Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTIVIRAL COMPOSITIONS, METHODS OF MAKING AND USING SUCH COMPOSITIONS, AND SYSTEMS FOR PULMONARY DELIVERY OF SUCH COMPOSITIONS
Document Type and Number:
WIPO Patent Application WO/2009/143011
Kind Code:
A1
Abstract:
A pharmaceutical composition comprises particles comprising an antiviral active, wherein the particles have a mass median aerodynamic diameter from about 1 µm to about 7 µm and a bulk density less than about 1.0 g/cm3. A pharmaceutical composition comprises a powder including an effective amount of antiviral and pharmaceutically acceptable excipient, wherein the powder comprises particles comprising having a mass median aerodynamic diameter from about 1 µm to about 7 µm, and a bulk density less than about 1.0 g/cm3. Also provided are pharmaceutical compositions comprising combinations of two or more antiviral actives. Also provided are unit dosage forms, methods of making and using such compositions, and methods and systems for pulmonary delivery of such compositions

Inventors:
KUO MEI-CHANG (US)
LALONDE GUY (US)
SAHNER DAVID (US)
BUECHE BLAINE (US)
Application Number:
PCT/US2009/044122
Publication Date:
November 26, 2009
Filing Date:
May 15, 2009
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NOVARTIS AG (CH)
KUO MEI-CHANG (US)
LALONDE GUY (US)
SAHNER DAVID (US)
BUECHE BLAINE (US)
International Classes:
A61K9/72; A61K31/196; A61K31/575
Domestic Patent References:
WO2000027359A12000-05-18
WO2007070875A12007-06-21
Foreign References:
US6518239B12003-02-11
US20050147567A12005-07-07
EP0267050A21988-05-11
US20070178166A12007-08-02
Attorney, Agent or Firm:
MANZARI, Oona A. (Corporate Intellectual PropertyOne Health Plaza Building 10, East Hanover New Jersey, US)
Download PDF:
Claims:

WHAT IS CLAIMED IS:

1. A pharmaceutical composition comprising particles comprising an effective amount of an antiviral and a pharmaceutically acceptable excipient, wherein the particles have a mass median aerodynamic diameter (MMAD) from about 1 μm to about 7 μm, and a bulk density of less than about 1.0 g/cm 3 .

2. The pharmaceutical composition of claim 1 wherein the antiviral is selected from the group consisting of a neuraminidase inhibitor, a hemagglutinin inhibitor, an M2 proton channel blocker, a nucleoside analog and any combination thereof.

3. The pharmaceutical composition of claim 2, wherein the antiviral is selected from the group consisting of a neuraminidase inhibitor, an M2 proton channel blocker, and a combination thereof.

4. The pharmaceutical composition of claim 2 or 3, wherein the neuraminidase inhibitor is selected from the group consisting of zanamivir, oseltamivir and peramivir.

5. The pharmaceutical composition of any of the preceding claims, wherein the M2 proton channel blocker is is selected from the group consisting of rimantadine and amantadine.

6. The pharmaceutical composition of any of the preceding claims, wherein the pharmaceutically acceptable excipient comprises at least one member selected from carbohydrate, lipid, amino acid, buffer, and salt.

7. The pharmaceutical composition of claim 5, wherein the pharmaceutically acceptable excipient comprises a phospholipid and optionally a metal ion.

8. The pharmaceutical composition of claim 5, wherein the pharmaceutically acceptable excipient comprises a di- or tri-peptide having at least two leucines.

9. The pharmaceutical composition of claim 8, wherein the pharmaceutically acceptable excipient comprises a dileucine, a trileucine or a combination thereof.

10. The pharmaceutical composition of any of the preceding claims, wherein the particles have a mass median aerodynamic diameter (MMAD) from about 1 μm to about 5 μm.

11. The pharmaceutical composition of any of the preceding claims, wherein the particles are spray dried particles which are either hollow, porous or both.

12. The pharmaceutical composition of any of the preceding claims, wherein the pharmaceutical composition is in a form selected from an inhalable powder, a dry powder composition, a propellant-containing aerosol and a propellant- free inhalable suspension.

13. The pharmaceutical composition of claim 12, wherein the pharmaceutical composition is in a form of dry powder composition.

14. The pharmaceutical composition of any of the preceding claims, wherein the pharmaceutical composition is suitable for inhalation by nasal or pulmonary route.

15. The pharmaceutical composition of any of the preceding claims, wherein the particles have a particle size distribution of at least 50% of the particles having an aerodynamic diameter of less than about 3 microns.

16. The pharmaceutical composition of any of the preceding claims, wherein the composition provides an emitted dose of the antiviral of at least about 50%.

17. A pharmaceutical composition according to claim 1 in the form of an inhalable powder comprising particles comprising an antiviral selected from zamanivir, rimandadine and a combination thereof, and an excipient selected from a phospholipid or a di- or tri- peptide comprising at least two leucines, wherein the particles have a particle size distribution of at least 50% of the particles having an aerodynamic diameter of less than about 3 microns, and wherein the composition provides an emitted dose of active of at

least about 50%.

18. A unit dosage form comprising a container containing a pharmaceutical composition according to any of the claims 1 to 17.

19. The unit dosage form of claim 18, wherein the container comprises a capsule.

20. A delivery system, comprising an inhaler and a pharmaceutical composition according to any of the claims 1 to 17.

21. The delivery system of claim 20, wherein the inhaler comprises a dry powder inhaler.

22. The delivery system of claim 20, wherein the inhaler comprises a nebulizer, and wherein the particles are suspended in a liquid.

23. A method of treating a viral infection, comprising administering an effective amount of a pharmaceutical composition according to any of the claims 1 to 17.

24. The method of claim 23, wherein the administration comprises pulmonary delivery by inhalation or nasal administration.

25. A method of making particles, comprising: suspending an antiviral and a pharmaceutically acceptable excipient in a liquid to form a feedstock; and removing the liquid from the feedstock to produce particles, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm, and a bulk density of less than about 1.0 g/cm 3 .

26. The method of claim 25, wherein the liquid removal comprises spray-drying.

27. The method of claim 26, wherein the feedstock further comprises a blowing agent.

28. The method of any of the claims 25 to 27, wherein the pharmaceutically acceptable excipient comprises a phospholipid.

29. The method of any of the claims 25 to 27, wherein the pharmaceutically acceptable excipient comprises a phospholipid and metal ion.

30. The method of any of the claims 25 to 27, wherein the pharmaceutically acceptable excipient comprises a di- or tri-peptide having at least two leucines.

31. The method of any of the claims 25 to 27, wherein the pharmaceutically acceptable excipient comprises a dileucine, a trileucine or a combination thereof.

32. The method of any of the claims 25 to 31 , wherein the spray-dried particles are either hollow, porous or both.

Description:

Antiviral Compositions, Methods of Making and Using Such Compositions, and Systems for Pulmonary Delivery of Such Compositions

BACKGROUND

[0001] The present invention relates to pharmaceutical compositions comprising an antiviral active, powder compositions comprising antiviral actives, and compositions comprising combinations of two or more antiviral actives. One or more embodiments of the present invention include methods of making and using such compositions, and methods and systems for pulmonary delivery of such compositions.

[0002] This invention relates to methods and compositions for treating viral infections, and has particular reference to the treatment of influenza.

[0003] Influenza, more commonly known as the flu, is an acute, viral infection that attacks mainly the upper respiratory tract-the nose, throat and bronchi and rarely also the lungs. Although the flu is considered to be an infection of the respiratory tract, individuals suffering from the flu usually become acutely ill with high fever, chills, headache, weakness, loss of appetite and aching joints. The typical length of time from when a person is exposed to influenza virus to when symptoms first occur ranges between one and five days, with an average of two days. Adults can be infectious (i.e., shedding virus) starting the day before the onset of symptoms begin until approximately 5 days after the onset of illness. Children can be infectious for longer periods of time. Systemic symptoms include abrupt onset of fever (e.g. usually 100-103 0 F in an adult and possibly higher in children), chills, headaches, myalgia and malaise.

[0004] Most people recover within one to two weeks without requiring any medical treatment. In the very young, the elderly and people suffering from medical conditions such as lung diseases, diabetes, cancer, kidney or heart problems, influenza poses a serious risk. In these people, the infection may lead to severe complications of underlying diseases, pneumonia and death.

[0005] Also, influenza infections are known to increase the susceptibility of an infected to particular bacterial infections caused by species of bacterial pathogens such as, pneumococcus, staphylococcus, mycoplasma, non-group H. influenza, and Moraxella catarrhalis. Secondary bacterial infections, such as, but not limited to, infections of the lower respiratory tract (e.g., pneumonia), middle ear infections (e.g., otitis media) and bacterial sinusitis are common complications of an infection with viral influenza.

[0006] Given that the flu and its associated complications (e.g. bacterial infections, viral pneumonia, and cardiac and other organ system abnormalities) represent the sixth leading cause of death in the world and the leading infectious cause of death, there is a clear need for improved therapeutics and methods for the treatment of viral diseases and disorders, such as the flu and its related conditions.

[0007] Aerosolized medicaments are used to treat patients suffering from a variety of ailments. Medicaments can be delivered directly to the lungs by having the patient inhale the aerosol through a tube and/or mouthpiece coupled to an aerosol generator. By inhaling the aerosolized medicament, the patient can quickly, safely and efficiently receive a dose of medicament.

[0008] Aerosolized medicaments can be administered directly to the lungs to treat diseases and/or conditions of the lung, and to treat diseases or conditions having a systemic effect or component thereof. Many medicaments cannot be administered orally, due to their sensitivity to metabolism and/or degradation and resulting inactivation in the gastrointestinal tract, thus pulmonary delivery avoids the need for intramuscular, subcutaneous or transdermal delivery and associated needles. Additionally or alternatively, it may be safer and/or more efficacious to deliver the medicament directly to the lungs and/or pulmonary system instead of other administration routes.

[0009] Pulmonary delivery by aerosol inhalation has received much attention as an attractive alternative to intravenous, intramuscular, and subcutaneous injection, since this approach eliminates the necessity for injection syringes and needles. Pulmonary delivery also limits irritation to the skin and body mucosa which are common side effects of transdermal^, iontophoretically, and intranasally delivered drugs, eliminates the need for nasal and skin penetration enhancers (typical components of intranasal and transdermal systems that often cause skin irritation/dermatitis), is economically attractive, is amenable to patient self-administration, and is often preferred by patients over other alternative modes of administration.

[0010] Pulmonary delivery may comprise aerosolized liquids, dispersions, or powder forms. The compositions may be delivered via liquid nebulizers, metered dose (pressurized) inhalers or dry-powder inhalers.

[0011] Dry powder inhalers are known in the art as disclosed, for example, in U.S.

Pat. Nos. 5,458,135; 5,740,794; 5,775,320; 5,785,049; 6,089,228; 6,257,233 and in US Patent Application Publications 2005-0016533; 2003-0150454; and 2003-0094173 all of which are hereby incorporated in their entirety by reference. In addition, U.S. Pat. No.

5,875,776 discloses a dry powder inhaler and discloses antibiotics as suitable for administration by the devices disclosed therein.

[0012] Existing antiviral therapies suffer from several deficiencies. Commercially- available products are limited to a single type or class of antiviral. Dosing of existing products is often problematic, in view of toxicity and/or side effects, resulting in generally low concentrations in the lung. Additionally, oral or peritoneal dosing tends to promote resistance.

[0013] In view of the known antiviral actives, compositions, and systems for administering antivirals, there remains a need for better efficacy, greater safety, fewer side or adverse effects, higher efficiency and more convenient delivery systems.

[0014] One or more embodiments of the present invention satisfy one or more of these needs.

SUMMARY OF THE INVENTION

[0015] The present invention relates to antiviral pharmaceutical compositions, methods of making and using such compositions, and systems for pulmonary delivery of such compositions.

[0016] One or more embodiments of the present invention relate to pharmaceutical compositions comprising particles comprising antiviral actives (i.e. drugs, pharmaceuticals, compounds, chemicals, metals, biologies and combinations having antiviral activity, individually and collectively referred to as antivirals.)

[0017] Other features and advantages of embodiments of the present invention will be set forth in the description of invention that follows, and in part will be apparent from the description or may be learned by practice of the invention. Embodiments of the invention will be realized and attained by the compositions and methods particularly pointed out in the written description and claims hereof.

[0018] In one aspect, the present invention relates to pharmaceutical compositions comprising particles comprising an effective amount of an antiviral and a pharmaceutically acceptable excipient, wherein the particles have a mass median aerodynamic diameter (MMAD) from about 1 μm to about 7 μm, and a bulk density of less than about 1.0 g/cm 3 .

[0019] In one aspect, the present invention is directed to a pharmaceutical composition for pulmonary delivery comprising particles comprising at least one antiviral

and a pharmaceutically acceptable excipient, wherein pulmonary distribution (i.e. to and throughout bronchi, bronchioles and alveoli) is very good.

[0020] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles comprising one or more antivirals selected from the group consisting of a neuraminidase inhibitor, a hemagglutinin inhibitor, an M2 proton channel blocker, a nucleoside analog, peptide analogs, protease inhibitors, SiRNAs, antibodies, antibody fragments, antibody constructs and glycodendritic structures or polymers and any combination thereof.

[0021] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles comprising one or more antivirals selected from the group consisting of a neuraminidase inhibitor, a hemagglutinin inhibitor, an M2 proton channel blocker, a nucleoside analog and any combination thereof.

[0022] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles comprising two or more antivirals selected from the group consisting of a neuraminidase inhibitor, a hemagglutinin inhibitor and a M2 proton channel blocker.

[0023] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles comprising an antiviral which is selected from the group consisting of a neuraminidase inhibitor, a hemagglutinin inhibitor, a M2 proton channel blocker, a nucleoside analog and combinations thereof, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm, and a bulk density of less than about 1.0 g/cm 3 .

[0024] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles comprising an antiviral which is selected from the group consisting of a neuraminidase inhibitor, a hemagglutinin inhibitor, a M2 proton channel blocker, a nucleoside analog and combinations thereof, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 5 μm and a bulk density of less than about 1.0 g/cm 3 .

[0025] In one aspect, the present invention is directed to one of the aforementioned pharmaceutical composition comprising particles comprising about 10-99 wt% of antiviral which is selected from a neuraminidase inhibitor, a hemagglutinin inhibitor, a M2 proton channel blocker, a nucleoside analog and combinations thereof.

[0026] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles comprising a neuraminidase inhibitor and a M2 proton

channel blocker, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 .

[0027] In one aspect, the present invention is directed to a pharmaceutical composition comprising porous and/or holiow particles comprising an antiviral.

[0028] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles as described above, wherein the composition is in the form of a powder.

[0029] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles comprising at least one antiviral selected from the group consisting of a neuraminidase inhibitor, a hemagglutinin inhibitor, a M2 proton channel blocker and a nucleoside analog, and at least one excipient, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 , a particle size distribution of at least 50% of the particles having an aerodynamic diameter of less than about 3 microns. In a further aspect, the the composition provides an emitted dose of active of at least about 50% of the antiviral.

[0030] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles comprising zamanivir and an excipient selected from a phospholipid or a di- or tri-peptide comprising at least two leucines, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 , a particle size distribution of at least 50% having an aerodynamic diameter less than about 3 microns, and wherein the composition further provides an emitted dose of active of at least about 50%.

[0031] In one aspect, the present invention is directed to a pharmaceutical composition comprising particles comprising rimantadine and a pharmaceutically acceptable excipient selected from a phospholipid or a di- or tri-peptide comprising at least two leucines, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 , a particle size distribution of at least 50% having an aerodynamic diameter less than about 3 microns, and wherein the composition further provides an emitted dose of active of at least about 50%.

[0032] In still another aspect, the present invention is directed to a unit dosage form, comprising a container containing a pharmaceutical composition comprising particles comprising an effective amount of antiviral and a pharmaceutically acceptable excipient,

wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 .

[0033] In still another aspect, the present invention is directed to a unit dosage form, comprising a container containing a pharmaceutical composition comprising particles comprising an effective amount of antiviral and a di or tri- peptide containing at least two leucines, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 .

[0034] In still another aspect, the present invention is directed to a unit dosage form, comprising a container containing a pharmaceutical composition comprising particles comprising an effective amount of antiviral and a phospholipid, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 .

[0035] In a further aspect, the present invention is directed to a delivery system, comprising an inhaler and a pharmaceutical composition comprising particles comprising antiviral and a pharmaceutically acceptable excipient as set out above.

[0036] In yet another aspect, the present invention is directed to a method of making particles, comprising suspending an antiviral in a liquid to form a feedstock and removing the liquid therefrom to produce particles, wherein the particles comprise an antiviral and have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 .

[0037] In yet another aspect, the present invention is directed to a method of making particles, comprising suspending an antiviral in a liquid to form a feedstock and spray drying the feedstock to produce spray-dried particles, wherein the particles comprise an antiviral and have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 .

[0038] In still another aspect, the present invention is directed to a method of treating a patient with a condition associated with a viral infection comprising administering an effective amount of a pharmaceutical composition comprising antiviral by inhalation to a patient, wherein the composition comprises particles comprising an antiviral a pharmaceutically acceptable excipient, such particles having a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 .

[0039] In another aspect, the present invention is directed to a method of treating viral infections by pulmonary administration of a pharmaceutical composition as set out

above comprising an antiviral combination, wherein an effective dose of the antiviral combination is at least about ten times lower than an effective dose of the same antiviral delivered orally.

[0040] In another aspect, the present invention is directed to a kit comprising a pharmaceutical composition comprising particles comprising an effective amount of antiviral and a pharmaceutically acceptable excipient, wherein the particles have a mass median aerodynamic diameter from about 1 μm to about 7 μm and a bulk density of less than about 1.0 g/cm 3 , and a delivery device for the composition.

DRAWINGS

[0041] Embodiments of the present invention are further described in the description of invention that follows, in reference to the noted plurality of non-limiting drawings, wherein:

[0042] Figs 1A-1E show a passive inhaler device.

[0043] Figs 2A-2D are photomicrographs showing particles made with varying amounts of antiviral and excipient, in accordance with one or more embodiments of the present invention.

[0044] Fig 3 is a graph showing particle size and particle size distribution for particles made in accordance with one or more embodiments of the present invention.

[0045] Fig 4 is a bar graph showing emitted dose showing particles made with varying amounts of antiviral and excipient, in accordance with one or more embodiments of the present invention

[0046] Figs 5A-5B are graphs showing emptying profiles of amorphous drug particles made with varying amounts of antiviral and excipient, in accordance with one or more embodiments of the present invention.

[0047] Fig 6 is a bar graph of drug delivered (as lung dose) for particles made with varying amounts of antiviral and excipient, in accordance with one or more embodiments of the present invention

DESCRIPTION

[0048] It is to be understood that unless otherwise indicated the present invention is not limited to specific formulation components, drug delivery systems, manufacturing techniques, administration steps, or the like, as such may vary. Unless otherwise stated, a reference to a compound or component includes the compound or component by itself, as well as the compound in combination with other compounds or components, such as mixtures of compounds.

[0049] Before further discussion, a definition of the following terms will aid in the understanding of embodiments of the present invention.

[0050] As used herein, the singular forms "a," "an," and "the" include the plural reference unless the context clearly dictates otherwise. Thus, for example, reference to "a phospholipid" includes a single phospholipid as well as two or more phospholipids in combination or admixture unless the context clearly dictates otherwise.

[0051] When referring to an active agent, the term encompasses not only the specified molecular entity, but also its pharmaceutically acceptable, pharmacologically active analogs, including, but not limited to, salts, esters, amides, hydrazides, N-alkyl derivatives, N-acyl derivatives, prodrugs, conjugates, active metabolites, and other such derivatives, analogs, and related compounds. Therefore, as used herein, the term "antiviral" refers to antivirals per se or derivatives, analogs, or related compounds noted above, as long as such antivirals derivatives, analogs, or related compounds exhibit antiviral activity.

[0052] As used herein, the terms "treating" and "treatment" refer to reduction in severity and/or frequency of symptoms, elimination of symptoms and/or underlying cause, reduction in likelihood of the occurrence of symptoms and/or underlying cause, and improvement or remediation of damage. Thus, "treating" a patient with an active agent as provided herein includes prevention of a particular condition, disease or disorder in a susceptible individual as well as treatment of a clinically symptomatic individual. [0053] As used herein, "effective amount" refers to an amount covering both therapeutically effective amounts and prophylactically effective amounts.

[0054] As used herein, "therapeutically effective amount" refers to an amount that is effective to achieve the desired therapeutic result. A therapeutically effective amount of a given active agent will typically vary with respect to factors such as the type and severity of the disorder or disease being treated and the age, gender, and weight of the patient.

[0055] As used herein, "prophylactically effective amount" refers to an amount that is effective to achieve the desired prophylactic result. Because a prophylactic dose is

administered in patients prior to onset of disease, the prophylactically effective amount typically is tess than the therapeutically effective amount.

[0056] As used herein, the term "respiratory infections" includes, but is not limited to upper respiratory tract infections such as sinusitis, pharyngitis, and influenza, and lower respiratory tract infections such as tuberculosis, bronchiectasis (both the cystic fibrosis and non-cystic fibrosis indications), bronchitis (both acute bronchitis and acute exacerbation of chronic bronchitis), and pneumonia (including various types of complications that arise from viral and bacterial infections including hospital-acquired and community-acquired infections).

[0057] As used herein, "mass median diameter" or "MMD" refers to the median diameter of a plurality of particles, typically in a polydisperse particle population, i.e., consisting of a range of particle sizes. MMD values as reported herein are determined by laser diffraction (Sympatec Helos, Clausthal-Zellerfeld, Germany), unless the context indicates otherwise. Typically, powder samples are added directly to the feeder funnel of the Sympatec RODOS dry powder dispersion unit. This can be achieved manually or by agitating mechanically from the end of a VIBRI vibratory feeder element. Samples are dispersed to primary particles via application of pressurized air (2 to 3 bar), with vacuum depression (suction) maximized for a given dispersion pressure. Dispersed particles are probed with a 632.8 nm laser beam that intersects the dispersed particles' trajectory at right angles. Laser light scattered from the ensemble of particles is imaged onto a concentric array of photomultiplier detector elements using a reverse-Fourier lens assembly. Scattered light is acquired in time-slices of 5 ms. Particle size distributions are back-calculated from the scattered light spatial/intensity distribution using a proprietary algorithm.

[0058] As used herein, "geometric diameter" refers to the diameter of a single particle, as determined by microscopy, unless the context indicates otherwise.

[0059] As used herein, "mass median aerodynamic diameter" or "MMAD" refers to the median aerodynamic size of a plurality of particles or particles, typically in a polydisperse population. The "aerodynamic diameter" is the diameter of a unit density sphere having the same settling velocity, generally in air, as a powder and is therefore a useful way to characterize an aerosolized powder or other dispersed particle or particle formulation in terms of its settling behavior. The aerodynamic diameter encompasses particle or particle shape, density, and physical size of the particle or particle. As used herein, MMAD refers to the median of the aerodynamic particle or particle size distribution

of an aerosolized powder determined by cascade impaction, unless the context indicates otherwise.

[0060] As used herein, the term "emitted dose" or "ED" refers to an indication of the delivery of dry powder from an inhaler device after an actuation or dispersion event from a powder unit or reservoir. ED is defined as the ratio of the dose delivered by an inhaler device to the nominal dose (i.e., the mass of powder per unit dose placed into a suitable inhaler device prior to firing). The ED is an experimentally determined amount, and may be determined using an in vitro device set up which mimics patient dosing. To determine an ED value, as used herein, a nominal dose of dry powder (as defined herein) is placed into a suitable inhaler device, for example, a Turbospin® DPI device (PH&T, Italy), described in U.S. Patent Nos. 4,069,819 and 4,995,385, which are incorporated herein by reference in their entireties. The inhaler device is actuated, dispersing the powder. The resulting aerosol cloud is then drawn from the device by vacuum (30 L/min) for 2.5 seconds after actuation, where it is captured on a tared glass fiber filter (Gelman, 47 mm diameter) attached to the device mouthpiece. The amount of powder that reaches the filter constitutes the delivered dose. For example, for a capsule containing 5 mg of dry powder that is placed into an inhalation device, if dispersion of the powder results in the recovery of 4 mg of powder on a tared filter as described above, then the ED for the dry powder composition is 80% [= 4 mg (delivered dose)/5 mg (nominal dose)].

[0061] As used herein, "passive dry powder inhaler" refers to an inhalation device that relies upon a patient's inspiratory effort to disperse and aerosolize a pharmaceutical composition contained within the device in a reservoir or in a unit dose form and does not include inhaler devices which comprise a means for providing energy, such as pressurized gas and vibrating or rotating elements, to disperse and aerosolize the drug composition.

[0062] As used herein, "active dry powder inhaler" refers to an inhalation device that does not rely solely on a patient's inspiratory effort to disperse and aerosolize a pharmaceutical composition contained within the device in a reservoir or in a unit dose form and does include inhaler devices that comprise a means for providing energy to disperse and aerosolize the drug composition, such as pressurized gas and vibrating or rotating elements.

[0063] Compositions including antivirals may include various forms and amounts of antivirals. For example, the antiviral may be present in an amount from, in weight percentage (wt%) at least about 0.01 , or 0.5 or 1 or 2 or 5 or 10 or 20 or 30 or 40 or 50 or

60 or 70 or 80 or 90 or 95 or 98 or 99 wt%, or in a range of any combination of the stated amounts.

[0064] The pharmaceutical composition according to one or more embodiments of the invention may comprise one or more antiviral and, optionally, one or more other active ingredients and/or pharmaceutically acceptable excipients. For example, the pharmaceutical composition may comprise neat particles of antiviral, may comprise neat particles of antiviral together with other particles, and/or may comprise particles comprising antiviral and one or more active ingredients and/or one or more pharmaceutically acceptable excipients. The particles may be as a dry powder, or may be suspended or dispersed in a liquid.

[0065] The pharmaceutical composition according to one or more embodiments of the invention may comprise one or more antiviral and, optionally, one or more other active ingredients and/or pharmaceutically acceptable excipients.

[0066] Thus, the pharmaceutical composition according to one or more embodiments of the invention may, if desired, contain a combination of antiviral and one or more other active ingredients. Examples of other active agents include, but are not limited to, agents that may be delivered to or through the lungs or nasal passages. For example, the other active agent(s) may be long-acting agents and/or active agents that are active against pulmonary and/or nasal infections such as antifungals and/or antibiotics.

[0067] In one or more embodiments, the present invention comprises a particulate formulation comprising at least one antiviral. In one or more embodiments, the present invention comprises a formulation comprising at least two antivirals. In one or more embodiments, the present invention comprises a formulation comprising at least two antivirals wherein the antivirals are of different classes. In one or more embodiments, the present invention comprises a formulation comprising at least three antivirals wherein the antivirals are of different classes.

[0068] Antivirals may be classified by a variety of schemes. One such scheme is based upon the target inhibited within the viral life cycle stage. In this classification scheme, antivirals may be conveniently divided into three classes, by approximate functional mode

Before cell entry

[0069] A very early stage of viral infection is viral entry, when the virus attaches to and enters the host cell. A number of entry-inhibitingor entry-blocking drugs exist or are

being developed. Two entry-blockers, amantadine and rimantadine, have been introduced to combat influenza. Entry-inhibiting drugs to combat hepatitis B and C virusus are under development.

[0070] At his initial phase, the virus must first bind to a specific receptor molecule on the surface of the host cell. Viruses that have a lipid envelope must also fuse their envelope with the target cell, or with a vesicle that transports them into the cell, before they can uncoat.

[0071] This stage of viral replication can be inhibited by using agents which mimic the virus-associated protein (VAP) and bind to the cellular receptors. This may include VAP anti-idiotypic antibodies, anti-receptor antibodies, and natural ligands of the receptor and anti-receptor antibodies. Agents which mimic the receptor and bind to the VAP also exist, such as anti-VAP antibodies, receptor anti-idiotypic antibodies, extraneous receptor and synthetic receptor mimics.

[0072] One entry-blocker is pleconaril, which works against rhinoviruses, by blocking a pocket on the surface of the virus that controls the uncoating process.

[0073] Hemagglutinin (HA) is an antigenic glycoprotein found on the surface of the influenza viruses (as well as many other bacteria and viruses). It is responsible for binding the virus to the cell that is being infected. HA binds to the monosaccharide sialic acid which is present on the surface of its target cells. This causes the viral particles to stick to the cell's surface. The cell membrane then engulfs the virus and the portion of the membrane that encloses it pinches off to form a new membrane-bound compartment within the cell (an endosome), which contains the engulfed virus. The cell then attempts to begin digesting the contents of the endosome by acidifying its interior and transforming it into a lysosome. However, as soon as the pH within the endosome drops to about 6.0, the original folded structure of the HA molecule becomes unstable, causing it to partially unfold, and releasing a very hydrophobic portion of its peptide chain that was previously hidden within the protein. This so-called "fusion peptide" inserts itself into the endosomal membrane and locks on. Then, when the rest of the HA molecule refolds into a new structure (which is more stable at the lower pH), it retracts the fusion peptide segment and pulls the endosomal membrane right up next to the virus particle's own membrane, causing the two to fuse together. Once this has happened, the contents of the virus, including its RNA genome, is free to pour out into the cell's cytoplasm.

During viral synthesis

[0074] A second approach is to target the processes that synthesize virus components after a virus invades a cell. One way of doing this is to develop nucleotide or nucleoside analogues that look like the building blocks of RNA or DNA, but deactivate the enzymes that synthesize the RNA or DNA once the analogue is incorporated.

[0075] Examples include acyclovir and zidovudine, which are effective against herpes virus infections.

[0076] Also in this class is lamivudine, approved to treat hepatitis B, which uses reverse transcriptase as part of its replication process. Inhibitors have been developed that do not look like nucleosides, but can still block reverse transcriptase.

[0077] Once a virus genome becomes operational in a host cell, it then generates messenger RNA (mRNA) molecules that direct the synthesis of viral proteins. Production of mRNA is initiated by proteins known as transcription factors. Several antivirals are now being designed to block attachment of transcription factors to viral DNA.

[0078] Antisense molecules are segments of DNA or RNA designed as mirror images to critical sections of viral genomes, and the binding of these antisense segments to these target sections blocks the operation of those genomes. A phospho roth bate antisense drug named fomivirsen is used to treat opportunistic eye infections in AIDS patients caused by cytomegalovirus. Morpholino oligos have been used to experimentally suppress many viral types including caliciviruses, flaviviruses (including WNV, Dengue and HCV), and corona viruses.

[0079] Yet another antiviral technique inspired by genomics is a set of drugs based on ribozymes, which are enzymes that will cut apart viral RNA or DNA at selected sites. A ribozyme antivirals exist or are under development to treat hepatitis C and HIV.

[0080] Some viruses include a protease that cuts viral protein chains apart so they can be assembled into their final configuration. Protease inhibitors can thus attack the virus at the assembly phase of its life-cycle.

[0081] The class of M2 channel blockers includes amantadine and rimantadine.

Release phase

[0082] The final stage in the life cycle of a virus is the release of completed viruses from the host cell, and this step has also been targeted by antiviral drug developers. Zanamivir (Relenza), oseltamivir (Tamiflu) and Peramivir have been introduced to treat influenza by preventing the release of viral particles by blocking the neuraminidase enzyme (neuraminidase inhibitors) that is found on the surface of flu viruses. Zanamivir may be formulated for as a powder for inhalation. A commercially-available inhalation

powder of Zanamivir (Relenza) is formulated with lactose, yielding relatively large particles.

[0083] Neuraminidase is a glycoside hydrolase enzyme (EC 3.2.1.18). It is frequently found as an antigenic glycoprotein and is best known as one of the enzymes found on the surface of the Influenza virus. Some variants of the influenza neuraminidase confer more virulence to the virus than others. At least four neuraminidases in the human genome have been described. Neuraminidase has functions that aid in the efficiency of virus release from cells. Neuraminidase cleaves terminal sialic acid residues from carbohydrate moieties on the surfaces of infected cells. This promotes the release of progeny viruses from infected cells. Neuraminidase also cleaves sialic acid residues from viral proteins, preventing aggregation of viruses. Administration of chemical inhibitors of neuraminidase is a treatment that limits the severity and spread of viral infections.

[0084] Antivirals may also be classified by chemical type, such as: nucleoside analogs; peptide anologs, neuraminic acid mimetics, proteins, triazoles, tricyclic amines, small cyclic molecules and kinase inhibitors, for example.

[0085] SiRNA (small interfering RNAs) may be employed as anti-infectives, especially as anti-virals.

[0086] In addition to those identified above, examples of antivirals include, but are not limited to, acyclovir, gangcyclovir, azidothymidine, cytidine arabinoside, ribavirin, rifampacin, iododeoxyuridine, poscamet, and trifluridine.

[0087] In one or more embodiments, the present invention comprises one or more antivirals formulated for inhalation in the form of particles to have a MMAD particle size of less than about 7 microns, such as less than about 6 or 5 or 4 or 3 or 2 microns.

[0088] In one or more embodiments, the antiviral composition comprises a powder which can be administered using an inhaler device. In one or more embodiments, the antiviral composition comprises particles which are sufficiently small to provide good lung distribution, through the lower airways and alveoli. Such distribution not only aids in therapeutic effect, but helps to mitigate, reduce or eliminate toxicity associated with uneven distribution of drug. Moreover, the specified small particle size helps to mitigate, reduce or eliminate bronchospasm.

[0089] In one or more embodiments, the antiviral composition comprises particles comprising at least two different antiviral actives and an excipient matrix such that the individual particles comprise predominantly all three components. In one or more embodiments, the antiviral composition comprises particles comprising a single antiviral

active and an excipient matrix such that the individual particles comprise predominantly only two components. In one or more embodiments, the antiviral composition comprises a mixture of first and second particles, wherein the first particle comprises a first antiviral active and an excipient matrix, and the second particle comprises a second antiviral active and an excipient matrix, and wherein the excipient may be the same or different, and the first and second particles may be of substantially similar physical characteristics, or may differ in one or more physical characteristics.

[0090] In one or more embodiments, the antiviral formulation of the present invention when administered by inhalation, affords a beneficial ratio of lung C max to serum C max . Thus a lung concentration is sufficiently high to provide therapeutic effectiveness, while a serum concentration is sufficiently low to eliminate or minimize side effects, unwanted effects, adverse reactions and/or toxicity. In one or more embodiments, a ratio of lung C m a x to serum C max is at least about 2000:1.

[0091] When a combination of active agents is used, the agents may be provided in combination in a single species of pharmaceutical composition or individually in separate species of pharmaceutical compositions. Further, the pharmaceutical composition may be combined with one or more other active or bioactive agents that provide the desired dispersion stability or powder dispersibility.

[0092] The amount of active agent{s), e.g., antiviral, in the pharmaceutical composition may vary. The amount of active agent(s) is typically at least about 0.5 wt%, such as at least about 1 wt%, at least about 2 wt%, at least about 5 wt%, at least about 10 wt%, at least about 20 wt%, at least about 30 wt%, at least about 40 wt%, at least about 50 wt%, at least about 60 wt%, at least about 70 wt%, or at least about 80 wt%, of the total amount of the pharmaceutical composition. The amount of active agent(s) generally varies between about 0.1 wt% to 100 wt%, such as about 1 wt% to about 95 wt%, about 2 wt% to about 90 wt%, about 30 wt% to about 80 wt%, about 40 wt% to about 70 wt%, about 50 wt% to about 60 wt%, about 1 wt% to about 20 wt%, about 2 wt% to about 10 wt%, about 5 wt% to about 50 wt%, or about 4 wt% to about 20 wt%.

[0093] As noted above, the pharmaceutical composition may include one or more pharmaceutically acceptable excipients. Examples of pharmaceutically acceptable excipients include, but are not limited to, lipids, metal ions, surfactants, amino acids, carbohydrates, buffers, salts, polymers, and the like, and combinations thereof.

[0094] Examples of lipids include, but are not limited to, phospholipids, glycolipids, ganglioside GM1 , sphingomyelin, phosphatide acid, cardiolipin; lipids bearing polymer

chains such as polyethylene glycol, chitin, hyaluronic acid, or polyvinylpyrrolidone; lipids bearing sulfonated mono-, di-, and polysaccharides; fatty acids such as palmitic acid, stearic acid, and oleic acid; cholesterol, cholesterol esters, and cholesterol hemisuccinate.

[0095] In one or more embodiments, the phospholipid comprises a saturated phospholipid, such as one or more phosphatidylcholines. Exemplary acyl chain lengths are 16:0 and 18:0 (i.e., palmitoyl and stearoyl). The phospholipid content may be determined by the active agent activity, the mode of delivery, and other factors.

[0096] Phospholipids from both natural and synthetic sources may be used in varying amounts. When phospholipids are present, the amount is typically sufficient to coat the active agent(s) with at least a single molecular layer of phospholipid. In general, the phospholipid content ranges from about 5 wt% to about 99.9 wt%, such as about 20 wt% to about 80 wt%.

[0097] Generally, compatible phospholipids comprise those that have a gel to liquid crystal phase transition greater than about 40 0 C, such as greater than about 60 0 C, or greater than about 80 0 C. The incorporated phospholipids may be relatively long chain {e.g., C 16 -C 22 ) saturated lipids. Exemplary phospholipids useful in the disclosed stabilized preparations include, but are not limited to, phosphoglycerides such as dipalmitoylphosphatidylcholine, distearoylphosphatidylcholine, diarachidoylphosphatidylcholine, dibehenoylphosphatidylcholine, dimyristoylphosphatidylcholine, diphosphatidyl glycerols, short-chain phosphatidylcholines, hydrogenated phosphatidylcholine, E-100-3 (available from Lipoid KG, Ludwigshafen, Germany), long-chain saturated phosphatidylethanolamines, long- chain saturated phosphatidylserines, long-chain saturated phosphatidylglycerols, long- chain saturated phosphatidylinositols, phosphatidic acid, phosphatidylinositol, and sphingomyelin.

[0098] Examples of metal ions include, but are not limited to, divalent cations, including calcium, magnesium, zinc, iron, and the like. For instance, when phospholipids are used, the pharmaceutical composition may also comprise a polyvalent cation, as disclosed in WO 01/85136 and WO 01/85137, which are incorporated herein by reference in their entireties. The polyvalent cation may be present in an amount effective to increase the melting temperature (T m ) of the phospholipid such that the pharmaceutical composition exhibits a T m which is greater than its storage temperature (T s ) by at least about 20 0 C, or 25°C or 30 0 C or 35°C or 40 0 C or 45 0 C or more. The molar ratio of polyvalent cation to phospholipid may be at least about 0.05:1, such as about 0.05:1 to about 2.0: 1 or about 0.25: 1 to about 1.0: 1. An example of the molar ratio of polyvalent

cation:phospholipid is about 0.50:1. When the polyvalent cation is calcium, it may be in the form of calcium chloride. Although metal ion, such as calcium, is often included with phospholipid, none is required.

[0099] One or more embodiments of the pharmaceutical composition may include one or more surfactants. For instance, one or more surfactants may be in the liquid phase with one or more being associated with solid particles or particles of the composition. By "associated with" it is meant that the pharmaceutical compositions may incorporate, sorb, adsorb, absorb, be coated with, or be formed by the surfactant. Surfactants include, but are not limited to, fluorinated and nonfluorinated compounds, such as saturated and unsaturated lipids, nonionic detergents, nonionic block copolymers, ionic surfactants, and combinations thereof. It should be emphasized that, in addition to the aforementioned surfactants, suitable fluorinated surfactants are compatible with the teachings herein and may be used to provide the desired preparations.

[00100] Examples of nonionic detergents include, but are not limited to, sorbitan esters including sorbitan trioleate (Span™ 85), sorbitan sesquioleate, sorbitan monooleate, sorbitan monolaurate, polyoxyethylene (20) sorbitan monolaurate, and polyoxyethylene (20) sorbitan monooleate, oleyl polyoxyethylene (2) ether, stearyf polyoxyethylene (2) ether, lauryl polyoxyethylene (4) ether, glycerol esters, and sucrose esters. Other suitable nonionic detergents can be easily identified using McCutcheon's Emulsifiers and Detergents (McPublishing Co., Glen Rock, New Jersey), which is incorporated by reference herein in its entirety.

[00101] Examples of block copolymers include, but are not limited to, diblock and triblock copolymers of polyoxyethylene and polyoxypropylene, including poloxamer 188 (Pluronic™ F-68), poloxamer 407 (Pluronic™ F-127), and poloxamer 338.

[00102] Examples of ionic surfactants include, but are not limited to, sodium sulfosuccinate, and fatty acid soaps.

[00103] Examples of amino acids include, but are not limited to, hydrophobic amino acids. Use of amino acids as pharmaceutically acceptable excipients is known in the art as disclosed in WO 95/31479, WO 96/32096, and WO 96/32149, which are incorporated herein by reference.

[00104] Hydrophobic amino acids and lipids are capable of providing a particle surface of low surface energy. Magnesium stearate may also be used as an excipient to reduce surface energy.

[00105] Examples of carbohydrates include, but are not limited to, monosaccharides, disaccharides, and polysaccharides. For example, monosaccharides such as dextrose (anhydrous and monohydrate), galactose, mannitol, D-mannose, sorbitol, sorbose and the like; disaccharides such as lactose, maltose, sucrose, trehalose, and the like; trisaccharides such as raffinose and the like; and other carbohydrates such as starches (hydroxyethylstarch), cyclodextrins and maltodextrins.

[00106] Examples of buffers include, but are not limited to, tris, citrate, acetate, phosphate, TES and MES.

[00107] Examples of acids include, but are not limited to, organic acids such as carboxylic acids, in particular mono and di- carboxylic acids. .

[00108] Examples of salts include, but are not limited to, sodium chloride, salts of carboxylic acids, (e.g., sodium citrate, sodium ascorbate, magnesium gluconate, sodium gluconate, tromethamine hydrochloride, etc.), ammonium carbonate, ammonium acetate, ammonium chloride, and the like.

[00109] The excipients may be glass forming excipients providing an amorphous glass, e.g., with a glass transition temperature that is at least 20 0 C greater than the storage temperature. Glass forming systems are disclosed in U.S. Patent Nos. 6,258,341; 5,098,893; 5,928,469; and 6,071 ,428, which are incorporated herein by reference.

[00110J The pharmaceutical composition of one or more embodiments of the present invention may also include a biocompatible, such as biodegradable polymer, copolymer, or blend or other combination thereof. In this respect useful polymers comprise polylactides, polylactide-glycolides, cyclodextrins, polyacrylates, methylcellulose, carboxymethylcellulose, polyvinyl alcohols, polyanhydrides, polylactams, polyvinyl pyrrolidones, polysaccharides (dextrans, starches, chitin, chitosan, etc.), hyaluronic acid, proteins, (albumin, collagen, gelatin, etc.). Those skilled in the art will appreciate that, by selecting the appropriate polymers, the delivery efficiency of the composition and/or the stability of the dispersions may be tailored to optimize the effectiveness of the active agent(s).

[00111] Besides the above mentioned pharmaceutically acceptable excipients, it may be desirable to add other pharmaceutically acceptable excipients to the pharmaceutical composition to improve particle rigidity, production yield, emitted dose and deposition, shelf-life, and patient acceptance. Such optional pharmaceutically acceptable excipients include, but are not limited to: coloring agents, taste masking agents, buffers, hygroscopic agents, antioxidants, and chemical stabilizers. Further, various pharmaceutically

acceptable excipients may be used to provide structure and form to the particle compositions (e.g., latex particles). In this regard, it will be appreciated that the rigidifying components can be removed using a post-production technique such as selective solvent extraction.

[00112] The pharmaceutical compositions may also include mixtures of pharmaceutically acceptable excipients. For instance, mixtures of carbohydrates and amino acids are within the scope of the present invention. Other combinations of excipients include, but are not limited to, (a) distearoylphosphatidylcholine to calcium chloride (e.g., in a 2:1 molar ratio); (b) core-shell particles comprised of a shell of trileucine, and a core comprised of glass forming excipients, including sodium citrate and trehalose.

[00113] As noted above, in one or more embodiments, the present invention may comprise one or more antivirals combined with one or more antibiotics, such as an antifungal and/or antbiotic.

[00114] Examples of antifungals include, but are not limited to, azoles (e.g., imidazoles, itraconazole, pozaconazole), micafungin, caspafungin, salicylic acid, oxiconazole nitrate, ciclopirox olamine, ketoconazole, miconazole nitrate, and butoconazofe nitrate.

[00115] Examples of antibiotics include, but are not limited to, penicillin and drugs of the penicillin family of antimicrobial drugs, including but not limited to penicillin-G, penicillin-V, phenethicillin, ampicillin, amoxacillin, cyclacillin, bacampicillin, hetacillin, cloxacillin, dicloxacillin, methicillin, nafcillin, oxacillin, azlocillin, carbenicillin, mezlocillin, piperacillin, ticaricillin, and imipenim; cephalosporin and drugs of the cephalosporin family, including but not limited to cefadroxil, cefazolin, caphalexn, cephalothin, cephapirin, cephradine, cefaclor, cefamandole, cefonicid, cefoxin, cefuroxime, ceforanide, cefotetan, cefinetazole, cefoperazone, cefotaxime, ceftizoxime, ceftizone, moxalactam, ceftazidime, and cefixime; aminoglycoside drugs and drugs of the aminoglycoside family, including but not limited to streptomycin, neomycin, kanamycin, gentamycin, tobramycin, amikacin, and netilmicin; macrolide and drugs of the macrolide family, exemplified by amphotericin B 1 azithromycin, clarithromycin, roxithromycin, erythromycin, lincomycin, and clindamycin; tetracyclin and drugs of the tetracyclin family, for example, tetracyclin, oxytetracyclin, democlocyclin, methacyclin, doxycyclin, and minocyclin; quinoline and quinoline-like drugs, such as, for example, naladixic acid, cinoxacin, norfloxacin, ciprofloxacin, ofloxicin, enoxacin, and pefloxacin; antimicrobial peptides, including but not limited to polymyxin B, colistin, and bacatracin, as well as other antimicrobial peptides such as defensins,

magainins, cecropins, and others, provided as naturally-occurring or as the result of engineering to make such peptides resistant to the action of pathogen-specific proteases and other deactivating enzymes; other antimicrobial drugs, including chloramphenicol, vancomycin, rifampicin, metronidazole, voriconazole, fluconazole, ethambutol, pyrazinamide, sulfonamides, isoniazid, and erythromycin.

[00116] The compositions of one or more embodiments of the present invention may take various forms, such as dry powders, capsules, tablets, reconstituted powders, suspensions, or dispersions comprising a non-aqueous phase, such as propellants (e.g., chlorofluorocarbon, hydrofluoroalkane). The moisture content of dry powder may be less than about 15 wt%, such as less than about 10 wt%, less than about 5 wt%, less than about 2 wt%, less than about 1 wt%, or less than about 0.5 wt%. Such powders are described in WO 95/24183, WO 96/32149, WO 99/16419, WO 99/16420, and WO 99/16422, which are incorporated herein by reference in their entireties.

[00117] One or more embodiments of the invention involve homogeneous compositions of antiviral incorporated in a matrix material with little, if any, unincorporated antiviral. For instance, at least about 40 wt%, at least about 50 wt%, at least about 60 wt%, at least about 70%, at least about 80%, at least about 90 wt%, at least about 95 wt%, or at least about 99 wt%, of the composition may comprise particles including both antiviral and matrix material.

[00118] It is particularly advantageous for the particle size (such as a mass median diameter, and/or a geometric diameter, and or aerodynamic diameter) of the particles to be below 3.0 microns, preferably below 2.5 microns, and more preferably below about 2.0 microns, in order to provide highly dispersible, homogenous compositions of active agent incorporated into the matrix material. Accordingly, a preferred embodiment is directed to homogeneous compositions of active agent incorporated in a matrix material without any unincorporated active agents particles. In some cases, however, a heterogeneous composition may be desirable in order to provide a desired pharmacokinetic profile of the antiviral to be administered, and in these cases, a large antiviral particle (e.g., mass median diameter of about 3 μm to about 10 μm, or larger) may be used.

[00119] In one or more versions, the pharmaceutical composition comprises an antiviral incorporated into a phospholipid matrix. The pharmaceutical composition may comprise phospholipid matrices that incorporate the active agent and that are in the form of particles that are hollow and/or porous microstructures, as described in the aforementioned WO 99/16419, WO 99/16420, WO 99/16422, WO 01/85136, and WO 01/85137, US 20040156792; and US 20050214224, all of which are incorporated herein

by reference in their entireties. The hollow and/or porous microstructures are useful in delivering the antiviral to the lungs because the density, size, and aerodynamic qualities of the hollow and/or porous microstructures facilitate transport into the deep lungs during a user's inhalation. In addition, the phospholipid-based hollow and/or porous microstructures reduce the attraction forces between particles, making the pharmaceutical composition easier to deagglomerate during aerosolization and improving the flow properties of the pharmaceutical composition making it easier to process.

[00120] In one or more versions, the pharmaceutical composition is composed of hollow and/or porous microstructures having a bulk density less than about 1.0 g/cm 3 , less than about 0.8 g/cm 3 , less than about 0.5 g/cm 3 , less than about 0.3 g/cm 3 , less than about 0.2 g/cm 3 , or less than about 0.1 g/cm 3 . For example, small porous particles of the present invention may have a bulk density ranging from 0.01 g/cm 3 to 0.4 g/cm 3 , such as from 0.03 g/cm 3 to 0.25 g/cm 3 . Particle density can be controlled by controlling the drying rate and surface composition of spray-dried particles, or by inclusion of a specific pore forming agent in the formulation. Preferred pore-forming agents are medium chain fluorocarbons such as perfluorooctyl bromide (PFOB), perfluorodecalin (PFD), and perfluorooctyl ethane (PFOE). Bulk densities may be determined by simply weighing a No. 2 capsule (0.37 ml fill volume) filled with powder, subtracting the weight of the capsule and dividing by the volume.

[00121] By providing low bulk density particles or particles, the minimum powder mass that can be filled into a unit dose container is reduced, which eliminates the need for carrier particles. That is, the relatively low density of the powders of one or more embodiments of the present invention provides for the reproducible administration of relatively low dose pharmaceutical compounds. Moreover, the elimination of carrier particles will potentially reduce throat deposition and any "gag" effect or coughing, since large carrier particles, e.g., lactose particles, will impact the throat and upper airways due to their size.

[00122] In one or more versions, the pharmaceutical composition is in dry powder form and is contained within a unit dose receptacle which may be inserted into or near the aerosolization apparatus to aerosolize the unit dose of the pharmaceutical composition. This version is useful in that the dry powder form may be stably stored in its unit dose receptacle for a long period of time. The pharmaceutical compositions of one or more embodiments of the present invention may be stable for at least about 2 years. In some versions, no refrigeration may be required to obtain stability. In other versions, reduced

temperatures, e.g., at 2-8 0 C, may be used to prolong stable storage. In many versions, the storage stability allows aerosolization with an external power source.

[00123] It will be appreciated that the pharmaceutical compositions disclosed herein may comprise a structural matrix that exhibits, defines or comprises voids, pores, defects, hollows, spaces, interstitial spaces, apertures, perforations or holes. The absolute shape (as opposed to the morphology) of the perforated microstructure is generally not critical and any overall configuration that provides the desired characteristics is contemplated as being within the scope of the invention. Accordingly, some embodiments comprise approximately spherical shapes. However, non-spherical shapes, or amorphous shapes, such as collapsed, deformed or fractured spherical-shaped particles are also within the scope of the invention.

[00124] In some versions the antiviral is incorporated in a matrix that forms a discrete particle, and the pharmaceutical composition comprises a plurality of the discrete particles. The discrete particles may be sized so that they are effectively administered and/or so that they are available where needed. For example, for an aerosolizable pharmaceutical composition, the particles are of a size that allows the particles to be aerosolized and delivered to a user's respiratory tract during the user's inhalation.

[00125] In some versions, the pharmaceutical composition comprises particles having a mass median diameter less than about 20 μm, such as less than about 10 μm, less than about 7 μm, or less than about 5 μm, and may, e.g., range from 1 μm to 10 μm, such as from 1 μm to 5 μm. The particles may have a mass median aerodynamic diameter (MMAD) ranging from about 1 μm to about 6 μm, such as about 1.5 μm to about 5 μm, or about 2 μm to about 4 μm. The particle size and/or size distribution are selected to maximize and/or optimize the number and/or mass of particles that will reach the deep lung. The particle size and/or size distribution are additionally or alternatively selected to minimize or optimize the number and/or mass of particles that may be exhaled.

[00126] The matrix material may comprise a hydrophobic or a partially hydrophobic material. For example, the matrix material may comprise a lipid, such as a phospholipid, and/or a hydrophobic amino acid, such as leucine or tri-leucine. Examples of phospholipid matrices are described in WO 99/16419, WO 99/16420, WO 99/16422, WO 01/85136, and WO 01/85137 and in U.S. Patent Nos. 5,874,064; 5,855,913; 5,985,309; and 6,503,480, and in U.S. Application Publication No. 20040156792, all of which are incorporated herein by reference in their entireties. Examples of hydrophobic amino acid matrices are described in U.S. Patent Nos. 6,372,258 and 6,358,530, and in U.S.

Application Publication No. 20020177562, each of which are incorporated herein by reference in their entireties.

[00127] When phospholipids are utilized as the matrix material, the pharmaceutical composition may also comprise a polyvalent cation, as disclosed in WO 01/85136 and WO 01/85137, which are incorporated herein by reference in their entireties.

[00128] According to another embodiment, release kinetics of the active agent(s) containing composition is controlled. According to one or more embodiments, the compositions of the present invention provide immediate release of the active agent(s). Alternatively, the compositions of other embodiments of the present invention may be provided as non-homogeneous mixtures of active agent incorporated into a matrix material and unincorporated active agent in order to provide desirable release rates of antifungal agent. According to this embodiment, active agents formulated using the emulsion-based manufacturing process of one or more embodiments of the present invention have utility in immediate release applications when administered to the respiratory tract. Rapid release is facilitated by: (a) the high specific surface area of the low density porous powders and; (c) the low surface energy of the particles.

[00129] Alternatively, it may be desirable to engineer the particle matrix so that extended release of the active agent(s) is effected. This may be particularly desirable when the active agent(s) is rapidly cleared from the lungs or when sustained release is desired. For example, the nature of the phase behavior of phospholipid molecules is influenced by the nature of their chemical structure and/or preparation methods in spray- drying feedstock and drying conditions and other composition components utilized. In the case of spray-drying of active agent(s) solubilized within a small unilamellar vesicle (SUV) or multilamellar vesicle (MLV), the active agent(s) are encapsulated within multiple bilayers and are released over an extended time.

[00130] In contrast, spray-drying of a feedstock comprised of emulsion droplets and dispersed or dissolved active agent(s) in accordance with the teachings herein leads to a phospholipid matrix with less long-range order, thereby facilitating rapid release. While not being bound to any particular theory, it is believed that this is due in part to the fact that the active agent(s) are never formally encapsulated in the phospholipid, and the fact that the phospholipid is initially present on the surface of the emulsion droplets as a monolayer (not a bilayer as in the case of liposomes). The spray-dried particles prepared by the emulsion-based manufacturing process of one or more embodiments of the present invention often have a high degree of disorder. Also, the spray-dried particles typically have low surface energies, where values as low as 20 mN/m have been observed for

spray-dried DSPC particles (determined by inverse gas chromatography). Small angle X- ray scattering (SAXS) studies conducted with spray-dried phospholipid particles have also shown a high degree of disorder for the lipid, with scattering peaks smeared out, and length scales extending in some instances only beyond a few nearest neighbors.

[00131] It should be noted that a matrix having a high gel to liquid crystal phase transition temperature is not sufficient in itself to achieve sustained release of the active agent(s). Having sufficient order for the bilayer structures is also important for achieving sustained release. To facilitate rapid release, an emulsion-system of high porosity (high surface area), and minimal interaction between the drug substance and phospholipid may be used. The pharmaceutical composition formation process may also include the additions of other composition components (e.g., small polymers such as Pluronic F-68; carbohydrates, salts, hydrotropes) to break the bilayer structure are also contemplated. [00132] To achieve a sustained release, incorporation of the phospholipid in bilayer form may be used, especially if the active agent is encapsulated therein. In this case increasing the T m of the phospholipid may provide benefit via incorporation of divalent counterions or cholesterol. As well, increasing the interaction between the phospholipid and drug substance via the formation of ion-pairs (negatively charged active + steaylamine, positively charged active + phosphatidylglycerol) would tend to decrease the dissolution rate. If the active is amphiphilic, surfactant/surfactant interactions may also slow active dissolution.

[00133] The addition of divalent counterions (e.g., calcium or magnesium ions) to long-chain saturated phosphatidylcholines results in an interaction between the negatively charged phosphate portion of the zwitterionic headgroup and the positively charged metal ion. This results in a displacement of water of hydration and a condensation of the packing of the phospholipid lipid headgroup and acyl chains. Further, this results in an increase in the Tm of the phospholipid. The decrease in headgroup hydration can have profound effects on the spreading properties of spray-dried phospholipid particles on contact with water.

[00134] For example, upon reconstitution, the surface tension of spray-dried DSPC/Ca mixtures at the air/water interface decreases to equilibrium values (about 20 mN/m) as fast as a measurement can be taken. In contrast, liposomes of DSPC decrease the surface tension (about 50 mN/m) very little over a period of hours, and it is likely that this reduction is due to the presence of hydrolysis degradation products such as free fatty acids in the phospholipid. Single-tailed fatty acids can diffuse much more rapidly to the air/water interface than can the hydrophobic parent compound.

[00135] In one or more embodiments, compositions of the invention may comprise one or more di- or tripeptides containing two or more leucine residues. Such di-leucyl- containing dipeptides (e.g., dileucine) and tripeptides are superior in their ability to increase the dispersibility of powdered compositions, and, as demonstrated in the Examples, are better than leucine in improving aerosol performance.

[00136] Di-leucyl containing tripeptides for use in the invention are tripeptides having the formula, X-Y-Z, where at least X and Y or X and Z are feucyl residues (i.e., the leucyl residues can be adjacent to each other (at the 1 and 2 positions), or can form the ends of the trimer (occupying positions 1 and 3). The remaining amino acid contained in the trimer can be any amino acid as defined in section I above. Suitable are amino acids such as glycine (gly), alanine (ala), valine (val), leucine (leu), isoleucine (ile), methionine (met), proline (pro), phenylalanine (phe), trytophan (trp), serine (ser), threonine (thr), cysteine (cys), tyrosine (tyr), asparagine (asp), glutamic acid (glu), lysine (lys), arginine (arg), histidine (his), norleucine (nor), and modified forms thereof. Preferably, for di-leucyl containing trimers, the third amino acid component of the trimer is one of the following: leucine (leu), valine (val), isofeucine (isoleu), tryptophan (try) alanine (ala), methionine (met), phenylalanine (phe), tyrosine (tyr), histidine (his), and proline (pro). Exemplary trimers for use in the invention include but are not limited to the following: leu-leu-gly, leu- leu-ala, leu-leu-val, !eu-leu-leu, leu-leu-ile, leu-leu-met, leu-leu-pro, leu-leu-phe, leu-leu- trp, leu-leu-ser, leu-leu-thr, leu-leu-cys, leu-leu-tyr, leu-leu-asp, leu-leu-glu, leu-leu-lys, leu-leu-arg, leu-leu-his, leu-leu-nor, leu-gly-leu, leu-ala-leu, leu-val-leu, leu-ile-leu, leu- met-leu, leu-pro-leu, leu-phe-leu, leu-trp-leu, leu-ser-leu, leu-thr-leu, leu-cys-leu, leu-try- leu, leu-asp-leu, leu-glu-leu, leu-lys-leu, leu-arg-leu, leu-his-leu, and leu-nor-leu. The foregoing trimers in reverse order are also suitable. Particularly preferred peptides are dileucine and trileucine.

[00137] In one or more embodiments, additional dispersibility-enhancing peptides for use in the invention are 4-mers and 5-mers containing two or more leucine residues. The leucine residues may occupy any position within the peptide, and the remaining (i.e., non- leucyl) amino acids positions are occupied by any amino acid as described above, provided that the resulting 4-mer or 5-mer has a solubility in water of at least about 1 mg/ml. Preferably, the non-leucyl amino acids in a 4-mer or 5-mer are hydrophilic amino acids such as lysine, to thereby increase the solubility of the peptide in water. Also suitable are di- and tripeptides having a glass transition temperature greater than about 40DC.

[00138] As disclosed in US 6518239 and US 6835372, both to Kuo et al, the disclosures of which are fully incorporated herein for all purposes by reference, preferred di- and tripeptides for use in the present invention are those peptides that are surface active. Dileucine and trileucine are extremely effective, even when present in low concentrations, at significantly depressing the surface tension of water. The noted Kuo et al patents show that dipeptides and tripeptides containing two or more leucines have a much greater surface activity than dipeptides and tripeptides composed of fewer than two leucyl residues. Due to their highly surface active nature, the di- and tripeptides of the invention, when contained in dry powder compositions, tend to concentrate on the surface (enriching the surface) of the powder particles, thereby imparting to the resulting particles high dispersivities.

[00139] In embodiments comprising di- or tri- peptide, the compositions of the invention will contain from about 1% to about 99% by weight di- or tripeptide, preferably from about 2% to about 75% by weight di- or tripeptide, and even more preferably from about 5% to about 50% by weight di- or tripeptide. Typically, the optimal amount of di- or tripeptide is determined experimentally, i.e., by preparing compositions containing varying amounts of di- or tripeptide (ranging from low to high), examining the dispersibilities of the resulting compositions, and further exploring the range at which optimal aerosol performance is attained. Generally, for trileucine containing dry powder formulations, an optimal amount of trileucine appears to be around 22-25% by weight.

[00140] In other versions, the pharmaceutical composition comprises low density particles achieved by co-spray-drying with a perfluorocarbon-in-water emulsion. Examples of perfluorocarbons include, but are not limited to, perfluorohexane, perfluorooctyl bromide, perfluorooctyl ethane, perfluorodecalin, perfluorobutyi ethane.

[00141] In accordance with the teachings herein the particle compositions will preferably be provided in a "dry" state. That is, in one or more embodiments, the particles will possess a moisture content that allows the powder to remain chemically and physically stable during storage at ambient or reduced temperature and remain dispersible. In this regard, there is little or no change in primary particle size, content, purity, and aerodynamic particle size distribution.

[00142] As such, the moisture content of the particles is typically less than about 10 wt%, such as less than about 6 wt%, less than about 3 wt%, or less than about 1 wt%. The moisture content is, at least in part, dictated by the composition and is controlled by the process conditions employed, e.g., inlet temperature, feed concentration, pump rate, and blowing agent type, concentration and post drying. Reduction in bound water leads to

significant improvements in the dispersibility and flowability of phospholipid based powders, leading to the potential for highly efficient delivery of powdered lung surfactants or particle composition comprising active agent dispersed in the phospholipid. The improved dispersibility allows simple passive DPI devices to be used to effectively deliver these powders.

[00143] Yet another version of the pharmaceutical composition includes particle compositions that may comprise, or may be partially or completely coated with, charged species that prolong residence time at the point of contact or enhance penetration through mucosae. For example, anionic charges are known to favor mucoadhesion while cationic charges may be used to associate the formed particle with negatively charged bioactive agents such as genetic material. The charges may be imparted through the association or incorporation of polyanionic or polycationic materials such as polyacrylic acids, polylysine, polylactic acid, and chitosan.

[00144] These unit dose pharmaceutical compositions may be contained in a container. Examples of containers include, but are not limited to, capsules, blisters, vials, ampoules, or container closure systems made of metal, polymer (e.g., plastic, elastomer), glass, or the like.

[00145] The container may be inserted into an aerosol ization device. The container may be of a suitable shape, size, and material to contain the pharmaceutical composition and to provide the pharmaceutical composition in a usable condition. For example, the capsule or blister may comprise a wall which comprises a material that does not adversely react with the pharmaceutical composition. In addition, the wall may comprise a material that allows the capsule to be opened to allow the pharmaceutical composition to be aerosolized. In one version, the wall comprises one or more of gelatin, hydroxypropyl methylcellulose (HPMC), polyethyleneglycol-compounded HPMC, hydroxyproplycellulose, agar, aluminum foil, or the like. In one version, the capsule may comprise telescopically adjoining sections, as described for example in U.S. Patent No. 4,247,066 which is incorporated herein by reference in its entirety. The size of the capsule may be selected to adequately contain the dose of the pharmaceutical composition. The sizes generally range from size 5 to size 000 with the outer diameters ranging from about 4.91 mm to 9.97 mm, the heights ranging from about 11.10 mm to about 26.14 mm, and the volumes ranging from about 0.13 mL to about 1.37 mL Suitable capsules are available commercially from, for example, Shionogi Qualicaps Co. in Nara, Japan and Capsugel in Greenwood, South Carolina. After filling, a top portion may be placed over the bottom portion to form a capsule shape and to contain the powder within the capsule, as

described in U.S. Patent Nos. 4,846,876 and 6,357,490, and in WO 00/07572, which are incorporated herein by reference in their entireties. After the top portion is placed over the bottom portion, the capsule can optionally be banded.

[00146] In one or more versions, the pharmaceutical composition comprising antiviral is aerosolizable so that it may be delivered to the lungs of a patient during the patient's inhalation. In this way the antiviral in the pharmaceutical composition is delivered directly to the site of infection. This is advantageous over systemic administration. Because the active agent(s) often have renal or other toxicity, minimizing systemic exposure is typically preferred. Therefore, the amount of active agent(s) that may be delivered to the lungs is preferably limited to the minimum pharmacologically effective dose. By administering the active agent(s) directly to the lungs, a greater amount may be delivered to the site in need of the therapy while significantly reducing systemic exposure.

[00147J The pharmaceutical compositions of one or more embodiments of the present invention lack taste. In this regard, although taste masking agents are optionally included within the composition, the compositions often lack taste even without a taste masking agent.

[00148] The particles, particles, and compositions of one or more embodiments of the present invention may be made by any of the various methods and techniques known and available to those skilled in the art. The pharmaceutical composition may be produced using various known techniques. For example, the composition may be formed by spray drying, lyophilization, milling (e.g., wet milling, dry milling), and the like.

[00149] A liquid solution, suspension or dispersion of one or more antiviral actives, and optional excipient or excipients in an appropriate solvent may be made, and then converted to a powder form by a liquid or solvent removal process. Such a process may comprise a supercritical solvent extraction, spray-drying, or other solvent removal process.

[00150] In spray drying, the preparation to be spray-dried or feedstock can be any solution, coarse suspension, slurry, colloidal dispersion, or paste that may be atomized using the selected spray drying apparatus. In the case of insoluble agents, the feedstock may comprise a suspension as described above. Alternatively, a dilute solution and/or one or more solvents may be utilized in the feedstock. In one or more embodiments, the feed stock will comprise a colloidal system such as an emulsion, reverse emulsion, microemulsion, multiple emulsion, particle dispersion, or slurry.

[00151] In one version, the antiviral and the matrix material are added to an aqueous feedstock to form a feedstock solution, suspension, or emulsion. The feedstock is then spray dried to produce dried particles comprising the matrix material and the antiviral. Suitable spray-drying processes are known in the art, for example as disclosed in WO 99/16419 and U.S. Patent Nos. 6,077,543; 6,051,256; 6,001 ,336; 5,985,248; and 5,976,574, which are incorporated herein by reference in their entireties.

[00152] Whatever components are selected, the first step in particle production typically comprises feedstock preparation. If a phosphoiipids-based particle is intended to act as a carrier for the antiviral, the selected active agent(s) may be introduced into a liquid, such as water, to produce a concentrated suspension. The concentration of antiviral and optional active agents typically depends on the amount of agent required in the final powder and the performance of the delivery device employed, e.g., the fine particle dose for a metered dose inhaler (MDI) or a dry powder inhaler (DPI).

[00153] Any additional active agent(s) may be incorporated in a single feedstock preparation and subjected to a solvent removal process (e.g. spray drying) to provide a single pharmaceutical composition species comprising a plurality of active agents. Conversely, individual active agents could be added to separate stocks and subjected to a solvent removal process (e.g. spray drying) separately to provide a plurality of pharmaceutical composition species with different compositions. These individual species could be added to the suspension medium or dry powder dispensing compartment in any desired proportion and placed in the aerosol delivery system as described below.

[00154] Polyvalent cation may be combined with the antiviral suspension, combined with the phospholipid emulsion, or combined with an oN-in-water emulsion formed in a separate vessel. The antiviral may also be dispersed directly in the emulsion.

[00155] For example, polyvalent cation and phospholipid may be homogenized in hot distilled water (e.g., 70 0 C) using a suitable high shear mechanical mixer (e.g., Ultra-Turrax model T-25 mixer) at 8000 rpm for 2 to 5 min. Typically, 5 to 25 g of fluorocarbon is added dropwise to the dispersed surfactant solution while mixing. The resulting polyvalent cation-containing perfluorocarbon in water emulsion may then be processed using a high pressure homogenizer to reduce the particle size. Typically, the emulsion is processed for five discrete passes at 12,000 to 18,000 PSI and kept at about 50 0 C to about 80 0 C.

[00156] When the polyvalent cation is combined with an oil-in-water emulsion, the dispersion stability and dispersibility of the spray dried pharmaceutical composition can be improved by using a blowing agent, as described in US 6565885, which is incorporated

herein by reference in its entirety. This process forms an emulsion, optionally stabilized by an incorporated surfactant, typically comprising submicron droplets of water immiscible blowing agent dispersed in an aqueous continuous phase. The blowing agent may be a fluorinated compound (e.g. perfluorohexane, perfluorooctyl bromide, perfluorooctyl ethane, perfluorodecalin, perfluorobutyl ethane) which vaporizes during the spray-drying process, leaving behind generally hollow, porous aerodynamically light particles. Other suitable liquid blowing agents include non-fluorinated oils, chloroform, Freon® fluorocarbons, ethyl acetate, alcohols, hydrocarbons, nitrogen, and carbon dioxide gases. The blowing agent may be emulsified with a phospholipid.

[00157] Although the pharmaceutical compositions may be formed using a blowing agent as described above, it will be appreciated that, in some instances, no additional blowing agent is required and an aqueous dispersion of the antiviral and/or pharmaceutically acceptable excipients and surfactant(s) are spray dried directly. In such cases, the pharmaceutical composition may possess certain physicochemical properties (e.g., elevated melting temperature, surface activity, etc.) that make it particularly suitable for use in such techniques.

[00158] As needed, cosurfactants such as poloxamer 188 or span 80 may be dispersed into this annex solution. Additionally, pharmaceutically acceptable excipients such as sugars and starches can also be added.

[00159] The feedstock(s) may then be fed into a spray dryer. Typically, the feedstock is sprayed into a current of warm filtered air that evaporates the solvent and conveys the dried product to a collector. The spent air is then exhausted with the solvent. Commercial spray dryers manufactured by Bϋchi Ltd. or Niro Corp. may be modified for use to produce the pharmaceutical composition. Examples of spray drying methods and systems suitable for making the dry powders of one or more embodiments of the present invention are disclosed in U.S. Patent Nos. 6,077,543; 6,051 ,256; 6,001 ,336; 5,985,248; and 5,976,574, which are incorporated herein by reference in their entireties.

[00160] Operating conditions of the spray dryer such as inlet and outlet temperature, feed rate, atomization pressure, flow rate of the drying air, and nozzle configuration can be adjusted in order to produce the required particle size, and production yield of the resulting dry particles. The selection of appropriate apparatus and processing conditions are within the purview of a skilled artisan in view of the teachings herein and may be accomplished without undue experimentation. Exemplary settings are as follows: an air inlet temperature between about 60°C and about 170 0 C, such as between 80 0 C and 120 0 C; an air outlet between about 40 0 C to about 120 0 C, such as about 50 0 C and 70°C; a

feed rate between about 3 mL/min to about 15 mL/min; an aspiration air flow of about 300 L/min; and an atomization air flow rate between about 25 L/min and about 50 L/min. The solids content in the spray-drying feedstock will typically be in the range from 0.5 wt% to 10 wt%, such as 1.0 wt% to 5.0 wt%. The settings will, of course, vary depending on the type of equipment used. In any event, the use of these and similar methods allow formation of aerodynamically light particles with diameters appropriate for aerosol deposition into the lung.

[00161] Hollow and/or porous microstructures may be formed by spray drying, as disclosed in WO 99/16419, which is incorporated herein by reference. The spray-drying process can result in the formation of a pharmaceutical composition comprising particles having a relatively thin porous wall defining a large internal void. The spray-drying process is also often advantageous over other processes in that the particles formed are less likely to rupture during processing or during deagglomeration.

[00162] Pharmaceutical compositions useful in one or more embodiments of the present invention may additionally or alternatively formed by lyophilization. Lyophilization is a freeze-drying process in which water is sublimed from the composition after it is frozen. The lyophilization process is often used because biologicals and pharmaceuticals that are relatively unstable in an aqueous solution may be dried without exposure to elevated temperatures, and then stored in a dry state where there are fewer stability problems. With respect to one or more embodiments of the instant invention, such techniques are particularly compatible with the incorporation of peptides, proteins, genetic material and other natural and synthetic macromolecules in pharmaceutical compositions without compromising physiological activity. Lyophilized cake containing a fine foam-like structure can be micronized using techniques known in the art to provide particles of the desired size.

[00163] In some embodiments, a formulation comprising two or more antiviral actives may be produced by preparing individual feedstocks comprising a single antiviral active in an appropriate liquid. Each feedstock is then subjected to a liquid removal process with characteristics selected to yield particles of appropriate physical characteristics. For example, a feedstock may be spray-dried, supertically processed or other liquid removal process. The resulting particles may then be dry blended to produce a powder having a combination of two or more antiviral actives. In some embodiments, the liquid removal step and powder blending steps may occur simultaneously, for example, by spray drying two or more feedstocks with a multiple nozzle spray drier, leading to a common powder collector.

[00164] The compositions of one or more embodiments of the present invention may be administered by known techniques, such as inhalation, oral, intramuscular, intravenous, intratracheal, intraperitoneal, subcutaneous, and transdermal. [00165] For example, the pharmaceutical compositions of one or more embodiments of the invention are effective in the treatment, including adjunctive treatment, of viral diseases or conditions.

[00166] In one or more versions, the compositions, when inhaled, penetrate into the nasal cavities and/or airways of the lungs to achieve effective antiviral concentrations.

[00167] In one or more versions, the compositions, either powder or liquid, may include a bronchodilator, and/or other adjuncts intended to improve drug or delivery effectiveness, patient compliance or safety.

{00168] In one or more embodiments of the invention, a pharmaceutical composition comprising antiviral is administered to the lungs of a patient in need thereof. For example, the patient may have been diagnosed with a viral infection or the patient may be determined to be susceptible to a viral infection.

[00169] Thus, the pharmaceutical compositions of one or more embodiments of the present invention can be used to treat and/or provide prophylaxis for a broad range of patients. A suitable patient for receiving treatment and/or prophylaxis as described herein is any mammalian patient in need thereof, preferably such mammal is a human. Examples of patients include, but are not limited to, pediatric patients, adult patients, and geriatric patients.

[00170] In some versions, an aerosolizeable pharmaceutical composition comprising antiviral is administered to the lungs and/or nasal cavity of a patient in a manner that results in an effective antiviral concentration.

[00171] In some versions, the pharmaceutical composition comprising antiviral is administered so that a target concentration is maintained over a desired period of time. For example, it has been determined that an administration routine that maintains a target concentration of antiviral is effective in treating and/or providing prophylaxis. In some embodiments, the antiviral concentration is maintained at the target lung concentration for a period of at least about 1 week, or about 2 weeks, or about 3 weeks.

[00172] The dosage necessary and the frequency of dosing for maintaining the antiviral concentration within the target concentration depends on the composition and concentration of the antiviral within the composition. In each of the administration regimens, the dosages and frequencies are determined to give a lung antiviral

concentration that is maintained within a certain target range. In one or more versions, the antiviral may be administered daily. In such versions, the daily dosage of antiviral may range from about 2 mg to about 75 mg, such as about 3 mg to about 50 mg, about 4 mg to about 25 mg, about 5 mg to about 20 mg, and about 7 mg to about 10 mg.

[00173] In some versions, a unit dosage of antiviral may comprise from about 20 mg to about 60 mg, such as about 30 mg to about 40 mg. In one or more versions, a pharmaceutical formulation comprises at least 10 or 20 or 30 or 40 or 45 or 50 or 55 or 60 mg of antiviral. In one or more versions, a human dose of the pharmaceutical formulation comprises at least about 0.1 or 0.2 or 0.3 or 0.4 or 0.5 or 0.6 or 0.7 or 0.8 or 0.9 or 1.0 or 1.5 or 2.0 mg/Kg of antiviral.

[00174] The drug loading in the small porous particles of the present invention depends upon factors including: (a) the volume of the unit dose (blister or capsule); (b) the lung delivery efficiency achieved with the device; (c) factors related to the mechanism of device emptying. In one or more versions, pulmonary delivery efficiency for the powder formulations of the present invention with portable, passive dry powder inhalers will be 40%-80%. In such versions, this would suggest a powder loading of 125 to 250% of the target lung dose. In some embodiments, optimal performance of capsule-based devices (e.g., the inhaler shown in Figs. 1A-1 E) depends at least in part, upon having sufficient mass in the capsule to facilitate proper capsule spinning and emptying characteristics. [00175] In one or more embodiments, a drug loading (i.e. active drug as a percentage of powder composition) can be from 1% to 100%, such as 5% to 90%, or any intermediate amount.

[00176] In some embodiments, a higher drug loading can be achieved with a blister- based inhaler. An example of such an inhaler is disclosed in international application published as WO2008/051621 , the disclosure of which is fully incorporated herein by reference. This device comprises a smaller volume for loading powder. In this case, the loading will typically range from 5 wt% to 90 wt%, such as 10 wt% to 80 wt%.

[00177] In one or more embodiments, a drug loading will provide for delivery of between about 10 mg to 100 mg, such as between about 32 mg to 75 mg, of active in a single puff (i.e. a single inhalation) from a dry powder inhaler. The reduction in administration time is anticipated to improve patient compliance.

[00178] The dose may be administered during a single inhalation or may be administered during several inhalations. The fluctuations of lung antiviral concentration can be reduced by administering the pharmaceutical composition more often or may be

increased by administering the pharmaceutical composition less often. Therefore, the pharmaceutical composition of one or more embodiments of the present invention may be administered from about three times daily to about once every two days.

[00179] The amount per dose of antiviral is be an amount that is effective, especially therapeutically-effective. In one or more embodiments, a dose ranges from about 0.01 mg/kg to about 5.0 mg/kg, such as about 0.4 mg/kg to about 4.0 mg/kg, or about 0.7 mg/kg to about 3.0 mg/kg.

[00180] Thus, in one or more versions, the pharmaceutical composition may be delivered to the lungs of a patient in the form of a dry powder. Accordingly, the pharmaceutical composition comprises a dry powder that may be effectively delivered to the deep lungs or to another target site. This pharmaceutical composition is in the form of a dry powder comprising particles or particles having a size selected to permit penetration into the alveoli of the lungs.

[00181] In some instances, it is desirable to deliver a unit dose, such as powder doses of 5 mg or 10 mg or greater of antiviral to the lung in a single inhalation. The above described dry powder particles allow for powder doses of about 5 mg or greater, in some embodiments greater than about 10 mg, and in some embodiments greater than about 25 mg, to be delivered in a single inhalation and in an advantageous manner. Alternatively, a dosage may be delivered over two or more inhalations. For example, a 10 mg powder dosage may be delivered by providing two unit doses of 5 mg each, and the two unit doses may be separately inhaled.

[00182] The dispersions or powder pharmaceutical compositions may be administered using an aerosolization device. The aerosolization device may be a nebulizer, a metered dose inhaler, a liquid dose instillation device, or a dry powder inhaler. The powder pharmaceutical composition may be delivered by a nebulizer as described in WO 99/16420, by a metered dose inhaler as described in WO 99/16422, by a liquid dose instillation apparatus as described in WO 99/16421 , and by a dry powder inhaler as described in U.S. Patent Application No. 09/888,311 filed on June 22, 2001, in WO 99/16419, in WO 02/83220, in U.S. Patent No. 6,546,929, and in U.S. Patent Application No. 10/616,448, filed on July 8, 2003, which are incorporated herein by reference in their entireties. As such, an inhaler may comprise a canister containing the particles or particles and propellant, and wherein the inhaler comprises a metering valve in communication with an interior of the canister. The propellant may be a hydrofluoroalkane.

[00183] Suitable passive dry powder inhalers include both capsule-based inhalers and blister-based inhalers. Suitable capsule-based inhalers include: devices by Nektar Therapeutics disclosed in U.S. Application Nos. 10/298,177; 10/295,783; 10/821 ,652; 10/821 ,624; 10/822,850; 10/704,160; 10/714,511 ; and 10/313,419, which are incorporated herein by reference. Devices sold or marketed under the following tradenames an/or trademarks may also be suitable: Handihaler (Boehringer Ingelheim), Eclipse (Aventis), AIR inhaler (Alkermes), Cyclohaler (Plastiape), Flowcaps (Hovione), Turbospin (PH&T), Monohaler (Pfizer), Spinhaler (Aventis), Rotahaler (GSK). Suitable blister-based inhalers include: the Diskus (GSK), the device of Nektar Therapeutics disclosed in WO 2008/051621, which is incorporated herein by reference, Gyrohaler (Vectura), E-Flex, Microdrug, Diskhaler (GSK). Also contemplated are active dry powder inhalers including: the inhalation device described in U.S. Patent No. 6,257,233, Aspirair (Vectura), and Microdose inhaler (Microdose).

[00184] The pharmaceutical composition of one or more embodiments of the present invention typically has improved emitted dose efficiency. Accordingly, high doses of the pharmaceutical composition may be delivered using a variety of aerosol ization devices and techniques.

[00185] The emitted dose (ED) of these powders may be greater than about 30%, such as greater than about 40%, or 45%, or 50%, or 55% or 60%, or 65% or 70% or 75% or 80% or 85% or 90% or 95%.

[00186] An example of a dry powder aerosol ization apparatus particularly useful in aerosolizing a pharmaceutical composition 100 according to one or more embodiments of the present invention is shown schematically in Fig. 1A. The aerosolization apparatus 200 comprises a housing 205 defining a chamber 210 having one or more air inlets 215 and one or more air outlets 220. The chamber 210 is sized to receive a capsule 225 which contains an aerosolizable pharmaceutical composition comprising antiviral. A puncturing mechanism 230 comprises a puncture member 235 that is moveable within the chamber 210. Near or adjacent the outlet 220 is an end section 240 that may be sized and shaped to be received in a user's mouth or nose so that the user may inhale through an opening 245 in the end section 240 that is in communication with the outlet 220.

[00187] The dry powder aerosolization apparatus 200 utilizes air flowing through the chamber 210 to aerosolize the pharmaceutical composition in the capsule 225. For example, Figs. 1A-1E illustrate the operation of a version of an aerosolization apparatus 200 where air flowing through the inlet 215 is used to aerosolize the pharmaceutical composition and the aerosolized pharmaceutical composition flows through the outlet 220

so that it may be delivered to the user through the opening 245 in the end section 240. The dry powder aerosolization apparatus 200 is shown in its initial condition in Fig. 1A. The capsule 225 is positioned within the chamber 210 and the pharmaceutical composition is contained within the capsule 225.

[00188] To use the aerosolization apparatus 200, the pharmaceutical composition in the capsule 225 is exposed to allow it to be aerosolized. In the version of Figs. 1 A-1 E, the puncture mechanism 230 is advanced within the chamber 210 by applying a force 250 to the puncture mechanism 230. For example, a user may press against a surface 255 of the puncturing mechanism 230 to cause the puncturing mechanism 230 to slide within the housing 205 so that the puncture member 235 contacts the capsule 225 in the chamber 210, as shown in Fig. 1B. By continuing to apply the force 250, the puncture member 235 is advanced into and through the wall of the capsule 225, as shown in Fig, 1 C. The puncture member may comprise one or more sharpened tips 252 to facilitate the advancement through the wall of the capsule 225. The puncturing mechanism 230 is then retracted to the position shown in Fig. 1 D, leaving an opening 260 through the wall of the capsule 225 to expose the pharmaceutical composition in the capsule 225.

[00189] Air or other gas then flows through an inlet 215, as shown by arrows 265 in Fig. 1E. The flow of air causes the pharmaceutical composition to be aerosolized. When the user inhales 270 through the end section 240 the aerosolized pharmaceutical composition is delivered to the user's respiratory tract. In one version, the air flow 265 may be caused by the user's inhalation 270. In another version, compressed air or other gas may be ejected into the inlet 215 to cause the aerosolizing air flow 265.

[00190] A specific version of a dry powder aerosolization apparatus 200 is described in U.S. Patent Nos. 4,069,819 and 4,995,385, which are incorporated herein by reference in their entireties. In such an arrangement, the chamber 210 comprises a longitudinal axis that lies generally in the inhalation direction, and the capsule 225 is insertable lengthwise into the chamber 210 so that the capsule's longitudinal axis may be parallel to the longitudinal axis of the chamber 210. The chamber 210 is sized to receive a capsule 225 containing a pharmaceutical composition in a manner which allows the capsule to move within the chamber 210. The inlets 215 comprise a plurality of tangentially oriented slots. When a user inhales through the endpiece, outside air is caused to flow through the tangential slots. This airflow creates a swirling airflow within the chamber 210. The swirling airflow causes the capsule 225 to contact a partition and then to move within the chamber 210 in a manner that causes the pharmaceutical composition to exit the capsule 225 and become entrained within the swirling airflow. This version is particularly effective

in consistently aerosolizing high doses of the pharmaceutical composition. In one version, the capsule 225 rotates within the chamber 210 in a manner where the longitudinal axis of the capsule is remains at an angle less than 80 degrees, and preferably less than 45 degrees from the longitudinal axis of the chamber. The movement of the capsule 225 in the chamber 210 may be caused by the width of the chamber 210 being less than the length of the capsule 225. In one specific version, the chamber 210 comprises a tapered section that terminates at an edge. During the flow of swirling air in the chamber 210, the forward end of the capsule 225 contacts and rests on the partition and a sidewall of the capsule 225 contacts the edge and slides and/or rotates along the edge. This motion of the capsule is particularly effective in forcing a large amount of the pharmaceutical composition through one or more openings 260 in the rear of the capsule 225.

[00191] In another passive dry powder inhaler version, the dry powder aerosolization apparatus 200 may be configured differently than as shown in Figs. 1A-1E. For example, the chamber 210 may be sized and shaped to receive the capsule 225 so that the capsule 225 is orthogonal to the inhalation direction, as described in U.S. Patent No. 3,991 ,761, which is incorporated herein by reference in its entirety. As also described in U.S. Patent 3,991 ,761 , the puncturing mechanism 230 may puncture both ends of the capsule 225. In another version, the chamber may receive the capsule 225 in a manner where air flows through the capsule 225 as described for example in U.S. Patent Nos. 4,338,931 and 5,619,985. In another version, the aerosolization of the pharmaceutical composition may be accomplished by pressurized gas flowing through the inlets, as described for example in U.S. Patent Nos. 5,458,135; 5,785,049; and 6,257,233, or propellant, as described in WO 00/72904 and U.S. Patent No. 4,114,615, which are incorporated herein by reference. These types of dry powder inhalers are generally referred to as active dry powder inhalers.

[00192] In one or more embodiments, a blister-based inhaler device can achieve a high drug loading loading. A specific example of such a device is that disclosed in the WO 2008/051621. This device typically operates with a smaller volume for loading powder. In one or more embodiments with such device, the loading will typically range from 5 wt% to 50 wt%, such as 4 wt% to 20 wt%. In one or more embodiments with such device, the loading will typically range from about 0.7 to 8 mg per blister, such as from about 4 to 6 mg per blister. In one or more embodiments, such drug loading and device will provide for delivery of up to about 4 mg (of antiviral active) in a single puff (or inhalation) from a dry powder inhaler.

[00193] The pharmaceutical composition disclosed herein may also be administered to the pulmonary and/or nasal air passages of a patient via aerosolization, such as with a metered dose inhaler. The use of such stabilized preparations provides for superior dose reproducibility and improved lung deposition as disclosed in WO 99/16422, which is incorporated herein by reference in its entirety. MDIs are well known in the art and could be employed for administration of the antiviral. Breath activated MDIs, as well as those comprising other types of improvements which have been, or will be, developed are also compatible with the pharmaceutical composition of one or more embodiments of the present invention.

[00194] Nebulizers are known in the art and can be employed for administration of the antiviral dosage forms herein by making a dispersion or aerosol thereof. Breath activated nebulizers, as well as those comprising other types of improvements which have been, or will be, developed are also compatible with dispersions, which are contemplated as being with in the scope of one or more embodiments of the present invention. Along with the aforementioned embodiments, the dispersions of one or more embodiments of the present invention may also be used in conjunction with nebulizers as disclosed in WO 99/16420, which is incorporated herein by reference in its entirety, in order to provide an aerosolized medicament that may be administered to the pulmonary and/or nasal air passages of a patient in need thereof.

[00195] Along with DPIs, MDIs and nebulizers, it will be appreciated that the stabilized dispersions, suspensions or solutions of one or more embodiments of the present invention may be used in conjunction with liquid dose instillation or LDI techniques as disclosed in, for example, WO 99/16421 , which is incorporated herein by reference in its entirety. Liquid dose instillation involves the direct administration of a stabilized dispersion to the lung. In this regard, direct pulmonary and/or nasal administration of bioactive compounds is particularly effective in the treatment of disorders especially where poor vascular circulation of diseased portions of a lung reduces the effectiveness of intravenous drug delivery. With respect to LDI the stabilized dispersions are preferably used in conjunction with partial liquid ventilation or total liquid ventilation. Moreover, one or more embodiments of the present invention may further comprise introducing a therapeutically beneficial amount of a physiologically acceptable gas (such as nitric oxide or oxygen) into the pharmaceutical microdispersion prior to, during or following administration.

[00196] The time for dosing is typically short. For a single capsule (e.g., 5 mg powder dose), the total dosing time is normally less than about 1 minute. A two capsule

close (e.g., 10 mg powder) usually takes about 1 min. A five capsule dose (e.g., 25 mg powder) may take about 3.5 min to administer. Thus, the time for dosing may be less than about 5 min, such as less than about 4 min, less than about 3 min, less than about 2 min, or less than about 1 min.

100197] Alternatively or additionally, the pharmaceutical composition may comprise a liquid form and may be aerosolized using an aerosol generator nebulizer. Examples of nebulizers include, but are not limited to, the Aeroneb®Go or Aeroneb®Pro nebulizers, available from Stamford Ltd, of Galway, Ireland.

[00198] The nebulizer (i.e., aerosol generator) thus may be of the type, for example, where a vibratable member is vibrated at ultrasonic frequencies to produce liquid droplets. In one or more embodiments, the ultrasonic frequency of vibration comprises at least about 45 kHz. Some specific, non-limiting examples of technologies for producing fine liquid droplets is by supplying liquid to an aperture plate having a plurality of tapered apertures and vibrating the aperture plate to eject liquid droplets through the apertures. Such techniques are described generally in U.S. Patent Nos. 5,164,740; 5,938,117; 5,586,550; 5,758,637, 6,014,970, and 6,085,740, the complete disclosures of which are incorporated by reference. In one or more embodiments of the present invention, the aerosol generator comprises a vibrating mesh type, wherein vibrational energy is supplied via a piezoelectric element in communication (directly or indirectly) with the mesh element.

[00199] The aerosolization element may be constructed of a variety of materials, comprising metals, which may be electroformed to create apertures as the element is formed, as described, for example, in U.S. patent No. 6,235,177 assigned to the present assignee and incorporated by reference herein in its entirety. Palladium is believed to be of particular usefulness in producing an electroformed, multi-apertured aerosolization element, as well as in operation thereof to aerosolize liquids. Other metals that can be used are palladium alloys, such as PdNi, with, for example, 80 percent palladium and 20% nickel. Other metals and materials may be used without departing from the present invention.

[00200] In one or more embodiments, the aerosol generator comprises a tube core design, as described in WO 2006/127181, assigned to the same assignee as the invention herein, and incorporated by reference herein in its entirety for all purposes.

[00201] The foregoing description will be more fully understood with reference to the following Examples. Such Examples, are, however, merely representative of methods of

practicing one or more embodiments of the present invention and should not be read as limiting the scope of the invention.

EXAMPLES

Example I - Preparation of Neυraminidase/M2 Channel Blocker Powder by Spray- Drying

[00202] The following materials were obtained: zanamivir from LGM Pharmaceuticels, Batch# 071201 ; rimantadine from Aldrich; trileucine from BaChem (Lot# 7848-1); ethanol from Sigma 99.5% (Batch* 003595E); and water: (Fisher)

[00203] Seven feedstock formulations of rimantadine/zanamivir (R/Z) powders were prepared at solids percentages of trileucine, and ethanol, and at different R:Z ratios These formulations are shown in Table 1. Ethanol was added to decrease the solubility of the trileucine and to cause early shell formation. Four of the feedstocks were stable over the course of the experiment; 5976-56, -59, -61 and -67. The other three precipitated. Specifically, trileucine was very sensitive to the presence of ethanol. At 2.5% ethanol, and 1.5% solids, 30% trileucine could be added, while at 1.75% solids only 20% trileucine could be added to the feedstock. The feedstock was stable at 4% solids even without trileucine.

[00204] Feed stock preparation: All feedstock component ratios, including liquids were based upon mass. Trileucine was dissolved in the specified mass of water/ethanol. Zanamivir and rimantadine were added and the solution was adjusted to pH 6.5 - 6.6. The compositions of all feed stocks are shown in Table 1 below.

Table 1

[00205] Spray Drying: Powders were spray dried on a Buchi Spray dryer using a standard Buchi nozzle, and nitrogen as the atomization and drying gas. Feed stock feed rate was 5.0 ml/min. Atomization pressures were 20 and 40 psi and outlet temperature was 7O 0 C.

[00206] Yields of the spray-dried feed stocks were 67% to 71 %. The powders containing trileucine had similar "dimpled" or "raisin-like" morphologies, as shown in Figs 2B-2C. Specifically, Fig 2A shows formulation 5976- 56, which lacks trileucine. Figs 2B, 2C and 2D show the -59, -61 and -67 formulations, respectively. The powder lacking trileucine contained more spherical particles, which posses less favorable aerodynamic properties. A graph of particle size distributions for Sample 5976-67 {25% trileucine, 9% zanamivir and 66% rimantadine) is shown in Fig 3.

[00207] Table 2 shows median particles sizes (in microns) for Sample No. 5976-67. In the Table, particles sizes are given as below the 10, 16, 50, 84 and 90% distribution ranges.

Table 2: Median particles sizes (in microns) for Sample No. 5976-67

Example Il - Aerosol Properties

[00208] The powders of Example I above were hand filled into capsules and aerosol tested. Capsule emptying volume was determined using a photo-detector system. Aerosol results are given in Table 3, ED representing the emitted dose. Table 3 also shows resulting particle sizes (MMAD = mass median aerodynamic diameter), and aerodynamic particle size distributions (aPSD), for four different formulations: Nos. 5976- 61 , 5976-59; 5976-56 and 5976-67. It can be seen that the formulations including the trileucine excipient provided emitted doses of greater than 80% as well as Fine Particle Doses (FPD) < 3 . 3 microns of at least about 40%.

Table 3:

Conditions Me.in ED oPSO

I Tiiiget ED Capsule Rimantadine Zamanttivir ftimantadiii Zaiiamavit ACI Noun. • Fill SO Retention Retention MMAD FPD<2.3μm FPD^.Sμm e FPD^ s^ FPD< 3 3lm Recoveiy Recovery Lot * [mq| ED [%| P 1 I N SD [>:,] fμni] |ιiiq] [ι«fll [(HCJl IM PlI

5976-61 . _^ 50_ UL- 6_ _ _ 5 _ 1.9 1331 4.44 19.22 6.41 72 86 20% TriLeu 20 13.18 " 4.33 " 1922 6.41 72 " 87

2.0 13.73 4.58 19.79 6.B0 77 92 average 2.0 13.41 447 19.41 6.47 74 89 set θ.0 0.29 0.10 0.33 0.11 3 3

5976-59 . 50 93 2 1 0 2.0 13.76 3.75 19.16 5.23 85 92

30% TriLeu ; 2.0 13.78 3.76 19.31 5.27

I se 93

2.1 13.27 3.62 18.96 r 5.17 ' " 92 average 2.0 13.60 3.71 19.15 5.22 85 92

Sd 0.0 0.29 0.08 0.17 0.05 0 1

1 5976-56 I 20 43 13 21 12 2.7 1.81 ; 0.60 3.23 6.60 33 77 ■ " NeaT

5976-67 _\_ 50 94 2 2 1 2.3 1400 1.91 21.99 1 3.00 86 92

25%TriLeu i 2.3 13 90 1.90 21.86 2.98 94 average 2,3 13.95 1.90 21.93 2.99 87 * 93 sd 0.0 0.07 0.01 0.09 0.01 1 1

[00209] Trileucine appears to result in good aerosol performance. The emitted dose for Sample No. 5976-59 (30% trileucine) and No. 5976-67 (25% trileucine) were above 90% with %SD of 2% and capsule retentions of 1% and 2% respectively. At 20% trileucine (Sample 5976-61) the ED was still 83% with %SD of 6 and capsule retention of 6%. The formulation without trileucine (Sample 5976-56) the ED was only 43%, %SD of 13% and capsule retention of 21%. These results are summarized also in Figure 4. In the Figure, "ED" = Emitted Dose, and "CR" = Capsule Retained.

[00210] All three powders with trileucine performed nearly equally well achieving drug delivery targets. Lot Nos. 5976-59, 5976-61 and 5976-67 delivered 17.3 mg (13.6 mg Rimantadine + 3.71 mg Zamamavir), 17.9 mg (13.41 mg Rimantadine + 4.47 mg Zamamavir),, and 15.9 mg (13.95 mg Rimantadine + 1.9 mg Zamamavir) of powder smaller thant 2.3 microns, respectively. In this assay, No. 5976-61 was slightly better than the other powders possibly because the aerosol particle size was small and the drug loading was higher than the other powders. These results are summarized also in Figure 6. Fig 6 shows estimated delivered doses of rimantidine and zanamivir for the three formulations (5976-59, -61 and -67) containing trileucine, wherein FDP stands for Fine Particle Doses.

[00211] The foregoing demonstrates that powders can be prepared in commercially reasonable yield. Where about 25% or more di- or tri-peptide containing at least two leucines was added to the formulation, aerosol performance achieved drug delivery

targets. Trileucine appeared to improve powder flow properties within the capsule, increasing the emitted dose and decreased ED variability.

Example III - Aerosol pulse measurements

[00212] Figs 5A-5B show aerosol pulse measurement of two powders, wherein Fig 5 A refers to Sample No. 5976-67, exiting an inhaler device mouthpiece, demonstrating times required to complete emptying formulations of the present invention. It can be seen that peak flow rates reach about 60L/min, thus a 2L volume is emptied in less than about 2 to 2.5 seconds.

Example IV - Preparation of a M2 Blocker Powder

[00213] The following materials were obtained: rimantadine from Aldrich; trileucine from BaChem (Lot# 7848-1 ); ethanol from Sigma 99.5% {Batch# 003595E); and water: (Fisher).

[00214] An aqueous feedstock formulation of rimantadine was prepared by dissolving 80% rimandadine and 20% trileucine in a sufficient amount of a 95% water, 5% ethanol solution to yield a 2% solids level. All feedstock component ratios, including liquids, were based upon mass. The solution was adjusted to pH 6.5. Ethanol was used to decrease the solubility of the trileucine and to cause early shell formation.

[00215] The feedstock was spray dried on a Buchi Spray dryer using a standard Buchi nozzle, and nitrogen as the atomization and drying gas. Feed stock feed rate was 5.0 ml/min, Atomization pressures were 20 and 40psi and outlet temperature was 6O 0 C or 72 0 C. The feedstock formulation was divided into two lots: one was spray dried and at 60 0 C (lot 11/60) and the other at 72°C (lot 11/72) outlet temperatures.

[00216] The spray-dried powders were transferred into a glovebox with a relative humidity less than 5% and placed into unit dosage forms (capsules or blisters) suitable for use in a dry powder inhaler device as described herein, for example, as described in U.S. Patent No. 4995305.

[00217] Aerosol Results are shown in Table 4 below, with ED = emitted dose, MMAD - mass median aerodynamic diameter, aPSD - aerodynamic particle size distributions, FDP = fine particle doses.

Table 4

Example V - Preparation of Neuraminidase Inhibitor Powder

[00218] The following materials are obtained: zanamivir from LGM Pharmaceuticels, Batch# 071201 ; trϋeucine from BaChem (Lot# 7848-1 ); ethanol from Sigma 99.5% (Batch# 003595E); and water: (Fisher).

[00219] An aqueous feedstock formulation of zanamivir is prepared by dissolving 80% zanamivir and 20% trileucine in a sufficient amount of a 95% water, 5% ethanol solution to yield a 2% solids level. The solution is adjusted to pH 6.5. Ethanol is used to decrease the solubility of the trileucine and to cause early shell formation. All feedstock component ratios, including liquids, are based upon mass.

[00220] The feedstock is spray dried on a Buchi Spray dryer using a standard Buchi nozzle, and nitrogen as the atomization and drying gas. Feed stock feed rate is 5.0 ml/min, Atomization pressures are 20 and 40 psi and outlet temperature is 7O 0 C.

[00221] The powders are transferred into a glovebox with a relative humidity less than 5% and placed into unit dosage forms (capsules or blisters) suitable for use in a dry powder inhaler device as described herein, for example, as described in U.S. Patent No. 4995305.

[00222] Although the present invention has been described in considerable detail with regard to certain preferred versions thereof, other versions are possible, and alterations, permutations and equivalents of the version shown will become apparent to those skilled in the art upon a reading of the specification and study of the drawings. Also, the various features of the versions herein can be combined in various ways to provide additional versions of the present invention. Furthermore, certain terminology has been

used for the purposes of descriptive clarity, and not to limit the present invention. Therefore, any appended claims should not be limited to the description of the preferred versions contained herein and should include all such alterations, permutations, and equivalents as fall within the true spirit and scope of the present invention.