WO2020123610A1 - Compounds and methods for treating copd and/or lung fibrosis - Google Patents

Compounds and methods for treating copd and/or lung fibrosis Download PDF

Info

Publication number
WO2020123610A1
WO2020123610A1 PCT/US2019/065652 US2019065652W WO2020123610A1 WO 2020123610 A1 WO2020123610 A1 WO 2020123610A1 US 2019065652 W US2019065652 W US 2019065652W WO 2020123610 A1 WO2020123610 A1 WO 2020123610A1
Authority
WO
WIPO (PCT)
Prior art keywords
subject
isothiazol
agonist
mif
benzo
Prior art date
Application number
PCT/US2019/065652
Other languages
French (fr)
Inventor
Patty J. LEE
Richard Bucala
Maor SAULER
Original Assignee
Yale University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Yale University filed Critical Yale University
Publication of WO2020123610A1 publication Critical patent/WO2020123610A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/428Thiazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41921,2,3-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/423Oxazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents

Definitions

  • FIGS. 2A-2C illustrate that smokers with COPD have decreased circulating MIF and gene expression.
  • Tire articles“a” and“an” are used herein to refer to one or to more than one f to at least one) of the grammatical object of the article.
  • “an element” means one element or more than one element.
  • MIF-mediated disease refers to any disease or disorder in which MIF contributes, directly or indirectly, to the pathology of the disease or any disease or di sorder which may be treated or prevented by altering the acti vity of MIF.
  • the MIF-mediated disease is associated with pathologically low levels of MIF activity.
  • the MIF-mediated disease is COPD.
  • the MIF-mediated disease is a fibrotic disease.
  • the MIF-mediated disease is lung fibrosis.
  • “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 tire 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.
  • treating a disease or disorder means reducing the frequency with which a symptom of the disease or disorder is experienced by a patient.
  • Disease and disorder are used interchangeably herein.
  • Ranges throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description m 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 subranges 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 subranges such as from ! 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, 5.3, and 6. This applies regardless of the breadth of die range.
  • the invention provides a method of treating chronic obstructive pulmonary disease (COPD) in a female subject in need thereof, the method comprising administering to the female subject an effective amount of at least one MIF agonist.
  • COPD chronic obstructive pulmonary disease
  • MIF agonist emphysema type COPD.
  • the compounds of the invention may possess one or more stereocenters, and each stereocenter may exist independently in either the (R) or (S) configuration.
  • compounds described herein are present in optically active or racemic forms. It is to be understood that the compounds described herein encompass racemic, optically-active, regioisomeric and stereoisomeric forms, or combinations thereof that possess the
  • optically active forms 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.
  • a mixture of one or more isomer is utilized as the therapeutic compound described herein.
  • compounds described herein contain one or more chiral centers. These compounds are prepared by any means, including
  • the compounds of the invention may exist as tautomers.
  • “Tautomerization” is a form of isomerization involving the migration of a proton
  • tautomerization e.g., in solution
  • a chemical equilibrium of tautomers can be reached.
  • tautomerization is between a ketone and its corresponding enol.
  • Heterocycles may form tautomers such as the interconversion of pyrrolidinone and hydroxy pyrrole. Ail tautomers are included within the scope of the compounds presented herein.
  • Compounds of the invention can in certain embodiments form acids or bases.
  • the invention contemplates acid addition salts.
  • the invention contemplates base addition salts.
  • the invention contemplates pharmaceutically acceptable acid addition salts.
  • the invention contemplates pharmaceutically acceptable base addition salts.
  • Pharmaceutically acceptable salts refer to salts of those bases or acids that are not toxic or otherwise biologically undesirable.
  • Suitable pharmaceutically acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid.
  • inorganic acids include hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric (including sulfate and h drogen sulfate), and phosphoric acids (including hydrogen phosphate and dihydrogen phosphate).
  • Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, malonic, saccharin, fumaric, pyruvic, aspartic, glutamic, benzoic, anthramlic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfomc, ethanes ulfonic, benzenesulfonic, pantothenic, trifluoromethanesulfonic, 2- hydroxyethanesu!fonie, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic,
  • the invention provides a method of treating a bacterial respirator ⁇ ' infection in a subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist.
  • the bacterial respirator ⁇ ' infection is a cigarette smoke- associated respirator ⁇ ' infection.
  • the term cigarette smoke-associated respirator ⁇ ' infection is a respiratory infection that is typically associated with or aggravated by exposure to cigarette smoke.
  • the bacterial respirator ⁇ ' infection is Streptococcus pneumonia.
  • Formulations may be employed in admixtures with conventional excipients, i.e., pharmaceutically acceptable organic or inorganic carrier substances suitable for oral, parenteral, nasal, intravenous, subcutaneous, enteral, or any other 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., other analgesic agents.
  • the compounds of the invention may be in the form of tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e g. , polyvinylpyrrolidone, hydroxypropy!cel!u!ose or hydroxypropyl methylcellulose); fillers (e.g., cornstarch, lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrates (e.g., sodium starch gly collate); or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e g. , polyvinylpyrrolidone, hydroxypropy!cel!u!ose or hydroxypropyl methylcellulose
  • fillers e.g., cornstarch, lactose, microcrystalline cellulose or calcium phosphate
  • lubricants e.g., magnesium stearate, talc, or silica
  • the tablets may be coated using suitable methods and coating materials such as OPADRYTM film coating systems available from Coiorcon, West Point, Pa. (e.g., OPADRYTM OY Type, OYC Type, Organic Enteric OY-P Type, Aqueous Enteric OY-A Type, OY-PM Type and OPADRYTM White, 32K18400).
  • OPADRYTM film coating systems available from Coiorcon, West Point, Pa.
  • Liquid preparation for oral administration may be in the form of solutions, syrups or suspensions.
  • the liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g, sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agent (e.g , lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and
  • suspending agents e.g, sorbitol syrup, methyl cellulose or hydrogenated edible fats
  • emulsifying agent e.g , lecithin or acacia
  • non-aqueous vehicles e.g., almond oil, oily esters or ethyl alcohol
  • preservatives e.g. methyl or propyl p-hydroxy benzoates or sorbic acid.
  • 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 that is longer that the same amount of agent administered in bolus form.
  • the compounds may be formulated with a suitable polymer or hydrophobic material that provides sustained release properties to the compounds.
  • the compounds for use the method of the invention may be administered in the form of microparticles, for example, by injection or m the form of wafers or discs by implantation.
  • delayed release is used herein in its conventional sense to refer to a drag formulation that provides for an initial release of the drag after some delay following drug administration and that mat, although not necessarily, includes a delay of from about 10 minutes up to about 12 hours.
  • Actual dosage levels of the cells in the pharmaceutical formulations of this invention may be varied so as to obtain an amount of the compositi on that are effective to achieve the desired therapeutic response for a particular subject, composition, and mode of
  • Lungs were removed from control and experimental mice and inflated slowly at a constant pressure of 25 cm. Lung volume was assessed by volume displacement.
  • HUVEC was treated with 0% or 2.5% CSE along with the following 1) recombinant M1F (rMIF; lOOng/ml), 2) MIF silencing RNA (siMIF), 3) neutralizing MIF antibody (MIF nAb). 4) MIF098 - a MIF antagonist, or 5) MIF20 - a MIF agonist. After 24 hours, gene expression and SA- -gal activity were determined. To determine the mechanism of MIF20, U0126, an Erk inhibitor, was treated 1 hour prior to MIF20 treatment. ABT263, a known senolytic agent, were used as positive control.
  • CD74 expression in endothelial cells were increased in people with COPD.
  • Activation of MIF signaling pathway attenuated CSE-induced pl6 and p21 expression as well as SA-b- gal activity. These protective effects were abolished by ERK inhibitor.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pulmonology (AREA)
  • Engineering & Computer Science (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

In various aspects and embodiments the invention provides methods for the treatment of a MIF-mediated disease. In one aspect, the invention provides a method of treating a fibrotic disease in a male subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist. In various embodiments, the fibrotic disease is lung fibrosis. In another aspect, the invention provides a method of treating chronic obstructive pulmonary disease (COPD) in a female subject in need thereof, the method comprising administering to the female subject an effective amount of at least one MIF agonist. In various embodiments, the COPD is emphysema type COPD.

Description

TiTLE OF THE INVENTION
Compounds and Methods for Treating COPD and/or Lung Fibrosis
CROSS-REFERENCE TO RELATED APPLICATIONS
The present application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Patent Application No. 62/778,187 filed December 11, 2018 and U.S. Provisional Patent Application No. 62/910,164 filed October 3, 2019, each of which is incorporated herein by reference in its entirely.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR
DEVELOPMENT
Tins invention was made with government support under W81XWH-16-1 -0680 awarded by United States Army Medical Research and Material Command and under HL 135402 awarded by N ational Institutes of Health. The government has certain rights in the invention.
BACKGROUND OF THE INVENTION
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide and is especially common in military members and Veterans. COPD is a progressive, destructive process of airflow obstruction, leading to respiratory failure. A major cause of mortality in people with COPD is bacterial pneumonia, such as those caused by Streptococcus pneumonia ( S. pneumoniae). In addition to aging, a history of primary cigarette smoke exposure (CSE), or significant second-hand CSE, is the most common environmental risk factor for developing COPD - even years after smoking exposure ceases.
There is a need in the ait for compositions and methods for treatment or prevention of COPD and other diseases. This disclosure addresses that need.
SUMMARY OF THE INVENTION
In one aspect, the invention provides a method of treating a fibrotic disease in a male subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist.
In various embodiments, the fibrotic disease is lung fibrosis. In another aspect, the invention provides a method of treating chronic obstructive pulmonary disease (COPD) in a female subject in need thereof, the method comprising administering to the female subject an effective amount of at least one MIF agonist.
In various embodiments, the COPD is emphysema type COPD.
In various embodiments, the at least one MIF agonist is selected from the group consisting of:
O
2-(4-chlorophenyl)-6-fluorobenzo[d]isothiazol-3(2H)-one
Figure imgf000004_0001
2-(4-chloropheny!)-5-fluorobenzo[djisothiazo!-3(2H)-one
Figure imgf000004_0002
2-(4-chlorophenyl)-3-oxo-2,3-dihydrobenzo[d|isothiazole-5-carbonitrile
Figure imgf000004_0003
O
5-fluoro-2-(4-methoxyphenyl)benzo[d]isothia ol-3(2H)-one
Figure imgf000004_0004
5-fluoro-2~(3-methoxyphenyl)benzo[d]isothiazol-3(2H)-one
Figure imgf000004_0005
2-(4-chlorophenyl)-5,6-difluorobenzo[d]isotbiazol-3(2H)-one
Figure imgf000004_0006
7-fluoro-2-(3-methoxyphenyl)benzo[d]isothiazol-3(2H)-one
Figure imgf000004_0007
6-fluoro-3-(3-hydroxybenzyl)benzo[d]oxazol-2(3H)-one
Figure imgf000004_0008
5-fluoro-3 -(3 -methoxy benzy J)benzo[ d| oxazol-2(3H)-one
Figure imgf000005_0001
Figure imgf000005_0002
4-(4-(pyridin-3-yl)- 1H- 1 ,2,3-triazoJ-l -yljphenol
Figure imgf000005_0003
4-(4~(isoquinolin-4-yl)~ 1H- 1 ,2,3-triazol- 1 -yljphenol
Figure imgf000005_0004
or a salt or solvate thereof, and any mixtures thereof.
In various embodiments, the MIF agonist is 4-(4-(py ri din-3 -yl)-l H- 1,2,3 -triazol-1 - yljphenol, or a salt or solvate thereof.
In various embodiments, the at least one agonist is formulated in a composition further comprising at least one pharmaceutically acceptable excipient.
In various embodiments, the at least one agonist is administered by at least one route selected from the group consisting of inha!ationa!, oral, rectal, vaginal, parenteral, intracranial, topical, transdermal, pulmonary , intranasal, buccal, ophthalmic, intrathecal, and intravenous.
In various embodiments, the subject is a mammal.
In various embodiments, the subject is a human.
In another aspect, the invention provides a method of treating a bacterial respiratory' infection in a subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist. In various embodiments, the bacterial respiratory infection is a cigarette smoke- associated respiratory infection.
In various embodiments, the bacterial respiratory infection is Streptococcus pneumonia.
In various embodiments, method of treating bacterial pneumonia in a subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist.
In various embodiments, the subject has COPD.
BRIEF DESCRIPTION OF THE DRAWINGS
The following detailed description of selected embodiments of the invention will be better understood when read in conjunction with the appended drawings. For the purpose of illustrating the invention, selected 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 m the drawings.
FIG. I illustrates that COPD is increasing as a source of mortality in proportion to other leading causes of death in the United States.
FIGS. 2A-2C illustrate that smokers with COPD have decreased circulating MIF and gene expression. FIG. 2A shows current and formal smokers (n=72, >65 years of age mean and 95% confidence limits). Results adjusted for age (years), sex (male/female), smoking history' (formal, current, never) and race (white, nonwhite). FIGS. 2B-2C show the correlation of MIF gene expression with FEV) (FIG. 2B) and percent emphysema (FIG. 2C) in a subset of COPDgene participants that underwent expression array analysis of peripheral blood mononuclear cells (n=! 36).
FIGS. 3A-3C illustrate that genetic MIF deficiencies in mice result in emphysema (n=4-9 /group; aging mice).
FIGS. 4A-4D illustrate the effect of 4-(4-(pyridin-3-yl)-lH-l,2,3-triazol-l -yl)phenol on pres sure- volume (PV) loops in short-term cigarete smoke (CS)-exposed mice. The mice were exposed to CS in Teague machine for 3 months (6 h/day, 5 day s/week). After anesthesia and paralysis the mice, PV loops were measured by FlexiVent (Scireq, Montreal, Canada). FIGS. 4A-4B depict CS exposure induced upward-shift of P V loops in both female and male mice compared to those of room-air (RA) group. FIGS. 4C-4D show Female (FIG. 4C) and male (FIG. 4D) mice that were orally treated vehicle (V), 25, 50 or 100 mg/kg of 4-(4- (pyridin-3-yl)-lH-l,2,3-triazol-l-yl)phenol during the last 1 month. The data are shown as mean ± SEM (n=5 per group). *p<0.05 vs. RA group.
FIGS. 5A-5D show that CD74 expression is increased in endothelial cells isolated from subjects with COPE).
FIGS. 6A-6J show that MIF signaling prevents CSE-induced senescence in endothelial cells.
FIGS. 7A-7E show that M1F20 shows antisenescence effect through ERK signaling in endothelial cells.
FIGS. 8A-8B show that MIF2Q attenuates CS-induced left-shift of PV loop.
FIG. 9 elucidates the protective mechanisms of MIF signaling against endothelial senescence in smoke-induced COPD.
FIGS. 10A and IQB: Effect of Streptococcus pneumoniae (Spneu) on body weight and survival rate in mice. C57BL/6J mice were injected Spneu (1 x 10s CFUs/50 uL/mouse; ATCC #6303) into the trachea. FIG. 10A: Body weight was measured once a day after infection. Data are shown as mean ± SEM. */><0.05, ** ><0.01 compared to control group (student /-test). FIG. 10B: Survival was monitored once a day after infection. Data are shown as percentage of survival rate. *p< 0.05 compared to control group (Log-rank, Mantel-Cox test).
FIGS. 11A and 11B: Effect of macrophage migration inhibitory factor (MIF) agonist on cigarette smoke (CS) and Streptococcus pnuemoniae (Spneu)-induced body weight loss and mortality. C57BL/6J mice were exposed to CS in a smoking machine (Teague TE-10) for 4 weeks. After the last CS exposure, mice were injected Spneu (1 x 105 CFUs/50 uL/mouse; ATCC #6303) into the trachea. MIF20 (M), a MIF agonist, was administered to mice once a day after Spneu infection (3 mg/150 uL/mouse, oral treatment). FIG. 11 A: Body weight was measured once a day after infection. Data are shown as mean ± SEM. FIG. 1 IB: Survival was monitored once a day after infection. Data are shown as percentage of survi val rate. Statistic analysis was performed by Log-rank, Mantel-Cox test. n:::6 for CS + Spneu, n=7 for CS + Spneu + M group.
FIGS. 12A-12C: Effect of macrophage migration inhibitory factor (MIF) agonist on cigarette smoke (CS)-induced pressure-volume (PV) loops shift in mice. AKR/J mice were exposed to cigarette smoke in smoking machine (Teague TE-10) for 4 months. MIF20 (M), a MIF agonist, was injected to mice once a day during the last 2 months of CS exposure (3 mg/150 uL/mouse, oral treatment). PV loops ware determined using a FlexiVent in (FIG.
12A) male and (FIG. 12B) female mice, and (FIG. I2C) pooled the data (n=T3, 7, 1 1 , and 12 for RA, RA+M, CS, and CS+M, respectively). Data are shown as mean ± SEM. *p<0.05, **p<0.01 , ***p<Q.00l compared to RA group;†p<0.05 compared to CS group (2way ANQVA). RA=room-air.
DETAILED DESCRIPTION
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 the invention pertains. Although any methods and materials similar or equivalent to those described herein can be used in the practice for testing of the present invention, selected materials and methods are described herein. In describing and claiming the present invention, the following terminology will be used.
It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting.
Tire articles“a” and“an” are used herein to refer to one or to more than one f
Figure imgf000008_0001
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.
“About” as used herein 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%, more preferably ±5%, even more preferably ±1%, and still more preferably ±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.
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, subcutaneous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration.
An“effective amount” or“therapeutically effective amount” of a compound is that amount of compound that is sufficient to provide a beneficial effect to the subject to which the compound is administered. An“effective amount” of a delivery vehicle is that amount sufficient to effectively bind or deliver a compound. As used herein,“macrophage migration inhibitory factor’' or“MIF” refers to macrophage migration inhibitor)' factor (MIF) protein, the macrophage MIF gene (UniProt - PI 4174), any mRNA encoded by the gene or any homolog thereof. MIF is an innate immune protein that binds CD74, and has the amino acid sequence:
SEQ ID NO: 1
MPMFIVNTNVPRASVPDGFLSELTQQLAQATGKPPQYIAVHWPDQLMAFGG
SSEPCALCSLHSIGKIGGAQNRSYSKLLCGLLAERLRISPDRVYINYYDMNA
ANVGWNNSTFA
the term may also refer to the mature, bioactive protein which lacks the N-terminal methionine.
As used herein, the term“MIF-mediated disease’ refers to any disease or disorder in which MIF contributes, directly or indirectly, to the pathology of the disease or any disease or di sorder which may be treated or prevented by altering the acti vity of MIF. In various embodiments, the MIF-mediated disease is associated with pathologically low levels of MIF activity. In various embodiments, the MIF-mediated disease is COPD. In various embodiments, the MIF-mediated disease is a fibrotic disease. In various embodiments, the MIF-mediated disease is lung fibrosis.
The terms“patient,”“subject,”“individual,” and the like 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 certain non-limiting embodiments, 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 com 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 tire 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,“treating a disease or disorder” means reducing the frequency with which a symptom of the disease or disorder is experienced by a patient. Disease and disorder are used interchangeably herein.
As used herein, the term“treatment” or“treating” encompasses prophylaxis anchor therapy. Accordingly, the compositions and methods of the present invention are not limited to therapeutic applications and can be used in prophylactic ones. Therefore “treating” or“treatment” of a state, disorder or condition includes: (i) preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in a subject that may be afflicted with or predisposed to the slate, disorder or condition but does not yet experience or display clinical or subclinicaJ symptoms of the state, disorder or condition, (ii) inhibiting the state, disorder or condition, i.e., arresting or reducing the development of the disease or at least one clinical or subclimcai symptom thereof, or (iii) relieving the disease, i.e. causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms. The usage of compounds (e.g. MIF agonists) includes, but not limited to, co-treatment with other drugs and therapeutic interventions.
Ranges: throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description m 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 subranges 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 subranges such as from ! 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, 5.3, and 6. This applies regardless of the breadth of die range.
Figure imgf000011_0001
Methods of Treating Disease
Without wishing to be limited by theory, the invention is based in part on the discovery of a gender specific response to M1F agonists (FIGS. 4A-4D). In one aspect, the invention provides a method of treating a fibrotic disease in a male subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist. In various embodiments, the fibrotic disease is lung fibrosis.
In another aspect, the invention provides a method of treating chronic obstructive pulmonary disease (COPD) in a female subject in need thereof, the method comprising administering to the female subject an effective amount of at least one MIF agonist. In various embodiments, COPD is emphysema type COPD.
In various embodiments, the at least one MIF agonist is any of the compounds recited in U.S. Patent Nos. 6,774,227; Bemhagen, et al, 1993, Nature 365:756-759; S enter, et al, 2002, Free Natl Acad Sci USA 99: 144-149; Dios, et al, 2002, J. Med Chem 45:2410-2416; Lubetsky, et al, 2002, J Biol Chem 277:24976-24982; which are hereby incorporated by reference.
In various embodiments, the at least one MIF agoni st is any of the MIF agonists recited in US 2018/0162813, which is incorporated herein by reference. In various embodiments, the at least one MIF agonist is selected from the group consisting of: O
2-(4-chlorophenyl)-6-fluorobenzo d]isothiazol-3(2H)-one
Figure imgf000012_0001
O
2-(4-chlorophenyl)-5-fluorobenzo[d]isothiazol-3(2H)-one
Figure imgf000012_0002
2-(4-chlorophenyl)-3-oxo-2,3-dihydrobenzo[d]isothiazole-5-carbonitrile
Figure imgf000012_0003
O 5-fluoro-2-(4-methoxyphenyl)benzo[d]isothiazol-3(2H)-one
Figure imgf000012_0004
5-fluoro-2-(3-methoxyphenyl)benzo[d]isothiazol-3(2H)-one
Figure imgf000012_0005
2-(4-chlorophenyl)-5,6-dif!uorobenzo[d]isothiazol-3(2H)-one
Figure imgf000012_0006
7-fluoro-2-(3-methoxyphenyl)benzo d]isothiazol-3(2H)-one
Figure imgf000012_0007
6-fluoro-3-(3-hydroxybenzyl)benzo[d]oxazol-2(3H)-one
Figure imgf000012_0008
5-fluoro-3-(3-methoxybenzyl)benzo[d]oxazol-2(3H)-one
Figure imgf000012_0009
2-phenylhenzo[
Figure imgf000012_0010
6-methyl-2-phenylbenzo[d]thiazole Me
Figure imgf000013_0001
4-(l -(2-methoxy benzyl)-! H-l ,2,3-tnazol-4-y l)phenol
Figure imgf000013_0002
4-(4-(pyridin-3-yl)-lH-l,2,3-triazol-l -yl)phenol
Figure imgf000013_0003
4-(4-(isoquinolin-4-yl)-lH-l,2,3-triazol-I-yl)phenol
Figure imgf000013_0004
;
or a salt or solvate thereof, and any mixtures thereof.
In various embodiments, the MIF agonist is 4-(4-(pyridin-3-yl)-lH-l,2,3-triazoI-l- yi)phenoi, or a salt or solvate thereof.
In various embodiments, the composition further comprises at least one
pharmaceutically acceptable excipient. Further details regarding dosage and administration are discussed elsewhere herein. In various embodiments, the subject is a mammal. In various embodiments, the subject is a human.
The compounds of the invention may possess one or more stereocenters, and each stereocenter may exist independently in either the (R) or (S) configuration. In certain embodiments, compounds described herein are present in optically active or racemic forms. It is to be understood that 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 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, enantiosel active synthesis and/or separation of a mixture of enantiomers and/ or diastereomers. 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. All possible stereochemical configurations of a given compound containing chiral center(s) are contemplated. All possible mixtures enriched with a particular enantiomer or diasteromer(s) are contemplated. Ail pure individual enantiomers or diastereomers are contemplated.
In certain embodiments, the compounds of the invention may exist as tautomers. “Tautomerization” is a form of isomerization involving the migration of a proton
accompanied by changes in bond order, often the interchange of a single bond with an adjacent double bond. Where tautomerization is possible (e.g., in solution), a chemical equilibrium of tautomers can be reached. One well known example of tautomerization is between a ketone and its corresponding enol. Heterocycles may form tautomers such as the interconversion of pyrrolidinone and hydroxy pyrrole. Ail tautomers are included within the scope of the compounds presented herein.
In certain embodiments, compounds described herein are prepared as prodrugs. A “prodrug” refers to an agent that is converted into the parent drug in vivo. In certain 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.
Compounds of the invention can in certain embodiments form acids or bases. In certain embodiments, the invention contemplates acid addition salts. In other embodiments, the invention contemplates base addition salts. In yet other embodiments, the invention contemplates pharmaceutically acceptable acid addition salts. In yet other embodiments, the invention contemplates pharmaceutically acceptable base addition salts. Pharmaceutically acceptable salts refer to salts of those bases or acids that are not toxic or otherwise biologically undesirable.
Suitable pharmaceutically acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric (including sulfate and h drogen sulfate), and phosphoric acids (including hydrogen phosphate and dihydrogen phosphate). Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, malonic, saccharin, fumaric, pyruvic, aspartic, glutamic, benzoic, anthramlic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfomc, ethanes ulfonic, benzenesulfonic, pantothenic, trifluoromethanesulfonic, 2- hydroxyethanesu!fonie, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, aigmic, b-hydroxybutyric, salicylic, galactaric and galacturonic acid.
Suitable pharmaceutically acceptable base addition salts of compounds of the invention include, for example, metallic salts including alkali metal, alkaline earth metal and transition metal salts such as, for example, calcium, magnesium, potassium, sodium, lithium and copper, iron and zinc salts. Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, N,N’-dibenzylethylene-diamine, chloroprocame, choline, dietha olamine, ethy!enediamine, meglumine (iV-methylglucamine) and procaine. All of these salts may be prepared from the corresponding compound by reacting, for example, the appropriate acid or base with the compound.
Methods of Treating Bacterial Respiratory Infection
t has now been unexpectedly discovered that treatment with the above described MIF agonists improves resistance to bacterial respiratory' infection in subjects exposed to cigarette smoke as shown m FIGS. iOA-IOB and 1 lA-1 IB. Accordingly, in one aspect the invention provides a method of treating a bacterial respirator}' infection in a subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist. In various embodiments, the bacterial respirator}' infection is a cigarette smoke- associated respirator}' infection. As used herein, the term cigarette smoke-associated respirator}' infection is a respiratory infection that is typically associated with or aggravated by exposure to cigarette smoke. In various embodiments, the bacterial respirator}' infection is Streptococcus pneumonia.
In another aspect, the invention provides a method of treating bacterial pneumonia m a subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist. In various embodiments, the subject has COPD.
A dministration/Dosing
In clinical settings, deliver}' systems for the compositions described herein can be introduced into a subject by any of a number of methods, each of which is familiar in the art. For instance, a pharmaceutical formulation of the composition can be administered by inhalation or systemically, e.g. by intravenous injection. 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 manifestation of symptoms associated with the disease or condition. 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 composition of the present invention to a subject, preferably a mammal, more preferably a human, may be carried out using known procedures, at dosages and for periods of time effective to treat a disease or condition in the subject. An effective amount of the composition necessary' to achieve a therapeutic effect may vary according to factors such as the time of administration; the duration of administration; other drugs, compounds or materials used in combination with the composition; the state of the disease or disorder; age, sex, weight, condition, general health and prior medical history of the subject being treated; and like factors well-known in the medical arts. 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. 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 composition without undue experimentation. Formulations may be employed in admixtures with conventional excipients, i.e., pharmaceutically acceptable organic or inorganic carrier substances suitable for oral, parenteral, nasal, intravenous, subcutaneous, enteral, or any other 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., other analgesic agents.
Routes of administration of any of the compositions of the invention include oral, nasal, 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, (trails jurethral, vaginal (e.g., trans- and perivagmally), (intrajnasai and (trans)rectal), intravesical, intrapul monaiy, intraduodenal, intragastricaJ, intrathecal, subcutaneous, intramuscular, iniraderma!, intra-arterial, intravenous, intrabronchial, inhalation, and topical administration.
Suitable compositions and dosage forms include, for example, tablets, capsules, caplets, pills, gel caps, troches, dispersions, suspensions, solutions, syrups, granules, beads, transdermal patches, gels, powders, pellets, magmas, lozenges, creams, pastes, plasters, lotions, discs, suppositories, liquid sprays for nasal or oral administration, dry powder or aerosolized formulations for inhalation, compositions and formulations for intravesical administration 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.
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 un coated 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.
For oral administration, the compounds of the invention may be in the form of tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e g. , polyvinylpyrrolidone, hydroxypropy!cel!u!ose or hydroxypropyl methylcellulose); fillers (e.g., cornstarch, lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrates (e.g., sodium starch gly collate); or wetting agents (e.g., sodium lauryl sulphate). If desired, the tablets may be coated using suitable methods and coating materials such as OPADRY™ film coating systems available from Coiorcon, West Point, Pa. (e.g., OPADRY™ OY Type, OYC Type, Organic Enteric OY-P Type, Aqueous Enteric OY-A Type, OY-PM Type and OPADRY™ White, 32K18400). Liquid preparation for oral administration may be in the form of solutions, syrups or suspensions. The liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g, sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agent (e.g , lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and
preservatives (e.g. methyl or propyl p-hydroxy benzoates or sorbic acid).
Parenteral Administration
For parenteral administration, the compounds of the invention may be formulated for injection or infusion, for example, intravenous, intramuscular or subcutaneous injection or infusion, or for administration in a bolus dose and/or continuous infusion. Suspensions, solutions or emulsions in an oily or aqueous vehicle, optionally containing other formulatory agents such as suspending, stabilizing and/or dispersing agents may be used.
Controlled Release Formulations arid 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 that 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 m the form of wafers or discs by implantation.
In certain embodiments, 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 drag formulation that provides for an initial release of the drag after some delay following drug administration and that mat, 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 drag 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.
Dosing
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 herein in the patient being treated. The skilled artisan is able to determine appropriate dosages depending on these and other factors.
A suitable dose of a compound of the present invention may he in the range of from about 0.001 rng to about 5,000 mg per day, such as from about 0 01 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 m 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 12-hour interval between doses.
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 maybe 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.
Actual dosage levels of the cells in the pharmaceutical formulations of this invention may be varied so as to obtain an amount of the compositi on that are effective to achieve the desired therapeutic response for a particular subject, composition, and mode of
administration, without being toxic to the subject.
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 LD50 (tire dose lethal to 5G% of the population) and the ED50 (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 LD50 and ED50 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 preferably within a range of circulating concentrations that include the ED50 with minimal toxicity. The dosage optionally varies within this range depending upon the dosage form employed and the route of administration utilized.
EXPERIMENTAL EXAMPLES
The invention is further described in detail by reference to the following experimental examples. These examples are provided for purposes of illustration only, and are not intended to be limiting unless otherwise specified. Thus, the invention should m no way be construed as being limited to the following examples, but rather, should be construed to encompass any and all variations which become evident as a result of the teaching provided herein.
Without further description, it is believed that one of ordinal}' skill in the art can, using the preceding description and the following illustrative examples, make and utilize the compounds of the present invention and practice the claimed methods. The following working examples therefore, specifically point out selected embodiments of the present invention, and are not to be construed as limiting in any way the remainder of the disclosure.
Animals
Animal protocols were reviewed and approved by the Animal Care and Use Committee at Yale University. Wild-type (WT) 8- to 10-wk-old C57BL/6J mice were purchased from Jackson Laboratories (Bar Harbor, ME, USA) and bred in facility. Miff , Ddi ' and Cd74 / mice were generated. The animals were housed in cages located in temperature- and humidity-controlled rooms with a 12 h light-dark cycle and received water and food ad libitum.
Cigarette smoke (CS) exposure
The mice were exposed in a Teague machine to CS from scientific reference cigarettes (3R4F, University of Kentucky , Lexington, KY) at 140 mg/m3 total suspended particulates for 3 months (6 h/day, 5 days/week).
MIF20 administration
Mice were orally administered vehicle (1% carboxymethyl cellulose in water) or 25, 50, 100 mg/kg MIF20 once a day during the last 1 month. PV loops
At the end of CS exposure, mice were anesthetized by urethane and paralyzed with a pancuronium bromide (intraperitoneal injection). After confirming complete paralysis, the lungs were ventilated with a Constant Flow ventilator using the FlexiVent software to measure PV loops. PV loops data were analyzed by the student /-test, and statistical differences between the groups were considered significant at p<0.05.
Lung volume
Lungs were removed from control and experimental mice and inflated slowly at a constant pressure of 25 cm. Lung volume was assessed by volume displacement.
Chord lengths
The right main bronchus was ligated and the left lung was inflated with 0.5% low- temperature melting agarose at a constant pressure of 25 cm as previously described. The lungs were then fixed, paraffin embedded, and stained with hematoxylin and eosin. A minimum of six random fields were evaluated by microscopic projection and the NIH image program; alveolar size was estimated from the mean linear intercept (I,m) of the air space as described previously.
Human model
HUVEC was treated with 0% or 2.5% CSE along with the following 1) recombinant M1F (rMIF; lOOng/ml), 2) MIF silencing RNA (siMIF), 3) neutralizing MIF antibody (MIF nAb). 4) MIF098 - a MIF antagonist, or 5) MIF20 - a MIF agonist. After 24 hours, gene expression and SA- -gal activity were determined. To determine the mechanism of MIF20, U0126, an Erk inhibitor, was treated 1 hour prior to MIF20 treatment. ABT263, a known senolytic agent, were used as positive control.
Mouse model
AKR/J mice were exposed to CS for 4 months, and MIF20 was orally administered (3 mg/mouse) to mice every day for the last 2 months of CS exposure. PV loop was measured by FlexiVent in CS-exposed AKR/J and MIF knockout mice (FIGS. 8A-8B).
Example 1:
Cohort study
A cohort of 72 adults without prior testing undement spirometry to confirm airflow obstruction (FIG. 2A). In an atempt to determine changes that occur with aging, the inclusion criteria were adults between the ages of 18-45 or older than 65 years of age.
Additionally, recruited persons were required to provide a smoking history and agreed to be contacted for the study. The exclusion criteria were 1) immunocompromised adults (HIV, malignancy diagnosis in past 5 years, systemic chemotherapy, end-stage renal failure); 2) adults with other obstructive lung disease (history of asthma, cystic fibrosis, bronchiectasis, bronchiolitis obliterans, or vocal cord dysfunction); 3) adults who could not give informed consent because of language, cognitive, or other barriers; and 4) adults who could not perform spirometry because of cognitive, health, or other reasons. Participants completed a questionnaire to assess sociodemographic and self-reported health status, including comorbid conditions and medications, and underwent phlebotomy to obtain plasma samples. COPD was defined by GOLD criteria, which are based on a forced expiratory volume in 1 s (FEVi) obtained by spirometry. Because many persons with COPD had quit smoking, participants with COPD include both current smokers, defined as having a greater than 10 pack-year lifetime history and have smoked in the past 11 months, or former smokers (quit for over 1 year but have a greater than 10 pack -year history). Covariates were age expressed in years, sex, smoking status, and race as self-identified white or self-identified as other than white. The subgroup of adults 65 years of age (an age at which COPD is common) was then subdivided into those who were current or former smokers and by the presence or absence of COPD to determine whether differences in plasma MIF were evident (FIG. 2A). In FIGS. 2B-2C, pubiically available gene expression profiles that were previously obtained using Affymetrix U133 2.0 microarrays performed on peripheral blood mononuclear cells obtained from patients in the COPDgene study (n=T36) were analyzed.
Example 2
CD74 expression in endothelial cells were increased in people with COPD. Activation of MIF signaling pathway attenuated CSE-induced pl6 and p21 expression as well as SA-b- gal activity. These protective effects were abolished by ERK inhibitor.
Example 3
MIF knockout mice showed increased PV loop and mean chord lengths, while MIF20 prevented PV loop in CS-exposed mice.
The disclosures of each and ever}' patent, patent application, and publication cited herein are hereby incorporated herein by reference in their entirety.
Wlule 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.

Claims

CLAIMS What is claimed is:
1. A method of treating a fibrotic disease in a male subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist.
2. The method according to claim 1, wherein the fibrotic disease is lung fibrosis.
3. A method of treating chronic obstructive pulmonary disease (COPD) in a female subject in need thereof, the method comprising administering to the female subject an effective amount of at least one MIF agonist.
4. The method according to claim 3, wherein the COPD is emphysema Ape COPD.
5. The method according to any one of claims 1-4, wherein the at least one MIF agonist is selected from the group consisting of:
O
2-(4-chlorophenyl)-6-fluorobenzo[d]isothiazol-3(2H)-one
Figure imgf000024_0001
o
2-(4-chlorophenyl)-5-fluorobenzo[d]isothiazol-3(2H)-one
Figure imgf000024_0002
2-(4-chlorophenyl)-3-oxo-2,3-dihydrobenzo[d]isothiazole-5-carboni trite
Figure imgf000024_0003
5-fluoro-2-(4-rnethoxyphenyl)benzo[d]isothiazol-3(2H)-one
Figure imgf000024_0004
5-fluoro-2-(3-methoxyphenyl)benzo[d]isothiazol-3(2H)-one
Figure imgf000024_0005
O
2-(4-chlorophenyl)-5,6-difluorobenzo[d]isothiazol-3(2H)-one
Figure imgf000025_0001
7-fluoro-2-(3-methoxyphenyl)benzo[d]isothiazol-3(2H)-one
Figure imgf000025_0002
6-fluoro-3-(3-hydroxybenzyl)benzo| d]oxazol-2(3H)-one
Figure imgf000025_0003
5-f]uoro-3-(3-methoxybenzyl)benzo[d]oxazol-2(3H)-one
Figure imgf000025_0004
Figure imgf000025_0005
4-(4-(pyridin-3-yl)- 1H- 1 ,2,3-triazol-i -yl)phenol
Figure imgf000025_0006
4-(4-(isoquinolin-4-yl)-lH-l,2,3-triazol-l-yl)phenol
Figure imgf000025_0007
or a salt or solvate thereof, and any mixtures thereof.
6. The method according to claim 5, wherein the M1F agonist is 4-(4-(pyridin-3-yl)-lH- l,2,3-triazol-l-yi)phenol, or a salt or solvate thereof.
7. The method according to any one of claims 1-6, wherein the at least one agonist is formulated in a composition further comprising at least one pharmaceutically acceptable excipient.
8. The method according to any one of claims 1-7, wherein the at least one agonist is administered by at least one route selected from the group consisting of inhalational, oral, rectal, vaginal, parenteral, intracranial, topical, transdermal, pulmonary, mtranasal, buccal, ophthalmic, intrathecal, and intravenous.
9. The method according to any one of claims 1-8, wherein the subject is a mammal.
10. The method according to any one of claims 1-9, wherein the subject is a human.
11. A method of treating a bacterial respiratory infection in a subject m need thereof, the method comprising administerin to the male subject an effective amount of at least one MIF agonist.
12. The method according to claim 11, wherein the bacterial respiratory infection is a cigarette smoke-associated respirator}' infection.
13. The method according to claim 11, wherein the bacterial respiratory infection is Streptococcus pneumonia.
14. A method of treating bacterial pneumonia in a subject in need thereof, the method comprising administering to the male subject an effective amount of at least one MIF agonist.
15. The method according to claim 14, wherein the subject has CQPD.
16. The method according to any one of claims 1 1-15, wherein the at least one MIF agonist is selected from the group consisting of:
O
2-(4-chlorophenyl)-6-fluorobenzo[d]isothiazol-3(2H)-one
Figure imgf000026_0001
-(4-chlorophenyl)-5-fluorobenzo[d]isothiazol-3(2H)-one
Figure imgf000027_0001
-(4-chlorophenyl)-3-oxo-2,3-dihydrobenzo[d|isothiazole-5-carbonitrile
Figure imgf000027_0002
-f]uoro~2-(4-meihoxyphenyr)benzofd]isothiazol~3(2H)~one
Figure imgf000027_0003
-fluoro-2-(3-methoxyphenyl)benzo[d]isotbiazol-3(2H)-one
Figure imgf000027_0004
·
O -(4-chlorophenyl)-5,6-difluorobenzo[d]isothiazol-3(2H)-one
Figure imgf000027_0005
-fluoro-2-(3-methoxyphenyl)benzo[d]isothiazol-3(2H)-one
Figure imgf000027_0006
-fluoro-3-(3-hydroxybenzyl)benzo| d]oxazol-2(3H)-one
Figure imgf000027_0007
-fluoro-3-(3-methoxybenzyl)benzo[d]oxazol-2(3H)-one
Figure imgf000027_0008
-phenylbenzoj d]thi azole
Figure imgf000027_0009
-methy 1 -2-pheny Ibenzo [ d j thi azole
Figure imgf000027_0010
4-(l -(2-methoxy benzyl)-! H-l ,2,3-triazol-4-y l)phenol
Figure imgf000028_0001
4-(4-(pyridin-3-yl)-lH-l,2,3-triazol-l -yl)phenol
Figure imgf000028_0002
4-(4-(isoquinolin-4-yl)-lH-l,2,3-triazol-l-yl)phenol
Figure imgf000028_0003
or a salt or solvate thereof, and any mixtures thereof.
17. The method according to claim 16, wherein the MIF agonist is 4-(4-(pyridin-3-yl)- lH-l,2,3-triazol-l-yl)phenol, or a salt or solvate thereof.
18. The method according to any one of claims 11-17, wherein the at least one agonist is formulated in a composition further comprising at least one pharmaceutically acceptable excipient.
19. The method according to any one of claims 11-18, wherein the at least one agonist is administered by at least one route selected from the group consisting of inhaiationai, oral, rectal, vaginal, parenteral, intracranial, topical, transdermal, pulmonary, intranasal, buccal, ophthalmic, intrathecal, and intravenous.
20. The method according to any one of claims 11-19, wherein the subject is a mammal.
21. The method according to any one of claims 11-20, wherein the subject is a human.
PCT/US2019/065652 2018-12-11 2019-12-11 Compounds and methods for treating copd and/or lung fibrosis WO2020123610A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862778187P 2018-12-11 2018-12-11
US62/778,187 2018-12-11
US201962910164P 2019-10-03 2019-10-03
US62/910,164 2019-10-03

Publications (1)

Publication Number Publication Date
WO2020123610A1 true WO2020123610A1 (en) 2020-06-18

Family

ID=71076676

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/065652 WO2020123610A1 (en) 2018-12-11 2019-12-11 Compounds and methods for treating copd and/or lung fibrosis

Country Status (1)

Country Link
WO (1) WO2020123610A1 (en)

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JORGENSEN, W.L. ; GANDAVADI, S. ; DU, X. ; HARE, A.A. ; TROFIMOV, A. ; LENG, L. ; BUCALA, R.: "Receptor agonists of macrophage migration inhibitory factor", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, AMSTERDAM, NL, vol. 20, no. 23, 1 December 2010 (2010-12-01), AMSTERDAM, NL, pages 7033 - 7036, XP027459351, ISSN: 0960-894X, DOI: 10.1016/j.bmcl.2010.09.118 *
LEE PATTY J: "MIF-Based Therapies in Cigarette Smoke-Related COPD and Pneumonia", pages 1 - 23, XP055720395, Retrieved from the Internet <URL:https://apps.dtic.mil/dtic/tr/fulltext/u2/1074300.pdf> *
PAUL A. BEAVIS, UPULIE DIVISEKERA, CHRISTOPHE PAGET, MELVYN T. CHOW, LIZA B. JOHN, CHRISTEL DEVAUD, KAREN DWYER, JOHN STAGG, MARK : "Blockade of A 2A receptors potently suppresses the metastasis of CD73 + tumors", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, vol. 110, no. 36, 3 September 2013 (2013-09-03), pages 14711 - 14716, XP055459265, ISSN: 0027-8424, DOI: 10.1073/pnas.1308209110 *

Similar Documents

Publication Publication Date Title
Corcoran et al. Pharmacological inhibition of the nod-like receptor family pyrin domain containing 3 inflammasome with MCC950
KR102034703B1 (en) Drug for treatment of nonalcoholic fatty liver disease
US10086001B2 (en) Composition for the treatment of cystic fibrosis
Ito et al. Steroid-resistant neutrophilic inflammation in a mouse model of an acute exacerbation of asthma
US20060194756A1 (en) Enhancing treatment of HIF-1 mediated disorders with adenosine A3 receptor agonists
JP2008520746A (en) Accelerated treatment of cancer and HIF-1 mediated diseases using an adenosine A3 receptor antagonist
US20210008020A1 (en) Methods of Treating or Preventing Acute Respiratory Distress Syndrome
US11660281B2 (en) Compositions and methods of treating or preventing fibrotic lung diseases
WO2018131626A1 (en) Prophylactic and therapeutic drug for nonalcoholic fatty liver disease
US11524056B2 (en) Thymosin alpha 1 for use in treatment of cystic fibrosis
US7417072B2 (en) Blockade of Pin1 prevents cytokine production by activated immune cells
US20120046333A1 (en) Methods and Compositions of PI-3 Kinase Inhibitors for Treating Fibrosis
ELDesoky et al. Variability in response to cardiovascular drugs
WO2020123610A1 (en) Compounds and methods for treating copd and/or lung fibrosis
Hsiao et al. Inhibitory mechanisms of YC-1 and PMC in the induction of iNOS expression by lipoteichoic acid in RAW 264.7 macrophages
US11253506B2 (en) Compounds, compositions and methods for preventing and/or treating chlorine inhalation toxicity and/or injury
EP3355909A1 (en) Methods for treating diseases mediated by erbb4-positive pro-inflammatory macrophages
Blot MRSA pneumonia: better outcome through continuous infusion of vancomycin?
EP3781166A1 (en) Pharmaceutical composition, methods for treating and uses thereof
CN106714842B (en) Methods of treatment and compositions comprising dual PI3K delta-gamma kinase inhibitors and corticosteroids
EP4374868A1 (en) Pharmaceutical composition or drug kit for alleviating or treating fibrotic diseases, and use thereof
EP4008325A1 (en) Cxcl8 inhibitors for use in the treatment of covid-19
JP7233087B2 (en) antiarteriosclerotic
EP3868368A1 (en) Cxcl8 (interleukin-8) activity inhibitor and corticosteroid combination and pharmaceutical composition and use thereof
EP4032534A1 (en) Lonafarnib for use in the treatment of viral infections

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19896655

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19896655

Country of ref document: EP

Kind code of ref document: A1