Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
Methods of Treating or Preventing Acute Respiratory Distress Syndrome
Document Type and Number:
WIPO Patent Application WO/2019/178023
Kind Code:
A1
Abstract:
The invention includes a method of preventing or treating acute respiratory distress syndrome (ARDS) in a subject, comprising administering to the subject a thyroid hormone and/or a thyroid receptor β-agonist.

Inventors:
KAMINSKI NAFTALI (US)
LEE PATTY (US)
YU GUOYING (US)
ZHANG YI (US)
Application Number:
PCT/US2019/021750
Publication Date:
September 19, 2019
Filing Date:
March 12, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV YALE (US)
International Classes:
A61K38/22; A61K45/06
Foreign References:
US20170105956A12017-04-20
CN107469086A2017-12-15
US20110044981A12011-02-24
US4997644A1991-03-05
Other References:
See also references of EP 3765059A4
Attorney, Agent or Firm:
DOYLE, Kathryn et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed:

1. A method of preventing or treating acute respiratory distress syndrome (ARDS) in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of at least one compound selected from the group consisting of a thyroid hormone and a thyroid receptor (TR) b-agonist, wherein the at least one thyroid hormone is administered to the subject using a route selected from the group consisting of nasal, inhalational, intratracheal, intrapulmonary, intrabronchial, and inhalation.

2. The method of claim 1, wherein the hormone comprises T3 hormone or T4 hormone.

3. The method of claim 2, wherein the hormone is T3 hormone or T4 hormone.

4. The method of claim 1, wherein the thyroid hormone is administered to the subject using an inhaler.

5. The method of claim 1, wherein the thyroid hormone is formulated as a dry powder blend.

6. The method of claim 1, wherein the TR b-agonist is selected from the group consisting of GC-l (sobetirome or 2-(4-(4-hydroxy-3-isopropylbenzyl)-3, 5-dimethyl phenoxyjacetic acid), KB-2115 (eprotirome or 3-[3,5-dibromo-4-(4-hydroxy-3-propan-2- ylphenoxy)anilino]-3-oxopropanoic acid), KB-141 (({3,5-dichloro-4-[4-hydroxy-3-(propan- 2-yl)phenoxy]phenyl} acetic acid), MB07811 ((4S)-4-(3-chlorophenyl)-2-[(3,5-dimethyl-4-(4- hydroxy-3-isopropylbenzyl)phenoxy)methyl]-2-oxido-[l,3,2]-dioxaphosphonane), MB07344 (3,5-dimethyl-4-(4’-hydroxy-3’-isopropylbenzyl)phenoxy methylphosphonic acid),

MGL3196 (2-[3,5-dichloro-4-[[l,6-dihydro-5-(l -methyl ethyl)-6-oxo-3- pyridazinyl]oxy]phenyl]-2,3,4,5-tetrahydro-3,5-dioxo-l,2,4-triazine-6-carbonitrile),

MGL3745, VK2809 (4-(3-chlorophenyl)-2-((4-(4-hydroxy-3-isopropylbenzyl)-3,5- dimethylphenoxy)methyl)-l,3,2-dioxaphosphinane 2-oxide), VK0214, an ester, salt or solvate thereof, and any mixtures thereof.

7. The method of claim 1, wherein the TR b-agonist is administered to the subject through a route selected from the group consisting of oral, parenteral, nasal, intravenous, subcutaneous, enteral, pulmonary, aerosol, ophthalmic, inhalational, intratracheal, intrabronchial, and topical.

8. The method of claim 1, wherein administration of the at least one compound does not cause significant or undesirable cardiac stimulation, significant or undesirable blood lipid decrease, and/or significant or undesirable weight loss.

9. The method of claim 1, wherein the subject is further administered at least one additional agent that treats, prevents or reduces the symptoms of ARDS.

10. The method of claim 1, wherein the at least one compound is administered to the subject at a frequency selected from the group consisting of about three times a day, about twice a day, about once a day, about every other day, about every third day, about every fourth day, about every fifth day, about every sixth day, and about once a week.

11. The method of claim 1, wherein the subject is a mammal.

12. The method of claim 11, wherein the mammal is a human.

Description:
TITLE OF THE INVENTION

Methods of Treating or Preventing Acute Respiratory Distress Syndrome

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Patent Application No. 62/641,643, filed March 12, 2018, which is incorporated herein by reference in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR

DEVELOPMENT

This invention was made with government support under HL123851 and AG049484 awarded by National Institutes of Health and under W81XWH- 16- 1-0646 and W8XHW-16- 1-0680 awarded by the United States Medical Research and Material Command. The government has certain rights in the invention.

BACKGROUND OF THE INVENTION

Acute respiratory distress syndrome (ARDS), also known as acute lung injury (ALI), is a medical condition occurring in critically ill patients characterized by widespread inflammation in the lungs. In fact, ARDS is not a particular disease, but rather a clinical syndrome triggered by various pathologies such as trauma, pneumonia, and/or sepsis.

ARDS is associated with fluid accumulation in the lungs that is not explained by heart failure (noncardiogenic pulmonary edema), and is typically provoked by an acute injury to the lungs. This results in flooding of the lungs’ microscopic air sacs, partial collapse of the lungs, and low levels of oxygen in the blood (hypoxemia). ARDS is associated with pathological findings including pneumonia, eosinophilic pneumonia, cryptogenic organizing pneumonia, acute fibrinous organizing pneumonia, and diffuse alveolar damage (DAD, which is characterized by a diffuse inflammation of lung tissue). The triggering insult to the tissue is often inflammation or mechanical stress in the lung, causing an initial release of chemical signals and other inflammatory mediators secreted by local epithelial and endothelial cells. Neutrophils and T-lymphocytes migrate into the inflamed lung tissue and amplify the syndrome.

ARDS impairs the lungs’ ability to exchange oxygen and carbon dioxide with the blood across a thin layer of the alveoli. A subject with ARDS presents diffuse injury to cells forming the barrier of the microscopic air sacs of the lungs, surfactant dysfunction, activation of the innate immune system response, and dysfunction of the body’s regulation of clotting and bleeding. Signs and symptoms of ARDS can include shortness of breath, fast breathing, and a low oxygen level in the blood due to abnormal ventilation. ARDS has a death rate between 20 and 50%.

Diagnostic criteria for ARDS were updated as the“Berlin definition” in 2012. Under that definition, ARDS is characterized by: lung injury of acute onset, within 1 week of an apparent clinical insult and with progression of respiratory symptoms; bilateral opacities on chest imaging not explained by other lung pathology; respiratory failure not explained by heart failure or volume overload; and decreased Pa0 2 /Fi0 2 ratio (reduced arterial

oxygenation from available inhaled gas).

ARDS is usually treated with mechanical ventilation in the intensive care unit, but treatment of the underlying cause is crucial. If infection of the lugs is suspected, the patient must be aggressively treated with antibiotics as soon as possible. However, no actual treatment of the syndrome itself has been proven effective so far.

There is thus a need in the art to identify novel therapeutic treatments that can be used to treat or prevent ARDS in an afflicted subject. The present invention addresses and meets this need.

BRIEF SUMMARY OF THE INVENTION

The invention provides a method of preventing or treating acute respiratory distress syndrome (ARDS) in a subject. In certain embodiments, the subject is in need of such prevention and/or treatment.

In certain embodiments, the method comprises administering to the subject a therapeutically effective amount of at least one compound, which can be a thyroid hormone and/or a thyroid receptor (TR) b-agonist. In other embodiments, the method comprises administering to the subject a therapeutically effective amount of a thyroid hormone. In yet other embodiments, the method comprises administering to the subject a therapeutically effective amount of a TR b-agonist.

In certain embodiments, the at least one thyroid hormone is administered to the subject using a route selected from the group consisting of nasal, inhalational, intratracheal, intrapulmonary, intrabronchial, and inhalation.

In certain embodiments, the hormone comprises T3 hormone or T4 hormone. In other embodiments, the hormone is T3 hormone or T4 hormone. In yet other embodiments, the thyroid hormone is administered to the subject using an inhaler. In certain embodiments, the thyroid hormone is formulated as a dry powder blend.

In certain embodiments, the TR b-agonist is GC-l (sobetirome or 2-(4-(4-hydroxy-3- isopropylbenzyl)-3, 5-dimethyl phenoxy)acetic acid), an ester, salt or solvate thereof, and any mixtures thereof. In certain embodiments, the TR b-agonist is KB-2115 (eprotirome or 3- [3,5-dibromo-4-(4-hydroxy-3-propan-2-ylphenoxy)anilino]-3-ox opropanoic acid), an ester, salt or solvate thereof, and any mixtures thereof. In certain embodiments, the TR b-agonist is KB-141 (({3,5-dichloro-4-[4-hydroxy-3-(propan-2-yl)phenoxy]phenyl}a cetic acid), an ester, salt or solvate thereof, and any mixtures thereof. In certain embodiments, the TR b-agonist is MB07811 ((4S)-4-(3-chlorophenyl)-2-[(3,5-dimethyl-4-(4-hydroxy-3-iso propylbenzyl) phenoxy)methyl]-2-oxido-[l,3,2]-dioxaphosphonane), an ester, salt or solvate thereof, and any mixtures thereof. In certain embodiments, the TR b-agonist is MB07344 (3,5-dimethyl- 4-(4’-hydroxy-3’-isopropylbenzyl)phenoxy methylphosphonic acid), an ester, salt or solvate thereof, and any mixtures thereof. In certain embodiments, the TR b-agonist is MGL3196 (2- [3, 5-dichloro-4-[[l,6-dihydro-5-(l -methyl ethyl)-6-oxo-3-pyridazinyl]oxy]phenyl]-2, 3,4,5- tetrahydro-3,5-dioxo-l,2,4-triazine-6-carbonitrile), an ester, salt or solvate thereof, and any mixtures thereof. In certain embodiments, the TR b-agonist is MGL3745, an ester, salt or solvate thereof, and any mixtures thereof. In certain embodiments, the TR b-agonist is VK2809 (4-(3-chlorophenyl)-2-((4-(4-hydroxy-3-isopropylbenzyl)-3,5- dimethylphenoxy)methyl)-l,3,2-dioxaphosphinane 2-oxide), an ester, salt or solvate thereof, and any mixtures thereof. In certain embodiments, the TR b-agonist is VK0214, an ester, salt or solvate thereof, and any mixtures thereof.

In certain embodiments, the TR b-agonist is administered to the subject through a route selected from the group consisting of oral, parenteral, nasal, intravenous, subcutaneous, enteral, pulmonary, aerosol, ophthalmic, inhalational, intratracheal, intrabronchial, and topical.

In certain embodiments, administration of the at least one compound does not cause significant or undesirable cardiac stimulation, significant or undesirable blood lipid decrease, and/or significant or undesirable weight loss.

In certain embodiments, the subject is further administered at least one additional agent that treats, prevents, or reduces the symptoms of ARDS.

In certain embodiments, the at least one compound is administered to the subject at a frequency selected from the group consisting of about three times a day, about twice a day, about once a day, about every other day, about every third day, about every fourth day, about every fifth day, about every sixth day, and about once a week. In certain embodiments, the subject is a mammal. In other embodiments, the mammal is a human.

BRIEF DESCRIPTION OF THE DRAWINGS

The following detailed description of specific embodiments of the invention will be better understood when read in conjunction with the appended drawings. For the purpose of illustrating the invention, specific embodiments are shown in the drawings. It should be understood, however, that the invention is not limited to the precise arrangements and instrumentalities of the embodiments shown in the drawings.

FIG. 1 illustrates an exemplary T3 administration regime in mice. WT mice were nebulized with triiodothyronine (T3, 40pg/kg) for 1 day or 3 days or propylthiouracil (PTU, l00pg/kg) for 3 days prior to 72h continuous hyperoxia exposure.

FIGs. 2A-2F illustrates the finding that T3 attenuates hyperoxia-induced lung injury, inflammation, and oxidants in WT mice. WT mice were nebulized with T3 (40pg/kg) for 1 day or 3 days, or PTU (lOOpg/kg) for 3 days prior to 72h continuous hyperoxia exposure. Control mice were exposed to room air (RA). FIG. 2A: Total cells recovered from BAL were counted. FIG. 2B: Lung permeability was assessed by BAL protein content. FIG. 2C: LDH activity assays were performed on BAL fluid. FIG. 2D: Oxidant generation was detected by Amplex Red from BAL fluid. IL- 1 b (FIG. 2E) and IL-6 (FIG. 2F) were detected by ELISA in BALF. The values are expressed as mean+SD and analyzed by Mann-Whitney test n = 6 for each group). */><0.05, us RA; **/ <0.05, us hyperoxia without pretreatment.

FIG. 3 illustrates the finding that T3 pretreatment increases mitochondrial

biogenesis/fusion/mitophagy and decreases ER stress & apoptosis. WT mice were nebulized with T3 (40pg/kg) for 1 day or 3 days or PTU (lOOpg/kg) for 3 days prior to 72h continuous hyperoxia exposure. Lysates were isolated and immunoblotted against antibodies as listed. b-Actin was used as protein loading control.

FIGs. 4A-4F illustrate the finding that PINK1, not Parkin, mediates T3 effects against hyperoxia-induced lung injury. WT, PINKT 7 , and Parkin 7 mice were nebulized with T3 (40pg/kg) for 3 days or no pretreatment prior to 72h continuous hyperoxia exposure. FIG. 4A: Total cells recovered from BAL were counted. FIG. 4B: Lung permeability was assessed by BAL protein content. FIG. 4C: LDH activity assays were performed on BAL fluid. FIG. 4D: Oxidant generation was detected by Amplex Red from BAL fluid. IL- 1 b (FIG. 4E) and IL-6 (FIG. 4F) were detected by ELISA in BALF. The values are expressed as mean+SD and analyzed by Mann-Whitney test n = 6 for each group). *p< 0.05, us WT no T3 RA; **/ <0.05, US WT no T3 hyperoxia; # p< 0.05, us corresponding RA; u p< 0.05, us corresponding no T3 hyperoxia.

FIG. 5 illustrates the finding that T3 pretreatment increased Mito anti-ROS potential, biogenesis and mitophagy via PINK1. WT and PINK1-KO mice were nebulized with T3 (40pg/kg) for 3 days prior to 72h continuous hyperoxia exposure. Lysates were isolated and immunoblotted against antibodies as listed. b-Actin was used as protein loading control.

FIGs. 6A-6F illustrate the finding that GC-l pretreatment prevents hyperoxia-induced lung injury and accelerates recovery. WT mice were orally gavaged with GC-l (40mg/kg) and then exposed to RA or to continuous hyperoxia for 72 h (H3). Recovery phase was initiated after 72 h hyperoxia (Rl-3, 1, 2 3 and 6 days post-hyperoxia). Cells recovered from BAL were counted as BAL total cell counts (FIG. 6A). FIG. 6B: Lung permeability was assessed by BAL protein content. FIG. 6C: Lactate dehydrogenase (LDH) activity assays were performed on BAL fluid. FIG. 6D: Oxidant generation was detected by Amplex Red from BAL fluid. IL-6 (FIG. 6E) and IL- 1 b (FIG. 6F) was detected by ELISA in BALF. The values are expressed as mean+SD and analyzed by Mann-Whitney test (n = 4 for each group). *p < 0.05 versus no GC-l RA; **p < 0.05 versus no GC-l H3; #p < 0.05 versus corresponding no GC-l.

FIG. 7 illustrates the finding that GC-l pretreatment decreased hyperoxia-induced Mito dysfunction, ER stress, and apoptosis. WT mice were orally gavaged with GC-l (40mg/kg) and exposed to RA or to hyperoxia for 72 h (H3). A time course of recovery after 72 h hyperoxia is shown (Rl-3, 1, 2 3 and 6 days post-hyperoxia). Lysates from mouse lungs were immunoblotted against the listed antibodies (Abs).

FIG. 8 illustrates the finding that GC-l pretreatment increases Tie2 activation and decreases Ang2. WT mice were orally gavaged with GC-l (40mg/kg) and exposed to RA or to hyperoxia for 72 h (H3). A time course of recovery after 72 h hyperoxia is shown (Rl-3, 1, 2 3 and 6 days post-hyperoxia). Lysates from mouse lungs were immunoblotted against the listed antibodies.

DETAILED DESCRIPTION OF THE INVENTION

The present invention relates in part to the unexpected discovery that ARDS can be treated or prevented by administration of a thyroid hormone and/or a thyroid receptor (TR) b- agonist to the subject. In certain embodiments, delivery of a thyroid hormone by aerosol, and/or delivery of a TR b-agonist, reduces injury in a hyperoxia model of lung injury. Non-limiting examples of thyroid hormones contemplated within the invention include, but are not limited to, T3 hormone, T4 hormone, or a salt or solvate thereof. In certain embodiments, administration of the thyroid hormone is targeted to at least a portion of the lungs. In other embodiments, the thyroid hormone is aerosolized. In yet other embodiments, the thyroid hormone T3 is directly delivered into the lung using an inhaler. In yet other embodiments, the thyroid hormone T4 is directly delivered into the lung using an inhaler. In yet other embodiments, this allows for effective delivery of an optimal drug dose within areas of injured lung, maximizing its therapeutic effects, and minimizing potential side effects arising from systemic administration.

Non-limiting examples of TR b-agonists contemplated within the invention include, but are not limited to, GC-l (also known as sobetirome or 2-(4-(4-hydroxy-3- isopropylbenzyl)-3,5-dimethylphenoxy)acetic acid), KB-2115 (also known as eprotirome or 3 - [3 , 5 -dibromo-4-(4-hydroxy-3 -propan-2-ylphenoxy)anilino] -3 -oxopropanoic acid), KB - 141 (({3,5-dichloro-4-[4-hydroxy-3-(propan-2-yl)phenoxy]phenyl}a cetic acid), MB07811 ((4S)- 4-(3 -chlorophenyl)-2- [(3 , 5 -dimethyl-4-(4-hydroxy-3 -i sopropylb enzyl)phenoxy)m ethyl] -2- oxido-[l,3,2]-dioxaphosphonane), MB07344 (3,5-dimethyl-4-(4’-hydroxy-3’- i sopropylb enzyl)phenoxy methylphosphonic acid), MGL3196 (2-[3,5-dichloro-4-[[l,6- dihydro-5-(l-methylethyl)-6-oxo-3-pyridazinyl]oxy]phenyl]-2, 3,4,5-tetrahydro-3,5-dioxo- l,2,4-triazine-6-carbonitrile), MGL3745 (Madrigal Pharmaceuticals, West Conshohocken, PA), VK2809 (4-(3-chlorophenyl)-2-((4-(4-hydroxy-3-isopropylbenzyl)-3,5- dimethylphenoxy)methyl)-l,3,2-dioxaphosphinane 2-oxide), VK0214 (Viking Therapeutics, San Diego, CA), or an ester, salt or solvate thereof.

Esters contemplated in the invention include, but are not limited to, alkyl esters or cycloalkyl esters, such as for example methyl ester, ethyl ester, «-propyl ester, isopropyl ester, «-butyl ester, sec-butyl ester, isobutyl ester, tert- butyl ester, and so forth.

In certain embodiments, administration of the TR b-agonist does not cause significant or undesirable cardiac stimulation, such as but not significantly or undesirably elevated heart rate, significant or undesirable blood lipid decrease, and/or significant or undesirable weight loss. In certain embodiments, the TR b-agonist is administered to the subject at a frequency selected from the group consisting of about three times a day, about twice a day, about once a day, about every other day, about every third day, about every fourth day, about every fifth day, about every sixth day and about once a week. In other embodiments, the TR b-agonist is administered to the subject through a route selected from the group consisting of oral, parenteral, nasal, intravenous, subcutaneous, enteral, pulmonary, aerosol, ophthalmic, inhalational, intratracheal, intrabronchial, and topical.

In certain embodiments, the subject is a mammal. In other embodiments, the mammal is a human.

Definitions

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, non-limiting methods and materials are described.

As used herein, each of the following terms has the meaning associated with it in this section.

As used herein, the articles“a” and“an” are used to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example,“an element” means one element or more than one element.

As used herein,“about,” when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of ±20% or ±10%, in certain embodiments ±5%, in certain embodiments ±1%, and in certain embodiments ±0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.

A disease or disorder is“alleviated” if the severity of a symptom of the disease or disorder, the frequency with which such a symptom is experienced by a patient, or both, is reduced.

In one aspect, the terms“co-administered” and“co-administration” as relating to a subject refer to administering to the subject a compound of the invention or salt thereof along with a compound that may also treat the disorders or diseases contemplated within the invention. In certain embodiments, the co-administered compounds are administered separately, or in any kind of combination as part of a single therapeutic approach. The co- administered compound may be formulated in any kind of combinations as mixtures of solids and liquids under a variety of solid, gel, and liquid formulations, and as a solution.

As used herein, the term“composition” or“pharmaceutical composition” refers to a mixture of at least one compound useful within the invention with a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, nasal, pulmonary and topical administration.

A“disease” as used herein is a state of health of an animal wherein the animal cannot maintain homeostasis, and wherein if the disease is not ameliorated then the animal’s health continues to deteriorate.

A“disorder” as used herein in an animal is a state of health in which the animal is able to maintain homeostasis, but in which the animal’s state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the animal’s state of health.

As used herein, the terms“effective amount,”“pharmaceutically effective amount” and“therapeutically effective amount” refer to a nontoxic but sufficient amount of an agent to provide the desired biological result. That result may be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.

“Instructional material,” as that term is used herein, includes a publication, a recording, a diagram, or any other medium of expression that can be used to communicate the usefulness of the composition and/or compound of the invention in a kit. The instructional material of the kit may, for example, be affixed to a container that contains the compound and/or composition of the invention or be shipped together with a container that contains the compound and/or composition. Alternatively, the instructional material may be shipped separately from the container with the intention that the recipient uses the instructional material and the compound cooperatively. Delivery of the instructional material may be, for example, by physical delivery of the publication or other medium of expression

communicating the usefulness of the kit, or may alternatively be achieved by electronic transmission, for example by means of a computer, such as by electronic mail, or download from a website.

The terms“patient,”“subject” or“individual” are used interchangeably herein, and refer to any animal, or cells thereof whether in vitro or in situ , amenable to the methods described herein. In a non-limiting embodiment, the patient, subject or individual is a human.

As used herein, the term“pharmaceutically acceptable” refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.

As used herein, the term“pharmaceutically acceptable carrier” means a

pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the invention within or to the patient such that it may perform its intended function. Typically, such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be“acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound useful within the invention, and not injurious to the patient. Some examples of materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil;

glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid;

pyrogen-free water; isotonic saline; Ringer’s solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.

As used herein,“pharmaceutically acceptable carrier” also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound useful within the invention, and are physiologically acceptable to the patient. Supplementary active compounds may also be incorporated into the compositions. The“pharmaceutically acceptable carrier” may further include a pharmaceutically acceptable salt of the compound useful within the invention.

Other additional ingredients that may be included in the pharmaceutical compositions used in the practice of the invention are known in the art and described, for example in Remington’s Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference. As used herein, the language“pharmaceutically acceptable salt” refers to a salt of the administered compounds prepared from pharmaceutically acceptable non-toxic acids, including inorganic acids, organic acids, solvates, hydrates, or clathrates thereof.

The term“prevent,”“preventing” or“prevention,” as used herein, means avoiding or delaying the onset of symptoms associated with a disease or condition in a subject that has not developed such symptoms at the time the administering of an agent or compound commences.

As used herein, the term“T3” refers to (A')-tri i odothyroni ne, liothyronine, (S)-2- amino-3-[4-(4-hydroxy-3-iodophenoxy)-3,5-diiodophenyl]propan oic acid, or an ester, salt or solvate thereof.

As used herein, the term“T4” refers to (S)-thyroxine, (¾ ) -2-amino-3-[4-(4-hydroxy- 3,5-diiodophenoxy)-3,5-diiodophenyl]propanoic acid, or an ester, salt or solvate thereof.

A“therapeutic” treatment is a treatment administered to a subject who exhibits signs of pathology, for the purpose of diminishing or eliminating those signs.

As used herein, the term“TR” refers to thyroid receptor.

As used herein, the term“treatment” or“treating” is defined as the application or administration of a therapeutic agent, i.e., a compound of the invention (alone or in combination with another pharmaceutical agent), to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient ( e.g ., for diagnosis or ex vivo applications), who has a condition contemplated herein, a symptom of a condition contemplated herein or the potential to develop a condition contemplated herein, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect a condition contemplated herein, the symptoms of a condition contemplated herein or the potential to develop a condition contemplated herein. Such treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics.

By the term“specifically bind” or“specifically binds,” as used herein, is meant that a first molecule preferentially binds to a second molecule (e.g., a particular receptor or enzyme), but does not necessarily bind only to that second molecule.

Throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed sub-ranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.1, 5.3, 5.5, and 6. This applies regardless of the breadth of the range.

Compounds and Compositions

In certain embodiments, thyroid hormones are useful within the methods of the invention. Non-limiting examples of thyroid hormones contemplated within the invention include, but are not limited to, T3 hormone, T4 hormone, or a salt or solvate thereof.

In certain embodiments, thyroid receptor b-agonists are useful within the methods of the invention. Non-limiting examples of thyroid hormones contemplated within the invention include, but are not limited to, sobetirome or GC-l, eprotirome or KB-2115, KB-141, MB07811, MB07344, MGL3196, MGL3745, VK2809, VK0214, or an ester, salt or solvate thereof.

Esters contemplated within the invention include, but are not limited to, methyl, ethyl, «-propyl, isopropyl, «-butyl, .sec-butyl, isobutyl, /-butyl, «-pentyl, and the like. The compounds used in the methods described herein may form salts with bases, and such salts are included in the present invention. In certain other embodiments, the salts are pharmaceutically acceptable salts. The term“salts” embraces addition salts of free acids and/or bases that are useful within the methods of the invention. Pharmaceutically unacceptable salts may nonetheless possess properties such as high crystallinity, which have utility in the practice of the present invention, such as for example utility in process of synthesis, purification or formulation of compounds useful within the methods of the invention.

Suitable pharmaceutically acceptable base addition salts of compounds used in the methods of the invention include, for example, ammonium salts, metallic salts including alkali metal, alkaline earth metal and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts. Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, ammonium, N,N’-dibenzylethylene-diamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. All of these salts may be prepared from the corresponding compound by reacting, for example, the appropriate base with the compound. Salts may be comprised of a fraction of less than one, one, or more than one molar equivalent of base with respect to any compound of the invention.

In certain other embodiments, the at least one compound of the invention is in a pharmaceutical composition further including at least one pharmaceutically acceptable carrier. Certain formulations useful within methods of the invention can be found in US 2014/0017329 and WO 2009/089093, which are incorporated herein in their entireties by reference.

The methods and formulations described herein include the use of crystalline forms (also known as polymorphs), solvates, amorphous phases, and/or pharmaceutically acceptable salts of compounds having the structure of any compound of the invention, as well as metabolites and active metabolites of these compounds having the same type of activity. Solvates include water, ether ( e.g ., tetrahydrofuran, methyl tert- butyl ether) or alcohol (e.g, ethanol) solvates, acetates and the like. In certain other embodiments, the compounds described herein exist in solvated forms with pharmaceutically acceptable solvents such as water, and ethanol. In other embodiments, the compounds described herein exist in unsolvated form.

The compounds used in the methods of the invention may possess one or more stereocenters, and each stereocenter may exist independently in either the ( R ) or (S) configuration. In certain other embodiments, compounds described herein are present in optically active or racemic forms. The compounds described herein encompass racemic, optically-active, regioisomeric and stereoisomeric forms, or combinations thereof that possess the therapeutically useful properties described herein. Preparation of optically active forms is achieved in any suitable manner, including by way of non-limiting example, by resolution of the racemic form with recrystallization techniques, synthesis from optically- active starting materials, chiral synthesis, or chromatographic separation using a chiral stationary phase. In certain other embodiments, a mixture of one or more isomer is utilized as the therapeutic compound described herein. In other embodiments, compounds described herein contain one or more chiral centers. These compounds are prepared by any means, including stereoselective synthesis, enantioselective synthesis and/or separation of a mixture of enantiomers and/ or diastereoisomers. Resolution of compounds and isomers thereof is achieved by any means including, by way of non-limiting example, chemical processes, enzymatic processes, fractional crystallization, distillation, and chromatography.

In certain other embodiments, the compounds of the invention exist as tautomers. All tautomers are included within the scope of the compounds recited herein.

In certain other embodiments, compounds described herein are prepared as prodrugs. A“prodrug” is an agent converted into the parent drug in vivo. In certain other embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound. In other embodiments, a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound.

In certain other embodiments, sites on, for example, the aromatic ring portion of compounds of the invention are susceptible to various metabolic reactions. Incorporation of appropriate substituents on the aromatic ring structures may reduce, minimize or eliminate this metabolic pathway. In certain other embodiments, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a deuterium, a halogen, or an alkyl group.

Compounds described herein also include isotopically-labeled compounds wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds described herein include and are not limited to ¾ ¾ U C, 13 C, 14 C, 36 Cl, 18 F, 123 I, 125 I, 13 N, 15 N, 15 0, 17 0, 18 0, 32 P, and 35 S. In certain other embodiments, isotopically-labeled compounds are useful in drug and/or substrate tissue distribution studies. In other embodiments, substitution with heavier isotopes such as deuterium affords greater metabolic stability (for example, increased in vivo half-life or reduced dosage requirements). In yet other embodiments, substitution with positron emitting isotopes, such as C, F, O and N, is useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds are prepared by any suitable method or by processes using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.

In certain other embodiments, the compounds described herein are labeled by other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.

The compounds described herein, and other related compounds having different substituents are synthesized using techniques and materials described herein and in the art. General methods for the preparation of compound as described herein are modified by the use of appropriate reagents and conditions, for the introduction of the various moieties found in the formula as provided herein.

Methods

The invention includes a method of preventing or treating ARDS in a subject in need thereof. In certain embodiments, the method comprises administering to the subject therapeutically effective amounts of at least one thyroid hormone and/or TR b-agonist. In other embodiments, the at least thyroid hormone is administered through a route selected from the group consisting of nasal, inhalational, intratracheal, intrapulmonary, intrabronchial, and inhalation. In yet other embodiments, the TR b-agonist is administered to the subject through a route selected from the group consisting of oral, parenteral, nasal, intravenous, subcutaneous, enteral, pulmonary, aerosol, ophthalmic, inhalational, intratracheal, intrabronchial, and topical.

In certain embodiments, the thyroid hormone comprises T3 hormone and/or T4 hormone. In other embodiments, the thyroid hormone is T3 hormone and/or T4 hormone.

In certain embodiments, the TR b-agonist comprises at least one selected from the group consisting of GC-l (2-(4-(4-hydroxy-3-isopropylbenzyl)-3,5-dimethylphenoxy) acetic acid), KB-2115 (3-[3,5-dibromo-4-(4-hydroxy-3-propan-2-ylphenoxy)anilino]-3 - oxopropanoic acid), KB-141 (({3,5-dichloro-4-[4-hydroxy-3-(propan-2-yl)phenoxy]phenyl} acetic acid), MB07811 ((4S)-4-(3-chlorophenyl)-2-[(3,5-dimethyl-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)methyl]-2-oxido-[l,3,2]-dioxaphospho nane), MB07344 (3,5- dimethyl-4-(4’-hydroxy-3’-isopropylbenzyl)phenoxy methylphosphonic acid), MGL3196 (2- [3, 5-dichloro-4-[[l,6-dihydro-5-(l -methyl ethyl)-6-oxo-3-pyridazinyl]oxy]phenyl]-2, 3,4,5- tetrahydro-3,5-dioxo-l,2,4-triazine-6-carbonitrile), MGL3745 (Madrigal Pharmaceuticals, West Conshohocken, PA), VK2809 (4-(3-chlorophenyl)-2-((4-(4-hydroxy-3- isopropylbenzyl)-3,5-dimethylphenoxy)methyl)-l,3,2-dioxaphos phinane 2-oxide), VK0214 (Viking Therapeutics, San Diego, CA), or any salt, ester or solvate thereof.

In certain embodiments, the compositions of the invention are administered to the subject about three times a day, about twice a day, about once a day, about every other day, about every third day, about every fourth day, about every fifth day, about every sixth day and/or about once a week.

In certain embodiments, the subject is further administered at least one additional bioactive agent that treats, prevents or reduces the symptoms of ARDS.

In certain embodiments, the subject is a mammal. In other embodiments, the mammal is a human.

Kits

The invention includes a kit comprising at least one thyroid hormone and/or TR b- agonist, an applicator, and an instructional material for use thereof. The instructional material included in the kit comprises instructions for preventing or treating ARDS in a subject. The instructional material recites the amount of, and frequency with which the at least one thyroid hormone and/or TR b-agonist should be administered to the subject. In other embodiments, the kit further comprises at least one additional bioactive agent that treats, prevents or reduces the symptoms of ARDS.

Combination Therapies

In certain embodiments, the compounds of the invention are useful in the methods of the invention in combination with at least one additional compound useful for treating or preventing ARDS. This additional compound may comprise compounds identified herein or compounds, e.g ., commercially available compounds, known to treat, prevent or reduce the symptoms of ARDS.

A synergistic effect may be calculated, for example, using suitable methods such as, for example, the Sigmoid-E max equation (Holford & Scheiner, 19981, Clin. Pharmacokinet. 6: 429-453), the equation of Loewe additivity (Loewe & Muischnek, 1926, Arch. Exp. Pathol Pharmacol. 114: 313-326) and the median-effect equation (Chou & Talalay, 1984, Adv. Enzyme Regul. 22:27-55). Each equation referred to above may be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination. The corresponding graphs associated with the equations referred to above are the

concentration-effect curve, isobologram curve and combination index curve, respectively.

Administration/Dosage/Formulations

The regimen of administration may affect what constitutes an effective amount. The therapeutic formulations may be administered to the subject either prior to or after the onset of a disease or disorder contemplated in the invention. Further, several divided dosages, as well as staggered dosages may be administered daily or sequentially, or the dose may be continuously infused, or may be a bolus injection. Further, the dosages of the therapeutic formulations may be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.

Administration of the compositions of the present invention to a patient, such as a mammal, such as a human, may be carried out using known procedures, at dosages and for periods of time effective to treat a disease or disorder contemplated in the invention. An effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the state of the disease or disorder in the patient; the age, sex, and weight of the patient; and the ability of the therapeutic compound to treat a disease or disorder contemplated in the invention. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. A non-limiting example of an effective dose range for a therapeutic compound of the invention is from about 1 and 5,000 mg/kg of body weight/per day. One of ordinary skill in the art would be able to study the relevant factors and make the

determination regarding the effective amount of the therapeutic compound without undue experimentation.

Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.

The therapeutically effective amount or dose of a compound of the present invention depends on the age, sex and weight of the patient, the current medical condition of the patient and the progression of a disease or disorder contemplated in the invention. A medical doctor, e.g, physician or veterinarian, having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.

A suitable dose of a compound of the present invention may be in the range of from about 0.01 mg to about 5,000 mg per day, such as from about 0.1 mg to about 1,000 mg, for example, from about 1 mg to about 500 mg, such as about 5 mg to about 250 mg per day.

The dose may be administered in a single dosage or in multiple dosages, for example from 1 to 4 or more times per day. When multiple dosages are used, the amount of each dosage may be the same or different. For example, a dose of 1 mg per day may be administered as two 0.5 mg doses, with about a l2-hour interval between doses.

Compounds of the invention for administration may be in the range of from about 1 pg to about 10,000 mg, about 20 pg to about 9,500 mg, about 40 pg to about 9,000 mg, about 75 pg to about 8,500 mg, about 150 pg to about 7,500 mg, about 200 pg to about 7,000 mg, about 3050 pg to about 6,000 mg, about 500 pg to about 5,000 mg, about 750 pg to about 4,000 mg, about 1 mg to about 3,000 mg, about 10 mg to about 2,500 mg, about 20 mg to about 2,000 mg, about 25 mg to about 1,500 mg, about 30 mg to about 1,000 mg, about 40 mg to about 900 mg, about 50 mg to about 800 mg, about 60 mg to about 750 mg, about 70 mg to about 600 mg, about 80 mg to about 500 mg, and any and all whole or partial increments there between. In some embodiments, the dose of a compound of the invention is from about 1 mg and about 2,500 mg.

In certain embodiments, the compositions of the invention are administered to the patient in dosages that range from one to five times per day or more. In other embodiments, the compositions of the invention are administered to the patient in range of dosages that include, but are not limited to, once every day, every two, days, every three days to once a week, and once every two weeks. It is readily apparent to one skilled in the art that the frequency of administration of the various combination compositions of the invention varies from individual to individual depending on many factors including, but not limited to, age, disease or disorder to be treated, gender, overall health, and other factors. Thus, the invention should not be construed to be limited to any particular dosage regime and the precise dosage and composition to be administered to any patient is determined by the attending physical taking all other factors about the patient into account.

It is understood that the amount of compound dosed per day may be administered, in non-limiting examples, every day, every other day, every 2 days, every 3 days, every 4 days, or every 5 days. For example, with every other day administration, a 5 mg per day dose may be initiated on Monday with a first subsequent 5 mg per day dose administered on

Wednesday, a second subsequent 5 mg per day dose administered on Friday, and so on.

In the case wherein the patient’s status does improve, upon the doctor’s discretion the administration of the inhibitor of the invention is optionally given continuously; alternatively, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a“drug holiday”). The length of the drug holiday optionally varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days,

5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days. The dose reduction during a drug holiday includes from 10%- 100%, including, by way of example only, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.

Once improvement of the patient’s conditions has occurred, a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, is reduced, as a function of the disease or disorder, to a level at which the improved disease is retained. In certain embodiments, patients require intermittent treatment on a long term basis upon any recurrence of symptoms and/or infection.

The compounds for use in the method of the invention may be formulated in unit dosage form. The term“unit dosage form” refers to physically discrete units suitable as unitary dosage for patients undergoing treatment, with each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, optionally in association with a suitable pharmaceutical carrier. The unit dosage form may be for a single daily dose or one of multiple daily doses ( e.g ., about 1 to 4 or more times per day). When multiple daily doses are used, the unit dosage form may be the same or different for each dose.

Toxicity and therapeutic efficacy of such therapeutic regimens are optionally determined in cell cultures or experimental animals, including, but not limited to, the determination of the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population). The dose ratio between the toxic and therapeutic effects is the therapeutic index, which is expressed as the ratio between LD 50 and ED 50. The data obtained from cell culture assays and animal studies are optionally used in formulating a range of dosage for use in human. The dosage of such compounds lies in certain embodiments within a range of circulating concentrations that include the ED 50 with minimal toxicity. The dosage optionally varies within this range depending upon the dosage form employed and the route of administration utilized.

In certain embodiments, the compositions of the invention are formulated using one or more pharmaceutically acceptable excipients or carriers. In certain embodiments, the pharmaceutical compositions of the invention comprise a therapeutically effective amount of a compound of the invention and a pharmaceutically acceptable carrier.

The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity may be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms may be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal and the like. In many cases, it is advisable to include isotonic agents, for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol, in the composition.

In certain embodiments, the present invention is directed to a packaged

pharmaceutical composition comprising a container holding a therapeutically effective amount of a compound of the invention, alone or in combination with a second

pharmaceutical agent; and instructions for using the compound to treat, prevent, or reduce one or more symptoms of a disease or disorder contemplated in the invention.

Formulations may be employed in admixtures with conventional excipients, i.e., pharmaceutically acceptable organic or inorganic carrier substances suitable for any suitable mode of administration, known to the art. The pharmaceutical preparations may be sterilized and if desired mixed with auxiliary agents, e.g ., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure buffers, coloring, flavoring and/or aromatic substances and the like. They may also be combined where desired with other active agents, e.g. , analgesic agents.

Routes of administration of any of the compositions of the invention include oral, nasal, pulmonary, rectal, intravaginal, parenteral, buccal, sublingual, or topical. The compounds for use in the invention may be formulated for administration by any suitable route, such as for oral or parenteral, for example, transdermal, transmucosal ( e.g ., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and perivaginally), (intra)nasal and (trans)rectal), intravesical, intrapulmonary, intraduodenal, intragastrical, intrathecal, subcutaneous, intramuscular, intradermal, intra-arterial, intravenous, intrabronchial, inhalation, and topical administration. In certain embodiments, routes of administration of any of the compositions of the invention include nasal, inhalational, intratracheal, intrapulmonary, intrabronchial, and inhalation.

Suitable compositions and dosage forms include, for example, dispersions, suspensions, solutions, syrups, granules, beads, powders, pellets, liquid sprays for nasal or oral administration, dry powder or aerosolized formulations for inhalation, and the like. It should be understood that the formulations and compositions that would be useful in the present invention are not limited to the particular formulations and compositions that are described herein.

Powdered and granular formulations of a pharmaceutical preparation of the invention may be prepared using known methods. Such formulations may be administered directly to a subject, used, for example, to form a material that is suitable to administration to a subject. Each of these formulations may further comprise one or more of dispersing or wetting agent, a suspending agent, and a preservative. Additional excipients, such as fillers and sweetening, flavoring, or coloring agents, may also be included in these formulations.

Oral Administration

For oral application, particularly suitable are tablets, dragees, liquids, drops, suppositories, or capsules, caplets and gelcaps. The compositions intended for oral use may be prepared according to any method known in the art and such compositions may contain one or more agents selected from the group consisting of inert, non-toxic pharmaceutically excipients that are suitable for the manufacture of tablets. Such excipients include, for example an inert diluent such as lactose; granulating and disintegrating agents such as cornstarch; binding agents such as starch; and lubricating agents such as magnesium stearate. The tablets may be uncoated or they may be coated by known techniques for elegance or to delay the release of the active ingredients. Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert diluent.

Parenteral Administration

As used herein,“parenteral administration” of a pharmaceutical composition includes any route of administration characterized by physical breaching of a tissue of a subject and administration of the pharmaceutical composition through the breach in the tissue. Parenteral administration thus includes, but is not limited to, administration of a pharmaceutical composition by injection of the composition, by application of the composition through a surgical incision, by application of the composition through a tissue-penetrating non-surgical wound, and the like. In particular, parenteral administration is contemplated to include, but is not limited to, subcutaneous, intravenous, intraperitoneal, intramuscular, intrasternal injection, and kidney dialytic infusion techniques.

Buccal, Pulmonary, Inhalational, Intranasal Administration, and So Forth

A pharmaceutical composition of the invention may be prepared, packaged, or sold in a formulation suitable for pulmonary administration via the buccal cavity. Such a

formulation may comprise dry particles that comprise the active ingredient and have a diameter in the range from about 0.5 to about 7 nanometers, and in certain embodiments from about 1 to about 6 nanometers. Such compositions are conveniently in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant may be directed to disperse the powder or using a self-propelling solvent/powder-dispensing container such as a device comprising the active ingredient dissolved or suspended in a low-boiling propellant in a sealed container. In certain embodiments, such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nanometers and at least 95% of the particles by number have a diameter less than 7 nanometers. In certain embodiments, at least 95% of the particles by weight have a diameter greater than 1 nanometer and at least 90% of the particles by number have a diameter less than 6 nanometers. Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.

Low boiling propellants generally include liquid propellants having a boiling point of below 65 °F at atmospheric pressure. Generally the propellant may constitute 50 to 99.9% (w/w) of the composition, and the active ingredient may constitute 0.1 to 20% (w/w) of the composition. The propellant may further comprise additional ingredients such as a liquid non-ionic or solid anionic surfactant or a solid diluent (in certain embodiments having a particle size of the same order as particles comprising the active ingredient).

Pharmaceutical compositions of the invention formulated for pulmonary delivery may also provide the active ingredient in the form of droplets of a solution or suspension. Such formulations may be prepared, packaged, or sold as aqueous or dilute alcoholic solutions or suspensions, optionally sterile, comprising the active ingredient, and may conveniently be administered using any nebulization or atomization device. Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, or a preservative such as methylhydroxybenzoate. The droplets provided by this route of administration in certain embodiments have an average diameter in the range from about 0.1 to about 200 nanometers.

The pharmaceutical composition of the invention may be delivered using an inhalator such as those recited in U.S. Patent No. US 8,333,192 B2, which is incorporated herein by reference in its entirety.

The formulations described herein as being useful for pulmonary delivery are also useful for intranasal delivery of a pharmaceutical composition of the invention.

Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 to 500 micrometers. Such a formulation is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nares. Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of the active ingredient, and may further comprise one or more of the additional ingredients described herein.

Additional Administration Forms

Additional dosage forms of this invention include dosage forms as described in U.S. Patents Nos. 6,340,475; 6,488,962; 6,451,808; 5,972,389; 5,582,837; and 5,007,790.

Additional dosage forms of this invention also include dosage forms as described in U.S. Patent Applications Nos. 20030147952; 20030104062; 20030104053; 20030044466;

20030039688; and 20020051820. Additional dosage forms of this invention also include dosage forms as described in PCT Applications Nos. WO 03/35041; WO 03/35040; WO 03/35029; WO 03/35177; WO 03/35039; WO 02/96404; WO 02/32416; WO 01/97783; WO 01/56544; WO 01/32217; WO 98/55107; WO 98/11879; WO 97/47285; WO 93/18755; and WO 90/11757.

Controlled Release Formulations and Drug Delivery Systems

In certain embodiments, the formulations of the present invention may be, but are not limited to, short-term, rapid-offset, as well as controlled, for example, sustained release, delayed release and pulsatile release formulations.

The term sustained release is used in its conventional sense to refer to a drug formulation that provides for gradual release of a drug over an extended period of time, and that may, although not necessarily, result in substantially constant blood levels of a drug over an extended time period. The period of time may be as long as a month or more and should be a release which is longer that the same amount of agent administered in bolus form.

For sustained release, the compounds may be formulated with a suitable polymer or hydrophobic material that provides sustained release properties to the compounds. As such, the compounds for use the method of the invention may be administered in the form of microparticles, for example, by injection or in the form of wafers or discs by implantation.

In certain embodiments of the invention, the compounds of the invention are administered to a patient, alone or in combination with another pharmaceutical agent, using a sustained release formulation.

The term delayed release is used herein in its conventional sense to refer to a drug formulation that provides for an initial release of the drug after some delay following drug administration and that may, although not necessarily, includes a delay of from about 10 minutes up to about 12 hours.

The term pulsatile release is used herein in its conventional sense to refer to a drug formulation that provides release of the drug in such a way as to produce pulsed plasma profiles of the drug after drug administration.

The term immediate release is used in its conventional sense to refer to a drug formulation that provides for release of the drug immediately after drug administration.

As used herein, short-term refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes and any or all whole or partial increments thereof after drug administration after drug administration.

As used herein, rapid-offset refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes, and any and all whole or partial increments thereof after drug administration.

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures, embodiments, claims, and examples described herein. Such equivalents were considered to be within the scope of this invention and covered by the claims appended hereto. For example, it should be understood, that modifications in reaction conditions, including but not limited to reaction times, reaction size/volume, and experimental reagents, such as solvents, catalysts, pressures, atmospheric conditions, e.g ., nitrogen atmosphere, and reducing/oxidizing agents, with art- recognized alternatives and using no more than routine experimentation, are within the scope of the present application.

It is to be understood that wherever values and ranges are provided herein, all values and ranges encompassed by these values and ranges, are meant to be encompassed within the scope of the present invention. Moreover, all values that fall within these ranges, as well as the upper or lower limits of a range of values, are also contemplated by the present application.

The following examples further illustrate aspects of the present invention. However, they are in no way a limitation of the teachings or disclosure of the present invention as set forth herein.

EXAMPLES

The invention is now described with reference to the following Examples. These Examples are provided for the purpose of illustration only and the invention should in no way be construed as being limited to these Examples, but rather should be construed to encompass any and all variations which become evident as a result of the teaching provided herein.

Example 1:

FIG. 1 illustrates an exemplary T3 administration regime in mice. WT mice were nebulized with triiodothyronine (T3, 40pg/kg) for 1 day or 3 days or propylthiouracil (PTU, l00pg/kg) for 3 days prior to 72h continuous hyperoxia exposure.

FIGs. 2A-2F illustrate the finding that T3 attenuates hyperoxia-induced lung injury, inflammation, and oxidants in WT mice. WT mice were nebulized with T3 (40pg/kg) for 1 day or 3 days, or PTU (lOOpg/kg) for 3 days prior to 72h continuous hyperoxia exposure. Control mice were exposed to room air (RA). As illustrated in FIG. 2A, total cells recovered from BAL were counted. As illustrated in FIG. 2B, lung permeability was assessed by BAL protein content. As illustrated in FIG. 2C, LDH activity assays were performed on BAL fluid. As illustrated in FIG. 2D, oxidant generation was detected by Amplex Red from BAL fluid. IL- 1 b (FIG. 2E) and IL-6 (FIG. 2F) were detected by ELISA in BALF. The values are expressed as mean+SD and analyzed by Mann-Whitney test n = 6 for each group). *p< 0.05, vs RA; **/ <0.05, US hyperoxia without pretreatment.

FIG. 3 illustrates the finding that T3 pretreatment increases mitochondrial

biogenesis/fusion/mitophagy and decreases ER stress & apoptosis. WT mice were nebulized with T3 (40pg/kg) for 1 day or 3 days or PTU (lOOpg/kg) for 3 days prior to 72h continuous hyperoxia exposure. Lysates were isolated and immunoblotted against antibodies as listed. b-Actin was used as protein loading control.

FIGs. 4A-4F illustrate the finding that PINK1, not Parkin, mediates T3 effects against hyperoxia-induced lung injury. WT, PINKT 7 , and Parkin 7 mice were nebulized with T3 (40pg/kg) for 3 days or no pretreatment prior to 72h continuous hyperoxia exposure. As illustrated in FIG. 4A, total cells recovered from BAL were counted. As illustrated in FIG. 4B, lung permeability was assessed by BAL protein content. As illustrated in FIG. 4C, LDH activity assays were performed on BAL fluid. As illustrated in FIG. 4D, oxidant generation was detected by Amplex Red from BAL fluid. IL- 1 b (FIG. 4E) and IL-6 (FIG. 4F) were detected by ELISA in BALF. The values are expressed as mean+SD and analyzed by Mann- Whitney test n = 6 for each group). *p< 0.05, vs WT no T3 RA; **p< 0.05, vs WT no T3 hyperoxia; # p< 0.05, us corresponding RA; m p< 0.05, us corresponding no T3 hyperoxia.

FIG. 5 illustrates the finding that T3 pretreatment increased Mito anti-ROS potential, biogenesis and mitophagy via PINKT. WT and PINKT -KO mice were nebulized with T3 (40pg/kg) for 3 days prior to 72h continuous hyperoxia exposure. Lysates were isolated and immunoblotted against antibodies as listed. b-Actin was used as protein loading control.

FIGs. 6A-6F illustrate the finding that GC-l pretreatment prevents hyperoxia-induced lung injury and accelerates recovery. WT mice were orally gavaged with GC-l (40mg/kg) and then exposed to RA or to continuous hyperoxia for 72 h (H3). Recovery phase was initiated after 72 h hyperoxia (Rl-3, 1, 2 3 and 6 days post-hyperoxia). Cells recovered from BAL were counted as BAL total cell counts (FIG. 6A). As illustrated in FIG. 6B, lung permeability was assessed by BAL protein content. As illustrated in FIG. 6C, lactate dehydrogenase (LDH) activity assays were performed on BAL fluid. As illustrated in FIG. 6D, oxidant generation was detected by Amplex Red from BAL fluid. IL-6 (FIG. 6E) and PMb (FIG. 6F) was detected by ELISA in BALF. The values are expressed as mean+SD and analyzed by Mann-Whitney test (n = 4 for each group). *p < 0.05 versus no GC-l RA; **p < 0.05 versus no GC-l H3; #p < 0.05 versus corresponding no GC-L

FIG. 7 illustrates the finding that GC-l pretreatment decreased hyperoxia-induced Mito dysfunction, ER stress and apoptosis. WT mice were orally gavaged with GC-l (40mg/kg) and exposed to RA or to hyperoxia for 72 h (H3). A time course of recovery after 72 h hyperoxia is shown (Rl-3, 1, 2 3 and 6 days post-hyperoxia). Lysates from mouse lungs were immunoblotted against the listed antibodies (Abs).

FIG. 8 illustrates the finding that GC-l pretreatment increases Tie2 activation and decreases Ang2. WT mice were orally gavaged with GC-l (40mg/kg) and exposed to RA or to hyperoxia for 72 h (H3). A time course of recovery after 72 h hyperoxia is shown (Rl-3, 1, 2 3 and 6 days post-hyperoxia). Lysates from mouse lungs were immunoblotted against the listed antibodies.

Enumerated Embodiments

The following enumerated embodiments are provided, the numbering of which is not to be construed as designating levels of importance.

Embodiment 1 provides a method of preventing or treating acute respiratory distress syndrome (ARDS) in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of at least one compound selected from the group consisting of a thyroid hormone and a thyroid receptor (TR) b-agonist, wherein the at least one thyroid hormone is administered to the subject using a route selected from the group consisting of nasal, inhalational, intratracheal, intrapulmonary, intrabronchial, and inhalation.

Embodiment 2 provides the method of Embodiment 1, wherein the hormone comprises T3 hormone or T4 hormone.

Embodiment 3 provides the method of any of Embodiments 1-2, wherein the hormone is T3 hormone or T4 hormone.

Embodiment 4 provides the method of any of Embodiments 1-3, wherein the thyroid hormone is administered to the subject using an inhaler.

Embodiment 5 provides the method of any of Embodiments 1-4, wherein the thyroid hormone is formulated as a dry powder blend.

Embodiment 6 provides the method of any of Embodiments 1-5, wherein the TR b- agonist is selected from the group consisting of GC-l (sobetirome or 2-(4-(4-hydroxy-3- isopropylbenzyl)-3, 5-dimethyl phenoxyjacetic acid), KB-2115 (eprotirome or 3-[3,5- dibromo-4-(4-hydroxy-3-propan-2-ylphenoxy)anilino]-3-oxoprop anoic acid), KB-141 (({3,5- dichloro-4-[4-hydroxy-3-(propan-2-yl)phenoxy]phenyl}acetic acid), MB07811 ((4S)-4-(3- chlorophenyl)-2-[(3,5-dimethyl-4-(4-hydroxy-3-isopropylbenzy l)phenoxy)methyl]-2-oxido- [l,3,2]-dioxaphosphonane), MB07344 (3,5-dimethyl-4-(4’-hydroxy-3’- isopropylbenzyl)phenoxy methylphosphonic acid), MGL3196 (2-[3,5-dichloro-4-[[l,6- dihydro-5-(l-methylethyl)-6-oxo-3-pyridazinyl]oxy]phenyl]-2, 3,4,5-tetrahydro-3,5-dioxo- l,2,4-triazine-6-carbonitrile), MGL3745, VK2809 (4-(3-chlorophenyl)-2-((4-(4-hydroxy-3- isopropylbenzyl)-3,5-dimethylphenoxy)methyl)-l,3,2-dioxaphos phinane 2-oxide), VK0214, an ester, salt or solvate thereof, and any mixtures thereof. Embodiment 7 provides the method of any of Embodiments 1-6, wherein the TR b- agonist is administered to the subject through a route selected from the group consisting of oral, parenteral, nasal, intravenous, subcutaneous, enteral, pulmonary, aerosol, ophthalmic, inhalational, intratracheal, intrabronchial, and topical.

Embodiment 8 provides the method of any of Embodiments 1-7, wherein

administration of the at least one compound does not cause significant or undesirable cardiac stimulation, significant or undesirable blood lipid decrease, and/or significant or undesirable weight loss.

Embodiment 9 provides the method of any of Embodiments 1-8, wherein the subject is further administered at least one additional agent that treats, prevents or reduces the symptoms of ARDS.

Embodiment 10 provides the method of any of Embodiments 1-9, wherein the at least one compound is administered to the subject at a frequency selected from the group consisting of about three times a day, about twice a day, about once a day, about every other day, about every third day, about every fourth day, about every fifth day, about every sixth day, and about once a week.

Embodiment 11 provides the method of any of Embodiments 1-10, The method of claim 1, wherein the subject is a mammal.

Embodiment 12 provides the method of any of Embodiments 1-11, wherein the mammal is a human.

The disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated herein by reference in their entirety.

While this invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of this invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims are intended to be construed to include all such embodiments and equivalent variations.