Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TREATING ACUTE RESPIRATORY DISTRESS
Document Type and Number:
WIPO Patent Application WO/2021/183774
Kind Code:
A1
Abstract:
Provided herein are technologies for treating one or more respiratory diseases, disorders or conditions by administering (E)-3-[2-(2-thienyl)vinyl]-1H-pyrazole.

Inventors:
PAKA SWARNALATHA (US)
GOLDBERG ITZHAK D (US)
VENKATESAN JAY R (US)
NEYLAN III (US)
ASLAM SHAKIL (US)
Application Number:
PCT/US2021/021926
Publication Date:
September 16, 2021
Filing Date:
March 11, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ANGION BIOMEDICA CORP (US)
International Classes:
C07D231/12; C07D401/06; C07D403/06
Foreign References:
US20140256780A12014-09-11
US20050004029A12005-01-06
US20130302283A12013-11-14
US7879898B12011-02-01
US20100143312A12010-06-10
Attorney, Agent or Firm:
D'AMATO, Erica M. et al. (US)
Download PDF:
Claims:
CLAIMS

1. A method of treating acute lung injury or acute respiratory distress syndrome in a subject in need thereof, the method comprising administering to the subject a composition that provides (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole.

2. A method comprising administering a composition that provides (E)-3-[2-(2- thienyl)vinyl]-lH-pyrazole to a subject suffering from or susceptible to an acute lung injury or acute respiratory distress syndrome.

3. A method comprising administering a composition that provides (E)-3-[2-(2- thienyl)vinyl]-lH-pyrazole to a subject suffering from or susceptible to pneumonia.

4. The method of any one of the preceding claims, wherein the subject is suffering from pneumonia confirmed by chest imaging.

5. The method of any one of the preceding claims, wherein the subject is suspected of having or has been diagnosed with a viral infection.

6. The method of any one of the preceding claims, wherein the subject is suspected of having or has been diagnosed with a respiratory viral infection.

7. The method of any one of the preceding claims, wherein the subject is suspected of having or has been diagnosed with COVID-19.

8. The method of any one of the preceding claims, wherein the subject is hospitalized with COVID-19 pneumonia.

9. The method of any one of the preceding claims, wherein the subject has a score of 4 or 5 on the WHO’s disease severity scale assessment 8-point Ordinal Scale (e.g., as described in Example 38).

10. The method of any one of the preceding claims, wherein the subject has a fraction of inspired oxygen (FiCte) of greater than 40%.

11. The method of any one of the preceding claims, wherein the subject is receiving or has received mechanical ventilation.

12. The method of any one of the preceding claims, wherein the subject is receiving or has received extracorporeal membrane oxygenation (ECMO).

13. The method of any one of the preceding claims, wherein the subject has impaired kidney function.

14. The method of any one of the preceding claims, wherein the subject is receiving or has received renal replacement therapy.

15. The method of any one of the preceding claims, wherein the subject is not suffering from an active malignancy and has not suffered from a solid or hematologic malignancy.

16. The method of any one of claims 1-14, wherein the subject is suffering from an active malignancy or has suffered from a solid or hematologic malignancy.

17. The method of any one of the preceding claims, wherein the subject has not been assessed for an active malignancy or a history of a solid or hematologic malignancy.

18. The method of any one of the preceding claims, wherein the subject is suffering from or susceptible to pulmonary edema, shock-associated acute lung injury, chemically induced acute lung injury, radiation-induced acute lung injury, acute lung injury associated with hemorrhagic shock, blunt trauma injury to the lung, ischemia-reperfusion lung injury, or thermally induced acute lung injury.

19. The method of any one of the preceding claims, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, increased lung function relative to a comparable reference population.

20. The method of any one of the preceding claims, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, one or more of:

(i) increased proportion of patients alive without need for mechanical ventilation and free of need for renal replacement therapy on an ongoing basis at Day 28;

(ii) reduced all-cause mortality;

(iii) increased proportion of patients not requiring mechanical ventilation at Day 28;

(iv) increased proportion of patients not requiring renal replacement therapy on an ongoing basis at Day 28;

(v) increased number of ventilator-free days through Day 28;

(vi) reduced proportion of patients requiring initiation of mechanical ventilation and/or ECMO through Day 28;

(vii) reduced proportion of patients requiring initiation of renal replacement therapy through Day 28;

(viii) reduced number of days to renal recovery in subjects who were on RRT at time of randomization;

(ix) reduced number of ICU days through Day 28;

(x) reduced score on an ordinal scale at Day 28; and

(xi) reduced number of days to hospital discharge from randomization, relative to a comparable reference population.

21. The method of any one of the preceding claims, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, increased kidney function relative to a comparable reference population.

22. The method of any one of the preceding claims, wherein the composition is administered intravenously.

23. The method of any one of the preceding claims, wherein the composition is administered in a dose of 2 mg/kg.

24. The method of any one of the preceding claims, wherein the composition is administered once daily.

25. The method of any one of the preceding claims, wherein the composition is administered once daily for four days.

26. The method of any one of the preceding claims, wherein the composition comprises: about 6 mg/mL (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole; about 20% (w/v) to about 40% (w/v) polyethylene glycol 300; about 5% (w/v) to about 15% (w/v) polysorbate 80; and one or more aqueous components selected from phosphate buffered saline and normal saline.

Description:
TREATING ACUTE RESPIRATORY DISTRESS

RELATED APPLICATIONS

[0001] This application claims priority to U.S. Provisional Appl. No. 62/988,905, filed March 12, 2020, U.S. Provisional Appl. No. 62/991,512, filed March 18, 2020, U.S. Provisional Appl. No. 63/008,256, filed April 10, 2020, U.S. Provisional Appl. No. 63/042,484, filed June 22, 2020, and U.S. Provisional Appl. No. 63/089,927, filed October 9, 2020, the entire contents of each of which are hereby incorporated by reference.

BACKGROUND

[0002] Respiratory disease affects millions of people each year. Acute lung injury (ALI) and the more severe acute respiratory distress syndrome (ARDS) represent a spectrum of lung disease characterized by the sudden onset of pulmonary edema, inflammatory cell infiltration, and impaired oxygenation. ARDS can be associated with various events and/or conditions, including infections, and can be associated with significant mortality. Incidence of ARDS is between 150,000 and 200,000 cases per year in the United States, resulting in between 40,000 and 80,000 deaths per year. Additionally, more than 200 million people worldwide are affected by asthma, and more than 60 million suffer from COPD.

SUMMARY

[0003] The present disclosure provides certain technologies for treating certain respiratory diseases, disorders and conditions, in some embodiments including acute respiratory distress syndrome (“ARDS”). Current supportive measures for patients in acute respiratory distress are often associated with adverse side effects; there thus remains a need for new therapies.

[0004] In some embodiments, the present disclosure provides methods of treating (e.g., lessening the severity of such as by delaying onset and/or reducing degree and/or frequency of one or more features of) a respiratory disease, disorder or condition (e.g., ARDS), which methods may comprise, for example administering a small molecule mimetic of hepatocyte growth factor (HGF, also known as scatter factor (SF)). In some embodiments, the present disclosure provides methods of treating COVID-19 pneumonia, such methods comprising administering a small molecule mimetic of HGF/SF. [0005] HGF/SF is a pleiotropic growth factor that stimulates cell growth, cell motility, morphogenesis, and angiogenesis (see, for example, Gazdhar, A. et al. Hum. Gene Ther. 2013 Jan;24(l): 105-16; Watanabe, M. et al. MolTher. 2005 July; 12(1)58-67; Kato, N. et al. Centr. Nerv. Syst. Agents Med. Chem.2009 Dec;9(4):300-6; Romero-Vasquez, F. et al. Biochim. Biophys. Acta 2012 Oct;1822(10):1590-9; Sala, V. et al. CellMol. Life Sci. 2011 May;58(10: 1703-17; Aharinejad, S. et al. Lancet 2004 May 8;363(9420): 1503-8; Vivekananda,

J. et al. Am. J. Physiol. Lung Cell Mol. Physiol. 2000 Feb;278(2):L382-92; Calvi, C. et al. PLoS Genet. 2013;9(2):el003228; Liu, Y. et al. Kidney Int. 1999 Feb;55(2):442-53; Ono, K. et al. Circulation 1997 Jun3;95(l l):2552-8; Nakamura, T. et al. J. Gastroenterol. Hepatol. 2011 Jane; 26 suppl 1:188-202; Wang, H. et al. Hum. Gen. Ther. 2013 Mar 4, each of which is incorporated herein by reference). Certain small molecule mimetics of HGF/SF have been shown to be useful for treating or lessening severity of a variety of diseases, disorders, and conditions.

[0006] The present disclosure provides an insight that Compound 1 can enhance oxygenation, gas exchange, and recovery in subjects suffering from or susceptible to respiratory disease(s), disorder(s) and/or condition(s) (e.g., as may be associated with and/or result from damage, injury, infection, etc.), specifically including ARDS. In some such embodiments, subjects are receiving therapy with, or experience lung function that would benefit from, extracorporeal membrane oxygenation (ECMO).

[0007] Literature reports have presented contradictory descriptions of HGF’ s impact on lung function. For example, certain reports (e.g., Vivekananda 2000) have posited that HGF may impair surfactant production. On the other hand, certain literature has demonstrated that HGF can contribute to mesenchymal stem cell’s (MSCs) ability to protect the lung (Hu, S., et al. Stem Cell Res. Ther. (2016) 7:66), that HGF induces AT2 proliferation (Mason, R.J., et al. Am. J. Respir. Cell Mol. Biol. 1994 Nov; 11(5):561-7), and that HGF may mitigate both the decline in surfactant proteins and the inhibition of AT2 cell proliferation caused by hyperoxia (Zhong, L.L., et al. J. Cent. S. Univ. Med. Sci. 2007 Nov 30; 32(6): 1051-57) Furthermore, the present disclosure appreciates that certain studies have indicated a potential role for HGF in lung repair and recovery from ALLARDS (Panganiban, R.A.M., et al., ActaPharm. Sinica 2011; 32:12-20; Yanagita, K., et al. J. Biol. Chem. 1993 Oct 5; 268(28)21212-7, Ware, L.B., et al. Am. J.

Physiol. Lung Cell Mol. Physiol. 2002 May;282(5):L924-40).As noted herein, Compound 1 is an HGF mimic. HGF has been established to activate the c-Met receptor, and to stimulate various regenerative downstream processes; it is not thought to activate FGF receptors. The present disclosure demonstrates, in a variety of respiratory (e.g., ARDS) models, that pathway activation (e.g., by Compound 1) achieves improved outcomes.

[0008] Among other things, the present disclosure demonstrates certain beneficial effects of Compound 1 relevant to respiratory diseases, disorders and conditions, and provides technologies for treating such diseases, disorders and conditions with Compound 1 therapy as described herein.

BRIEF DESCRIPTION OF THE DRAWINGS [0009] FIG. 1A is a graph of proliferation of endothelial cells treated with HGF vs. Compound 1. FIG. IB is a graph of proliferation of bronchial cells treated with HGF vs. Compound 1. FIG. 1C is a graph of proliferation of fibroblasts treated with HGF vs. Compound 1

[0010] FIG. 2A is a graph showing concomitant Compound 1 therapy reduces lung weight wet/dry ratio in a bleomycin mouse model. FIG. 2B depicts exemplary H&E stained lung tissue samples from a bleomycin mouse model. FIG. 2C is a graph showing delayed Compound 1 therapy reduces lung weight wet/dry ratio in a bleomycin mouse model.

[0011] FIG. 3A is a graph showing survival of mice in which TGFpi expression was induced. FIG. 3B is a graph showing that Compound 1 treatment decreased pulmonary cell death in a mouse model of induced TGFpi expression. FIG. 3C is a graph showing that Compound 1 treatment improved pulmonary epithelial regeneration (by PCNA) in a mouse model of induced TGF i expression.

[0012] FIG. 4A is a graph of mean lung injury score from lung tissue samples from a LPS- induced shock associated ALI mouse model. FIG. 4B depicts exemplary H&E stained lung tissue samples from a LPS-induced shock associated ALI mouse model. FIG. 4C is a graph showing that Compound 1 treatment decreased apoptotic cell death in a LPS-induced shock associated ALI mouse model.

[0013] FIG. 5A is a graph of survival in Compound 1 vs. vehicle cohorts in mice with Ch- induced lung injury. FIG. 5B is a graph of BALF protein concentration in Compound 1 vs. vehicle cohorts in mice with Ch-induced lung injury. [0014] FIG. 6A is a graph showing increased pulmonary output in Compound 1 vs. vehicle cohorts in a PPE-induced emphysema rat model. FIG. 6B is a graph showing increased arterial oxygen concentration in Compound 1 vs. vehicle cohorts in a PPE-induced emphysema rat model.

[0015] FIG. 7A is a graph of mean lung injury score from lung tissue samples in a hemorrhagic shock-induced lung injury rat model. FIG. 7B depicts exemplary H&E stained lung tissue samples from a hemorrhagic shock-induced lung injury rat model.

[0016] FIG. 8A is a graph of end expiration volume in a pulmonary ischemia-reperfusion rat model. FIG. 8B is a graph of blood pH in a pulmonary ischemia-reperfusion rat model. FIG.

8C and FIG. 8D are graphs of blood oxygen levels in a pulmonary ischemia-reperfusion rat model. FIG. 8E is a graph of pulmonary epithelial regeneration (PNCA) in a pulmonary ischemia-reperfusion rat model.

[0017] FIG. 9A is a graph of lung weight wet/dry ratio after Compound 1 vs vehicle treatment in canines with warm lung ischemia-reperfusion injury. FIG. 9B is a graph of lung function after Compound 1 vs vehicle treatment in canines with warm lung ischemia-reperfusion injury. FIG. 9C is a graph of IL-1 levels after Compound 1 vs vehicle treatment in canines with warm lung ischemia-reperfusion injury. FIG. 9D is a graph of IL-6 levels after Compound 1 vs vehicle treatment in canines with warm lung ischemia-reperfusion injury. FIG. 9E depicts exemplary H&E stained lung sections from canines with warm lung ischemia-reperfusion injury. [0018] FIG. 10 is a graph showing lung weight wet/dry ratio after delayed Compound 1 treatment vs. vehicle in a bleomycin mouse model.

[0019] FIG. 11A is a graph showing survival in mice treated with Compound 1 vs. vehicle in a murine model of pulmonary TGFp l induction. FIG. 1 IB is a graph showing that Compound 1 decreased pulmonary cell death in murine model of pulmonary TGF l induction. FIG. 11C is a graph showing that Compound 1 increased pulmonary epithelial regeneration (PCNA) in murine model of pulmonary TGFp l induction. FIG. 11D depicts exemplary micro CT images of inflated and formalin fixed lungs, which suggest that treatment with Compound 1 preserves pulmonary microarchitecture murine model of pulmonary TGF l induction.

[0020] FIG. 12A is a graph showing Compound 1 attenuated mortality in sheep subjected to smoke inhalation and burn injury. FIG. 12B is a graph showing Compound 1 improved pulmonary gas exchange in sheep subjected to smoke inhalation and bum injury. [0021] FIG. 13A is a graph showing lung hydroxyproline levels in Compound 1 vs. vehicle cohorts for a genetic (TSK1/+) mouse model of systemic sclerosis including pulmonary fibrosis. FIG. 13B is a graph showing extent of Sirius red staining in lung tissue samples from a genetic (TSK1/+) mouse model of systemic sclerosis including pulmonary fibrosis. FIG.13C depicts exemplary Sirius red stained lung tissue samples from a genetic (TSK1/+) mouse model of systemic sclerosis including pulmonary fibrosis.

[0022] FIG. 14 is a graph showing lung hydroxyproline levels in a TGFpi -induced lung fibrosis in mouse model.

[0023] FIG. 15A is a graph showing pulmonary apoptosis (determined by TUNEL) in mice exposed to ionizing radiation. FIG. 15B is a graph showing caspase 3 levels in mice exposed to ionizing radiation. FIG. 15C is a graph showing lung inflammation as judged by staining for F4/80 in mice exposed to ionizing radiation.

[0024] FIG. 16A is a graph showing lung mass in mice exposed to radiation that received Compound 1 vs vehicle. FIG. 16B is a graph showing lung collagen-1 levels in mice exposed to radiation that received Compound 1 vs vehicle. FIG. 16C is a graph showing extent of Sirius red staining in lung tissue samples from mice exposed to radiation that received Compound 1 vs vehicle. FIG. 16D is a graph showing lung TGFpi levels in mice exposed to radiation that received Compound 1 vs vehicle.

[0025] FIG. 17A is a graph showing kidney TGFfFl levels in mice exposed to radiation that received Compound 1 vs vehicle. FIG. 17B is a graph showing liver TGFpi levels in mice exposed to radiation that received Compound 1 vs vehicle. FIG. 17C is a graph showing liver aSMA levels in mice exposed to radiation that received Compound 1 vs vehicle.

[0026] FIG. 18A is a graph showing arterial Pa02 for rats treated with Compound 1 vs vehicle in a (PPE)-induced emphysema rat model. FIG. 18B is a graph showing arterial PaC02 for rats treated with Compound 1 vs vehicle in a (PPE)-induced emphysema rat model.

[0027] FIG. 19A is a graph showing pulmonary apoptosis (caspase 3 staining) at 24 hr reperfusion in rats with ischemia-reperfusion injury. FIG. 19B depicts exemplary H&E stained lung tissue samples from rats with ischemia-reperfusion injury.

[0028] FIG. 20A is a graph showing end-expiration air volume following pulmonary 90 min ischemia-reperfusion in rats treated with Compound 1 vs vehicle. FIG. 20B is a graph showing blood pH prior to sacrifice in rats with ischemia-reperfusion injury treated with Compound 1 vs vehicle. FIG. 20C is a graph showing blood oxygen tension (p02) prior to sacrifice in rats with ischemia-reperfusion injury treated with Compound 1 vs vehicle. FIG. 20D is a graph showing blood oxygen saturation (s02) prior to sacrifice in rats with ischemia-reperfusion injury treated with Compound 1 vs vehicle. FIG. 20E is a graph showing pulmonary epithelial regeneration (PCNA immunoreactivity) in rats with ischemia-reperfusion injury treated with Compound 1 vs vehicle. FIG. 20F is a graph showing pulmonary cell death in rats with ischemia-reperfusion injury treated with Compound 1 vs vehicle. FIG. 20G depicts exemplary lung tissue samples from rats with ischemia-reperfusion injury treated with Compound 1 vs vehicle.

[0029] FIG. 21A depicts exemplary scans of rats with syngeneic lung transplantation injury that received Compound 1 vs vehicle. FIG. 21B is a graph showing blood pH in rats with syngeneic lung transplantation injury that received Compound 1 vs vehicle. FIG. 21C is a graph showing pC02 in rats with syngeneic lung transplantation injury that received Compound 1 vs vehicle. FIG. 21D is a graph showing p02 in rats with syngeneic lung transplantation injury that received Compound 1 vs vehicle. FIG. 21E is a graph showing s02 in rats with syngeneic lung transplantation injury that received Compound 1 vs vehicle. FIG. 21F depicts exemplary H&E stained lung tissue samples from rats with syngeneic lung transplantation injury that received Compound 1 vs vehicle.

[0030] FIG. 22A is a graph showing survival in sheep subjected to smoke inhalation and 40% body surface burn injury treated with Compound 1 vs vehicle. FIG. 22B is a graph showing pulmonary gas exchange in sheep subjected to smoke inhalation and 40% body surface burn injury treated with Compound 1 vs vehicle. FIG. 22C is a graph showing cumulative fluid balance in sheep subjected to smoke inhalation and 40% body surface burn injury treated with Compound 1 vs vehicle. FIG. 22D depicts exemplary H&E stained lung tissue samples from sheep subjected to smoke inhalation and 40% body surface bum injury treated with Compound 1 vs vehicle.

[0031] FIG. 23A is a graph showing lung weight wet/dry ratio in mice treated with Compound 1 vs vehicle in a bleomycin model. FIG. 23B depicts exemplary H&E stained lung tissue samples from mice treated with Compound 1 vs. vehicle in a bleomycin mouse model. [0032] FIG. 24 is a graph showing lung weight wet/dry ratio in mice treated with Compound 1 vs vehicle 24 hours after administration of bleomycin in a bleomycin model. [0033] FIG. 25A is a graph showing pulmonary hydroxyproline levels in mice treated with Compound 1 vs vehicle in a mouse model of pulmonary fibrosis. FIG. 25B is a graph showing extent of pulmonary Sirius red staining in mice treated with Compound 1 vs vehicle in a mouse model of pulmonary fibrosis.

[0034] FIG. 26A is a graph showing endothelial (HUVEC) cell proliferation stimulated by HGF/SF or Compound 1. FIG. 26B is a graph showing MRC-5 proliferation in the presence of HGF/SF or Compound 1.

[0035] FIG. 27 is graph showing human bronchial epithelial cell (HBEC) proliferation stimulated by HGF/SF or Compound 1.

[0036] FIG. 28A is a graph showing knockdown of c-Met mRNA levels in bovine pulmonary endothelial cells (bPAEC). C = control. FIG. 28B is a graph showing proliferation of knockdown bPAECs treated with Compound 1. FIG. 28C is a graph showing proliferation of knockdown bPAECs treated with HGF/SF.

[0037] FIG. 29 is a graph showing bPAEC migration when treated with Compound 1 or HGF/SF.

[0038] FIG. 30 depicts exemplary human bronchial epithelial cells stained with FITC- labeled Annexin-V, treated with either Compound 1 or vehicle.

[0039] FIG. 31A is a graph showing expiration volume of rats treated with Compound 1 (15 mg/kg or 45 mg/kg) vs vehicle in a rodent model of elastase-induced emphysema. FIG. 31B is a graph showing arterial oxygen concentration of rats treated with Compound 1 (15 mg/kg or 45 mg/kg) vs vehicle in a rodent model of elastase-induced emphysema. FIG. 31C is a graph showing mean linear intercept of rats treated with Compound 1 (15 mg/kg or 45 mg/kg) vs vehicle in a rodent model of elastase-induced emphysema. FIG. 31D exemplary lung tissue samples stained with H&E obtained from rats treated with Compound 1 (15 mg/kg or 45 mg/kg) vs vehicle in a rodent model of elastase-induced emphysema.

[0040] FIG. 32A is a graph showing serum creatinine levels in dogs subjected to renal ischemia-reperfusion and treated with immediate or delayed Compound 1. FIG. 32B is a graph showing BUN levels in dogs subjected to renal ischemia-reperfusion and treated with immediate or delayed Compound 1. [0041] FIG. 33A is a graph showing percentage of FhC -exposed Annexin-V positive cells treated with Compound 1 vs vehicle. FIG. 33B is a graph showing percentage of Annexin-V and Propidium Iodide negative (healthy live) cells treated with Compound 1 vs vehicle.

[0042] FIG. 34 is a graph showing Annexin-V and propidium iodide staining of 4MBr-5 cells challenged with H2O2 and treated with Compound 1 or HGF.

[0043] FIG. 35 is a graph showing mouse NIH/3T3 cell proliferation when exposed to Compound 1 or HGF.

[0044] FIG. 36A shows survival of mice exposed to radiation that were treated with Compound 1 vs vehicle. FIG. 36B is a graph showing BALF turbidity in radiation-exposed mice that were treated with Compound 1 vs vehicle.

[0045] FIG. 37A is a graph showing PCNA staining in lung tissue samples from mice in a TGFbl-induced acute lung injury model. (NS = not significant; p-values vs T GF(31 vehicle group). FIG. 37B is a graph showing caspase-3 staining in lung tissue samples from mice in a TGFpi-induced acute lung injury model. (NS = not significant; p-values vs TGFpi vehicle group).

[0046] FIG. 38 is a Kaplan-Meier survival graph from a TGFpi -induced acute lung injury mouse model.

[0047] FIG. 39A shows percent survival of Compound 1 vs vehicle treatment in a first study of an orthotopic glioma model. FIG. 39B shows percent survival of Compound 1 vs vehicle treatment in a second study of an orthotopic glioma model.

[0048] FIG. 40A shows colon tumor size of Compound 1 vs vehicle treatment in a human colon tumor xenograft model. FIG. 40B shows colon tumor weight of Compound 1 vs vehicle treatment in a human colon tumor xenograft model (ns = not significant).

[0049] FIG. 41A shows pancreatic tumor volume of Compound 1 vs vehicle treatment in a human pancreatic tumor xenograft model. FIG. 41B shows pancreatic tumor weight of Compound 1 vs vehicle treatment in a human pancreatic tumor xenograft model.

[0050] FIG. 42 provides XRPD pattern of Compound 1 Lot I.

[0051] FIG. 43 provides TGA curve of Compound 1 Lot I.

[0052] FIG. 44 provides DSC thermogram of Compound 1 Lot I.

[0053] FIG. 45 provides single crystal X-ray crystallography of Compound 1 Form A. N and S atoms are labeled; unlabeled non-hydrogen atoms are carbon. [0054] FIG. 46 provides XRPD pattern of Compound 1 Form A calculated from single crystal X-ray diffraction data.

[0055] FIG. 47 provides XRPD pattern of Compound 1 Form A.

[0056] FIG. 48 provides TGA curve of Compound 1 Form A.

[0057] FIG. 49 provides DSC thermogram of Compound 1 Form A.

[0058] FIG. 50 provides a comparison of XRPD patterns of Compound 1 Lot I and Compound 1 Single Crystal Form A.

DETAILED DESCRIPTION

Definitions

[0059] The term “about”, when used herein in reference to a value, refers to a value that is similar, in context to the referenced value. In general, those skilled in the art, familiar with the context, will appreciate the relevant degree of variance encompassed by “about” in that context. For example, in some embodiments, the term “about” may encompass a range of values that is within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less of the referred value.

[0060] As used herein, the term “administering” or “administration” typically refers to the administration of a composition to a subject to achieve delivery of an active agent to a site of interest (e.g., a target site which may, in some embodiments, be a site of disease or damage, and/or a site of responsive processes, cells, tissues, etc.) As will be understood by those skilled in the art, reading the present disclosure, in some embodiments, one or more particular routes of administration may be feasible and/or useful in the practice of the present disclosure. For example, in some embodiments, administration may be parenteral. In some embodiments, administration may be oral. In some embodiments, administration may involve only a single dose. In some embodiments, administration may involve application of a fixed number of doses. In some embodiments, administration may involve dosing that is intermittent (e.g., a plurality of doses separated in time) and/or periodic (e.g., individual doses separated by a common period of time) dosing. In some embodiments, administration may involve continuous dosing (e.g., perfusion) for at least a selected period of time. As described herein, in many embodiments, administration is parenteral, e.g., via intravenous (IV) administration, which in some embodiments may be or comprise IV perfusion); in some embodiments, one or more instances of perfusion may be performed. In some embodiments, amount perfused and/or rate of perfusion may be selected, for example, in light of a characteristic such as subject weight, age, presence and/or extent of one or more relevant symptom(s), timing relative to transplant procedure, etc. [0061] As used herein, the term “comparable” refers to two or more agents, entities, situations, sets of conditions, circumstances, individuals, or populations, etc., that may not be identical to one another but that are sufficiently similar to permit comparison there between so that one skilled in the art will appreciate that conclusions may reasonably be drawn based on differences or similarities observed. In some embodiments, comparable agents, entities, situations, sets of conditions, circumstances, individuals, or populations are characterized by a plurality of substantially identical features and one or a small number of varied features. Those of ordinary skill in the art will understand, in context, what degree of identity is required in any given circumstance for two or more such agents, entities, situations, sets of conditions, circumstances, individuals, or populations, etc. to be considered comparable. For example, those of ordinary skill in the art will appreciate that sets of circumstances, agents, entities, situations, individuals, or populations are comparable to one another when characterized by a sufficient number and type of substantially identical features to warrant a reasonable conclusion that differences in results obtained or phenomena observed under or with different agents, entities, situations sets of circumstances, individuals, or populations are caused by or indicative of the variation in those features that are varied.

[0062] As used herein, the term “pharmaceutical composition” refers to a composition comprising a pharmaceutical active (which may be, comprise, or otherwise become an active agent upon administration of the composition), formulated together with one or more pharmaceutically acceptable carriers. In some embodiments, a pharmaceutical composition is or comprises a pharmaceutical active present in unit dose amount appropriate for administration in a therapeutic regimen that shows a statistically significant probability of achieving a predetermined therapeutic effect when administered to a relevant population. In some embodiments, pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin, lungs, or oral cavity; intravaginally or intrarectally, for example, as a pessary, cream, or foam; sublingually; ocularly; transdermally; or nasally, pulmonary, and to other mucosal surfaces. In some embodiments, as described herein, a pharmaceutical composition is formulated for parenteral administration {e.g., for IV administration such as by infusion).

[0063] The term “pharmaceutically acceptable salt form,” as used herein, refers to a form of a relevant compound as a salt appropriate for use in pharmaceutical contexts, i.e., salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and/or lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977).

[0064] As used herein, the term “reference” describes a standard or control relative to which a comparison is performed. For example, in some embodiments, an agent, individual, population, sample, sequence or value of interest is compared with a reference or control agent, individual, population, sample, sequence or value. In some embodiments, a reference or control is tested and/or determined substantially simultaneously with the testing or determination of interest. In some embodiments, a reference or control is a historical reference or control, optionally embodied in a tangible medium. Typically, as would be understood by those skilled in the art, a reference or control is determined or characterized under comparable conditions or circumstances to those under assessment. Those skilled in the art will appreciate when sufficient similarities are present to justify reliance on and/or comparison to a particular possible reference or control.

[0065] As will be understood from context, “risk” of a disease, disorder, and/or condition refers to a likelihood that a particular individual will develop the disease, disorder, and/or condition. In some embodiments, risk is expressed as a percentage. In some embodiments, risk is from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90 up to 100%. In some embodiments risk is expressed as a risk relative to a risk associated with a reference sample or group of reference samples. In some embodiments, a reference sample or group of reference samples have a known risk of a disease, disorder, condition and/or event. In some embodiments a reference sample or group of reference samples are from individuals comparable to a particular individual. In some embodiments, relative risk is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more.

[0066] As used herein, the term “subject” refers an organism, typically a mammal (e g., a human). In some embodiments, a subject is suffering from a relevant disease, disorder or condition. In some embodiments, a human subject is an adult, adolescent, or pediatric subject.

In some embodiments, a subject is at risk of (e g., susceptible to), e.g., at elevated risk of relative to an appropriate control individual or population thereof, a disease, disorder, or condition. In some embodiments, a subject displays one or more symptoms or characteristics of a disease, disorder or condition. In some embodiments, a subject does not display any symptom or characteristic of a disease, disorder, or condition. In some embodiments, a subject is someone with one or more features characteristic of susceptibility to or risk of a disease, disorder, or condition. In some embodiments, a subject is an individual to whom diagnosis and/or therapy and/or prophylaxis is and/or has been administered. The terms “subject” and “patient” are used interchangeably herein.

Hepatocyte Growth Factor Mimetics

[0067] PCT Application No. PCT/US2003/040917, fried December 19, 2003 and published as W02004/058721 on July 15, 2004, the entirety of which is hereby incorporated by reference, describes certain compounds that act as HGF/SF mimetics. Such compounds include Compound 1: or a pharmaceutically acceptable salt thereof (i.e., Compound 1 in a pharmaceutically acceptable salt form). Compound 1 is useful in methods provided herein, e.g., for treating acute lung injury and/or acute respiratory distress syndrome (ARDS).

[0068] Compound 1 has a CAS Registry No. of 1070881-42-3 and is also known by at least the following names:

• Terevalefim; • 3-[(lE)-2-(thiophen-2-yl)ethen-l-yl]-lH-pyrazole; and

• (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole.

[0069] A synthesis of Compound 1, (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole, is described in detail in Example 7 of W02004/058721, as well as in Example 1 herein. An alternative synthesis of Compound 1 is provided in Example 47 herein.

[0070] Those skilled in the art will appreciate that Compound 1 has a structure that can exist in various tautomeric forms, including (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole and (E)-5-[2-(2- thienyl)vinyl]-lH-pyrazole, or any mixture thereof. Moreover, those skilled in the art, reading the present disclosure will appreciate that, in many embodiments, teachings described herein are not limited to any particular tautomeric form. Accordingly, in some embodiments, Compound 1 may be referred to as (E)-3(5)-[2-(2-thienyl)vinyl]-lH-pyrazole. The present disclosure contemplates use of all tautomeric forms of Compound 1.

[0071] Compound 1 has been demonstrated to be remarkably useful for treatment of a variety of conditions including, for example, fibrotic liver disease, ischemia-reperfusion injury, cerebral infarction, ischemic heart disease, renal disease, lung fibrosis, damaged and/or ischemic organs, transplants or grafts, stroke, cerebrovascular disease, and renal fibrosis, among others (see, for example, WO 2004/058721, WO 2010/005580, US 2011/0230407, US 7879898, WO 2009/064422, the entirety of each of which is hereby incorporated by reference). In particular, Compound 1 is or has been the subject of clinical trials for delayed graft function in recipients of a deceased donor kidney (Clinicaltrials.gov identifier: NCT02474667), as well as acute kidney injury after cardiac surgery involving cardiopulmonary bypass (Clinicaltrials.gov identifier: NCT02771509) and COVID-19 pneumonia (Clinicaltrials.gov identifier: NCT04459676). Compound 1 has also been demonstrated to mitigate post-ischemic kidney injury ( see Narayan, P., et al. Am. J. Physiol. Renal Physiol. 311 :F352-F361, 2016). Without wishing to be bound by any particular theory, it is believed that Compound 1 ’ s HGF mimic capability imparts a variety of beneficial attributes and activities. Thus, certain subjects may receive additional benefit (e.g., improved kidney function) from therapy as described herein.

[0072] In some embodiments, Compound 1 is provided and/or utilized (e.g., for inclusion in a composition and/or for delivery to a subject) in accordance with the present disclosure in a form such as a salt form. As already noted herein, pharmaceutically acceptable salts are well known in the art. [0073] In some embodiments, Compound 1 is provided and/or utilized (e.g., for inclusion in (e.g., during one or more steps of manufacturing of) a composition and/or for delivery to a subject) in accordance with the present disclosure in a form such as a solid form. Certain solid forms of Compound 1 are described in PCT Application No. PCT7US2020/027710, filed April 10, 2020 and published as WO 2020/210657 on October 15, 2020, the entirety of each of which is hereby incorporated by reference. In some embodiments, Compound 1 is provided and/or utilized in accordance with the present disclosure in an amorphous solid form, in a crystalline solid form, or in a mixture thereof. In some embodiments, a composition is substantially free of amorphous Compound 1. As used herein, the term “substantially free” means lacking a significant amount (e.g., less than about 10%, less than about 5%, less than about 3%, less than about 2%, or less than about 1%). In some embodiments, a composition comprises at least about 90% by weight of crystalline Compound 1. In some embodiments, a composition comprises at least about 95% by weight of crystalline Compound 1. In some embodiments, a composition comprises at least about 97%, about 98%, or about 99% by weight of crystalline Compound 1.

In some embodiments, a crystalline solid form may be or comprise a solvate, hydrate, or an unsolvated form. The use of any and all such forms are contemplated by the present disclosure. [0074] In some embodiments, a crystalline solid form of Compound 1 is Form A. In some embodiments, Form A of Compound 1 is unsolvated (e.g., anhydrous). In some embodiments, a Form A of Compound 1 is prepared according to the procedure described in Example 48.

[0075] In some embodiments, Form A is characterized by one or more peaks in its XRPD pattern selected from those at about 8.64, about 11.04, about 17.34, about 25.06, and about 25.70 degrees 2-theta. In some embodiments, Form A is characterized by two or more peaks in its XRPD pattern selected from those at about 8.64, about 11.04, about 17.34, about 25.06, and about 25.70 degrees 2-theta. In some embodiments, Form A is characterized by three or more peaks in its XRPD pattern selected from those at about 8.64, about 11.04, about 17.34, about 25.06, and about 25.70 degrees 2-theta.

[0076] In some embodiments, Form A is characterized by peaks in its XRPD pattern at about 8.64, about 11.04, about 17.34, about 25.06, and about 25.70 degrees 2-theta. In some embodiments, Form A is characterized by peaks in its XRPD pattern at about 8.64, about 11.04, about 17.34, about 25.06, and about 25.70 degrees 2-theta, corresponding to d-spacing of about 10.22, about 8.01, about 5.11, about 3.55, and about 3.46 angstroms. [0077] In some embodiments, Form A is characterized by substantially all of the peaks (degrees 2-theta) in its XRPD pattern, optionally corresponding to d-spacing (angstroms), at about:

[0078] In some embodiments, Form A is characterized by one or more of the following:

(i) an XRPD pattern substantially similar to that depicted in FIG. 46 and/or FIG. 47;

(ii) a TGA pattern substantially similar to that depicted in FIG. 48;

(iii) a DSC pattern substantially similar to that depicted in FIG. 49; and

(iv) a melting point of about 116.42 °C.

[0079] As used herein, the term “about” when used in reference to a degree 2-theta value refers to the stated value ± 0.2 degree 2-theta. In some embodiments, “about” refers to the stated value ± 0.1 degree 2-theta.

[0080] Unless otherwise indicated, as used herein “Compound 1” refers to (E)-3-[2-(2- thienyl)vinyl]-lH-pyrazole in any available form, such as, e.g., a tautomer, salt form, and/or solid form thereof.

[0081] Certain liquid (e.g., for intravenous or intraperitoneal administration) and solid (e.g., for oral administration) formulations of Compound 1 have been described. See, for example, PCT Application No. PCT/US2009/004014, filed July 9, 2009 and published as W02010/005580 on January 14, 2010, the entirety of which is hereby incorporated by reference. [0082] In some embodiments, Compound 1 is provided and/or utilized in accordance with the present disclosure as a liquid formulation. In some embodiments, a liquid formulation comprises Compound 1 in a concentration of from about 0.8 mg/mL to about 10 mg/mL.

[0083] In some embodiments, a liquid formulation comprising Compound 1 further comprises polyethylene glycol (e.g., polyethylene glycol 300). In some embodiments, a liquid formulation comprises from about 40% (w/v) to about 60% (w/v) polyethylene glycol (e.g., polyethylene glycol 300). In some embodiments, a liquid formulation comprises about 50% (w/v) polyethylene glycol (e.g., polyethylene glycol 300).

[0084] In some embodiments, a liquid formulation comprising Compound 1 further comprises polysorbate (e.g., polysorbate 80). In some embodiments, a liquid formulation comprises from about 5% (w/v) to about 15% (w/v) polysorbate (e.g., polysorbate 80). In some embodiments, a liquid formulation comprises about 10% (w/v) polysorbate (e.g., polysorbate 80). [0085] In some embodiments, a liquid formulation comprising Compound 1 is aqueous. In some embodiments, a liquid formulation comprising Compound 1 further comprises saline solution, buffer, or buffered saline solution (e.g., phosphate-buffered saline).

[0086] In some embodiments, a liquid formulation comprises Compound 1 and further comprises about 50% (w/v) polyethylene glycol (e.g., polyethylene glycol 300) and about 10% (w/v) polysorbate (e.g., polysorbate 80). In some such embodiments, the liquid formulation is aqueous. In some such embodiments, the liquid formulation further comprises phosphate- buffered saline and/or normal saline.

[0087] In some embodiments, Compound 1 is provided and/or utilized in accordance with the present disclosure as a formulation comprising: about 6 mg/mL to about 10 mg/mL Compound 1; about 20% (w/v) to about 60% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 5% (w/v) to about 15% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline.

[0088] In some embodiments, Compound 1 is provided and/or utilized in accordance with the present disclosure as a formulation comprising: about 10 mg/mL Compound 1; about 50% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 10% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline.

[0089] In some embodiments, Compound 1 is provided and/or utilized in accordance with the present disclosure as a formulation comprising: about 10 mg/mL Compound 1; about 40% (w/v) to about 60% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 5% (w/v) to about 15% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline.

[0090] In some embodiments, Compound 1 is provided and/or utilized in accordance with the present disclosure as a formulation comprising: about 10 mg/mL Compound 1; about 50% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 10% (w/v) polysorbate (e.g., polysorbate 80); and about 40% (w/v) phosphate buffered saline.

[0091] In some embodiments, Compound 1 is provided and/or utilized in accordance with the present disclosure as a formulation comprising: about 6 mg/mL Compound 1; about 20% (w/v) to about 40% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 5% (w/v) to about 15% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline.

[0092] In some embodiments, Compound 1 is provided and/or utilized in accordance with the present disclosure as a formulation comprising: about 6 mg/mL Compound 1; about 30% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 6% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline.

[0093] In some embodiments, Compound 1 is provided and/or utilized in accordance with the present disclosure as a formulation comprising: about 6 mg/mL Compound 1; about 30% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 6% (w/v) polysorbate (e.g., polysorbate 80); about 24% (w/v) phosphate buffered saline; and about 40% (w/v) normal saline.

[0094] In some embodiments, liquid formulations of Compound 1 provided herein are prepared by a process comprising a step of combining: crystalline Compound 1 (e.g., Form A); polyethylene glycol (e.g., polyethylene glycol 300); polysorbate (e.g., polysorbate 80); and one or more aqueous components (e.g., phosphate buffered saline and/or normal saline) to obtain the formulation.

[0095] In some embodiments, a liquid formulation has a pH of about 5 to about 9. In some embodiments, a liquid formulation has a pH of about 6 to about 8. In some embodiments, a liquid formulation has a pH of about 7 (e.g., about 7.4). In some embodiments, a liquid formulation has a pH of about 6.4 to about 8.4 or about 7.4 to about 7.9.

[0096] In some embodiments, a liquid formulation is suitable for intravenous administration. In some embodiments, a liquid formulation is suitable for intravenous administration over about 10 min, about 20 min, about 30 min, or about 40 min. In some embodiments, a liquid formulation is suitable for intravenous administration of about 0.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 6 mg/kg, or about 8 mg/kg Compound 1.

Provided Methods

[0097] Provided herein are methods of treating a subject or a population of subjects comprising administering Compound 1 (e.g., by administering a composition that comprises and/or delivers Compound 1 as described herein) to the subject(s) in need thereof. In some embodiments, such administering is achieved by administering a composition that delivers Compound 1 (e.g., in some embodiments, a composition that is or comprises Compound 1, or a composition that otherwise delivers Compound 1 - e.g., that is or comprises a prodrug of Compound 1, a complex or other entity that releases Compound 1 upon administration, etc.). [0098] In some embodiments, the present disclosure provides methods of treating a respiratory disease, disorder or condition comprising administering to a subject or a population susceptible to or suffering from a respiratory disease, disorder or condition a composition that provides Compound 1.In some embodiments, the present disclosure provides methods of treating acute lung injury or acute respiratory distress syndrome in a subject or population in need thereof comprising administering to the subject a composition that provides Compound 1. In some embodiments, the present disclosure provides methods of treating acute lung injury in a subject or population in need thereof comprising administering to the subject a composition that provides Compound 1. In some embodiments, the present disclosure provides methods of treating acute respiratory distress syndrome in a subject or population in need thereof comprising administering to the subject a composition that provides Compound 1. [0099] Acute lung injury and the more severe acute respiratory distress syndrome (ARDS) represent a spectrum of lung disease characterized by the sudden onset of pulmonary edema, inflammatory cell infiltration and impaired oxygenation. Current treatment strategies for severe ARDS include mechanical ventilation which, while potentially life-saving, can exacerbate lung injury, and antibiotics, which are the standard of care under certain treatment guidelines. Antibiotics are generally given prophylactically to prevent secondary infection related to ARDS as opposed to treating the ARDS itself. There are no approved pharmacologic options for ARDS. Common causes of ALI and ARDS include aspiration pneumonia, viral or bacterial pneumonia, sepsis, inhalational injury (e.g., from smoke or chemicals), trauma, and blood transfusions.

[0100] ARDS is generally defined by the 2012 ARDS Task Force “Berlin” definition. Key components of the Berlin definition are acute hypoxemia in ventilated patients receiving certain levels of positive end expiratory pressure and demonstration of non-cardiogenic bilateral opacities on imaging studies, with severity graded into mild, moderate, and severe ARDS, based on the PaCk/FiO ratio.

[0101] In some embodiments, the present disclosure provides methods comprising administering to a subject or population who is suffering from or susceptible to a respiratory disorder a composition that provides Compound 1. In some embodiments, a subject or population is suffering from or susceptible to acute lung injury. In some embodiments, a subject or population is suffering from or susceptible to acute respiratory distress syndrome.

[0102] In some embodiments, the present disclosure provides methods of administering Compound 1 to a population of subjects who are suffering from or susceptible to a respiratory disease, disorder or condition as described herein, for example by administering a composition that provides Compound 1, e.g., according to a dosing regimen described herein. In some embodiments, a population is a population of subjects who are suffering from or susceptible to acute lung injury. In some embodiments, a population is a population of subjects who are suffering from or susceptible to acute respiratory distress syndrome.

[0103] In some embodiments, the present disclosure provides methods of administering Compound 1 to a population of subjects who are suffering from or susceptible to a respiratory disease, disorder or condition as described herein, for example by administering a composition that provides Compound 1, e.g., according to a regimen established to achieve one or more desirable outcomes. In some embodiments, a population is a population of subjects who are suffering from or susceptible to acute lung injury. In some embodiments, a population is a population of subjects who are suffering from or susceptible to acute respiratory distress syndrome. In some embodiments, a regimen is or has been established to achieve one or more desirable outcomes in a population to which Compound 1 has been administered, relative to a comparable reference population that has not received Compound 1 (e g., that has received a reference composition which does not deliver Compound 1).

[0104] In some embodiments, in methods provided herein, a reference population has not received a composition that provides Compound 1. In some embodiments, in methods provided herein, a reference population has received an otherwise comparable reference composition that does not provide Compound 1 (e.g., a placebo, such as normal saline). In some embodiments, in methods provided herein, a reference composition may be or comprise normal saline. In some embodiments, in methods provided herein, a reference composition may be or may have been administered at the same intervals and/or volumes as a composition that provides Compound 1. [0105] In some embodiments, certain parameters may be evaluated to determine if a desirable outcome is achieved. Any one or more of parameters such as these may, in some embodiments, be useful for determining short-term and/or long-term efficacy of Compound 1 administered to the patient population. Alternatively or additionally, any one or more of such parameters may be assessed to monitor patient response to Compound 1 therapy.

[0106] In some embodiments, the present disclosure provides a method comprising: administering to a subject, or to a population of subjects, suffering from or susceptible to a respiratory disease, disorder or condition as described herein a composition that provides Compound 1 according to a regimen established to achieve one or more of: decreased lung injury score, reduced mortality, reduced lung histopathology, better cytokine evaluation, better lung MPO measurement, improved pulmonary function, reduced lung obstruction in bronchus and bronchioles, and better cardiopulmonary variables, relative to a comparable reference population.

[0107] In some embodiments, the present disclosure provides methods of improving outcomes for patients suffering from or susceptible to a disease, disorder or condition that is (i.e., is statistically and/or is in fact for the particular patient) associated with one or more undesirable respiratory features such as for example inflammation in the lungs, fluid in the lungs, fibroids in the lungs, etc., by administration of Compound 1 (e.g., alone and/or in combination with other therapy, for example directed at the underlying disease, disorder or condition, or otherwise being used in treatment of the patient(s)).

[0108] In some embodiments, the disease, disorder or condition being treated in methods provided herein is characterized by pulmonary edema, pulmonary epithelial cell apoptosis, inflammatory cell infdtration, impaired oxygenation, hypoxemia and/or lung fibrosis.

[0109] In some embodiments, a composition that provides Compound 1 is administered according to a regimen established to achieve a particular effect, e.g., at a particular time point (e.g., about 7 days, about 14 days, about 28 days, about 6 months and/or about 12 months after injury and/or randomization and/or first administration of Compound 1). In some embodiments, a composition that provides Compound 1 is administered according to a regimen established to achieve a particular effect, e.g., at a particular time point, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1, which comparable population may, in some embodiments, have received a reference composition that is otherwise comparable but does not provide Compound 1 upon administration).

[0110] In some embodiments, a particular effect may be or comprise: decreased lung injury score, reduced mortality, reduced lung histopathology, better cytokine evaluation, better lung MPO measurement, improved pulmonary function, reduce lung obstruction in bronchus and bronchioles, and/or better cardiopulmonary variables.

[0111] In some embodiments, a particular effect may be or comprise decreased mortality rate relative to a comparable reference population.

[0112] In some embodiments, a particular effect may be or comprise increased lung function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise one or more of: increased pulmonary output, increased arterial oxygen (PaC ), decreased arterial carbon dioxide (PaCCh), increased ratio of PaCh/FiCh, decreased lung injury score, decreased lung hydroxyproline concentration, decreased lung collagen level, decreased lung TGFp l concentration, and increased blood pH, relative to a comparable reference population.

[0113] In some embodiments, a particular effect may be or comprise increased kidney function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise less deterioration of kidney function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise one or more of: decreased serum creatinine concentration, increased estimated glomerular fdtration rate (eGFR), decreased blood urea nitrogen concentration, increased urine output, decreased kidney TGFp l concentration, and lesser incidence of dialysis, relative to a comparable reference population. In some embodiments, a particular effect may be or comprise one or more of: decreased serum creatinine concentration, reduced change in serum creatinine concentration (e g., over a particular period of time), increased estimated glomerular filtration rate (eGFR), reduced change in eGFR (e.g., over a particular period of time), increased measured glomerular filtration rate, reduced change in measured glomerular filtration rate (e.g., over a particular period of time), decreased blood urea nitrogen concentration, reduced change in blood urea nitrogen concentration (e.g., over a particular period of time), increased urine output, decreased kidney TGF i concentration, and lesser incidence of dialysis, relative to a comparable reference population. Kidney function can be evaluated using any method known in the art, such as one or more Study Assessments described in Example 46 herein. In some embodiments, kidney function is evaluated based on one or more of blood urea nitrogen concentration, serum creatinine concentration, eGFR, measured glomerular filtration rate, serum albumin concentration, urinalysis, renal clearance, renal imaging, renal histology, etc.

[0114] In some embodiments, a particular effect may be or comprise improved heart function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise less deterioration of heart function relative to a comparable reference population. Heart function can be evaluated using any method known in the art, such as one or more Study Assessments described in Example 46 herein. In some embodiments, heart function is evaluated based on one or more of troponin I levels, 12-lead electrocardiogram, echocardiogram, radiographic or nuclear medicine imaging, cardiac histology, etc.

[0115] In some embodiments, a particular effect may be or comprise improved liver function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise less deterioration of liver function relative to a comparable reference population. Liver function can be evaluated using any method known in the art, such as one or more Study Assessments described in Example 46 herein. In some embodiments, liver function is evaluated based on one or more of serum albumin concentration; total, direct, and/or indirect bilirubin levels; aspartate aminotransferase levels; alanine aminotransferase levels; alkaline phosphatase levels; gamma-glutamyl transpeptidase levels; imaging; histology, etc.

[0116] In some embodiments, the particular effect may be or comprise, for example, one or more effects as described in the Examples herein.

[0117] In some embodiments, Compound 1 is useful in treating a disorder or condition selected from acute lung injury, acute respiratory distress syndrome, pneumonia (e.g., influenza- associated pneumonia or COVID-19-associated pneumonia), pulmonary edema, TGFp i -induced lung injury, emphysema, chemically-induced (e.g., chlorine gas) lung injury, thermally-induced (e.g., smoke or burn) lung injury, shock-induced lung injury (e.g., lipopolysaccharide-induced shock), ischemic reperfusion lung injury, hemorrhagic shock lung injury, radiation-induced lung injury, blunt trauma to lung, and lung transplantation injury (see, for example, Huang C, e al. The Lancet. 2020 Jan 24.; Chen N, et al. The Lancet. 2020 Jan 30; Hess, D R. Semin. Respir. Crit. Care Med. 2014 Aug;35(4):418-30; Hess, D.R. Respir Care. 2011 Oct;56(10): 1555-72; Brower, R.G., Fessler, H.E. Clin. Chest Med. 2000 Sep;21(3):491-510; Haas, C.F. Crit. Care Clin. 2011 Jul;27(3):469-86; Siobal, M.S., Hess, D.R. Respir. Care 2010 Feb;55(2): 144-57, discussion 157- 61; Izumida, H., Fujishima, S. Masui 2013 May;62(5)541-6; Wang D et al. JAMA Published online February 7, 2020; Zhu, et al. Zhonghua Liu Xing Bing Xue Za Zhi. 2020;41(2): 145—151; Xu, Z., et al. Lancet Respir. Med. 2020 Feb 18; and Brauer, R. et al. Amer. J. Respir. Crit. Care Med. 2016 Aug 1; 194(3), the entirety of each of which is incorporated herein by reference). In some embodiments, Compound 1 is useful in treating a disorder or condition selected from acute lung injury, acute respiratory distress syndrome, pneumonia (e.g., influenza-associated pneumonia or COVID-19-associated pneumonia or aspiration pneumonia), pulmonary edema, chemically-induced (e.g., chlorine gas) lung injury, thermally-induced (e.g., smoke or bum) lung injury, shock-induced lung injury (e.g., septic shock or lipopolysaccharide-induced shock or cardiogenic shock), ischemic reperfusion lung injury, hemorrhagic shock lung injury, radiation- induced lung injury, blunt trauma to lung, and lung transplantation injury.

[0118] It will be appreciated that acute organ injury is the rapid deterioration of organ function and viability. Acute organ injury triggers immediate activation of repair pathways, which help to restore function and facilitate recovery of the injured organ. Without wishing to be bound by theory, it is believed that the HGF/c-Met pathway is the most important repair pathway triggered in response to an acute organ injury. [0119] In some embodiments, provided methods are useful for treating an acute organ injury (e.g., an acute injury of the lung). In some embodiments, provided methods comprise administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population who has experienced or is experiencing acute organ injury (e.g., an acute injury of the lung). In some embodiments, acute organ injury is caused by ischemia/hypoxia (i.e., oxygen deprivation of the organ). In some embodiments, acute organ injury is caused by reperfusion injury, which can lead to hemodynamic shear. In some embodiments, acute organ injury is caused by viral infection, such as by H1N1, a coronavirus (e.g., SAR-CoV-2, MERS-CoV, or SARS-CoV), influenza, etc. In some embodiments, acute organ injury is caused by traumatic injury such as by blunt trauma, thermal burns, chemical burns or injury, etc.

[0120] In some embodiments, the present disclosure provides a method of protecting an organ (e.g., the lung) from injury, the method comprising administering to a subject or population an HGF/SF mimetic (e.g., Compound 1). In some embodiments, the present disclosure provides a method of promoting alveolar regeneration, the method comprising administering to a subject or population an HGF/SF mimetic (e.g., Compound 1).

[0121] In some embodiments, the present disclosure provides a method of administering a HGF/SF mimetic (e.g., Compound 1) according to a regimen established to achieve one or more of: reduction of ongoing apoptosis and injury, thus maintaining the alveolar barrier integrity; mitigation of a rise in alveolar wall permeability and fluid extravasation in the alveolar space; improvement in gas exchange due to preservation of alveolar wall and reducing the alveolar wall and space edema; reduction in the inflammatory cell infiltration of the lungs; and induction of proliferation and regeneration of the alveolar epithelial cells, relative to a comparable reference population.

[0122] In some embodiments, the present disclosure provides a method of stimulating human endothelia and/or bronchial cell proliferation without creating fibroblasts, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some such embodiments, the present disclosure provides a method of increasing the levels of endothelial and/or bronchial cell proliferation, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. [0123] In some embodiments, provided methods are useful for treating pulmonary edema. In some embodiments, provided methods are useful for treating a disease, disorder, or condition characterized by pulmonary edema. In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects suffering from or susceptible to pulmonary edema. In some embodiments, the present disclosure provides a method of decreasing or attenuating pulmonary edema, relative to a comparable reference population, in a subject or population suffering from acute lung injury, the method comprising administering to the subject or population a HGF/SF mimetic (e.g., Compound 1). In some embodiments, the present disclosure provides a method of preventing, attenuating, or reducing red cell and/or neutrophil infdtration in alveolar spaces, relative to a comparable reference population, in a subject or population suffering from acute lung injury, the method comprising administering to the subject or population a HGF/SF mimetic (e.g., Compound 1). In some such embodiments, the HGF/SF mimetic (e.g., Compound 1) is administered 12 or 24 hours, or more, following lung injury.

[0124] A variety of pulmonary insults stimulates production and release of TGFp 1 into the pulmonary parenchyma, leading to acute cell death. Transforming growth factor beta (TGFpl) is a protein that controls proliferation, cellular differentiation, and other functions in most cells and is known to be a critical mediator in acute lung injury. Accordingly, provided methods are useful in treating TGFpl -induced acute lung injury. In some embodiments, the present disclosure provides a method of preventing, delaying the onset of, or reducing the severity of TGFpi- induced acute lung injury, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of decreasing pulmonary cell death, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of improving pulmonary epithelial regeneration, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of preserving or maintaining pulmonary architecture and/or reducing alveolar flooding, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0125] In some embodiments, provided methods are useful for treating shock-associated acute lung injury. Shock-associated acute lung injury can have a variety of underlying causes, including septic shock, LPS-induced shock, hemorrhagic shock, or cardiogenic shock. In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects suffering from or susceptible to shock-associated acute lung injury (e.g., acute lung injury associated with septic shock, LPS-induced shock, hemorrhagic shock, or cardiogenic shock). In some embodiments, the present disclosure provides a method of attenuating shock-associated acute lung injury, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of preventing, attenuating, mitigating, or reducing histopathological lung injury score, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of preventing, attenuating, mitigating, or reducing apoptotic cell death, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0126] In some embodiments, provided methods are useful for treating a chemically induced acute lung injury. In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects suffering from or susceptible to a chemically induced acute lung injury. In some embodiments, a subject or population has been exposed to chlorine gas, phosgene gas, or other inhaled toxin. In some embodiments, the present disclosure provides a method of preventing, delaying the onset of, or reducing the severity of chemically induced acute lung injury, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of preventing, delaying the onset of, or reducing the severity of pulmonary infiltration, relative to a comparable reference population, in a subject or population suffering from chemically induced acute lung injury, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0127] In some embodiments, the present disclosure provides a method of maintaining, enhancing, or increasing pulmonary output and/or arterial oxygen levels, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of maintaining, enhancing, or increasing arterial oxygen levels, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof

[0128] In some embodiments, provided methods are useful for treating acute lung injury associated with hemorrhagic shock (e.g., hemorrhagic shock from trauma). In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects suffering from or susceptible to acute lung injury associated with hemorrhagic shock (e.g., hemorrhagic shock from trauma). In some embodiments, the present disclosure provides a method of attenuating and/or decreasing lung injury associated with hemorrhagic shock, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0129] In some embodiments, provided methods are useful for treating ischemia-reperfusion lung injury. In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects suffering from or susceptible to ischemia-reperfusion lung injury (e.g., a subject or population of subjects who have undergone lung transplantation). In some embodiments, the present disclosure provides a method of attenuating ischemia-reperfusion lung injury, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of attenuating alveolar thickening, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some such embodiments, the HGF/SF mimetic (e.g., Compound 1) protects or preserves pulmonary architecture following ischemic reperfusion. In some embodiments, the present disclosure provides a method of preventing, attenuating, delaying the onset of, or mitigating IL-1 and/or IL-6 bronchoalveolar lavage fluid (BALF) accumulation, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0130] In some embodiments, provided methods are useful for treating emphysema. In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects suffering from or susceptible to emphysema. In some embodiments, the present disclosure provides a method of preventing, delaying the onset of, or reducing the severity of emphysema, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of increasing arterial levels of PaC , relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of decreasing arterial levels of PaCCh, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0131] In some embodiments, provided methods are useful for treating a thermally induced acute lung injury (e.g., an acute lung injury associated with smoke inhalation and/or thermal burn). In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects suffering from or susceptible to a thermally induced acute lung injury (e.g., an acute lung injury associated with smoke inhalation and/or thermal burn). In some embodiments, the present disclosure provides a method of attenuating lung injury associated with or resulting from a thermal injury (i.e., smoke or burn), relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of increasing or improving lung gas exchange, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0132] In some embodiments, provided methods are useful for treating radiation-induced acute lung injury. In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects suffering from or susceptible to a radiation-induced acute lung injury. In some embodiments, a subject or population has been exposed to ionizing radiation. In some embodiments, the present disclosure provides a method of attenuating or decreasing radiation-induced pulmonary apoptosis and/or inflammation, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. [0133] In some embodiments, provided methods are useful for treating acute lung injury associated with lung transplantation. In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects who have undergone a lung transplantation and/or who are susceptible to an associated acute lung injury. In some embodiments, the present disclosure provides a method of attenuating lung injury associated with transplantation, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of increasing blood pH and/or oxygen levels, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of increasing or improving alveolar air space, relative to a comparable reference population, the method comprising administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0134] In some embodiments, provided methods are useful for treating acute lung injury associated with blunt trauma to the lung. In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects who have experienced a blunt trauma to the lung.

[0135] In some embodiments, provided methods are useful for treating acute lung injury and/or ARDS associated with pneumonia. In some embodiments, pneumonia is viral pneumonia (e.g., influenza-associated pneumonia or COVID-19-associated pneumonia. In some embodiments, pneumonia is bacterial pneumonia. In some embodiments, pneumonia is aspiration pneumonia. In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population of subjects who are suffering from or susceptible to pneumonia (e.g., viral pneumonia, bacterial pneumonia, or aspiration pneumonia). [0136] In some embodiments, the present disclosure provides methods comprising administering to a subject who is receiving or has received therapy with extracorporeal membrane oxygenation (ECMO) a composition that provides Compound 1.

[0137] In some embodiments, provided methods comprise administering an HGF/SF mimetic (e.g., Compound 1) to a subject or population who is receiving therapy with ECMO and is at risk for acute lung injury. In some embodiments, the present disclosure provides a method of preserving or maintaining kidney function in a subject or population receiving ECMO relative to a comparable reference population, the method comprising administering an HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of mitigating, ameliorating, minimizing, or reducing damage to the kidneys in a subject or population receiving ECMO relative to a comparable reference population, the method comprising administering an HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of improving lung function in a subject or population receiving ECMO relative to a comparable reference population, the method comprising administering an HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0138] The present disclosure also provides methods of administering Compound 1 (e.g., by administering a composition that comprises and/or delivers Compound 1 as described herein) to a subject or population of subjects in need thereof, regardless of the subject’s malignancy status. Among other things, HGF/SF is known to stimulate c-MET (e.g., in injured organ tissues), which leads to activation of various cellular pathways, including, e.g., those involved in tissue repair.

In addition, it is suspected that uncontrolled activation of c-MET can initiate tumorigenesis and/or stimulate and/or promote tumor growth. As described above, Compound 1 is a HGF/SF mimetic, and as such, without wishing to be bound by theory, administration of Compound 1 under certain conditions might be expected to promote initiation or growth of cancer and/or other malignancies. Yet, experiments described herein (e.g., Example 45) demonstrate that, surprisingly, this may not be the case when Compound 1 is administered according to methods provided herein. Accordingly, the present disclosure encompasses the recognition that Compound 1 can be administered to a subject or population of subjects in need thereof, regardless of the subject’s malignancy status. It will be appreciated that such insight may be applicable not only to methods of treating indications described herein (e.g., treating acute lung injury, acute respiratory distress syndrome, COVID-19 pneumonia, etc.), but also to methods of treating any indication for which Compound 1 therapy is suitable.

[0139] In some embodiments, the present disclosure provides methods comprising administering a composition that provides Compound 1 (e.g., as provided herein) to a subject or population of subjects in need thereof, wherein the subject is suffering from an active malignancy or has suffered from a solid, metastatic or hematologic malignancy (e.g., within 5 years prior to administration of the composition). In some such embodiments, the subject has not suffered from a basal or squamous cell carcinoma of the skin that has been treated and/or removed. In some embodiments, the subject is suffering from or has suffered from glioma, colon cancer, or pancreatic cancer.

[0140] In some embodiments, the present disclosure provides methods comprising administering a composition that provides Compound 1 (e.g., as provided herein) to a subject or population of subjects in need thereof, wherein the subject has not been assessed for an active malignancy or a history of a solid, metastatic or hematologic malignancy. In some embodiments, a subject has not been assessed for an active malignancy or a history of a solid, metastatic or hematologic malignancy if prior to administration of Compound 1 (e.g., within about 1 year, about 6 months, about 3 months, about 2 months, about 1 month, about 2 weeks, or about 1 week), the subject has not been questioned about and/or screened for an active malignancy or a history of a solid, metastatic or hematologic malignancy. In some embodiments, a subject has not been assessed for an active malignancy or a history of a solid, metastatic or hematologic malignancy if a decision to administer Compound 1 therapy (e.g., by a physician) did not depend upon questioning of and/or screening of the subject for an active malignancy or a history of a solid, metastatic or hematologic malignancy.

COVID-19 Pneumonia and Other Viral Pneumonias

[0141] Over the past 15 years, three respiratory viruses have attracted significant attention because of the high proportion of affected patients who develop critical illness and ARDS: influenza, (particularly influenza A H1N1 2009); Middle Eastern respiratory syndrome coronavirus (MERS-CoV); and SARS coronavirus (SARS-CoV). Recently, a fourth virus emerged, the SARS coronavirus 2 (SARS-CoV-2), the proximate cause of COVID-19. COVID- 19 is a respiratory tract infection caused by SARS-CoV-2 (2019-nCoV). According to WHO Interim Guidance, the most common diagnosis in severe COVID-19 patients is severe pneumonia. It is estimated that approximately 14% of people with COVID-19 develop severe disease requiring hospitalization and oxygen support and 5% require admission to an intensive care unit. In severe cases, COVID-19 can be complicated by ARDS, sepsis and septic shock, and multiorgan failure, including AKI, neurological injuries, and cardiac injury.

[0142] Clinical presentation among reported cases of COVID-19 varies in severity from asymptomatic infection to mild illness to severe or fatal illness. Acute respiratory distress syndrome (ARDS) developed in 17-29% of hospitalized patients, and secondary infection developed in 10% of patients (Huang C, e al. The Lancet. 2020 Jan 24.; Chen N, et al. The Lancet. 2020 Jan 30, each of which is hereby incorporated by reference in its entirety). In one report, the median time from symptom onset to ARDS was 8 days (Wang D et al. JAMA Published online February 7, 2020, which is hereby incorporated by reference in its entirety). Most patients with ARDS develop respiratory failure severe enough to require mechanical ventilator support in an intensive care unit (ICU). Mechanical ventilation can exacerbate lung injury (Hess DR. Semin Respir Crit Care Med. 2014 Aug;35(4):418-30; Hess DR. Respir Care. 2011 Oct;56(10): 1555-72; Brower RG, Fessler HE. Clin Chest Med 2000 Sep;21(3):491-510, each of which is hereby incorporated by reference in its entirety). Approximately 20-30% of hospitalized patients with COVID-19 and pneumonia have required intensive care for respiratory support (Huang C, e al. The Lancet. 2020 Jan 24.; Wang D et al. JAMA Published online February 7, 2020). Among critically ill patients admitted to an intensive care unit, 11-64% received high-flow oxygen therapy and 47-71% received mechanical ventilation; some hospitalized patients have required advanced organ support with endotracheal intubation and mechanical ventilation (4-42%) (Chen N, et al. Lancet. 2020 Jan 30; Wang D et al. JAMA Published online February 7, 2020; Zhu, et al. Zhonghua Liu Xing Bing Xue Za Zhi.

2020;41(2): 145-151, which are hereby incorporated by reference in their entirety). A small proportion of patients have also been supported with extracorporeal membrane oxygenation (ECMO) (3-12%) (ChenN, et al. The Lancet. 2020 Jan 30; Wang D et al. JAMA Published online February 7, 2020; Zhu, et al. Zhonghua Liu Xing Bing Xue Za Zhi. 2020;41(2): 145—151). Other reported complications include cardiac injury, arrhythmia, septic shock, liver dysfunction, acute kidney injury, and multi-organ failure. Post-mortem examinations in one patient who died of ARDS reported pulmonary findings of diffuse alveolar damage. [0143] Accordingly, in some embodiments, provided methods comprise administering a HGF/SF mimetic (e.g., Compound 1) to a subject or population who is characterized by one or more of the following: admitted to an intensive care unit; receiving endotracheal intubation; receiving mechanical ventilation; receiving extracorporeal membrane oxygenation (ECMO); suspected of having, or has been diagnosed with, viral-induced lung injury; suspected of having, or has been diagnosed with, COVID-19-associated pneumonia; suspected of having, or has been diagnosed with, viral influenza-associated pneumonia; suspected of having, or has been diagnosed with, co-morbidities COVID-19 and pneumonia; suspected of having, or has been diagnosed with, co-morbidities viral influenza and pneumonia; and suspected of having, or has been diagnosed with, one or more additional complications selected from diffuse alveolar damage (DAD), cardiac injury, arrhythmia, septic shock, liver dysfunction, acute kidney injury, and multi-organ failure.

[0144] The FDA has only approved one drug for treating COVID-19, the antiviral agent remdesivir. Remdesivir is recommended only for hospitalized patients who require supplemental oxygen; however, it is not typically recommended for patients on mechanical ventilation due to limited benefits at an advanced stage of disease. Clinical management includes prompt implementation of recommended infection prevention and control measures and supportive management of complications, including advanced organ support. Existing drugs for other indications, including lopinavir-ritonavir (HIV protease inhibitors), chloroquine/hydroxychloroquine (an immunosuppressive drug and anti-parasite), are being used in the clinics for the treatment of COVID-19, but their efficacy remains to be established in clinical trials (Al-Tawfiq, Al-Homoud, and Memish 2020; Cao et al. 2020; Cortegiani et al.

2020; Gordon et al. 2020; Ko et al. 2020; Wang, Cao, et al. 2020). In a recently published clinical trial, in adult patients with severe COVID-19, no benefit was observed with lopinavir- ritonavir treatment beyond standard care in hospitalized adult patients with severe COVID-19 (Cao et al. 2020). Vaccines against SARS-CoV-2 are beginning to become available to certain populations of subjects; however, widespread vaccine availability remains to be realized. Therefore, there is an urgent unmet medical need to develop new drugs for COVID-19 infection and associated lung injury.

[0145] While there is a critical need to develop anti-viral drugs to treat COVID-19, it is also crucial to develop therapies that reduce organ injury induced by SARS-CoV-2 and further amplified by a dysregulated immune and inflammatory response. China’s National Health Commission treatment guidelines recommend treatment with IL-6 inhibitor, tocilizumab, for patients with severe COVID-19 and elevated IL-6 levels. This agent and another IL-6 inhibitor, sarilumab are currently being evaluated in clinical trials in the US. Cytokine induced organ injury and inflammation, “cytokine storm”, increase vascular permeability and exacerbate impaired pulmonary gas exchange.

[0146] The present disclosure encompasses the recognition that Compound 1, through activation of c-Met, a crucial pathway that limits organ injury and promotes organ repair, has a great therapeutic potential in COVID-19. Many patients with COVID-19 have evidence of injury to the heart and kidneys, in addition to the lungs, presumably mediated by viral binding to ACE2, which is heavily expressed in all of these organs. In particular, recent multi-site studies from the United States show that 22% to 36% of patients hospitalized with COVID-19 had acute kidney injury and 3% to 14% required dialytic intervention. Compound 1 is effective in reducing injury to these organs in various animal models while preserving tissue architecture, function, and improving animal survival. Thus, Compound 1 provides a unique opportunity to improve clinical outcomes by exerting beneficial effects on multiple organs affected by COVID- 19.

[0147] In some embodiments, the present disclosure provides methods of treating (e.g., reducing severity and/or progression of) pulmonary dysfunction in patients (e g., adult patients) hospitalized with COVID-19 pneumonia. In some embodiments, the present disclosure provides methods of treating (e.g., reducing severity and/or progression of) pulmonary and/or renal dysfunction in patients (e.g., adult patients) hospitalized with COVID-19 pneumonia. In some embodiments, such methods comprise administering intravenously 2 mg/kg Compound 1 twice daily for 5 days. In some embodiments, such methods comprise administering intravenously 2 mg/kg Compound 1 once daily for 4 days. In some embodiments, such methods comprise administering intravenously 2 mg/kg Compound 1 once daily for 3 days. [0148] In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population suffering from or susceptible to pulmonary and/or renal dysfunction (e.g., a subject or population hospitalized with COVID-19 pneumonia). In some embodiments, provided methods comprise administering a composition that provides Compound 1 to a subject or population who is hospitalized with COVID-19 pneumonia.

[0149] In some embodiments, the present disclosure provides methods of administering Compound 1 to a subject or a population suffering from or susceptible to pulmonary and/or renal dysfunction (e.g., a subject or population hospitalized with COVID-19 pneumonia), e.g., according to a dosing regimen described herein. In some embodiments, the present disclosure provides methods of administering Compound 1 to a subject or a population who is hospitalized with COVID-19 pneumonia, e.g., according to a dosing regimen described herein.

[0150] In some embodiments, the present disclosure provides methods of administering Compound 1 to a subject or population who is suffering from or susceptible to pulmonary and/or renal dysfunction (e.g., a subject or population hospitalized with COVID-19 pneumonia), for example by administering a composition that provides Compound 1, e.g., according to a regimen established to achieve one or more desirable outcomes. In some embodiments, the present disclosure provides methods of administering Compound 1 to a subject or a population who is hospitalized with COVID-19 pneumonia, for example by administering a composition that provides Compound 1, e.g., according to a regimen established to achieve one or more desirable outcomes. In some embodiments, the regimen is or has been established to achieve one or more desirable outcomes in a population to which Compound 1 has been administered, relative to a comparable reference population that has not received Compound 1 (e.g., that has received a reference composition which does not deliver Compound 1).

[0151] In some embodiments, certain parameters may be evaluated to determine if a desirable outcome is achieved. Any one or more of parameters such as these may, in some embodiments, be useful for determining short-term and/or long-term efficacy of Compound 1 administered to the patient population. Alternatively or additionally, any one or more of such parameters may be assessed to monitor patient response to Compound 1 therapy.

[0152] In some embodiments, the present disclosure provides a method comprising: administering to a subject, or to a population of subjects, suffering from viral pneumonia (e.g., hospitalized with COVID-19 pneumonia) a composition that provides Compound 1 according to a regimen established to achieve one or more of: decreased lung injury score, reduced mortality, reduced lung histopathology, better cytokine evaluation, better lung MPO measurement, improved pulmonary function, reduce lung obstruction in bronchus and bronchioles, andbetter cardiopulmonary variables, relative to a comparable reference population.

[0153] In some embodiments, the present disclosure provides methods of improving outcomes for patients suffering from or susceptible to a disease, disorder or condition that is (i.e., is statistically and/or is in fact for the particular patient) associated with one or more undesirable respiratory features such as for example inflammation in the lungs, fluid in the lungs, fibroids in the lungs, etc., by administration of Compound 1 (e.g., alone and/or in combination with other therapy, for example directed at the underlying disease, disorder or condition, or otherwise being used in treatment of the patient(s)).

[0154] In some embodiments, in methods provided herein, a reference population has not received a composition that provides Compound 1. In some embodiments, in methods provided herein, a reference population has received an otherwise comparable reference composition that does not provide Compound 1 (e.g., a placebo, such as normal saline). In some embodiments, in methods provided herein, a reference composition may be or comprise normal saline. In some embodiments, in methods provided herein, a reference composition may be or may have been administered at the same intervals and/or volumes as a composition that provides Compound 1. [0155] As used herein, “mean” may refer to an average and/or a least squares mean (LS mean). In some embodiments, “mean” may refer to a LS mean (e.g., a MMRM LS mean).

[0156] In some embodiments, a composition that provides Compound 1 is administered according to a regimen established to achieve a particular effect, e.g., at a particular time point (e.g., about 7 days, about 14 days, about 28 days, about 6 months and/or about 12 months after injury and/or randomization and/or first administration of Compound 1). In some embodiments, a composition that provides Compound 1 is administered according to a regimen established to achieve a particular effect, e.g., at a particular time point, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1, which comparable population may, in some embodiments, have received a reference composition that is otherwise comparable but does not provide Compound 1 upon administration).

[0157] In some embodiments, a particular effect may be achieved within a particular time frame or by a particular time point. In some embodiments, such time point may be, for example, about 7 days, about 14 days, about 28 days, about 6 months and/or about 12 months after initiation of Compound 1 therapy as described herein. In some embodiments, a particular effect may be achieved at about 28 days after initiation of Compound 1 therapy as described herein. [0158] In some embodiments, a particular effect may be or comprise a particular proportion of patients alive, without the need for mechanical ventilation and free of the need for renal replacement therapy (on an ongoing basis), e.g., at Day 28. In some embodiments, a particular effect may be or comprise an increased proportion of patients alive, without the need for mechanical ventilation and free of the need for renal replacement therapy (on an ongoing basis), e g., at Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0159] In some embodiments, a particular effect may be or comprise a particular proportion of patients alive, without the need for mechanical ventilation and free of the need for renal replacement therapy, e.g., at Day 28. In some embodiments, a particular effect may be or comprise an increased proportion of patients alive, without the need for mechanical ventilation and free of the need for renal replacement therapy, e.g., at Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0160] In some embodiments, a particular effect may be or comprise a particular all-cause mortality. In some embodiments, a particular effect may be or comprise a reduced all-cause mortality, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0161] In some embodiments, a particular effect may be or comprise a particular proportion of patients not requiring mechanical ventilation, e.g., at Day 28. In some embodiments, a particular effect may be or comprise an increased proportion of patients not requiring mechanical ventilation, e.g., at Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1). [0162] In some embodiments, a particular effect may be or comprise a particular proportion of patients not requiring renal replacement therapy on an on-going basis, e.g., at Day 28. In some embodiments, a particular effect may be or comprise an increased proportion of patients not requiring renal replacement therapy on an on-going basis, e.g., at Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0163] In some embodiments, a particular effect may be or comprise a particular number of ventilator-free days, e.g., through Day 28. In some embodiments, a particular effect may be or comprise an increased number of ventilator-free days, e.g., through Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0164] In some embodiments, a particular effect may be or comprise a particular proportion of patients requiring initiation of mechanical ventilation and/or ECMO, e.g., through Day 28. In some embodiments, a particular effect may be or comprise a reduced proportion of patients requiring initiation of mechanical ventilation and/or ECMO, e.g., through Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0165] In some embodiments, a particular effect may be or comprise a particular proportion of patients requiring initiation of renal replacement therapy (RRT), e.g., through Day 28. In some embodiments, a particular effect may be or comprise a reduced proportion of patients requiring initiation of renal replacement therapy, e.g., through Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0166] In some embodiments, a particular effect may be or comprise a particular number of days to renal recovery (defined as freedom from further RRT on an ongoing basis) in subjects who were on RRT at time of randomization. In some embodiments, a particular effect may be or comprise a reduced number of days to renal recovery (defined as freedom from further RRT on an ongoing basis) in subjects who were on RRT at time of randomization, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1). [0167] In some embodiments, a particular effect may be or comprise a particular number of ICU days, e.g., through Day 28. In some embodiments, a particular effect may be or comprise a reduced number of ICU days, e.g., through Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0168] In some embodiments, a particular effect may be or comprise a particular score on an ordinal scale (e.g., the ordinal scale of Example 38), e.g., at Day 28. In some embodiments, a particular effect may be or comprise a reduced score on an ordinal scale (e.g., the ordinal scale of Example 38), e.g., at Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0169] In some embodiments, a particular effect may be or comprise a particular change (e.g., a reduction) in score on an ordinal scale (e.g., the ordinal scale of Example 38), e.g., at Day 28. In some embodiments, a particular effect may be or comprise a greater change (e.g., a greater reduction) in score on an ordinal scale (e.g., the ordinal scale of Example 38), e.g., at Day 28, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0170] In some embodiments, a particular effect may be or comprise a particular number of days to hospital discharge from an initiating event (e.g., randomization in a clinical trial, admission to a hospital, admission to an ICU, etc.). In some embodiments, a particular effect may be or comprise a particular number of days to hospital discharge from randomization. In some embodiments, a particular effect may be or comprise a reduced number of days to hospital discharge from randomization, relative to an appropriate reference as described herein (e.g., as is observed in a comparable population who has not received a composition that provides Compound 1).

[0171] In some embodiments, a particular effect may be or comprise, decreased lung injury score, reduced mortality, reduced lung histopathology, better cytokine evaluation, better lung MPO measurement, improved pulmonary function, reduce lung obstruction in bronchus and bronchioles, or better cardiopulmonary variables.

[0172] In some embodiments, a particular effect may be or comprise a better cytokine evaluation (e.g., as evidenced by reduced or resolved systemic or localized inflammation). [0173] In some embodiments, a particular effect may be or comprise decreased mortality rate relative to a comparable reference population.

[0174] In some embodiments, a particular effect may be or comprise reduced time to improvement in oxygenation for at least 48 hours (e.g., reduced time to achieve an increase in Sp02/Fi02 of 50 or greater compared to the nadir Sp02/Fi02), reduced mean change from baseline on an 8-point Ordinal Scale (e.g., the ordinal scale described in Example 38), reduced time to improvement from admission in one category on an 8-point Ordinal Scale (e.g., the ordinal scale described in Example 38), reduced number of days with hypoxemia, reduced time to improvement in oxygenation for at least 48 hours by clinical severity (e.g., reduced time to achieve an increase in Sp02/Fi02 of 50 or greater compared to the nadir Sp02/Fi02), increased number of ventilator free days in the first 30 days, reduced number of patients requiring initiation of mechanical ventilation and/or ECMO, reduced number of patients admitted into an intensive care unit, reduced number of days of hospitalization (e.g., among survivors), and/or reduced number of patients with secondary bacterial and/or fungal infection.

[0175] In some embodiments, a particular effect may be or comprise increased lung function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise one or more of: increased pulmonary output, increased arterial oxygen (PaCh), decreased arterial carbon dioxide (PaCCh), increased ratio of PaCh/FiCh, decreased lung injury score, decreased lung hydroxyproline concentration, decreased lung collagen level, decreased lung TGFp l concentration, and increased blood pH, relative to a comparable reference population.

[0176] In some embodiments, a particular effect may be or comprise increased kidney function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise less deterioration of kidney function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise one or more of: decreased serum creatinine concentration, increased estimated glomerular filtration rate (eGFR), decreased blood urea nitrogen concentration, increased urine output, decreased kidney TGFp l concentration, and lesser incidence of dialysis, relative to a comparable reference population. In some embodiments, a particular effect may be or comprise one or more of: decreased serum creatinine concentration, reduced change in serum creatinine concentration (e.g., over a particular period of time), increased estimated glomerular filtration rate (eGFR), reduced change in eGFR (e.g., over a particular period of time), increased measured glomerular fdtration rate, reduced change in measured glomerular filtration rate (e.g., over a particular period of time), decreased blood urea nitrogen concentration, reduced change in blood urea nitrogen concentration (e.g., over a particular period of time), increased urine output, decreased kidney TGF i concentration, and lesser incidence of dialysis, relative to a comparable reference population. Kidney function can be evaluated using any method known in the art, such as one or more Study Assessments described in Example 46 herein. In some embodiments, kidney function is evaluated based on one or more of blood urea nitrogen concentration, serum creatinine concentration, eGFR, measured glomerular filtration rate, serum albumin concentration, urinalysis, renal clearance, renal imaging, renal histology, etc.

[0177] In some embodiments, a particular effect may be or comprise improved heart function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise less deterioration of heart function relative to a comparable reference population. Heart function can be evaluated using any method known in the art, such as one or more Study Assessments described in Example 46 herein. In some embodiments, heart function is evaluated based on one or more of troponin I levels, 12-lead electrocardiogram, echocardiogram, radiographic or nuclear medicine imaging, cardiac histology, etc.

[0178] In some embodiments, a particular effect may be or comprise improved liver function relative to a comparable reference population. In some embodiments, a particular effect may be or comprise less deterioration of liver function relative to a comparable reference population. Liver function can be evaluated using any method known in the art, such as one or more Study Assessments described in Example 46 herein. In some embodiments, liver function is evaluated based on one or more of serum albumin concentration; total, direct, and/or indirect bilirubin levels; aspartate aminotransferase levels; alanine aminotransferase levels; alkaline phosphatase levels; gamma-glutamyl transpeptidase levels; imaging; histology, etc.

[0179] In some embodiments, the disease, disorder or condition being treated in methods provided herein is characterized by pulmonary edema, pulmonary epithelial cell apoptosis, inflammatory cell infiltration, impaired oxygenation, hypoxemia and/or lung fibrosis.

[0180] In some embodiments, the present disclosure provides methods of administering Compound 1 to a subject or a population of subjects who are suffering from or susceptible to a respiratory disease, disorder or condition such as, e.g., COVID-19 lung injury. [0181] In some embodiments, the present disclosure provides methods comprising administering to a subject who is receiving or has received therapy with extracorporeal membrane oxygenation (ECMO) a composition that provides Compound 1.

[0182] In some embodiments, provided methods comprise administering an HGF/SF mimetic (e.g., Compound 1) to a subject or population who is receiving therapy with ECMO and is at risk for acute lung injury. In some embodiments, the present disclosure provides a method of preserving or maintaining kidney function in a subject or population receiving ECMO relative to a comparable reference population, the method comprising administering an HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of mitigating, ameliorating, minimizing, or reducing damage to the kidneys in a subject or population receiving ECMO relative to a comparable reference population, the method comprising administering an HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof. In some embodiments, the present disclosure provides a method of improving lung function in a subject or population receiving ECMO relative to a comparable reference population, the method comprising administering an HGF/SF mimetic (e.g., Compound 1) to a subject or population in need thereof.

[0183] In some embodiments, provided methods comprise administering to a subject or population an HGF/SF mimetic (e.g., Compound 1) and one or more anti-viral agents. In some embodiments, such anti-viral agents are selected from oseltamivir, lopinavir, ritonavir, chloroquine, hydroxychloroquine, and remdesivir, and combinations thereof. In some embodiments, provided methods comprise administering to a subject or population an HGF/SF mimetic (e.g., Compound 1) and anti-viral therapy. In some such embodiments, anti-viral therapy comprises hydroxychoroquine and azithromycin.

[0184] In some embodiments, provided methods comprise administering to a subject or population an HGF/SF mimetic (e.g., Compound 1) and one or more IL-6 inhibitors. Nonlimiting examples of IL-6 inhibitors include tocilizumab and sarilumab.

Subjects to Be Treated

[0185] In some embodiments, one or more subjects or populations selected to receive Compound 1 (e.g., according to provided methods) are characterized by one or more factors described herein. It will be appreciated that, in some embodiments, a subject or population is characterized by multiple (i.e., more than one) factors described herein.

[0186] In some embodiments, one or more subjects or populations selected to receive Compound 1 as described herein are characterized by one or more factors such as, for example, one or more of: presence of one or more risk factors or characteristics of a respiratory disease, disorder, or condition. In some embodiments, such subject(s) or population(s) may display, for example, one or more features of lung inflammation, fluid in the lungs, fibroids in the lungs, difficulty breathing, etc. Alternatively or additionally, in some embodiments, such subject(s) or population(s) may be suffering from an underlying condition (e.g., infection, trauma, etc.) that is associated with (e.g., established to be correlated with) a respiratory disease, disorder or condition.

[0187] In some embodiments, a respiratory disease, disorder or condition may involve one or more of airway (i .e., may affect tubes that carry gases such as oxygen into and out of the lungs), lung tissue (i.e., may involve inflammation and/or scarring of lung tissue), and/or circulation (i.e., may involve clotting, inflammation, and/or scarring of blood vessels in the lungs).

[0188] In some embodiments, a subject or population is suffering from or susceptible to acute respiratory distress (e.g., ARDS as defined by the Berlin criteria). In some embodiments, a subject or population is suffering from or susceptible to mild ARDS (e.g., as defined by the Berlin criteria). In some embodiments, a subject or population is suffering from or susceptible to moderate ARDS (e.g., as defined by the Berlin criteria). In some embodiments, a subject or population is suffering from or susceptible to severe ARDS (e.g., as defined by the Berlin criteria). In some embodiments, a subject or population is suffering from or susceptible to ARDS, e.g., mild ARDS or moderate ARDS (as defined in the Berlin criteria using Pa02/Fi02). [0189] In some embodiments, a subject or population is suffering from acute lung injury.

[0190] In some embodiments, a subject or population is suffering from or susceptible to ALI or ARDS secondary to, induced by, or otherwise associated with one or more of ischemia, drugs and/or toxins, neonatal status, radiation, etc.

[0191] In some embodiments, a subject or population is suffering from or susceptible to a chronic respiratory disease, disorder or condition.

[0192] In some embodiments, a subject or population is suffering from or susceptible to pulmonary edema. [0193] In some embodiments, a subject or population is suffering from or susceptible to shock-associated acute lung injury (e.g., acute lung injury associated with septic shock, LPS- induced shock, hemorrhagic shock, or cardiogenic shock). In some embodiments, a subject or population is in shock (e.g., septic shock, LPS-induced shock, hemorrhagic shock, or cardiogenic shock).

[0194] In some embodiments, a subject or population is suffering from or susceptible to a chemically induced acute lung injury. In some embodiments, a subject or population is suffering from or susceptible to a radiation-induced acute lung injury. In some embodiments, a subject or population may be or have been exposed to one or more drugs and/or toxins (e.g., chlorine gas or phosgene gas) or radiation. In some embodiments, a subject or population has been exposed to chlorine gas, phosgene gas, or other inhaled toxin. In some embodiments, a subject or population has been exposed to ionizing radiation.

[0195] In some embodiments, a subject or population is suffering from or susceptible to acute lung injury associated with hemorrhagic shock (e.g, hemorrhagic shock from trauma). In some embodiments, a subject or population has experienced a traumatic injury (e.g., to the lung). [0196] In some embodiments, a subject or population is suffering from or susceptible to acute lung injury associated with blunt trauma to the lung. In some embodiments, a subject or population has experienced a blunt trauma injury of the lung.

[0197] In some embodiments, a subject or population is suffering from or susceptible to ischemia-reperfusion lung injury. In some embodiments, a subject or population is suffering from or susceptible to acute lung injury associated with lung transplantation. In some embodiments, a subject or population has undergone a lung transplantation.

[0198] In some embodiments, a subject or population is suffering from or susceptible to emphysema.

[0199] In some embodiments, a subject or population is suffering from or susceptible to a thermally induced acute lung injury (e.g., an acute lung injury associated with smoke inhalation and/or thermal burn). In some embodiments, a subject or population is suffering from or has suffered from smoke inhalation and/or bum.

[0200] In some particular embodiments, a subject or population is suffering from or susceptible to a pneumonia (e.g., a viral pneumonia, bacterial pneumonia, or aspiration pneumonia). In some embodiments, a subject or population is suffering from pneumonia (e.g., as confirmed by, e.g., chest imaging). In some embodiments, a subject or population is suffering from COVID-19 pneumonia (e.g., as confirmed by chest imaging). In some embodiments, a subject or population is suffering from or susceptible to COVID-19 pneumonia (e.g., as confirmed by chest imaging).

[0201] Alternatively or additionally, in some particular embodiments, a subject or population may be suffering from or susceptible to infection, for example viral infection, e.g., with a respiratory virus such as respiratory syncytial virus (RSV), influenza, and/or a coronavirus (e.g., COVID-19).

[0202] Still further alternatively or additionally, in some particular embodiments, a subject or population may be suffering from or susceptible to one or more of a common cold, pneumonia, lung cancer, pulmonary embolism allergy, asthma, bronchiostasis or bronchitis, chronic obstructive pulmonary disease (COPD), a cold, obstructive sleep apnea syndrome, pulmonary hypertension, tuberculosis, or a viral infection (e.g., with a coronavirus (e.g., COVID-19), an influenza virus, an RSV, etc.).

[0203] In some embodiments, a subject or population is or has been a smoker. In some embodiments, a subject or population is not and has never been a smoker.

[0204] In some embodiments, a subject or population is in respiratory failure.

[0205] In some embodiments, a subject or population may be in the presence of or have been exposed to one or more risk factors such as, for example, allergens, air pollution (indoor and/or outdoor), smoking, infection (e.g., with a bacterial, viral, or fungal pathogen whose infection is associated with respiratory symptom(s)), or gas agents (e.g., chlorine gas or phosgene gas).

[0206] In some embodiments, a subject or population may display one or more symptoms or characteristics selected from the group consisting of chills, cough, difficulty breathing, fever, headache, etc. In some particular embodiments, a subject or population may display cough (e.g., dry cough) and fever. In some embodiments, a subject or population may display one or more of symptoms or features selected from labored breathing, rapid breathing, muscle fatigue, general weakness, low blood pressure, shortness of breath, and confusion, and combinations thereof. [0207] In some embodiments, a subject or population may be or have been diagnosed with an infectious disease (e.g., infection with a microbe or virus), for example through detection of a nucleic acid and/or antigen characteristic of a particular infectious agent in a sample(s) (e.g., that is or comprises blood, feces, saliva, serum, sputum, sweat, tears, urine, etc.) from the subject(s). [0208] In some embodiments, a subject or population has been admitted to an intensive care unit.

[0209] In some embodiments, a subject or population is or was on a ventilator and/or is or was receiving supplemental oxygen.

[0210] In some embodiments, a subject or population is resistant to oxygen therapy.

[0211] In some embodiments, a subject or population is receiving or has received endotracheal intubation.

[0212] In some embodiments, a subject or population may be having, have had, or be at risk of having a myocardial infarction.

[0213] Without wishing to be bound by any particular theory, subjects receiving therapy with an HGF/SF mimetic (e.g., Compound 1) may benefit from therapy with one or more additional agents. See , for example, Narasaraju, T., et al. Curr. Mol. Med. 2014;(14)690-702. In some embodiments, a subject or population is receiving or has received one or more additional therapies. In some embodiments, a subject or population is receiving or has received one or more antibiotics, antivirals, corticosteroids, and painkillers, and combinations thereof. In some embodiments, a subject or population is receiving or has received one or more antivirals. In some such embodiments, a subject or population is diagnosed with or suspected of having COVID-19 and is receiving or has received one or more antivirals. In some embodiments, a subject or population is receiving or has received antiviral therapy selection from oseltamivir, lopinavir, ritonavir, chloroquine, hydroxychloroquine, and remdesivir, and combinations thereof. In some embodiments, a subject or population is receiving or has received antiviral therapy comprising hydroxychloroquine and azithromycin. In some embodiments, a subject or population is receiving or has received one or more IL-6 inhibitors. In some embodiments, a subject or population is receiving or has received one or more IL-6 inhibitors selected from tocilizumab and sarilumab.

[0214] Those skilled in the art, reading the present disclosure will appreciate that, in some embodiments, appropriate subjects or populations to receive Compound 1 therapy as described herein may be those described in one or more of Combes, et al. Extracorporeal Membrane Oxygenation for Severe Acute Respiratory Distress Syndrome, N Engl. J. Med. 378;21, May 24, 2018, 1965; JAMA. 2009;302(17): 1888-1895; or Peek et al., Efficacy and economic assessment of conventional ventilatory support versus extracorporeal membrane oxygenation for severe adult respiratory failure (CESAR): a multicentre randomised controlled trial. Lancet, September 16, 2009, DOI:10.1016/S0140-6736(09)61069-2 (the entirety of each of which is incorporated herein by reference).

[0215] In some embodiments, the present disclosure provides the recognition that subjects or populations that receive therapy with extracorporeal membrane oxygenation (ECMO) benefit from treatment with an HGF/SF mimetic (e.g., Compound 1). ECMO stands for extracorporeal membrane oxygenation. In some embodiments, subjects or populations who need ECMO have a severe and life-threatening illness that stops their heart or lungs from working properly. In some embodiments, ECMO is used during life-threatening conditions such as severe lung damage from, e.g., infection, or shock after a massive heart attack.

[0216] In some embodiments, a subject or population is receiving or has received therapy with ECMO. In some embodiments, subjects or populations are supported by an ECMO machine for only a few hours. In some embodiments, subjects or populations are supported by an ECMO machine for one or more days. In some embodiments, subjects or populations are supported by an ECMO machine for one or more weeks. In some embodiments, a subject is a child, or a population consisting of children, of less than 18 years. In some embodiments, a subject is an adult or an adult population. In some embodiments, a subject is an adult, or a population consisting of adults, of 55 years or older. In some embodiments, a subject or population receives treatment, or is eligible for treatment, with ECMO wherein the subject’s or population’s lungs cannot provide enough oxygen to the body even when given extra oxygen. In some embodiments, a subject or population receives treatment, or is eligible for treatment, with ECMO wherein the subject’s or population’s lungs cannot expel carbon dioxide even with help from a mechanical ventilator.

[0217] Without wishing to be bound by any particular theory, patients on ECMO are particularly susceptible to acute kidney injury. Accordingly, in some embodiments, a subject or population who is receiving ECMO, or is eligible for ECMO, is at risk for acute kidney injury. [0218] In some embodiments, a subject or population is further characterized by impaired kidney function. In some embodiments, a subject is suffering from acute kidney injury (e.g., secondary to ECMO therapy). In some embodiments, a subject or population is suffering from a renal ischemia/reperfusion injury, renal failure, renal fibrosis, or a renal trauma. In some embodiments, a subject or population is receiving or has received dialysis (e.g., 1, 2, 3, 4, or 5 or more sessions in the last 1 week, 2 weeks, 3 weeks or more). In some embodiments, a subject or population has undergone renal transplantation (e.g., in the last 1 day, 2 days, 3 days, 1 week, 2 weeks, or more).

[0219] In some embodiments, a subject or population is experiencing respiratory complications, e.g., associated with COVID-19, influenza, chemical or thermal injury, chemical or thermal burns, etc. In some embodiments, a subject or population is suspected of suffering from a coronavirus. In some such embodiments, a subject or population is suspected of suffering from COVID-19. In some embodiments, a subject or population is or has been diagnosed with a coronavirus. In some such embodiments, a subject or population is or has been diagnosed with COVID-19.

[0220] Without wishing to be bound by any particular theory, subjects suffering from COVID-19 may be particularly susceptible to heart and/or kidney injuries, in addition to lung injury, presumably due to viral binding to ACE2, which is heavily expressed in all of these organs. Accordingly, in some embodiments, a subject or population is suffering from or susceptible to heart dysfunction, e.g., in addition to pulmonary dysfunction. In some embodiments, a subject or population suffering from or susceptible to heart dysfunction is characterized using one or more Study Assessments described in Example 46 herein or any other method known in the art (e.g., troponin I levels, 12-lead electrocardiogram, echocardiogram, radiographic or nuclear medicine imaging, cardiac histology, etc.). In some embodiments, a subject or population is suffering from or susceptible to renal dysfunction, e.g., in addition to pulmonary dysfunction. In some embodiments, a subject or population suffering from or susceptible to renal dysfunction is characterized using one or more Study Assessments described in Example 46 herein or any other method known in the art (e.g., blood urea nitrogen concentration, serum creatinine concentration, eGFR, measured glomerular filtration rate, serum albumin concentration, urinalysis, renal clearance, renal imaging, renal histology, etc.).

[0221] Further, without wishing to be bound by any particular theory, subjects suffering from COVID-19 may be particularly susceptible to liver injuries, in addition to lung and/or heart and/or kidney injuries. Accordingly, in some embodiments, a subject or population is suffering from or susceptible to liver dysfunction, e.g., in addition to pulmonary dysfunction. In some embodiments, a subject or population suffering from or susceptible to liver dysfunction is characterized using one or more Study Assessments described in Example 46 herein or any other method known in the art (e.g., serum albumin concentration; total, direct, and/or indirect bilirubin levels; aspartate aminotransferase levels; alanine aminotransferase levels; alkaline phosphatase levels; gamma-glutamyl transpeptidase levels; imaging; histology, etc.).

[0222] In some embodiments, a subject or population is hospitalized (e.g., hospitalized with COVID-19 pneumonia).

[0223] In some embodiments, a subject or population has provided a sample (e.g., a respiratory tract sample) that tests positive using a reverse-transcriptase-polymerase-chain- reaction (RT-PCR) assay for SARS-CoV-2. In some such embodiments, the sample has tested positive during the same hospitalization in which the subject or population is receiving or has received therapy with Compound 1.

[0224] In some embodiments, a subject or population has an ordinal score of 4 or 5, e.g., on the ordinal scale described in Example 38. In some embodiments, a subject or population has an ordinal score of 1, 2, or 3, e.g., on the ordinal scale described in Example 38. In some embodiments, a subject or population has an ordinal score of 6 or 7, e.g., on the ordinal scale described in Example 38. In some embodiments, a subject or population has an ordinal score of 5, e.g., on the ordinal scale described in Example 38. In some embodiments, a subject or population has an ordinal score of 4, e.g., on the ordinal scale described in Example 38. In some embodiments, a subject or population has an ordinal score of 5, e.g., on the ordinal scale described in Example 38.

[0225] In some embodiments, a subject or population is receiving or has received non- invasive ventilation and/or high-flow oxygen. In some embodiments, a subject or population is receiving or has received non-invasive ventilation and/or high-flow oxygen and has an ordinal score of 5, e.g., on the ordinal scale described in Example 38.

[0226] In some embodiments, a subject or population has a fraction of inspired oxygen (FiCh) of greater than about 40%. Fraction of inspired oxygen can be measured by any method known in the art. In some embodiments, a FiCh of greater than about 40% corresponds to greater than about 5 L/min with a nasal cannula and/or greater than about 10 L/min with a venturi mask and/or greater than about 8 L/min with a conventional mask and/or using a mask with oxygen reservoir. In some embodiments, a subject or population has a FiCh of greater than about 40% and an ordinal score of 4, e.g., on the ordinal scale described in Example 38. [0227] In some embodiments, a subject or population has a fraction of inspired oxygen (FiCk) of less than about 40%. Fraction of inspired oxygen can be measured by any method known in the art. In some embodiments, a FiCk of less than about 40% corresponds to less than about 5 L/min with a nasal cannula and/or less than about 10 L/min with a venturi mask and/or less than about 8 L/min with a conventional mask and/or using a mask with oxygen reservoir. In some embodiments, a subject or population has a FiCk of less than about 40% and an ordinal score of 4, e.g., on the ordinal scale described in Example 38.

[0228] In some embodiments, a subject or population has an oxygen saturation (Sa02) of about 94% or less (e.g., about 93%, about 92%, about 91%, about 90%, about 89%, about 88%, about 87%, about 86%, or about 85%, or less), e.g., while breathing ambient air.

[0229] In some embodiments, a subject or population has a ratio of partial pressure of oxygen (Pa02) to fraction of inspired oxygen (Fi02) that is about 300 mmHg or less (e.g., less than about 300 mmHg, less than about 200 mmHg, or less than about 100 mmHg).

[0230] In some embodiments, a subject or population is not suffering from or susceptible to a malignancy (e.g., an active malignancy). In some embodiments, a subject or population is not receiving treatment for a malignancy (e.g., an active malignancy). In some embodiments, a subject or population does not have a history of solid or hematological malignancies (e.g., within the past 5 years). In some embodiments, a subject or population is not suffering from or susceptible to a malignancy (e.g., an active malignancy) other than a basal or squamous cell carcinoma-in-situ of the skin that was diagnosed more than 2 years prior. In some embodiments, a subject or population thereof is not suffering from an active malignancy or has not suffered from a solid, metastatic or hematologic malignancy (e.g., within 5 years prior to administration of Compound 1 therapy). In some embodiments, a subject or population thereof has suffered from a basal or squamous cell carcinoma of the skin that has been treated and/or removed.

[0231] In some embodiments, a subject or population is suffering from or susceptible to a malignancy (e.g., an active malignancy). In some embodiments, a subject or population is receiving treatment for a malignancy (e.g., an active malignancy). In some embodiments, a subject or population has a history of solid or hematological malignancies (e.g., within the past 5 years). In some embodiments, a subject or population is suffering from or susceptible to a malignancy (e.g., an active malignancy) other than a basal or squamous cell carcinoma-in-situ of the skin that was diagnosed more than 2 years prior. In some embodiments, a subject or population thereof is suffering from an active malignancy or has suffered from a solid, metastatic or hematologic malignancy (e.g., within 5 years prior to administration of Compound 1 therapy). In some embodiments, a subject or population thereof has not suffered from a basal or squamous cell carcinoma of the skin that has been treated and/or removed. In some embodiments, a subject or population thereof is suffering from or has suffered from glioma, colon cancer, or pancreatic cancer.

[0232] In some embodiments, a subject or population thereof has not been assessed for an active malignancy or a history of solid or hematological malignancies. In some embodiments, a subject or population thereof has not been assessed for an active malignancy or a history of a solid, metastatic, or hematologic malignancy. In some embodiments, a subject or population thereof has an unknown malignancy status (i.e., an unknown medical history with respect to malignancies).

[0233] In some embodiments, a subject or population does not have an alanine aminotransferase (ALT) level greater than three times upper limit of normal (ULN) at baseline. [0234] In some embodiments, a subject or population does not have an aspartate transaminase (AST) level greater than three times ULN at baseline.

[0235] In some embodiments, a subject or population does not have a total bilirubin level greater than two times ULN at baseline.

[0236] In some embodiments, a subject or population does not require treatment with CYP1A2 inhibitors. In some embodiments, a subject or population does not require treatment with ciprofloxacin and/or fluvoxamine. In some embodiments, a subject or population is not receiving a CYP1A2 inhibitor. In some embodiments, a subject or population is not receiving ciprofloxacin or fluvoxamine. In some embodiments, a subject or population has not received ciprofloxacin or fluvoxamine (e.g., on the day(s) Compound 1 is administered and/or for 24 hours after last infusion of Compound 1). In some embodiments, a subject or population has not consumed a caffeinated beverage (e.g., on the day(s) Compound 1 is administered and/or for 24 hours after last infusion of Compound 1).

[0237] Non-limiting examples of CYP1A2 inhibitors include alosetron, caffeine, ciprofloxacin, duloxetine, fluvoxamine, melatonin, ramelteon, selegiline, tacrine, tasimelteon, tizanidine, and theophylline. [0238] In some embodiments, a subject or population is not receiving or has not received any other investigational drug product or procedure.

[0239] In some embodiments, a subject or population is not a recipient of a solid organ and/or hematopoietic cell transplantation.

[0240] In some embodiments, a subject or population is not suffering from end stage renal disease. In some embodiments, a subject or population is not being treated with maintenance hemodialysis or peritoneal dialysis, e.g., prior to the same hospitalization in which the subject or population is receiving Compound 1 therapy. In some embodiments, renal replacement therapy (RRT) is initiated during the same hospitalization in which the subject or population is receiving Compound 1 therapy.

[0241] In some embodiments, a subject or population is male and/or nonpregnant females.

In some embodiments, a subject or population is not pregnant or breastfeeding.

[0242] In some embodiments, a subject or population is adult (e.g., 18 years of age or older).

Administration

[0243] In some embodiments, a composition that provides Compound 1, as described herein, can be administered in accordance with methods (e.g., according to a regimen) provided herein. [0244] In some embodiments, a composition that provides Compound 1 is administered intravenously. In some embodiments, a composition that provides Compound 1 is administered (e.g., intravenously) over about 10 min, about 20 min, about 30 min, or about 40 min. In some embodiments, a composition that provides Compound 1 is administered intravenously in an amount suitable to provide about 0.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 6 mg/kg, or about 8 mg/kg Compound 1. In some embodiments, a composition that provides Compound 1 is administered intravenously at an infusion rate suitable to provide about 0.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 6 mg/kg, or about 8 mg/kg Compound 1 over about 10 min, about 20 min, about 30 min, or about 40 min. In some embodiments, provided formulations are administered as an infusion over about 30 min in an amount suitable to provide about 2 mg/kg Compound 1.

[0245] In some embodiments, methods provided herein comprise periodic administration of Compound 1 (e.g., three or four infusions of Compound 1 separated by 24 (± 2) hours). In some embodiments, methods provided herein comprise administration of one, two, three, four or five infusions of Compound 1 separated by a regular interval. In some embodiments, methods provided herein comprise administration of six, seven, eight, nine, or ten infusions of Compound 1 separated by a regular interval. In some such embodiments, a regular interval can be about 24 hours, about 30 hours, or about 36 hours. In some such embodiments, a regular interval can be about 12 hours (e.g., 12 hours ± 2 hours).

[0246] In some embodiments, methods provided herein comprise periodic administration of Compound 1 throughout a course of treatment (e.g., a course of treatment of about 1, about 2, about 3, about 4, or about 5 days). In some embodiments, Compound 1 is administered once daily throughout a course of treatment (e.g., a course of treatment of about 1, about 2, about 3, about 4, or about 5 days). In some embodiments, a course of treatment is about 3 days. In some embodiments, a course of treatment is about 4 days.

[0247] In some embodiments, methods provided herein comprise administration (e.g., intravenous administration) of Compound 1 (e.g., 2 mg/kg) twice a day for 5 days. In some embodiments, a composition that provides Compound 1 is first administered within about 6 hours of an initiating event (e.g., randomization or acute injury). In some embodiments, a composition that provides Compound 1 is administered for a second time within about 12 hours (e.g., within 10 hours, within 12 hours, or within 14 hours) from the time of first administration. In some embodiments, a composition that provides Compound 1 is administered for a third, fourth, fifth, sixth, seventh, eighth, ninth, and/or tenth time within about 12 hours (e.g., within 10 hours, within 12 hours, or within 14 hours) from the time of a prior administration.

[0248] In some embodiments, methods provided herein comprise administration (e.g., intravenous administration) of Compound 1 (e.g., 2 mg/kg) once a day for 4 days. In some embodiments, a composition that provides Compound 1 is first administered within about 6 hours, about 12 hours, about 18 hours, about 24 hours, about 30 hours, about 36 hours, or about 42 hours of an initiating event (e.g., randomization or acute injury). In some embodiments, a composition that provides Compound 1 is administered for a second time within about 20 hours, about 22 hours, about 24 hours, about 26 hours, or about 28 hours from the time of first administration. In some embodiments, a composition that provides Compound 1 is administered for a third time within about 20 hours, about 22 hours, about 24 hours, about 26 hours, or about 28 hours from the time of second administration. In some embodiments, a composition that provides Compound 1 is administered for a fourth time within about 20 hours, about 22 hours, about 24 hours, about 26 hours, or about 28 hours from the time of third administration.

[0249] In some embodiments, methods provided herein comprise administration (e.g., intravenous administration) of Compound 1 (e.g., 2 mg/kg) once a day for 3 days.

[0250] In some embodiments, a composition that provides Compound 1 is first administered within about 12 hours, about 18 hours, about 24 hours, about 30 hours, about 36 hours, or about 42 hours of an initiating event (e.g., randomization or acute injury). In some embodiments, a composition that provides Compound 1 is administered for a second time within about 20 hours, about 22 hours, about 24 hours, about 26 hours, or about 28 hours from the time of first administration. In some embodiments, a composition that provides Compound 1 is administered for a third time within about 20 hours, about 22 hours, about 24 hours, about 26 hours, or about 28 hours from the time of second administration.

[0251] In some embodiments, the present disclosure provides a method comprising intravenously administering to a subject or population a formulation comprising: about 6 mg/mL Compound 1; about 20% (w/v) to about 40% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 5% (w/v) to about 15% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline.

[0252] In some embodiments, the present disclosure provides a method comprising intravenously administering to a subject or population a formulation comprising: about 6 mg/mL Compound 1; about 30% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 6% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline.

[0253] In some embodiments, the present disclosure provides a method comprising steps of: (i) providing a first formulation of Compound 1; (ii) diluting the first formulation with normal saline to give a second formulation of Compound 1; and (iii) administering the second formulation to a subject or population in need thereof (e.g., as described herein). In some such embodiments, a first formulation of Compound 1 is more concentrated (e.g., 10 mg/mL) than a second formulation of Compound 1 (e.g., 6 mg/mL). In some embodiments, provided methods further comprise diluting the first formulation under aseptic conditions. In some embodiments, provided methods further comprise diluting the first formulation within 1 day, 2 days, or 3 days prior to administering the second formulation.

[0254] In some embodiments, a first formulation of Compound 1 comprises: about 10 mg/mL Compound 1; about 40% (w/v) to about 60% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 5% (w/v) to about 15% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline; and a second formulation of Compound 1 comprises: about 6 mg/mL Compound 1; about 20% (w/v) to about 40% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 5% (w/v) to about 15% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline.

[0255] In some embodiments, a first formulation of Compound 1 comprises: about 10 mg/mL Compound 1; about 50% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 10% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline; and a second formulation of Compound 1 comprises: about 6 mg/mL Compound 1; about 30% (w/v) polyethylene glycol (e.g., polyethylene glycol 300); about 6% (w/v) polysorbate (e.g., polysorbate 80); and one or more aqueous components selected from phosphate buffered saline and normal saline.

[0256] In some embodiments, Compound 1 therapy as described herein is initiated within 24, 20, 28, 26, 24, 12, 10, 8, 6, 4, 2, or 1 hours of initiation of mechanical ventilation; in some embodiments, Compound 1 therapy is initiated within 10, 8, 6, 4, or 2 hours of such ventilation initiation; in some embodiments within 4 hours.

[0257] In some embodiments, Compound 1 is administered by IV infusion and/or at a dose of about 2 mg/kg over a period of time (e.g., about 30 min).

[0258] In some embodiments, Compound 1 is administered in a plurality of doses, e.g., of fixed doses.

[0259] In some embodiments, Compound 1 is administered according to a dosing regimen over a period of time, for example during some or all of which the patient(s) is/are ventilated. In some embodiments, such period of time may be 1, 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 days or more; in some embodiments, such period of time may be about 10 days or fewer (e.g., about 8, 7, 6, 5, or 4 days).

[0260] In some embodiments, one or more of the following is monitored before, during, and/or after Compound 1 therapy: arterial blood gasses (ABGs) or draws from an arterial line to calculate the A-a gradient, A-a ratio, or the P/F ratio (Pa02/Fi02), in some embodiments at a plurality of time points (e.g., over the first 24-38 hours), frequently for the first 48 hours; in some embodiments such assessment s) may establish impact on (e.g., improvement of) gas exchange ventilator (in hours), days in ICU, days in hospital, % of patients discharged alive, or % of patients who progress to multi-organ failure.

[0261] In some embodiments, one or more of the following is monitored before, during, and/or after Compound 1 therapy: score on an 8-point ordinal scale (e.g., as described in Example 38), Sp02/Fi02, high-sensitivity C-reaction protein (HS-CRP), absolute lymphocyte count, serum ferritin, serum interleukin-6 (H-6), serum myoglobin, D-dimer, creatine phosphokinase (CPK), CPK-MB, troponin (e.g., troponin I), and LDH. In some embodiments, such assessment s) may establish impact on (e.g., improvement of) oxygenation, days with hypoxemia, ventilator-free days, percentage of patients discharged alive, percentage of patients requiring mechanical ventilation and/or ECMO, percentage of patients in ICU, days in ICU, days in hospital (e.g., among survivors), and/or percentage of patients with secondary bacterial and/or fungal infections.

[0262] In some embodiments, Compound 1 therapy as described herein is not administered to patients who have multi-organ failure. Exemplary Embodiments

[0263] The following numbered embodiments, while non-limiting, are exemplary of certain aspects of the disclosure:

1. A method of treating a respiratory disease, disorder or condition, the method comprising: administering to a subject susceptible to or suffering from a respiratory disease, disorder or condition a composition that provides (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole.

2. The method of embodiment of 1, wherein the disease, disorder or condition is characterized by pulmonary edema, pulmonary epithelial cell apoptosis, inflammatory cell infiltration, impaired oxygenation, hypoxemia and/or lung fibrosis.

3. The method of embodiment 1 or 2, wherein the subject shows one or more symptoms or features of the disease, disorder or condition.

4. The method of embodiment 3, wherein the disease, disorder or condition is or comprises acute lung injury or acute respiratory distress syndrome and the one or more symptoms or features are selected from labored breathing, rapid breathing, muscle fatigue, general weakness, low blood pressure, shortness of breath, and confusion, and combinations thereof.

5. The method of any one of the preceding embodiments, wherein the subject is resistant to oxygen therapy.

6. A method of treating acute lung injury or acute respiratory distress syndrome in a subject in need thereof, the method comprising administering to the subject a composition that provides (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole.

7. A method comprising: administering to a subject who is suffering from or susceptible to a respiratory disorder a composition that provides (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole.

8. A method comprising: administering to a subject who is receiving or has received therapy with extracorporeal membrane oxygenation a composition that provides (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole.

9. The method of any one of the preceding embodiments, wherein the subject is suffering from or susceptible to acute lung injury.

10. The method of any one of the preceding embodiments, wherein the subject is suffering from or susceptible to acute respiratory distress syndrome. 11. The method of any one of the preceding embodiments, wherein the subject is suffering from or susceptible to pneumonia.

12. The method of embodiment 11, wherein the pneumonia has been confirmed by chest imaging.

13. The method of any one of the preceding embodiments, wherein the subject has an oxygen saturation of 94% or less when breathing ambient air.

14. The method of any one of the preceding embodiments, wherein the subject has a ratio of partial pressure of oxygen to fraction of inspired oxygen of 300 mm Hg or less.

15. The method of any one of the preceding embodiments, wherein the subject is suffering from or susceptible to pulmonary dysfunction and is hospitalized with COVID-19 pneumonia.

16. The method of any one of the preceding embodiments, wherein the subject has received a positive reverse-transcriptase-polymerase-chain-reaction assay for SARS-CoV-2 from a respiratory tract sample.

17. The method of any one of the preceding embodiments, wherein the subject is suspected of having or has been diagnosed with a viral infection.

18. The method of embodiment 17, wherein the viral infection is a respiratory viral infection.

19. The method of embodiment 17 or 18, wherein the viral infection is a coronavirus infection.

20. The method of embodiment 19, wherein the coronavirus infection is COVID-19 infection.

21. The method of embodiment 17, wherein the viral infection is an influenza infection.

22. The method of embodiment 17, wherein the subject is receiving or has received antiviral therapy.

23. The method of embodiment 22, wherein the antiviral therapy is selected from lopinavir, ritonavir, chloroquine, hydroxychloroquine, azithromycin, and redesivir, and combinations thereof.

24. The method of any one of the preceding embodiments, wherein the subject is suffering from or has experienced a traumatic lung injury.

25. The method of embodiment 24, wherein the traumatic lung injury is a blunt trauma, a thermal burn or a chemical burn. 26. The method of any one of the preceding embodiments, wherein the subject is receiving or has received therapy with extracorporeal membrane oxygenation.

27. The method of any one of the preceding embodiments, wherein the subject is or has been admitted to an intensive care unit.

28. The method of any one of the preceding embodiments, wherein the subject is receiving or has received supplemental oxygen.

29. The method of any one of the preceding embodiments, wherein the subject is receiving or has received mechanical ventilation.

30. The method of any one of the preceding embodiments, wherein the subject is receiving or has received endotracheal intubation.

31. The method of any one of the preceding embodiments, wherein the subject is further characterized by impaired kidney function.

32. The method of any one of the preceding embodiments, wherein the subject is suffering from or susceptible to renal dysfunction.

33. The method of any one of the preceding embodiments, wherein the subject is suffering from acute kidney injury.

34. The method of any one of the preceding embodiments, wherein the subject is suffering from a renal ischemia/reperfusion injury, renal failure, renal fibrosis, or a renal trauma.

35. The method of any one of the preceding embodiments, wherein the subject is receiving or has received dialysis.

36. The method of any one of the preceding embodiments, wherein the subject has undergone renal transplantation.

37. The method of any one of the preceding embodiments, wherein the subject is not suffering from an active malignancy and has not suffered from a solid or hematologic malignancy.

38. The method of any one of embodiments 1-37, wherein the subject has suffered from a basal or squamous cell carcinoma-in-situ of the skin that was diagnosed more than 2 years prior to administration of the composition providing (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole.

39. The method of any one of embodiments 1-38, wherein the subject has not suffered from a solid or hematologic malignancy within 5 years prior to administration of the composition providing (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole. 40. The method of any one of embodiments 1-36, wherein the subject is suffering from an active malignancy or has suffered from a solid or hematologic malignancy.

41. The method of any one of embodiments 1-36 and 40, wherein the subject has not suffered from a basal or squamous cell carcinoma-in-situ of the skin that was diagnosed more than 2 years prior to administration of the composition providing (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole.

42. The method of any one of embodiments 1-36, 40 and 41, wherein the subject has suffered from a solid or hematologic malignancy within 5 years prior to administration of the composition providing (E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole.

43. The method of any one of embodiments 1-36, wherein the subject has not been assessed for an active malignancy or a history of a solid or hematologic malignancy.

44. The method of any one of the preceding embodiments, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, a decreased mortality rate relative to a comparable reference population.

45. The method of any one of the preceding embodiments, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, increased lung function relative to a comparable reference population.

46. The method of any one of the preceding embodiments, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, one or more of:

(i) increased pulmonary output;

(ii) increased arterial oxygen (PaCh);

(iii) decreased arterial carbon dioxide (PaCCh);

(iv) increased ratio of PaCh/FiC ;

(v) decreased lung injury score;

(vi) decreased lung hydroxyproline concentration;

(vii) decreased lung collagen level;

(viii) decreased lung TGFp l concentration; and (ix) increased blood pH, relative to a comparable reference population. 47. The method of any one of the preceding embodiments, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, one or more of:

(i) reduced time to improvement in oxygenation for at least 48 hours;

(ii) reduced mean change from baseline on an 8-point Ordinal Scale;

(iii) reduced time to improvement from admission in one category on an 8-point Ordinal Scale;

(iv) reduced number of days with hypoxemia;

(v) reduced time to improvement in oxygenation for at least 48 hours by clinical severity;

(vi) increased number of ventilator free days in the first 30 days;

(vii) reduced number of patients requiring initiation of mechanical ventilation and/or ECMO;

(viii) reduced number of patients admitted into an intensive care unit;

(ix) reduced number of days of hospitalization; and

(x) reduced number of patients with secondary bacterial and/or fungal infection, relative to a comparable reference population.

48. The method of any one of the preceding embodiments, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, one or more of:

(i) increased proportion of patients alive without need for mechanical ventilation and free of need for renal replacement therapy on an ongoing basis at Day 28;

(ii) reduced all-cause mortality;

(iii) increased proportion of patients not requiring mechanical ventilation at Day 28;

(iv) increased proportion of patients not requiring renal replacement therapy on an ongoing basis at Day 28;

(v) increased number of ventilator-free days at Day 28;

(vi) reduced proportion of patients requiring initiation of mechanical ventilation and/or ECMO at Day 28;

(vii) reduced proportion of patients requiring initiation of renal replacement therapy at Day 28; (viii) reduced number of days to renal recovery in subjects who were on RRT at time of randomization;

(ix) reduced number of ICU days at Day 28;

(x) reduced score on an ordinal scale at Day 28; and

(xi) reduced number of days to hospital discharge from randomization, relative to a comparable reference population.

49. The method of any one of the preceding embodiments, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, increased kidney function relative to a comparable reference population.

50. The method of any one of the preceding embodiments, wherein the composition is administered according to a regimen established to achieve, when administered to a relevant population, one or more of:

(i) decreased serum creatinine concentration;

(ii) increased estimated glomerular filtration rate;

(iii) decreased blood urea nitrogen concentration;

(iv) increased urine output;

(v) decreased kidney TGF i concentration; and

(vi) lesser incidence of dialysis, relative to a comparable reference population.

51. The method of any one of the preceding embodiments, wherein the composition is administered intravenously.

52. The method of any one of the preceding embodiments, wherein the composition is administered in a dose of about 2 mg/kg.

53. The method of any one of the preceding embodiments, wherein the composition is administered twice daily.

54. The method of any one of the preceding embodiments, wherein the composition is administered for 5 consecutive days.

55. The method of any one of the preceding embodiments, wherein the composition is administered once daily.

56. The method of any one of the preceding embodiments, wherein the composition is administered for 4 consecutive days. 57. The method of any one of the preceding embodiments, wherein the composition comprises:

(E)-3-[2-(2-thienyl)vinyl]-lH-pyrazole; polyethylene glycol; and poly sorb ate.

[0264] Many modifications and variations of the embodiments described herein may be made without departing from the scope, as is apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only.

EXAMPLES

Example 1. Synthesis of Compound 1

[0265] As described in Example 7 of WO 2004/058721, Compound 1 was synthesized according to Scheme 1.

Scheme 1

[0266] To a solution of diethoxyphosphorylacetaldehyde tosylhydrazone (3, 75 g) in 400 mL of THF was added 11.6 g of 60% NaH in portions, and the solution was stirred for 15 min. The solution was cooled to 0 °C, and then a solution of 3-(2-thienyl)acrylaldehyde (2) in 100 mL THF was added dropwise. The reaction was then stirred at room temperature for 1 hour, then at reflux for 1 hour. The reaction mixture was partitioned between 5% NaH PCh and ethyl acetate. The organic layer was separated, washed with water and brine, dried over magnesium sulfate, filtered and concentrated to provide the crude product as a brown oil. Purification via silica gel column chromatography afforded 8.3 g of a yellow powder. Trituration with dichlormethane/hexane afforded 4.4 g of yellow powder having >98% purity: 'H NMR (CDCb) d 6.47 (d, 1 H, J= 1.5 Hz), 6.93 (d, 1 H, J= 9.9 Hz), 6.99 (dd, 1 H, J= 3.9, 2.1 Hz), 7.06 (d, 1 H, J= 2.1 Hz), 7.20 (d, 1 H, J= 3.9 Hz), 7.22 (d, 1 H, 7= 9.9 Hz), 7.57 (d, 1 H, 7= 1.5 Hz). [0267] The conversion of 3-arylacrylaldehydes into substituted pyrazoles via treatment with diethoxyphosphorylacetaldehyde tosylhydrazone (3) is described in the literature (Almirante, N.; Cerri, A.; Fedrizzi, G.; Marazzi, G.; Santagostino, M. Tetrahedron Lett. 1998, 39, 3287). 3-(2- thienyl)acrylaldehyde (2) was prepared from 2-thienaldehyde and acetaldehyde as described in Heskin, H, Miller, R. E., Nord, F. F. J Org. Chem. 1951, 16, 199.

Example 2. Compound 1 Stimulated Endothelial Cell and Bronchial Cell Proliferation but not Fibroblast Cell Proliferation

[0268] An important pulmotrophic activity of HGF is induction of endothelial and epithelial cell proliferation but not that of fibroblasts. Endothelial cells (HUVECs), bronchial epithelial cells (HBECs) and lung fibroblasts (MRC-5) were grown to semi confluence in serum medium for 24 hours. Cells were then serum starved for 2 hours, followed by the addition of vehicle, HGF (50 ng/ml) or Compound 1 (36 mM) for 24 hours. The cells were washed with PBS and [3H]-thymidine incorporation determined as a measure of proliferation. Compound 1 produced a similar increase in endothelial (FIG. 1A) and bronchial (FIG. IB) cells proliferation as HGF, but not fibroblasts (FIG. 1C).

Example 3. Compound 1 Decreased Pulmonary Edema

[0269] Adult male C57BL/6 mice were intratracheally (IT) instilled with 50 pL of saline (sham) or bleomycin (1 mg/kg). The bleomycin cohort was randomized to vehicle (n=20) or Compound 1 treatment (2 mg/kg, IP, QD, n=20). Mice were sacrificed at 96 hr. Pulmonary edema was determined by lung wet/dry wt ratio. Other lungs were formalin fixed to obtain H&E sections. As seen in FIG. 2A, treatment with Compound 1 significantly attenuated pulmonary edema. H&E sections showed significant red cell and neutrophil infiltration in the vehicle cohort accompanied by protein flooding in the alveolar spaces. The Compound 1 treated cohort was markedly free of these abnormalities (FIG. 2B).

[0270] Delayed/therapeutic Compound 1 treatment also attenuated pulmonary edema. Adult male C57BL/6 mice were IT instilled with 50 pL of saline or bleomycin (1 mg/kg). The bleomycin cohort was randomized to vehicle (n=16) or Compound 1 (n=16; 2 mg/kg, IP, QD) started 24 hr after bleomycin and continued daily for 3 days. Mice were sacrificed 96 hours after bleomycin instillation. Pulmonary edema was determined by lung wet/dry wt ratio as above. As seen in FIG. 2C delayed treatment of Compound 1 attenuated pulmonary edema.

Example 4. Compound 1 protects against TGFpi-induced acute lung injury.

[0271] A variety of pulmonary insults stimulate production and release of TGFpi into the pulmonary parenchyma, which lead to acute cell death. Transforming growth factor beta (TGFp l ) is a protein that controls proliferation, cellular differentiation and other functions in most cells, and it is known to be a critical mediator in acute lung injury. A small colony of mice with a lung-specific TGFpi transgene that is regulated by a doxycycline-(dox) induced promoter was obtained. Dox-fed mice (0.5 mg/mL in water) were treated with Compound 1 (2 mg/kg, IP, QD, n=8) starting 3 hours after the induction of TGFpi expression. Compound 1 significantly improved the survival of mice in a 10 day treatment period (FIG. 3 A). Some mice were sacrificed 3 days after TGFpi induction, and the Compound 1 treated cohort was shown to have decreased pulmonary cell death (FIG. 3B) and improved pulmonary epithelial regeneration (by PCNA) vs vehicle cohort (FIG. 3C).

Example 5. Compound 1 attenuates lipopolysaccharide (LPS) induced shock associated ALI in a mouse model.

[0272] Bacterial endotoxin (LPS; E.coli, Sigma) was prepared in sterile PBS and 50 pL instilled IT (2.5 mg/kg body weight) to adult male C57BL/6 mice. A sham group of mice (n=4) were instilled with 50 pL PBS. In a pilot study, LPS challenged mice (n=8/group) were randomized to vehicle or Compound 1 (2 mg/kg, IP, QD) cohorts and treated for 10 days. Then lungs were evaluated for histopathological injury score by two independent observers (FIG. 4B) and TUNEL for apoptosis. Compound 1 treatment significantly attenuated LPS/shock associated lung injury score (FIG. 4A) and decreased apoptotic cell death (FIG. 4C) compared to the vehicle cohort.

Example 6. Compound 1 protects against chlorine (CL) toxicity

[0273] The acute effects of Ch inhalation can range from mild respiratory mucus membrane irritation to marked denudation of the mucosa, pulmonary infiltration, pulmonary edema and death. Recovery from Ck-induced lung injury requires repair and/or regeneration of the epithelial layer. To assess whether Compound 1 can aid in the prevention or recovery from Ch- induced pulmonary damage, C57BL/6 mice were exposed to Ch. Mortality of 27% was observed after 5 days and a significant increase in pulmonary infiltration was determined by an increase in the protein concentration in the bronchial alveolar lavage (BAL) fluid. Treatment with Compound 1 (2 mg/kg, IP, QD, initiated 3 hours after Ch exposure) resulted in a marked increase in animal survival (FIG. 5A) and a significant reduction in pulmonary infiltration (FIG. 5B).

Example 7. Compound 1 enhances pulmonary output and arterial oxygen levels in established emphysema model

[0274] We assessed the efficacy of Compound 1 in two studies using the porcine pancreatic elastase (PPE)-induced emphysema rat model. PPE was prepared in PBS at 250 U/kg/body weight and 250 uL instilled intratracheally. A PBS-instilled group was also set up. Two weeks after PPE, established emphysema was verified by a marked induction of turbidity in BAL, indicating inflammatory infiltration. Animals were then randomized into vehicle or Compound 1 (15 mg/kg, 45 mg/kg, PO, QD) groups. After three weeks of treatment and immediately prior to sacrifice, pulmonary output was measured by determining end-expiration volume. Lung function in anesthetized but unassisted (no ventilator) animals was obtained using a vertical U-tube manometer. PPE-instilled animals had significantly reduced expiration volume compared to untreated animals, and Compound 1 was found to significantly increased pulmonary output in the 45 mg/kg group compared to the vehicle control (p=0.003) (FIG. 6A). Both dose levels of Compound 1 markedly increased the arterial oxygen levels as compared to the vehicle-treated group, and restored Pa02 to normal levels (FIG. 6B).

Example 8. Compound 1 reduces hemorrhagic shock (HS) induced lung injury in a rat model.

[0275] Rats were exposed to 90 min ischemia by withdrawing 30 mL of blood from the carotid artery. Vehicle or Compound 1 (2 mg/kg, IV, n=6/group) was given one hour after the onset of reperfusion during 180 min reperfusion with 30 mL of lactated Ringer’s, and again 12 hours later. At 24 hrs, Compound 1 decreased lung histopathology (FIG. 7A and FIG. 7B). Example 9. Compound 1 attenuates pulmonary 90 min ischemia-72 hr reperfusion injury. [0276] Adult male Sprague Dawley rats were subjected to 90 min left pulmonary global normothermic ischemia and 72 hr reperfusion. At the onset of reperfusion, animals (n=5/group) were randomized to vehicle or Compound 1 (2 mg/kg, IV) administration, once daily. Lung function in anesthetized but unassisted (no ventilator) animals was obtained using a vertical U- tube manometer. Briefly, tracheal airflow was connected using a Luer-lock adaptor to one arm of the tube. The U-tube was partially filled with water allowing for a large volume of respiratory air reserve. The U-tube was placed against a graded scale such that excursion of water in the other arm of the tube could be recorded. End-expiration air volume was calculated by the equations shown below:

[0277] Single lung function immediately prior to sacrifice was also evaluated by obtaining blood gas levels (Fi02 =1.0) from the left pulmonary vein using an Abbot I-STAT blood gas analyzer. Lungs were evaluated histopathologically for pulmonary epithelial regeneration (PCNA). Treatment with Compound 1 was associated with significant improvement in postischemic end expiration air volume (FIG. 8A), blood pH (FIG. 8B), blood oxygen levels (FIG. 8C and FIG. 8D), and enhanced pulmonary epithelial regeneration and preservation of lung microarchitecture (FIG. 8E).

Example 10. Compound 1 attenuates canine warm lung ischemia-reperfusion (I-R) injury [0278] This example simulates a “marginal lung” from a cardiac arrest donor. Adult male dogs were exposed to 90 min of left lung normothermic ischemia by pulmonary artery occlusion and 180 min reperfusion. Seven subjects were treated with Compound 1 (10 mg/kg, iv) and six were treated with vehicle at reperfusion. Since preclinical pharmacokinetic (PK) data indicate that the plasma Compound 1 level (Cmax and AUC) in rats is ~ 5X higher than in dogs, due to more avid clearance in dogs, a Compound 1 dose, of 10 mg/kg (5X higher than in rats) was used. Treatment with Compound 1 decreased pulmonary edema (FIG. 9A) and improved arterial oxygenation levels (FIG. 9B). Additionally, there was a marked attenuation of alveolar thickening in dogs treated with Compound 1 as represented in H&E lung sections, indicating protection of the pulmonary architecture at 180 minutes post-reperfusion (FIG. 9E). Compound 1 treatment also decreased inflammatory cytokines IL-1 (FIG. 9C) and IL-6 (FIG. 9D), known promoters of lung graft injury, in BAL significantly.

Example 11. Determination the dose response and therapeutic time window of Compound 1 in a two-hit HS+LPS model of ALI/ARDS

[0279] Pre-catheterized rats are used. Hemorrhagic shock (HS) is initiated by blood withdrawal and reduction of the mean arterial pressure (MAP) to 40 mmHg within 15 min. After a hypotensive period of 60 min, rats are resuscitated by transfusion of the shed blood and Ringer’s lactate (RL) in a volume equal to that of shed blood, over a period of 2 h. At 1 hour after resuscitation, a tracheotomy is performed with intratracheal (IT) instillation of either LPS or saline (sham), followed by mechanically ventilated breaths using a rodent ventilator. In a pilot study, a small group of sham, HS+LPS challenged rats are sacrificed after 6 hr to evaluate the extent of acute lung injury and histopathological score. After confirmation of injury score, series- 1 study is conducted.

[0280] Dose response study. After 6 hr of LPS+HS challenge, pre-catheterized rats are randomized to vehicle and/or Compound 1 cohorts at 1, 3 and 10 mg/kg, (IV, QD n=16/dose) and dosing daily for 7 days. At the end of treatment, rats are sacrificed for analysis of lung injury.

[0281] Mortality, body weights, and lung weights are recorded, and serum is collected from all animals and stored at -70 °C until used. Histological scoring of lung injury from H&E stained sections is evaluated for all groups. Based on the lung injury scores, in order to gain a better understanding of the efficacy - exposure relationship of Compound 1, only lung tissues from the vehicle and optimally efficacious dose groups are further processed for TUNEL to evaluate protective effects of Compound 1 on epithelial cell apoptosis, and serum levels of Compound 1 determined by LC-MS/MS from the optimal Compound 1 dose groups. Immuno-histochemical staining is performed for c-Met upregulation and for a key inflammatory marker protein, F4/80, from the formalin preserved tissue sections. Left lungs are evaluated for MPO activity indicating PMN infiltration. The proinflammatory cytokines in the serum and lung tissue lysates (IL- 1 />, IL- 6, IL-10 and TNF-a) are evaluated from animals from the optimally efficacious dose groups. [0282] Therapeutic window study. In a pilot study, a small group of sham, HS+LPS challenged rats are sacrificed after 24 hrs to evaluate the extent of established acute lung injury and histopathological score. After confirmation of established lung injury, series-2 study is conducted. HS+LPS challenged rats after 24 hrs, are randomized to vehicle or Compound 1 groups. The optimal dose of Compound ldetermined from series-1 study is administered via the IV route daily for 7 days and 14 days.

[0283] In addition to the series- 1 study described end points, at the end of treatment, MAP and pulmonary function tests are performed using the Flexivent system to obtain ventilator mechanics: peak airway pressure, pause airway pressure, lung static and dynamic pressure. Once the collection of functional data from each animal has been confirmed, blood is collected for gas analysis, arterial and venous oxygenation (p02), arterial and venous saturation (PC02), pH, base excess electrolytes (sodium, calcium and potassium), hematocrit, hemoglobin, complete blood count with differential. Animals are then sacrificed and the efficacy measures as in series-1 study are conducted.

[0284] Following the treatment period, rats are anesthetized, tracheostomized, and placed on the FlexiVent system for forced oscillatory measurements. The use of the Flexivent system (SCIREQ, Montreal, QC, Canada) includes onsite demonstration and training using sentinel animals to assure proficiency in conducting lung function measurements. Rats are ventilated with a tidal volume of 10 mL/kg at a frequency of 150 breaths/min and a PEEP of 3 cm H2O to prevent alveolar collapse. Total lung capacity (TLC), Snapshot, Quickprime-3, and pressure- volume (PV) loops with constant increasing pressure (PVr-P) are consecutively performed using the Flexivent system according to the manufacturer’s protocols. A Snapshot perturbation maneuver uses a three-cycle sinusoidal wave of inspiration and expiration to measure total respiratory system resistance (R), dynamic compliance (C), and elastance (E). A Quickprime-3 perturbation, which produces a broadband frequency (0.5 to 19.75 Hz) over 3 seconds, measures Newtonian resistance which is a measure of central airway resistance (Rn), inertance (I), tissue damping (G), tissue elastance (H) and hysteresivity (eta). PV loops are generated between 30 cm H2O to -30 cm H2O pressure to obtain vital capacity (A), the upper portion of the deflation PV curve (K), quasi-static compliance (Cst) and elastance (Est), and the area of PV loop (Area). All perturbations are performed until three acceptable measurements with coefficient of determination (COD) > 0.9 recorded in each individual rat. In addition, forced vital capacity (FVC) and peak expiratory flow (PEF) are measured using the Flexivent system.

[0285] The primary endpoints for the evaluation of efficacy are evidence of decrease in pathological lung injury score, normalized MAP and improvement in pulmonary function in the Compound 1 treated groups compared to the vehicle cohort. Observed efficacy is expected to be due to decreases in apoptotic cell death, cellular infiltrates, proinflammatory cytokines in the BAL and tissue lysates compared to the vehicle cohort. Efficacy and optimal therapeutic time window for Compound 1 in a disease-relevant model of HS+LPS induced ARDS is expected to be defined using the study described above.

Example 12. Evaluating efficacy of Compound 1 in a smoke inhalation and burn induced ALI/ARDS sheep model

[0286] The efficacy of Compound 1 is tested in a smoke inhalation + skin burn induced trauma associated with ARDS sheep model. The pulmonary function and pathophysiology of acute lung injury secondary to burn and smoke inhalation have been well studied in this large animal model, particularly within 48 hours after injury. The well-established ovine model is clinically relevant because of similarities between human and ovine pulmonary physiology. [0287] Sheep are anesthetized with isoflurane and given a flame burn (40% total body surface area, third degree) and subjected to smoke inhalation injury (48 breaths of cotton smoke, <40°C). The arterial carboxyhemoglobin (COHb) level is determined immediately after the smoke inhalation. Six hours after the skin bum and smoke inhalation injury, sheep are randomized to vehicle and Compound 1 cohorts. A predetermined dose of Compound 1 is administered daily via the IV route for 5 days during which lung functional measurements and regular collection and analysis of blood and urine are performed, then the animals are sacrificed. [0288] Blood and urine are collected every 6 hours. Cardiopulmonary hemodynamics, MAP, pulmonary artery pressure, central venous pressure, left atrium pressure, heart rate, cardiac output, blood gas analysis, arterial and venous oxygenation (P02), arterial and venous saturation (PC02), pH, electrolytes (sodium, calcium and potassium), hematocrit, hemoglobin, CBC with differential, ventilatory mechanics: peak airway pressure, pause airway pressure, and lung static and dynamic pressure are measured at 48 hr and 5 day time points. Then sheep are sacrificed. Lung tissues are frozen and formalin fixed for lung histology.

[0289] Burn and smoke inhalation injury in sheep model. Adult female Merino sheep (body weight, 30-40 kg) are acclimatized at a facility with free access to food and water. Then sheep are surgically prepared under isoflurane anesthesia with a right femoral artery catheter (Intracath, 16GA, 24IN, BD Vascular Access, Sandy, UT), a thermodilution catheter (Model 131F7, Edwards Lifesciences LLC, Irvine, CA), and a left atrial catheter (0.062 in. ID, 0.125 in. OD; Dow Corning, Midland, MI). Animals are given a flame burn (40% total body surface area, third degree) and inhalation injury (48 breaths of cotton smoke, <40°C). The arterial carboxyhemoglobin (COHb) level is determined immediately after the smoke inhalation. After the bum and smoke inhalation injury, all surviving sheep are placed on a ventilator with positive end expiratory pressure set to 5 cm H2O and tidal volume maintained at 15 ml/kg. The sheep are ventilated (Servo Ventilator 300, Siemens-El ema AB, Sweden) with 100% oxygen for the first 3 hrs after injury for rapid clearance of CO in order to reduce COHb. Following this procedure, the fraction of inspired oxygen (Fi02) is adjusted according to blood gas analysis to maintain Pa02 above 80 mm Hg. Respiratory rate is initially set at 20 breaths/minute and there after adjusted to keep PaC02 between 25-35 mm Hg. All sheep are resuscitated with Ringer’s solution, using the formula 4 mL/kg/% burned body surface for 24 hrs and 2 mL/kg/% burned body surface from 24 to 48 hours. After 6 hours of initial skin bum and smoke inhalation injury, sheep are randomized to vehicle and Compound 1 cohorts daily for 5 days and then sacrificed.

Example 13. Compound 1 delayed treatment decreases pulmonary edema.

[0290] Adult male C57BL/6 mice were intratracheally instilled with saline or bleomycin (1 mg/kg). Bleomycin cohort was randomized to vehicle (n= 16) or Compound 1 (2 mg/kg, IP, QD) 24 hr later. Mice were sacrificed at 96 hr and lung wet weight was taken and then kept in oven for 48 hr at 100 °C and dry weight was taken. Pulmonary edema was determined by lung wet/dry weight ratio. Treatment with Compound 1 significantly attenuated pulmonary edema (FIG. 10).

Example 14. Compound 1 attenuates TGFpi-induced mortality and acute lung injury. [0291] A variety of pulmonary insults stimulate production and release of TGFpi into the pulmonary parenchyma which leads to acute cell death. In a genetic murine model of pulmonary TGF i induction (mice with a lung-specific TGFBl transgene under a doxycycline-(dox) inducible promoter, see Vicencio, A.G., et al. Am. J. Respir. Cell. Mol. Biol. 2004 Dec;31(6) 650-56), mice treated with Compound 1 (2 mg/kg, IP, QD, n=8) exhibited improved survival (FIG. 11 A), decreased pulmonary cell death (FIG. 1 IB) and improved pulmonary epithelial regeneration (PCNA) (FIG. 11C) at day 3 (following TGFp l induction). Micro CT images of inflated and formalin fixed lungs suggest that treatment with Compound 1 preserves pulmonary microarchitecture in this model (FIG. 11D).

Example 15. Compound 1 attenuates acute lung injury in a sheep model of smoke inhalation and burn injury.

[0292] Adult female sheep were subjected to smoke inhalation + burn injury and randomized 3 hr later to vehicle or Compound 1 (2 mg/kg, IV, QD). Survivors were sacrificed at 96 hr following injury induction. Treatment with Compound 1 attenuated mortality (FIG. 12A) and improved pulmonary gas exchange (FIG. 12B).

Example 16. Compound 1 prevents progression to pulmonary fibrosis.

[0293] The effect of Compound 1 on pulmonary collagen accumulation was evaluated in a genetic (TSK1/+) mouse model of systemic sclerosis including pulmonary fibrosis. Compound 1 treatment (2 mg/kg, IP, QD, n=5/group) for 12 days reduced pulmonary hydroxyproline content (FIG. 13A) and pulmonary Sirius red staining (FIG. 13B and FIG. 13C), sensitive indices of pulmonary collagen accumulation (fibrosis).

Example 17. Compound 1 is orally efficacious in a TGFpi-induced lung fibrosis in mouse model.

[0294] To evaluate Compound 1 as a potential anti-fibrotic agent in pulmonary fibrosis, Compound 1 was tested to determine if it could affect TGFp l -induced pulmonary fibrosis. Mice (n=12/group) containing a TGFp l transgene under a lung-specific doxycycline-inducible promoter were fed with dox (0.5 mg/mL in water) for 4 weeks. After lung fibrosis was established (Pre R) at 4 weeks, vehicle, Compound 1, and a clinically advanced compound imatinib were administered at 50 mg/kg (PO) for 4 weeks. Compound 1 treatment decreased lung hydroxyproline (HYP) and fibrosis significantly compared to vehicle cohort and imatinib cohort (FIG. 14).

Example 18. Compound 1 treatment mitigates ionizing radiation induced pulmonary cell apoptosis and inflammation.

[0295] In a pilot study, adult male mice were exposed to 7 Gy (@ 1 Gy/min, 137 Cs, at Cold Spring Harbor Laboratories (CSHL), NY and treated 3 hr later with Compound 1 (2 mg/kg, IP, QD) for 7 days. Lung tissue samples were analyzed for pulmonary cell apoptosis (determined by TUNEL), caspase-3, and staining for F4/80 for cellular infiltration and inflammation. Compound 1 treatment decreased pulmonary cell apoptosis (FIG. 15A and FIG. 15B) and inflammation (FIG. 15C) by two to three fold.

Example 19. Compound 1 treatment mitigates radiation induced lung fibrosis.

[0296] In another study, mice were exposed to 6 Gy and treated with Compound 1 (2 mg/kg, IP, QD) for 4.5 months. Compound 1 treatment mitigated the pulmonary fibrosis, as judged by reduced lung mass (FIG. 16 A), collagen- 1 levels (FIG. 16B), Sirius red staining (FIG. 16C), and TGF bΐ levels (FIG. 16D).

Example 20. Compound 1 treatment decreases radiation induced kidney and liver injury. [0297] In the above chronic radiation experiment, Compound 1 treatment also decreased kidney and liver fibrotic markers including TGFpi (FIG. 17A and FIG. 17B) and aSMA (FIG. 17C) significantly.

Example 21. Evaluation of the radiation dose and time course of c-Met upregulation in lung tissues after thoracic irradiation of mice.

[0298] Rationale. Thoracic radiation exposure can result in acute respiratory distress and death. In radiation induced post-injury, tissue-specific c-Met upregulation remains elevated for several days, affording a unique and expanded opportunity for therapeutic intervention. This Example is designed to evaluate the radiation dose and time course of HGF and c-met upregulation and the relation to pathological sequelae of radiation pneumonitis following thoracic irradiation of mice. This study is expected to guide the selection of radiation dose and time window for therapeutic intervention with Compound 1.

[0299] Experimental strategy and methods. C57L/J mice closely resemble humans with respect to sensitivity to radiation and susceptibility to pneumonitis as well as pulmonary fibrosis. C57L/J mice are exposed to 10, 12.5 and 17.5 Gy of thoracic irradiation (whole thoracic lesion irradiation, or WTLI) using 137 Cs source @ 1 Gy/min. These three doses have been shown to generate three representative pathological courses with increased severity following radiation. The mice are sacrificed after 1, 3, 7, 14, 30 and 60 days. A time course of HGF and its receptor, c-Met upregulation in lungs at both transcriptional level (by RTPCR) and translational level (by Western blots), and the pathological sequelae of pneumonitis after irradiation (mortality, BAL and pulmonary edema, tissue injury, apoptosis, and inflammation) are evaluated. A non-exposed remote organ, i.e., kidneys, are collected and frozen as a negative control.

Example 22. Efficacy of Compound 1 in two studies using the porcine pancreatic elastase (PPE)-induced emphysema rat model.

[0300] PPE was prepared in PBS at 250 U/kg body weight and 250 pL instilled IT. A PBS- instilled group was included. The day after PPE-instillation, the rats were randomly divided into two groups, Vehicle and Compound 1 (2 mg/kg, IP) QD for 6 weeks and arterial blood was drawn from an abdominal artery of each rat with an arterial blood sampler (Quick ABG, Vital Signs Colorado, Inc.), and quickly applied to aNPT7 analyzer (Radiometer/Copenhagen). Compound 1 significantly increased Pa02 (p<0.05) (FIG. 18A) and decreased PaC02 (p<0.05) (FIG. 18B) as compared to the vehicle treated group.

Example 23. Compound 1 Attenuates Pulmonary 30 min Ischemia-24 hr Reperfusion Injury.

[0301] Adult male Sprague-Dawley rats were subjected to 30 min normothermic left lung global ischemia and 24 hr reperfusion. Animals were randomized (n=6/group) to vehicle or Compound 1 (2 mg/kg, iv) at reperfusion. Vehicle or Compound 1 was administered again 20 hr following onset of reperfusion and animals were sacrificed at 24 hr reperfusion. Lungs from sham-operated rats served as normal nonischemic controls. Lungs were sectioned and stained for evidence of pulmonary epithelial cell death (caspase 3 immunoreactivity) and pulmonary microarchitecture (H&E). Slides were read by a pathologist blinded to the treatment groups. Treatment with Compound 1 attenuates pulmonary epithelial cell death observed in the vehicle cohort and preserves pulmonary microarchitecture.

[0302] Result. Compound 1 Mitigates Pulmonary Ischemia-reperfusion Injury: Treatment with Compound 1 immediately following 30 min pulmonary ischemia in the rat attenuated pulmonary apoptosis (caspase 3 staining) at 24 hr reperfusion (FIG. 19A, ^Compound 1 <0.05 vs vehicle). Representative sections from normal (sham), postischemic vehicle and postichemic Compound 1 -treated lungs showing necroinflammatory response in vehicle that is absent in Compound 1 -treated group (FIG. 19B).

Example 24. Compound 1 Attenuates Pulmonary 90 min Ischemia-72 hr Reperfusion Injury.

[0303] In this series, adult male Sprague Dawley rats were subjected to 90 min left pulmonary global normothermic ischemia and 72 hr reperfusion. At the onset of reperfusion, animals (n=5/group) were randomized to vehicle or Compound 1 (2 mg/kg, iv) administration, once daily. Lung function in anesthetized but unassisted (no ventilator) animals was obtained using a vertical U-tube manometer. Briefly, tracheal airflow was connected using a Luer-lock adaptor to one arm of the tube. The U-tube was partially fdled with water allowing for a large volume of respiratory air reserve. The U-tube was placed against a graded scale such that excursion of water in the other arm of the tube could be recorded. End-expiration air volume was calculated by the equations shown below: [0304] Single lung function immediately prior to sacrifice was also evaluated by obtaining blood gas levels from the left pulmonary vein using an Abbot I-STAT blood gas analyzer. Lungs were evaluated histopathologically for pulmonary epithelial regeneration (PCNA immunoreactivity) and microarchitecture. Treatment with Compound 1 was associated with significant improvement in postischemic end-expiration air volume, blood pH, and blood oxygen levels, enhanced pulmonary epithelial regeneration, reduced pulmonary cell death, and preservation of lung microarchitecture.

[0305] Result. Compound 1 Mitigates Pidmonary Ischemia-reperfusion Injury: Compound 1 improves end-expiration air volume following pulmonary 90 min ischemia-reperfusion (FIG. 20A). Prior to sacrifice, Compound 1 -treated cohort exhibited improved blood pH (FIG. 20B), blood oxygen tension (p02) (FIG. 20C) and blood oxygen saturation (s02) (FIG. 20D). Compound 1 treatment was also associated with improved pulmonary epithelial regeneration following reduced pulmonary parenchymal inflammation and neutrophil invasion. 8, p<0.05 vs vehicle (FIG. 20E and FIG. 20G). Compound 1 treatment additionally reduced pulmonary cell death (FIG. 20F).

Example 25. Compound 1 Attenuates Syngeneic Rat Lung Transplantation Injury.

[0306] The effects of Compound 1 were examined on lung function in a model of 3 hr lung cold preservation followed by transplantation in syngeneic Lewis rats. Recipients were sacrificed 3 days following transplantation. This model is unique in that few, if any, studies have examined lung function and histopathology several days following transplantation. In fact, the majority of studies described within the literature subject rodent lungs to cold preservation followed by transplantation with graft evaluation in the recipient conducted in situ (open chest) over only 2-6 hrs. Adult male Lewis recipient rats (n=5/group) were randomized to vehicle or Compound 1 (2 mg/kg, iv) immediately following lung transplantation. Drug administration was once daily. A panel of clinically relevant endpoints was examined to determine drug efficacy. Compound 1 treatment in the setting of lung transplantation was associated with reduced roentgenographic alveolar infiltrate, improved blood gas (obtained from left pulmonary vein) and preservation of alveolar air space integrity.

[0307] Result. Compound 1 Attenuates Lung Transplantation Injury. In rats receiving lung transplant and treated with Compound 1, roentgenographic infiltrate (dotted circular area) on day 3 following transplantation was reduced relative to the vehicle cohort (FIG. 21A). Compound 1 markedly improved blood pH (FIG. 21B) and oxygen levels (FIG. 21C, FIG. 21D, and FIG.

21E), preserved alveolar air space (FIG. 21F, H&E stained lung tissue samples) and attenuated the parenchymal collapse and neutrophil invasion observed in the vehicle cohort.

Example 26. Compound 1 Attenuates Acute Lung Injury in a Sheep Model of Smoke Inhalation + Burn Injury.

[0308] Patients presenting with smoke inhalation + body surface burn injury are especially susceptible to ALI and ARDS. Effects of Compound 1 were therefore evaluated in a large animal model of ALI/ARDS. A study evaluating Compound 1 pharmacokinetics (PK) was first conducted in 3 sheep. Results from this study indicated that the pharmacokinetic profile of Compound 1 in sheep is similar to its profile in rat and man (T50 ~ 2.6 hr).

[0309] Next, adult female sheep were subjected to smoke inhalation (48 puffs of cotton smoke) and 40% body surface bum injury and then randomized to vehicle or Compound 1 (2 mg/kg, iv, QD, n=3). Survivors were sacrificed at 96 hr following injury induction. Vehicle- treated sheep exhibited significant mortality within 96 hr. By contrast, all Compound 1 -treated sheep were alive at 96 hr. Treatment with Compound 1 also attenuated acute lung injury, improving pulmonary gas exchange, preserving fluid balance and preserving pulmonary parenchyma.

[0310] Result. Compound 1 Attenuates Ovine Acute Lung Injury: In sheep subjected to smoke inhalation and 40% body surface burn injury, treatment with Compound 1 attenuated mortality (FIG. 22A), improved pulmonary gas exchange (FIG. 22B) and preserved fluid balance (FIG. 22C). Examination of H&E-stained lung section indicated that Compound 1 treatment (B1 and B2) is associated with decreased alveolar flooding and decreased inflammation compared to vehicle cohort (VI and V2) (FIG. 22D).

Example 27. Compound 1 Decreases Pulmonary Edema.

[0311] Adult male C57BL/6 mice were intratracheally instilled with saline (sham) or bleomycin (1 mg/kg). Bleomycin cohort was immediately randomized to vehicle (n=20) or Compound 1 (2 mg/kg, iv, QD, n=20). Mice were sacrificed at 96 hr and the ratio of lung wet to dry weight was obtained. In some mice, lungs were formalin fixed and subjected to micro CT scan (Numira) and read by a veterinary radiologist. Other lungs were formalin fixed to obtain H&E sections. Treatment with Compound 1 significantly attenuated pulmonary edema (FIG.

23 A). H&E sections showed significant red cell and neutrophil infiltration in the bleomycin+vehicle cohort (vl; v2) accompanied by protein flooding in the alveolar spaces (FIG. 23B). The bleomycin + Compound 1 -treated cohort was markedly free of these abnormalities. Blinded radiographic observations suggested near complete filling of airspace with fluid in bleomycin+vehicle cohort; only mild to moderate filing in bleomycin + Compound 1 cohort and none in the sham (saline) cohort.

Example 28. 24-hr Delayed Compound 1 Treatment Attenuates Pulmonary Edema.

[0312] The study described in Example 27 was repeated except that mice were randomized to vehicle (n=16) or Compound 1 (2 mg/kg, iv QD, n=16) starting 24 hrs later. Mice were sacrificed at 96 hr following intratracheal instillation of bleomycin. 24-hr delayed Compound 1 treatment attenuates pulmonary edema (FIG. 24).

Example 29. Compound 1 Reverses Pulmonary Fibrosis.

[0313] Effects of Compound 1 on pulmonary collagen catabolism were evaluated in a genetic (TSK1/+) mouse model of pulmonary fibrosis. Compound 1 treatment (2 mg/kg, ip, QD, n=5/group) for 12 days reduced pulmonary hydroxyproline content (FIG. 25 A) and pulmonary Sirius red staining (FIG. 25B), sensitive indices of pulmonary interstitial collagen deposition.

Example 30. Compound 1 is Non-Mitogenic for Fibroblasts.

[0314] As a growth factor, an important aspect of SF/HGF activity is to induce endothelial cell proliferation. By contrast, SF/HGF is non-mitogenic for fibroblasts. We examined the effects of Compound 1 on the proliferation of human umbilical vein endothelial cell (HUVECs) and MRC-5 fibroblasts. 3H-thymidine incorporation, which is a measure of the rate of DNA synthesis, was used as a marker of cell proliferation. Compound 1 and SF/HGF stimulated HUVEC proliferation (n=3) (FIG. 26A). By contrast, neither SF/HGF nor Compound 1 stimulated proliferation of MRC-5 fibroblast cells (n=3) (FIG. 26B). Example 31. Compound 1 Stimulates Bronchial Epithelial Cell Proliferation.

[0315] Another important pulmotrophic aspect of SF/HGF activity is to induce epithelial cell proliferation. Compound 1 was tested to determine if, like SF/HGF, it exhibits this activity. Human bronchial epithelial cells (HBECs) were grown to 30-50% confluence in medium containing complete serum for 16-24 hours. Cells were then serum starved for 1-2 hours, followed by the addition of vehicle, Compound 1 or SF/HGF (positive control) for 16-24 hours. [3H]-thymidine was added to the medium and incubation continued for another 4-5 hours. The cells were washed three times with PBS and [3H]-thymidine incorporation determined as a measure of proliferation (increased DNA synthesis). Basal levels of incorporation averaged between 100 to 500 cpm/well and stimulation by SF/HGF averaged 2-fold above baseline. Compound 1 produced a similar increase in incorporation of 3H-thymidine incorporation, indicating that both SF/HGF and Compound 1 activate pulmonary epithelial cell proliferation (FIG. 27).

[0316] Result. Compound 1 Stimulates Bronchial Epithelial Cell Proliferation. Subconfluent HBECs in 48 well plates were treated with vehicle, Compound 1 (36 mM) or SF/HGF (25 ng/mL) for 24 hours in serum free medium. 3H-thymidine incorporation into newly synthesized DNA was assessed and compared to the vehicle controls (FIG. 27).

Example 32. c-Met Dependent Proliferation of Pulmonary Endothelial Cells by Compound

1.

[0317] Proliferation of pulmonary endothelial cells is an important pulmotrophic activity of SF/HGF. To test if Compound 1, like SF/HGF, exhibits proliferative activity and whether the activity is mediated through the SF/HGF receptor c-Met, a cell proliferation assay in bovine pulmonary endothelial cells (bPAEC) was conducted in combination with a siRNA technique. First, c-Met siRNAs and a lipo-transfecting reagents were used to knock down the c-Met mRNA level in bPAEC cells, and real time RT-PCR was performed to measure this effect. The c-Met siRNAs knocked down ~ 90% of c-Met mRNA at a c-Met siRNAs concentration as low as 50 nM. Next, siRNA knockdown conditions established from the first step to decrease >=90% of c- Met mRNA in bPAEC by using 100 nM of the c-Met siRNAs were used, and Compound 1- or SF/HGF-driven 3H-Thymidine incorporation in the bPAEC cells was subsequently measured. The results show that Compound 1, similar to SF/HGF, promotes proliferation of bPAEC cells through the SF/HGF receptor c-Met.

[0318] Result. siRNAs Knockdown c-Met mRNA in Pulmonary Endothelial Cells. -200,000 bovine pulmonary endothelial cells (bPAEC) (CAMBREX, Maryland.) were plated in a 6-well plate and incubated at 5% C02, 37 °C, overnight to -70% confluency. The next day, the cells were washed once with no-serum medium; 0, 10, 50, 100, 200 nM of c-Met siRNAs (Dharmacon) mixed with Lipofectamine (Invitrogene) following the manufacturer’s instruction were added, and the cells were incubated for another 48h; RNA was then prepared using RNeasy (Quiagen). The mRNA level of c-Met was measured by real-time RT-PCR using an ABI 7700 Sequence Detector and specific primers and probes for c-Met and GAPDH (internal control, C), respectively (FIG. 28A).

[0319] Compound 1 and SF/HGF Proliferation of Pulmonary Endothelial Cells is c-Met

Dependent. FIG. 28B and FIG. 28C show the effects of knocking down c-Met mRNA on Compound 1 and SF/HGF-driven pulmonary endothelial cell proliferation.

Example 33. Compound 1 Stimulates Migration of Pulmonary Endothelial Cells.

[0320] Another important pulmotrophic activity of SF/HGF is to stimulate migration of pulmonary endothelial cells. bPAEC cells were used to test if Compound 1, like SF/HGF, can stimulate migration of the cells. The CytoSelect 24-well cell migration assay (Cell Biolabs, Inc.) was used to measure the migration of bPAEC cells according to the kit manual. The results indicate that Compound 1 stimulates bPAEC cells migration, similar to SF/HGF.

[0321] Result. Compound 1 Stimulates Migration in Pulmonary Endothelial Cells. CytoSelect 24-well cell migration assay kit (Cell Biolabs, Inc.) was used. Briefly, -100,000 cells/chamber were seeded in serum-free medium and put into a larger well containing medium with no-serum (C), 10% serum (Sera), HGF, or Compound 1 at multiple concentrations (see FIG. 29), and incubated for 24h. Then the medium was aspirated from the chamber and the cells carefully and completely removed from the inner surface of the membrane with cotton and the membrane washed twice. The migrated cells were stained and quantified by measuring OD at 560nM. The mean values from duplicate wells for each compound are shown in FIG. 29. Example 34. Compound 1 Protects Bronchial Epithelial Cells Against Apoptosis.

[0322] Pulmonary ischemia-reperfusion injury is frequently associated with apoptosis of pulmonary epithelial cells. SF/HGF treatment in this setting is associated with reduced pulmonary epithelial death. We tested whether Compound 1 protects epithelial cells against apoptosis. Human bronchial epithelial cells were grown to 80% confluence and then treated with bleomycin (to induce apoptosis) + vehicle or Compound 1 or SF/HGF for 24 to 48 hrs. Cells were then washed with serum free media and stained with FITC-labeled Annexin-V. Both Compound 1 and SF/HGF protected epithelial cells from apoptosis (FIG. 30)

[0323] Result. Compound 1 Protects Bronchial Epithelial Cells Against Apoptosis. Human bronchial epithelial cells were grown to 80% confluence and then treated with bleomycin 10 (pg/mL) + vehicle, bleomycin plus Compound 1 (15 pg/mL) or bleomycin plus SF/HGF (80 ng/mL) for 24 to 48 hrs. Cells were then washed with serum free media and the cells were stained with FITC-labeled Annexin-V. FITC labeled apoptotic cells were visualized and quantified under a confocal microscope (Olympus) (FIG. 30).

Example 35. Oral Compound 1 Reverses Emphysema.

[0324] Emphysema is a chronic obstructive pulmonary disease often caused by exposure to toxic chemicals, including long-term exposure to smoke. Effects of Compound 1 were evaluated in a standard rodent model of elastase-induced emphysema. Rats were randomized to vehicle or Compound 1 (15 or 45 mg/kg, PO, QD, n=8/group) two weeks following onset of emphysema. The study was completed six weeks following randomization. Treatment with Compound 1 is associated with both functional and histopathological benefit in emphysema, including increased expiration volume (FIG. 31 A), increased arterial oxygen concentration (FIG. 3 IB), and improved mean linear intercept (FIG. 31C). Mean linear intercept is used to evaluate alveolar pathology. See, e.g., Hsia, C.C.W., et al. Am. J. Respir. Crit. Care Med. 2010 Feb 15;181(4):394-418. Additionally, lung tissue samples stained with H&E demonstrated improved are shown in FIG. 3 ID. Example 36. Immediate and Delayed Treatment with Compound 1 in a Dog Model of 120- Minute Renal Ischemia and 7-Day Reperfusion.

[0325] Male beagle dogs were subjected to 120-minute left kidney ischemia and 7-day reperfusion. At the onset of reperfusion, the contralateral (right) kidney was excised. Dogs were randomized to one of the following groups: 1) vehicle (IV, QD, n=4), 2), Compound 1 (10 mg/kg, IV, QD, n=4), both started at the onset of reperfusion, and 3) Compound 1 delayed treatment (10 mg/kg, IV, QD, n=5) started 1 day post ischemia-reperfusion. Dogs were dosed QD until and including Day 4. Blood was collected every 24 hours from an indwelling catheter for a total of 8 days that included a day prior to ischemia-reperfusion. Renal function (serum creatinine and blood urea nitrogen (BUN)) was measured.

[0326] Results are expressed as mean ± standard error of the mean (SEM) for each group. Two-way ANOVA was used to compare the BUN and creatinine time course in the dog ischemia-reperfusion study. Results are considered significant when p<0.05.

[0327] In the dog model of 120-minute renal ischemia followed by 7-day reperfusion, a group of beagle dogs was dosed with vehicle or 10 mg/kg Compound 1 at the onset of reperfusion (immediate treatment) while another group was dosed with 10 mg/kg Compound 1 at 1 day post reperfusion (delayed treatment). Dogs were dosed daily until and including Day 4. Blood was drawn daily for assessment of renal function. Both immediate and delayed Compound 1 treatments significantly reduced serum creatinine (FIG. 32A) and BUN levels (FIG. 32B) (pO.OOOl) compared to the vehicle treatment, indicating an improvement in renal function. [0328] In a study of normothermic renal ischemia and reperfusion in beagle dogs, treatment with Compound 1 reduced serum creatinine and BUN levels, indicating an improvement in renal function. Compound 1 treatment was efficacious, both at the onset of the ischemic injury and when treatment was delayed 1 day post ischemic injury.

Example 37. Compound 1 Protects Bronchial Epithelial Cells from IhC -Induced Apoptosis

[0329] Bronchial epithelial cells (4MBr-5) were grown to sub-confluence and then treated with vehicle or Compound 1 (10 mM) for 24 hours. Cells were then challenged with H2O2 (200 mM) for 1 hour, washed with serum-free media and stained with FITC-labeled Annexin-V and Propidium Iodide. Cells were analyzed by FACS and the percentage of Annexin-V positive cells in vehicle controls was found to be 34.8% of the total number of cells. Compound 1 reduced the percentage of all Annexin-V positive (apoptotic) cells that were fCCfe-exposed to 14.7% (p=0.0015 compared to vehicle control by analyzing the frequency distributions using Fisher’s exact test), indicating that Compound 1 protects bronchial epithelial cells from undergoing oxidative stress-induced apoptosis (FIG. 33A). In the same experiment, the percentage of Annexin-V and Propidium Iodide negative (healthy live) cells as a percentage of the total cell population was increased by Compound 1 from 42.0% to 79.6% (p < 0.0001 by analyzing the frequency distributions using Fisher’s exact test), indicating marked cytoprotection of bronchial epithelial cells by Compound 1 (FIG. 33B).

Example 38. A Multicenter, Randomized, Open-Label Phase 2 Study to Assess Safety and Efficacy of Compound 1 in Patients Hospitalized with COVID-19 Pneumonia.

Objectives:

[0330] Primary: To assess the clinical efficacy of Compound 1 relative to the standard of care arm in reducing the severity and progression of pulmonary dysfunction in adult patients hospitalized with COVID-19 pneumonia.

[0331] Secondary: To assess efficacy to prevent progression to requiring mechanical ventilation and/or ECMO; to assess efficacy to reduce the number of days on mechanical ventilation and/or ECMO; to assess efficacy to reduce the number of ICU days; to assess the safety of Compound 1 in patients hospitalized with COVID-19 pneumonia.

Study Design:

[0332] This is a randomized, open-label, parallel-arm, multicenter study which will assess the efficacy and safety of Compound 1 plus the standard of care (SOC) vs. SOC in patients with COVID-19 pneumonia. Patients hospitalized with confirmed pneumonia with COVID-19 who meet the inclusion criteria but none of the exclusion criteria will be eligible to participate in this study. The eligible subjects will be randomized in 1:1 ratio to the standard of care treatment (SOC) or SOC plus treatment with Compound 1.

[0333] Compound 1 will be administered by 2 daily intravenous (IV) infusions of 2 mg/kg for a total of 10 doses. The first dose will be started within 6 hours of randomization. The subsequent doses will be administered 12 ± 2 hours after the previous dose. [0334] Patients will be followed for safety and efficacy up to Day 30, with Day 1 being the day of the first infusion of study drug. Patients will be assessed daily through Day 14 and then on Days 21 and 30.

[0335] Efficacy endpoints include assessment of the clinical severity on an 8-point Ordinal Scale as recommended by the World Health Organization’s task force on COVID-19 efficacy endpoints:

[0336] There will be approx up to 50 patients per treatment group (i.e., a total of approx up to 100 patients).

Eligibility Criteria:

[0337] Inclusion Criteria:

• Male and nonpregnant female patients 18 years of age or older.

• A positive reverse-transcriptase-polymerase-chain-reaction (RT-PCR) assay for SARS- CoV-2- in a respiratory tract sample.

• Have pneumonia confirmed by chest imaging.

• Ordinal score of 4 or 5.

• Have an oxygen saturation (Sao2) of 94% or less while they were breathing ambient air or a ratio of the partial pressure of oxygen (Pao2) to the fraction of inspired oxygen (Fio2) (Pao2:Fio2) at or below 300 mm Hg.

• Ability to provide informed consent signed by study patient or legally acceptable representative.

• Willingness and ability to comply with study-related procedures/assessments.

[0338] Exclusion Criteria:

• An active malignancy or patients undergoing treatment of a malignancy. • Pregnancy or breast-feeding.

• In the opinion of the investigator, unlikely to survive fo >48 hours from screening.

• Any physical examination findings and/or history of any illness that, in the opinion of the study investigator, might confound the results of the study or pose an additional risk to the patient by their participation in the study.

Product, Dosage, and Mode of Administration:

[0339] Compound 1 for IV administration is a solution with a concentration of 10 mg/mL. The solution also contains 50% (w/v) polyethylene glycol 300 NF, 10% (w/v) polysorbate 80 NF, and phosphate buffered saline.

[0340] Patients will receive 2 mg/kg of Compound 1 via IV infusion over 30 minutes twice daily for 5 days. The first dose will be started within 6 hours of randomization. The subsequent doses will be administered 12 ± 2 hours after the previous dose for a total of 10 doses.

Criteria for Evaluation:

[0341] Efficacy:

[0342] Primary Endpoint:

• Percentage of patients reporting each severity rating on an 8-point ordinal scale at Day 14 of randomization

[0343] Secondary Endpoints:

• All-cause mortality (ACM)

• Time to improvement in oxygenation for at least 48 hours. Increase in Sp02/Fi02 of 50 or greater compared to the nadir Sp02/Fi02

• Mean change in the 8-point ordinal scale from the baseline

• Time to improvement in one category from admission using the 8-point ordinal scale

• Number of days with hypoxemia

• Time to improvement in oxygenation for at least 48 hours by clinical severity. Increase in Sp02/Fi02 of 50 or greater compared to the nadir Sp02/Fi02

• Number of ventilator free days in the first 30 days

• Number of patients requiring initiation of mechanical ventilation and/or ECMO

• Number of patients admitted into an intensive care unit (ICU)

• Number of days of hospitalization among survivors

• Number of patients with secondary bacterial and/or fungal infections [0344] Exploratory Endpoints:

• high-sensitivity C-reaction protein (HS-CRP), absolute lymphocyte count, serum ferritin, semm interleukin-6 (IL-6), semm myoglobin, D-dimer, creatine phosphokinase (CPK), CPK-MB, troponin, and LDH drawn within 12 hours of randomization and at days 3-5 and 14

[0345] Safety:

• Collection of adverse events (AEs) emerging during treatment, Grade 3 or greater, serious adverse events (SAEs), and AEs leading to discontinuation of study treatment

• Laboratory parameters (hematology, chemistry, hepatic, coagulation, urinalysis)

• Vital signs Statistical Methods:

[0346] Efficacy analysis:

[0347] Continuous variables will be summarized with descriptive statistics (the number of non-missing values [n], mean, median, standard deviation [SD], minimum, and maximum). All categorical variables will be summarized with frequency counts and percentages, as applicable. Time to event variables will be analyzed using Kaplan-Meier (K-M) survival estimates. The K- M survival curves will be compared between treatment groups using Log-rank test. K-M estimates including 25th, 50th, and 75th percentiles, 95% confidence intervals (CIs), and number and percent censored will be presented. Student’s t test will be carried out for normally distributed variables. Wilcoxon rank-sum test will be used for non-normally distributed variables, Difference in proportions between treatment groups will be analyzed using Fisher’s exact test.

[0348] Safety analysis:

[0349] All patients randomized and receiving any part of at least one infusion of study treatment (Compound 1 or Placebo) will be evaluated for safety. The safety analyses will include evaluation of the incidence of treatment-emergent AEs, Grade 3 or greater AEs, serious adverse events (SAEs), and AEs leading to discontinuation of study treatment. Laboratory and vital signs assessments will be evaluated over time on study using descriptive statistics. Shift analyses of relevant clinical laboratory parameters will be produced showing shifts across low, normal, and high categories. Example 39. A Multicenter, Prospective, Randomized, Double-Blind, Placebo-Controlled Phase 2 Study to Assess Safety and Efficacy of Compound 1 in Patients Hospitalized with Confirmed COVID-19 Pneumonia.

Primary Objective:

[0350] To assess clinical efficacy of Compound 1 relative to placebo plus standard of care in reducing the severity and progression of pulmonary and renal dysfunction and mortality in adult patients hospitalized with COVID-19 pneumonia.

Secondary Objectives:

[0351] To assess safety of Compound 1 in patients hospitalized with COVID-19 pneumonia. [0352] To assess efficacy to prevent progression to requiring mechanical ventilation and/or extracorporeal membrane oxygenation (ECMO) and/or renal replacement therapy.

[0353] To assess efficacy to reduce number of days on mechanical ventilation and/or ECMO.

[0354] To assess efficacy of Compound 1 to reduce ICU length of stay in patients with

COVID-19 pneumonia.

Study Design:

[0355] This is a randomized, prospective, double-blind, placebo-controlled, parallel-arm, multicenter study which will assess the efficacy and safety of Compound 1 plus the standard of care (SOC) vs. placebo plus SOC in patients with COVID-19 pneumonia. Patients hospitalized with confirmed pneumonia with COVID-19 who meet the inclusion criteria but none of the exclusion criteria are eligible to participate in this study. The eligible patients will be randomized in 1 : 1 ratio to Compound 1 + SOC or placebo + SOC.

[0356] Compound 1 is administered by once daily intravenous (IV) infusions of 2 mg/kg for a total of 4 doses. The first dose is started within 6 hours of randomization. Subsequent doses are administered 24 ± 4 hours after the previous dose.

[0357] Patients are followed for safety and efficacy up to Day 28 ± 2 days, with Day 1 being the day of randomization. Patients are assessed daily until their discharge from the hospital or Day 28 after randomization.

[0358] Safety is monitored by an independent Safety Review Committee on an ongoing basis. The following 8-point Ordinal Scale as recommended by the World Health Organization (WHO’s) task force on COVID-19 efficacy endpoints are used for inclusion/exclusion and some of the secondary efficacy endpoints:

[0359] There will be approx up to 50 patients per treatment group (i.e., a total of approx up to 100 patients).

Elmbility Criteria:

[0360] Inclusion Criteria:

• Patient is a male or nonpregnant female patients 18 years of age or older.

• Patient has a positive reverse-transcriptase-polymerase-chain-reaction (RT-PCR) assay for SARS-CoV-2 in a respiratory tract sample during the current hospital admission.

• Patient has pneumonia confirmed by chest imaging.

• Patient has score of 5 on WHO’s disease severity scale assessment 8-point Ordinal Scale at time of randomization.

• Patient has ability to provide informed consent signed by study patient or legally acceptable representative.

• Patient has willingness and ability to comply with study-related procedures/assessments.

[0361] Exclusion Criteria:

• Has an active malignancy or history of solid or hematological malignancies within 5 years prior to enrollment in the study. Patients who had basal or squamous cell carcinoma-in-situ of the skin that was diagnosed > 2 years prior to the study enrollment and not currently being treated are eligible for study enrollment.

• Patient is pregnant or breast-feeding.

• Patient, in the opinion of the investigator, is unlikely to survive for > 48 hours from the time of screening. • Patient has any physical examination findings and/or history of any illness that, in the opinion of the study investigator, might confound the results of the study or pose an additional risk to the patient by their participation in the study.

• Patient with alanine aminotransferase (ALT) or aspartate transaminase (AST) > 3x upper limit of normal (ULN) and/or total bilirubin > 2x ULN at baseline.

• Requires treatment with the cytochrome P450 1A2 (CYP1A2) inhibitors, ciprofloxacin and/or fluvoxamine.

• Patients participating in any other clinical trial with an investigational drug product or procedure.

• Recipients of solid organ and/or hematopoietic cell transplantation.

• Patient is known to have End Stage Renal Disease (ESRD) and was being treated with maintenance hemodialysis or peritoneal dialysis prior to the current hospitalization. (Patients who initiated renal replacement therapy due to Acute Kidney Injury during their current hospitalization are eligible for the study.)

Drug Product, Dosage, and Mode of Administration:

[0362] Compound 1 for intravenous (IV) administration is a sterile solution with a concentration of 10 mg/mL. The solution also contains 50% weight per volume (w/v) PEG 300 national formulary, 10% w/v polysorbate 80 NF, and phosphate buffered saline.

[0363] Patients received 2 mg/kg Compound 1 or placebo (equivalent volume of normal saline), via IV infusion over 30 minutes, once-daily for 4 days. The first dose is started within 6 hours of randomization. The subsequent doses are administered 24 ± 4 hours after the previous dose for a total of 4 doses. A regular schedule for administering subsequent doses every 24 hours is established.

[0364] If a patient misses a scheduled dose of Compound 1, the missed dose may be administered as long as there are at least 12 hours between the end of the infusion of the “make up” dose and the next scheduled dose of Compound 1.

[0365] The 10 mg/mL stock solution is diluted with normal saline before being administered. Volume is administered according to the patient’s weight. An equivalent volume of normal saline is used as placebo.

Duration of Treatment:

[0366] Once-daily for 4 days. Reference Therapy Dosage and Mode of Administration:

[0367] Patients receive SOC as adopted by the participating institution plus placebo. Normal saline is used as placebo. Patients receive a volume of normal saline equivalent to that containing active drug on a mL/kg basis.

Criteria for Evaluation:

[0368] Primary Endpoint:

• Proportion of patients alive, without need for mechanical ventilation and free of the need for renal replacement therapy (RRT) (on an ongoing basis) at Day 28.

[0369] Secondary Endpoints:

• All-cause mortality.

• Proportion of patients not requiring mechanical ventilation at Day 28.

• Proportion of patients not requiring RRT on an on-going basis at Day 28.

• Number of ventilator free days in the first 28 days.

• Proportion of patients requiring initiation of mechanical ventilation and/or ECMO through Day 28.

• Proportion of patients requiring initiation of renal replacement therapy through Day 28.

• Number of days to renal recovery (defined as freedom from further RRT on an ongoing basis) in subjects who were on RRT at the time of randomization.

• Number of ICU days from randomization to Day 28.

• Change in WHO 8-point ordinal scale from randomization to Day 28.

• Number of days to hospital discharge from randomization.

[0370] Exploratory Endpoints:

• Mean change from baseline in biomarkers: high-sensitivity C-reactive protein (HS-CRP), D-dimer, absolute lymphocyte count, ferritin, myoglobin, troponin, lactate dehydrogenase (LDH)

[0371] Safety:

• Collection of Adverse Events (AEs) emerging during treatment, Grade 3 or greater, serious adverse events (SAEs), and AEs leading to discontinuation of study treatment.

• Laboratory parameters (hematology, chemistry, troponin, hepatic, coagulation, urinalysis)

• Vital signs • Electrocardiogram Statistical Methods:

[0372] Efficacy Analysis:

[0373] Sample size: Approximately 100 patients are enrolled in this study.

[0374] Methods: Continuous variables are summarized with descriptive statistics (number of non-missing values [n], mean, median, standard deviation [SD], minimum, and maximum). All categorical variables are summarized with frequency counts and percentages, as applicable.

Time to event variables are analyzed using Kaplan-Meier (K-M) survival estimates. The K-M survival curves will be compared between treatment groups using Log-rank test K-M estimates including 25 th , 50 th , and 75 th percentiles, 95% confidence intervals (CIs), and number and percent censored are presented. A Mixed Model Repeated Measures (MMRM) analysis or Analysis of Covariance (ANCOVA) are carried out for continuous variables, depending on the number of assessments post-baseline. For binary endpoints, difference in proportions between treatment groups are analyzed using a Mantel-Haenszel Test, stratifying on baseline severity. Analyses are carried out on the Full Analysis Set.

[0375] Interim Analysis: Formal interim analysis is not planned, however safety data are reviewed on an ongoing basis by an independent Safety Review Committee.

[0376] Safety Analysis: All patients randomized and who received SOC plus placebo or received SOC and any part of at least one infusion of study treatment (Compound 1) are evaluated for safety. The safety analyses include evaluation of the incidence of treatment- emergent AEs, Grade 3 or greater AEs, SAEs, and AEs leading to discontinuation of study treatment. Laboratory and vital signs assessments are evaluated over time on study using descriptive statistics. Shift analyses of relevant clinical laboratory parameters are produced showing shifts across low, normal, and high categories.

Example 40. Effect of Compound 1 on EhC -Induced Apoptosis in Bronchial Epithelial Cells

[0377] Bronchial epithelial cells (4MBr-5) were grown to sub-confluence and treated with vehicle or Compound 1 (10 mM) or HGF (50 ng/mL) for 24 hours. Cells were then challenged with H2O2 (200 mM) for 1 hour, washed with serum-free media and stained with FITC-labeled Annexin-V and propidium iodide (PI). Cells were analyzed for Annexin-V and PI stained fractions with a fluorescence-activated cell sorter and cells were divided in four staining categories based on Annexin and PI staining intensity. Annexin-V staining precedes the loss of membrane integrity which accompanies the latest stages of cell death resulting from either apoptotic or necrotic processes. Therefore, staining with Annexin-V is used in conjunction with a vital dye such as propidium iodide to identify early apoptotic cells (PI negative, Annexin-V positive). Viable cells with intact membranes exclude PI, whereas the membranes of dead and damaged cells are permeable to PI. Cells that are considered viable are Annexin-V and PI negative; cells that are in early apoptosis are Annexin-V positive and PI negative; and cells that are in late apoptosis or already dead are both Annexin-V and PI positive. As shown in FIG. 34, both treatment with Compound 1 and with HGF changed the overall distribution of cells amongst different staining categories.

Example 41. Effect of Compound 1 vs HGF on Mouse NIH/3T3 Fibroblast Proliferation [0378] NIH/3T3 cells, a mouse fibroblast cell line, was maintained and cultured using DMEM with 10% FBS added. Cells were grown in culture flasks (T75 and T150) and incubated at 37 °C in an incubator conditioned with 5% CC . To harvest cells, growth medium was removed and the cell monolayer was rinsed with PBS and treated with 0.25% trypsin until the cell monolayer was detached, which was monitored under a microscope. Cells were dispersed by gentle pipetting and split into new flasks.

[0379] For cell proliferation assays, NIH/3T3 cells were seeded at 1000 to 1500 cells per well in 48-well plates in complete medium containing 10% serum, and grown to 30 to 40% confluency for 24 hours. Cells were then serum-starved for 1 to 2 hours in RPMI medium containing 1% BSA, followed by treatment with HGF (25 ng/mL), Compound 1 (17 mM), or DMSO for 16 to 24 hours. [ 3 H]-Thymidine was added at 10 pCi/mL to the medium and incubation continued for another 4 to 5 hours. Cells were washed 3 times with PBS and lysed with 0.5 mL of alkaline lysis buffer containing 0.5 N NaOH and 1 % SDS. After incubation on a shaker for 30 minutes to 1 hour at room temperature, the cell lysates were placed in scintillation vials to which 3.5 mL of scintillation fluid was added. The radioactivity was measured using a beta counter. The [ 3 H] signal, representing thymidine incorporation into to newly synthesized DNA (a measure of cell proliferation), was counted using a beta counter.

[0380] The cell proliferative effects of Compound 1 versus HGF were compared in mouse NIH/3T3 fibroblasts (FIG. 35). NIH/3T3 fibroblasts were incubated with Compound 1 (5 mM) or HGF (25 ng/mL), and [ 3 H]-thymidine incorporation was used as a measure of cell proliferation. In NIH/3T3 fibroblasts, which do not express the c-Met receptor, neither HGF nor Compound 1 stimulated cell proliferation. As a positive control, serum, which contains many growth stimulating factors, was evaluated in parallel and was found to stimulate NIH/3T3 cell proliferation.

Example 42. Compound 1 Protects Against Radiation-Induced Injury [0381] Eight- to ten-week old male C57BL/6 mice were anesthetized with a mixture of ketamine (100 mg/kg) + xylazine (10 mg/kg)) via intraperitoneal injection. Mice were aligned on a prone position and exposed to total body irradiation (TBI) at 1 Gy /min for 7 min (7 Gy) using an MDS Nordion Gamma Cell-40 Exactor/Research Irradiator (Cs-137 source, Cold Spring Harbor Laboratories, NY) with a beam of collimator. Sham-irradiated mice were treated in the same way, except that the radiation source was not turned on. After irradiation, when the mice were fully awake and mobile, they were transported to the animal facility (15-minute drive).

[0382] A total of 24 mice were irradiated and three hours later, mice were randomized to vehicle (n=12; IP; RAD Vehicle) and Compound 1 (n=12; 2 mg/kg; IP; RAD Cmpd 1) once daily (QD) for 6 days. Mortality was followed for 6 days and surviving animals were then sacrificed. Two age and gender matched sham-irradiated animals served as controls (Control). Animals were sacrificed in the afternoon, after receiving a final dose in the morning. Bronchoalveolar lavage fluid (BALF) was collected immediately prior to sacrifice to evaluate pulmonary cell infiltration by spectrophotometry. [0383] BALF was collected at the time of sacrifice. After anesthetizing the animals, 1 mL of 0.9% saline was injected with a 2 mL syringe through the trachea and gently flushed 2 to 3 times through the lungs to collect the lavage fluid into the syringe. An aliquot (200 pL) of this lavage fluid was transferred to a 96-well plate and the optical density at 450 nm was measured. The % transmission is an indicator of BAL turbidity, which is a readout of the number of all infiltrating white cells. Results are expressed as % transmittance readings and as D transmittance (the difference in % transmittance between experimental samples and background), which is directly proportional to BAL turbidity.

[0384] At the time of sacrifice, 6 days after irradiation, the number of animals in radiation- exposed vehicle-treated animals was 2. The percentage of surviving animals in this group was 16.6% (2 out of 12). At the time of sacrifice, the number of surviving animals in radiation-exposed Compound 1 -treated animals was 6. The percentage of surviving animals in this group was 50% (6 out of 12). Lischer’s exact test analysis indicated that the difference between Compound 1 and vehicle treatment groups was not statistically significant. However, Compound 1 did appear to increase survival after radiation exposure (FIG. 36A).

[0385] Bronchoalveolar lavage fluid (BALF) was collected at the time of sacrifice and BALF turbidity was determined by measuring % transmission at 450 nm. The difference in transmittance between experimental samples and background was calculated (A transmittance). The A transmittance is proportional to BALF turbidity and an indicator of infiltration. As shown below, BALF turbidity was markedly increased in radiation-exposed vehicle-treated animals, indicating significant pulmonary infiltration as a result of radiation exposure. In radiation-exposed animals that were treated with Compound 1, turbidity was significantly reduced compared to the vehicle-treated cohort (FIG. 36B). This indicates that Compound 1 significantly reduced pulmonary infiltration resulting from radiation exposure.

Example 43. Compound 1 Protects Against TGFpi-Induced Acute Lung Injury (3-day study)

[0386] Six-to-eight week old TGFpl -transgenic male and female mice were administered doxy cy dine (dox) in the drinking water (0.5 mg/mL dox and 20 g/L sucrose) to induce high level lung-specific expression of TGFpi. One day after initiation of dox administration, animals were randomized to the vehicle (n=8; 4 male and 4 female) and Compound 1 group (n=8; 4 male and 4 female; 2 mg/kg), and both groups received once daily intraperitoneal injections of Compound 1 or vehicle and continued on dox feeding (0.5 mg/mL dox and 20 g/L sucrose in drinking water) before sacrifice in the afternoon of day 3 after initiation of doxycycline administration, with a last compound or vehicle administration in the morning of the day of sacrifice. Four non-induced control animals (2 male and 2 female) were processed in parallel.

[0387] To evaluate the effect of Compound 1 treatment on lung histopathology, lung tissue sections were immunohistologically stained with an antibody against proliferating cell nuclear antigen (PCNA), a marker of cell proliferation, and an antibody against Caspase-3, a marker of cell apoptosis.

[0388] Compound 1 -treated doxy cy cline-exposed animals showed a significant increase in PCNA staining compared to vehicle-treated animals (FIG. 37A). This indicates that Compound 1 stimulates lung cell proliferation after TGF 1 -induced pulmonary damage. It should be noted that no difference in PCNA staining between lungs from Control animals and TGF i Vehicle animals was expected, and that lung regeneration was only enhanced in the TGF i Compound 1 group. [0389] Compound 1 -treated animals showed a significant decrease in Caspase-3 staining compared to vehicle-treated animals (FIG. 37B). This indicates that Compound 1 protects lung cells from TGF l -induced apoptosis.

Example 44. Compound 1 Protects Against TGFpi-Induced Acute Lung Injury (10-day study)

[0390] Eight-to-10 week old TGFpi -transgenic male and female mice (approximately 25 g body weight) were administered doxycycline (dox) in the drinking water (0.5 mg/mL dox and 20 g/L sucrose) to induce high level lung-specific expression of TGFpi. One day after initiation of dox administration, animals were randomized to the vehicle group (n=15; 10 male and 5 female) and Compound 1 group (n=15; 10 male and 5 female; 2 mg/kg) and both groups received once daily intraperitoneal injections and continued dox feeding (0.5 mg/mL dox and 20 g/L sucrose in drinking water) for 10 days.

[0391] As illustrated below, vehicle-treated doxycycline-exposed animals showed marked mortality after initiation of doxycycline administration. Mortality in this group after 10 days was 73.3% (11 out of 15 animals died). In Compound 1 -treated doxycycline-exposed animals, mortality after 10 days was 13.3% (2 out of 15 animals died).

[0392] The survival data for the 10-day survival study were analyzed using Fischer’s exact test to evaluate the proportions of surviving animals at day 10 (FIG. 38). Compound 1 treatment conferred a significant survival benefit (p= 0.0025) compared to vehicle-treated animals. A Kaplan-Meier analysis of the survival data showed that vehicle-treated animals had a median survival time of 6.0 days, while the median survival time for Compound 1-treated animals could not be calculated due to the limited mortality in this group (>10 days). A Log-rank (Mantel-Cox) test comparing the two treatment cohorts showed a significant survival benefit as a result of Compound 1 treatment compared to vehicle treatment (p=0.0008).

Example 45. Effect of Compound 1 on Growth of c-Met-expressing Human Tumor Cells in Immunocompromised Mice

[0393] The effect of Compound 1 on growth of c-Met-expressing human tumor cells was evaluated in immunocompromised mice using xenograft models of pancreatic, colon, and brain cancer. As detailed below, Compound 1 treatment did not adversely affect survival in an in vivo model of c-Met-expressing glioma in immunocompromised mice, nor did it promote tumor growth in in vivo models of c-Met-expressing colon cancer and pancreatic cancer in immunocompromised mice. Thus, exposure to Compound 1 was not associated with increases in tumor size or tumor weight, nor was it associated with a decrease in survival compared to vehicle-treated animals.

Glioma Orthograft Model

[0394] Human glioma cells U87-MG (obtained from American Type Culture Collection [ATCC]), which express c-Met (La, B., et al. Clin. Cancer Res. 2005 Jun 15;ll(12):4479-86), were used in the glioma orthotopic model. U87-MG cells were maintained and cultured using protocols provided by ATCC and summarized as follows. Cells were grown in culture flasks (T75 and T150) and incubated at 37 °C in an incubator conditioned with 5% CCh. The culture medium used was Eagle's minimum essential medium (EMEM) modified with Eagle's balanced salt solution and 2 mM L-glutamine, 1 mM sodium pyruvate, 0.1 mM nonessential amino acids, and 1.5 g/L sodium bicarbonate, supplemented with 10% fetal bovine serum (FBS). To harvest cells, growth medium was removed and cell monolayer was rinsed with phosphate buffered saline (PBS) and treated with trypsin-ethylenediaminetetraacetic acid (EDTA) solution (0.25% w/v trypsin, 0.03% w/v EDTA) until the cell monolayer was detached as monitored under a microscope. Cells were separated from each other by gentle pipetting. Cells in culture were split twice a week at a ratio of 1:3 to 1:5. Harvested cells were suspended in EMEM medium for animal injection. Cells (2 x 105 cells in 10 pL) were injected intracranially into BALB/c nude mice. Tumors were allowed to grow for 7 days, then animals were treated daily until day 28 with 2 mg/kg Compound 1 or vehicle administered via 50 pL IP injection. Survival of the mice was monitored daily until all mice died.

[0395] Compound 1 was evaluated in two independent studies in the orthotopic glioma model described above, and the results are summarized in FIG. 39A and FIG. 39B. FIG. 39A shows percent survival of Compound 1 vs vehicle treatment in a first study of an orthotopic glioma model. FIG. 39B shows percent survival of Compound 1 vs vehicle treatment in a second study of an orthotopic glioma model. Compound 1 treatment did not decrease survival rate, as compared with a vehicle control group for each of the two studies (log-rank test, A and B, p=0.1930 and p=0.8215, respectively). Colon Tumor Xenograft Model

[0396] Human colon cancer cells HT29 (obtained from ATCC), which express c-Met (Wielenga, V. J., et al. Am. J. Pathol. 2000 Nov; 157(5): 1563-73), were used in the colon tumor xenograft model. HT-29 cells were maintained and cultured using protocols provided by ATCC and summarized as follows. Cells were grown in culture flasks (T75 and T150) and incubated at 37 °C in an incubator conditioned with 5% CCh. The culture medium used was McCoy's 5A medium modified to contain 1.5 mM L-glutamine and 2200 mg/L sodium bicarbonate, supplemented with 10% FBS. To harvest cells, growth medium was removed and the cell monolayer was rinsed with PBS and treated with trypsin-EDTA solution (0.25% w/v trypsin, 0.03% w/v EDTA) until the cell monolayer was detached as monitored under a microscope.

Cells were separated from each other by gentle pipetting. Cells in culture were split twice a week at a ratio of approximately 1:5. Harvested cells were suspended in growth medium containing no serum for animal injection. Cells (1 x 106 cells in 100 pL) were injected SC in the right hind flank of BALB/c nude mice. Tumors were allowed to grow for 7 days, then animals were treated daily until Day 27 with 2 mg/kg Compound 1 or vehicle administered via 50 pL IP injection. Tumor size was measured with dial calipers on Days 7, 12, 15, 19, 22, 27 and survival was recorded. Animals were sacrificed on Day 27 and tumor size and weight was recorded.

[0397] Compound 1 was evaluated in a human colon tumor xenograft model described above, and the results are summarized in FIG. 40A and FIG. 40B. FIG. 40A shows colon tumor size of Compound 1 vs vehicle treatment in a human colon tumor xenograft model. FIG. 40B shows colon tumor weight of Compound 1 vs vehicle treatment in a human colon tumor xenograft model (ns = not significant). Compound 1 treatment did not increase the size (FIG. 40A) or weight (FIG. 40B) of the colon tumor xenograft (p=0.7065 [ANOVA] and p=0.4677 [T- test], respectively).

Pancreatic Tumor Xenograft Model

[0398] Human pancreatic ductal carcinoma cells SUIT-2 (obtained from the Japanese Collection of Research Bioresources [JCRB] Cell Bank), which express c-Met (Maehara, N., et al. ChBr J. Cancer 2001 Mar 23;84(6):864-73), were used in the pancreatic tumor xenograft model. SUIT-2 cells were maintained and cultured using protocols provided by the JCRB Cell Bank and summarized as follows. Cells were grown in culture flasks (T75 and T150) and incubated at 37 °C in an incubator conditioned with 5% CO2. The culture medium used was Roswell Park Memorial Institute (RPMI) with 10% FBS. To harvest cells, growth medium was removed and the cell monolayer was rinsed with PBS and treated with 0.25% trypsin in PBS until the cell monolayer was detached as monitored under a microscope. Cells were separated from each other by gentle pipetting. Cells in culture were split weekly. Harvested cells were suspended in growth medium containing no serum for animal injection. Cells (5 x 106 cells in 100 pL) were injected SC in the right hind flank of male BALB/c nude mice. Tumors were allowed to grow for 12 days, then animals were treated daily, 5 days a week, for 3 weeks with 2 mg/kg Compound 1 or vehicle administered via 50 pL IP injection. Tumor volume was measured twice weekly using dial calipers, and tumor weight was measured at sacrifice. Animals were sacrificed after the last dose administration.

[0399] Compound 1 was evaluated in a human pancreatic tumor xenograft model, and the results are summarized in FIG. 41A and FIG. 41B. FIG. 41A shows pancreatic tumor volume of Compound 1 vs vehicle treatment in a human pancreatic tumor xenograft model. FIG. 41B shows pancreatic tumor weight of Compound 1 vs vehicle treatment in a human pancreatic tumor xenograft model. Compound 1 treatment did not increase the size (FIG. 41 A) or weight (FIG. 41B) of the pancreatic tumor. In fact, Compound 1 reduced the size of the tumor (p=0.0078, ANOVA), although it did not significantly reduce the weight of the tumor (p=0.3584, T-test).

Example 46. A Multicenter, Prospective, Randomized, Double-Blind, Placebo-Controlled Phase 2 Study to Assess Safety and Efficacy of Compound 1 in Patients Hospitalized with Confirmed COVID-19 Pneumonia.

Primary Objective:

[0400] To assess clinical efficacy of Compound 1 plus standard of care relative to placebo plus standard of care in reducing the severity and progression of pulmonary and renal dysfunction and mortality in adult patients hospitalized with COVID-19 pneumonia.

Secondary Objectives:

[0401] To assess safety of Compound 1 in patients hospitalized with COVID-19 pneumonia. [0402] To assess efficacy to prevent progression to requiring mechanical ventilation and/or extracorporeal membrane oxygenation (ECMO) and/or renal replacement therapy.

[0403] To assess efficacy to reduce number of days on mechanical ventilation and/or ECMO. [0404] To assess efficacy of Compound 1 to reduce ICU length of stay in patients with COVE ) - 19 pneumonia.

Study Desisn:

[0405] This is a randomized, prospective, double-blind, placebo-controlled, parallel-arm, multicenter study which assesses the efficacy and safety of Compound 1 plus the standard of care (SOC) vs. placebo plus SOC in patients with COVID-19 pneumonia. Patients hospitalized with confirmed pneumonia with COVID-19 who meet the inclusion criteria but none of the exclusion criteria were eligible to participate in this study. The eligible patients are randomized in 1 : 1 ratio to Compound 1 + SOC or placebo + SOC. Subjects were stratified at randomization by disease severity (moderate [Score 4] vs. severe [Score 5]) based on the WHO’s disease severity scale assessment 8-point Ordinal Scale.

[0406] Compound 1 was administered by once daily intravenous (IV) infusions of 2 mg/kg for a total of 4 doses. The first dose was started within 6 hours of randomization. Subsequent doses were administered 24 ± 4 hours after the previous dose.

[0407] Patients were followed for safety and efficacy up to Day 28 ± 2 days, with Day 1 being the day of randomization. Patients were assessed daily until their discharge from the hospital or Day 28 after randomization.

[0408] Safety was monitored by an independent Safety Review Committee on an ongoing basis. The following 8-point Ordinal Scale as recommended by the World Health Organization (WHO’s) task force on COVID-19 efficacy endpoints was used for inclusion/exclusion and some of the secondary efficacy endpoints:

[0409] There were approx up to 50 patients per treatment group (i.e., a total of approx up to 100 patients). too Eligibility Criteria:

[0410] To be eligible for the study, participants were required to meet all of the following inclusion criteria:

• Patient is a male or nonpregnant female patients 18 years of age or older.

• Patient has a positive reverse-transcriptase-polymerase-chain-reaction (RT-PCR) assay for SARS-CoV-2 in a respiratory tract sample during the current hospital admission.

• Patient has pneumonia confirmed by chest imaging.

• Patient has moderate to severe disease based on WHO’s disease severity scale assessment 8-point Ordinal Scale at time of randomization defined as: o Score 4, only those with F1O2 >40% (FiCh >40% defined as nasal cannula > 5 L/min, venturi mask > 10 L/min, conventional mask > 8 L/min, or mask with oxygen reservoir). o Score 5 (Non-invasive ventilation or high-flow oxygen).

• Patient has ability to provide informed consent signed by study patient or legally acceptable representative.

• Patient has willingness and ability to comply with study-related procedures/assessments. [0411] A participant who met any of the following exclusion criteria were excluded from the study:

• Has an active malignancy or history of solid or hematological malignancies within 5 years prior to enrollment in the study. Patients who had basal or squamous cell carcinoma-in-situ of the skin that was diagnosed > 2 years prior to the study enrollment and not currently being treated are eligible for study enrollment.

• Patient is pregnant or breast-feeding.

• Patient, in the opinion of the investigator, is unlikely to survive for > 48 hours from the time of screening.

• Patient has any physical examination findings and/or history of any illness that, in the opinion of the study investigator, might confound the results of the study or pose an additional risk to the patient by their participation in the study.

• Patient with alanine aminotransferase (ALT) or aspartate transaminase (AST) > 3x upper limit of normal (ULN) and/or total bilirubin > 2x ULN at baseline. • Requires treatment with the cytochrome P450 1A2 (CYP1A2) inhibitors, ciprofloxacin and/or fluvoxamine.

• Patients participating in any other clinical trial with an investigational drug product or procedure.

• Recipients of solid organ and/or hematopoietic cell transplantation.

• Patient is known to have End Stage Renal Disease (ESRD) and was being treated with maintenance hemodialysis or peritoneal dialysis prior to the current hospitalization. (Patients who initiated renal replacement therapy due to Acute Kidney Injury during their current hospitalization are eligible for the study.)

Drue Product, Dosage, and Mode of Administration:

[0412] Compound 1 for intravenous (IV) administration is a sterile solution with a concentration of 10 mg/mL. The solution also contains 50% weight per volume (w/v) PEG 300 national formulary, 10% w/v polysorbate 80 NF, and phosphate buffered saline.

[0413] Patients received 2 mg/kg Compound 1 or placebo (equivalent volume of normal saline), via IV infusion over 30 minutes, once-daily for 4 days. The first dose was started within 6 hours of randomization. The subsequent doses were administered 24 ± 4 hours after the previous dose for a total of 4 doses. A regular schedule for administering subsequent doses every 24 hours was established.

[0414] If a patient misses a scheduled dose of Compound 1, the missed dose may be administered as long as there are at least 12 hours between the end of the infusion of the “make up” dose and the next scheduled dose of Compound 1.

[0415] The 10 mg/mL stock solution was diluted with normal saline to a concentration of 6 mg/mL before being administered. Volume was administered according to the patient’s weight. An equivalent volume of normal saline was used as placebo.

Duration of Treatment:

[0416] Once-daily for 4 days.

Reference Therapy, Dosaee, and Mode of Administration:

[0417] Patients received SOC as adopted by the participating institution plus placebo.

Normal saline was used as placebo. Patients received a volume of normal saline equivalent to that containing active drug on a mL/kg basis.

Criteria for Evaluation: [0418] Primary Endpoint:

• Proportion of patients alive, without need for mechanical ventilation and free of the need for renal replacement therapy (RRT) (on an ongoing basis) at Day 28.

[0419] Secondary Endpoints:

• All-cause mortality.

• Proportion of patients not requiring mechanical ventilation at Day 28.

• Proportion of patients not requiring RRT on an on-going basis at Day 28.

• Number of ventilator free days in the first 28 days.

• Proportion of patients requiring initiation of mechanical ventilation and/or ECMO through Day 28.

• Proportion of patients requiring initiation of renal replacement therapy through Day 28.

• Number of days to renal recovery (defined as freedom from further RRT on an ongoing basis) in subjects who were on RRT at the time of randomization.

• Number of ICU days from randomization to Day 28.

• Change in WHO 8-point ordinal scale from randomization to Day 28.

• Number of days to hospital discharge from randomization.

[0420] Exploratory Endpoints:

• Mean change from baseline in biomarkers: high-sensitivity C-reactive protein (HS-CRP), D-dimer, absolute lymphocyte count, ferritin, myoglobin, troponin, lactate dehydrogenase (LDH)

[0421] Safety:

• Collection of Adverse Events (AEs) emerging during treatment, Grade 3 or greater, serious adverse events (SAEs), and AEs leading to discontinuation of study treatment.

• Laboratory parameters (hematology, chemistry, troponin, hepatic, coagulation, urinalysis)

• Vital signs

• Electrocardiogram Statistical Methods:

[0422] Efficacy Analysis:

[0423] Sample size: Approximately 100 patients were enrolled in this study. [0424] Methods: Continuous variables were summarized with descriptive statistics (number of non-missing values [n], mean, median, standard deviation [SD], minimum, and maximum).

All categorical variables were summarized with frequency counts and percentages, as applicable. Time to event variables were analyzed using Kaplan-Meier (K-M) survival estimates. The K-M survival curves were compared between treatment groups using Log-rank test. K-M estimates including 25 th , 50 th , and 75 th percentiles, 95% confidence intervals (CIs), and number and percent censored were presented. A Mixed Model Repeated Measures (MMRM) analysis or Analysis of Covariance (ANCOVA) was carried out for continuous variables, depending on the number of assessments post-baseline. For binary endpoints, difference in proportions between treatment groups were analyzed using a Mantel-Haenszel Test, stratifying on baseline severity. Analyses were carried out on the Full Analysis Set.

[0425] Interim Analysis: Formal interim analysis was not planned, however safety data were reviewed on an ongoing basis by an independent Safety Review Committee.

[0426] Safety Analysis: All patients randomized and who received SOC plus placebo or received SOC and any part of at least one infusion of study treatment (Compound 1) were evaluated for safety. The safety analyses include evaluation of the incidence of treatment- emergent AEs, Grade 3 or greater AEs, SAEs, and AEs leading to discontinuation of study treatment. Laboratory and vital signs assessments were evaluated over time on study using descriptive statistics. Shift analyses of relevant clinical laboratory parameters were produced showing shifts across low, normal, and high categories.

Study Assessments

[0427] Demographics and Disease Characteristics: The patient’s date of birth, sex, race and ethnicity were recorded at the Screening visit. Disease characteristics were recorded including date of first contact with the virus (i.e., exposure), date of symptom onset, date SARS-CoV-2 virus test (RT-PCR)/NAT positive.

[0428] Medical History: A detailed medical history for each patient was obtained at Screening. All relevant past and present conditions, as well as prior malignancies and/or surgical procedures, were recorded for the main body systems. Key clinical features and symptoms of COVID-19 collected at screening were not considered adverse events until the definition of an AE was met. [0429] Physical Examination: A complete physical examination was performed at Screening which included the following: an examination of the skin, general appearance, neck (including thyroid), eyes, ears, nose, throat, lymph nodes, chest (lungs), heart, abdomen, musculoskeletal system, neurological system and any additional assessments needed to establish baseline status or evaluate symptoms or adverse events. Symptom-directed physical examinations were performed as needed for safety evaluations on other study visits.

[0430] Smoking History: Smoking history was collected at Screening.

[0431] Height and Weight: Height and weight were collected on Day 1. Weight was required within prior 72 hours prior to randomization and was used for dosing calculations.

[0432] Vital Signs: Vital signs (systolic and diastolic blood pressure, pulse, respiratory rate, and temperature) were collected at all study visits. On days when Compound 1 is administered, vital signs were collected just prior to the infusion of Compound 1, at the completion of infusion, and then at 4 hours post completion of each infusion. At all other study visits, vital signs were collected once.

[0433] 12-Lead Electrocardiogram: A 12-lead ECG was performed as part of Screening. Standard of care results can be used for screening if taken during the current hospitalization. A 12-lead ECG was performed at Days 2, 4, 14, and 28.

[0434] Laboratory Assessments: Laboratory assessment was measured at Screening through Day 14 while the patient is hospitalized, and during follow-up visit on Day 28±2. Hematology included hemoglobin, hematocrit, red blood cells (RBC), white blood cells (WBC) with differential (including bands, if available), and platelet count. Blood chemistry included glucose, phosphorus, total protein, blood urea nitrogen (BUN), creatinine, albumin, and electrolytes (sodium, potassium, calcium, bicarbonate, chloride). Hepatic profile included total, direct and indirect bilirubin, aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), gamma- glutamyl transpeptidase (GGTP). Coagulation profile included International Normalized Ratio (INR) and activated partial thromboplastin time (APTT). Troponin I was collected on Days 2, 4, 6, 9, 11,

14. Urinalysis measured urine pH, specific gravity, protein, glucose, ketones, bilirubin, blood, microscopic. Women of childbearing potential must have had a negative serum/urine pregnancy screen at the Screening visit. A serum/urine pregnancy screen was also collected at Day 28. Additional Laboratory assessments including HS-CRP, D-dimer, ferritin, LDH, and myoglobin were collected at Baseline, Days 6, 9, and 14. [0435] CT Scan (as clinically indicated): Computed tomography (CT) scan was conducted at Screening/Baseline and at Day 28±2.

[0436] Chest X-rav: Chest x-ray was conducted at Screening/Baseline and as indicated per soc.

[0437] WHO Ordinal Disease Severity Assessment: Disease severity was assessed by the WHO COVID-19 Task Force 8-point Ordinal Scale for Clinical Improvement. Disease severity was assessed at Screening/Baseline, daily through Day 14 while the patient was hospitalized,

Day 15-28 if patient was hospitalized and during follow-up visits Day 14 and Day 28±2.

[0438] Oxygen Administration and Assessment of Oxygenation: Use of supplemental oxygen administration including the type, percent, flow start date/time, and flow end date/time and patient’s oxygen saturation (Sa02) was recorded at Screening/Baseline, daily through Day 14 while the patient was hospitalized, Day 15-28 if patient was hospitalized and during follow up visits Day 14 and Day 28±2. Patient Sa02, Pa02, and Fi02 was measured as available.

[0439] Mechanical Ventilation: Use of any ventilatory support type was recorded at Screening/Baseline, daily through Day 14 while the patient was hospitalized, Day 15-28 if patient was hospitalized and during follow-up visits Day 14 and Day 28±2.

[0440] Record Hospital Admission and Discharge Dates: Hospital admission and discharge dates on or before Day -28/Final Visit were recorded.

[0441] Intensive Care Unit Admission and Discharge Dates: Intensive care unit admission and discharge dates on or before Day-28/Final Visit were recorded.

[0442] SARS-CoV-2 Viral Load: Test for confirmation of positive or negative for COVID19 SARS-CoV-2 virus by NAT or PCR (RT-PCR) was collected at baseline and then as indicated as SOC through Day 28±/ End of Study visit.

Example 47. Alternative Synthesis of Compound 1

OCH 3

Step 1. Synthesis of (E)-4-(thiophen-2-yl)but-3-en-2-one (2.1)

[0443] To a solution of thiophene-2-carboxaldehyde (1.1, 40 kg) in 70 L of acetone was added 80 kg of 0.4 M aqueous NaOH slowly at 5 °C. The solution was warmed to room temperature and stirred for 2-3 hours. Diehl or omethane (approx. 4.5 volumes) was then added to the reaction mixture, the layers were allowed to separate, and the organic layer was removed.

The aqueous layer was extracted with dichloromethane (approx. 0.7 volumes), the layers were allowed to separate, and the organic layer was combined with the first organic layer. Water (approx. 0.8 volumes) was added, the layers were allowed to separate, and the organic layer was combined with the first and second organic layers. The combined organic layers were then concentrated and diluted with toluene, which was then distilled with a Dean Stark apparatus to remove water. The mixture was then polish filtered and further concentrated to 3-4 volumes to provide a solution of (E)-4-(thiophen-2-yl)but-3-en-2-one (2.1) in toluene, which was carried forward into the next step without further purification.

Step 2. Synthesis of (lE,4E)-l-(dimethylamino)-5-(thiophen-2-yl)penta-l,4-dien-3- one (2.3) [0444] To a solution of 2.1 in toluene from the previous step, N,N-dimethylformamide dimethylacetal (2.2a, 97 kg), was added and the reaction mixture heated at reflux for 36 hours. The reaction mixture was then concentrated to 2.5-3.5 volumes. The resulting slurry was cooled to room temperature and ethyl acetate was added. The resulting solids were filtered, washed with ethyl acetate, and dried under vacuum to afford (lE,4E)-l-(dimethylamino)-5-(thiophen-2- yl)penta-l,4-dien-3-one (2.3) in 55% yield from thiophene-2-carboxaldehyde (2.1). [0445] In some instances, a slurry of seed material of 2.3 in ethyl acetate was added after concentration of the reaction mixture. Seed material of 2.3 can be prepared using the process described above.

Step 3. Synthesis of Compound 1

[0446] 2.3 (40 kg) was dissolved in 130 kg of isopropyl alcohol under nitrogen, and the mixture was cooled to 10 °C. Acetic acid (13 kg) was then added at 5-25 °C. The solution was then cooled to 5-15 °C, followed by slow addition of hydrazine hydrate (11.5 kg) while maintaining a temperature of 10-25 °C. The reaction mixture was then stirred at 20 °C. Once the reaction was judged to be complete (e.g., using HPLC or thin layer chromatography), water was added, and the resulting solids were filtered, washed with a mixture of isopropyl alcohol and water (1 :4), and dried to afford crude Compound 1 in 90% yield.

[0447] Crude Compound 1 (26 kg) was dissolved in 42 kg acetonitrile at 65-75 °C and then slowly cooled to 15-25 °C to induce crystallization. The resulting solids were filtered and washed with a mixture of acetonitrile and water, and then dried under vacuum. The solids were then dissolved in ethyl acetate at 55-65 °C and polish filtered. n-Heptane (200 kg) was then added to the mixture at 50-60 °C. The mixture was cooled to 15-25 °C, the resulting solids were isolated by filtration, washed with n-heptane, and dried under vacuum to afford crystalline Compound 1 in 67% yield.

Example 48. Preparation and Characterization of Compound 1 Solid Form A General Methods:

X-Ray Powder Diffraction (XRPD)

[0448] XRPD patterns of samples from scaled-up preparations were recorded at ambient temperature on a Bruker D8 Advance X-ray diffractometer (Karlsruhe, Germany) using Cu Ka radiation (l = 1.54 A) at 40 kV, 40 mA passing through a Vario monochromator (Karlsruhe, Germany). The sample was loaded on a zero-background holder and gently pressed by a clean glass slide to ensure co-planarity of the powder surface with the surface of the holder. Data were collected in a continuous scan mode with a step size of 0.05° and dwell time of 1 s over an angular range of 3° to 40° 20. Obtained diffractograms were analyzed with DIFFRAC.EVA diffraction software (Bruker, Wisconsin, USA). [0449] In some cases, the X-ray intensity data were measured on a Bruker D8 Eco diffractometer system equipped with a graphite monochromator and a Cu Ka Sealed tube (l = 1.54 A). The sample was loaded in a polyimide capillary and collected data in transmission mode. Bruker’ s APEX3 software suite (Bruker, Wisconsin, USA) was used to collect and extract the intensity data. Obtained diffractograms were analyzed with TOPAS software (Bruker, Wisconsin, USA). Mercury 4.2.0 software (Build 257471, Cambridge Crystallographic Data Centre, UK) was used to calculate the XRPD patterns from single crystal data. Thermogravimetric Analysis (TGA)

[0450] TGA was performed using a Discovery TGA 5500 (TA® Instruments, New Castle, Delaware, USA) instrument operating with TRIOS software (Version 5.0). The sample was placed in an aluminum pan. The sample cell was purged with dry nitrogen at a flow rate of 15 mL/min. A heating rate of 10 °C/min from 25 °C to desired temperature was used in all the experiments.

Differential Scanning Calorimetry (DSC)

[0451] Conventional DSC experiments were performed using a Discovery DSC 250 (TA® Instruments, New Castle, Delaware, USA) instrument equipped with a refrigerated cooling system (RCS90) and operating with TRIOS software (Version 5.0). The sample cell was purged with dry nitrogen at a flow rate of 50 mL/min. Accurately weighed samples (2-5 mg) placed in TZero pans with a pin hole were scanned at a heating rate of 10 °C/min over a temperature range of 25 °C to desired temperature was used in all the experiments.

Compound 1 Lot I

[0452] Compound 1 was provided (e.g., via the method of Example 47) in a form with an XRPD as shown in FIG. 42, a TGA as shown in FIG. 43, and a DSC as shown in FIG. 44. Herein, this material is referred to as “Compound 1 Lot I.”

Form A

[0453] Compound 1 Form A was synthesized by recrystallizing Compound 1 Lot I from methanol. In a typical reaction, -450 mg of Compound 1 Lot I was dissolved in 2 mL of methanol while heating at 50 °C. Resultant solution was kept at room temperature and allowed for slow evaporation of the solvent. Crystals suitable for single crystal X-ray diffraction were obtained within one day. [0454] Compound 1 Form A bulk powder was prepared as follows: ~ 5 g of Compound 1 Lot I was suspended in 5 mL of methanol and slurried at room temperature for two days. The resulting solid was filtered using 0.45 pm PTFE syringe filter.

[0455] Single crystal X-ray diffraction of Compound 1 Form A was obtained (FIG. 45). Crystal data and structure refinement parameters are summarized below:

[0456] The XRPD pattern of Compound 1 Form A calculated from single crystal X-ray diffraction data is shown in FIG. 46 and is summarized below:

[0457] The XRPD pattern of Compound 1 Form A bulk powder is shown in FIG. 47. Comparison of observed XRPD pattern of Compound 1 Form A bulk powder corresponds well with the calculated XRPD pattern (FIG. 46).

[0458] TGA of Compound 1 Form A is shown in FIG. 48. A weight loss of 0.6% was observed up to 150 °C.

[0459] DSC of Compound 1 Form A is shown in FIG. 49. One endotherm was observed at 116.42 °C.

[0460] A comparison of Compound 1 Lot I is shown in FIG. 50 and verifies that Compound

1 Lot I matches Form A.

Example 49. Preparation of a Compound 1 Formulation

[0461] Polyethylene glycol 300 (175 kg) and polysorbate 80 (35 kg) were combined in a 440 L vessel. Compound 1 (3.5 kg, adjusted for purity, water content, and residual solvent) was added and stirred for 60 minutes. Phosphate buffered saline was filtered and added to the solution until the total mass of the compounding mixture was 381.5 kg. 1.0 N HC1 and/or 1.0 N NaOH was added to bring pH to 7.5-7.9. The resulting solution was filtered through 0.45 pm

Ill and 0.22 mih polish filters in succession, then filled into 20 mL Type 1 glass vials with a target fill weight of 25.12 g per vial and nitrogen over-fill. Vials were capped with B2-40 West stoppers and sealed.