
ATE215357T1 (en) 2002-04-15
JP4489716B2 (en) 2010-06-23
WO1997044013A1 (en) 1997-11-27
EP0907356B1 (en) 2002-04-03
JP2000511189A (en) 2000-08-29
CA2255564A1 (en) 1997-11-27
ES2175420T3 (en) 2002-11-16
US6399102B1 (en) 2002-06-04
US5874064A (en) 1999-02-23
JP4315468B2 (en) 2009-08-19
US20020141947A1 (en) 2002-10-03
US20040047811A1 (en) 2004-03-11
US20050158249A1 (en) 2005-07-21
PT907356E (en) 2002-07-31
US6635283B2 (en) 2003-10-21
DK0907356T3 (en) 2002-07-29

Similar Documents

Publication Publication Date Title
CA2255564C (en) Aerodynamically light particles for pulmonary drug delivery
US6503480B1 (en) Aerodynamically light particles for pulmonary drug delivery
US6740310B2 (en) Porous particles comprising excipients for deep lung delivery
US20050244341A1 (en) Aerodynamically light particles for pulmonary drug delivery
US5855913A (en) Particles incorporating surfactants for pulmonary drug delivery
USRE37053E1 (en) Particles incorporating surfactants for pulmonary drug delivery
US6652837B1 (en) Preparation of novel particles for inhalation
EP0954282B1 (en) Preparation of particles for inhalation
US5985309A (en) Preparation of particles for inhalation
EP1089712B1 (en) Large porous particles emitted from an inhaler
US20020159954A1 (en) Aerodynamically light vaccine for active pulmonary immunization
CA2403349C (en) Preparation of particles for inhalation
EP1273290A1 (en) Large porous particles emitted from an inhaler
EP1498115A1 (en) Preparation of particles for inhalation

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20170523