CA2350074C - Aerosols comprising nanoparticle drugs - Google Patents

Aerosols comprising nanoparticle drugs Download PDF

Info

Publication number
CA2350074C
CA2350074C CA002350074A CA2350074A CA2350074C CA 2350074 C CA2350074 C CA 2350074C CA 002350074 A CA002350074 A CA 002350074A CA 2350074 A CA2350074 A CA 2350074A CA 2350074 C CA2350074 C CA 2350074C
Authority
CA
Canada
Prior art keywords
drug
less
nanoparticulate
particles
aerosol
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 - Fee Related
Application number
CA002350074A
Other languages
French (fr)
Other versions
CA2350074A1 (en
Inventor
H. William Bosch
Kevin D. Ostrander
Eugene R. Cooper
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.)
Alkermes Pharma Ireland Ltd
Original Assignee
Elan Pharma International Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Elan Pharma International Ltd filed Critical Elan Pharma International Ltd
Publication of CA2350074A1 publication Critical patent/CA2350074A1/en
Application granted granted Critical
Publication of CA2350074C publication Critical patent/CA2350074C/en
Anticipated expiration legal-status Critical
Expired - Fee Related 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
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • 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
    • 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
    • A61K9/008Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy comprising drug dissolved or suspended in liquid propellant for inhalation via a pressurized metered dose inhaler [MDI]
    • 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
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/146Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic macromolecular compounds
    • 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
    • A61K9/1682Processes
    • A61K9/1694Processes resulting in granules or microspheres of the matrix type containing more than 5% of excipient
    • 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
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Abstract

The invention discloses aqueous dispersions of nanoparticulate aerosol formulations, dry powder nanoparticulate aerosol formulation, propellant-based aerosol formulations, methods of using the formulations in aerosol delivery devices, and methods of making such formulations. The nanoparticles of the aqueous dispersions or dry powder formulations comprise insoluble drug particles having a surface modifier on the surface thereof.

Description

AEROSOLS COMPRISING NANOPARTICLE DRUGS
FIELD OF THE INVENTION
The present invention is directed to aerosol formulations of nanoparticulate drug compositions, and methods of making and using such aerosol formulations.
BACKGROUND OF THE INVENTION
The route of administration of a drug substance can be critical to its pharmacological effectiveness. Various routes of administration exist, and all have their to own advantages and disadvantages. Oral drug delivery of tablets, capsules, liquids, and the like is the most convenient approach to drug delivery, but many drug compounds are not amenable to oral administration. For example, modern protein drugs which are unstable in the acidic gastric environment or which are rapidly degraded by proteolytic enzymes in the digestive tract are poor candidates for oral administration.
Similarly, 15 poorly soluble compounds which do not dissolve rapidly enough to be orally absorbed are likely to be ineffective when given as oral dosage forms. Oral administration can also be undesirable because drugs which are administered orally are generally distributed to all tissues in the body, and not just to the intended site of pharmacological activity.
Alternative types of systemic administration are subcutaneous or intravenous inj ection.
2o This approach avoids the gastrointestinal tract and therefore can be an effective route for delivery of proteins and peptides. However, these routes of administration have a low rate of patient compliance, especially for drugs such as insulin which must be administered one or more times daily. Additional alternative methods of drug delivery have been developed including transdermal, rectal, vaginal, intranasal, and pulinonary 25 delivery.
Nasal drug delivery relies on inhalation of an aerosol through the nose so that active drug substance can reach the nasal mucosa. Drugs intended for systemic activity can be absorbed into the bloodstream because the nasal mucosa is highly vascularized. Alternatively, if the drug is intended to act topically, it is delivered directly 3o to the site of activity and does not have to distribute throughout the body; hence, relatively low doses may be used. Examples of such drugs are decongestants, antihistamines, and anti-inflammatory steroids for seasonal allergic rhinitis.
Pulmonary drug delivery relies on inhalation of an aerosol through the mouth and thmat so that the drug substance can reach the lung. For systemically active drugs, it is desirable for the drug particles to reach the alveolar region of the lung, whereas drugs which act on the smooth muscle of the conducting airways should preferentially deposit in the bronchiole region. Such drugs can include beta-agonists, anticholinergics, and corticosteroids.
1o Devices Used For Nasal and Pulmonary Drus Delivery Drugs intended for intranasal delivery (systemic and local) can be administered as aqueous solutions or suspensions, as solutions or suspensions in halogenated hydrocarbon propellants (pressurized metered-dose inhalers), or as dry powders. Metered-dose spray pumps for aqueous formulations, pMDIs, and DPIs for ~5 nasal delivery, are available from, for example, Valois of America or Pfeiffer of America.
Drugs intended for pulmonary delivery can also be administered as aqueous formulations, as suspensions or solutions in halogenated hydrocarbon propellants, or as dry powders. Aqueous formulations must be aerosolized by liquid nebulizers employing either hydraulic or ultrasonic atomization, propellant-based systems 2o require suitable pressurized metered-dose inhalers (pMDIs), and dry powders require dry powder inhaler devices (DPIs) which are capable of dispersing.the drug substance effectively. For aqueous and other non-pressurized liquid systems, a variety of nebulizers (including small volume nebulizers) are available to aerosolize the formulations. Compressor-driven nebulizers incorporate jet technology and use 25 compressed air to generate the liquid aerosol. Such devices are commercially available from, for example, Healthdyne Technologies, Inc.; Invacare, Inc.; Mountain Medical Equipment, Inc.; Pari Respiratory, Inc.; Mada Medical, Inc.; Puritan-Bennet;
Schuco, Inc., DeVilbiss Health Care, Inc.; and Hospitak, Inc. Ultrasonic nebulizers rely on mechanical energy in the form of vibration of a piezoelectric crystal to generate respirable liquid droplets and are commercially available from, for example, Omron Heathcare, Inc.
and DeVilbiss Health Care, Inc.
A propellant driven inhaler (pMDi) releases a metered dose of medicine upon each actuation. The medicine is formulated as a suspension or solution of a dnzg substance in a suitable propellant such as a halogenated hydrocarbon. pMDIs are described in, for example, Newman, S. P., Aerosols and the Lung, Clarke et al., eds., pp.
197-224 (Butterworths, London, England, 1984).
Dry powder inhalers (DPIs), which involve deaggregation and aerosolization of dry powders, normally rely upon a burst of inspired air that is drawn to through the unit to deliver a drug dosage. Such devices are described in, for example, U.S. Pat. No. 4,807,814, which is directed to a pneumatic powder ejector having a suction stage and an injection stage; SU 628930 (Abstract), describing a hand-held powder disperser having an axial air flow tube; Fox et al., Powder and Bulk Engineering, pages 33-36 (March 1988), describing a venturi eductor having an axial air inlet tube upstream 15 of a venturi restriction; EP 347 779, describing a hand-held powder disperser having a collapsible expansion chamber; and U.S. Pat. No. 5,785,049, directed to dry powder delivery devices for drugs.
Dronlet/Particle Size Determines Deposition Site 2o In developing a therapeutic aerosol, the aerodynamic size distribution of the inhaled particles is the single most important variable in defining the site of droplet or particle deposition in the patient; in short, it will determine whether drug targeting succeeds or fails. See P. Byron, "Aerosol Formulation, Generation, and Delivery Using Nonmetered Systems," Respiratory Drug Delivery, 144-151, 144 (CRC Press, 1989).
25 Thus, a prerequisite in developing a therapeutic aerosol is a preferential particle size. The deposition of inhaled aerosols involves different mechanisms for different size particles.
D. Swift (1980); Parodi et al., "Airborne Particles and Their Pulmonary Deposition," in Scientific Foundations of Respiratory Medicine, Scaddings et al. (eds.), pp.

(W.B. Saunders, Philadelphia, 1981); J. Heyder, "Mechanism of Aerosol Particle Deposition," Chert, 80:820-823 ( 1981 ).
Generally, inhaled particles are subject to deposition by one of two mechanisms: impaction, which usually predominates for larger particles, and sedimentation, which is prevalent for smaller particles. Impaction occurs when the momentum of an inhaled particle is large enough that the particle does not follow the air stream and encounters a physiological surface. In contrast, sedimentation occurs' primarily in the deep lung when very small particles which have traveled with the inhaled air stream encounter physiological surfaces as a result of random diffusion within the air 1o stream. For intranasally administered drug compounds which are inhaled through the nose, it is desirable for the drug to impact directly on the nasal mucosa;
thus, large (ca. S
to 100 Vim) particles or droplets are generally preferred for targeting of nasal delivery.
Pulmonary drug delivery is accomplished by inhalation of an aerosol through the mouth and throat. Particles having aerodynamic diameters of greater than about 5 microns generally do not reach the lung; instead, they tend to impact the back of the throat and are swallowed and possibly orally absorbed. Particles having diameters of about 2 to about 5 microns are small enough to reach the upper- to mid-pulmonary region (conducting airways), but are too large to reach the alveoli. Even smaller particles, i.e., about 0.5 to about 2 microns, are capable of reaching the alveolar region.
Particles having 2o diameters smaller than about 0.5 microns can also be deposited in the alveolar region by sedimentation, although very small particles rnay be exhaled.
Problems with Conventional Aerosol Compositions and Methods Conventional techniques are extremely inefficient in delivering agents to the lung for a variety of reasons. Prior to the present invention, attempts to develop respirable aqueous suspensions of poorly soluble drugs have been largely unsuccessful.
For example, it has been reported that ultrasonic nebulization of a suspension containing fluorescein and latex drug spheres, representing insoluble drug particles, resulted in only 1% aerosolization of the particles, while air jet nebulization resulted in only a fraction of particles being aerosolized. Susan L. Tiano, "Functionality Testing Used to Rationally Assess Performance of a Model Respiratory Solution or Suspension in a Nebulizer,"
1o Dissertation Abstracts International, 56/12-B, pp. 6578 (1995). Another problem encountered with nebulization of liquid formulations prior to the present invention was the long (4-20 min) period of time required for administration of a therapeutic dose. Long administration times are required because conventional liquid formulations for nebulization are very dilute solutions or suspensions of micronized drug substance.
~5 Prolonged administration times are undesirable because they lessen patient compliance and make it difficult to control the dose administered. Lastly, aerosol formulations of micronized drug are not feasible for deep lung delivery of insoluble compounds because the droplets needed to reach the alveolar region (0.5 to 2 microns) are too small to accommodate micronized drug crystals, which are typically 2-3 microns or more in 2o diameter.
Conventional pMDIs are also inefficient in delivering drug substance to the lung. In most cases, pMDIs consist of suspensions of micronized drug substance in halogenated hydrocarbons such as chlorofluorocarbons (CFCs) or hydrofluoroalkanes (IRAs). Actuation of the pMDI results in delivery of a metered dose of drug and 25 propellant, both of which exit the device at high velocities because of the propellant pressures. The high velocity and momentum of the drug particles results in a high degree of oropharyngeal impaction as well as loss to the device used to deliver the agent. These losses lead to variability in therapeutic agent levels and poor therapeutic control. In addition, oropharyngeal deposition of drugs intended for topical administration to the CA 02350074 2001-05-10 -' 15 ~ ~~ °2000 PGTIU599126799 DESC , ,; , , "
r, .
Attorney Docket No.: 029318/0520 conducting airways (such as corticosteroids) can lead to systemic absorption with resultant undesirable side effects. Additionally, conventional micronization (air-j et milling) of pure drug substance can reduce the drug particle size to no less than about 2-3 microns.
Thus, the micronized material typically used in pMDIs is inherently unsuitable for delivery to the alveolar region and is not expected to deposit below the central bronchiole region of the lung.
Prior to the present invention, delivery of dry powders to the lung typically used micronized drug substance. In the dry powder form, micronized substances tend to have substantial interparticle electrostatic attractive forces which prevent the powders from flowing smoothly and generally make them difficult to disperse. Thus, two key challenges to pulmonary delivery of dry powders are the ability of the device to accurately meter the intended dose and the ability of the device to fully disperse the micronized particles. For many devices and formulations, the extent of dispersion is dependent upon the patient's inspiration rate, which itself may be variable and can lead to a variability in the delivered dose.
Delivery of drugs to the nasal mucosa can also be accomplished with aqueous, propellant-based, or dry powder formulations. However, absorption of poorly soluble drugs can be problematic because of mucociliary clearance which transports deposited particles from the nasal mucosa to the throat where they are swallowed. Complete clearance generally occurs within about 15-20 minutes. Thus, poorly soluble drugs which do not dissolve within this time frame are unavailable for either local or systemic activity.
Background Relating to Nanoparticnlate Drugs Non-aerosol nanoparticulate compositions of submicron sized drugs are described in U.S.
Patent No. 5,518,738. This reference describes a composition comprising a crystalline NSA117, polyvinylpyrrolidone as a surface stabilizer, hygroscopic sugar, and sodium Iauryl sulfate. The composition has an effective average particle size of less than about 1000 nm and exhibits greatly reduced gastric irritation following oral administration andlor hastened onset of action.
WO 98/35666 is directed to pharmaceutical formulations of nanoparticulate naproxen compressed into tablets exhibiting an increased rate of dissolution in vivo when compared to conventional micronized naproxen solid dose formulations. The formulations are made by compressing a solid dose form of a nanoparticulate drug formulation comprising into tablets.

SUBSTITUTE SHEET
~~.=, Pr~rited 23 '11 2000 Co red from PC'TIUS99I22799 on 22 ~ '~ 21340' ;1 a P :: .

~1~ 11=2e0y .r~~i.~~5~~mt~~~ 'DESC
Finally, liquid aerosol dispersions comprising nanoparticulate drugs are described in WO
96/25918.
The development of aerosol drug delivery systems has been hampered by the inherent instability of aerosols, the di~culty of formulating dry powder and aqueous aerosols of water-insoluble drugs, and the difficulty of designing an optimal drug particle size for an aerosol drug delivery system. There is a need in the art for aerosols that deliver an optimal dosage of essentially insoluble drugs throughout the respiratory tract or nasal cavity.
The present invention satisfies these needs.

SUBSTITUTE SHEET
~~pr~
anted 23=j 1 2000. ~o E~i #r~m P~CTllIS9912279~ ~~ 22-~ ~ 2pa~ 2 ;' _: . . , ....:r. ... ... _.~. _... .::.. ..: P . . . . .. , 28516-45(S) SUMMARY OF "f~E TNVENrrION
The present:, invention. is cii:r:v~~:.~ted t.o a~queotxs, propellant-based, arid. dry powcxez: ,ac~r~,~:=ical.~ ~.:rf naruop<zrt::ic:ulate compositions' for puLmona~-y aznc~ uu.,~:aa:l. r_u.celife:r~y' in wl~ic:~h essentially every inhaled particle cont.Griras <a.t least one nanoparticulate drug parti-e.le. "fire drug is lz:ighly water-insoluble. F~refer~abi.y' t: he z-z~3zm,~::mt ~::1 c i.a:l.atrv~ ci..rug hay;
an.
effective average pazwticl..e si:,i-: o:f_ <::u~c~m:t. l rn:;_crc7n ox°
less.
This invention is an improvement ~~:fv ~..h;e nanc:.3particul.ate aerosol formulations described ixz U.S. Patent No. 6,264,922 issued July 24, 2001, fox "~.e:rc~7~~;01:~G~.:'c~r~~t~.airuimc~
Nanoparticulate ~ispersior~as" . ~~ca:rr-ae~r~~~;01 preparat::ic>ns of ~~ubmicron sized water-~insolub:).e da:~ug~~ are ~:~escri.bEad in U.S.
Patent No. 5,145,684.
Tn one aspect, the ~:~re sent invent.ican provides an aerosol composition comprising: (a) aggregates of a spray-dried powder comprisirng rzanopar~tic:ulat..e drug particles, whez°ein the nanoparr: icu:f ate c~.z-~a.g ~>~ax:wti c~°::L.e:~
: ~,' i. ) c.~c,mprise a poo:r:ly soluble c:ryst~alli.r~e <:~r~.rg, ~n.m~i.ry~ ;_d a.;<:ol~abi.lity in at least one liquid dispersion meth-um of l::ss than about 10 mg/ml, (ii) at least 50% of: ~~he parv:it~les have a particle :~i2fe of: less than aho~,xt 1(.)00 rkna, <~raci ~~:i i:i;i ~o.sve a surface rnod:ifier adsorbed orz t:: he :3urf~zcc_~ r~hex-eca:fi; ~arvd (b) the aggregates of spray--dried drug parti.c.l~=;::::a aa:°~: less than or equal to about 100 microns in diameter, winerein the dry powder aggregates az_~e formulated into <~rr ~zcrx:~oso1 comfaosit ion .
Tn another aspect, tl~e pres': nt ia~v~exxtion provides an aerosol cornposil=~.on comp:ri4~zra.g: (~~; ~:~ggz"egates of a freeze-dried powder ccampr~_sinc~ rAarr.opa:e-t:.::a.c:u~.ate drug particles, wherein the aggregates of freeze.-dried dru<~ are 28516-45(S) less than or equal to aboaat 100 mi~,~r_r~ns i.n diameter and the narLoparticulate drug part,i.c°le.: ( ~ j comprise a poor=~y soluble crystalline drug, ~iav:i.rncy~ c:a >calubility .a.zn at l.ea.st one liquid dispersion medium of less than about 10 mg/ml, (iii at least SOo of the ~nart:~~:~le~.~ rmavE, a L>2~r.r_l:z.cvle :~_.ze of less than about 1000 nrn, and ~z~.i.) have a :;r.ar_face modifier adsorbed on the surface thereof, wlle~:eim the freeze-dried pow;ier aggregates area fox~mula::~~c~. :Lluto are aero~so composition.
In a further aspect, the present invention provrides an aerosol ccompc>si.t.i<=yrr f:~o u:.r~~ ia:n a pxo:>pellant.-bast~d pMDI comprising: !.~~.) d:cy pc>wdex~ agg:z;egate:s cf particles of a nanoparticulate poorly sulut~le crystalline dru~_l having a solubility iz-a at. 4.ea.St ron~: :I_:i.c~uz.ci dispersion medium of less than about 10 nrg/ml, whereim the aggregates are les~a than or equa:i. tc~ abol*k. 1.~4)c:) rn:z.c:::r-on.; in diameter, and wherein the drug particles : i. ) i:~ra we a ~>u~W f-ar.'e moc~.if ier adsorbed on the surface thereof, and (ii) ;:rt least 500 of the particles have s.~ particl,e s:x.~e c;f lk:ss t~a~an about 1000 rzm, and (b) a non-aqueous propellant, w't~zPreain the dry powder aggregates and non-aqueous px:~op~:ll.a~:zt aa:w~ t'.orrnulated into a dry powder aerosol for use im:.~ px~c~pell:rnt...basEa pMDI.
In yet. a f=urther ,~sp~:c;t., t:.he ~~re;ent. invention pro~rides an aerosol compcsitioi:. of an aa:luec:>us dispersion of nanoparticulate drug L;aart.i.w.Le~;, wh<<rein: i,a) essential:Ly each droplet of the a~arosol comprises ,-z t:. l.c.~ast cne rranoparticulate pocau~ly sol°,rble drey par!~ic:=Le; (b) the c~roL~lets of the ae:rcas<~~1. Ylawe a rruzM~~ mec:~:i_aru aerodynamic diameter (MMAD) less than or equal to about:. :100 microns; and (c) the nanoparticul.at:e drug ~>art:ic~~_E=s c:~urnl:ari~e a poo~~ly soluble drug having a solub:il.ity :in at ?..ea.:st one liquid dispersion medium of less than about 10 rogr'rrrl, at least 50 0 ~~' cz 28516-45 (S) of the particles have a particle size of less than about 1000 nm, and have a surface modifier adsorbed on the surface of the drug.
In yet a. furthEax~ as'.~-~ecvt: , tire px:~es ent. unventy icon provides a nanoparticulate ae~::oso:1 c:c.amF>c~sitic~n f or_ u;ae in a propellant-based pP~tUI comprising: i:a) a n:anoparticu~ate poorly soluble crystallizne dr~.zcw~ l.~r<:rvir:rc~ a s~~lubi.lit:y ~.n at lea;~t one liquid d.ispersi.on rnc~d~.l.~m of less than about;
mg/m1, wherein the drug has a surface modifier adsorbed 10 ~on t, he surface thereao~, arud a: l.e,x:-~t: ~a0'aof t:he part:p.c~l.es have a particle size of less t.lraz°a abau~t: l.OtlO nm, (b) essentially each droplet of the aerosa_1 comprises at least one nanoparticulat~: dru~:~ paz:~t:icle, wnez°eir~ t;he droplets of the aerosol genera:ated :k:~y ta~n~~s pMDI have a diameter less than ar equal tcs about 100 rnicx,ons, and (c) a non--aqueous propellant:..
rn another aspect, the presean~ iravention provides a method of making a:~n aerosol c~c:~rrrpc~~s.t.. i.::~rl c~otnpri sing (a) forming an aqueous nanoparticulate dispersion of a poorly soluble drug having a ;:7c~lu~lilit.y in at. least one Liq~.z:id dispersion ms::=dium c>f lc ~:~ t~.r~ar~ ~Gw~c~au'. 1.C> mg/n~I, whex-ein: ( l ) the dispers ion compri. ses *aoorl.y sclu.ble cryStall ine drug part cycles arxcl a surf a~:~ rn<adi f ier adsorbed on t:he surface thereof, arzd (a_i.) ~~xt, rLe~~,:;t, ~:>0~ of the particles have a particle size of less ~harn about 1000 nm;
(b) spray-drying t'ne nanoparticulate dispevsic:an to form a dry powder of aggrega~t~es c~f t;.1 e- :rrt:rrcopa~:~::icr.tlate drug and surface modifier pax.°ti.cles, where:irG the ag<~regates ha~,re a diameter of less than or equal_ to about 10i~ microns; and i;c) formulating the dry Pc:'wdex 4~c~gx°c~gat~ø-~:~ .:r~t_o an aerosol composition.
'7 b 28616-45(S) In a further aspect, the present invention prcwides a rrrethod of rnakirzg a~z s;z~:~:r_wasc.~.l. c~omposi.t:ion corr.prising: (a) rrrill_:ing r.irnde~ r°m:rz-pressurized <~onditeions in a non-aqueous medium Izaviz:uc~ a rx:i.c~r:r bc:a:i1 inca .~7c,3i.z~t: t;he following: (i.) a pc,c:u:rly ~~a.lul~,l~a c::x~y~:>t.al.7.i.n~:= drlzg lnaz;,in.g a solubility in at least one li~:~ui.d dispersi;an medium <::f less than about 10 mg/ml , arid ( i. z. ) ~:.~ =stzx,.f a~~: rr~o~i:i.f~ier, t:o obtain a nanoparti culate drug cornpos ition wrxexvein at least ~:~0 0 of t: he particles have a particle ~i ~:r~ c>f Less tta.arz about: 1000 nm, (b) evaporating the non-aqueous medium to obtain a dry powder of aggregates c~f c~.rug a:;~r~.c~ :~r.4r~f,~c~.=' mc~cii.fa.E~r paz:°ti.cles, wherein the aggregates have a ciiarrrc.tez- of less than c>r equal to about 100 microns; anr~ (c°) foro.raLat:ing thze dry ~>owder aggregates into an <~ex:osc>l cony>~~>sit::.ic~n.
In yet a :f:urther as~.aec.~t., t.r~e= ~pre~~~erut: invent. ion provides a method of making azn aex:~osol. cYorn~aosition comprising: (a) rni:Lling r.xndex ~:ore~~m.ari..aed c~c:~x:rc~ition~~, in a non--aqueous medium the fol:Low_~.rrg: (:i.) z pc::~or~l.y soluble crystalline drug having a solubil.i.t:y in ;~t least. one liquid dispersion medium of Less trzar: abc::o_at ~_0 rry~a"ml, and. (ii.) a suraace modifier, to obtain a drug particle wherein at least ~~0% of the pa:rticl.~a; have a pa:rt:ir:::l.e w;~L~c~ ~afv less than about 1000 nm, (b) evaporating the raon-aqueau; medium to obtain a dry powder of aggregate" caf dx_ug ~~.:~<i s~,.z:e:~:f:~c.:~e modifier particles, wherein the agg:regat.es -:rave 4~ d.ameter of Less than or eq~zal to about;: 10C) rtuic::ro:°arz~~; a.nt:i (~:,'~
formulating the dry powder aggregates into am <~c~>rosol c;c::>rrrpc:>sition.
r:n anot:her- aspect, tfrE_= ~:a.r~esezzt:: ir:~~,rention prr:w:i.des a method of making az~x aerosol composit.ic~x~z comprising:
(a) forming an aqueous narropart:. ic~..xlat:~_= ~:~ia~:~ersion of a poorly solublE~ drug having <:~ ~~~:~:J.ubiLit.y irr at least ore l.iqu.id dispersion med:i.um of less than azbout 10 mg/ml, ~1 ii 28516-45(S) wherein: (i.) the d:i::~persion s.vcarrr~.m~:isE;y, L:~ccrl.y ;~«lub~.c~:
crystalline drug part:iclec>>, wizex:eir~ ar_ least. 50'p of t:he drug particles have a particle size of _ess than about 10C?0 nm, and (ii) a surface madifiex:~ ac:xsc~r°ka~.ci ..~,r:, the sur:~ac~e i::.hereof;
(b) freeze-drying the nancpart::~c~L?:l.~.rt:.~~ c~ispersiorx to norm a dry powder of aggregates of the raanoparticalate drug and surface modifier particles, wherein t.'tle aggregates have a diameter of less than cr ec~Lia::. t.c~ <abc;>ut ;107 rc~_Lcr:-oms; and (c) formulating the freeze-drec3 powder ~.~~,:~regates i.nt.o an aerosol composition.
In a further aspect, the present invention provides a method of maki.n.g ar: aex~r_~scl corycasition comprising an aqueo..rs di:per's:i ;>~u r:>f roar?~~p<~:r~i::~_c:~.v.r1 ate ci.rug particles, wherein said nanoparv,icmlate dr~.~g particles comprise a poorly soluble druc, having a sc.iubility irr at leapt one liquid di~~.pers:ion mE:,>;ia.urr? c~~f: l.c.~s~ than about. 10 rng%rnl, at least 50% c:c~ th~.= pa~:-t:i.c~:7.e~s ll;ave <~ .~:~artic'le :size of less than about 1000 nm, and brave a non-cr«sslinked surface modifier adsorbed orn the suz°fac~triexec~f, vvrierein the rruethod <comprises: (a) pxaviding are. acluec::u.r~ ai~,pea:s:i.c>ri of said nanc~parti.culate drug parta.cles; ar?d (b) forming an aerosol comprising liquid droplets of said dispF.rs.'~on, wherein:
(i) essentia.lly each drcplat c~f i~he ac~;vc~5c->1. comprises at least one nanoparticrulat:e poox l~~ ;c>lLUblzdwg ga.rtiele and at least one surface modifier adsorbed Tic the surface of the dru<~ particle, and !;ii) the liquid droplet's forming t.fze aerc7sol have <~ mas;~ rrm~an aeroc~yma3.xnic ~;~.:~;rme~i-er of less than about 100 microns.
In yet a further :~s~7~~c°t , tha 1-~re:m>ent invention provides a method of making are aerosol composition of nanopart.iculate drug parti.c_Le~, l:c:a:r~ ..zse :c..n <-s. propellant.: -based pMD7, wherein said nar°mpax~t~c:~u.~at.r> ,~rua.c p~~xticles comL>rise a "; c1 28516-45 (S) poorly soluble drug hxavimg a ;~clubilit~.v in at least cane liauid dispersion medium of: lc::a~ ~.::klar:r ~~bout ~1.« rng/ml., at least 500 of the particles have ~ pa:r~t.~~cl.e sire of le::ss~ than about 1000 nm, and have a non--crossl.ink:ed surface modifier adsorbed on the su.rf~ace thcre~~i, w~mr°~~i.n t"m rnet:hod comprises: (a) provi..ding a d::~;~pe:r:r_sic~x:, ref ;"~ic~
nanoparticulate drug particles in a :Liquid propellant; and (b) forming an aerosol corr~pxvi~;::Lxxg 1 ~.qt::~..d d:~c~plet~s of said dispersion, wherein : ( i) e~ss<~~a: t, i.rt~l 1.y e:~r;h ciropiet: c9 f': the aerosol comprises at least one nancjparticulate poorly soluble drug particle and at :' east oxwe surface modifier adsorbed to the surface ref t.hE~ ci~:u~r parti.r_~~F:s, axad dii.,) the liquid droplets forming the aerosol. have a mass mean aerodynamic diameter of less ttiatx about 10~:~ microns.
A. Aqueous Aerosol Formulations The present invention encompasses aqueous formulations containing r~xarrop~rti~~ulate dr~:rg particles. For aqueous aerosol formulati.oxi.~, t.ue drug ~r~ay be present at a com:.entration of about 0.05 mg/mL up to about 500 mg/mL.
Such formulatiorxs provide effectir~je delive~~y to appropriate area:rs of_ the lung or nasal.. .at,~i.ti.E~=:~. Ir3 acid.it.i.on, th.e more concentrated aeroso_L t:.ormulatons l; .l . a . , fr~r aqueous aerosol forrnulati_ons, about 10 mg/rnL up to about 6t.)0 mg/mL) have the add:i.tional ad~Vanta~~e of ena~l.ixvc:~ 1.~.~,.rge ~u.a,utit.ies of c:irug substance to be delivered to the ~.urng iii ,~ very short period of time, e.g., about 1 to abou 2 seconds i:1. puff) as compared to the converrt:iorral. ~ -<~:() ruin. <xdrn::.nistration period.
B. Dry Powder Aerosol Formulations Anol~her embodiment c:f t~~e i.:nwc~nt~i,orr. is direc°:.~ted to dry powder aerosol formulations compri.:~:i.nc-~ drug particles T ~>

28516-45(S) for pulmonary and nasal administz~at.ic~n.. Dry powders, which can be used in both DPIs and pMDIs, can be made by spray-drying aqueous nanoparticulatc~ dr°ug dis persia~ns .
Alternatively, dry pc>wder~s ~:corit:,z:i.r-xa_zn~~ r ~,~ra.op4~ri~.iculat~:
drug can be made by free~Ea-.drying n:~anop,:~rt:i.r.°ul.st~.~ c~~~ug dispersions.
"f Combinations of spray-dried and freeze-dried nanoparticulate drug powders can be used in DPIs and pMDIs. For dry powder aerosol formulations, the drug may be present at a concentration of about 0.05 mg/g up to about 990 mg/g. In addition, the more concentrated aerosol formulations (i.e., for dry powder aerosol formulations about 10 mg/g up to about 990 mg/g) have the additional advantage of enabling large quantities of drug substance to be delivered to the lung in a very short period of time, e.g., about 1 to about 2 seconds (1 puff).
1. Spray-Dried Powders Containing Nanoparticulate Drug t o Powders comprising nanoparticulate drug can be made by spray-drying aqueous dispersions of a nanoparticulate drug and a surface modifier to form a dry powder which consists of aggregated drug nanoparticles. The aggregates can have a size of about 1 to about 2 microns which is suitable for deep lung delivery. The aggregate particle size can be increased to target alternative delivery sites, such as the upper 15 bronchial region or nasal mucosa by increasing the concentration of drug in the spray-dried dispersion or by increasing the droplet size generated by the spray dryer.
Alternatively, the aqueous dispersion of drug and surface modifier can contain a dissolved diluent such as lactose or mannitol which, when spray dried, forms respirable diluent particles, each of which contains at least one embedded drug 2o nanoparticle and surface modifier. The diluent particles with embedded drug can have a particle size of about 1 to about 2 microns, suitable for deep lung delivery.
In addition, the diluent particle size can be increased to target alternate delivery sites, such as the upper bronchial region or nasal mucosa by increasing the concentration of dissolved diluent in the aqueous dispersion prior to spray drying, or by increasing the droplet size 25 generated by the spray dryer.
Spray-dried powders can be used in DPIs or pMDIs, either alone or combined with freeze-dried nanoparticulate powder. In addition, spray-dried powders containing drug nanoparticles can be reconstituted and used in either jet or ultrasonic nebulizers to generate aqueous dispersions having respirable droplet sizes, where each droplet contains at least one drug nanoparticle. Concentrated nanoparticulate dispersions may also be used in these aspects of the invention.
2. Freeze-Dried Powders Containing Nanoparticulate Drug Nanoparticulate drug dispersions can also be freeze-dried to obtain powders suitable for nasal or pulmonary delivery. Such powders may contain aggregated nanoparticulate drug particles having a surface modifier. Such aggregates may have sizes within a respirable range, i.e., about 2 to about 5 microns. Larger aggregate particle sizes can be obtained for targeting alternate delivery sites, such as the nasal mucosa.
1o Freeze dried powders of the appropriate particle size can also be obtained by freeze drying aqueous dispersions of drug and surface modifier, which additionally contain a dissolved diluent such as lactose or mannitoi. In these instances the freeze dried powders consist of respirable particles of diluent, each of which contains at least bne embedded drug nanoparticle.
~5 Freeze-dried powders can be used in DPIs or pMDIs, either alone or combined with spray-dried nanoparticulate powder. In addition, freeze-dried powders containing drug nanoparticles can be reconstituted and used in either jet or ultrasonic nebulizers to generate aqueous dispersions having respirable droplet sizes, where each droplet contains at least one drug nanoparticle. Concentrated nanoparticulate dispersions 20 may also be used in these aspects of the invention.
C. Propellant-Based Formulations Yet another embodiment of the invention is directed to a process and composition for propellant-based systems comprising nanoparticulate drug particles and a 25 surface modifier. Such formulations may be prepared by wet milling the coarse drug substance and surface modifier in liquid propellant, either at ambient pressure or under high pressure conditions. Alternatively, dry powders containing drug nanoparticles may be prepared by spray-drying or freeze-drying aqueous dispersions of drug nanoparticles and the resultant powders dispersed into suitable propellants for use in conventional pMDIs. Such nanoparticulate pMDI formulations can be used for either nasal or pulinonary delivery. For pulmonary administration, such formulations afford increased delivery to the deep lung regions because of the small (i.e., about 1 to about 2 microns) particle sizes available from these methods. Concentrated aerosol formulations can also be employed in pMDIs.
D. Methods of Making Aerosol Formulations The invention also provides methods for making an aerosol of nanoparticulate compositions. The nanoparticulate dispersions used in making aqueous 1o aerosol compositions can be made by wet milling or by precipitation methods known in the art. Dry powders containing drug nanoparticles can be made by spray drying or freeze-drying aqueous dispersions of drug nanoparticles. The dispersions used in these systems may or may not contain dissolved diluent material prior to drying.
Additionally, both pressurized and non-pressurized milling operations can be employed to make 15 nanoparticulate drug compositions in non-aqueous systems.
In a non-aqueous, non-pressurized milling system, a non-aqueous liquid which has a vapor pressure of 1 afro or less at room temperature is used as a milling medium and may be evaporated to yield dry nanoparticulate drug and surface modifier.
The non-aqueous liquid may be, for example, a high-boiling halogenated hydrocarbon.
2o The dry nanoparticulate drug composition thus produced may then be mixed with a suitable propellant or propellants and used in a conventional pMDI.
Alternatively, in a pressurized milling operation, a non-aqueous liquid which has a vapor pressure > 1 afro at room temperature is used as a milling medium for making a nanoparticulate drug and surface modifier composition. Such a liquid may be, 25 for example, a halogenated hydrocarbon propellant which has a low boiling point. The resultant nanoparticulate composition can then be used in a conventional pNiDI
without further modification, or can be blended with other suitable propellants.
Concentrated aerosols may also be made via such methods.

E. Methods of Using Nanoparticulate Aerosol Formulations In yet another aspect of the invention, there is provided a method of treating a mammal comprising: (1) forming an aerosol of a dispersion (either aqueous or powder) of nanoparticles, wherein the nanoparticles comprise an insoluble drug having a surface modifier on the surface thereof, and (2) administering the aerosol to the pulmonary or nasal cavities of the mammal. Concentrated aerosol formulations may also be used in such methods.
Another embodiment of the invention provides a method of diagnosing a mammal comprising: (1) forming an aerosol of a dispersion (either aqueous or dry) of to nanoparticles, wherein the nanoparticles comprise an insoluble diagnostic agent having a surface modifier; (2) administering the aerosol to the pulmonary or nasal cavities of the mammal; and (3) imaging the diagnostic agent in the pulinonary or nasal system.
Concentrated aerosol formulations can also be employed in such diagnostic methods.
Both the foregoing general description and the following detailed ~5 description are exemplary and explanatory and are intended to provide fiirther explanation of the invention as claimed. Other objects, advantages, and novel features will be readily apparent to those skilled in the art from the following detailed description of the invention.
2o BRIEF DESCRIPTION OF THE FIGURES
Figure 1: Shows an in vitro deposition pattern of a concentrated aerosolized beclomethasone dipropionate dispersion from an ultrasonic nebulizer.
Figure 2: Shows an in vitro deposition pattern of a concentrated aerosolized beclomethasone dipropionate dispersion from a jet nebulizer.
25 Figure 3: Shows the aerodynamic volume distribution diameter of a spray-dried naproxen aerosol (2% (w/w) naproxen).
Figure 4: Shows a scanning electron micrograph of spray-dried naproxen aerosol particles (aggregated naproxen/polyvinylpyrrolidone (surface modifier) nanoparticles, demonstrating the overall uniformity of size and the spherical nature of the particles.
Figure 5: Shows the aerodynamic volume distribution diameter of a spray-dried naproxen aerosol (5% (w/w) naproxen).
Figure 6: Shows the aerodynamic volume distribution diameter of a spray-dried triamcinolone acetonide (TA) aerosol (10% (w/w) TA).
Figure 7: Shows two photomicrographs: Fig. 8(A) shows spray-dried nanoparticulate budesonide particles, and Fig. 8(B) shows particles of micronized budesonide.
to Figure 8: Shows the particle size distribution (by volume) of a reconstituted freeze-dried anti-emetic aerosol containing dextrose diluent.
Figure 9: Shows the particle size distribution of a reconstituted freeze-dried .anti-emetic aerosol containing mannitol diluent.
Figure 10: Shows a scanning electron micrograph of nanoparticulate TA milled in a non-pressurized propellant system.
DETAILED DESCRIPTION OF THE INVENTION
A. Aerosol Formulations The compositions of the invention are aerosols which contain drug nanoparticles. Aerosols can be defined as colloidal systems consisting of very finely divided liquid droplets or dry particles dispersed in and surrounded by a gas.
Both liquid and dry powder aerosol compositions are encompassed by the invention.

1. Nanoparticulate Drug and Surface Modifier Particle Size Preferably, the compositions of the invention contain nanoparticles which have an effective average particle size of less than about 1000 nm, more preferably less than about 400 nm, less than about 300 nm, iess than about 250 nm, less than about 100 nm, or less than about 50 nm, as measured by light-scattering methods. By "an effective average particle size of less than about 1000 nm" it is meant that at least 50% of the drug particles have a weight average particle size of less than about 1000 nm when measured by light scattering techniques. Preferably, at least 70% of the drug particles have an average particle size of less than about 1000 nm, more preferably at least 90%
of the drug 1o particles have an average particle size of less than about 1000 nm, and even more preferably at least about 95% of the particles have a weight average particle size of less than about 1000 nm.
2. Concentration of Nanoparticulate Drug For aqueous aerosol formulations, the nanoparticulate agent is present at a concentration of about 0.05 mg/mL up to about 600 mg/mL. For dry powder aerosol formulations, the nanoparticulate agent is present at a concentration of about 0.05 mg/g up to about 990 mg/g, depending on the desired drug dosage. Concentrated nanoparticulate aerosols, defined as containing a nanoparticulate drug at a concentration of about 10 mg/mL up to about 600 mg/mL for aqueous aerosol formulations, and about 10 mg/g up to about 990 mg/g for dry powder aerosol formulations, are specifically encompassed by the present invention. Such formulations provide effective delivery to appropriate areas of the lung or nasal cavities in short administration times, i. e., less than about 15 seconds as compared to administration times of up to 4 to 20 minutes as found in conventional pulmonary nebulizer therapies.
3. In Vivo Deposition of Inhaled Aerosols Aerosols intended for delivery to the nasal mucosa are inhaled through the nose. For optimal delivery to the nasal cavities, inhaled particle sizes of about 5 to about 100 microns are useful, with particle sizes of about 30 to about 60 microns being preferred. For nasal delivery, a larger inhaled particle size is desired to maximize impaction on the nasal mucosa and to minimize or prevent pulmonary deposition of the administered formulation. Inhaled particles may be defined as liquid droplets containing dissolved drug, liquid droplets containing suspended drug particles (in cases where the drug is insoluble in the suspending medium), dry particles of pure drug substance, aggregates of drug nanoparticles, or dry particles of a diluent which contain embedded drug nanoparticles.
For delivery to the upper respiratory region, inhaled particle sizes of about 2 to about 10 microns are preferred, more preferred is about 2 to about 6 microns.
1o Delivery to the upper respiratory region may be desirable for drugs such as bronchodilators or corticosteroids that are to act locally. This is because drug particles deposited in the upper respiratory tract can dissolve and act on the smooth muscle of the airway, rather than being absorbed into the bloodstream of the patient.
However, the goal for some inhaled drugs is systemic delivery, such as in cases of proteins or peptides which 15 are not amenable to oral administration. It is preferred that drugs intended for systemic administration be delivered to the alveolar region of the lung, because 99.99%
of the available surface area for drug absorption is located in the peripheral alveoli. Thus, with administration to the alveolar region, rapid absorption can be realized. For delivery to the deep lung (alveolar) region, inhaled particle sizes of less than about 2 microns are 20 preferred.
4. Aqueous Aerosols Aqueous formulations of the present invention consist of colloidal dispersions of water-insoluble nanoparticulate drug in an aqueous vehicle which are aerosolized using air jet or ultrasonic nebulizers. The advantages of the present invention 25 can best be understood by comparing the sizes of nanoparticuiate and conventional micronized drug particles with the sizes of liquid droplets produced by conventional nebulizers. Conventional micronized material is generally about 2 to about 5 microns or more in diameter and is approximately the same size as the liquid droplet size produced by medical nebulizers. In contrast, nanoparticulate drug particles are substantially smaller than the droplets in such an aerosol. Thus, aerosols containing nanoparticulate drug particles improve drug delivery efficiency because they contain a higher number of drug particles per unit dose such that each aerosolized droplet contains active drug substance.
Empty aqueous droplets . ~ . ~ ~.
Micronized ~ ~ ~ ~~ Aqueous droplets, each drug crystal ~ O ~. ~ containing many nanoparticulate drug particles 2 to 5 pm Aqueous Suspension of Colloidal Dispersion of Micronized Drug Substance Drug Nauoparticles Thus, with administration of the same dosages of nanoparticulate and micronized drug, more lung or nasal cavity surface area is covered by the aerosol formulation containing nanoparticulate drug.
Another advantage of the present invention is that it permits water-insoluble drug compounds to be delivered to the deep lung via nebulization of aqueous formulations. Conventional micronized drug substance is too large to reach the peripheral lung regardless of the size of the droplet produced by the nebulizer, but the present invention permits nebulizers which generate very small (about 0.5 to about 2 microns) aqueous droplets to deliver water-insoluble drugs in the form of nanoparticles to the alveoli. One example of such devices is the Circulaire~ (Westmed Corp., Tucson, AZ).
Yet another advantage of the present invention is that ultrasonic nebulizers 2s can be used to deliver water-insoluble drugs to the lung. Unlike conventional micronized material, nanoparticulate drug particles are readily aerosolized and show good in vitro deposition characteristics. A specific advantage of the present invention is that it permits water-insoluble drugs to be aerosolized by ultrasonic nebulizers which require the drug substance to pass through very fine orifices to control the size of the aerosolized droplets.
While conventional drug material would be expected to occlude the pores, nanoparticulate drug particles are much smaller and can pass through the pores without difficulty.

Another advantage of the present invention is the enhanced rate of dissolution of water-insoluble drugs. Since dissolution rate is a function of the total surface area of drug substance to be dissolved, more finely divided drug particles (e.g., nanoparticies) have much faster dissolution rates than conventional micronized drug particles. This can result in more rapid absorption of inhaled drugs. For nasally administered drugs it can result in more complete absorption of the dose, since with a nanoparticulate drug dose the particles can dissolve rapidly and completely before being cleared via the mucociliary mechanism.
l0 5. Dry Powder Aerosol Formulations The invention is also directed to dry powders which contain nanoparticulate compositions for pulinonary or nasal delivery. The powders may consist of respirable aggregates of nanoparticulate drug particles, or of respirable particles of a diluent which contains at least one embedded drug nanoparticle. Powders containing 15 nanoparticulate drug particles can be prepared from aqueous dispersions of nanoparticles by removing the water via spray-drying or lyophilization (freeze drying).
Spray-drying is less time consuming and less expensive than freeze-drying, and therefore more cost-effective. However, certain drugs, such as biologicals benefit from lyophilization rather than spray-drying in making dry powder formulations.
2o Dry powder aerosol delivery devices must be able to accurately, precisely, and repeatably deliver the intended amount of drug. Moreover, such devices must be able to fully disperse the.dry powder into individual particles of a respirable size.
Conventional micronized drug particles of 2-3 microns in diameter are often difficult to meter and disperse in small quantities because of the electrostatic cohesive forces inherent 25 in such powders. These difficulties can lead to loss of drug substance to the delivery device as well as incomplete powder dispersion and sub-optimal delivery to the lung.
Many drug compounds, particularly proteins and peptides, are intended for deep lung delivery and systemic absorption. Since the average particle sizes of conventionally prepared dry powders are usually in the range of 2-3 microns, the fraction of material which actually reaches the alveolar region may be quite small. Thus, delivery of micronized dry powders to the lung, especially the alveolar region, is generally very inefficient because of the properties of the powders themselves.
The dry powder aerosols which contain nanoparticulate drugs can be made smaller than comparable micronized drug substance and, therefore, are appropriate for efficient delivery to the deep lung. Moreover, aggregates of nanoparticulate drugs are spherical in geometry and have good flow properties, thereby aiding in dose metering and deposition of the administered composition in the lung or nasal cavities.
a::
::a::: ..'~.i::R.
;;;;;. .,::: , ;f:
'... :t : .~_ ~ .=.,:
;::: . . , .::_:::
.. .. ~ ;._:~:
w Micronized Drug Substance Respirable Aggregates of Nanoparticles Respirable Diluent Particles Coataining not less than 2 ~m in diameter Less than 2 ~m to 100 ~m in diameter Embedded of Nanoparticles. Less than 2 ~m to 100 ~m is diameter 2o Dry nanoparticulate compositions can be used in both DPIs and pMDIs.
(In this invention, "dry" refers to a composition having less than about 5%
water.) 6. Propellant-Based Aerosols Another embodiment of the invention is directed to a process and composition for propellant-based MDIs containing nanoparticulate drug particles. pMDIs can comprise either discrete nanoparticles of drug and surface modifier, aggregates of nanoparticles of d=ug and surface modifier, or inactive diluent particles containing embedded nanoparticles. pMDIs can be used for targeting the nasal cavity, the conducting airways of the lung, or the alveoli. Compared to conventional formulations, 3o the present invention affords increased delivery to the deep lung regions because the inhaled nanoparticulate drug particles are smaller than conventional micronized material (< 2 ~tm) and are distributed over a Larger mucosal or alveolar surface area as compared to micronized drugs.

Nanoparticulate drug pMDIs of the present invention can utilize either chlorinated or non-chlorinated propellants. Concentrated nanoparticulate aerosol formulations can also be employed in pMDIs.
B. Methods of Making Aerosol Formulations The nanoparticulate drug compositions for aerosol administration can be made by, for example, ( 1 ) nebulizing an aqueous dispersion of nanoparticulate drug, obtained by either grinding or precipitation; (2) aerosolizing a dry powder of aggregates of nanoparticulate drug and surface modifier (the aerosolized composition may additionally contain a diluent); or (3) aerosolizing a suspension of nanoparticulate drug or drug aggregates in a non-aqueous propellant. The aggregates of nanoparticulate drug and surface modifier, which may additionally contain a diluent, can be made in a non-pressurized or a pressurized non-aqueous system. Concentrated aerosol formulations may 15 also be made via such methods.
1. Aqueous Milling to obtain Nanoparticulate Drug Dispersions Milling of aqueous drug to obtain nanoparticulate drug is described in the '684 patent. In sum, drug particles are dispersed in a liquid dispersion medium and 2o mechanical means is applied in the presence of grinding media to reduce the particle size of the drug to the desired effective average particle size. The particles can be reduced in size in the presence of one or more surface modifiers. Alternatively, the particles can be contacted with one or more surface modifiers after attrition. Other compounds, such as a diluent, can be added to the drug/surface modifier composition during the size reduction 25 process. Dispersions can be manufactured continuously or in a batch mode.
2. Precipitation to Obtain Nanoparticulate Drug Compositions Another method of forming the desired nanoparticle dispersion is by microprecipitation. This is a method of preparing stable dispersions of drugs in the 30 presence of one or more surface modifiers and one or more colloid stability enhancing surface active agents free of any trace toxic solvents or solubilized heavy metal impurities. Such a method comprises, for example, (1) dissolving the drug in a suitable solvent with mixing; (2) adding the formulation from step (1) with mixing to a solution comprising at least one surface modifier to form a clear solution; and (3) precipitating the formulation from step (2) with mixing using an appropriate nonsolvent. The method can be followed by removal of any formed salt, if present, by dialysis or diafiltration and concentration of the dispersion by conventional means. The resultant nanoparticulate drug dispersion can be utilized in liquid nebulizers or processed to form a dry powder for use in a DPI or pMDI.
3. Non-Aqueous Non-Pressurized Milling Systems In a non-aqueous, non-pressurized milling system, a non-aqueous liquid having a vapor pressure of about 1 atm or less at room temperature and in which the drug substance is essentially insoluble is used as a wet milling medium to make a i5 nanoparticulate drug composition. In such a process, a slurry of drug and surface modifier is milled in the nonaqueous medium to generate nanoparticulate drug particles.
Examples of suitable non-aqueous media include ethanol, trichloromonofluoromethane (CFC-1 I), and dichlorotetrafluoroethane (CFC-114). An advantage of using CFC-1 I is that it can be handled at only marginally cool room temperatures, whereas CFC-requires more controlled conditions to avoid evaporation. Upon completion of milling the liquid medium may be removed and recovered under vacuum or heating, resulting in a dry nanoparticulate composition. The dry composition may then be filled into a suitable container and charged with a final propellant. Exemplary final product propellants, which ideally do not contain chlorinated hydrocarbons, include HFA-134a (tetrafluoroethane) 2s and HFA-227 (heptafluoropropane). While non-chlorinated propellants may be preferred for environmental reasons, chlorinated propellants may also be used in this aspect of the invention.

4. Non-Aqueous Pressurized Milling System In a non-aqueous, pressurized milling system, a non-aqueous liquid medium having a vapor pressure significantly greater than 1 afro at room temperature is used in the milling process to make nanoparticulate drug compositions. If the milling medium is a suitable halogenated hydrocarbon propellant, the resultant dispersion may be filled directly into a suitable pMDI container. Alternately, the milling medium can be removed and recovered under vacuum or heating to yield a dry nanoparticulate composition. This composition can then be filled into an appropriate container and charged with a suitable propellant for use in a pMDI.
5. Spray-Dried Powder Aerosol Formulations Spray drying is a process used to obtain a powder containing nanoparticulate drug particles following particle size reduction of the drug in a liquid medium. In general, spray-drying is used when the liquid medium has a vapor pressure of less than about 1 afro at room temperature. A spray-dryer is a device which allows for liquid evaporation and drug powder collection. A liquid sample, either a solution or suspension, is fed into a spray nozzle. The nozzle generates droplets of the sample within a range of about 20 to about 100 ~m in diameter which are then transported by a carrier gas into a drying chamber. The carrier gas temperature is typically between about 80 and 2o about 200°C. The droplets are subjected to rapid liquid evaporation, leaving behind dry particles which are collected in a special reservoir beneath a cyclone apparatus.
If the liquid sample consists of an aqueous dispersion of nanoparticles and surface modifier, the collected product will consist of spherical aggregates of the nanoparticulate drug particles. If the liquid sample consists of an aqueous dispersion of nanoparticles in which an inert diluent material was dissolved (such as lactose or mannitol), the collected product will consist of diluent (e.g., lactose or mannitol) particles which contain embedded nanoparticulate drug particles. The final size of the collected product can be controlled and depends on the concentration of nanoparticulate drug and/or diluent in the liquid sample, as well as the droplet size produced by the spray-dryer nozzle. For deep lung delivery it is desirable for the collected product size to be less than about 2 ~m in diameter; for delivery to the conducting airways it is desirable for the collected product size to be about 2 to about 6 wm in diameter, and for nasal delivery a collected product size of about S to about 100 pm is preferred. Collected products may then be used in conventional DPIs for pulinonary or nasal delivery, dispersed in propellants far use in pMDIs, or the particles may be reconstituted in water for use in nebulizers.
In some instances it may be desirable to add an inert carrier to the spray-dried material to improve the metering properties of the final product. This may 1 o especially be the case when the spray dried powder is very small (less than about 5 ltm) or when the intended dose is extremely small, whereby dose metering becomes difficult. In general, such carrier particles (also known as bulking agents) are too large to be delivered to the lung and simply impact the mouth and throat and are swallowed. Such carriers typically consist of sugars such as lactose, mannitol, or trehalose. Other inert materials, including polysaccharides and cellulosics, may also be useful as carriers.
:_,~'~ ::~. p~~ O O
.v .. ~O . O
:;; :., :;:
::: ...:::. D D D O
.. ' ' o~ o0 O O
O O
.. .. D D D
Respirable aggregates of Respirable diluent particles containing nanoparticles mixed with embedded nanoparticles mixed with an inert an inert tamer carrier.
Spray-dried powders containing nanoparticulate drug particles may used in conventional 3o DPIs, dispersed in propellants for use in pMDIs, or reconstituted in a liquid medium for use with nebulizers.
6. Freeze-Dried Nanoparticulate Compositions For compounds that are denatured or destabilized by heat, such as compounds having a low melting point (i.e., about 70 to about 150°C), or for example, biologics, sublimation is preferred over evaporation to obtain a dry powder nanoparticulate drug composition. This is because sublimation avoids the high process temperatures associated with spray-drying. In addition, sublimation, also known as freeze-drying or lyophilization, can increase the shelf stability of drug compounds, particularly for biological products. Freeze-dried particles can also be reconstituted and 1o used in nebulizers. Aggregates of freeze-dried nanoparticulate drug particles can be blended with either dry powder intermediates or used alone in DPIs and pMDIs for either nasal or pulmonary delivery.
Sublimation involves freezing the product and subjecting the sample to strong vacuum conditions. This allows for the formed ice to be transformed directly from a solid state to a vapor state. Such a process is highly efficient and, therefore, provides greater yields than spray-drying. The resultant freeze-dried product contains drug and modifier(s). The drug is typically present in an aggregated state and can be used for inhalation alone (either pulmonary or nasal), in conjunction with diluent materials (lactose, mannitol, etc.), in DPIs or pMDIs, or reconstituted for use in a nebulizer.
C. Methods of Using Nanoparticulate Drug Aerosol Formulations The aerosols of the present invention, both aqueous and dry powder, are particularly useful in the treatment of respiratory-related illnesses such as asthma, emphysema, respiratory distress syndrome, chronic bronchitis, cystic fibrosis, chronic obstructive pulmonary disease, organ-transplant rejection, tuberculosis and other infections of the lung, fungal infections, respiratory illness associated with acquired immune deficiency syndrome, oncology, and systemic administration of an anti-emetic, analgesic, cardiovascular agent, etc. The formulations and method result in improved lung and nasal surface area coverage by the administered drug.

In addition, the aerosols of the invention, both aqueous and dry powder, can be used in a method for diagnostic imaging. Such a method comprises administering to the body of a test subject in need of a diagnostic image an effective contrast-producing amount of the nanoparticulate aerosol diagnostic image contrast composition.
Thereafter, at least a portion of the body containing the administered contrast agent is exposed to x-rays or a magnetic field to produce an x-ray or magnetic resonance image pattern corresponding to the presence of the contrast agent. The image pattern can then be visualized.
1o D. Summary of Advantaees of the Compositions and Methods of the Invention Using the compositions of the invention, essentially water-insoluble drugs can be delivered to the deep lung. This is either not possible or extremely difficult using aerosol formulations of micronized water-insoluble drugs. Deep lung delivery is necessary for drugs that are intended for systemic administration, because deep lung 15 delivery allows rapid absorption of the drug into the bloodstream via the alveoli, thus enabling rapid onset of action.
The present invention increases the number of drug particles per unit dose and results in distribution of the nanoparticulate drug particles over a larger physiological surface area as compared to the same quantity of delivered micronized drug.
For systemic 2o delivery via the pulmonary route, this approach takes maximum advantage of the extensive surface area presented in the alveolar region - thus producing more favorable drug delivery profiles, such as a more complete absorption and rapid onset of action.
Moreover, in contrast to micronized aqueous aerosol dispersions, aqueous dispersions of water-insoluble nanoparticulate drugs can be nebulized ultrasonically.
25 Micronized drug is too large to be delivered efficiently via an ultrasonic nebulizer.
Droplet size determines in vivo deposition of a drug, l. e., very small particles, about < 2 microns, are delivered to the alveoli; larger particles, about 2 to about microns, are delivered to the bronchiole region; and for nasal delivery, particles of about 5 to about 100 microns are preferred. Thus, the ability to obtain very small drug 3o particle sizes which can "fit" in a range of droplet sizes allows more effective and more efficient (i.e., dose uniformity) targeting to the desired delivery region.
This is not possible using micronized drug, as the particle size of such drugs is too large to target areas such as the alveolar region of the lung. Moreover, even when micronized drug is incorporated into larger droplet sizes, the resultant aerosol formulation is heterogeneous (i.e., not all droplets contain drug), and does not result in such the rapid and efficient drug delivery enabled by the nanoparticulate aerosol formulations of the invention.
The present invention also enables the aqueous aerosol delivery of high doses of drug in an extremely short time period, i.e., 1-2 seconds (1 puff).
This is in contrast to the conventional 4-20 min. administration period observed with pulmonary to aerosol formulations of micronized drug.
Furthermore, the dry aerosol nanoparticulate powders of the present invention are spherical and can be made smaller than micronized material, thereby producing aerosol compositions having better flow and dispersion properties, and capable of being delivered to the deep lung.
15 Finally, the aerosol compositions of the present invention enable rapid nasal delivery. Nasal delivery of such aerosol compositions will be absorbed more rapidly and completely than micronized aerosol compositions before being cleared by the mucociliary mechanism.
20 * * * **
Drug Particles The nanoparticles of the invention comprise a therapeutic or diagnostic agent, which in the invention are collectively are referred to as a "drug." A
therapeutic agent can be a pharmaceutical, including biologics such as proteins and peptides, and a 25 diagnostic agent is typically a contrast agent, such as an x-ray contrast agent, or any other type of diagnostic material. The drug exists as a discrete, crystalline phase.
The crystalline phase differs from a non-crystalline or amorphous phase which results. from precipitation techniques, such as those described in EPO 275,796.
The invention can be practiced with a wide variety of drugs. The drug is 3o preferably present in an essentially pure form, is poorly soluble, and is dispersible in at least one liquid medium. By "poorly soluble" it is meant that the drug has a solubility in the liquid dispersion medium of less than about I O mg/mL, and preferably of less than about 1 mg/mL.
Suitable drugs include those intended for pulmonary or intranasal delivery.
Pulmonary and intranasal delivery are particularly useful for the delivery of proteins and polypeptides which are difficult to deliver by other routes of administration.
Such pulmonary or intranasal delivery is effective both for systemic delivery and for localized delivery to treat diseases of the air cavities.
Preferable drug classes include proteins, peptides, bronchodilators, 1o corticosteroids, elastase inhibitors, analgesics, anti-fungals, cystic-fibrosis therapies, asthma therapies, emphysema therapies, respiratory distress syndrome therapies, chronic bronchitis therapies, chronic obstructive pulmonary disease therapies, organ-transplant rejection therapies, therapies for tuberculosis and other infections of the lung, fungal infection therapies, and respiratory illness therapies associated with acquired immune 15 deficiency syndrome, oncology therapies, systemic administration of anti-emetics, analgesics, cardiovascular agents, etc.
The drug can be selected from a variety of known classes of drugs, including, for example, analgesics, anti-inflammatory agents, anthelmintics, anti-arrhythmic agents, antibiotics (including penicillins), anticoagulants, antidepressants, 2o antidiabetic agents, antiepileptics, antihistamines, antihypertensive agents, antimuscarinic agents, antimycobacterial agents, antineoplastic agents, immunosuppressants, antithyroid agents, antiviral agents, anxiolytic sedatives (hypnotics and neuroleptics), astringents, beta-adrenoceptor blocking agents, blood products and substitutes, cardiac inotropic agents, contrast media, corticosteroids, cough suppressants (expectorants and mucolytics), 25 diagnostic agents, diagnostic imaging agents, diuretics, dopaminergics (antiparkinsonian.
agents), haemostatics, immuriological agents, lipid regulating agents, muscle relaxants, parasympathomimetics, parathyroid calcitonin and biphosphonates, prostaglandins, radio-pharmaceuticals, sex hormones (including steroids), anti-allergic agents, stimulants and anoretics, sympathomimetics, thyroid agents, vasodilators and xanthines.

28516-45 (S) ' A description of these classes of drugs and a listing of secies within each class can be found in Martindale, The Extra Pharmacopoeia, Twenty-ninth Edition (The Pharmaceutical Press, London, 1989). The drugs are commercially available and/or can be prepared by techniques known in the art.
Preferred contrast agents are taught in the '684 patent, which is specifically incorporated by reference.
Suitable diagnostic agents are also disclosed in the U.S.
Patent No. 5,260,478; U.S. Patent No. 5,264,610; U.S. Patent No. 5,300,739.
Surface Modifiers Suitable surface modifiers can preferably be selected from known organic and inorganic pharmaceutical excipients. Such excipients include various polymers, low molecular weight oligomers, natural products, and surfactants. Preferred surface modifiers include nonionic and ionic surfactants. Two or more surface modifiers can be used in combination.
Representative examples of surface modifiers include cetyl pyridinium chloride, gelatin, casein, lecithin (phosphatides), dextran, glycerol, gum acacia, cholesterol, tragacanth, stearic acid, benzalkonium chloride, calcium stearate, glycerol monostearate, cetostearyl alcohol, cetomacrogol emulsifying wax, sorbitan esters, polyoxyethylene alkyl ethers (e.g., macrogol ethers such as cetomacrogol 1000), polyoxyethylene castor oil derivatives, polyoxyethylene sorbitan fatty acid esters (e.g., the commercially available Tweens~ such as e.g., Tween 20° and Tween 80° (ICI Specialty Chemicals)); polyethylene glycols (e. g., Carbowaxs 3350 and 1450, and Carbopol 934 (Union 28516-45(S) Carbide)), dodecyl trimethyl ammonium bromide, polyoxymethylene stearates, colloidal silicon dioxide, phosphates, sodium dodecylsulfate, carboxymethylcellulose calcium, hydroxypropyl cellulose (HPC, HPC-SL, and HPC-L), hydroxypropyl methylcellulose (HPMC), carboxymethylcellulose sodium, methylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethyl-cellulose phthalate, noncrystalline cellulose, magnesium aluminum silicate, triethanolamine, polyvinyl alcohol (PVA), polyvinylpyrrolidone (PVP), 4-(1,1,3,3-tetramethylbutyl)-phenol polymer with ethylene oxide and formaldehyde (also known as tyloxapol, superione, and triton), poloxamers (e. g., Pluronics F68° and F108~, which are block copolymers of ethylene oxide and propylene oxide); poloxamines (e. g., Tetronic 908°, also known as Poloxamine 908~, which is a tetrafunctional block copolymer derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine (BASF Wyandotte Corporation, Parsippany, N.J.)); a charged phospholipid such as dimyristoyl phosphatidyl glycerol, dioctylsulfosuccinate (DOSS);
Tetronic 1508° (T-1508) (BASF Wyandotte Corporation), dialkylesters of sodium sulfosuccinic acid (e. g., Aerosol OT~, which is a dioctyl ester of sodium sulfosuccinic acid (American Cyanamid)); Duponol P~, which is a sodium lauryl sulfate (DuPont); Tritons X-200~, which is an alkyl aryl polyether sulfonate (Rohm and Haas); Crodestas F-110°, which is a mixture of sucrose stearate and sucrose distearate (Croda Inc.); p-isononylphenoxypoly-(glycidol), also known as Olin-lOG~ or Surfactant 10-G° (Olin Chemicals, Stamford, CT); Crodestas SL-40~ (Croda, Inc.); and SA90HC0, which is C18H37CH2 (CON (CH3) -CH2 (CHOH) 4 (CHZOH) 2 (Eastman Kodak Co. ) ;
decanoyl-N-methylglucamide; n-decyl ,Q-D-glucopyranoside; n-decyl ~i-D-maltopyranoside; n-dodecyl ,Q-D-glucopyranodside;

28516-45(S) ri-dodecyl a-D-maltoside; heptanoyl-N-methylglucamide; n-heptyl-a-D-glucopyranoside; n-heptyl a-D-thioglucoside; n-hexyl ~-D-glucopyranoside; nonanoyl-N-methylglucamide; n-noyl-~-D-glucopyranoside; octanoyl-N-methylglucamide; n-octyl-a-D-glucopyranoside; octyl a-D-thioglucopyranoside;
and the like. Tyloxapol is a particularly preferred surface modifier for the pulmonary or intranasal delivery of steroids, even more so for nebulization therapies.
Most of these surface modifiers are known pharmaceutical excipients and are described in detail in the Handbook of Pharmaceutical Excipients, published jointly by the American Pharmaceutical Association and The Pharmaceutical Society of Great Britian (The Pharmaceutical Press, 1986). The surface modifiers are commercially available and/or can be prepared by techniques known in the art.
27a Ratios The relative amount of drug and surface modifier can vary widely and the optimal amount of the surface modifier can depend upon, for example, the particular drug and surface modifier selected, the critical micelle concentration of the surface modifier if it forms micelles, the hydrophilic-lipophilic-balance (HLB) of the surface modifier, the melting point of the surface modifier, the water solubility of the surface modifier and/or drug , the surface tension of water solutions of the surface modifier, etc.
In the present invention, the optimal ratio of drug to surface modifier is about 1% to about 99% drug, more preferably about 30% to about 90% drug.
1o ****
The following examples are given to illustrate the present invention. It should be understood, however, that the invention is not to be limited to the specific 15 conditions or details described in these examples.
Example 1 The purpose of this example was to demonstrate the ability to aerosolize a concentrated nanoparticulate dispersion in an ultrasonic nebulizer which incorporates a 2o fine mesh screen in its design. An additional purpose of this example was to demonstrate that a therapeutic quantity of a concentrated nanoparticulate corticosteroid can be aerosolized in a very short period of time; e.g., two seconds or less.
Two different nanoparticulate dispersions of beclomethasone dipropionate (BDP) (1.25% and 10% BDP) were aerosolized using an ultrasonic nebulizer (Omron NE-25 U03 MicroAir~. The nebulizer generated droplets on a piezoelectric crystal and extruded them through a screen which contains ultrafine laser-drilled holes, producing an aerosol which has a very narrow particle size distribution in the range of approximately 1-5 Vim.
The device was connected to an Andersen cascade impactor with a flow rate at 28.3 liters per minute. For each formulation, the nebulizer was actuated for two seconds using a 30 programmable timer. The actuation time roughly corresponds to one inhalation cycle with a pMDI. After actuation, each stage of the impactor was analyzed for drug deposition by HPLC analysis.
The data indicate that substantial quantities of drug substance were found on stages 3-6 of the cascade impactor, corresponding to aerodynamic droplet sizes of about 0.7 to 4.7 Vim. The total amount of drug in the respirable droplet size range for deep lung delivery (i.e., particles less than about 2 microns; Stages 5, 6, and '~ was 11.72 pg for the 1.25% BDP (w/w) dispersion and 18.36 ~g for the 10% BDP (w/w) dispersion.
The total amount of drug in the respirable droplet size range for upper pulinonary delivery (i.e. particles about 2 to 5 microns; Stages 2, 3, 4, and 5) was 17.26 ~.g for the 1.25% BDP
1 o dispersion and 178.40 ~g for the 10% BDP dispersion.
One advantage provided by nanoparticulate formulations is that the drug particles are small enough to pass through the finer mesh channels of the nebulizer. In contrast, conventional micronized drug material would be expected to clog the orifices in the screen. Cascade impactor data from an in vitro deposition study of a nanoparticulate BDP dispersion aerosolized by an Omron NE-U03 Ultrasonic Nebulizer are summarized in Table I below:

TABLE I
Observed In-Vitro Deposition Pattern of an Aerosolized Nanoparticulate BDP Dispersion Deposition Droplet Size 1.25% BDP" 10% BDP' Site/ Range (pg Collected)(pg Collected) Impactor Area (pm)' Stage 0 9.0 -10.0 4.76 19.30 Stage 1 5.8 - 9.0 1.95 37.50 Stage 2 4.7 - 5.8 0.75 42.00 Stage 3 3.3 - 4.7 1.73 79.40 Stage 4 2.1 - 3.3 5.97 45.20 Stage 5 1.1 - 2.1 8.81 11.80 Stage 6 0.7 - 1.1 2.09 3.59 Stage 7 0.4 - 0.7 0.82 2.97 After Filter <0.4 2.25 18.70 Collar N/A 0.00 N/A

Induction PortN/A 4.10 22.40 Adapter N/A N/A N/A

Tube N/A N/A 10.98 -~rti resutcs oases on c secona acnxauon mm me umron N~-UUS.
dParticie Size of concentrate BDP 1.25% (w/w): mean of 171 nm, 90% < 234 nm, standard deviation 30 nm 'Particle Size of concentrate BDP 10% (w/w): mean of 94 nm, 90% < 130 nm, standard deviation 30 nm The results, which are graphically depicted in Figure 1, show substantial deposition of drug at Stages 2, 3, 4, and 5. This corresponds to delivery to conducting s airways. Most of the drug substance is found in droplets of about 2 to about 6 Vim, which are ideal for delivery to the bronchiole region.
Example 2 The purpose of this example was to demonstrate aerosolization of a l0 nanoparticulate dispersion using a using a jet nebulizer (Circulaire~, Westmed, Inc., Tucson, AZ), which can produce aqueous droplets in the size range of 0.5-2.0 pm. Such droplet sizes are suitable for delivery to the alveolar region of the lung, l.
e., deep lung delivery.
A nanoparticulate dispersion of BDP was prepared by wet milling 15 micronized drug substance in an aqueous tyloxapol surface modifier solution until a satisfactory particle size distribution had been obtained. The formulation was evaluated by light scattering methods (Microtrac UPA, Leeds & Northrop) and was found to have a mean particle size of 139 nm, with 90% of the particles being less than 220 nm (volume statistics).
The delivery performance of the BDP/surface modifier dispersion in a jet nebulizer was evaluated as follows: Approximately 3.5 ml of the BDP/surface modifier dispersion (2 mg/ml) was added to the nebulizer bowl, and the nebulizer mouthpiece was connected to the throat of a cascade impa.ctor apparatus with an airtight seal. The nebulizer and cascade impactor were then operated under suitable pressure and flow conditions for approximately 4 minutes using a 4 seconds on/4 seconds off cycle. Upon completion of the nebulization, each section of the apparatus was rinsed with acetonitrile and the washings diluted volumetrically.
The quantity of drug substance present in each section of the apparatus was determined by high performance liquid chromatography.
Results Analysis of the chromatograms showed that relatively little drug substance was deposited in the higher regions of the cascade impactor apparatus, while substantial quantities of material appeared on stages S-7, as well as on the exit filter.
In Experiment 1, approximately 92% of the emitted dose (ex-device) was contained in droplets < 2.1 ~n in diameter; in Experiment 2 the value was 86%. The results indicate that substantial quantities of drug substance were found on cascade impactor stages 5, 6, and 7, corresponding to droplet sizes of about 0.43 to about 2.1 microns. The smallest drug particle size normally accessible by conventional micronization methods for raw materials is about 2 to 3 microns, which is clearly larger than the droplets generated by this jet nebulizer. Detailed results of the cascade impactor study are presented Table II below, and graphically in Figure 2.

TABLE II
Observed In Vitro Deposition Pattern of a Nanoparticulate BDP Suspension Deposition Droplet Size Experiment Experiment Site Range (pm) 1' 2' Throat 33.13 36.00 Preselector, > 9.0 17.64 65.27 Stage 0 Stages 1 and 4.7 - 9.0 19.90 80.69 Stage 3 3.3 - 4.7 8.76 55.59 Stage 4 2.1- 3.3 2.13 17.90 Stage S 1.1- 2.1 122.41 336.16 Stage 6 0.65 -1.1 354.20 580.20 Stage 7 0.43 - 0.65 286.42 376.11 filter < 0.43 297.60 297.15 TOTAL 1142.19 1845.07 'fig Of BDP COlleCted In contrast to Example 1, which used an ultrasonic nebulizer (Omron NE-U03 MicroAir~ that generates droplets in the range of 2-6 ~.m, this example used a jet nebulizer that generates droplets in the range of < 2 Vim. The successful deposition of aerosol drug particles at Stages 6 and 7 demonstrates the effectiveness of using such compositions for deep lung delivery.
to Example 3 The purpose of this example was to demonstrate the preparation of a nanoparticulate dry powder for use in a DPI.
40.0% (w/w) naproxen, 4.00% (w/w) PVP K29/30 {a surface modifier), and 56.0% (w/w) deionized water were milled with 500 pm SDy-20 polymeric media for 7.5 hours to achieve a mean particle size of 254 nm, with 90% of the particles having a size of less than 335 nm. The material was diluted to 20% (w/w) naproxen and further milled with 50 p,m SDy-20 media for a period of 6 hours to yield a mean particle size of 155 nm, with 90% of the particles having a particle size of less than 212 nm.
The nanoparticulate dispersion was then diluted to 2% (w/w) naproxen with sufficient quantities of Sterile Water for Injection. The suspension was then spray-dried using a Yamato GB-22 operating with the following parameters:

Inlet Temp.: 130C

Outlet Temp.: 71 - 76C

Drying Air. 0.37 m3/min.

Atom. Air: 2 M Pa Pump Speed: ca. 8.4 mL/min.

The resultant nanoparticulate powder possessed a M1VIAD of 1.67 Vim, with 90% of the particles having a NIMAD of less than 2.43 p,m, as determined by a time-of flight particle sizing instrument. See Figure 3, which shows the volume distribution by the aerodynamic diameter of the spray-dried naproxen aerosol. Thus, all particles fell 1o within the respirable size range required for pulmonary deposition.
Additionally, greater than 50 percent of the particle population fell within the size required for peripheral lung deposition (alveolar, < 2 pm).
Interestingly, the spray-dried drug particles also demonstrated a spherical shape, which will improve the flow properties of the powder (as compared to prior micronized spray-dried powder formulations). The electron micrograph of Figure clearly shows the overall uniformity of size and the spherical nature of the particles. In addition, the exterior surface of the drug particle, which is composed of the polymeric stabilizer, may have advantages in limiting moisture uptake upon storage.
Lastly, to demonstrate that these spray-dried particles are constructed of 2o aggregates of the original nanoparticulate drug, reconstitution in a liquid medium resulted in the return to the original nanoparticulate dispersion, with a mean particle size of 184 nm, and 90 % of the particles having a size of less than 255 nm.
Example 4 The purpose of this example was to fiirther demonstrate the ability to influence the aerodynamic size of the spray-dried nanoparticulate composition by using a different concentration of nanoparticulate drug dispersion.
The concentration of naproxen and surface modifier (PVP K29/30) was the same as in Example 5, except that the composition was diluted with Sterile Water for Injection to achieve a 5 % (w/w) naproxen suspension. The spray-drier used was the Yamato GB-22, with the same operating parameters used in Example 4.

The resultant powder was composed of nanoparticulate aggregates with a MMAD of 2.91 pm, with 90% of the drug particles having a M1VIAD of less than 4.65 pm. This material is within the desired range for inhaled pulmonary deposition and may be more suitable for central airway targeting, i.e., within a range of 2 to 6 pm. See Figure 5, which shows the volume distribution by the aerodynamic diameter of the spray-dried naproxen aerosol.
Example 5 The purpose of this example was to produce a spray-dried nanoparticulate to powder for aerosol administration.
20.0% (w/w) triamcinolone acetonide (TA), 2.00% (w/w) HPC-SL (a surface modifier), 0.01% (w/w) benzalkonium chloride (BKC), and 76.9% (w/w) deionized water was milled in the presence of 500 ~,m SDy-20 polymeric media for approximately one hour. The final drug mean particle size was 169 nm, with 90 % of the drug particles having a size of less than 259 nm. The nanoparticulate drug dispersion was then diluted to 10% (w/w) TA with a 0.01 % BKC solution. The dispersion was then spray-dried using a Buchi B-191 spray-drier at the following settings:
Inlet Temp.: 130°C
Outlet Temp. 76°C
2o Aspirator setting: 90% capacity Product feed: 18% capacity The resultant nanoparticulate powder possessed aggregates of nanoparticulate TA particles with a MMAD of 5.54 p,m, and 90 % of the TA
particles had a MMAD of less than 9.08 ~m via a time-of flight measuring system. Thus, 50 percent of the particles fall within the respirable range for central airway (bronchiole deposition).
See Figure 6, which shows the volume distribution by the aerodynamic diameter of the spray-dried TA aerosol. In addition, the TA powder was of spherical shape as compared to the jet-milled drug, thus affording improved flow properties. Lastly, the powder redisperses in liquid medium to achieve well-dispersed nanoparticles of drug at a mean 3o particle size of 182 nm.

Ezample 6 The purpose of this example was to produce a spray-dried nanoparticulate drug/surface modifier powder for aerosol administration, wherein the composition lacks a diluent. In addition, this example compares the deposition of the nanoparticulate powder with the deposition of a micronized drug substance in a dry-powder delivery device.
10% (w/w) budesonide, 1.6% (w/w) HPMC (surface modifier), and 88.4% (w/w) deionized water were milled in the presence of 500 pm SDy-20 polymeric media for 1.5 hours. The resultant mean particle size was 166 nm, with 90% of the particles having a size of less than 233 nm. The nanoparticulate dispersion was then diluted to 0.5% (w/w) 1o budesonide with deionized water. The dispersion was spray-dried using a Yamato GB-22 spray-dryer operating at the following parameters:
Inlet Temperature: 125°C
Drying Air: 0.40 m3/minute Atomizing Air: 0.2 MPa Outlet Temperature: 60-61 °C
The resultant nanoparticulate aggregates possessed a MMAD of 1.35 pm, with 90% of the particles having a MIVIAD of less than 2.24 ~.m, as measured by time-of flight methodology.
2o A final powder blend was made, composed of 4% (w/w) nanoparticulate budesonide/surface modifier (3.2% (w/w) drug) and 96% lactose. The mixing was carried out using a Patterson-Kelley V-Blender with Lexan shell. The same procedure was followed for micronized budesonide at 3.4% (w/w) drug (Sicor, Via Terrazano 77, Italy).
Each drug powder - the nanoparticulate and the micronized - was then loaded into a ClickhalerTM (NIL, Laboratories plc, England), having a 1.5 mm3 dosing chamber. Each unit was evaluated using an Andersen cascade impactor operating at appmxirnately 60 liters per minute. Five actuations were delivered to the impactor and the unit was then disassembled and the collection plates analyzed via HPLC.
This was performed in triplicate. The data as percent of emitted dose from the DPI is shown below 3o in Table III.

TABLE III
In vitro Deposition of Nanoparticulate Budesonide vs Micronized Budesonide in a DPI' Impactor RegionAerodynamic Nanoparticulate Micronized Particle Size Budesonide Bndesonide Range tl~m) Stage 0 5.9 - 10.0 14.1 16.7 Stage 1 4.1 - 5.9 1.03 5.31 Stage 2 3.2 - 4.1 3.09 4.76 Stage 3 2.1 - 3.2 14.9 7.74 Stage 4 1.4 - 2.1 26.7 5.73 Stage 5 0.62 - 1.4 12.1 3.48 Stage 6 0.35 - 0.62 2.22 N/D

Stage 7 0.15 - 0.35 0.39 N/D

After Filter <0.15 N/D N/D

Total Respirable<5.9 60.4 27.0 Total Systemic <2.1 41.4 9.21 Cone N/A 0.40 0.94 Induction Port N/A 12.7 44.0 Adapter N/A 12.4 11.3 'As percent of etmttea lose tnrougn aevice. ~ascaae lmpactor operates at ca.
au min.
The results indicate that the nanoparticulate budesonide powder delivered 60.4% of the dose to the respirable regions of the impactor, while only 27% of the micronized drug was delivered to the same region. Furthermore, 41.4% of the nanoparticulate aggregates were found in the region corresponding to alveolar lung deposition, in contrast to only 9.21 % for the micronized material. Thus, the spray-dried nanoparticulate aggregates were more efficiently aerosolized than the micronized drug.
~o About 450% more in vitro deposition was observed within the systemic region for the nanoparticulate aggregates as compared to the micronized drug blend (measured as percent of delivered dose). Electron micrographs of the nanoparticulate and micronized dry substance formulations are shown in Figure 7.

E~amole 7 The purpose of this example was to demonstrate the production of freeze-dried nanoparticulate drug compositions for use in aerosol formulations.
10.0% (w/w) of a novel anti-emetic, 2.00% (w/w) of Poloxamer 188~ (a surface modifier), 0.500% (w/w) PVP C-15, and 87.5% (w) of Sterile Water for Injection was milled in the presence of 500 p,m SDy-20 polymeric media for a period of 2 hours. A
composition having a mean particle size of 286 nm, with 90% of the particles having a size of less than 372 nm, was determined via the Horiba LA-910 particle sizer.
This material was then diluted to 5 % (w/w) drug with Sterile Water for Injection and subjected to 60 minutes milling with 50 p,m SDy-20 media. The final particle size obtained was 157 nm, with 90 % of the drug particles having a size of less than 267 nm, as determined via the Horiba LA-910. This dispersion was then utilized in a series of freeze-drying experiments below.
The freeze-dryer utilized was an FTS Dura-Stop system with operating parameters as follows:
Product freeze temperature: -30°C (2 hours hold) Primary Drying:

1. Shelf temperature-25C
set:

Chamber vacuum:100 mT

Hold time: 2000 min.

2. Shelf temp.: -10C

Chamber vacuum:100 mT

Hold time: 300 min.

3. Shelf temp.: 0C

Chamber vacuum:100 mT

Hold time: 300 min.

4. Shelf temp.: 20C

Chamber vacuum:50 mT

Hold time: 800 min.

Example 7A
The following freeze-dried material was reconstituted in deionized water and examined for particle size distribution via the Horiba LA-910 particle analyzer:
5.00% (w/w) novel anti-emetic, 5.00% (w/w) dextrose, 1.00% (w/w) Poloxamer 188~, 0.250% (w/w) PVP C-15, and 88.8% {w/w) Sterile Water for Injection.
The average particle size of the reconstituted nanoparticulate dispersion was 4.23 pm, with 90% of the particles having an average particle size of less than 11.8 p.m. The resultant material demonstrates that aggregates were present in. the freeze-dried material having suitable particle sizes for pulmonary deposition. See Figure 8, which 1o shows the particle size distribution of the freeze-dried anti-emetic aerosol. (For this example, the particle sizes were measured by weight.) Example 7B
The following freeze-dried material was reconstituted in deionized water 15 and examined for particle size distribution via the Horiba LA-910 particle analyzer:
1.00% (w/w) novel anti-emetic, 5.00% (w/w) mannitol, 0.200% (w/w) Poloxamer 188, 0.050% (w/w) PVP C-15, and 93.8% (w/w) Sterile Water for Injection.
The resultant powder when reconstituted demonstrated an average particle size of 2.77 Vim, with 90% of the drug particles having an average particle size of less 20 than 7.39 Vim. Thus, aggregates of the nanoparticulate anti-emetic have a particle size within an acceptable range for pulmonary deposition after patient inhalation.
See Figure 9, which shows the particle size distribution of the freeze-dried anti-emetic aerosol. Also, if larger aggregates are generated (beyond about S to about 10 p,m), jet-milling may be employed to decrease the particle size distribution of the system for pulmonary 25 indications.
All of the dry powder inhalation systems can be utilized in either unit dose or mull-dose delivery devices, in either DPIs or pMDIs, and in nebulizer systems.

Example 8 The purpose of this prophetic example is to demonstrate the production of a propellant-based pMDI. This aerosol dosage form for pulmonary deposition has been the most routinely prescribed for asthma indications. The system is pressurized by using a propellant, such as a CFC or HFA (hydrofluorinated alkane), which functions as the delivery medium for a micronized drug. Additionally, a valve lubricant is present. These are typically the only components for suspension-based pMDIs. The micronized drug is jet-milled to the appropriate size for lung deposition (about 3 to about 5 p,m).
In contrast, the present invention is directed to the use of either discrete 1o nanoparticles or aggregates of nanoparticles. For preparation of discrete nanoparticulate drug, a non-aqueous milling medium is used, comprised of a high boiling point propellant. By employing a CFC-11 or trichloromonofluoromethane milling medium, nanoparticulate drug with suitable modifier can be made in a non-pressurized milling system. For example, the boiling point of CFC-11 is 23.7°C (according to the Merck 15 index). Thus, by maintaining the milling chamber temperature below 23.7°C, the CFC-11 remains intact during the size reduction process without developing internal pressure.
After the size reduction process, the propellant can be evaporated and reclaimed in a condenser. The resultant powder of nanoparticulate drug and surface modifier can then be resuspended in non-CFC propellants. Compounds HFA-134a 20 (tetrafluoroethane) and HFA-227 (heptafluoropropane) (Solway Fluorides, Inc., Greenwich, CT; Dupont Fluorochemicals, Wilmington, DE) are the most widely recognized non-CFC propellants. These can be pressure-filled into canisters containing the nanoparticulate drug and surface modifier.
25 Example 8A
The purpose of this example was to prepare a nanoparticulate aerosol formulation in a non-aqueous, non-pressurized milling system.
The following material was subjected to milling for 1.5 hrs with SDy-20 500 ~m polymeric media: 5.00% (w/w) triamcinolone acetonide (TA), 0.500% (w/w) 3o Span 85~ (surface modifier), and 94.5% (w/w) CFC-11. The resultant dispersion was then harvested and the propellant evaporated. A scanning electron microgragh was taken of the resultant powder to inspect for size reduction of the drug crystals.
See Figure 10.
Significant size reduction of drug particles was observed, and a large population of smaller drug crystals was found to be present. This material is of sufficient size to be respirable for inhaled administration via a pMDI or DPI system.
An exemplary corticosteroid formulation can comprise the following:
0.066%-(w/w) nanoparticulate TA, 0.034% (w/w) Span 85, and 99.9% HFA-134a.
Assuming a product density of 1.21 g/ml and a 50 p,1 metering valve, a theoretical delivery of 40 p,g TA is achieved. If necessary, this quantity can be modified to compensate for actuator efficiency. Ideally, the nanoparticulate powder can be dispensed into an appropriate container, followed by pressurized propellant filling, or a bulk slurry can be prepared and introduced into the final form by cold filling or pressure filling.
Example 9 15 The purpose of this example was to describe the use of a nanoparticulate aerosol in a propellant system operating at pressurized conditions. A
pressurized system allows the processing to progress at ambient room temperature.
The milling is conducted using either ball milling with ceramic/glass media or high-energy Dyno-milling with modifications to contain approximately 2o psig. The intent is to load the unit with chilled propellant and seal the sample ports. Thus, if the mill or roller bottle is at room temperature, the propellant will vaporize to maintain equilibrium within the containment system. A balance will be made between propellant in a liquid state and in a vapor state. This allows for milling in a liquid medium (the propellant) at temperatures above the propellant's boiling point.
25 Exemplary useful non-chlorinated propellants include HFA-134a (tetrafluoroethane), comprising about 50 to about 99.9% of final product weight, milling within pressure at/below 100 psig, and temperatures atJbelow 25°C; and (heptafluoropropane), comprising about 50 to about 99.9% of final product weight, milling within pressure at/below 53 psig, and temperatures at/below 25°C. In addition, 3o chlorinated propellants can be used in this embodiment. Exemplary chlorinated propellants include Freon-12 (dichlordifluoromethane), comprising about 50 to about 99.9% of milling composition, processed within pressure atlbelow 85 psig, and temperatures at/below 25°C; and Freon-114 (dichlorotetrafluoroethane), comprising about 50 to about 99.9% of milling slurry, processed at pressure at/below 19 psig, and s temperatures atJbelow 25°.
Example 9A
In this prophetic example, the following compounds can be combined for an exemplary budesonide aerosol composition to be used in a propellant system operating l0 at pressurized conditions: 5.00% (w/w) budesonide, 0.500% PVP C-15, and 94.5% (w/w) HFA-134a.
The nanoparticulate aerosol composition would be further diluted as necessary to obtain desired delivery doses.
~ 5 Example 9B
In this prophetic example, the following compounds can be combined for an exemplary TA aerosol composition to be used in a propellant system operating at pressurized conditions: 5.00% (w/w) TA, 0.500% PEG-400, and 94.5% (w/w) HFA-227.
The nanoparticulate aerosol composition would be further diluted as 20 necessary to obtain desired delivery doses.
Example 10 The purpose of this example was to demonstrate the use of powders comprising spray-dried or freeze-dried nanoparticulate aggregates or discrete 25 nanoparticulate particles in propellant systems for inhalation. The NiIVIAD
of the nanoparticulate aggregates would be about 0.5 p,m to about 6.0 wm, and the mean particle diameter of the discrete nanoparticulate drug particles would be about <1000 nm.. This allows for aqueous milling and subsequent water removal. The remaining powder can then be reconstituted with a propellant, such as those listed above.

The following can be combined for use in a propellant based system for inhalation: 0.704% (w/w) nanoparticulate agentlsurface modifier and 99.3%
(w/w) HFA-227. The resultant nanoparticulate powder is a spray-dried aggregate with a MMAD of 2.0 Vim. Based on a theoretical product density of 1.42 g/ml and a metering valve of 100 p,1, a dose of 1000 ~g could be expected through-the-valve.
****
It will be apparent to those skilled in the art that various modifications and variations can be made in the methods and compositions of the present invention without 1 o departing from the spirit or scope of the invention. Thus, it is intended that the present invention cover the modifications and variations of this invention provided they come within the scope of the appended claims and their equivalents.

Claims (28)

CLAIMS:
1. An aerosol composition comprising:
(a) aggregates of a spray-dried powder comprising nanoparticulate drug particles, wherein the nanoparticulate drug particles:
(i) comprise a poorly soluble crystalline drug, having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, (ii) at least 50% of the particles have a particle size of less than about 1000 nm, and (iii) have a surface modifier adsorbed on the surface thereof; and (b) the aggregates of spray-dried drug particles are less than or equal to about 100 microns in diameter, wherein the dry powder aggregates are formulated into an aerosol composition.
2. An aerosol composition comprising:
(a) aggregates of a freeze-dried powder comprising nanoparticulate drug particles, wherein the aggregates of freeze-dried drug are less than or equal to about 100 microns in diameter and the nanoparticulate drug particles:
(i) comprise a poorly soluble crystalline drug, having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, (ii) at least 50% of the particles have a particle size of less than about 1000 nm, and (iii) have a surface modifier adsorbed on the surface thereof, wherein the freeze-dried powder aggregates are formulated into an aerosol composition.
3. The aerosol composition of claim 2, further comprising spray-dried nanoparticulate drug powder, wherein the drug of the freeze-dried nanoparticulate drug powder is either the same or different from the drug of the spray-dried nanoparticulate drug powder.
4. The aerosol composition of claim 1 or 2, further comprising a diluent.
5. The aerosol composition of claim 4, wherein~
essentially every diluent particle comprises at least one embedded nanoparticulate drug particle having a surface modifier adhered to the surface of the drug particle.
6. An aerosol composition for use in a propellant-based pMDI comprising:
(a) dry powder aggregates of particles of a nanoparticulate poorly soluble crystalline drug having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, wherein the aggregates are less than or equal to about 100 microns in diameter, and wherein the drug particles:
(i) have a surface modifier absorbed on the surface thereof, and (ii) at least 50% of the particles have a particle size of less than about 1000 nm, and (b) a non-aqueous propellant, wherein the dry powder aggregates and non-aqueous propellant are formulated into a dry powder aerosol for use in a propellant-based pMDI.
7. The aerosol composition of claim 6, wherein the propellant is a non-CFC propellant.
8. An aerosol composition of an aqueous dispersion of nanoparticulate drug particles, wherein:
(a) essentially each droplet of the aerosol comprises at least one nanoparticulate poorly soluble drug particle;

(b) the droplets of the aerosol have a mass median aerodynamic diameter (MMAD) less than or equal to about 100 microns; and (c) the nanoparticulate drug particles comprise a poorly soluble drug having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, at least 50% of the particles have a particle size of less than about 1000 nm, and have a surface modifier adsorbed on the surface of the drug.
9. A nanoparticulate aerosol composition for use in a propellant-based pMDI comprising:

(a) a nanoparticulate poorly soluble crystalline drug having a solubility in at least one liquid dispersion medium of less than about 10 mg.ml, wherein the drug has a surface modifier adsorbed on the surface thereof, and at least 50% of the particles have a particle size of less than about 1000 nm, (b) essentially each droplet of the aerosol comprises at least one nanoparticulate drug particle, wherein the droplets of the aerosol generated by the pMDI
have a diameter less than or equal to about 100 microns, and (c) a non-aqueous propellant.
10. The aerosol composition of claim 9, wherein the propellant is a non-CFC propellant.
11. The aerosol composition of any one of claims 1 to 10, wherein the nanoparticulate drug particles have an effective average particle size selected from the group consisting of less than about 400 nm, less than about 300 nm, less than about 250 nm, less than about 100 nm, and less than about 50 nm.
12. The aerosol composition of any one of claims 1 to 11, wherein the aerosol comprises a concentration of a drug selected from the group consisting of about from about 0.05 mg/g up to about 900 mg/g; about 10 mg/g or more, about 100 mg/g or more, about 200 mg/g or more, about 400 mg/g or more, about 600 mg/g or more, and about 900 mg/g.
13. The aerosol composition of any one of claims 1 to 12, wherein the aggregates of the nanoparticulate drug particles, or the droplets of the aerosol, have a mass median aerodynamic diameter selected from the group consisting of about 5 to about 100 microns, about 30 to about 60 microns, about 2 to about 10 microns, about 2 to about 6 microns, and less than about 2 microns.
14. The aerosol composition of any one of claims 1 to 13, wherein the drug is selected from the group consisting of proteins, peptides, bronchodilators, corticosteroids, elastase inhibitors, analgesics, anti-fungals, cystic-fibrosis therapies, asthma therapies, emphysema therapies, respiratory distress syndrome therapies, chronic bronchitis therapies, chronic obstructive pulmonary disease therapies, organ-transplant rejection therapies, therapies for tuberculosis and other infections of the lung, fungal infection therapies, and respiratory illness therapies associated with acquired immune deficiency syndrome, an oncology drug, an anti-emetic, an analgesic, and a cardiovascular agent.
15. An aerosol composition according to any one of claims 1 to 14, wherein the aerosol comprises drug at a concentration of 10 mg/g or greater, and wherein the composition is for administration to a patient utilizing a delivery time of about 15 seconds or less.
16. A method of making an aerosol composition comprising:
(a) forming an aqueous nanoparticulate dispersion of a poorly soluble drug having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, wherein:
(i) the dispersion comprises poorly soluble crystalline drug particles and a surface modifier adsorbed on the surface thereof, and (ii) at least 50% of the particles have a particle size of less than about 1000 nm;
(b) spray-drying the nanoparticulate dispersion to form a dry powder of aggregates of the nanoparticulate drug and surface modifier particles, wherein the aggregates have a diameter of less than or equal to about 100 microns; and (c) formulating the dry powder aggregates into an aerosol composition.
17. The method of claim 16, further comprising adding a diluent to the nanoparticulate dispersion prior to spray-drying, wherein following spray-drying essentially every diluent particle contains at least one embedded drug particle and a surface modifier.
18. A method of making an aerosol composition comprising:
(a) milling under non-pressurized conditions in a non-aqueous medium having a high boiling point the following:
(i) a poorly soluble crystalline drug having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, and (ii) a surface modifier, to obtain a nanoparticulate drug composition wherein at least 50% of the particles have a particle size of less than about 1000 nm, (b) evaporating the non-aqueous medium to obtain a dry powder of aggregates of drug and surface modifier particles, wherein the aggregates have a diameter of less than or equal to about 100 microns; and (c) formulating the dry powder aggregates into an aerosol composition.
19. A method of making an aerosol composition comprising:
(a) milling under pressurized conditions in a non-aqueous medium the following:

(i) a poorly soluble crystalline drug having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, and (ii) a surface modifier, to obtain a drug particle wherein at least 50% of the particles have a particle size of less than about 1000 nm, (b) evaporating the non-aqueous medium to obtain a dry powder of aggregates of drug and surface modifier particles, wherein the aggregates have a diameter of less than or equal to about 100 microns; and (c) formulating the dry powder aggregates into an aerosol composition.
20. A method of making an aerosol composition comprising:
(a) forming an aqueous nanoparticulate dispersion of a poorly soluble drug having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, wherein:
(i) the dispersion comprises poorly soluble crystalline drug particles, wherein at least 50% of the drug particles have a particle size of less than about 1000 nm, and (ii) a surface modifier adsorbed on the surface thereof;
(b) freeze-drying the nanoparticulate dispersion to form a dry powder of aggregates of the nanoparticulate drug and surface modifier particles, wherein the aggregates have a diameter of less than or equal to about 100 microns;
and (c) formulating the freeze-dried powder aggregates into an aerosol composition.
21. The method of claim 20, further comprising adding a diluent to the nanoparticulate dispersion prior to freeze-drying, wherein following freeze-drying essentially every diluent particle contains at least one embedded drug particle and a surface modifier.
22. A method of making an aerosol composition comprising an aqueous dispersion of nanoparticulate drug particles, wherein said nanoparticulate drug particles comprise a poorly soluble drug having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, at least 50% of the particles have a particle size of less than about 1000 nm, and have a non-crosslinked surface modifier adsorbed on the surface thereof, wherein the method comprises:
(a) providing an aqueous dispersion of said nanoparticulate drug particles; and (b) forming an aerosol comprising liquid droplets of said dispersion, wherein:
(i) essentially each droplet of the aerosol comprises at least one nanoparticulate poorly soluble drug particle and at least one surface modifier adsorbed to the surface of the drug particle, and (ii) the liquid droplets forming the aerosol have a mass mean aerodynamic diameter of less than about 100 microns.
23. A method of making an aerosol composition of nanoparticulate drug particles for use in a propellant-based pMDI, wherein said nanoparticulate drug particles comprise a poorly soluble drug having a solubility in at least one liquid dispersion medium of less than about 10 mg/ml, at least 50% of the particles have a particle size of less than about 1000 nm, and have a non-crosslinked surface modifier adsorbed on the surface thereof, wherein the method comprises:
(a) providing a dispersion of said nanoparticulate drug particles in a liquid propellant; and (b) forming an aerosol comprising liquid droplets of said dispersion, wherein:
(i) essentially each droplet of the aerosol comprises at least one nanoparticulate poorly soluble drug particle and at least one surface modifier adsorbed to the surface of the drug particle, and (ii) the liquid droplets forming the aerosol have a mass mean aerodynamic diameter of less than about 100 microns.
24. The method of claim 16, 18, 19, 20, 22 or 23, wherein the nanoparticulate drug particles have an effective average particle size selected from the group consisting of less than about 400 nm, less than about 300 nm, less than about 250 nm, less than about 100 nm, and less than about 50 nm.
25. The method of claim 16, 18, 19, 20, 22 or 23, wherein the aerosol comprises a concentration of a drug selected from the group consisting of about from about 0.05 mg/g up to about 900 mg/g; about 10 mg/g or more, about 100 mg/g or more, about 200 mg/g or more, about 400 mg/g or more, about 600 mg/g or more, and about 900 mg/g.
26. The method of claim 16, 18, 19, 20, 22 or 23, wherein the aggregates of the nanoparticulate drug particles, or the droplets of the aerosol, have a mass median aerodynamic diameter selected from the group consisting of about 5 to about 100 microns, about 30 to about 60 microns, about 2 to about 10 microns, about 2 to about 6 microns, and less than about 2 microns.
27. The method of claim 16, 18, 19, 20, 22 or 23, wherein the drug is selected from the group consisting of proteins, peptides, bronchodilators, corticosteroids, elastase inhibitors, analgesics, anti-fungals, cystic-fibrosis therapies, asthma therapies, emphysema therapies, respiratory distress syndrome therapies, chronic bronchitis therapies, chronic obstructive pulmonary disease therapies, organ-transplant rejection therapies, therapies for tuberculosis and other infections of the lung, fungal infection therapies, and respiratory illness therapies associated with acquired immune deficiency syndrome, an oncology drug, an anti-emetic, an analgesic, and a cardiovascular agent.
28. The method according to claim 16, 18, 19, 20, 22 or 23, wherein the aerosol comprises drug at a concentration of 10 mg/g or greater, and wherein the composition is for administration to a patient utilizing a delivery time of about 15 seconds or less.
CA002350074A 1998-11-12 1999-11-12 Aerosols comprising nanoparticle drugs Expired - Fee Related CA2350074C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/190,138 1998-11-12
US09/190,138 US7521068B2 (en) 1998-11-12 1998-11-12 Dry powder aerosols of nanoparticulate drugs
PCT/US1999/026799 WO2000027363A1 (en) 1998-11-12 1999-11-12 Aerosols comprising nanoparticle drugs

Publications (2)

Publication Number Publication Date
CA2350074A1 CA2350074A1 (en) 2000-05-18
CA2350074C true CA2350074C (en) 2004-03-23

Family

ID=22700162

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002350074A Expired - Fee Related CA2350074C (en) 1998-11-12 1999-11-12 Aerosols comprising nanoparticle drugs

Country Status (12)

Country Link
US (4) US7521068B2 (en)
EP (2) EP1128814B1 (en)
JP (1) JP4758548B2 (en)
AT (2) ATE493971T1 (en)
AU (1) AU1346900A (en)
CA (1) CA2350074C (en)
CY (2) CY1111344T1 (en)
DE (1) DE69943112D1 (en)
DK (2) DK2263652T3 (en)
ES (2) ES2387749T3 (en)
PT (1) PT2263652E (en)
WO (1) WO2000027363A1 (en)

Families Citing this family (213)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0810853B1 (en) * 1995-02-24 2004-08-25 Elan Pharma International Limited Aerosols containing nanoparticle dispersions
UA72189C2 (en) 1997-11-17 2005-02-15 Янссен Фармацевтика Н.В. Aqueous suspensions of 9-hydroxy-risperidone fatty acid esters provided in submicron form
US20080102121A1 (en) * 1998-11-02 2008-05-01 Elan Pharma International Limited Compositions comprising nanoparticulate meloxicam and controlled release hydrocodone
US7521068B2 (en) * 1998-11-12 2009-04-21 Elan Pharma International Ltd. Dry powder aerosols of nanoparticulate drugs
US6375986B1 (en) 2000-09-21 2002-04-23 Elan Pharma International Ltd. Solid dose nanoparticulate compositions comprising a synergistic combination of a polymeric surface stabilizer and dioctyl sodium sulfosuccinate
US6969529B2 (en) 2000-09-21 2005-11-29 Elan Pharma International Ltd. Nanoparticulate compositions comprising copolymers of vinyl pyrrolidone and vinyl acetate as surface stabilizers
US20020061281A1 (en) * 1999-07-06 2002-05-23 Osbakken Robert S. Aerosolized anti-infectives, anti-inflammatories, and decongestants for the treatment of sinusitis
US6576224B1 (en) * 1999-07-06 2003-06-10 Sinuspharma, Inc. Aerosolized anti-infectives, anti-inflammatories, and decongestants for the treatment of sinusitis
AU2001234005B2 (en) * 2000-02-28 2006-01-19 Pharmakodex Limited Improvements in or relating to the delivery of oral drugs
GB0009773D0 (en) * 2000-04-19 2000-06-07 Univ Cardiff Particulate composition
US8512718B2 (en) 2000-07-03 2013-08-20 Foamix Ltd. Pharmaceutical composition for topical application
DE10043509A1 (en) * 2000-09-01 2002-03-14 Asta Medica Ag Solid peptide preparations for inhalation and their manufacture
US6613308B2 (en) * 2000-09-19 2003-09-02 Advanced Inhalation Research, Inc. Pulmonary delivery in treating disorders of the central nervous system
US7198795B2 (en) 2000-09-21 2007-04-03 Elan Pharma International Ltd. In vitro methods for evaluating the in vivo effectiveness of dosage forms of microparticulate of nanoparticulate active agent compositions
US7998507B2 (en) 2000-09-21 2011-08-16 Elan Pharma International Ltd. Nanoparticulate compositions of mitogen-activated protein (MAP) kinase inhibitors
DE10058772A1 (en) * 2000-11-27 2002-06-06 Falk Pharma Gmbh Process for the preparation of readily soluble pharmaceutical formulations and corresponding formulations
DK2283818T3 (en) * 2000-11-30 2017-10-16 Vectura Ltd Process for preparing particles for use in a pharmaceutical composition
WO2002043700A2 (en) 2000-11-30 2002-06-06 Vectura Limited Particles for use in a pharmaceutical composition
US20040052733A1 (en) * 2000-11-30 2004-03-18 Staniforth John Nicholas Pharmaceutical compositions for inhalation
ES2474199T3 (en) * 2000-11-30 2014-07-08 Vectura Limited Pharmaceutical compositions for inhalation
US9700866B2 (en) 2000-12-22 2017-07-11 Baxter International Inc. Surfactant systems for delivery of organic compounds
US8067032B2 (en) 2000-12-22 2011-11-29 Baxter International Inc. Method for preparing submicron particles of antineoplastic agents
GB0107106D0 (en) * 2001-03-21 2001-05-09 Boehringer Ingelheim Pharma Powder inhaler formulations
US20040101483A1 (en) * 2001-03-30 2004-05-27 Rudi Muller-Walz Medical aerosol formulations
WO2002094229A1 (en) * 2001-05-24 2002-11-28 Alexza Molecular Delivery Corporation Delivery of muscle relaxants through an inhalation route
WO2002094246A2 (en) * 2001-05-24 2002-11-28 Alexza Molecular Delivery Corporation Delivery of nonsteroidal antiinflammatory drugs through an 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
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
DK1429731T3 (en) * 2001-09-19 2007-05-14 Elan Pharma Int Ltd Nanoparticle formulations containing insulin
BR0212833A (en) 2001-09-26 2004-10-13 Baxter Int Preparation of submicron sized nanoparticles by dispersion and solvent or liquid phase removal
US20060003012A9 (en) 2001-09-26 2006-01-05 Sean Brynjelsen Preparation of submicron solid particle suspensions by sonication of multiphase systems
EP1458361A4 (en) * 2001-11-20 2007-04-25 Advanced Inhalation Res Inc Compositions for sustained action product delivery
WO2003057188A1 (en) 2001-11-21 2003-07-17 Alexza Molecular Delivery Corporation Delivery of caffeine through an inhalation route
US20030129242A1 (en) * 2002-01-04 2003-07-10 Bosch H. William Sterile filtered nanoparticulate formulations of budesonide and beclomethasone having tyloxapol as a surface stabilizer
EP3536344B1 (en) 2002-03-01 2020-02-19 Chiesi Farmaceutici S.p.A. Formoterol superfine formulation
CA2479665C (en) * 2002-03-20 2011-08-30 Elan Pharma International Ltd. Nanoparticulate compositions of angiogenesis inhibitors
US20030185762A1 (en) * 2002-03-22 2003-10-02 Cowan Siu Man L. Highly aqueous liquid carrier formulations
GB0219815D0 (en) 2002-08-24 2002-10-02 Accentus Plc Preparation of small crystals
FI20020857A (en) * 2002-05-07 2003-11-08 Focus Inhalation Oy Conversion of amorphous material to a corresponding crystalline material by spray drying and utilization of the crystalline spray-dried material in drug formulations
IL165291A0 (en) * 2002-06-17 2005-12-18 Epigenesis Pharmaceuticals Llc Dihydrate dehydroepiandorosterone and methods of treating asthma or chronic obstructive pulmonary disease using compositions thereof
US20040258757A1 (en) * 2002-07-16 2004-12-23 Elan Pharma International, Ltd. Liquid dosage compositions of stable nanoparticulate active agents
US20040045546A1 (en) * 2002-09-05 2004-03-11 Peirce Management, Llc Pharmaceutical delivery system for oral inhalation through nebulization consisting of inert substrate impregnated with substance (S) to be solubilized or suspended prior to use
EP1556091A1 (en) * 2002-10-04 2005-07-27 Elan Pharma International Limited Gamma irradiation of solid nanoparticulate active agents
IL152486A0 (en) 2002-10-25 2003-05-29 Meir Eini Alcohol-free cosmetic and pharmaceutical foam carrier
US10117812B2 (en) 2002-10-25 2018-11-06 Foamix Pharmaceuticals Ltd. Foamable composition combining a polar solvent and a hydrophobic carrier
US8119109B2 (en) 2002-10-25 2012-02-21 Foamix Ltd. Foamable compositions, kits and methods for hyperhidrosis
US7704518B2 (en) 2003-08-04 2010-04-27 Foamix, Ltd. Foamable vehicle and pharmaceutical compositions thereof
EP1556009B2 (en) 2002-10-25 2021-07-21 Foamix Pharmaceuticals Ltd. Cosmetic and pharmaceutical foam
US8486376B2 (en) 2002-10-25 2013-07-16 Foamix Ltd. Moisturizing foam containing lanolin
US20080138296A1 (en) 2002-10-25 2008-06-12 Foamix Ltd. Foam prepared from nanoemulsions and uses
US9265725B2 (en) 2002-10-25 2016-02-23 Foamix Pharmaceuticals Ltd. Dicarboxylic acid foamable vehicle and pharmaceutical compositions thereof
US7700076B2 (en) 2002-10-25 2010-04-20 Foamix, Ltd. Penetrating pharmaceutical foam
US9668972B2 (en) 2002-10-25 2017-06-06 Foamix Pharmaceuticals Ltd. Nonsteroidal immunomodulating kit and composition and uses thereof
US8119150B2 (en) 2002-10-25 2012-02-21 Foamix Ltd. Non-flammable insecticide composition and uses thereof
US9211259B2 (en) 2002-11-29 2015-12-15 Foamix Pharmaceuticals Ltd. Antibiotic kit and composition and uses thereof
US8900554B2 (en) 2002-10-25 2014-12-02 Foamix Pharmaceuticals Ltd. Foamable composition and uses thereof
US7820145B2 (en) 2003-08-04 2010-10-26 Foamix Ltd. Oleaginous pharmaceutical and cosmetic foam
US20040208833A1 (en) * 2003-02-04 2004-10-21 Elan Pharma International Ltd. Novel fluticasone formulations
JP4707937B2 (en) * 2003-02-28 2011-06-22 ホソカワミクロン株式会社 Method for producing drug-containing composite particles and transpulmonary preparation
US8912174B2 (en) 2003-04-16 2014-12-16 Mylan Pharmaceuticals Inc. Formulations and methods for treating rhinosinusitis
US9808471B2 (en) * 2003-04-16 2017-11-07 Mylan Specialty Lp Nasal pharmaceutical formulations and methods of using the same
US7811606B2 (en) * 2003-04-16 2010-10-12 Dey, L.P. Nasal pharmaceutical formulations and methods of using the same
US7575739B2 (en) 2003-04-28 2009-08-18 Foamix Ltd. Foamable iodine composition
ES2370395T3 (en) 2003-05-21 2011-12-15 Alexza Pharmaceuticals, Inc. USE OF A SOLID FUEL LAYER, MANUFACTURING PROCEDURE AND CORRESPONDING HEATING UNIT.
CA2523035C (en) * 2003-05-22 2011-04-26 Elan Pharma International Ltd. Sterilization of dispersions of nanoparticulate active agents with gamma radiation
US20040242729A1 (en) * 2003-05-30 2004-12-02 3M Innovative Properties Company Stabilized particle dispersions containing surface-modified inorganic nanoparticles
US7459146B2 (en) * 2003-05-30 2008-12-02 3M Innovative Properties Company Stabilized aerosol dispersions
US7109247B2 (en) * 2003-05-30 2006-09-19 3M Innovative Properties Company Stabilized particle dispersions containing nanoparticles
DE10325989A1 (en) * 2003-06-07 2005-01-05 Glatt Gmbh Process for the preparation of and resulting micropellets and their use
US20060251584A1 (en) * 2003-06-11 2006-11-09 Nara Machinery Co., Ltd. Drug nano-particle, method and apparatus for preparing pharmaceutical preparation using the particle
WO2004112799A1 (en) * 2003-06-13 2004-12-29 Chrysalis Technologies Incorporated Methods and apparatus for producing nanoscale particles
US8486374B2 (en) 2003-08-04 2013-07-16 Foamix Ltd. Hydrophilic, non-aqueous pharmaceutical carriers and compositions and uses
US8795693B2 (en) 2003-08-04 2014-08-05 Foamix Ltd. Compositions with modulating agents
US20060292081A1 (en) * 2003-09-15 2006-12-28 Vectura Limited Methods for preparing pharmaceutical compositions
GB0327723D0 (en) * 2003-09-15 2003-12-31 Vectura Ltd Pharmaceutical compositions
US20060263350A1 (en) * 2003-09-26 2006-11-23 Fairfield Clinical Trials Llc Combination antihistamine medication
GB0323684D0 (en) 2003-10-09 2003-11-12 Jagotec Ag Improvements in or relating to organic compounds
WO2005077339A1 (en) * 2004-02-10 2005-08-25 E.I. Dupont De Nemours And Company Process for preparing stable sol of pharmaceutical ingredients and hydrofluorocarbon comprising milling the said sol, then transfert it into a canister
AU2005219413A1 (en) * 2004-03-02 2005-09-15 Massachusetts Institute Of Technology Nanocell drug delivery system
US20070053845A1 (en) * 2004-03-02 2007-03-08 Shiladitya Sengupta Nanocell drug delivery system
AU2005244128B2 (en) * 2004-05-07 2009-06-25 President And Fellows Of Harvard College Pulmonary malarial vaccine
US20060009435A1 (en) * 2004-06-23 2006-01-12 Joseph Kaspi Synthesis and powder preparation of fluticasone propionate
GB0418791D0 (en) * 2004-08-23 2004-09-22 Glaxo Group Ltd Novel process
US9061027B2 (en) * 2004-08-27 2015-06-23 Board Of Regents, The University Of Texas System Enhanced delivery of drug compositions to treat life threatening infections
US20060073173A1 (en) * 2004-10-04 2006-04-06 Maria Banach Large-scale manufacturing process for the production of pharmaceutical compositions
EP1809252A2 (en) 2004-10-29 2007-07-25 Presidents and Fellows of Harvard College Particles for treatment of pulmonary infection
US9492400B2 (en) 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
EP2623095A1 (en) * 2004-11-16 2013-08-07 Elan Pharma International Limited Injectable nanoparticulate olanzapine formulations
GB0425758D0 (en) 2004-11-23 2004-12-22 Vectura Ltd Preparation of pharmaceutical compositions
GB0426301D0 (en) * 2004-11-30 2004-12-29 Vectura Ltd Pharmaceutical formulations
UA89513C2 (en) * 2004-12-03 2010-02-10 Элан Фарма Интернешнл Лтд. Nanoparticulate raloxifene hydrochloride composition
CA2590675A1 (en) * 2004-12-15 2006-06-22 Elan Pharma International Ltd. Nanoparticulate tacrolimus formulations
WO2006069098A1 (en) * 2004-12-22 2006-06-29 Elan Pharma International Ltd. Nanoparticulate bicalutamide formulations
WO2006074218A2 (en) * 2005-01-06 2006-07-13 Elan Pharma International Ltd. Nanoparticulate candesartan formulations
US20060198896A1 (en) 2005-02-15 2006-09-07 Elan Pharma International Limited Aerosol and injectable formulations of nanoparticulate benzodiazepine
US20060204588A1 (en) * 2005-03-10 2006-09-14 Elan Pharma International Limited Formulations of a nanoparticulate finasteride, dutasteride or tamsulosin hydrochloride, and mixtures thereof
US20090110633A1 (en) * 2005-03-14 2009-04-30 Shiladitya Sengupta Nanocells for Diagnosis and Treatment of Diseases and Disorders
JP2008533174A (en) * 2005-03-16 2008-08-21 エラン ファーマ インターナショナル リミテッド Nanoparticulate leukotriene receptor antagonist / corticosteroid preparation
NZ561666A (en) * 2005-03-17 2010-05-28 Elan Pharma Int Ltd Nanoparticulate biphosphonate compositions
BRPI0609700A2 (en) * 2005-03-23 2010-04-20 Elan Pharma Int Ltd nanoparticulate corticosteroid and antihistamine formulations
US20060246141A1 (en) * 2005-04-12 2006-11-02 Elan Pharma International, Limited Nanoparticulate lipase inhibitor formulations
MX2007012778A (en) * 2005-04-12 2008-01-11 Elan Pharma Int Ltd Nanoparticulate quinazoline derivative formulations.
US20090081297A1 (en) * 2005-04-27 2009-03-26 Cook Robert O Use of surface tension reducing agents in aerosol formulations
DE112006001606T5 (en) 2005-06-08 2009-07-09 Elan Pharma International Ltd., Athlone Nanoparticulate and controlled release composition comprising cefditoren
US7947308B2 (en) * 2005-07-20 2011-05-24 Raimar Loebenberg Effervescent powders for inhalation
JP2007079857A (en) * 2005-09-13 2007-03-29 Canon Inc Server apparatus, client apparatuses and those control methods, computer program, storage medium
EP2279727A3 (en) 2005-09-15 2011-10-05 Elan Pharma International Limited Nanoparticulate aripiprazole formulations
EP1954246A4 (en) * 2005-11-11 2012-01-18 Biolab Sanus Farmaceutica Ltda Solid pharmaceutical composition comprising agglomerate nanoparticles and a process for producing the same
WO2007070682A2 (en) 2005-12-15 2007-06-21 Massachusetts Institute Of Technology System for screening particles
AU2006330627A1 (en) * 2005-12-16 2007-07-05 University Of Kansas Nanoclusters for delivery of therapeutics
EP1798504A1 (en) * 2005-12-19 2007-06-20 Koninklijke Philips Electronics N.V. Method of making dried particles
US20070178051A1 (en) * 2006-01-27 2007-08-02 Elan Pharma International, Ltd. Sterilized nanoparticulate glucocorticosteroid formulations
US20070202182A1 (en) * 2006-02-26 2007-08-30 Kane Kevin M Preparing solid formulation of nanoparticles of pharmaceutical ingredient, including at least micron-sized particles
US8367112B2 (en) * 2006-02-28 2013-02-05 Alkermes Pharma Ireland Limited Nanoparticulate carverdilol formulations
FI20060428L (en) * 2006-05-03 2007-11-04 Esko Kauppinen Surface-modified aerosol particles, method and device for their production and powders and dispersions containing said particles
WO2007133807A2 (en) 2006-05-15 2007-11-22 Massachusetts Institute Of Technology Polymers for functional particles
WO2007150030A2 (en) 2006-06-23 2007-12-27 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
AU2007264418B2 (en) 2006-06-30 2012-05-03 Iceutica Pty Ltd Methods for the preparation of biologically active compounds in nanoparticulate form
US20080260655A1 (en) 2006-11-14 2008-10-23 Dov Tamarkin Substantially non-aqueous foamable petrolatum based pharmaceutical and cosmetic compositions and their uses
CA2678455C (en) 2007-01-10 2019-02-12 Board Of Regents The University Of Texas System Enhanced delivery of immunosuppressive drug compositions for pulmonary delivery
WO2008098165A2 (en) 2007-02-09 2008-08-14 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
JP2008222567A (en) * 2007-03-08 2008-09-25 Iwate Univ Method for producing fine particles
WO2008112661A2 (en) 2007-03-09 2008-09-18 Alexza Pharmaceuticals, Inc. Heating unit for use in a drug delivery device
WO2008124632A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Amphiphilic compound assisted nanoparticles for targeted delivery
EP2142209B1 (en) * 2007-04-18 2018-10-31 Particle Sciences, Inc. Aerosol dispersions of particles with active pharmaceutical ingredients
EP2155166A2 (en) * 2007-05-11 2010-02-24 F. Hoffmann-Roche AG Pharmaceutical compositions for poorly soluble drugs
GB0712454D0 (en) * 2007-06-27 2007-08-08 Generics Uk Ltd Pharmaceutical compositions
US8633152B2 (en) 2007-08-07 2014-01-21 Nanomaterials Technology Pte Ltd Process for making micro-sized protein particles
US8636982B2 (en) 2007-08-07 2014-01-28 Foamix Ltd. Wax foamable vehicle and pharmaceutical compositions thereof
WO2009049159A1 (en) 2007-10-10 2009-04-16 Parion Sciences, Inc. Delivering osmolytes by nasal cannula
EP3424525A1 (en) 2007-10-12 2019-01-09 Massachusetts Institute Of Technology Vaccine nanotechnology
WO2009049304A1 (en) * 2007-10-12 2009-04-16 Map Pharmaceuticals, Inc. Inhalation drug delivery
US9439857B2 (en) 2007-11-30 2016-09-13 Foamix Pharmaceuticals Ltd. Foam containing benzoyl peroxide
WO2009073843A1 (en) * 2007-12-06 2009-06-11 Cytotech Labs, Llc Inhalable compositions having enhanced bioavailability
WO2009072007A2 (en) 2007-12-07 2009-06-11 Foamix Ltd. Carriers, formulations, methods for formulating unstable active agents for external application and uses thereof
WO2010041141A2 (en) 2008-10-07 2010-04-15 Foamix Ltd. Oil-based foamable carriers and formulations
US20090238867A1 (en) * 2007-12-13 2009-09-24 Scott Jenkins Nanoparticulate Anidulafungin Compositions and Methods for Making the Same
JP5570996B2 (en) 2007-12-14 2014-08-13 エアロデザインズ インコーポレイテッド Delivery of aerosolizable foodstuffs
CA2712120A1 (en) 2008-01-14 2009-07-23 Foamix Ltd. Poloxamer foamable pharmaceutical compositions with active agents and/or therapeutic cells and uses
EP2095816A1 (en) * 2008-02-29 2009-09-02 Schlichthaar, Rainer, Dr. Nanosuspension with antifungal medication to be administered via inhalation with improved impurity profile and safety
WO2009117410A2 (en) * 2008-03-17 2009-09-24 Board Of Regents, The University Of Texas System Formation of nanostructured particles of poorly water soluble drugs and recovery by mechanical techniques
WO2009158300A1 (en) * 2008-06-26 2009-12-30 3M Innovative Properties Company Dry powder pharmaceutical compositions for pulmonary administration, and methods of manufacturing thereof
US8277812B2 (en) 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
US8343498B2 (en) 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8591905B2 (en) 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
US8343497B2 (en) 2008-10-12 2013-01-01 The Brigham And Women's Hospital, Inc. Targeting of antigen presenting cells with immunonanotherapeutics
KR20100052262A (en) * 2008-11-10 2010-05-19 (주)아모레퍼시픽 Process for preparing powder comprising nanoparticles of sparingly soluble drug, powder prepared by same process and pharmaceutical composition comprising same powder
US20100119558A1 (en) * 2008-11-10 2010-05-13 Scandivir Ab Ngna compositions and methods of use
AU2010210620B2 (en) 2009-02-03 2016-02-25 Microbion Corporation Bismuth-thiols as antiseptics for epithelial tissues, acute and chronic wounds, bacterial biofilms and other indications
US9028878B2 (en) 2009-02-03 2015-05-12 Microbion Corporation Bismuth-thiols as antiseptics for biomedical uses, including treatment of bacterial biofilms and other uses
EP2400950B1 (en) 2009-02-26 2019-05-22 Glaxo Group Limited Pharmaceutical formulations comprising 4-{(1 r)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol
WO2010102065A1 (en) 2009-03-05 2010-09-10 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives
JP6027890B2 (en) 2009-04-24 2016-11-16 イシューティカ ピーティーワイ リミテッド New formulation of indomethacin
US20120097160A1 (en) * 2009-04-24 2012-04-26 Schering Corporation Agglomerate formulations including active pharmaceutical agents with targeted particle sizes
CA2760186C (en) 2009-04-28 2019-10-29 Foamix Ltd. Foamable vehicle and pharmaceutical compositions comprising aprotic polar solvents and uses thereof
JP6072539B2 (en) 2009-05-27 2017-02-01 アルカーメス ファーマ アイルランド リミテッド Reduction of flaky aggregation in nanoparticulate active agent compositions
AU2010254549B2 (en) 2009-05-27 2016-10-20 Selecta Biosciences, Inc. Nanocarriers possessing components with different rates of release
US20100310660A1 (en) * 2009-06-08 2010-12-09 Taipei Medical University Dry powder microparticles for pulmonary delivery
WO2011013008A2 (en) 2009-07-29 2011-02-03 Foamix Ltd. Non surface active agent non polymeric agent hydro-alcoholic foamable compositions, breakable foams and their uses
WO2011013009A2 (en) 2009-07-29 2011-02-03 Foamix Ltd. Non surfactant hydro-alcoholic foamable compositions, breakable foams and their uses
US20110033545A1 (en) * 2009-08-06 2011-02-10 Absize, Inc. Topical pharmaceutical preparations having both a nanoparticle solution and a nanoparticle suspension and methods for the treatment of acute and chronic pain therewith
US8871184B2 (en) 2009-10-02 2014-10-28 Foamix Ltd. Topical tetracycline compositions
US9849142B2 (en) 2009-10-02 2017-12-26 Foamix Pharmaceuticals Ltd. Methods for accelerated return of skin integrity and for the treatment of impetigo
GB0918149D0 (en) 2009-10-16 2009-12-02 Jagotec Ag Improved medicinal aerosol formulation
US8174881B2 (en) 2009-11-24 2012-05-08 Micron Technology, Inc. Techniques for reducing disturbance in a semiconductor device
GB0921075D0 (en) 2009-12-01 2010-01-13 Glaxo Group Ltd Novel combination of the therapeutic agents
GB201001070D0 (en) 2010-01-22 2010-03-10 St George's Hospital Medical School Theraputic compounds and their use
SG10202010355PA (en) 2010-03-12 2020-11-27 Berg Llc Intravenous formulations of coenzyme q10 (coq10) and methods of use thereof
ES2689520T3 (en) 2010-04-23 2018-11-14 Kempharm, Inc. Therapeutic formulation to reduce drug side effects
WO2011146583A2 (en) 2010-05-19 2011-11-24 Elan Pharma International Limited Nanoparticulate cinacalcet formulations
MX2012013713A (en) 2010-05-26 2013-01-28 Selecta Biosciences Inc Nanocarrier compositions with uncoupled adjuvant.
GB201011411D0 (en) 2010-07-06 2010-08-18 St Georges Hosp Medical School Therapeutic compounds and their use
US8815294B2 (en) 2010-09-03 2014-08-26 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives and a carrier material
AU2011305566A1 (en) * 2010-09-22 2013-05-02 Map Pharmaceuticals, Inc. Aerosol composition for administering drugs
EP2627317A4 (en) * 2010-10-15 2014-08-20 Glaxo Group Ltd Aggregate nanoparticulate medicament formulations, manufacture and use thereof
JOP20120023B1 (en) 2011-02-04 2022-03-14 Novartis Ag Dry powder formulations of particles that contain two or more active ingredients for treating obstructive or inflammatory airways diseases
GB201102237D0 (en) * 2011-02-09 2011-03-23 Kuecept Ltd Particle formulation
RU2013139701A (en) 2011-02-17 2015-03-27 Ф. Хоффманн-Ля Рош Аг METHOD FOR CONTROLLED CRYSTALLIZATION OF AN ACTIVE PHARMACEUTICAL INGREDIENT FROM THE STATE OF A COOLED LIQUID BY EXTRUSION OF HOT DECAY
US9084727B2 (en) 2011-05-10 2015-07-21 Bend Research, Inc. Methods and compositions for maintaining active agents in intra-articular spaces
CN103732213A (en) 2011-06-07 2014-04-16 帕里昂科学公司 Methods of treatment
JO3410B1 (en) * 2011-06-07 2019-10-20 Otsuka Pharma Co Ltd Freeze-dried aripiprazole formulation
US8945605B2 (en) * 2011-06-07 2015-02-03 Parion Sciences, Inc. Aerosol delivery systems, compositions and methods
MX351781B (en) 2011-06-17 2017-10-30 Berg Llc Inhalable pharmaceutical compositions.
EA201490381A1 (en) 2011-07-29 2014-06-30 Селекта Байосайенсиз, Инк. SYNTHETIC NANOSEAGES WHICH STIMULATE THE FORMATION OF HUMORAL IMMUNE RESPONSE AND IMMUNE RESPONSE MEDIATED BY CYTOTOXIC T-LYMPHOCYTES (CTL)
WO2013059676A1 (en) * 2011-10-21 2013-04-25 Subhash Desai Compositions for reduction of side effects
EP2601973A1 (en) 2011-12-09 2013-06-12 Laboratoires SMB SA Dry powder formulation of azole derivative for inhalation
KR20140135197A (en) * 2012-02-16 2014-11-25 카프니아, 인코포레이티드 Gas dispenser with diffusing nosepiece
CA2869849A1 (en) * 2012-04-13 2013-10-17 Glaxosmithkline Intellectual Property Development Limited Aggregate particles comprising nanoparticulate drug particles of umeclidinium bromide, vilanterol trifenatate and fluticasone furoate
BR112015009708A2 (en) * 2012-11-01 2017-07-04 Prec Valve Corporation free flow aerosol valve
US9757395B2 (en) 2012-12-20 2017-09-12 Otitopic Inc. Dry powder inhaler and methods of use
US9757529B2 (en) 2012-12-20 2017-09-12 Otitopic Inc. Dry powder inhaler and methods of use
GB201307659D0 (en) * 2013-04-26 2013-06-12 Korea Coast Guard Commissioner Preparation of drug particles by micronisation
EP3607941A1 (en) 2013-04-30 2020-02-12 Otitopic Inc. Dry powder formulations and methods of use
WO2015089105A1 (en) 2013-12-09 2015-06-18 Respira Therapeutics, Inc. Pde5 inhibitor powder formulations and methods relating thereto
JP6701086B2 (en) 2014-02-20 2020-05-27 オティトピック インコーポレイテッド Dry powder formulation for inhalation
US9526734B2 (en) 2014-06-09 2016-12-27 Iceutica Pty Ltd. Formulation of meloxicam
CA3026452C (en) 2015-06-04 2023-03-21 Crititech, Inc. Nozzle assembly and methods for use
WO2017176628A1 (en) 2016-04-04 2017-10-12 Crititech, Inc. Methods for solid tumor treatment
GB201609925D0 (en) * 2016-06-07 2016-07-20 Novabiotics Ltd Formulation
MX2020012139A (en) 2016-09-08 2021-01-29 Vyne Pharmaceuticals Inc Compositions and methods for treating rosacea and acne.
JP2020523285A (en) 2017-06-09 2020-08-06 クリチテック,インコーポレイテッド Treatment of epithelial cysts by intracystic injection of antitumor particles
KR102303762B1 (en) 2017-06-14 2021-09-23 크리티테크, 인크. Methods of treating lung disorders
CN111278436A (en) 2017-10-03 2020-06-12 克里蒂泰克公司 Local delivery of anti-tumor particles in combination with systemic delivery of immunotherapeutic agents for the treatment of cancer
US11534397B2 (en) * 2017-11-09 2022-12-27 The Board Of Regents Of The University Of Oklahoma Nanocrystal microparticles of poorly soluble drugs and methods of production and use thereof
WO2019126703A1 (en) 2017-12-22 2019-06-27 Ecolab Usa Inc. Antimicrobial compositions with enhanced efficacy
CN108651937B (en) * 2018-04-20 2021-12-24 大连工业大学 Method for producing soup-stock seasoning powder based on ultrasonic spray freeze drying
WO2019211492A1 (en) 2018-05-04 2019-11-07 Tollys Tlr3 ligands that activate both epithelial and myeloid cells
EP3827260A4 (en) * 2018-07-24 2022-05-04 Board of Regents, The University of Texas System Compositions of surface-modified therapeutically active particles by ultra-rapid freezing
AU2019315509A1 (en) * 2018-07-31 2021-02-18 Microbion Corporation Bismuth-thiol compositions and methods of use
WO2020028561A1 (en) 2018-07-31 2020-02-06 Microbion Corporation Bismuth-thiol compositions and methods for treating wounds
WO2022189861A1 (en) 2021-03-08 2022-09-15 Tollys Carbohydrate conjugates of tlr3 ligands and uses thereof
WO2023112966A1 (en) * 2021-12-14 2023-06-22 ダイキン工業株式会社 Spray device and air treatment device

Family Cites Families (115)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3269798A (en) * 1962-01-26 1966-08-30 Preston John Miles Stabilized phosphoric acid
NL289785A (en) * 1962-11-29
US3692532A (en) * 1970-10-27 1972-09-19 David R Shenkenberg Milk-fruit juice beverage and process for preparing same
SU628930A1 (en) 1974-11-26 1978-10-25 Московский научно-исследовательский институт туберкулеза Device for introducing medicinal powders
DE3013839A1 (en) 1979-04-13 1980-10-30 Freunt Ind Co Ltd METHOD FOR PRODUCING AN ACTIVATED PHARMACEUTICAL COMPOSITION
JPS5726615A (en) 1980-07-23 1982-02-12 Grelan Pharmaceut Co Ltd Improving method for absorbability of slightly soluble drug
US4389397A (en) * 1980-08-04 1983-06-21 Merck & Co., Inc. Solubilization of ivermectin in water
JPS5846019A (en) * 1981-09-14 1983-03-17 Kanebo Ltd Nifedipine preparation with prolonged action
JPS59101423A (en) * 1982-12-02 1984-06-12 Takada Seiyaku Kk Novel solid pharmaceutical preparation of nifedipine
DE3318649A1 (en) * 1983-05-21 1984-11-22 Bayer Ag, 5090 Leverkusen TWO-PHASE FORMULATION
US4917816A (en) * 1984-01-03 1990-04-17 Abco Industries, Inc. Stabilized peroxide compositions and process for producing same
DE3421468A1 (en) * 1984-06-08 1985-12-19 Dr. Rentschler Arzneimittel Gmbh & Co, 7958 Laupheim LIPID NANOPELLETS AS A CARRIER SYSTEM FOR MEDICINAL PRODUCTS FOR PERORAL USE
US4757059A (en) * 1984-08-14 1988-07-12 International Copper Research Association Method for treating convulsions and epilepsy with organic copper compounds
US4663364A (en) 1984-09-05 1987-05-05 Kao Corporation Biocidal fine powder, its manufacturing method and a suspension for agricultural use containing the above powder
FR2575678B1 (en) * 1985-01-04 1988-06-03 Saint Gobain Vitrage PNEUMATIC POWDER EJECTOR
US4727077A (en) 1985-02-20 1988-02-23 Ishihara Sangyo Kaisha Ltd. Benzoyl urea compounds, process for their production, and antitumorous compositions containing them
US4826821A (en) 1985-06-26 1989-05-02 The Regents Of The University Of California Lung surfactant compositions
IT1187751B (en) 1985-10-15 1987-12-23 Eurand Spa PROCEDURE FOR THE PREPARATION OF SOLID FORMULATIONS OF NIFEDIPINE WITH HIGH BIO AVAILABILITY AND WITH PROLONGED EFFECT AND FORMULATIONS SO OBTAINED
JPS62185013A (en) 1986-02-08 1987-08-13 Green Cross Corp:The Easily absorbable pharmaceutical composition
EP0262560A3 (en) 1986-09-29 1989-07-05 Ishihara Sangyo Kaisha, Ltd. Benzoyl urea compound
US4983605A (en) 1986-10-23 1991-01-08 Ishihara Sangyo Kaisha Ltd. Pharmaceutical composition
FR2608942B1 (en) * 1986-12-31 1991-01-11 Centre Nat Rech Scient PROCESS FOR THE PREPARATION OF COLLOIDAL DISPERSIBLE SYSTEMS OF A SUBSTANCE, IN THE FORM OF NANOCAPSULES
FR2608988B1 (en) 1986-12-31 1991-01-11 Centre Nat Rech Scient PROCESS FOR THE PREPARATION OF COLLOIDAL DISPERSIBLE SYSTEMS OF A SUBSTANCE, IN THE FORM OF NANOPARTICLES
IT1217890B (en) 1988-06-22 1990-03-30 Chiesi Farma Spa DOSED AEROSOL INHALATION DEVICE
JP2661709B2 (en) 1988-07-08 1997-10-08 ダウ・ケミカル日本株式会社 Production method of highly active pesticide wettable powder
FR2635459B1 (en) 1988-08-17 1992-06-19 Cird PROCESS FOR THE MANUFACTURE OF MICROSPHERES BY CROSSLINKING PROTEINS, MICROSPHERES THUS OBTAINED AND THEIR USES
DE68914929T2 (en) * 1988-09-29 1994-08-11 Shiseido Co Ltd Emulsified composition.
US5225183A (en) 1988-12-06 1993-07-06 Riker Laboratories, Inc. Medicinal aerosol formulations
JPH02196719A (en) 1989-01-24 1990-08-03 Green Cross Corp:The Powdery drug composition
JP2642486B2 (en) 1989-08-04 1997-08-20 田辺製薬株式会社 Ultrafine particle method for poorly soluble drugs
GB2237510B (en) 1989-11-04 1993-09-15 Danbiosyst Uk Small particle drug compositions for nasal administration
JPH05963A (en) 1990-04-13 1993-01-08 Toray Ind Inc Polypeptide composition
US5091188A (en) 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
US5091187A (en) 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
CA2053000C (en) 1990-10-15 1995-08-29 Michael J. Conder Biosynthetic production of 6(r)-[2-(8(s)-hydroxy-2(s), 6(r)-dimethyl-1,2,6,7,8,8a(r)-hexahydronaphthyl)-ethyl]-4 (r)-hydroxy-3,4,5,6-tetrahydro-2h-pyran-2-one triol acid by enzymatic hydrolysis of lovastatin acid using an enzyme derived from__lonostachys compactiuscula
GB9024365D0 (en) 1990-11-09 1991-01-02 Glaxo Group Ltd Medicaments
US5552160A (en) * 1991-01-25 1996-09-03 Nanosystems L.L.C. Surface modified NSAID nanoparticles
US5399363A (en) * 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
AU642066B2 (en) * 1991-01-25 1993-10-07 Nanosystems L.L.C. X-ray contrast compositions useful in medical imaging
US5145684A (en) * 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US5744123A (en) * 1991-12-12 1998-04-28 Glaxo Group Limited Aerosol formulations containing P134a and particulate medicaments
US5202110A (en) * 1992-01-22 1993-04-13 Virginia Commonwealth University Formulations for delivery of beclomethasone diproprionate by metered dose inhalers containing no chlorofluorocarbon propellants
JPH07112936A (en) 1992-04-27 1995-05-02 Philippe Perovich Preparation of component for medical treatment, especially component based on aspirin
US5300739A (en) 1992-05-26 1994-04-05 Otis Elevator Company Cyclically varying an elevator car's assigned group in a system where each group has a separate lobby corridor
US5719147A (en) * 1992-06-29 1998-02-17 Merck & Co., Inc. Morpholine and thiomorpholine tachykinin receptor antagonists
US5785049A (en) * 1994-09-21 1998-07-28 Inhale Therapeutic Systems Method and apparatus for dispersion of dry powder medicaments
FR2695563B1 (en) * 1992-09-11 1994-12-02 Pasteur Institut Microparticles carrying antigens and their use for the induction of humoral or cellular responses.
WO1994010982A1 (en) * 1992-11-17 1994-05-26 Yoshitomi Pharmaceutical Industries, Ltd. Sustained-release microsphere containing antipsychotic and process for producing the same
FR2698560B1 (en) * 1992-11-30 1995-02-03 Virbac Laboratoires Stabilized powdery active ingredients, compositions containing them, process for obtaining them and their applications.
US5298262A (en) * 1992-12-04 1994-03-29 Sterling Winthrop Inc. Use of ionic cloud point modifiers to prevent particle aggregation during sterilization
US5346702A (en) 1992-12-04 1994-09-13 Sterling Winthrop Inc. Use of non-ionic cloud point modifiers to minimize nanoparticle aggregation during sterilization
US5260478A (en) 1992-12-08 1993-11-09 Sterling Winthrop Inc. Iodinated aroyloxy carboxamides
US5429824A (en) 1992-12-15 1995-07-04 Eastman Kodak Company Use of tyloxapole as a nanoparticle stabilizer and dispersant
US5322679A (en) 1992-12-16 1994-06-21 Sterling Winthrop Inc. Iodinated aroyloxy esters
DE69433723T3 (en) 1993-02-22 2008-10-30 Abraxis Bioscience, Inc., Los Angeles PROCESS FOR IN VIVO ADMINISTRATION OF BIOLOGICAL SUBSTANCES AND COMPOSITIONS USED THEREFROM
US5916596A (en) * 1993-02-22 1999-06-29 Vivorx Pharmaceuticals, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
CA2091152C (en) 1993-03-05 2005-05-03 Kirsten Westesen Solid lipid particles, particles of bioactive agents and methods for the manfuacture and use thereof
US5264610A (en) 1993-03-29 1993-11-23 Sterling Winthrop Inc. Iodinated aromatic propanedioates
US5994341A (en) * 1993-07-19 1999-11-30 Angiogenesis Technologies, Inc. Anti-angiogenic Compositions and methods for the treatment of arthritis
DE4327063A1 (en) 1993-08-12 1995-02-16 Kirsten Dr Westesen Ubidecarenone particles with modified physicochemical properties
US6177104B1 (en) * 1994-01-27 2001-01-23 The Board Of Regents Of The University Of Oklahoma Particulate support matrix for making a rapidly dissolving dosage form
US5711934A (en) 1994-04-11 1998-01-27 Abbott Laboratories Process for the continuous milling of aerosol pharmaceutical formulations in aerosol propellants
US5521168A (en) * 1994-10-13 1996-05-28 Alcon Laboratories, Inc. Estrogen metabolites for lowering intraocular pressure
US5466440A (en) * 1994-12-30 1995-11-14 Eastman Kodak Company Formulations of oral gastrointestinal diagnostic X-ray contrast agents in combination with pharmaceutically acceptable clays
US5585108A (en) * 1994-12-30 1996-12-17 Nanosystems L.L.C. Formulations of oral gastrointestinal therapeutic agents in combination with pharmaceutically acceptable clays
JPH10511957A (en) 1995-01-05 1998-11-17 ザ ボード オブ リージェンツ オブ ザ ユニヴァーシティ オブ ミシガン Surface-modified nanoparticles and methods for their production and use
JPH08259460A (en) 1995-01-23 1996-10-08 Takeda Chem Ind Ltd Production of sustained release pharmaceutical preparation
US5503723A (en) * 1995-02-08 1996-04-02 Eastman Kodak Company Isolation of ultra small particles
US5518738A (en) 1995-02-09 1996-05-21 Nanosystem L.L.C. Nanoparticulate nsaid compositions
US5510118A (en) * 1995-02-14 1996-04-23 Nanosystems Llc Process for preparing therapeutic compositions containing nanoparticles
US5747001A (en) * 1995-02-24 1998-05-05 Nanosystems, L.L.C. Aerosols containing beclomethazone nanoparticle dispersions
EP0810853B1 (en) * 1995-02-24 2004-08-25 Elan Pharma International Limited Aerosols containing nanoparticle dispersions
EP0869772B1 (en) * 1995-12-27 2001-10-04 Janssen Pharmaceutica N.V. Bioadhesive solid dosage form
JPH09241178A (en) 1996-03-07 1997-09-16 Takeda Chem Ind Ltd Production of sustained release pharmaceutical preparation of adenocorticotropic hormone derivative
US5795909A (en) * 1996-05-22 1998-08-18 Neuromedica, Inc. DHA-pharmaceutical agent conjugates of taxanes
US5985309A (en) * 1996-05-24 1999-11-16 Massachusetts Institute Of Technology Preparation of particles for inhalation
DE59709127D1 (en) * 1996-07-08 2003-02-20 Ciba Sc Holding Ag Triazine derivatives as UV filters in sunscreens
JP2000516244A (en) * 1996-08-22 2000-12-05 リサーチ・トライアングル・ファーマシューティカルズ Composition containing fine particles of water-insoluble substance and method for producing the same
GB2316316A (en) 1996-08-23 1998-02-25 Int Centre Genetic Eng & Bio Sustained release of erythropoietin from microspheres
US5972389A (en) * 1996-09-19 1999-10-26 Depomed, Inc. Gastric-retentive, oral drug dosage forms for the controlled-release of sparingly soluble drugs and insoluble matter
IN186315B (en) * 1996-12-12 2001-08-04 Panacea Biotec Ltd
JPH10194996A (en) * 1996-12-25 1998-07-28 Janssen Pharmaceut Nv Acylated cyclodextrin-containing pharmaceutical composition
EP0971698A4 (en) 1996-12-31 2006-07-26 Nektar Therapeutics Aerosolized hydrophobic drug
US6458373B1 (en) * 1997-01-07 2002-10-01 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
FR2758461A1 (en) 1997-01-17 1998-07-24 Pharma Pass PHARMACEUTICAL COMPOSITION HAVING HIGH BIOAVAILABILITY AND PROCESS FOR PREPARING THE SAME
US6045829A (en) * 1997-02-13 2000-04-04 Elan Pharma International Limited Nanocrystalline formulations of human immunodeficiency virus (HIV) protease inhibitors using cellulosic surface stabilizers
WO1998035666A1 (en) 1997-02-13 1998-08-20 Nanosystems Llc Formulations of nanoparticle naproxen tablets
DE69818607T2 (en) * 1997-05-30 2004-07-29 Osmotica Corp. MULTILAYER OSMOSIS DEVICE
WO1999012571A1 (en) 1997-09-05 1999-03-18 Maruho Kabushikikaisha Nanocapsule preparations for treating intraarticular diseases
US20020002294A1 (en) * 1997-09-24 2002-01-03 D' Amato Robert J. Estrogenic compounds as antiangiogenic agents
GB9726543D0 (en) 1997-12-16 1998-02-11 Smithkline Beecham Plc Novel compositions
US6086376A (en) 1998-01-30 2000-07-11 Rtp Pharma Inc. Dry aerosol suspension of phospholipid-stabilized drug microparticles in a hydrofluoroalkane propellant
ZA991922B (en) 1998-03-11 1999-09-13 Smithkline Beecham Corp Novel compositions of eprosartan.
US6177103B1 (en) * 1998-06-19 2001-01-23 Rtp Pharma, Inc. Processes to generate submicron particles of water-insoluble compounds
US6241969B1 (en) * 1998-06-26 2001-06-05 Elan Corporation Plc Aqueous compositions containing corticosteroids for nasal and pulmonary delivery
US8293277B2 (en) * 1998-10-01 2012-10-23 Alkermes Pharma Ireland Limited Controlled-release nanoparticulate compositions
EP1117384A1 (en) 1998-10-01 2001-07-25 Elan Pharma International Limited Controlled release nanoparticulate compositions
US6375986B1 (en) 2000-09-21 2002-04-23 Elan Pharma International Ltd. Solid dose nanoparticulate compositions comprising a synergistic combination of a polymeric surface stabilizer and dioctyl sodium sulfosuccinate
US7521068B2 (en) * 1998-11-12 2009-04-21 Elan Pharma International Ltd. Dry powder aerosols of nanoparticulate drugs
ES2367352T3 (en) 1999-04-28 2011-11-02 Genetics Institute, Llc TREATMENT OF MEASURING FIBROSIS THE ANTAGONISM OF IL-3 AND THE IL-3 RECEPTOR CHAINS.
US6395300B1 (en) * 1999-05-27 2002-05-28 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
GB9913536D0 (en) * 1999-06-10 1999-08-11 Sterix Ltd Use
US6368620B2 (en) * 1999-06-11 2002-04-09 Abbott Laboratories Formulations comprising lipid-regulating agents
US6656504B1 (en) 1999-09-09 2003-12-02 Elan Pharma International Ltd. Nanoparticulate compositions comprising amorphous cyclosporine and methods of making and using such compositions
US6242003B1 (en) 2000-04-13 2001-06-05 Novartis Ag Organic compounds
US6316029B1 (en) * 2000-05-18 2001-11-13 Flak Pharma International, Ltd. Rapidly disintegrating solid oral dosage form
AR035642A1 (en) 2000-05-26 2004-06-23 Pharmacia Corp USE OF A CELECOXIB COMPOSITION FOR QUICK PAIN RELIEF
JP4295420B2 (en) 2000-06-01 2009-07-15 株式会社リコー Blank page printing method for printer control device
EP1292709B1 (en) 2000-06-02 2012-01-18 Eidgenössische Technische Hochschule Zürich Conjugate addition reactions for the controlled delivery of pharmaceutically active compounds
ES2284646T3 (en) 2001-02-22 2007-11-16 Jagotec Ag STATIN-FIBRATE COMBINATIONS WITH SECONDARY EFFECTS IN REDUCED FAST-FASTING.
US7112340B2 (en) * 2001-10-19 2006-09-26 Baxter International Inc. Compositions of and method for preparing stable particles in a frozen aqueous matrix
CA2479665C (en) * 2002-03-20 2011-08-30 Elan Pharma International Ltd. Nanoparticulate compositions of angiogenesis inhibitors
EP1800666A1 (en) 2002-03-20 2007-06-27 Elan Pharma International Limited Nanoparticulate compositions of angiogenesis inhibitors
ATE419835T1 (en) 2002-05-06 2009-01-15 Elan Pharma Int Ltd NYSTATIN NANOPARTICLE COMPOSITIONS
US20040033202A1 (en) 2002-06-10 2004-02-19 Elan Pharma International, Ltd. Nanoparticulate sterol formulations and novel sterol combinations

Also Published As

Publication number Publication date
JP2002529396A (en) 2002-09-10
US8124057B2 (en) 2012-02-28
EP1128814A1 (en) 2001-09-05
US20090181100A1 (en) 2009-07-16
ES2359298T3 (en) 2011-05-20
DE69943112D1 (en) 2011-02-17
WO2000027363A9 (en) 2000-11-30
WO2000027363B1 (en) 2000-07-13
EP2263652B1 (en) 2012-05-09
EP1128814B1 (en) 2011-01-05
CA2350074A1 (en) 2000-05-18
DK2263652T3 (en) 2012-08-20
ATE493971T1 (en) 2011-01-15
ES2387749T3 (en) 2012-10-01
US20120213829A1 (en) 2012-08-23
US7521068B2 (en) 2009-04-21
ATE556702T1 (en) 2012-05-15
AU1346900A (en) 2000-05-29
DK1128814T3 (en) 2011-03-07
WO2000027363A1 (en) 2000-05-18
EP2263652A1 (en) 2010-12-22
US6811767B1 (en) 2004-11-02
JP4758548B2 (en) 2011-08-31
CY1111344T1 (en) 2015-08-05
CY1112978T1 (en) 2016-04-13
US20020102294A1 (en) 2002-08-01
PT2263652E (en) 2012-08-09

Similar Documents

Publication Publication Date Title
CA2350074C (en) Aerosols comprising nanoparticle drugs
CA2213638C (en) Aerosols containing nanoparticle dispersions
EP1354583B1 (en) Aerosols containing beclomethasone nanoparticle dispersions
EP1196146B1 (en) Powder particles with smooth surface for use in inhalation therapy
KR100277622B1 (en) Inhalation ultrafine powder and its manufacturing method
EP1019022B2 (en) Method for forming a powder comprising perforated microparticles
WO1992000107A1 (en) Aerosol drug formulations
EP2010153A2 (en) Drug microparticles
MX2007011502A (en) Nanoparticulate leukotriene receptor antagonist/corticosteroid formulations.
EP1976534A1 (en) Sterilized nanoparticulate glucocorticosteroid formulations
JP2000507862A (en) Method and apparatus for inhaling granular drugs
JP4266399B2 (en) Pharmaceutical composition for inhalation in powder form
Klyashchitsky et al. Nebulizer-compatible liquid formulations for aerosol pulmonary delivery of hydrophobic drugs: glucocorticoids and cyclosporine

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20151112