Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
IL-1 RECEPTOR ANTAGONIST TREATMENT FOR NEUTROPHILIC LUNG DISEASE
Document Type and Number:
WIPO Patent Application WO/2024/077042
Kind Code:
A2
Abstract:
A method for treating a neutrophilic lung disease in a human subject in need thereof is disclosed, including administering a pharmaceutical composition including an effective amount of an IL-IRa (e.g., rhIL-1Ra) to the human subject, wherein the neutrophilic lung disease is selected from the group consisting of chronic obstructive pulmonary disease, steroid-resistant chronic obstructive pulmonary disease, bronchiectasis, neutrophilic asthma, acute respiratory distress syndrome, chemical lung injury, and rheumatoid arthritis-associated interstitial lung disease, and other neutrophilic lung disease.

Inventors:
WRING STEPHEN (US)
PALACIOS MICHELLE (US)
MCKERNAN COURTNEY (US)
CRIZER KATELYN (US)
Application Number:
PCT/US2023/075912
Publication Date:
April 11, 2024
Filing Date:
October 04, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ONSPIRA THERAPEUTICS INC (US)
International Classes:
A61K41/00
Attorney, Agent or Firm:
GENG, Deric, X. et al. (US)
Download PDF:
Claims:
What is claimed is:

1. A method for treating a neutrophilic lung disease in a human subject in need thereof, comprising administering a pharmaceutical composition comprising an effective amount of an interleukin 1 receptor antagonist (IL-IRa) to the human subject, wherein the neutrophilic lung disease is selected from the group consisting of chronic obstructive pulmonary disease, steroid-resistant chronic obstructive pulmonary disease, bronchiectasis, neutrophilic asthma, acute respiratory distress syndrome, chemical lung injury, and rheumatoid arthritis-associated interstitial lung disease.

2. The method of claim 1, wherein the IL-IRa is administered to the airways of the human subject.

3. The method of claim 2, wherein the IL-IRa is administered by inhalation.

4. The method of claim 2, wherein the IL-IRa is administered intranasally.

5. The method of claim 1, wherein the IL-IRa is delivered to the distal regions of the human subject’s lung.

6. The method of claim 1, wherein the IL-IRa is administered intratracheally.

7. The method of claim 1, wherein the IL-IRa is nebulized.

8. The method of claim 7, wherein the nebulized IL-IRa has a mass median aerodynamic diameter (MMAD) of about 1 pm to about 15 pm.

9. The method of claim 8, wherein the nebulized IL-IRa has a MMAD of about 3 pm.

10. The method of any one of claims 1 to 9, wherein the pharmaceutical composition comprises ALTA-2530.

11. The method of any one of claims 1 to 10, wherein the pharmaceutical composition comprises anakinra.

12. The method of claim 1, wherein the method reduces secretion of a chemokine(s) that promotes neutrophil recruitment.

13. The method of claim 10, wherein the chemokine is selected from the group consisting of CXCL1, CXCL2, CCL2, CCL4, and IL-8.

14. The method of claim 1, wherein the method reduces neutrophilic airway inflammation.

15. The method of claim 1, wherein the method reduces airway neutrophils.

16. The method of claim 15, wherein the method reduces airway neutrophils without eradicating at least one neutrophil.

17. The method of claim 1, wherein the method prevents or reduces alveolar edema and/or neutrophil trafficking.

18. The method of claim 1, wherein the IL-IRa in the pharmaceutical composition binds to an IL-1 receptor with an affinity higher than IL-ip.

19. The method of claim 1, wherein the IL-IRa in the pharmaceutical composition binds to an IL-1 receptor with an affinity higher than IL- la.

20. The method of any of claims 1 to 19, wherein the pharmaceutical composition further comprises a second therapeutic agent.

21. The method of claim 20, wherein the second therapeutic agent is selected from the group consisting of cyclosporine, steroids, antibiotics, and a combination thereof.

Description:
IL-1 RECEPTOR ANTAGONIST TREATMENT FOR NEUTROPHILIC LUNG DISEASE

[0001] This application claims the benefit of and priority under 35 U.S.C. § 1 19(e) to U.S. Serial No. 63/413,502 filed October 5, 2022, the contents of which is hereby incorporated by reference in its entirety.

[0002] This patent disclosure contains material that is subject to copyright protection.

The copyright owner has no objection to the facsimile reproduction of the patent document or the patent disclosure as it appears in the U.S. Patent and Trademark Office patent file or records, but otherwise reserves any and all copyright rights.

INCORPORATION BY REFERENCE

[0003] All documents cited herein are incorporated herein by reference in their entirety.

FIELD OF THE INVENTION

[0004] The invention relates generally to the field of pharmaceutical science. More particularly, the invention relates to compounds and compositions useful as pharmaceuticals for treating various lower airways disorders.

BACKGROUND

[0005] Lung neutrophilia is a common pathological feature of several diseases, including, for example, chronic obstructive pulmonary disease (COPD) (Peter J. Barnes, Immunology of asthma and chronic obstructive pulmonary disease, Nat. Rev. Immunol.

8(3): 183-92 (2008)), bronchiectasis (Pallavi Bedi et al., Blood Neutrophils Are Reprogrammed in Bronchiectasis, Am. J. Respir. Crit. Care Med. 198(7):880-90 (2018); Derek W. Russell et al., Neutrophil Fates in Bronchiectasis and Alpha-1 Antitrypsin Deficiency, Ann. Am. Thorac. Soc. 13 Suppl. 2, pp. S123-29 (2016)), neutrophilic asthma (F. Wang et al., Different inflammatory phenotypes in adults and children with acute asthma, Eur. Respir. J. 38(3):567-74 (2011)), rheumatoid arthritis-associated interstitial lung disease (RA-ILD) (A. Saku et al., Prognostic significance of peripheral blood monocyte and neutrophil counts in rheumatoid arthritis-associated interstitial lung disease, Respir Med. 182: 106420 (2021)), acute respiratory distress syndrome (ARDS) (K.P. Steinberg et al., Evolution of bronchoalveolar cell populations in the adult respiratory distress syndrome, Am. J. Respir. Crit. Care Med. 150(1): 113-22 (1994); Shun-Chin Yang et al., Understanding the role of neutrophils in acute respiratory distress syndrome, Biomed. J. 44(4):439-446 (2021)), and chemical lung injury (Javad Beheshti et al., Mustard lung secrets: long term clinicopathological study following mustard gas exposure, Pathol. Res. Pract. 202(10):739-44 (2006)).

[0006] Infiltrating neutrophils typically mediate the innate immune response and destroy invading pathogens in the lung through phagocytosis and degranulation. However, dysregulation of neutrophil activation and recruitment leads to tissue damage and inflammatory diseases (Oliver Soehnlein et al., Neutrophil secretion products pave the way for inflammatory monocytes, Blood 112(4): 1461-71 (2008); E. Abraham et al., Neutrophils as early immunologic effectors in hemorrhage- or endotoxemia-induced acute lung injury, Am. J. Physol. Lung Cell Mol. Physiol. 279:L1137-45 (2000); Shun-Chin Yang et al., Understanding the role of neutrophils in acute respiratory distress syndrome, Biomed. J. 44(4):439-446 (2021)). While steroids can be used to reduce lung inflammation in neutrophilic lung diseases, many patients are resistant to steroid treatment. For example, 10- 25% of asthmatic patients and up to 90% of COPD patients are resistant to steroid treatment (Stanly J. Szefler et al., Significant variability in response to inhaled corticosteroids for persistent asthma, J. Allergy Clin. Immunol., 109(3):410-l 8 (2002); Donald Y.M. Leung & John W. Bloom, Update on glucocorticoid action and resistance, J. Allergy Clin. Immunol., 111 (1): 3-22 (2003); Mark T.S. Chan et al., Difficult-to-control asthma: Clinical characteristics of steroid-insensitive asthma J . Allergy Clin. Immunol., 101 (5):p594-601 (1998)). In view of this, the need for new treatment options to treat steroid resistant lung inflammation, e.g., lung inflammation associated with neutrophilic lung diseases, has been recognized in the field (Peter J. Barnes, Immunology of asthma and chronic obstructive pulmonary disease , Nat. Rev. Immunol. 8(3): 183-92 (2008)).

[0007] Neutrophils play an important role in facilitating steroid resistance, potentially by increased proinflammatory cytokine release due to reduced neutrophil apoptosis (T. Kato et al., Inhibition by dexamethasone of human neutrophil apoptosis in vitro, Nat. Immun.

14(4): 198-208 (1995); K. Ito et al., Steroid-resistant neutrophilic inflammation in a mouse model of an acute exacerbation of asthma, Am. J. Respir. Cell Mol. Biol. 39(5):543-50 (2008); X. Wang et al., Diversity of TH cytokine profiles in patients with chronic rhinosinusitis: a multicenter study in Europe, Asia, and Oceania, J Allergy Clin. Immunol. 138(5): 1344-53 (2016)). Thus, targeting neutrophil activation and migration may be an effective treatment for neutrophilic lung diseases. [0008] During inflammation, a delicate balance of neutrophils must be maintained. Neutrophils are necessary to help clear pathogens, but an imbalance of neutrophils can lead to tissue damage and inflammatory disease.

[0009] Interleukin 1 alpha (IL- la) and interleukin 1 beta (IL-ip) are cytokines that are part of the interleukin-1 (IL-1) family and bind to the interleukin-1 type 1 receptor (IL-1R1) to activate pro-inflammatory signaling pathways. IL- la and IL-ip can stimulate neutrophil recruitment indirectly by enhancing expression of neutrophil chemo-attractants (T.

Yoshimura et al., Purification of a human monocyte-derived neutrophil chemotactic factor that has peptide sequence similarity to other host defense cytokines, Proc. Nat’l Acad. Sci. USA 84(24): 9233 -37 (1987); Susan J. Burke et al., Transcription of the gene encoding TNF- a is increased by IL-lf in rat and human islets and f-cell lines, Mol. Immunol. 62(1): 54-62 (2014); Christian D. Sadik et al., Neutrophils cascading their way to inflammation, Trends Immunol. 32(10):452-60 (2011)). In preclinical models, intratracheal administration of IL-la or IL-ip enhances the percentage of infiltrating lung neutrophils (J. A. Leff et al., Interleukin- 1 -induced lung neutrophil accumulation and oxygen metabolite-mediated lung leak in rats, Am. J. Physiol. 266:L2-L8 (1994); Patricia Dubin et al., Interleukin-23-Mediated Inflammation in Pseudomonas aeruginosa Pulmonary Infection, Infect. Immun. 80(l):398- 409 (2012)). Intranasal administration of IL-ip to mice increased expression of the neutrophil chemotactic factor CXCL1 as well as the number of neutrophils in the bronchoalveolar lavage fluid (BALF) (Pamela Gasse et al., IL-lRl/MyD88 signaling and the inflammasome are essential in pulmonary inflammation and fibrosis in mice, J. clin. Invest. 117(12):3786-99 (2007)). These studies show that overexpression of IL-la or IL-ip is sufficient to drive lung neutrophilia. In asthma patients, both BALF IL-la protein levels and IL-1R1 mRNA expression correlate with neutrophilia (Sucia Liu et al., Steroid resistance of airway type 2 innate lymphoid cells from patients with severe asthma: The role of thymic stromal lymphopoietin, J. Allergy Clin. Immunol. 141(l):257-68 (2018); Michael D. Evans et al., Sputum cell IL-1 receptor expression level is a marker of airway neutrophilia and airflow obstruction in asthmatic patients, J. Allergy Clin. Immunol. 142(2):415-23 (2018)). These data further support a role for IL-1 signaling in lung neutrophil infiltration in humans.

[0010] Inhibiting the IL-1 signaling pathway has been shown to reduce neutrophilia in vivo. In cigarette-smoke induced preclinical models of COPD, IL-1R1 or IL-la deficient mice had an approximately 100% and 75% reduction in the number of neutrophils in the BALF, respectively (Fernando M. Botelho et al., IL-la/IL-lRl Expression in Chronic Obstructive Pulmonary Disease and Mechanistic Relevance to Smoke-Induced Neutrophilia in Mice, PLoS One 6(12): 1-13 (2011); N.S. Pauwels et al., Role of IL-la and the Nlrp3/caspase-l/IL-lf axis in cigarette smoke-induced pulmonary inflammation and COPD, Eur. Resp. J. 38: 1019-18 (2011)). Neutralization with an IL-la antibody decreased the number of BALF neutrophils by 53% in an inflammatory COPD mouse model (Hannes Bucher et al., Neutralization of both IL-la/ IL-1 plays a major role in suppressing combined cigarette smoke/virus-induced pulmonary inflammation in mice, Pulm. Pharm. Therap. 44:96- 105 (2017)). An IL-ip neutralizing antibody, but not the steroid dexamethasone, significantly reduced BALF neutrophils in a severe steroid resistant asthma model (Richard Y. Kim et al., Role for NLRP 3 inflammasome -mediated, IL- 1 ^-dependent responses in severe, steroid-resistant asthma, Am. J. Resp. Crit. Care Med. 196(3):283-97 (2017)).

However, targeting both IL-la and IL-ip (e.g., by blockading IL-1R1) may elicit the greatest benefit in neutrophilic airway diseases. While an approved IL-1R1 receptor antagonist exists, it is systemically administered and achieves negligible exposure in lung tissue (Dong-Chool Kim et al., Kidney as a Major Clearance Organ for Recombinant Human Interleukin- 1 Receptor Antagonist, J. Pharm. Sci. 84(5):575-80 (1995); C. Cawthorne et al., Biodistribution, pharmacokinetics and metabolism of interleukin- 1 receptor antagonist (IL- 1RA) using [ 18 F] -IL- IRA and PET imaging in rats, Br. J. Pharmacol. 162(3):659-72 (2011)). Moreover, approved monoclonal antibody therapies target either IL-la or IL-ip , but not both, proteins; thus, the need exists for therapies that target both IL-la and IL-ip proteins. Surprisingly, it has been found in the present disclosure that this could be achieved by blocking their binding to IL-1R1 using the IL-IRa compositions disclosed herein.

BRIEF DESCRIPTION OF THE DRAWINGS

[0011] FIGS. 1 A-B show Surface Plasmon Resonance sensorgrams comparing the binding of human IL-ip to IL-1R1 (FIG. 1A) to the binding of the rhIL-IRa protein in the ALTA-2530 formulation to IL-1R1 (FIG. IB).

[0012] FIGS. 2A-B show Surface Plasmon Resonance sensorgrams comparing the binding of human IL-la to IL-1R1 (FIG. 2A) to the binding of the rhIL-IRa protein in ALTA-2530 formulation to IL-1R1 (FIG. 2B).

[0013] FIGS. 3A-B show immunohistochemistry of ALTA-2530 in lung samples from cynomolgus monkeys that received once-daily exposure to saline (FIG. 3A) or ALTA-2530 (FIG. 3B) via nebulizer for seven days. [0014] FIG. 4 shows immunohistochemistry of ALTA-2530 in lung samples from rats that received once-daily exposure to saline (left), low dose ALTA-2530 (middle), or high dose ALTA-2530 (right) via nebulizer for seven days.

[0015] FIG. 5 shows results from a human whole blood multiplex assay assessing the efficacy of ALTA-2530 in treating IL-ip stimulated cytokine release. The assays assess the ability of ALTA-2530 to inhibit IL-ip stimulated cytokines CXCL1, CXCL2, IL-6, IL-8, CCL2, and CCL4.

[0016] FIG. 6 shows results from a study in cynomolgus monkeys to assess rhIL-IRa levels in BALF following exposure of the subject to ALTA-2530. The dotted line represents the amount of ALTA-2530 needed to induce maximal inhibition of IL-ip-stimulated IL-6 release in monkey whole blood.

[0017] FIGS. 7A-C show histological examination results from a study comparing a healthy rat lung sample (FIG. 7A) to a lung sample from a rat exposed to sulfur mustard and treated with ALTA-2530 (FIG. 7B) and a to lung sample from a rat exposed to sulfur mustard and treated with saline (FIG. 7C). Lung tissues treated with ALTA-2530 show no alveolar edema and few neutrophils, indicating the healing process following sulfur mustard is proceeding at a more rapid pace than that displayed following saline treatment. In the saline treated animals alveoli depict significant alveolar edema (yellow arrows, wispy eosinophilic strands) and acute neutrophilic inflammation (red arrows).

[0018] FIG. 8 shows the primary, secondary, and exploratory endpoints from an inhaled lipopolysaccharide challenge in non-human primates. Monkeys were treated with vehicle or low or high dose ALTA-2530. ALTA-2530 treatment led to a significant decrease in the percentage of bronchoalveolar lavage fluid (BALF) neutrophils and a reduction in total neutrophil counts in the BALF. ALTA-2530 reduced BALF IL-1 [3 and IL-6 levels. Serum IL-6 and IL-8 levels were significantly decreased in ALTA-2530 treated animals. BALF TNF-a was significantly reduced in ALTA-2530 treated animals.

[0019] FIGS. 9A-B show results from a study assessing the ability of low dose ALTA- 2530 (top) and high dose ALTA-2530 (bottom) to reduce neutrophil infiltration following exposure to lipopolysaccharide in the 4 hours (FIG. 9A) and 24 hours (FIG. 9B) after lipopolysaccharide administration. In FIGS. 9A-B, n.s. indicates not significant, one asterisk (*) indicates p<0.05, two asterisks (**) indicate p<0.01, three asterisks (***) indicate p<0.001, and four asterisks (****) indicate p<0.0001. The results show that treatment with low-dose ALTA-2530 reduced the percent neutrophil infiltration in the BALF by an average of 32.7% at 4 hours after lipopolysaccharide administration and treatment with high-dose ALTA-2530 decreased the percent neutrophil infiltration by an average of 36.2% at 24 hours after lipopolysaccharide administration. Each line represents an individual animal.

[0020] FIGS. 10A-B show results from a study assessing the ability of low dose ALTA- 2530 (top) and high dose ALTA-2530 (bottom) to reduce the total cell count in BALF following exposure to lipopolysaccharide in the 4 hours (FIG. 10 A) and 24 hours (FIG. 10B) after lipopolysaccharide administration. In FIGS. 10A-B, n.s. indicates not significant, and one asterisk (*) indicates p<0.05. The results show that exposure to lipopolysaccharide increased the total cell count by up to four times in the BALF, and that treatment with both low-dose and high-dose ALTA-2530 reduced the total cell count at 4 hours and at 24 hours after lipopolysaccharide administration. Each line represents an individual animal. Total cell number in BALF was normalized to baseline for each individual animal.

[0021] FIGS. 11A-B show results from a study assessing the ability of low dose ALTA- 2530 (top) and high dose ALTA-2530 (bottom) to reduce absolute neutrophil counts in BALF following exposure to lipopolysaccharide in the 4 hours (FIG. 11 A) and 24 hours (FIG. 1 IB) after lipopolysaccharide administration. In FIGS. 11 A-B, n.s. indicates not significant, one asterisk (*) indicates p<0.05, and two asterisks (**) indicate p<0.01. The results show that treatment with both low-dose and high-dose ALTA-2530 reduced the absolute neutrophil counts in the BALF at 4 hours and at 24 hours after lipopolysaccharide administration. Each line represents an individual animal.

[0022] FIGS. 12A-B show results from a study assessing the ability of low dose ALTA- 2530 (top) and high dose ALTA-2530 (bottom) to reduce or attenuate IL-ip levels in BALF following exposure to lipopolysaccharide in the 4 hours (FIG. 12 A) and 24 hours (FIG. 12B) after lipopolysaccharide administration. In FIGS. 12A-B, n.s. indicates not significant, one asterisk (*) indicates p<0.05, and three asterisks (***) indicate p<0.001. The results show that exposure to lipopolysaccharide significantly increases IL-ip levels in BALF and that IL- 1R antagonism by treatment with both low-dose and high-dose ALTA-2530 attenuated the IL-ip levels in the BALF at 4 hours and at 24 hours after lipopolysaccharide administration. Each line represents an individual animal.

[0023] FIGS. 13 A-B show results from a study assessing the ability of low dose ALTA- 2530 (top) and high dose ALTA-2530 (bottom) to reduce or attenuate IL-6 levels in BALF following exposure to lipopolysaccharide in the 4 hours (FIG. 13 A) and 24 hours (FIG. 13B) after lipopolysaccharide administration. In FIGS. 13 A-B, n.s. indicates not significant, two asterisks (**) indicate p<0.01, and three asterisks (***) indicate p<0.001. The results show that exposure to lipopolysaccharide significantly increases IL-6 levels in BALF and that IL- 1R antagonism by treatment with both low-dose and high-dose ALTA-2530 attenuated the IL-6 levels in the BALF at 4 hours and at 24 hours after lipopolysaccharide administration. Each line represents an individual animal.

[0024] FIGS. 14A-B show results from a study assessing the ability of low dose ALTA- 2530 (top) and high dose ALTA-2530 (bottom) to reduce or attenuate IL-6 levels in serum following exposure to lipopolysaccharide in the 4 hours (FIG. 14 A) and 24 hours (FIG. 14B) after lipopolysaccharide administration. In FIGS. 14A-B, n.s. indicates not significant, one asterisk (*) indicates p<0.05, two asterisks (**) indicate p<0.01, three asterisks (***) indicate p<0.001, and four asterisks (****) indicate pO.OOOl. The results show that IL-1R antagonism by treatment with both low-dose and high-dose ALTA-2530 attenuated the IL-6 levels in the serum at 4 hours and at 24 hours after lipopolysaccharide administration. Each line represents an individual animal.

[0025] FIG. 15 shows results from a study assessing the ability of low dose ALTA-2530 to reduce or attenuate TNF-a levels in BALF following exposure to lipopolysaccharide in the 4 hours after lipopolysaccharide administration. In FIG. 15, one asterisk (*) indicates p<0.05 and two asterisks (**) indicate p<0.01. The results show that IL-1R antagonism by treatment with low-dose ALTA-2530 attenuated TNF-a levels in the BALF at 4 hours after lipopolysaccharide administration. Each line represents an individual animal.

[0026] FIG. 16 shows results from a study assessing the ability of low dose ALTA-2530 to reduce or attenuate IL-8 levels in serum following exposure to lipopolysaccharide in the 4 hours after lipopolysaccharide administration. In FIG. 16, n.s. indicates not significant and one asterisk (*) indicates p<0.05. The results show that IL-1R antagonism by treatment with low-dose ALTA-2530 attenuated the IL-8 levels in the serum at 4 hours after lipopolysaccharide administration. Each line represents an individual animal.

SUMMARY OF THE INVENTION

[0027] In one aspect, a method is described for the treatment of neutrophilic lung diseases. In some embodiments, the neutrophilic lung disease is characterized by tissue damage and inflammation associated with dysregulation of neutrophil activation and/or recruitment. In some embodiments, the treatment includes delivery (e.g., inhaled delivery) of an IL-1R antagonist (“IL-IRa,” e.g., recombinant human IL-IRa), to block the activity of IL- la and IL-ip and to thereby target neutrophil activation and migration.

[0028] In one aspect, the invention provides a method for treating a neutrophilic lung disease in a human subject in need thereof, comprising administering a pharmaceutical composition comprising an effective amount of an IL-IRa (e.g., recombinant human IL-IRa) to the human subject, wherein the neutrophilic lung disease is selected from the group consisting of steroid-resistant chronic obstructive pulmonary disease, bronchiectasis, neutrophilic asthma, acute respiratory distress syndrome, chemical lung injury, and rheumatoid arthritis-associated interstitial lung disease.

[0029] In certain embodiments, the IL-IRa (e.g., recombinant human IL-IRa) is administered to the airways of the human subject. In some cases, for example, the IL-IRa (e.g., recombinant human IL-IRa) is administered by inhalation. In some cases, the IL-IRa (e.g., recombinant human IL-IRa) is administered intranasally. In certain embodiments, the IL-IRa (e.g., recombinant human IL-IRa) is delivered to the distal regions of the human subject’s lung. In certain embodiments, the IL-IRa (e.g., recombinant human IL-IRa) is administered intratracheally.

[0030] In certain embodiments, the IL-IRa (e.g., recombinant human IL-IRa) is nebulized. In some cases, the nebulized IL-IRa (e.g., recombinant human IL-IRa) has a mass median aerodynamic diameter (MMAD) of about 1 pm to about 15 pm. For example, the nebulized IL-IRa (e.g., recombinant human IL-IRa) may have a MMAD of about 3 pm. [0031] In certain embodiments the pharmaceutical composition comprises ALTA-2530, a formulation of recombinant human IL-IRa. In some embodiments, the pharmaceutical composition comprises anakinra.

[0032] In certain embodiments, the methods described herein reduce secretion of a chemokine(s) that promotes neutrophil recruitment. In some cases, the chemokine is selected from the group consisting of CXCL1, CXCL2, CCL2, CCL4, and IL-8. In certain embodiments, the methods described herein reduce neutrophilic airway inflammation. In certain embodiments, the methods described herein reduce airway neutrophils. In certain embodiments, the methods described herein reduce airway neutrophils without eradicating the neutrophils. In certain embodiments, the methods described herein prevent or reduce alveolar edema and/or neutrophil trafficking.

[0033] In certain embodiments, the IL-IRa (e.g., recombinant human IL-IRa) in the pharmaceutical composition binds to an IL-1 receptor with an affinity higher than IL-ip. In certain embodiments, the IL-IRa (e.g., recombinant human IL-IRa) in the pharmaceutical composition binds to an IL-1 receptor with an affinity higher than IL- la.

DETAILED DESCRIPTION

Definitions

[0034] The following are definitions of terms used in the present specification. The initial definition provided for a group or term herein applies to that group or term throughout the present specification individually or as part of another group, unless otherwise indicated. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art.

[0035] The term “interleukin 1 receptor antagonist” or “IL-IRa” refers to any peptide or protein that inhibits or blocks (either competitively or non-competitively) the activity of an interleukin-1 receptor (e.g., IL-1 type 1 receptor). In some embodiments, the IL-IRa is a recombinant human IL-lra (rhIL-IRa).

[0036] The term “ALTA-2530” when used herein refers to or describes any inhaled (e.g., nebulized) pharmaceutical composition comprising an rhIL-IRa (e.g., anakinra, or a peptide IL-1R antagonist). In some embodiments, ALTA-2530 comprises a peptide IL-IRa of about 50 amino acids in length or less. In some embodiments, ALTA-2530 comprises anakinra. In some embodiments, ALTA-2530 comprises an anakinra-containing pharmaceutical composition.

[0037] The term “lower airways” or “lower respiratory tract” when used herein refers to or describes anatomic regions below the larynx including the trachea and lungs, as well as lower regions of the lung.

[0038] The term “upper airways” or “upper respiratory tract” when used herein refers to or describes the anatomic regions including the passageways from flares or nostrils to the soft palate and includes the sinuses.

[0039] The terms “treating,” “treatment,” and “therapy” as used herein refer to attempted reduction or amelioration of the progression, severity and/or duration of a disorder, or the attempted amelioration of one or more symptoms thereof resulting from the administration of one or more modalities (e.g., one or more therapeutic agents such as a compound or composition of the invention).

[0040] As used herein, the terms “prevent,” “preventing” and “prevention” refer to the prevention or inhibiting of the recurrence, onset, or development of a disorder or a symptom thereof in a subject resulting from the administration of a therapy e.g., a prophylactic or therapeutic agent), or the administration of a combination of therapies (e.g., a combination of prophylactic or therapeutic agents).

[0041] As used herein, “therapeutically effective amount” or “effective amount” refers to any amount that is necessary or sufficient for achieving or promoting a desired outcome. In some instances, an effective amount is a therapeutically effective amount. In some embodiments, a therapeutically effective amount is any amount that is necessary or sufficient for promoting or achieving a desired biological response in a subject. The effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular agent being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art can empirically determine the effective amount of a particular agent without necessitating undue experimentation.

[0042] As used herein, the terms “subject” and “patient” are used interchangeably herein. The terms “subject” and “subjects” refer to an animal, preferably a mammal including a nonprimate and a primate (e.g., a monkey such as a cynomolgus monkey, a chimpanzee, and a human), and more preferably a human. The term “animal” also includes, but is not limited to, companion animals such as cats and dogs; zoo animals; wild animals; farm or sport animals such as ruminants, non-ruminants, livestock and fowl (e.g., horses, cattle, sheep, pigs, turkeys, ducks, and chickens); and laboratory animals, such as rodents (e.g., mice, rats), rabbits; and guinea pigs, as well as animals that are cloned or modified, either genetically or otherwise (e.g., transgenic animals).

[0043] As used herein, “a” or “an” means at least one, unless clearly indicated otherwise. The term “about,” unless otherwise indicated, refers to a value that is no more than 10% above or below the value being modified by the term. For example, the term “about 5% (w/w)” may mean a range of from 4.5% (w/w) to 5.5% (w/w). In some embodiment, the term “about” refers to a value that is no more than 5% above or below the value being modified by the term.

[0044] As used herein, unless indicated otherwise, the terms “composition” and “composition of the invention”, are used interchangeably. Unless stated otherwise, the terms are meant to encompass, and are not limited to, pharmaceutical compositions and nutraceutical compositions containing drug substance (e.g., rhIL-IRa). The composition may also contain one or more “excipients” that are inactive ingredients or compounds devoid of pharmacological activity or other direct effect in the diagnosis, cure, mitigation, treatment, or prevention of disease or to affect the structure or any function of the human.

[0045] As used herein, the term “vehicle” refers to a diluent, adjuvant, excipient, carrier, or filler with which the compound or composition of the invention is stored, transported, and/or administered.

[0046] As used herein, the phrase “pharmaceutically acceptable salt” refers to pharmaceutically acceptable organic or inorganic salts of a compound of the invention. [0047] As used herein, the term “pharmaceutically acceptable solvate” refers to an association of one or more solvent molecules and a compound of the invention. Examples of solvents that form pharmaceutically acceptable solvates include, but are not limited to water, saline, water-salt mixtures, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, polyethylene glycol and ethanolamine.

[0048] As used herein, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.

Treatment for Neutrophilic Lung Disease

[0049] In one aspect, a method for treating a neutrophilic lung disease is described. In certain embodiments, the method includes administering a pharmaceutical composition comprising an effective amount of an interleukin 1 receptor antagonist (IL-IRa) (e.g., rhlL- IRa) to the human subject (e.g., to the airways of the human subject). For example, the method may include administering a pharmaceutical composition comprising an effective amount of rhIL-lRa. rhIL-IRa blocks the biologic activity of IL-la and IL-ip by competitively inhibiting IL-la and IL-ip’s binding to IL-1R1. In some cases, the method may include administering ALTA-2530.

[0050] RhIL-IRa in ALTA-2530 formulation may have a higher affinity for human IL- 1R1 than either or both IL-la and IL-ip. In some embodiments, it was surprisingly found that ALTA-2530 has a KD value of 2.5 x 10' 11 M, while IL-la and IL-ip have KD values of 5.47 x 10' 7 M and 5.75 x 10' 10 M, respectively. Moreover, in some embodiments, it was found, using surface plasmon resonance binding analysis, that ALTA-2530 can bind faster, stronger, and longer to IL-1R1 than IL-ip. Data from these studies are reported in FIGS. 1 A- B and FIGS. 2A-B. A comparison of the association and dissociation constants of ALTA- 2530 and IL-ip, as determined by these studies, are reported in Table 1.

[0051] In some embodiments, it has been surprisingly found that IL-IRa (e.g., rhIL-IRa) can be directly administered to a human subject, e.g., to the lower airways of a human subject having a neutrophilic lung disease to effectively treat the disease. Without wishing to be bound by any specific theory, it is believed that, in the human subject having the neutrophilic lung disease disclosed herein, IL- la and IL-ip bind to the interleukin- 1 type I receptor which triggers neutrophil recruitment and that IL-IRa (e.g., rhIL-IRa) blocks the activities of these IL-1 cytokines, reducing neutrophilia and thereby treating the neutrophilic lung disease. [0052] Thus, in one aspect, a method for treating a neutrophilic lung disease in a human subject in need thereof is described, including administering a pharmaceutical composition comprising an effective amount of IL-IRa (e.g., rhIL-IRa) to the human subject. In some embodiments, the neutrophilic lung disease may be obstructive pulmonary disease (e.g., COPD), bronchiectasis, neutrophilic asthma, rheumatoid arthritis-associated interstitial lung disease, acute respiratory distress syndrome (ARDS), or chemical lung injury, or combinations of these diseases. In some embodiments, the neutrophilic lung disease is obstructive pulmonary disease (e.g., COPD). In some embodiments, the neutrophilic lung disease is bronchiectasis. In some embodiments, the neutrophilic lung disease is neutrophilic asthma. In some embodiments, the neutrophilic lung disease is rheumatoid arthritis- associated interstitial lung disease. In some embodiments, the neutrophilic lung disease is acute respiratory distress syndrome (ARDS), e.g., COVID-related ARDS. In some embodiments, the neutrophilic lung disease is chemical lung injury. In some embodiments, the effective amount of IL-IRa (e.g., rhIL-IRa) is from about 0.1 mg to about 200 mg per day.

[0053] In some embodiments, the neutrophilic lung disease is COPD, e.g., steroid- resistant COPD. COPD is a chronic inflammatory lung disease that causes obstructed airflow from the lungs. Symptoms of COPD may include, for example, breathing difficulty, cough, mucus production, and wheezing. In some cases, COPD is caused by long-term exposure to irritating gases or particulate matter, e.g., cigarette smoke. In some cases, COPD is associated with an increased risk of developing heart disease and/or lung cancer. Certain steroids can reduce the inflammation in a patient’s lungs and are used in treating COPD. In some cases, steroid treatment can result in resistance. In some embodiments, the neutrophilic lung disease to be treated is steroid-resistant COPD. In some embodiments, the effective amount of IL-IRa (e.g., rhIL-IRa) is from about 0.1 mg to about 200 mg per day.

[0054] In some embodiments, the neutrophilic lung disease is asthma. Asthma is a chronic inflammatory airway disease with several distinct phenotypes (e.g., eosinophilic, neutrophilic, mixed granulocytic and paucigranulocytic asthma). In some embodiments, the neutrophilic lung disease is neutrophilic asthma. Neutrophilic asthma is a severe and persistent disease with frequent exacerbations and hospitalizations. Neutrophilic asthma is characterized by the presence of high levels of neutrophils in the lungs and airways and fixed airflow obstruction. Activated neutrophils release multiple proteinases, cytokines, chemokines, and reactive oxygen species which cause airway epithelial cell injury, inflammation, hyperresponsiveness and airway remodeling. In some cases, neutrophilic asthma is unresponsive to high dose inhaled corticosteroids and to novel monoclonal antibody therapies. In some embodiments, the effective amount of IL-IRa (e.g., rhIL-IRa) is from about 0.1 mg to about 200 mg per day.

[0055] In some embodiments, the neutrophilic lung disease is rheumatoid arthritis- associated interstitial lung disease. Rheumatoid arthritis (RA) is a systemic inflammatory disorder, with the most common extra-articular manifestation of RA being lung involvement. While any of the lung compartments can be affected and manifest as interstitial lung disease (ILD), pleural effusion, cricoarytenoiditis, constrictive or follicular bronchiolitis, bronchiectasis, pulmonary vasculitis, and pulmonary hypertension, RA-associated ILD is a leading cause of death in patients with RA and is associated with significant morbidity and mortality. In some embodiments, the effective amount of IL-IRa (e.g., rhIL-IRa) is from about 0.1 mg to about 200 mg per day.

[0056] In some embodiments, the neutrophilic lung disease is ARDS. ARDS is a lifethreatening disease which requires immediate mechanical ventilation to prevent lung failure. In some embodiments, the subject having ARDS suffers from neutrophilia mediated by IL-1 and IL-IRa (e.g., rhIL-IRa, such as in ALTA-2530 formulation) blocks the activities of IL- la and/or IL-ip local to the lower airways and thereby effectively treats ARDS. [0057] In some embodiments, the neutrophilic lung disease is a chemical lung injury. As used herein, a chemical lung injury includes any injury (e.g., inflammation or damage) to the lung as a result of inhalation of one or more foreign and/or toxic agents. In some embodiments, the subject having a chemical lung injury suffers from neutrophilia mediated by IL-la and IL-ip, and IL-IRa (e.g., rhIL-IRa, such as in ALTA-2530 formulation) blocks the activities of these cytokines local to the lower airways and thereby effectively treats neutrophilia and the chemical lung injury.

[0058] In some embodiments, the chemical lung injury is caused by inhalation of one or more chemical warfare agents. Non-limiting examples of the chemical warfare agents include chlorine gas and sulfur mustard. Sulfur mustard exposure, in particular, is associated with neutrophil infiltration into the airways. Persistence of blood neutrophilia was observed in Iranian sulfur mustard victims years after exposure (Javad Beheshti et al., Mustard lung secrets: long term clinicopathological study following mustard gas exposure, Pathol. Res. Pract. 202(10):739-44 (2006)). Other examples of the chemical warfare agents known in the art are contemplated. In some embodiments, the chemical lung injury is chlorine-induced bronchiolitis obliterans syndrome (BOS). In some embodiments, the chemical lung injury is sulfur mustard-induced BOS.

[0059] In other embodiments, the chemical lung injury is caused by inhalation of one or more environmental and/or industrial toxic agents. Various toxic agents exist in the environmental (natural or artificial) and industrial setting. A human subject may come into contact with and inhale these agents (e.g., while working) and suffer from injuries to the lung that lead to inflammation. For example, a human subject may come into contact with and inhale these agents through exposure to toxic vapors and/or gases produced from or associated with burn pits (e.g., as military burn pits). Non-limiting examples of the environmental and industrial toxic agents include isocyanate (e.g., toluene diisocyanate), nitrogen oxide, sulfuric acid, ammonia, phosgene, diacetyl, 2,3-pentanedione, 2,3- hexanedione, fly ash, fiberglass, silica, coal dust, asbestos, hydrogen cyanide, cadmium, acrolein, acetaldehyde, formaldehyde, aluminum, beryllium, iron, cotton, tin oxide, bauxite, mercury, sulfur dioxide, zinc chloride, polymer fumes, and metal fumes (e.g., fumes generated by copper, magnesium, nickel, silver, or zinc). In some specific embodiments, the chemical lung injury is pneumoconiosis. As used herein, pneumoconiosis refers to a class of interstitial lung diseases caused by inhalation of various solid particles. In some specific embodiments, the chemical lung injury is bronchiolitis obliterans, commonly referred to as “popcorn lung.” In some specific embodiments, the bronchiolitis obliterans is caused by the inhalation of one or more industrial toxic agents selected from the group consisting of acetaldehyde, formaldehyde, diacetyl, 2,3-pentanedione, and 2,3-hexanedione.

[0060] In still other embodiments, the chemical lung injury is a vaping-associated lung injury. Vaping, or using electronic cigarettes, may cause the user to inhale harmful chemicals and result in lung injuries. In recent years, there has been a significant growth in cases of vaping-associated lung injury reported in the U.S. In some embodiments, an electronic cigarette user inhales harmful chemicals found in the electronic cigarette liquid, e.g, diacetyl, which causes injuries, e.g, bum, inflammation, to the lung. Other non-limiting examples of harmful chemicals in the electronic cigarette causing vaping-associated lung injury include 2,3-pentanedione, nicotine, carbonyls, volatile organic compounds (e.g., benzene and toluene), trace metal elements, a-Tocopheryl acetate, and bacterial endotoxins and fungal glucans. In some embodiments, the vaping-associated lung injury is pneumonitis. In some specific embodiments, the vaping-associated lung injury is bronchiolitis obliterans, commonly referred to as “popcorn lung.”

[0061] In some embodiments, IL-IRa (e.g., rhIL-IRa, such as in ALTA-2530 formulation) is administered to the airways, e.g., lower airways, of the human subject. In some cases, for example, the IL-IRa (e.g., rhIL-IRa) may be administered directly to the airways, e.g., the lower airways, of the human subject. Non-limiting examples of administration of IL-IRa (e.g., rhIL-IRa) to the airways of the human subject include administration by inhalation and intranasal administration. Advantages of administration directly to the airways, e.g., local airways, include the lack of adverse effects due to systemic exposure of the active ingredient.

[0062] In some embodiments, IL-IRa (e.g., rhIL-IRa, such as in ALTA-2530 formulation) is delivered to the distal regions of the human subject’s lung(s).

[0063] In some embodiments, IL-IRa (e.g., rhIL-IRa, such as in ALTA-2530 formulation) is administered via inhalation or via direct instillation into the lower airways. In some embodiments, IL-IRa (e.g., rhIL-IRa) is administered intratracheally.

[0064] In certain embodiments, a delivery device is used to administer IL-IRa (e.g., rhIL- IRa, such as in ATLA-2530 formulation) directly to the lower airways. Non-limiting examples of the delivery devices include a nebulizer, an inhaler, and a subminiature aerolizer. In some specific embodiments, the delivery device is a dry powder inhaler. In some specific embodiments, the delivery device is a mesh nebulizer. In some specific embodiments, the delivery devise is a jet nebulizer.

[0065] The methods described herein beneficially treat the subject’s neutrophilic lung disease. The effectiveness of the treatment may be assessed by various factors, which those skilled in the art will readily understand. In some embodiments, the methods described herein reduce the secretion of a chemokine (or chemokines) that promotes neutrophil recruitment. In some cases, for example, the administration of IL-IRa (e.g., rhIL-IRa, such as in ALTA-2530 formulation) to the subject may block the IL-1R1 to reduce the secretion of IL-la and IL-ip. In some cases, the administration of IL-IRa (e.g., rhIL-IRa, such as in ALTA-2530 formulation) may reduce the secretion of one or more cytokines or chemokines downstream on IL-1 binding to the IL-1R including but not limited to CXCL1, CXCL2, CCL2, CCL4, and IL-8.

[0066] In some embodiments, the method described herein reduces neutrophilic airway inflammation in the subject. In some embodiments, the method described herein reduces airway neutrophils in the subject. In some cases, the methods may reduce airway neutrophils without eradicating the neutrophils. As noted above, neutrophils play an important role in the innate response, but the dysregulation of neutrophils (e.g., too many or too active neutrophils) lead to tissue damage and disease. Thus, the propensity of the methods described herein to reduce neutrophils (e.g., airway neutrophils) without eradicating the neutrophils advantageously addresses neutrophil dysregulation, and thereby the tissue damage and disease, without negatively impacting the role of neutrophils in the immune system. In some embodiments, the methods described herein prevent or reduce at least one of alveolar edema and neutrophil trafficking.

Pharmaceutical Compositions

[0067] In some aspects, the method for treating a neutrophilic lung disease described herein includes administering IL-IRa (e.g., rhIL-IRa) to a human subject. IL-IRa (e.g., rhIL- IRa) may be administered to the human subject as a component of a pharmaceutical composition. For example, the method may comprise administering a pharmaceutical composition comprising IL-IRa (e.g., rhIL-IRa), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition is a spray, aerosol, gel, solution, emulsion, or suspension.

[0068] In some embodiments, the pharmaceutical composition comprises ALTA-2530. In some embodiments, the pharmaceutical composition comprises anakinra. [0069] In some embodiments, the pharmaceutical composition further comprises a second therapeutic agent. The inclusion of a second therapeutic agent may improve the clinical benefit of the disclosed treatment method across indications. For example, the pharmaceutical composition may include, as a second therapeutic agent, cyclosporine, steroids, antibiotics, and combinations thereof.

[0070] The phrase “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body. Some examples of materials which can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as butylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer’s solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.

[0071] The term “carrier” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions also are capable of being comingled with the compounds of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency. The composition may also include additional agents such as an isotonicity agent, a preservative, a surfactant, and, a divalent cation, preferably, zinc.

[0072] In some embodiments, the pharmaceutically acceptable carrier is selected from the group consisting of saline, Ringer's solution, dextrose solution, and a combination thereof. Other suitable pharmaceutically acceptable carriers known in the art are contemplated. Suitable carriers and their formulations are described in Remington's Pharmaceutical Sciences, 2005, Mack Publishing Co. The pH of the solution is preferably from about 5 to about 8, and more preferably from about 7 to about 7.5. The formulation may also comprise a lyophilized powder. Further carriers include sustained release preparations such as semipermeable matrices of solid hydrophobic polymers, which matrices are in the form of shaped articles, e.g., films, liposomes or microparticles. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon, for instance, the route of administration and concentration of IL-IRa (e.g., rhIL-IRa) being administered. [0073] The pharmaceutical composition may also include an excipient, or an agent for stabilization of IL-IRa (e.g., rhIL-IRa), such as a buffer, a reducing agent, a bulk protein, amino acids (such as e.g., glycine or praline), or a carbohydrate. Bulk proteins useful in formulating IL-IRa (e.g., rhIL-IRa), as a non-limiting example, include albumin. Typical carbohydrates useful in formulating IL-IRa (e.g., rhIL-IRa) include, as non-limiting examples, mannitol, lactose, trehalose, and glucose.

[0074] The pharmaceutical composition may also include a surfactant. Surfactants may be used to prevent soluble and insoluble aggregation and/or precipitation of proteins (e.g., IL- IRa) included in the composition. Suitable surfactants include but are not limited to sorbitan trioleate, soya lecithin, and oleic acid. In certain cases, solution aerosols are preferred using solvents such as ethanol. Thus, the pharmaceutical composition comprising IL-IRa (e.g., rhIL-IRa) can also include a surfactant that can reduce or prevent surface-induced aggregation of IL-IRa (e.g., rhIL-IRa) caused by atomization of the solution in forming an aerosol. Various conventional surfactants can be employed, such as polyoxyethylene fatty acid esters and alcohols, and polyoxyethylene sorbitol fatty acid esters. Amounts will generally range between 0.001% and 4% by weight of the formulation. Especially preferred surfactants include, as non-limiting examples, polyoxyethylene sorbitan mono-oleate, polysorbate 80, and polysorbate 20. Additional agents known in the art can also be included in the pharmaceutical composition.

[0075] In some embodiments, the pharmaceutical compositions further comprise one or more compounds that reduce the rate by which IL-IRa (e.g., rhIL-IRa) will decay or will change in character. Such “stabilizers” or “preservatives” may include, but are not limited to, amino acids, antioxidants, pH buffers, or salt buffers. Non-limiting examples of antioxidants include butylated hydroxy anisole (BHA), ascorbic acid and derivatives thereof, tocopherol and derivatives thereof, butylated hydroxy anisole and cysteine. Non-limiting examples of preservatives include parabens, such as methyl or propyl p-hydroxybenzoate and benzalkonium chloride. Additional non-limiting examples of amino acids include glycine or proline. [0076] The pharmaceutical composition comprising IL-IRa (e.g., rhIL-IRa) may be formulated to be compatible with its intended route of administration. Examples of routes of administration include, but are not limited to, oral or nasal inhalation (e.g., inhalation of sufficiently small particles to be deposited expressly within the lower airways). In various embodiments, the pharmaceutical composition is sterile and in suitable form for administration to a (human) subject.

[0077] The dosage form of the pharmaceutical composition will typically vary depending on their use. Non-limiting examples of dosage forms include powders; solutions; aerosols (e.g., sprays, metered or nonmetered dose atomizers, oral or nasal inhalers including metered dose inhalers (MDI)); liquid dosage forms suitable for mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or a water-in-oil liquid emulsions), solutions, and sterile solids (e.g., crystalline or amorphous solids) that can also be reconstituted to provide liquid dosage forms suitable for lower airways administration.

Administration to the Human Subject

[0078] The amount of IL-IRa (e.g., rhIL-IRa) (or the pharmaceutical composition comprising IL-IRa, such at ALTA-2530) that will be effective to treat a neutrophilic lung disease will vary, e.g., with the nature and severity of the disease and the route or mode by which the IL-IRa (e.g., rhIL-IRa) is administered. The frequency and dosage will also vary according to factors specific for each subject, such as age, body, weight, response, and the past medical history of the subject. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. Suitable regimens can be selected by one skilled in the art by considering such factors and by following, for example, dosages reported in the literature and recommended in the Physician’s Desk Reference (71st ed., 2017).

[0079] In general, the recommended daily dose range of IL-IRa (e.g., rhIL-IRa) for the neutrophilic lung disease described herein lie within the range of from about 0.01 mg to about 200 mg per day, given as a single once-a-day dose preferably or as divided doses throughout a day. In one embodiment, the daily dose is administered twice daily in equally divided doses. Specifically, a daily dose range should be from about 100 micrograms to about 50 milligrams per day, more specifically, between about 500 micrograms and about 5 milligrams per day. In managing the patient, the therapy may be initiated at a lower dose, perhaps about 500 micrograms, and increased if necessary up to about 5.0 milligrams per day as either a single dose or divided doses, depending on the patient’s global response. It may be necessary to use dosages of IL-IRa (e.g., rhIL-IRa) outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art. Furthermore, it is noted that in instances where a clinician or treating physician is involved, such a person will know how and when to interrupt, adjust, or terminate therapy in conjunction with individual subject response.

[0080] In some embodiments, the effective amount of IL-IRa (e.g., rhIL-IRa) is from about 0.1 mg to about 100 mg per day, from about 0.1 mg to about 50 mg per day, or from about 0.1 mg to about 10 mg per day. Effective dosages and schedules for administering IL- IRa (e.g., rhIL-IRa) may be determined empirically, and making such determinations is within the skill in the art. Those skilled in the art will understand that the dosage of any composition administered will vary depending on, for example, the subject receiving the composition, the route of administration, the particular composition used including the coadministration of other drugs and other drugs being administered to the mammal. A typical daily dosage of IL-IRa (e.g., rhIL-IRa) used alone might range from about e.g., 0.25 mg to up to 20.0 mg per oral or nasal inhalation, or 0.125 mg to 25.0 mg per oral or nasal inhalation, however, depending on symptoms and body weight a higher or lower dosage may be appropriate.

[0081] Alternatively, the dosage administered can vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent, and its mode and route of administration; age, and health of the recipient; nature and extent of symptoms, kind of concurrent treatment, frequency of treatment, and the effect desired. As an example, treatment can be provided as a one-time or periodic dosage of IL-IRa (e.g., rhIL-IRa) from 0.01 to 200 mg, or 0.01 to 100 mg, such as 0.025, 0.05, 0.075, 0.1, 0.125, 0.25, 0.50, 0.75, 1.0, 1.125, 1.25, 1.5, 2, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200 mg, per day, on at least one of day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40, or alternatively or additionally, at least one of week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, or 52, or alternatively or additionally, at least one of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 years, or any combination thereof, using single, infusion or repeated doses. In some specific embodiments, the effective amount of IL-IRa (e.g., rhIL-IRa) is from about 0.125 mg to 20.0 mg per day. [0082] Different therapeutically effective amounts of IL-IRa (e.g., rhIL-IRa) may be applicable for different neutrophilic lung diseases, as will be readily known by those of skill in the art. Similarly, different therapeutically effective compounds may be included in a specific composition depending on the subject’s disease. Similarly, amounts sufficient to prevent, manage, treat or ameliorate such disease, but insufficient to cause, or sufficient to reduce, adverse effects associated with IL-IRa (e.g., rhIL-IRa) are also encompassed by the above described dosage amounts and dose frequency schedules. Further, when a subject is administered multiple dosages of IL-IRa (e.g., rhIL-IRa), not all of the dosages need be the same. For example, the dosage administered to the subject may be increased to improve the prophylactic or therapeutic effect of the compound or it may be decreased to reduce one or more side effects that a particular subject is experiencing.

Delivery Devices

[0083] As noted above, one aspect of the current invention is the local delivery, e.g., directly to the lower airways, of IL-IRa (e.g., rhIL-IRa) to the human subject and the delivery device that accomplishes said dosing. Delivery devices described herein may provide methods for the direct delivery of a pharmaceutical composition whereby IL-IRa (e.g., rhIL-IRa) may have local effects, e.g., directly in the vicinity of the mucosa of the lower airways. The advantages of local therapy for local disease include the lack of adverse effects due to systemic exposure of the active ingredient.

[0084] For pulmonary administration, preferably IL-IRa (e.g., rhIL-IRa) is delivered in a particle size effective for reaching the lower airways. There are a several desirable features of an inhalation device for administering IL-IRa (e.g., rhIL-IRa). To be specific, delivery by the inhalation device is generally reliable, reproducible, and accurate. The inhalation device can optionally deliver small dry particles, e.g., less than about 10 microns, preferably about 3 to 5 microns, for good respirability, or dry particles with small stokes radius. In some cases, the inhalation device can optionally deliver a wet mist of particles, e.g., droplets. Suitable liquid nebulizers include, for example, InnoSpire Go mesh nebulizer (Philips), iNeb AAD system (Philips) the iNeb Advance nebulizer (Philips), and PARI nebulizer (PARI).

[0085] In some embodiments, IL-IRa (e.g., rhIL-IRa) may be nebulized and achieve particles mass median aerodynamic diameter (MMAD) of about between about 1 pm and about 5 pm, between about 5 pm and between about 10 pm, between about 10 pm and 15 pm, or between about 15 pm and 20 pm. Preferably, the MMAD is about 3 pm, consistent with delivery to lower regions of the lung. An MMAD of about 3 gm permits a high delivered dose to the region of the respiratory tract most associated with neutrophilia. Further, inhaled delivery lowers systemic exposure with the potential benefit of reducing the risk of the adverse events associated with alternative administration routes, e.g., high dose IV infusion therapy.

[0086] According to the invention, IL-IRa (e.g., rhIL-IRa) can be delivered by any of a variety of inhalation devices known in the art for administration of a therapeutic agent by inhalation. These devices capable of depositing aerosolized formulations in the lower airways of a patient include but are not limited to metered dose inhalers, sprayers, nebulizers, and dry powder generators. Other devices suitable for pulmonary administration of proteins and small molecules, including IL-IRa (e.g., rhIL-IRa), are also known in the art. All such devices can use of formulations suitable for the dispensing of IL-IRa (e.g., rhIL-IRa) in an aerosol. Such aerosols can include nanoparticles, microparticles, solutions (both aqueous and nonaqueous), or solid particles.

[0087] Nebulizers like AERx Aradigm, the Ultravent nebulizer (Mallinckrodt), and the Acorn II nebulizer (Marquest Medical Products) (U.S. Pat. No. 5,404,871, WO 97/22376, entirely incorporated herein by reference), produce aerosols from solutions. In some embodiments, the nebulizer is Monaghan Aeroeclipse II Breath Activated Jet Nebulizer, or the Philips Innospire Go vibrating mesh nebulizer.

[0088] Metered dose inhalers such as the Ventolin metered dose inhaler, typically use a propellent gas and require actuation during inspiration see, e.g., WO 94/16970, WO 98/35888, entirely incorporated herein by reference).

[0089] Suitable dry powder inhalers like Turbuhaler (Astra), Rotahaler (Glaxo), Diskus (Glaxo), Spiros inhaler (Dura/Elan) devices, the Spinhaler powder inhaler (Fisons), use breath-actuation of a mixed powder (U.S. Pat. No. 4,668,218, EP 237507, WO 97/25086, WO 94/08552, U.S. Pat. No. 5,458,135, WO 94/06498, all of which are herein entirely incorporated by reference). Metered dose inhalers, dry powder inhalers and the like generate small particle aerosols.

[0090] These specific examples of commercially available inhalation devices are intended to be a representative of specific devices suitable for the practice of this invention and are not intended as limiting the scope of the invention. In some embodiments, a pharmaceutical composition comprising IL-IRa (e.g., rhIL-IRa) is delivered by a dry powder inhaler or a sprayer. In other embodiments, a pharmaceutical composition comprising IL- IRa (e.g., rhIL-IRa) is an aerosolized formulation delivered by an aerosolized nebulizer. [0091] The pharmaceutical composition comprising IL-IRa (e.g., rhIL-IRa) can be administered as a topical spray or powder to the lower airways of a human subject by a delivery device (e.g., oral or nasal inhaler, aerosol generator, oral dry powder inhaler, through a fiberoptic scope, or via syringe during surgical intervention). These numerous drug delivery devices capable of drug distribution to the lower airways can use a liquid, semisolid, and solid composition. Investigators have found the site of deposition in the lower airways and the deposition area depend on several parameters related to the delivery device, such as mode of administration, particle size of the formulation and velocity of the delivered particles. They describe several in vitro and in vivo methods that may be used by one of ordinary skill in the art to study distribution and clearance of therapeutics delivered to the lower airways, all of which is incorporated in its entirety, herein. Thus, any of these devices may be selected for use in the current invention, given one or more advantages for a particular indication, technique, and subject. These delivery devices include but are not limited to devices producing aerosols (metered-dose inhalers (MDIs)), nebulizers and other metered and nonmetered inhalers.

[0092] In general, current container-closure system designs for inhalation spray drug products include both premetered and device-metered presentations using mechanical or power assistance and/or energy from patient inspiration for production of the spray plume. Premetered presentations may contain previously measured doses or a dose fraction in some type of units (e.g., single, multiple blisters, or other cavities) that are subsequently inserted into the device during manufacture or by the patient before use. Typical device-metered units have a reservoir containing formulation sufficient for multiple doses that are delivered as metered sprays by the device itself when activated by the patient.

[0093] Administration of a pharmaceutical composition comprising IL-IRa (e.g., rhIL- IRa) as a spray can be produced by forcing a suspension or solution of IL-IRa (e.g., rhIL- IRa) inhibitor through a nozzle under pressure. The nozzle size and configuration, the applied pressure, and the liquid feed rate can be chosen to achieve the desired output and particle size to optimize deposition expressly in the lower airways. An electrospray can be produced, for example, by an electric field in connection with a capillary or nozzle feed. Advantageously, particles of IL-IRa (e.g., rhIL-IRa) delivered by a sprayer have a particle size less than about 20 microns, preferably in the range below 10 microns, and most preferably, about 3 to 5 microns, but other particle sizes may be appropriate depending on the device, composition, and subject needs.

[0094] Commercially available nebulizers for liquid formulations, including jet nebulizers, mesh nebulizers (e.g., vibrating mesh nebulizers), and ultrasonic nebulizers may also be useful for administration to the lower airways. Liquid formulations may be directly nebulized and lyophilized powder nebulized after reconstitution. Alternatively, the composition may be aerosolized using a metered dose inhaler, or inhaled as a lyophilized and milled powder. In addition, the liquid formulation of composition may be instilled through a bronchoscope, placed directly into the affected regions.

[0095] In one embodiment of the present invention, IL-IRa (e.g., rhIL-IRa) may be administered by a metered dose inhaler (MDI). The metered-dose inhaler can contain therapeutically active ingredients dissolved or suspended in a propellant, a mixture of propellants, or a mixture of solvents, propellants, and/or other excipients in compact pressurized aerosol dispensers. The MDI may discharge up to several hundred metered doses of the composition. Depending on the composition, each actuation may contain from a few micrograms (pg) up to milligrams (mg) of the active ingredients delivered in a volume typically between 25 and 100 micro liters. In an MDI, a propellant, IL-IRa (e.g., rhIL-IRa), and various excipients or other compounds are contained in a canister as a mixture including a liquified compressed gas (propellants). Actuation of the metering valve releases the mixture as an aerosol, preferably containing particles in the size range of less than about 20 microns. In some embodiments, the particle size is less than about 10 microns. In some embodiments, the particle size is below 5 microns. The desired aerosol particle size can be obtained by employing a formulation of antibody composition protein produced by various methods known to those of skill in the art, including jet-milling, spray drying, critical point condensation, or other methods well known to one of ordinary skill in the art.

[0096] Pharmaceutical compositions of IL-IRa (e.g., rhIL-IRa) for use with a metered- dose inhaler device can include a finely divided powder containing IL-IRa (e.g., rhIL-IRa) as a suspension in a non-aqueous medium, for example, suspended in a propellant with the aid of a surfactant or solubilizing agent. The propellant can be any conventional material including but not limited to chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichlorofluoromethane, dichlorodifluoromethane, di chlorotetrafluoroethanol and 1, 1,1,2-tetrafluoroethane, HFA- 134a (hydrofluroalkane-134a), HFA-227 (hydrofluroalkane-227). Hydrofluorocarbon is a preferred propellant. A surfactant can be chosen to stabilize the IL-IRa (e.g., rhIL-IRa) as a suspension in the propellant, to protect the active agent against chemical degradation. In some cases, solution aerosols are preferred using solvents such as ethanol for more water- soluble active agents. Additional agents including a protein can also be included in the composition.

[0097] One of ordinary skill in the art will recognize that the methods of the current invention can be achieved by lower airways administration of IL-IRa (e.g., rhIL-IRa) compositions via devices not described herein. The current invention also incorporates unitdose metering and nonmetering spray devices that are especially suited for single administration. These devices are typically used for acute short-term treatments (i.e., acute exacerbations) and single-dose delivery (i.e., long acting compositions) and can accommodate a liquid, powder, or mixture of both formulations of the composition. However, in certain circumstances, these unit dose devices may be preferred over multidose devices when used repeatedly in a particular way. Such uses may include but are not limited to repeated procedures where a sterile device is preferred.

[0098] In one embodiment, such an intrapulmonary aerosolizer comprises an aerosolizer attached to a pressure generator for delivery of liquid as an aerosol and which can be positioned in close proximity to the lungs by being inserted into the trachea directly or into an endotracheal tube or bronchoscope positioned within the trachea. Such an aerolizer may operate at pressures of up to about 2000 psi and produces particles with a medium particle size of 12 pm.

[0099] In an alternate embodiment, such an intrapulmonary aerosolizer comprises a substantially elongated sleeve member, a substantially elongated insert, and a substantially elongated body member. The sleeve member includes a threaded inner surface, which is adapted to receive the insert, which is a correspondingly threaded member. The threaded insert provides a substantially helical channel. The body member includes a cavity on its first end, which terminates by an end wall at its second end. The end wall includes an orifice extending therethrough. The body member is connected with the sleeve member to provide the aerosolizer of the present invention. The aerosolizer is sized to accommodate insertion into the trachea of a subject for administration of compositions containing IL-IRa (e.g., rhIL- IRa). For operation of the device, the aerosolizer is connected by a suitable tube with a liquid pressure driver apparatus. The liquid pressure driver apparatus is adapted to pass liquid material (e.g., a pharmaceutical composition comprising IL-IRa (e.g., rhIL-IRa)) therefrom which is sprayed from the aerosolizer. Due to the location of the device deep within the trachea, the liquid material is sprayed in close proximity to the lungs, with resulting improved penetration and distribution of the sprayed material in the lungs.

[0100] In an alternate embodiment, such an aerosolizer, sized for intratracheal insertion, is adapted for spraying a composition containing IL-IRa (e.g., rhIL-IRa) directly into the lower airways (e.g., in close proximity to the lungs). The aerosolizer is placed into connection with a liquid pressure driver apparatus for delivering of the liquid composition. The aerosolizer comprises a generally elongated sleeve member, which defines a first end and a second end and includes a longitudinally extending opening therethrough. The first end of the sleeve member is placed in connection with the liquid pressure driver apparatus. A generally elongated insert is also provided. The generally elongated insert defines a first end and a second end and is received within at least a portion of the longitudinally extending opening of the sleeve member. The insert includes an outer surface which has at least one substantially helical channel provided surrounding its outer surface which extends from the first end to the second end. The substantially helical channel of the insert is adapted to pass the liquid material, which is received by the sleeve member. A generally elongated body member is also included which is in connection with the sleeve member. The body member includes a cavity provided in its first end, which terminates at an end wall which is adjacent its second end. The end wall is provided having an orifice therein for spraying the liquid material, which is received from the insert. The portions of the sleeve member, insert and body member, in combination, are of sufficient size to allow for intratracheal insertion. A method of using such an aerosolizer includes the steps of connecting an aerosolizer with a first end of a hollow tube member and connecting the second end of the hollow tube member with the liquid pressure driver apparatus. The method further includes the steps of providing the aerosolizer in the trachea or into a member which is provided in the trachea, and then activating the liquid pressure driver apparatus for spraying a pharmaceutical composition comprising IL-IRa (e.g., rhIL-IRa) therefrom.

[0101] In an alternate embodiment, a powder dose composition containing IL-IRa (e.g., rhIL-IRa) is directly administered to the lower airways via use of a powder dispenser. Exemplary powder dispensers are disclosed in U.S. Pat. Nos. 5,513,630, 5,570,686 and 5,542,412, all of which are herein incorporated in their entirety. Such a powder dispenser is adapted to be brought into connection with an actuator, which introduces an amount of a gas for dispensing the powder dose. The dispenser includes a chamber for receiving the powder dose and a valve for permitting passage of the powder dose only when the actuator introduces the gas into the dispenser. The powder dose is passed from the dispenser via a tube to the lower airways of the subject. The powder dose may be delivered intratracheally, near the carina, which bypasses the potential for large losses of the powder dose to e.g., the mouth, throat, and trachea. In addition, in operation the gas passed from the actuator serves to slightly insufflate the lungs, which provides increased powder penetration. For the intratracheal insertion, the tube can be effected through an endotracheal tube in anesthetized, ventilated subjects, including animal or human patients, or in conscious subjects, the tube be inserted directly into the trachea preferably using a small dose of local anesthetic to the throat and/or a small amount of anesthetic on the tip of the tube, in order to minimize a “gag” response.

[0102] In one embodiment, a composition containing one or more therapeutic agents described herein is directly administered to the lower airways. Such administration may be carried out via use of an aerolizer, which create an aerosol containing the composition and which may be directly installed into the lower airways. Exemplary aerolizers are disclosed in U.S. Pat. Nos. 5,579,758; 6,041,775; 6,029,657; 6,016,800; 5,606,789; and 5,594,987 all of which are herein incorporated by reference in their entirety. The invention thus provides for the methods of administering pharmaceutical compositions containing IL-IRa (e.g., rhlL- IRa) directly to the lower airways by an aerolizer.

[0103] In particular, an embodiment of the present invention is a new use for the “intratracheal aerosolizer” device which methodology involves the generation of a fine aerosol at the tip of a long, relatively thin tube that is suitable for insertion into the trachea. Thus, the present invention provides a new method of use for this aerosolizer technology in a microcatheter as adapted herein, for use in the lower airways in the prevention, treatment, and care of lower airways disorders.

[0104] In another embodiment of the invention, an aerosolizing microcatheter is used to administer a composition containing a pro-inflammatory cytokine inhibitor. Examples of such catheters and their use, termed “intratracheal aerosolization,” which involves the generation of a fine aerosol at the tip of a long, relatively thin tube that is suitable for insertion into the trachea, are disclosed in U.S. Pat. Nos. 5,579,758; 5,594,987; 5,606,789; 6,016,800; and 6,041,775.

[0105] In a further embodiment, a new use for the microcatheter aerosolizer device (U.S. Pat. Nos. 6,016,800 and 6,029,657) is adapted for nasal and paranasal sinus delivery and uses to deliver bioactive agents (e.g., IL-IRa (e.g., rhIL-IRa)) in the treatment, prevention, and diagnosis of lower airways disorders. One advantage of this microcatheter aerosolizer is the potential small size (0.014" in diameter), and thus capable of being easily inserted into the working channel of a human flexible (1 to 2 mm in diameter) or ridged endoscope and thereby directed partially or completely into the ostium of a paranasal sinus.

[0106] One of ordinary skill in the art will recognize that the methods of the current invention can be achieved by lower airways administration of IL-IRa (e.g., rhIL-IRa) composition via devices not described herein.

EXAMPLES

Example 1. Distribution of ALTA-2530 Following Administration

[0107] To assess the distribution of IL-IRa (e.g., rhIL-IRa) following administration of ALTA-2530, immunohistochemistry studies were conducted. In one study, cynomolgus monkeys were administered ALTA-2530 or a saline control once daily for 7 days. Twenty- four hours after the last administration, lung tissue was harvested from the subjects.

Immunohistochemistry for rhIL-IRa was performed on lung sections. Positive staining was observed in the epithelium of the bronchioles, the alveolar septae, and the smooth muscle layer of the arteries in subjects that received ALTA-2530. Immunohistochemistry results for the saline-treated and the ALTA-2530-treated subjects are shown in FIG. 3A-B.

[0108] In another study, rats were administered a saline control, a low dose of ALTA- 2530, or a high dose of ALTA-2530 via nebulizer once daily for 7 days. Twenty-four hours after the last administration, lung tissue was harvested from the subjects.

Immunohistochemistry for rhIL-IRa was performed on lung sections. Positive staining was observed in the epithelium of the bronchioles, the alveolar septae, and the bronchus- associated lymphoid tissue in subjects that received ALTA-2530. Immunohistochemistry results are shown in FIG. 4.

[0109] The distribution of rhIL-IRa to the bronchioles and alveoli of rats and non-human primates, observed in these studies, supports the clinically relevant distribution in humans, as the airways in the human are larger in diameter than the airways of the nonclinical species studied.

Example 2. Efficacy of ALTA-2530 on IL-ip Stimulated Cytokine Release

[0110] To assess the efficacy of ALTA-2530 in treating IL-ip stimulated cytokine release, human whole blood assays were performed. In these assays, whole blood from healthy human donors was incubated in 96-well plates with increasing concentrations of ALTA-2530 30 minutes prior to IL-ip induction. Whole blood was treated with 1 ng/mL IL- ip for 24 hours. The supernatant was collected and analyzed for cytokine release by multiplex analysis.

[OHl] These assays showed that ALTA-2530 inhibits IL-ip stimulated cytokine and chemokine release from human whole blood in a dose dependent manner, with 50% maximal inhibition achieved at concentrations <100 ng/mL for most of these cytokines/chemokines. In particular, ALTA-2530 inhibits IL-6 (stimulates T- and B-cells and leads to monocyte recruitment; ICso 2.60 ng/mL), IL-8 (promotes neutrophil activation and recruitment; ICso 157.2 ng/mL), CCL2 (promotes leukocyte recruitment to site of inflammation; ICso 66.63 ng/mL), CCL4 (serves as a lymphocyte chemoattractant; ICso 79.21 ng/mL), CXCL1 (promotes neutrophil activation and recruitment; ICso 71.08 ng/mL), and CXCL2 (promotes neutrophil activation and recruitment; ICso 26.94 ng/mL). Results from these assays are shown in FIG. 5.

Example 3. ALTA-2530 Levels in BALE Following Administration

[0112] Another study was carried out in cynomolgus monkeys to assess ALTA-2530 levels in BALF following exposure of the subject to ALTA-2530. Cynomolgus monkeys received one dose of ALTA-2530. Twenty minutes after administration of ALTA-2530, BALF was collected, and the ALTA-2530 levels were quantified. BALF levels of ALTA- 2530 exceed the ICso value for IL-6, suggesting that ALTA-2530 reaches pharmacologically relevant exposures in vivo. Results of this study are shown in FIG. 6

Example 4. Efficacy of ALTA-2530 in Sulfur Mustard Induces Lung Injury

[0113] As noted above, sulfur mustard exposure is associated with neutrophil infiltration into the airways. ALTA-2530 was assessed in a sulfur mustard induced lung injury model. Rats were exposed to sulfur mustard and treated with saline or ALTA-2530 4 hours later. Histological examination (hematoxylin and eosin staining) of lung tissue showed relatively few neutrophils in lungs of rats treated with ALTA-2530, indicating treatment of lung neutrophilia. Moreover, while alveolar spaces showed an increase in alveolar macrophages, there was no alveolar edema. In contrast, the lungs of saline-treated rats had acute neutrophilic inflammation in the alveoli. In saline-treated rats, alveoli also depicted significant alveolar edema. Results from this study are shown in FIGS. 7A-C, which compares healthy (FIG. 7A), sulfur mustard exposed, ALTA-2530-treated (FIG. 7B), and sulfur mustard exposed, saline-treated (FIG. 7C) lung samples.

Example 5. Efficacy of ALTA-2530 in Lipopolysaccharide Modeled Disease

[0114] Lipopolysaccharide (LPS) is a major component of the outer membrane of Gramnegative bacteria. LPS binds to receptors present on the surface of several types of immune cells to promote an acute inflammatory response. Inhaled LPS serves as a model of acute neutrophilic airway inflammation (Ole Janssen et al., Low-dose endotoxin inhalation in healthy volunteers - a challenge model for early clinical drug development, BMC Pulm. Med. 13: 19 (2013)). The ability of ALTA-2530 to reduce neutrophilic airway inflammation was assessed in a non-human primate inhaled LPS model. The effect of ALTA-2530 treatment on the primary, secondary, and exploratory endpoints is shown in FIG. 8.

[0115] In this study, cynomolgus monkeys were administered vehicle or ALTA-2530 one hour prior to exposure to and inhalation of LPS via mask. Treatment with low-dose ALTA- 2530 reduced the percent neutrophil infiltration in the BALF by 32.7% at 4 hours post-LPS administration. High-dose ALTA-2530 decreased the percent neutrophil infiltration by 36.2% at 24 hours post-LPS administration. Treatment with low-dose ALTA-2350 resulted in a median change in percent neutrophil infiltration in the BALF, as compared to the vehicle control, of -28.01% at 4 hours post-LPS administration and -7.80% at 24 hours post-LPS administration. Treatment with high-dose ALTA-2530 resulted in a median change in percent neutrophil infiltration in the BALF, as compared to the vehicle control, of -23.25% at 4 hours post-LPS administration and -40.46% at 24 hours post-LPS administration. Results from this study, demonstrating reduction of the percent of neutrophil infiltration in the BALF, are shown in FIGS. 9A-B.

[0116] This study also demonstrated that exposure to lipopolysaccharide increased the total cell count by up to four times in the BALF, and that treatment with both low-dose and high-dose ALTA-2530 reduced the total cell count at 4 hours and at 24 hours after lipopolysaccharide administration. Results from this study, demonstrating reduction of the total cell count in the BALF, are shown in FIGS. 10A-B.

[0117] This study also demonstrated that treatment with both low-dose and high-dose ALTA-2530 reduced the absolute neutrophil counts in the BALF at 4 hours and at 24 hours after lipopolysaccharide administration. Treatment with low-dose ALTA-2350 resulted in a median reduction in neutrophil counts in the BALF, as compared to the vehicle control, of 19.76 x 10 4 cells/mL at 4 hours post-LPS administration and 40.47 x 10 4 cells/mL at 24 hours post-LPS administration. Treatment with high-dose ALTA-2530 resulted in a median reduction in neutrophil counts in the BALF, as compared to the vehicle control, of 4.08 x 10 4 cells/mL at 4 hours post-LPS administration and 43.52 x 10 4 cells/mL at 24 hours post-LPS administration. Results from this study, demonstrating reduction of the absolute neutrophil count in the BALF, are shown in FIGS. 11 A-B.

[0118] This study also demonstrated that exposure to lipopolysaccharide significantly increases IL-ip levels in BALF and that IL-1R antagonism by treatment with both low-dose and high-dose ALTA-2530 attenuated the IL-ip levels in the BALF at 4 hours and at 24 hours after lipopolysaccharide administration. Treatment with low-dose ALTA-2350 resulted in a median change in percent IL-ip in the BALF, as compared to the vehicle control, of -37.81% at 4 hours post-LPS administration. Treatment with high-dose ALTA-2530 resulted in a median change in percent IL-ip in the BALF, as compared to the vehicle control, of -57.59% at 4 hours post-LPS administration. Results from this study, demonstrating the reduction in IL-ip levels in BALF, are shown in FIGS. 12A-B.

[0119] This study also demonstrated that exposure to lipopolysaccharide significantly increases IL-6 levels in BALF and that IL-1R antagonism by treatment with both low-dose and high-dose ALTA-2530 attenuated the IL-6 levels in the BALF at 4 hours and at 24 hours after lipopolysaccharide administration. Treatment with low-dose ALTA-2350 resulted in a median change in percent IL-6 in the BALF, as compared to the vehicle control, of -50.00% at 4 hours post-LPS administration and -39.88% at 24 hours post-LPS administration. Treatment with high-dose ALTA-2530 resulted in a median change in percent IL-6 in the BALF, as compared to the vehicle control, of -64.28% at 4 hours post-LPS administration and -47.91% at 24 hours post-LPS administration. Results from this study, demonstrating the reduction in IL-6 levels in BALF, are shown in FIGS. 13A-B.

[0120] This study also demonstrated that IL-1R antagonism by treatment with both low- dose and high-dose ALTA-2530 attenuated the IL-6 levels in the serum at 4 hours and at 24 hours after lipopolysaccharide administration. Treatment with low-dose ALTA-2350 resulted in a median change in percent IL-6 in serum, as compared to the vehicle control, of -87.13% at 24 hours post-LPS administration. Treatment with high-dose ALTA-2530 resulted in a median change in percent IL-6 in serum, as compared to the vehicle control, of -54.38% at 4 hours post-LPS administration and -62.40% at 24 hours post-LPS administration. Results from this study, demonstrating the reduction in IL-6 levels in serum, are shown in FIGS.

14 A-B. [0121] This study also demonstrated that IL-1R antagonism by treatment with low-dose ALTA-2530 attenuated the TNF-a levels in the BALF at 4 hours after lipopolysaccharide administration. Results from this study demonstrating the reduction in TNF-a levels in BALF, are shown in FIG. 15.

[0122] This study also demonstrated that IL-1R antagonism by treatment with low-dose ALTA-2530 attenuated the IL-8 levels in the serum at 4 hours after lipopolysaccharide administration. Results from this study demonstrating the reduction in IL-8 levels in serum, are shown in FIG. 16.

[0123] The above data show that ALTA-2530 can significantly reduce neutrophilic airway inflammation. Furthermore, ALTA-2530 reduces airway neutrophils without eradicating them, which may protect from overactive neutrophilic lung damage. In particular, the above data demonstrate that ALTA-2530 inhibits IL-ip stimulated cytokine and chemokine release and that both low- and high-dose ALTA-2530 reduce neutrophilic airway inflammation (e.g., as quantified by reduction in neutrophil infiltration, IL-ip infiltration, and IL-6 levels) in an LPS model.