Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SMALL MOLECULE INHIBITORS OF PENDRIN ION EXCHANGE AND PHARMACEUTICAL COMPOSITIONS
Document Type and Number:
WIPO Patent Application WO/2017/147523
Kind Code:
A1
Abstract:
Provided herein are small molecule inhibitors of pendrin. More specifically, the small molecules disclosed herein may be used to inhibit pendrin-mediated anion exchange. These small molecules may be used for treatment of diseases and disorders that are treatable by inhibiting pendrin, such as cystic fibrosis, rhinitis, chronic rhinosinusitis, exposure to an industrial toxin, certain viral infections, certain bacterial infections, and chronic obstructive pulmonary disease. The small molecules may further be used to potentiate a diuretic effect of a diuretic compound. The small molecules may additionally be used to treat hypertension or thyroid conditions.

Inventors:
VERKMAN ALAN S (US)
HAGGIE PETER (US)
CIL ONUR (US)
Application Number:
PCT/US2017/019506
Publication Date:
August 31, 2017
Filing Date:
February 24, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV CALIFORNIA (US)
VERKMAN ALAN S (US)
International Classes:
A61K31/18; A61K31/4162; A61K31/4353
Foreign References:
EP2370437B12013-09-04
US20100331295A12010-12-30
US20090156611A12009-06-18
Other References:
DATABASE PUBCHEM 21 June 2011 (2011-06-21), XP055412960, Database accession no. 010-708-793
DATABASE PUBCHEM 21 June 2011 (2011-06-21), XP055412962, Database accession no. 010-708-795
DATABASE PUBCHEM 21 June 2011 (2011-06-21), XP055412963, Database accession no. 010-708-724
DATABASE PUBCHEM 21 June 2010 (2010-06-21), XP055412965, Database accession no. 45492877
DATABASE PUBCHEM 22 February 2011 (2011-02-22), XP055412967, Database accession no. 010-662-536
GENNARO: "Remington: The Science and Practice of Pharmacy", 2005, MACK PUB. CO.
See also references of EP 3419612A4
Attorney, Agent or Firm:
HAN, Hai et al. (US)
Download PDF:
Claims:
CLAIMS

1. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of Formula (I):

(D

wherein,

n is 0, 1, 2, or 3;

R1 at each occurrence is the same or different and independently optionally substituted alkyl, optionally substituted alkoxy, halo, trifluoroalkyl, or optionally substituted thioalky], and

R2 is optionally substituted aryl or optionally substituted aralkyl.

2. The pharmaceutical composition of claim 1 wherein each Rx is independently C C4 alkyl, C1 -C4 alkoxy, Cj-C4 thioalkvl, fluoro, trifluoroalkyl, or chloro.

3. The pharmaceutical composition of claim 2 wherein each R1 is independently methyl, ethyl, methoxy, thiomethyl, trifluoromethyl, fluoro or chloro.

4. The pharmaceutical composition of any one of claims 1-3, wherein R1 is not at the ortho-position.

5. The pharmaceutical composition of any one of claims 1 -4, wherein R2 is phenyl or substituted phenyl.

6. The pharmaceutical composition of claim 5, wherein R" is phenyl substituted with one or more fluoro.

7. The pharmaceutical composition of any one of claims 1 -4, wherein R2 is benzyl or substituted benzyl.

8. The pharmaceutical composition of claim 7, wherein R2 is benzyl substituted with methyl, fluoro, or a combination thereof.

9. The pharmaceutical composition of claim 1, wherein compound is represented by Formula (la):

wherein,

Rla and Rlb are the same or different and independently hydrogen, optionally substituted alkyl, optionally substituted alkoxy, halo, tnfluoroalkyl, or optionally substituted thioalkyl;

R3 is hydrogen or halo.

10. The pharmaceutical composition of claim 9 wherein R1" and Rl are the same or different and independently methyl, ethyl, methoxy, thiom ethyl, trifluoromethyl, fluoro, or chloro.

11. The pharmaceutical composition of claim 10 wherein Rla is hydrogen, Rlb is methyl, ethyl, methoxy, thiomethyi, or chloro.

12. The pharmaceutical composition of claim 10 wherein R is hydrogen, RIa is methyl, ethyl, methoxy, tnfluoromethyl, thiomethyl, fluoro, or chloro.

13. The pharmaceutical composition of any one of claims 9-12 wherein R3 is hydrogen or fluoro.

14. The pharmaceutical composition of any one of claims 9-12 comprising at last one compound of Table 1.

15. The pharmaceutical composition of claim 1, wherein

compound i s represented by Formula (lb):

wherein,

m is 0, 1 or 2;

Rla and Rlb are the same or different and independently hydrogen, optionally substituted alkyl, optionally substituted alkoxy, halo, trifluoroalkyl, or optionally substituted thioalkyl; and

each R4 is the same or different and independently optionally substituted alkyl or halo.

16. The pharmaceutical composition of claim 15 wherein Rla and Rlb are the same or different and independently methyl, ethyl, methoxy, thiomethyl, fluoro or chloro.

17. The pharmaceutical composition of claim 16 wherein R a is hydrogen, R is methyl, ethyl, methoxy, thiomethyl, or chloro,

18. The pharmaceutical composition of claim 16 wherein Rl is hydrogen, Rla is methyl, ethyl, methoxy, trifluoromethyi, thiomethyl, fluoro, or chloro.

19. The pharmaceutical composition of any one of claims 15-18 wherein each R4 is hydrogen or fluoro.

20. The pharmaceutical composition of any one of claims 15-19 comprising at last one compound of Table 2.

21. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and a pyrazolothiophenesulfonamide compound represented by Formula (II):

wherein,

R5a and R5 are the same or different and independently hydrogen, or optionally substituted alkyl;

R6 and R' are the same or different and independently hydrogen, optionally substituted alkyl, or -S(0)2TMHR, provided one of R6 and R7 is -S(0)2NHR; and

R is optionally substituted aryl or optionally substituted cycloalkyl.

22. The pharmaceutical composition of claim 21 wherein R~ d and R" are the same or different and independently hydrogen or methyl.

23. The pharmaceutical composition of any one of claims 21-22 wherein R6 is -S(0)2 HR, and R' is hydrogen or methyl.

24. The pharmaceutical composition of any one of claims 21 -22, wherein R'' is -S(0)2 HR, and R° is hydrogen or methyl.

25. The pharmaceutical composition of any one of claims 21-24 wherein R is optionally substituted aryi or optionally substituted cycloalkyl.

26. The pharmaceutical composition of any one of claims 21-25 wherein R is tetrahydronaphthalenyl.

27. The pharmaceutical composition of any one of claims 21-25 wherein R is phenyl optionally substituted by one or more alkyl, cyclohexyl or cyclopentyl.

28. The pharmaceutical composition of any one of claims 21-27 comprising at last one compound of Table 3.

29. A method of treating an airway disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the pharmaceutical composition of any one of claims 1 -28,

30. The method of claims 29 wherein the airway diseases is an inflammatory lung disease, rhinovirus infection, rhinitis, chronic rhinosinusitis, sinusitis, certain viral or bacterial infections, or caused by exposure to industrial toxins.

31. A method of treating cystic fibrosis in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the pharmaceutical composition of any one of claims 1-28.

32. A method for treating a disease or condition treatable by inhibiting pendrin, partially or completely, in a subject, the method comprising administering a pharmaceutical composition comprising a pharmaceutically acceptable excipient and the pharmaceutical composition of any one of claims 1-22,

33. A method of promoting diuresis to a subject in need thereof comprising:

administering to the subject a diuretic; and

administering to the subject a therapeutically effective amount of the pharmaceutical composition of any one of claims 1-28, wherein the pharmaceutical composition is administered prior to, contemporaneous with, or following

administration of the diuretic to the subject.

34. The method of claim 33 wherein the diuretic is a thiazide compound selected from the group consisting of furosemide, azosemide, bumelanide, piretanide, and torasemide.

35. The method of claim 33 wherein the diuretic is furosemide and the pharmaceutical composition com rises:

36. A method for treating hypertension or edema in a subject in need thereof comprising:

administering to the subject a diuretic, and

administering to the subject a therapeutically effective amount of the pharmaceutical composition of any one of claims 1-28, wherein the pharmaceutical composition is administered prior to, contemporaneous with, or following

administration of the diuretic to the subject.

37. Use of the pharmaceutical composition of any one of claims 1 -28 for airway disease therapy.

38. Use of the pharmaceutical composition of any one of claims 1-28 for combination therapy with a diuretic.

39. A method of treating hypertension comprising administering to a subject in need thereof a therapeutically effective amount of the pharmaceutical composition of any one of claims 1-28.

40. The method of claim 39, wherein the hypertension is salt- sensitive hypertension.

41. A method of treating a thyroid condition comprising administering to a subject in need thereof a therapeutically effective amount of the pharmaceutical composition of any one of claims 1-28.

42. A pharmaceutical composition of any one of claims 1-28 for use in therapy, wherein therapy includes treating hypertension, a thyroid condition, an airway disease, cystic fibrosis, or edema.

Description:
SMALL MOLECULE INHIBITORS OF PENDRIN ION EXCHANGE AND PHARMACEUTICAL COMPOSITIONS

CROSS REFERENCE TO RELATED APPLICATION

This application claims the benefit under 35 U.S.C. § 119(e) to U.S. Provisional Application 62/300,41 J filed February 26, 2016, which application is incorporated by reference herein in its entirety.

STATEMENT OF GOVERNMENT INTEREST

This invention was made with government support under NTH Grant Nos. P30 DK072517, DK101373, DK35124, DK99803, R37 EB00415, EY13574, All 11634, and P30CA082103. The government may have certain rights in this invention.

BACKGROUND Technical Field

This disclosure relates to pharmaceutical composition of small molecule pendrin inhibitors and methods for treating diseases and disorders related to aberrant ion exchange activity, such as cystic fibrosis, salt-sensitive hypertension, edema, cirrhosis, and certain renal disorders.

Description of the Related Art

Aberrant anion exchange is implicated in a variety of human diseases and disorders. For example, solute imbalances in plasma or urine may indicate renal, cardiac, or hepatic issues. In cystic fibrosis (CF), defective CI " and HCO 3 " transport alters the volume and composition of airway surface liquid (ASL), which may lead to a variety of complications such as impaired immune function and CD disease pathogenesis. Thus, cellular mechanisms involved in anion exchange are of interest as potential therapeutic targets. Pendrin, encoded by the SLC26A4 (solute carrier family 26, member 4) gene, is an anion exchanger expressed in a variety of tissues, including the epithelium of inflamed airways, the inner ear, the thyroid, the adrenal gland, and the kidneys.

Pendrin is implicated in airways diseases. Pendrin expression is upregulated in a variety of airway disease contexts, such as, for example: airway epithelial cultures that are chronically exposed to cytokines such as IL-13, IL-4, and IL- 17 A; rodent models of inflammatory lung disease, including allergen (ovalbumin, OVA)-induced asthma, chronic obstructive pulmonary disease (COPD), infection, and industrial toxin exposure, and, humans with rhinovirus infection, asthma, CF, rhinitis, and chronic rhinosinusitis. The best previously described pendrin inhibitor, niflumic acid, produces <40% inhibition at 100 μΜ and has poor specificity. Drug candidates capable of effecting anion exchangers such as pendrin are needed.

BRIEF SUMMARY

Provided herein are pharmaceutical compositions comprising

compounds that are potent inhibitors of pendrin activities. Also disclosed are methods for treating diseases, disorders, and conditions that are treatable by inhibiting pendrin- mediated anion exchange by administrating the pharmaceutical composition provided herein. The pharmaceutical compositions of the present disclosure may also be used in conjunction with a diuretic, such as a loop diuretic, to potentiate diuresis. The following embodiments are provided herein.

Some embodiments provide for pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a compound of of Formula (I):

wherein n is 0, 1 , 2, or 3; R 1 at each occurrence is the same or different and independently optionally substituted alkyl, optionally substituted alkoxv, halo, trifluoroalkyl, or optionally substituted thioaikyi; and R 2 is optionally substituted aryl or optionally substituted aralkyl .

In a more specific embodiment, the compound is represented by Formula

(la):

(la)

wherein,

R la and R lb are the same or different and independently hydrogen, optionally substituted alkyl, optionally substituted alkoxv, halo, trifluoroalkyl, or optionally substituted thioaikyi;

R 3 is hydrogen or halo.

In another more specific embodiment, the compound is represented by

Formula (lb):

wherein,

m is 0, 1 or 2;

R la and R lb are the same or different and independently hydrogen, optionally substituted alkyl, optionally substituted alkoxv, halo, trifluoroalkyl, or optionally substituted thioaikyi, and each R 4 is the same or different and independently optionally substituted aikyi or halo.

Other embodiments provide for pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a pyrazolothiophenesulfonamide compound represented by Formula i

wherein R 3a and R 5 are the same or different and independently hydrogen, or optionally substituted alkyl; R 6 and R are the same or different and independently hydrogen, optionally substituted alkyl, or -S(0) 2 HR, provided one ." and R 7 is ---S(C)) 2 HR; and

R is optionally substituted aryl or optionally substituted cycloalkyl.

Other embodiments provide for methods of treating an airway disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of one or more of the pharmaceutical compositions described above.

Still other embodiments provide for methods of treating cystic fibrosis in a subject in need thereof comprising administering to the subject a therapeutically effective amount of one or more of the pharmaceutical compositions described above.

Other embodiments provide for methods of treating a disease or condition treatable by inhibiting pendrin, partially or completely, in a subject in need thereof comprising administering to the subject a therapeutically effective amount of one or more of the pharmaceutical compositions described above.

Further embodiments provide for methods for promoting diuresis to a subject in need thereof comprising: administering to the subject a diuretic; and administering to the subject a therapeutically effective amount of of one or more of the pharmaceutical compositions described above, wherein the one or more pharmaceutical composition is adiminstered prior to, contemporaneous with, or following

administration of the diuretic to the subject.

Other embodiments provide methods for treating hypertension or edema in a subject in need thereof comprising: administering to the subject a diuretic; and administering to the subject a therapeutically effective amount of of one or more of the pharmaceutical compositions described above, wherein the one or more pharmaceutical composition is adiminstered prior to, contemporaneous with, or following

administration of the diuretic to the subject.

Further embodiments provide for use of one or more of the pharmaceutical compositions described above for airway disease therapy. Other embodiments provide for use of one or more of the pharmaceutical compositions described above for combination therapy with a diuretic.

Other embodiments provide for methods of of method of treating hypertension comprising administering to a subject in need thereof a therapeutically effective amount of the one or more of the pharmaceutical compositions described above.

Still other embodiments provide for methods of treating a thyroid condition comprising administering to a subject in need thereof a therapeutically effective amount one ormore of the pharmaceutical compositions described above. BRIEF DESCRIPTION OF THE DRAWINGS

Figures 1 A-1D illustrate pendrin-inhibiting compounds identified by high-throughput screening and summarize experimental approaches used to characterize the inhibitory activity. Figure 1A shows a schematic of assay showing extracellular addition of SCN " results in pendrin-mediated C17SCN " exchange and YFP quenching. Figure IB shows representative time course of YFP fluorescence quenching in cells expressing YFP alone (non-transfected, top) and YFP with human pendrin, showing curves for active and inactive compounds (and no test compound "control"). Figure IC shows the chemical structures of pendrin inhibitors identified from the screen. Figure I D shows the stmctural determinants of activity of compounds of Formula (I) (left) and compounds of Formula (II) (right).

Figures 2A-2D present data illustrating kinetics of PDSjn h -AOl and

PDSm h -COl inhibiting pendrin-mediated anion exchange. Figure 2A shows original fluorescence quenching curves for inhibition of pendrin-mediated C17SCN " (left) and C17T (right) exchange by PDSm h -AOl (top) and PDSm h -COl (bottom). Data shown in Figure 2B illustrates concentration-dependent inhibition of pendrin-mediated C17SCN ' , and C17F and C17N0 3 ' exchange by PDSm h -AOl (top) and PDSm h -COl (bottom). IC 5 o values for C17SCN * exchange for both compounds were ~9 μΜ. PDSm h -AOl and PDSm h -CO l inhibited C17F and C17N0 3 " exchange with IC 50 of -8 μΜ and ~5 μΜ, respectively. Figure 2C shows inhibition kinetics of pendrin-mediated C17SCN " exchange in which 10 μΜ PDSinh-AOl was added for indicated times (left) and summary (right) for PDSin h -AOl and PDSm h -COl . Figure 2D shows data from washout experiments illustrating that inhibition of pendrin by PDSm h -AOl and PDSm h -COl is reversible. All data shown as mean ± SEM (n : = 4).

Figures 3A-3D illustrate pendrin inhibition by PDSin h -AO l and PDSjn h - C01 and resultant effects on cytoplasmic pH. Figure 3A depicts a schematic assay showing replacement of extracellular CI " for gluconate in BCECF-ioaded FRT cells expressing human pendrin causes CI " efflux and HC0 3 " influx, resulting in cytoplasmic aikalinization. Figure 3B presents representative BCECF fluorescence data showing inhibition of C17HC0 3 " exchange by PDSm h -AOl and PDSm h -COl . Figure 3C.

Concentration-dependent inhibition of pendrin-mediated C17HC0 3 " exchange by PDSm h -AOl and PDS inh -C01 (mean ± SEM, n = 3-9 cultures). Figure 3D shows the control study done in non-transfected and pendrin expressing cells in the absence of HCOj " . BCECF ratio imaging indicated an initial cytoplasmic pH of -7.3 in all cultures studied.

Figures 4A-4B illustrate PDSin h -AOl selectivity for pendrin. Figure 4A shows short-circuit current (I sc ) in IL-13-treated human bronchial epithelial (HBE) cells in response to agonists and inhibitors that target key ion transport processes: 20 μΜ amiloride; 20 μΜ forskolin; 10 μΜ CFT inh-172; 100 mM ATP. Experiments were done in the absence (left, top) and presence (left, bottom) of PDS lall -A01 (25 μΜ, 30 minute pretreatment). (right) Summary of ,ul sc data (mean ± SEM, n = 3, differences not significant). Figure 4B shows YFP quenching assay indicating effects of PDSin - A01 (25 μΜ, 30 min does not inhibit SCN " transport by human SLC26A3 as assayed by YFP fluorescence responses after application of a 70 mM SCN " gradient. Differences not significant on SCN- transport by human SLC26A3 (the human protein most similar to pendrin in sequence; -50% identity) after application of a 70 mM SCN " gradient. Fluorescence responses (left) and data summary (right) are shown (mean ± SEM, n = 17-25 cells, differences not significant).

Figures 5 A-5C present data indicating the effects of PDS -. h -AOl on CI "

/HC0 3 ~ exchange and ASL pH in untreated and IL-13-treated primary HBE and CFBE cell cultures. Figure 5 A shows BCECF fluorescence in HBE and CFBE cells in response to extracellular CI ' replacement by gluconate in control (left) and IE- 13 (right) -treated cells, with PDSm -A01 (25 μΜ, 30 min) as indicated. Figure 5B presents a data summary (mean ± SEM, n = 3-4 cultures; * p < 0.05 comparing without vs. with PDSmh-AOl). Figure 5C. PDSM L -AOI (25 μΜ, 4 hours) does not alter ASL pH, with data shown for HBE and CFBE cells under control and IL-13-treated conditions (mean ± SEM, n = 3-7 cultures; * p < 0.005; ** p < 0.05). BCECF ratio imaging indicated an initial cytoplasmic pH of -7.3 in all cultures studied.

Figures 6A-6B present data demonstrating that IL-13 increases ASL depth in both primary HBE and CFBE ceil cultures, and that pendrin inhibition significantly increases ASL depth in IL-13 -treated cultures. Figure 6A shows reconstructions of confocal z-stacks showing rhodamine dextran-iabeled ASL in HBE and CFBE cells, without and with IL-13 -treatment. PBS^-AOl was used at 25 μΜ, Figure 6B shows a summary of ASL depth (mean ± SEM, n = 6-12 cultures; * p < 0.05 compared to HBE cultures (no IL-13); ** p < 0.05 compared to IL-13-treated cultures; IL-13 increased A SL depth for all conditions compared to no IL-13 -treatment.

Figures 7A-7F further characterize a pendrin-inhibiting compound of the present disclosure and show representative data from pendrin-inhibition experiments in FRT cells. Figure 7A. PDSinh-COl is comprised of a thiophene with a sulfonamide group and a pyrazole heterocycle linked at the 3- and 5-positions, respectively.

Structure-activity studies showed that changing the pyrazole from 5- to 4-position abolished activity and that a sulfonamide group at the 2- or 3-position was needed for inhibition activity. Limited substituents on the pyrazole were studied, with 3', 4'- dimethyl giving the most potent compounds followed by 3'-methyl and trifluoromethyi. Substitution on the sulfonamide affected activity, with electron-neutral rings such as tetrahydro-naphthalene and 2-ethylphenyl giving best activity, whereas halide- substituted phenyl ring reduced activity. Figure 7B. Functional studies of pendrin- mediated CI " exchange for Γ, SCN " and N()< " were done in FRT cells stably expressing murine pendrin and a YFP halide-sensing fluorescent indicator, (top) Schematic representation of fluorescence quenching assay for pendrin-mediated ion exchange in the presence of a pendrin inhibitor, (bottom) Fluorescence quenching data indicating ion exchange activity at various PDSinh-COl concentrations. Figure 7C. Summary of pendrin inhibition by PDSinh-COl . Figure 7D. Pendrin-mediated GVHCCV exchange was measured by the kinetics of intracellular H, using BCEC fluorescence as a cytoplasmic pH sensor, following extracellular addition of HC0 3 7C0 2 -containing buffer to drive CI " efflux, HC0 3 " influx, and consequent cytoplasmic alkalinization. Figure 7E. Concentration-dependent effects of PDSinh-COl on the kinetics of Cr/HCCV exchange, the activity of pendrin with relevance to kidney function, were measured in a concentration-dependent manner with IC50 -1.2 μΜ, Figure 7F. Selectivity data illustrating insignificant inhibition by PDSinh-COl of solute carrier proteins Sic4a (AE1), Slc26a3 (CLD/DRA) or N CC l (Slcl2a2).

Figures 8A-8D present PDSinh-COl pharmacokinetics data in mouse urine and plasma. These data were used to guide studies of diuretic efficacy. Figures 8A and 8C show original LC/MS data and linear standard curves in plasma and urine in which known amounts of PDSinh-COl were added to plasma and urine from untreated mice. Figures 8B and 8D summarize PDSinh-COl concentrations in plasma and urine following bolus interaperitoneal (ip) administration of 10 mg/kg PDSmh-COl, showing predicted therapeutic concentrations for several hours. Figures 9A-9D illustrate that pendrin inhibition alone by PDSin h -COl does not affect renal function. PDSin h -COl was administered to mice by ip injection as done in the pharmacokinetics measurements. Figure 9 A shows similar 3-h urine volume and osmolality in two different strains of mice treated with vehicle or PDSm h - C01, even at a very high dose of 50 mg/kg. PDSM I -CO I administration did not significantly change urine pH (Figure 9B) or blood gas values (Figure 9C), nor did it affect 3-h urinary salt excretion (Fig. 9D).

Figures 10A- 10E illustrate that pendrin inhibition potentiates the diuretic action of furosemide in an acute treatment model. Figure I OA. PDSinh-COl (10 mg/kg) significantly increased urine volume by -30% at each dose of furosemide tested, without effect on urine osmolality. The diuretic effect was significantly greater than that produced by maximal furosemide (50 mg/kg). Increasing PDS lnh -C01 dose to 50 mg/kg did not further potentiate the furosemide effect. PDSin h -COl, when given with 20 mg/kg furosemide, did not affect urine pH (Figure 10B), but produced a compensated metabolic alkalosis (Figure OC). To rule out an inhibitory effect of furosemide on pendrin activity that could confound the physiological data, in vitro measurements showed no effect of furosemide on pendrin activity (Figure 10E).

Figures 1 1 A- l 1C further illustrate that pendrin inhibition potentiates the diuretic action of furosemide in a chronical treatment model. Because chronic loop diuretic treatment upregulates renal pendrin expression, which might potentiate the diuretic efficacy of pendrin inhibition, the action of PDSin h -COl in a chronic furosemide treatment model was studied. After 8 days of furosemide treatment, PDSin -COl further potentiated the furosemide effect. Figure 1 IB shows a -60% increase in urine volume following PDSm h -COl in the chronically furosemi de-treated mice, without effect on urine osmolality. Figure 1 1C shows that PDSin h -COl significantly increased urinary Na + and K + excretion.

DETAILED DESCRIPTION

Small-molecule inhibitors of pendrin are described herein, as are pharmaceutical compositions compri sing the same. Various embodiments are directed to methods for treating a variety of airway diseases or conditions, such as, for example, CF, rhinovirus infection, asthma, rhinitis, chronic rhinosinusitis, certain viral or bacterial infections, and exposure to industrial toxins. Other embodiments are directed to methods of potentiating diuresis in a subject in need thereof, Pendrin Activities

Pendrin consists of 780 amino acids with a predicted cytoplasmic amino terminus and twelve membrane-spanning regions. Pendrin was identified by positional cloning in subjects with Pendred syndrome, an autosomal recessive disorder associated with congenital hearing loss and thyroid goiter.

Functional studies show that pendrin mediates electroneutral exchange of CI " with various anions, including Γ, HC0 3 " , OH " , N0 3 " , SCN " (thiocyanate) and HCO ? . " (formate). In the inner ear, pendrin is expressed in the apical membrane of epithelial cells in the sensor}' (saccule, utricle, and ampullae) and non-sensory

(endolymphatic sac) domains, and is implicated in determining inner ear luminal fluid volume and composition. In the thyroid, pendrin is expressed on the apical membrane of follicular cells, where it facilitates Γ efflux into the follicule colloid for thyroid hormone synthesis. In the kidney, pendrin is expressed in the apical plasma membrane of type B and non-A/non-B intercalated cel ls in the aldosterone-sensitive distal nephron, where it facilitates CI " absorption and HCO 3 " secretion, in addition to being involved in Na + absorption and acid-base balance.

Pendrin Inhibitors and Pharmaceutical Compositions

Various embodiments are directed to smal l molecules that selectively inhibit pendrin activities (e.g., pendrin-mediated anion exchange) at low micromolar potency. These compounds were identified by a functional, cell-based screen of a small molecule library and were found to produce strong and sustained impairment of pendrin-mediated ion exchange activity when pendrin showed elevated expression. Thus, pharmaceutical compositions comprising these pendrin inhibitors are suitable for treating diseases and disorders (including pulmonary and renal diseases) treatable by inhibiting pendrin. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a tetrahydropyrazolopyridine compounds represented by Formula (I):

wherein,

n is O, 1, 2, or 3;

R 1 at each occurrence is the same or different and independently optionally substituted alkyl, optionally substituted alkoxy, halo, trifluoroalkyl, or optionally substituted thioalkyl; and

R~ is optionally substituted aryl or optionally substituted aralkyl .

In various embodiments, R 1 is C 1 -C4 alkyl, or R 1 is Ci-C 4 alkoxy, or R 1 is C 1 -C4 thioalkyl.

In other embodiments, R 1 is fluoro, trifluoroalkyl, or c-hloro.

In preferred embodiments, R 1 is not at the ortho- position of the phenyl ring.

In certain embodiments, R' is phenyl or substituted phenyl.

In a further embodiment, R 2 is phenyl substituted with one or more halo {e.g., fluoro). In preferred embodiments, R 2 is phenyl substituted with fluoro.

In yet another embodiment, R 2 is benzyl or substituted benzyl. In a preferred embodiment, R 2 is benzyl.

In a further embodiment, R' is benzyl in which the phenyl moiety is substituted with one or more alkyl {e.g., C-. -C^ alkyl) or halo (e.g., fluoro). In preferred embodiments, R 2 is benzyl substituted with fluoro. In other embodiments, R 2 is benzyl substituted with methyl. In a more specific embodiment, the tetrahydropyrazoiopyridine compounds are represented by Formula (la):

wherein,

R la and R lb are the same or different and independently hydrogen, optionally substituted alkyl, optionally substituted alkoxy, halo, tnfluoroalkyl, or optionally substituted thioalkyl;

R J is hydrogen or halo.

In various embodiments, R la and R lb are independently Ci -C 4 alkyl, Ci- C 4 alkoxy, C 1 -C4 thioalkyl, trifluoroalkyl, or halo. In more specific embodiments, R la and R lb are the same or different and independently methyl, ethyl, methoxv, thiomethyl, trifluoromethyl, fluoro, or chloro.

In preferred embodiments, when R la is hydrogen, R lb is C 1 -C4 alkyl, Ci~ C 4 alkoxy, C 1 -C4 thioalkyl or halo. In more specific embodiments, when R la is hydrogen, R lb is methyl, ethyl, methoxy, thiomethyl, or chloro.

In other embodiments, when R l is hydrogen, R ld is Ci~C 4 alkyl, Cj-C 4 alkoxy, C 1 -C4 thioalkyl, trifluoroalkyl, or halo. In more specific embodiments, when R lb is hydrogen, R la is methyl, ethyl, methoxy, trifluoromethyl, thiomethyl, fluoro, or chloro.

In preferred embodiments, R' is hydrogen. In other preferred

embodiments, R 3 is fluoro, including 3-, or 4-substituted fluoro.

Table 1 provides specific compounds of Formula (la) and their respective percentage inhibition of human pendrin. These compounds demonstrated strong inhibition (at least 45% inhibition) at 25 μΜ. In comparison, the only known pendrin inhibitor, niflumic acid, showed less than 40% inhibition at 100 μΜ. TABLE 1

In another more specific embodiment, the tetrahydropyrazoiopyridine compounds are represented by Formula (lb):

(lb)

wherein,

m is 0, 1 or 2;

R la and R lb are the same or different and independently hydrogen, optionally substituted alkyl, optionally substituted al koxy, halo, tnfluoroalkyl, or optionally substituted thioalkyl; and

each R 4 is the same or different and independently optionally substituted alkyl or halo.

In various embodiments, R ! a and R lb are independently C-. -C^ alkyl, C \ - C 4 alkoxy, C 1 -C4 thioalkyl, trifluoroalkyl, or halo. In more specific embodiments, R la and R lb are the same or different and independently methyl, ethyl, methoxv, thiomethyl, fiuoro or chloro.

In preferred embodiments, when R la is hydrogen, R lb is C 1 -C4 alkyl, Ci~ C 4 alkoxy, C 1 -C4 thioalkyl or halo. In more specific embodiments, when R ! a is hydrogen, R lb is methyl, ethyl, methoxy, or chloro.

In other embodiments, when R lb is hydrogen, R la is -C 4 alkyl, Cj -C 4 alkoxy, C 1 -C4 thioalkyl, trifluoroalkyl, or halo. In more specific embodiments, when R lb is hydrogen, R la is methyl, ethyl, methoxy, fiuoro, or chloro.

Table 2 provides specific compounds of Formula (lb) and their respective percentage inhi bition of human pendrin. These compounds also

demonstrated strong inhibition (at least 45% inhibition) at 25 μΜ. TABLE 2

Yet another embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a

pyrazolothiophenesulfonamide compound represented by Formula (II):

wherein,

R 5a and R 5b are the same or different and independently hydrogen, or optionally substituted alkyl;

R 6 and R' are the same or different and independently hydrogen, optionally substituted alkyl, or -S(0) 2 TMHR, provided one of R 6 and R 7 is -S(0) 2 NHR; and

R is optionally substituted aryl or optionally substituted cycloalkyl. In various embodiments, R 5a and R ,b are the same or different and independently hydrogen or methyl.

In other embodiments, R 6 is -S(0) 2 NHR, and R 7 is hydrogen or methyl. In yet other embodiments, is -S(0) 2 NHR, and R° is hydrogen or methyl.

In various embodiments, R is optionally substituted aryl. In a particularly preferred embodiment, R is tetrahydronaphthalenyl {e.g., 1- tetrahydronaphthalenyl)

In other embodiments, R is phenyl optionally substituted by one or more alkyl (e.g., methyl or ethyl).

In further embodiments, R is optionally substituted cycloalkyl, including for example, cyclohexyl and cyclopentyl.

Table 3 provides specific compounds of Formula (II) and their respective percentage inhibition of human pendrin. These compounds also demonstrated strong inhibition (at least 45% inhibition) at 25 μΜ.

TABLE 3

The small molecule pendrin inhibitors of Formulae (I), (II) and substructures may be formulated into pharmaceutical composition by known methods in the art. The pharmaceutical composition additionally comprises a physiologically acceptable excipient (pharmaceutically acceptable or suitable excipient or carrier) (i.e., a non-toxic material that does not interfere with the activity of the active ingredient). Any suitable excipient or carrier known to those of ordinary skill in the art for use in pharmaceutical compositions may be employed in the compositions described herein. Excipients for therapeutic use are well known, and are described, for example, in Remington: The Science and Practice of Pharmacy (Gennaro, 21 bt Ed. Mack Pub. Co., Easton, PA (2005))

The pharmaceutical compositions disclosed herein may be a sterile aqueous or non-aqueous solution, suspension or emulsion. Such compositions may also be in the form of a solid, liquid, or gas (aerosol). Alternatively, compositions described herein may be formulated as a lyophilizate, or compounds may be encapsulated within liposomes using technology known in the art. Pharmaceutical compositions may also contain other components, which may be biologically active or inactive. Such components include, but are not limited to, buffers (e.g., neutral buffered saline or phosphate buffered saline), carbohydrates (e.g., glucose, mannose, sucrose or dextrans), mannitol, proteins, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione, stabilizers, dyes, flavoring agents, and suspending agents and/or preservatives.

A composition comprising any one of the compounds described herein may be formulated for sustained or slow release. Such compositions may generally be prepared using well known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site.

In a preferred embodiment, the pharmaceutical compositions described herein may be used in aerosol formulation to be administered via inhalation. The compounds may be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like. The aerosol formulation is particularly suitable for delivering the pendrin inhibitors to the airways. In another preferred embodiment, the pendrin inhibitor of the present disclosure can be combined with a loop diuretic (e.g., furosemide) in a pharmaceutical composition.

Treatment of Airway Diseases

In airway epithelial cells, pendrin is thought to facilitate€17 HC0 3 " exchange and may thereby affect the volume and composition of ASL. Inhibition of the pendrin activities in the airway epithelial cells is thus capable of regulating ASL volume, which could secondarily affect mucociliary clearance, bacterial colonization, and other mucosal immune responses associated with abnormal ASL volume.

Thus, one embodiment provides a method of treating an airway disease in a subject in need thereof compri sing administering to the subject a therapeutically effective amount of a pharmaceutical composition having a compound of any one of Formulae (I), ( la ), (lb) or Formula (II).

As used herein, "airway diseases" is characterized by low or abnormal ASL volume and may have a range of symptoms, including mucus production, cough, wheezing, and airway hyper-responsiveness. Airway diseases may take the forms of inflammatory lung diseases (e.g., cystic fibrosis, asthma, and chronical obstructive pulmonary disease), rhinovirus infection, rhinitis, chronic rhinosinusitis, sinusitis, certain viral or bacterial infections, and exposure to industrial toxins.

As demonstrated in further detail in Example 6, the pendrin inhibitors disclosed herein are efficacious in increasing ASL volume and improve ASL hydration in airway cell culture subjected to an inflammatory stimulant (IL-13). Increased airway hydration by pendrin inhibition represents a viable treatment of inflammatory lung diseases including CF. Although pendrin activity and increased ASL hydration were seen only in cell cultures after IL- 3 -treatment, asthma-like symptoms (cough, wheezing, airway hyper-responsiveness) and increased IL-13 are prevalent in subjects with CF, as well as other airway diseases.

Thus, a specific embodiment provides a method of treating cystic fibrosis in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition having a compound of any one of Formulae (I), (la), (lb) or Formula (II).

In still other embodiments, the present disclosure provides methods for treating a disease or condition treatable by inhibiting pend in, partially or completely, in a subject the method comprising administering a pharmaceutical composition comprising a pharmaceutically acceptable excipient and any one or more of the foregoing compounds of Formula (I), (la), (lb) or Formula (II) to the subject.

In the context of inhibiting pendrin, i.e., pendrin-mediated anion exchange, "inhibit" refers specifically to a statistically significant reduction or a decrease in pendrin-mediated anion exchange between a cell and its local environment, relative to a reference. Pendrin inhibition by the compounds of Formula (I), (la), (lb) or (II) can be assessed, at a given molar amount, in human pendrin. As used herein, the molar amount of the pendrin inhibitor may be 2.5μΜ, ΙΟμΜ or 25 μΜ. The compounds of Formula (I), (la), (lb) or (II) disclosed herein typically exhi bit percentage inhibition (at 25 μΜ) of at least 45%, or at least 50%, or at least 60%, or at least 70%, or at least 80% or at least 90% or fully blocking the pendrin activity (near 100% for PDSin h -AOl). Without wishing to be bound by theory, pendrin-mediated anion exchange may be inhibited by impairing an ani on-exchange functionality of pendrin, by reducing or decreasing pendrin protein expression, by reducing or decreasing pendrin protein translation, or by reducing or decreasing pendrin gene expression. Compounds of the present disclosure may be said to be "pendrin-inhibiting" as well as "inhibiting pendrin-mediated anion exchange". These phrases are intended to carry the same meaning; compounds of the present disclosure inhibit the ability of pendrin to perform or facilitate anion exchange. Treatment of Hypertension

Hypertension, also known as high blood pressure, is a long-term medical condition in which the blood pressure in the arteries is persistently elevated.

Hypertension is classified as either primary (essential) hypertension or secondary hypertension. Most cases are categorized as primary hypertension, which is defined as high blood pressure with no apparent underlying cause. The remaining cases (5-10%) are categorized as secondary hypertension, which is associated with an identifiable cause, such as narrowing of the aorta or kidney arteries, chronic kidney disease, or an endocrine disorder such as excess aldosterone, Cortisol, or catecholamines.

Both primary hypertension and secondary hypertension are linked with ionic imbalances. For example, considerable evidence suggests that disturbances in renal salt/water balance (particularly in the intrarenal renin-angiotensin system) may account for increased resistance to bloodflow. Similarly, secondary hypertension is commonly associated with kidney diseases such as renal stenosis. In another example, Liddle's syndrome, a rare inherited form of hypertension, is caused by mutations that affect sodium reabsorption in the renal distal tubule. Additionally, mice overexpressing pendrin have been shown to be hypertensive. Accordingly, pendrin inhibitors disclosed herein may be effective in treating one or more forms of hypertension, such as, for example, salt-sensitive hypertension.

Thus, a further embodiment provides a method for treating hypertension in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition having a compound of any one of Formulae (I), (la), (lb) or Formula ( ! ! ).

Treatment of Thyroid Conditions

Ionic imbalance has also been associated with thyroid issues, including goiter, nodules, tumors, and aberrant thyroid activity. For example, elevated anion gap, which is the difference between measured cation and anion levels in serum, plasma, or urine, has been investigated for association with thyroid dysfunction and nodular goiter. Metabolic acidosis, caused by excessive production of acid and/or deficient removal of acid by the kidneys, is found among patients experiencing hypo- or hyperthyroidism. Thus, pendrin inhibitors disclosed herein may be effective in treating thyroid conditions.

One embodiment provides a method of treating thyroid conditions in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition having a compound of any one of Formulae (I), (la), (lb) or Formula (II). Diuresis and Potentiation

Pendrin inhibitors disclosed herein may be combined with diuretic therapy to enhance diuresis and salt excretion. Such a combination therapy can be effective in treating hypertension and edema (including diuretic-resi stant edema),

Diuresis is the physiological process by which urine production in the kidneys is increased as part of the body' s homeostatic maintenance of fluid balance. A diuretic promotes urine output, osmolality and salt excretion. Although pendrin inhibitors disclosed herein do not appear to be diuretic in an animal model, when combined with a known loop diuretic, the pendrin inhibitors can enhance or potentiate the effect of the loop diuretic (e.g., urine output and Na + secretion were increased by more than 30%, or by more than 60% depending on the use of the diuretic).

Thus, one embodiment provides a method of promoting diuresis to a subject in need thereof comprising administering to the subject a diuretic; and administering to the subject a pendrin inhibitor of any one of Formula (I), (la), (lb) or (II). The pendrin inhibitor may be administered prior to, contemporaneous with, or following administration of the diuretic to the subject. In various embodiments, the diuretic may be admini stered to the subject acutely or chronical ly.

As used herein, a diuretic refers to a loop diuretic, which acts on the Na 7 ' K72Cl ~ cotransporter (N CC2) in the thick ascending limb of the loop of Henle to inhibit sodium, chloride and potassium reabsorption. Common loop diuretics include thiazide compounds such as furosemide, azosemide, bumelanide, piretanide, torasemide, and the li ke. In a preferred embodiment, the loop diuretic is furosemide.

Another embodiment provides a method for treating hypertension in a subject in need thereof comprising administering to the subject a diuretic, and administering to the subject a pendrin inhibitor of any one of Formula (I), (la), (lb) or (II).

Another embodiment provides a method for treating edema in a subject in need thereof comprising administering to the subject a diuretic; and administering to the subject a pendrin inhibitor of any one of Formula (I), (la), (lb) or (II).

ΔΔ Definitions

The terms below, as used herein, have the following meanings, unless indicated otherwise.

"Alkyl" refers to a straight or branched hydrocarbon chain radical which is saturated having from one to 12 carbon atoms, and which is attached to the rest of the molecule by a single bond. An alkyl comprising up to 12 carbon atoms is referred to as a C 1 -C 12 alkyl. Alkyls comprising other numbers of carbon atoms are represented similarly. Alkyl groups include, but are not limited to, Ci -Ci? alkyl, C 1 -C 10 alkyl, Ci-Cg alkyl, Ci-Ce alkyl, C1-C4 alkyl, C1-C3 alkyl, C1-C2 alkyl, C 2 -C 8 alkyl, C 3 -C 8 al kyl and C 4 -C 8 alkyl. Representative alkyl groups include, but are not limited to, methyl, ethyl, w-propyl, 1-methylethyl (/so-propyl), κ-butyl, / ' -butyl, 5-butyl, /i-pentyl,

1, 1 -dimethyl ethyl (/-butyl), 3 -methyl hexyl, 2-methylhexyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted as described below.

" Alkylene" or "alkylene chain" refers to a straight or branched dival ent hydrocarbon chain linking the rest of the molecule to a radical group. Alkylenes may ¬ be saturated or unsaturated (i.e., contains one or more double and/or triple bonds). Representative alkylenes include, but are not limited to, C \ -Cn alkylene, C]-C 8 alkylene, i-Ce alkylene, C1-C4 alkylene, C 1 -C 3 alkylene, C1-C2 al kylene, Ci alkylene, Representative alkylene groups include, but are not limited to, methylene, ethylene, propylene, «-butylene, ethenylene, propenylene, κ-butenylene, propynylene, ft-butynylene, and the like. The al kylene chain i s attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.

Unless stated otherwise specifically in the specification, an alkylene chain may be optionally substituted as described below.

"Aikoxy" refers to a radical of the formula -OR a where R a is an alkyl radical as defined. Aikoxy groups include, but are not limited to, Ci-C 12 aikoxy, C 1 -C30 aikoxy, Ci-Cg aikoxy, Cr-Ce aikoxy, C 1 -C4 aikoxy, C1-C3 aikoxy, C 1 -C 2 aikoxy, C 2 -C 8

Δ$ aikoxy, C 3 -C 8 alkoxy and C 4 -C 8 alkoxy. Unless stated otherwise specifically in the specification, an alkoxy group (i.e., the alkyl portion) may be optionally substituted as described below.

"Alkoxyalkyl" refers to a radical of the formula -RbOR a where R a is an alkyl radical as defined and where R¾ is an alkyl ene radical as defined. Unless stated otherwise specifically in the specification, an alkoxyalkyl group may be optionally substituted as described below.

"Aryl" refers to a radical derived from a hydrocarbon ring system comprising hydrogen, 6 to 30 carbon atoms and at least one aromatic ring. The aryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. Aryl radicals include, but are not limited to, aryl radicals derived from the hydrocarbon ring systems of aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as- indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pieiadene, pyrene, and triphenylene. As used herein, it is not required that the aryl radical is fully aromatic (i.e., with fully unlocalized electrons). Instead, the aryl radical comprises at least one aromatic ring, which may be fused to a non-aromatic ring. An example is tetrahydronaphthalene. Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-"(such as in "aralkyl") is meant to include aryl radicals that are optionally substituted.

"Aralkyl" refers to a radical of the formula -R b -R c where is an alkylene chain as defined above and R c is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl, trityi and the like. Unless stated otherwise specifically in the specification, an aralkyl group, i.e., the aryl moiety, may be optionally substituted.

"Cycloalkyl" refers to a stable 3-10 member non-aromatic ring radical comprising solely of carbon and hydrogen. The cycloalkyl radical may be a

monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. Examples of cycloalkyl radicals include, but are not limited to, cyclohexyl, cyclopentyl. Unless stated otherwise specifically in the specification, a cycloalkyi group may be optionally substituted.

"Halo" or "halogen" refers to bromo (Br), chloro (CI), fluoro (F) or iodo

(I)- "Haloaikyi" refers to an aikyi substituted with one or more halo, for example, trifluoroalkyl, wherein at three hydrogen atoms have been replaced with fluoro moieties. Trifluoroalkyl include trifluoromethyl and the like.

"Heterocyclyl", "heterocycle" or "heterocyclic ring" refers to a stable 3- to 24-membered non-aromatic ring radical comprising 2 to 23 carbon atoms and from one to 8 heteroatoms selected from the group consisting of nitrogen, oxygen, phosphorous and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized, the nitrogen atom may be optionally quatemized; and the heterocyclyl radical may be partially or fully saturated. Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl,

isothiazolidinyl, isoxazolidinyi, morpholinyi, octahydroindolyl, octahydroisoindoiyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrroiidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamo holinyl, 1 -oxo-thiomorpholinyl, 1,1-dioxo-thiomorpholinyl, 12-crown-4, 15-crown-5, 18- crown-6, 21-crown-7, aza-18-crown-6, diaza-18-crown-6, aza-21-crown-7, and diaza- 21-crown-7. Unless stated otherwise specifically in the specification, a heterocyclyl group may be optionally substituted.

"Heterocyclyialkyi" refers to a radical of the formula where R ¾ is an alkylene chain as defined above and R c is one or more heterocyclyl radicals as defined above, for example, tetrahydrofuranyl-methyl, tetrahydropyranyl-methyl and the like. A 6-membered heterocyclyialkyi refers to a heterocyclyialkyi, wherein the

5 heterocyclyl moiety has 6 atoms in the ring. Unless stated otherwise specifically in the specification, a heterocyclalkyl group may be optionally substituted,

"Heteroaryl" refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen, phosphorous and sulfur, and at least one aromatic ring. For purposes of this invention, the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized, the nitrogen atom may be optionally quaternized.

Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazoiyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyi,

benzothiazolyl, benzothiadiazolyl, benzo[£][l,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl,

di benzofuranyl, di benzothiophenyl, furanyl, furanonyl, isothiazolyi, imidazoiyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1- oxidopyridinyl, 1 -oxidopyrimidinyl , 1 -oxidopyrazinyl, 1 -oxidopyridazinyl,

1 -phenyl- 1/J-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolvl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl,

tetrahydroquinolinyl, thiazoiyl, thiadiazolyi, triazolyi, tetrazolyl, triazinyl, and thiophenyl (i.e., thienyl). Unless stated otherwise specifically in the specification, a heteroaryl group may be optionally substituted,

"Heteroarylalkyl" refers to a radical of the formula -R -Rc where ¾ is an alkylene chain as defined above and R c is one or more heteroaryl radicals as defined above, for example, furanyl-methyl, pyridyl -methyl and the like. A 6-membered heteroarylalkyl refers to a heteroarylalkyl, wherein the heteroaryl moiety has 6 atoms in the ring. Unless stated otherwise specifically in the specification, a heteroarylalk l group may be optionally substituted.

"Thioalkyi" refers to a radical of the formula -S-Ra where R a is an alkyl radical as defined. Thioalkyi groups include, but are not limited to, C 1 -C 12 thioalkyi, Cj-Cio thioalkyi, Ci-Cg thioalkyi, Cj-C 6 thioalkyi, C1-C4 thioalkyi, C C 3 thioalkyi, Ci- C 2 thioalkyi, C 2 -C 8 thioalkyi, C 3 -C 8 thioalkyi and C 4 -C 8 thioalkyi. Unless stated otherwise specifically in the specification, a thioalkyi group (i.e., the alkyl portion) may be optionally substituted.

All the above groups may be "optionally substituted," i.e., either substituted or unsubstituted. The term "substituted" as used herein means any of the above groups (i.e., alkyl, alkylene, alkoxy, alkoxyalkyl, aryl, aralkyl, heterocyciyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl and/or trifiuoroalkyl), may be further functionalized wherein at least one hydrogen atom is replaced by a bond to a non- hydrogen atom substituent. Unless stated specifically in the specification, a substituted group may include one or more substituents selected from: oxo, -C0 2 H, nitrile, nitro, -CONH 2 , hydroxy!, thiooxy, alkyl, alkylene, alkoxy, alkoxyalkyl, alkylcarbonyl, alkyloxycarbonyl, aryl, aralkyl, arylcarbonyl, aryloxycarbonyl, aralkylcarbonyl, aralkyloxycarbonyl, aryloxy, cycloaikyi, cyeioalkylalkyl, cycloalkylcarbonyl, cycloalkylalkylcarbonyl, cycloaikyi oxycarbonyl, heterocyciyl, heteroaryl ,

dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarvlsilyl groups, thioalkyi triarylsilyl groups, perfluoroalkyl or perfiuoroalkoxv, for example, trifluoromethyl or trifluoromethoxy. "Substituted" also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example, "substituted" includes any of the above groups in which one or more hydrogen atoms are replaced

with -OR g , -SR g , -S ORg, -S0 2 R g , -OS0 2 R g , -S0 2 OR g , -NS0 2 R g , and ~S0 2 NR g R h . "Substituted" also means any of the above groups in which one or more hydrogen atoms are replaced with -C(=0)Rg, -C(=0)ORg, -CH 2 S0 2 Rg, ~CH 2 S0 2 NR g R h , -SH, -SR g or -SSR g . In the foregoing, R g and ¾ are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroaryl alkyl. In addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.

The compounds described herein may generally be used as the free acid or free base. Alternatively, the compounds may be used in the form of acid or base addition salts. Acid addition salts of the free base amino compounds may be prepared according to methods well known in the art, and may be formed from organic and inorganic acids. Suitable organic acids include (but are not limited to) maleic, fumaric, benzoic, ascorbic, succinic, methanesulfonic, acetic, oxalic, propionic, tartaric, salicylic, citric, gluconic, lactic, mandelic, cinnamic, aspartic, stearic, palmitic, glycolic, glutamic, and benzenesulfonic acids. Suitable inorganic acids include (but are not limited to) hydrochloric, hydrobromic, sulfuric, phosphoric, and nitric acids. Base addition salts of the free acid compounds of the compounds described herein may also be prepared by methods well known in the art, and may be formed from organic and inorganic bases. Suitable inorganic bases included (but are not limited to) the hydroxide or other salt of sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like, and organic bases such as substituted ammonium salts. Thus, the term " ' pharmaceutically acceptable salt" of compounds of Structures I and II and substructures thereof, as well as any and all substructures and specific compounds described herein is intended to encompass any and all

pharmaceutically suitable salt forms.

With regard to stereoisomers, the compounds of structure (I) and structure (II), as well as any sub-structure herein, may have one or more chiral (or asymmetric) centers, and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or («5)~. When the compounds described herein contain olefmic double bonds or other centers of geometric asymmetiy, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers (e.g., cis or trans.) Likewise, ail possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included. It is therefore contemplated that various stereoisomers and mixtures thereof and includes "enantiomers," which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another. Thus, the compounds may occur in any isomeric form, including racemates, racemic mixtures, and as individual enantiomers or diastereomers.

Furthermore, some of the crystal line forms of any compound described herein may exist as polymorphs, which are also included and contemplated by the present disclosure. In addition, some of the compounds may form solvates with water or other organic solvents. Such solvates are similarly included within the scope of compounds and compositions described herein.

As used herein, "administer," "administering," or "to administer" refers to the giving or supplying of a medication, including in vivo administration, as well as administration directly to tissue ex vivo. Generally, compositions may be administered systemically orally, bucally, parenterally, topically, by inhalation or insufflation (i.e., through the mouth or through the nose) or rectally in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants and vehicles as desired, or may be locally administered by means such as, but not limited to, injection, implantation, grafting, topical application or parenterally.

The terms "agent" and "therapeutic agent" are used interchangeably herein to refer to a drug, molecule, composition, or other substance that provides a therapeutic effect. The term "active agent" as used herein, refers to the ingredient, component or constituent of the compositions of the described invention responsible for the intended therapeutic effect.

As used herein, the term "treat," "treating," or "to treat" as used herein, refers to accomplishing one or more of the following: (a) reducing the severity of a disorder; (b) limiting the development of symptoms characteristic of a disorder being treated; (c) limiting the worsening of symptoms characteri stic of a disorder being treated, (d) limiting the recurrence of a disorder in patients that previously had the disorder; and I limiting recurrence of symptoms in patients that were previously asymptomatic for the disorder. The term "treat", "treating" or "to treat" includes abrogating, substantially inhibiting, slowing or reversing the progression of a disease, condition or disorder, substantially ameliorating clinical or estheticai symptoms of a condition, substantially preventing the appearance of clinical or estheticai symptoms of a disease, condition, or disorder, and protecting from harmful or annoying symptoms.

The term "about" when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary between 1% and 15% of the stated number or numerical range. It should also be noted that the term "or" is generally employed in its sense including "and/or" unless the content clearly dictates otherwise.

Methods of Using Pharmaceutical Compositions

In pharmaceutical dosage forms, any one or more of the compounds of

Formula (I), (la), (lb) or (II), substructures, and specific structures described herein may be administered in the form of a pharmaceutically acceptable derivative, such as a salt, or they may also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds. An effective amount or therapeutically effective amount refers to an amount of a compound or a composition comprising one or more compounds administered to a subject, either as a single dose or as part of a series of doses, which is effective to produce a desired therapeutic effect.

Optimal doses may generall be determined using experimental models and/or clinical trials. The optimal dose may depend upon the body mass, weight, or blood volume of the subject. In general, the amount of a compound described herein, that is present in a dose, ranges from about 0.01 g to about 1000 g per kg weight of the host. The use of the minimum dose that is sufficient to provide effective therapy is usually preferred.

Pharmaceutical compositions may be administered in a manner appropriate to the disease or disorder to be treated as determined by persons skilled in the medical arts. Generally, compositions may be administered systemically orally, bucally, parenterally, topically, by inhalation or insufflation (i.e., through the mouth or through the nose). For airway diseases, the pharmaceutical composition disclosed herein may be administered directly into the airway. For diuretic combination therapy, the , the pharmaceutical composition disclosed herein may be administered orally.

The diuretic combination therapy may be administered concurrently when the pendrin inhibitors of Formula (I), (la), (lb) or (II) are formulated into the same pharmaceutical composition with the diuretic or into separate pharmaceutical compositions.

The diuretic combination therapy may be administered in sequential administration, whereby the pendrin inhibitors may be administered by the same route or by different routes from that of the diuretic.

Other embodiments and uses will be apparent to one skilled in the art in light of the present disclosures. The following examples are provided merely as illustrative of various embodiments and shall not be constmed to limit the claims in any ¬ way.

EXAMPLES EXAMPLE 1

HIGH-THROUGHPUT IDENTIFICATION OF SMALL MOLECULE PENDRIN INHIBITORS Cells for high-throughput screening

Fischer rat thyroid (FRT) cells were cultured in Kaign's modified Ham's Fl 2 medium supplemented with 10 % FBS, 2 mM L-glutamine, 100 U/ml penicillin, 100 mg/mi streptomycin, 18 mg ml myo-inositol and 45 mg/ml ascorbic acid. For high- throughput screening, FRT cells were stably transfected with EYFP-H148Q/I152L F46L (EYFP-HIF, in pcDNAB. !/Hygro (+)), isolated using 0.25 mg/ml hygromyciti B, and then transfected with pcDNAS. l (+) encoding human pendrin with clonal ceil lines selected using 0.5 mg/ml G418. Human bronchial epithelial cell cultures

Bronchial tissues were obtained from non-CF (without significant airway disease) and CF subjects following lung transplantation, or from lungs donated for transplantation but subsequently found to be unsuitable for that purpose. Non-CF and CF human bronchial epithelial (HBE and CFBE, respectively) cell cultures were grown at an air-liquid interface as described in detail. At 21 days post-seeding, cells typically formed a tight epithelium ( ¾Έ >1000 cm 1 ). In some experiments, the culture medium was supplemented with 10 ng/ml recombinant human IL-13 (Peprotech, Rocky Hill, NJ) for 7 days starting at day 21 of culture. As needed, the apical ceil surface was rinsed with PBS to remove excess mucus 24 hours or more before ASL measurements. Pendrin transcript levels in human primary ceils were measured by Taqman quantitative PGR using the Hs00166504_ml probe set (ThermoFisher Scientific, Grand Island, NY) at the UCSF Helen Diller Cancer Center Genome Analysis Core. Human tissues were acquired and used with approval from the University of California, San Francisco Committee on Human Research ,

Screening procedures

High-throughput screening was performed using a semi-automated screening platform (Beckman, Fullerton, CA) configured as described. FRT cells expressing human pendrin and EYFP-HIF were plated in 96-well black-walled, clear- bottom tissue culture plates (Coming, Corning, NY) at a density of 20,000 cells/well, and cultured to confluence over 48 h. For screening, plates were washed twice with PBS (in mM: 137 NaCi, 2.7 KC1, 1 . 1 Kl hPO ,. 8.1 Na 2 HP0 4 , 0.9 CaCk 0.5 MgCl 2 ) prior to addition of 100 μΐ PBS containing test compounds. Screening was done with -36,000 compounds from ChemDiv (San Diego, CA), TimTec (Newark, DE), and Asinex (Winston-Salem, NC). After 10 min incubation with test compound, plates were transferred to a Tecan Infinite Ml 000 plate reader (Tecan, Morrisville, NC) for fluorescence assay of pendrin activity. The assay duration was 10 s with initial fluorescence intensity recorded for 2 s prior to addition of 100 μΐ of NaSCN-substituted PBS (137 mM NaCl replaced with NaSCN). All assay plates contained negative controls (DM SO). In some experiments CI " exchange for Γ or N0 3 " was measured similarly using Nal and NaN0 3 -substituted PBS. Control experiments to measure basal ion transport rates were done in FRT cells expressing EYFP-HIF without pendrin.

Screening of -36,000 compounds produced several classes of compounds that inhibited pendrin by >50 % at 25 μΜ (Fig. 1C). Based on their potency two compound classes (A and C) were further characterized. A total of 188 commercially available class A (tetrahydropyrazolopyridines) analogs and 160 class C (pyrazolothiophenesulfonamide) analogs were tested, with structural determinants of activity summarized in Figure ID.

Compounds of Formula (I) ("class A inhibitors") contained a core tetrahydropyrazolopyridine heterocycle with a substituted aniline linked to the core ring at the 5-position via a carbamate linker, and with a substituted-methoxy linked at the 8- position of the core. Analogs with different substitution on the aniline ring (R 1 ) were explored. Para-substituted aniline gave best activity whereas ortho- sub situti on (including methyl, methoxy, halo, and trifluoroni ethyl substituents) abolished activity. The electronic property of the substituent on the aniline appeared to be less crucial, with electron-donating and neutral groups giving highest activity. Several R 2 groups on the 8-methoxy were also explored. In general, analogs with R 2 substituted with phenyl rings were more active than with benzyl rings. Halide-substituted benzyl rings greatly reduced activity.

Compounds of Formula ( f t ) ("class C inhibitors") are comprised of a thiophene with a sulfonamide group and a pyrazole heterocycle linked at the 3- and 5- positions, respectively. Changing the pyrazole from 5- to 4-position of the thiophene ring abolished activity. The sulfonamide group at the 2- or 3-position was tolerated. Substitution on the sulfonamide influenced activity, with primary sulfonamide bearing a bicyclic tetrahydronaphthalene ring giving best activity, whereas monocyclic ring or aikyi-substituted anilines generally reducing activity. Secondary sulfonamide, alkyl amines and electron-rich anilines had no activity. The most potent pendrin compound from each class, PDSm h -AOl and PDSjn h -COl (Figure 1C, top), were further studied. EXAMPLE 2

PDSi NH -AOl AND PDSi NH -COl REVERSIBLY INHIBIT PENDRIN-MED I ATED ANION

EXCHANGE

Cytoplasmic pH (pHi) was measured to assay pendrin-mediated CI " /HC0 3 ' exchange in pendrin-expressing FRT cells and primary cell cultures using the pH-sensitive fluorescent probe BCECF. Cells were loaded for 20 min using 10 μΜ BCECF -AM (Invitrogen, Carlsbad, CA) in HC0 3 ' -containing buffer (in mM: 120 NaCl, 5 KC1, 1 CaC-2, 1 MgS0 4 , 10 glucose, 5 Hepes, 25 NaHC0 3 " ; pH 7.4) at 27 °C in a tissue culture incubator. Cells were washed and transferred to the stage of a TE2000 microscope (Nikon, Melville, NY) equipped with a C9100 EM-CCD (Hamamatsu, San Jose, CA), XCite light source (Excelitas, Waltham, MA), Nikon 20x N.A. 0.75 S Fluor objective, Uniblitz shutter (Vincent Associates, Rochester, NY), and filters for BCECF fluorescence (Chroma, Bellows Fall, VT), where they were purged with 95 % 0 2 /5 % C0 2 . Pendrin-mediated C17HC0 3 ' exchange was initiated by apical addition of iX-free HCOs ' -containing buffer (Na gluconate replacing NaCl, pH 7.4; 95 % 0 2 /5 % C0 2 equilibrated) to generate a 100 mM gluconate gradient driving cytoplasmic CI " efflux. BCECF pH-sensitive fluorescence (490 nm excitation, 535 nm emission) was acquired at 0.5 Hz, with control experiments confirming that BCECF pH-insensitive

fluorescence was not altered during exchange protocols. In some experiments pHi changes in the absence of HC0 3 " were measured using buffers in which NaCl or Na gluconate replaced Nal ΚΌ To relate BCECF fluorescence to pH, a calibration curve over the quasi-linear range of BCECF sensitivity (pH 6.6-7.7) was generated as described.

FRT cells were chosen for screening due to their low intrinsic permeability to anions that are transported by pendrin. As depicted in Figures 1 A and 2 A, pendrin activity was assayed from the kinetics of the halide-sensitive fluorescent protein EYFP-HIF in response to addition of a SCN ' -containing solution to drive pendrin-mediated C17SCN " . Pendrin inhibition reduces the rate of EYFP-HIF quenching. PDSjnh-AOland PDSmh-COl decreased pendrin-mediated C17SCN " and C17F exchange in a concentration-dependent manner, as measured by YFP fluorescence quenching. These data are summarized, along with inhibition of C17N0 3 " exchange, in Figure 2B, together with data from a negative control (DMSO vehicle). Because the only previously-described pendrin inhibitor is the weakly active compound niflumic acid, no positive control was included in the screening plates.

IC 50 values for C17SCN ' exchange for both compounds were ~9 μΜ.

PDSin h -AOland PDS^-COl inhibited CIVr and C17N0 3 ' exchange with IC 50 of ~8 μΜ and ~5 μΜ, respectively. As shown in Figure 2C, kinetics of PDSmh-AO 1 and PDSin h -COlinhibition of pendrin-mediated C17SCN " exchange in which 10 μΜ

PDSm h -AOl was added for indicated times were measured {left). At right is a summary of these data in bar graph form. Both PDSjn h -AOl and PDSi nh -C01 displayed ~IC 50 at 5 minutes, and displayed substantially higher IC values by 7.5 minutes. Inhibition by both compounds is reversible by washout, as shown in Figure 2D.

EXAMPLE 3

PENDRIN INHIBITORS BLOCK CL7HC0 3 " EXCHANGE

C17HC0 3 " exchange is the relevant pendrin function in the airways during inflammatory responses. Pendrin-mediated C17HC0 3 " exchange was measured in pendrin-expressing FRT cells using BCECF to report cytoplasmic pH (pi I) (Figure 3 A). BCECF-iabeled cells were incubated in a Cl ' -containing HC0 3 " -buffered solution, and C17HC0 3 " exchange was initiated by application of a gluconate-containing HC0 3 " - buffered solution to drive CI " efflux and HCO 3 " influx, producing cytoplasmic aikalinization with a rate of -0.1 pH units per min (Figure 3B). Cytoplasmic alkalinization was strongly inhibited by 25 μΜ PDSm h -AOl or PDSm h -COl , with the fluorescence responses similar to those in cells not expressing pendrin (Fig. 3B, top), Fifty percent inhibition was seen at · 2.5 μΜ PDSm h -AOl (Figure 3C). In control experiments done in the absence of HC0 3 " , application of the gluconate-containing solution to ceils did not alter pFL . in pendrin-expressing or non-transfected cells, indicating that observed pH changes are HC0 3 " -dependent, as expected (Figure 3D). EXAMPLE 4

PD S ! H~ AO 1 -MEDIATED INHIBITIO IS SPECIFIC FOR PENDRIN ION EXCHANGE ACTIVITY OVER OTHER SOLUTE TRANSPORT PROTEINS AND OVER THE SEQUENCE- SIMILAR SLC26A3

Further experiments were done with PDSink-AOl because of its greater inhibition potency for C17HC0 3 " exchange than PDSin h -COl . To investigate PDSjn h -AOl specificity, the major ion transport pathways in HBE cells were studied by short-circuit current (I sc ) analysis. Cells were treated with EL- 13 to increase Ca 2" -activated CI " conductance (CaCC) activity. For I sc measurements, cells were sequentially treated with amiloride to inhibit the epithelial sodium channel (ENaC), forskolin to activate the cystic fibrosis transmembrane conductance regulator (CFTR.), CFTR mlr 172 to inhibit CFTR, and ATP to activate CaCC. In the absence of PDS mh -A01, ion channel modulators produced the anticipated changes in I sc (Figure 4A). Pre-treatment of cells for 30 min with 25 μΜ PDSm h -AOl did not alter the I sc responses, indicating that PDSiu h -AOl did not affect the activity of ENaC, CFTR or CaCC, the various other transporters (including K~ channels and NKCC1 cotransporter) required to support their activity, or tight junction conductance.

lL-13-treated HBE cells were cultured on Snapwell clear permeable supports (12 mm diameter, 0.4 μιη polyester membrane, Costar, Corning, Corning, NY). Short-circuit current was measured using symmetrical HC0 3 ' -buffered solutions (in ffiM: 120 NaCl, 5 KC1, 3 MgCl 2 , 1 CaCl 2 , 5 Flepes, 25 NaFIC0 3 ; pf l 7.4) as described, with ion transport modulators added to both apical and basolateral bathing solutions. Cells were equilibrated with 95 % 0 2 /5 % CO? and maintained at 37 °C during data acquisition. Statistical analysis was by Student's t-test.

Experiments were also done to investigate PDSm h -AOl effect on

SLC26A3 (CLD (congenital chloride diairhea) / DRA (downregulated in adenoma)), the protein most similar to pendrin in sequence (-50 % identity). SLC26A3 activity was measured using a SCN " transport assay that produced YFP fluorescence quenching (Figure 4B). YFP fluorescence quenching in SLC26 A3 -expressing cells in response to an SCN " gradient was not significantly inhibited by PDSin k -AOl . SLC26 A3 -mediated ion transport was measured in COST fibroblasts transiently transfected to express EYFP-HIF and human SLC26A3 (Origene, Rockville, MD). COS7 ceils were cultured in DMEM-H21 supplemented with 10 % FBS, 2 m\i glutamine, 100 U/ml penicillin and 100 mg/ml streptomycin and transfected using Lipofectamine 2000 (Invitrogen, Carlsbad, CA). SLC26 A3 -mediated transport was measured from EYFP-HIF fluorescence after addition of a NaSCN-substituted PBS solution to generate a 70 mM SCN " gradient. Control experiments were done using COS-7 cells expressing EYFP-HIF alone.

EXAMPLE 5

PD S IMH - AO 1 INHIBITS CL- HC 03 - EXCHANGE IN IE - 13 TREATED PRIMARY HUMAN

BRONCHIAL EPITHELIAL CELL CULTURES BUT DOES NOT CHANGE ASL PH

Pendrin-mediated C17HC0 3 " exchange was measured in BCECF-labeled HBE and CFBE cells under basal conditions and after pendrin upregulation by IL-13, a cytokine that is elevated in multiple airway disease states including CF. In control cultures (no added IL- 3) pH changes in response to apical application of a gluconate gradient to drive CI " efflux and HC0 3 " influx were not altered by PDSm h -AOl, suggesting little or no pendrin activity under basal conditions (Figure 5A, left).

However, treatment of cells with IL-13, which strongly upregulates pendrin expression, produced a robust intracellular alkalimzation in response to a gluconate gradient, which was inhibited by 25 μΜ PDSm h -A01 (Figure 5A, right, data summary in Figure 5B). Similar results were found in HBE and CFBE cultures. Quantitative PCR (qPCR) analysis indicated that pendrin transcript levels were increased by 30-35-fold in the HBE and CFBE cultures (data not shown), similar to fold-increases previously reported in similar cell systems.

To investigate the possible involvement of pendrin in ASL pH regulation, fluorescence ratio imaging was done of the ASL in HBE and CFBE cultures after staining with BCECF-dextran. In the absence of IL-13 -treatment, ASL pH was -0.3 pH units more acidic in CFBE than HBE cultures (Fig. 5C; pH -7.4 vs. -7.1), in agreement with prior studies showing relative ASL acidity in CF. PDSinh-AOl did not alter ASL pH under basal conditions. Treatment of HBE ceil cultures with IL-13 produced a significant decrease in ASL pH by -0.2 pH units, although no change in ASL pH was seen in IL- 13 -treated CF cultures. Incubation of IL- 13 -treated HBE and CFBE ceil cultures with PDS mh -AOl did not alter ASL pH. These studies indicate that pendrin is not a key determinant of ASL pH under basal or IL- 13 -stimulated conditions, suggesting that alternative pathways contribute to ASL pH homeostasis.

ASL pH was measured using the pH-sensitive fluorophore BCECF conjugated to 10 kDa dextran (Invitrogen, Carlsbad, CA) as described, which was added 4-6 h prior to measurement (20 μΐ of 0.5 mg/ml in PBS). Cell inserts were incubated in HC0 3 " -containing buffer (5 % C0 2 environment) and the ASL was imaged using a SMZ stereoscopic microscope (Nikon, Melville, NY) equipped with a C9100 EM-CCD, Exfo XCite light source and BCECF filters (440/1 OX, 490/20X, 535/25M, Chroma, Bellows Falls, VT). In some experiments ASL pH was measured after PDS m tr A01 treatment, as described for ASL depth measurements. To relate BCECF

fluorescence to pH, a calibration curve over the quasi-linear range of BCECF sensitivity was generated.

EXAMPLE 6

PDSi NH -AOl INCREASES ASL DEPTH IN IL-13 -TREATED PRIMARY HUMAN BRONCHIAL

EPITHELIAL CELL CULTURES ASL depth was measured in HBE and CFBE cell cultures under basal conditions and with chronic IL-13-treatment. CFBE cell cultures showed reduced ASL depth compared to HBE cell cultures in the absence of IL-13 -treatment (~8 μηι versus ~5 μηι; Figure 6A, left; summary in Figure 6B), in agreement with prior studies. Under these basal conditions, treatment of HBE or CFBE cell cultures with 25 μΜ POS^-AOl did not significantly change ASL depth. Treatment of HBE and CFBE cell cultures with IL-13 produced a significant increase in ASL depth to -16 μτη. Remarkably, treatment of IL-13 -treated HBE and CFBE ceil cultures with PDS mh -AOl produced a significant further increase in ASL depth of -8 μτη (Figure 6A, right; summary in Figure 6B). Pendrin inhibition thus causes ASL hydration in IL- 13 -treated HBE and CFBE ceil cultures, providing pharmacological evidence for pendrin as a key regulator of ASL hydration in airway cell cultures exposed to an inflammatory stimulant,

ASL depth was measured by confocai imaging of rhodamine B-dextran fluorescence (20 μΐ of 0.1 mg/ml in PBS) applied 4-6 h prior to measurement. Scanning confocai microscopy was done using an upright Nikon EZ-Cl confocai microscope equipped with a LUMplan FL N 60x/1.00w objective (Olympus, Waltham, MA). For measurements the transwell filter containing cells with iluorescently stained ASL was inverted, with the immersion objective contacting a droplet of PBS added onto the upper-facing basal surface of the transwell filter. In some experiments ASL. depth was measured after treatment with 25 μΜ PDS 1!3h ~A01 in the culture medium for 4-6 hours. ASL depth was measured using a multi-point histogram method in which each confocai z-stack was fitted to a quadratic function, as described and validated previously.

Without wishing to be bound by theory, it is believed that as pendrin mediates electroneutral CITHCO- ? ' exchange, pendrin activity will produce net osmol movement into the cell because some HCO 3 " in the ASL becomes protonated and liberates C0 2 ; pendrin inhibition may therefore increase ASL volume in the steady- state.

In CF, defective CI " and HCO3 " transport into the ASL alters salt and water transport, promoting ASL volume depletion. There is a considerable body of evidence linking ASL volume dysregulation in CF to impaired immune functions and CF disease pathogenesis. Rehydration of the airway surface is thus considered to be a useful therapeutic approach in CF. Evidence from confocai microscopy, transmission electron microscopy and fluorescence photobleaching indicate that the ASL consists of two discrete layers: a superficial mucus layer (ML) that contains large gel-forming mucins (MUC5AC and MUC5B) that trap particulates for subsequent mucociliary clearance, and in contact with cells and cilia, a peri ciliary layer (PCL) that contains large membrane-bound mucins (MUC 1 and MUC4). Classically, the ASL was considered to consist of a mucus gel supported on a water-like liquid layer surrounding cilia ( ' ' gel-on-liquid' model). More recently, Button and colleagues showed that tethered mucins in the PCL form a mesh to prevent entry of gel-forming mucins and support ciliary beating, and proposed a gel-on-bmsti model to describe the ASL,

Photobleaching studies support this concept and have established that the fluid phase viscosity of the ML and PCL are similar in non-CF subjects (7-10-times more viscous than saline), but significantly elevated in both regions in CF cultures (25-30-times more viscous than saline). In addition to forming a permeability barrier, tethered mucins in the PCL generate an osmotic pressure to regulate PCL hydration. The osmotic potential exerted by CF mucus (>8 % solids), but not healthy mucus (~2 % solids), was demonstrated to be sufficient to dehydrate the PCL and collapse cilia, and to restrict mobility of bacteria and foster formation of bacteri al biofilm precursor rnacrocolonies. Restoration of CFTR expression in CF primary epithelial cells by viral transduction significantly increases ASL depth, ciliary beat frequency and mucus transport, supporting the idea that loss of CFTR function mediates ASL dehydration. Therefore, the bulk of available evidence links reduced ASL volume to increased propensity for infection, with reduced CFTR ion transport leading to reduced mucin hydration in the ML followed by PCL dehydration, ciliary collapse, mucostasis and bacterial colonization.

EXAMPLE 7

FURTHER CHARACTERIZATION OF THE PENDRIN INHIBITOR PDS iN H-C01

PDSi NH -COl was further examined for pendrin inhibition and diuretic activity. PDSm h -COl was purchased from ChemDiv (San Diego CA, USA). Other chemicals were purchased from Sigma unless otherwise stated. A plasmid expressing murine pendrin was generated in pcD A3 , l (Invitrogen). A plasmid encoding murine Slcl2a3 (NCC) was purchased from Dharmacon, and plasmids encoding murine Slc4al (AEl ) and myc-tagged murine Slc26a3 (CLD/DRA) were from Origene. A plasmid encoding human N CCl (94 % sequence similarity to murine N CC 1) was provided by B. Forbush (Yale).

Functional studies of pendrin-mediated CI " exchange for I " , SCN " and N03 " were done in FRT cells stably expressing murine pendrin and a YFP halide- sensing fluorescent indicator (Figure 7B). Fischer rat thyroid (FRT) cells were cultured in Kaign's modified Ham's F12 medium supplemented with 10 % FBS, 2 mM L-glutamine, 100 U/ml penicillin, 100 mg/ml streptomycin, 18 mg/ml myo-inositol and 45 mg/ml ascorbic acid. FRT cells stably expressing murine pendrin (from plasmid) and EYFP-H148Q/I152L/F46L (referred to as YFP) (by lentivirus) were generated by limiting dilution and selection using 0.5 mg/mi G418. COS-7 fibroblasts were cultured in DMEM-H21 supplemented with 10 % FBS, 2 mM L-glutamine, 100 U/ml penicillin and 100 mg/ml streptomycin and transiently transfected with cDNAs encoding YFP and various membrane transporters using Lipofectamine 2000.

Pendrin-mediated exchange of CI " with SCN " , Γ or N0 3 " was measured from the kinetics of YFP fluorescence quenching in a BMG FLUOstar Omega platereader. Pendrin/YFP-expressing FRT cells were washed in PBS and transport measured following a 70-mM gradient of SCN " , Γ or N0 3 " . Pendrin-mediated C17HCCV exchange was measured from the kinetics of cytoplasmic pH (pHi) using BCECF-AM (Invitrogen). Pendrin-expressing FRT cells in HC0 3 " -containing buffer (in mM: 120 NaCl, 5 KCl, I CaCl 2 , 1 MgS0 4 , 10 glucose, 5 Hepes, 25 NaHCO, " ; pi t 7.4, 95 % 0,/5 % CO ? , equilibrated) were exposed to a Cl ' -free buffer (gluconate replacing CI " ) to drive CI " efflux and HCO 3 " influx.

As shown in Figure 7B, addition of Γ to the extracellular solution caused YFP fluorescence quenching in pendrin expressing cells, with near-zero quenching in cells expressing YFP alone. Pendrin inhibition by PDSin h -COl reduced the rate of fluorescence quenching ( as a function of CI " exchange with Γ, SCN " or N03 " ) in a concentration-dependent manner (Figure 7C). Pendrin-mediated O7HC0 3 " exchange was measured from the kinetics of intracellular pH, using BCECF fluorescence as a cytoplasmic pH sensor, following extracellular addition of gluconate in HC0 3 7C0 2 - containing buffer to drive CI- efflux, HC0 3 " influx, and consequent cytoplasmic aikalinization (Figure 7D). PDSin h -COl reduced the kinetics of CI7HCO 3 " exchange, the activity of pendrin of relevance to kidney function, in a concentration-dependent manner with IC 50 ~L2 μΜ (Figure 7E). In selectivity studies, PDSin h -COl at 25 μΜ did not significantly inhibit the transport activities of Slc4al (AE1), Slc26a3 (CLD/DRA) or KCCl (Slcl2a2) (Figure 7F). YFP-based fluorescence quenching assays in COS-7 cells were established to measure AE1 (Slc4al), CLD (Slc26a3) and NCC (Sicl2a3) activities. For AE1 assay, cells expressing YFP and AE1 were equilibrated in PBS and then subjected to a 70-mM SCN " gradient (SCN " replacing CI " ). For CLD assay, cells expressing YFP and CLD were subjected to a 102-niM SCN " gradient. For NCC activity, cells expressing YFP and NCC were equilibrated for 1 h in Na gluconate- substituted PBS and then subjected to a 70-mM Γ gradient. A YFP-V163 S fluorescence quenching assay in FRT cells was used to measure N CC1 activity in which cells equilibrated gluconate-substituted PBS buffer were exposed to a Cl ' -containing solution containing 75 mM Na T and 75 mM E . EXAMPLE 8

PHARMACOKINETICS OF PDS !NH -CQllN MICE

Pharmacokinetics measurements were done to guide studies of diuretic efficacy. Animal experiments were approved by UCSF Institutional Animal Care and Use Committee. Female CD1 mice (8-10 weeks) were injected with 10 mg/kg PDSjn h - C01 (in saline containing 5% DMSO and 10% Kolliphor HS) ip and blood was collected by orbital puncture at 15, 30, 60, 150 and 240 min, and centrifuged at 5000 rpm for 15 min to separate plasma. Urine was collected in metabolic cages. An LC/MS method was developed to measure PDSM I -CO I concentrations in mouse blood and urine. Plasma and urine samples (60 uL) were mixed with 300-uL acetomtrile and centrifuged at 13000 rpm for 20 min, and 90 uL of the supernatant was taken for liquid chromatography/mass spectrometry (LC/MS, Waters 2695 and Micromass ZQ). The solvent system consisted of a linear gradient from 5 to 95% acetonitriie over 16 min. Sodium and potassium concentrations in 3-h collected urine samples were measured by flame photometry (PFP7 Clinical Flame Photometer, Bibby Scientific Ltd, Stone.

Staffs, UK). For blood gas analysis, arterial blood was collected from the abdominal aorta under isoflurane anesthesia 3 h after treatment. Blood gas was analyzed by i STATl with CG4+ cartridges (Abbott Laboratories, Abbott Park, IL, USA). Urine pH was measured on freshly collected urine samples using an AB 15 pH Meter (Fisher Scientific, Pittsburgh, PA, US A). Experiments with two groups were analyzed with Student's t-test, and experiments with 3 or more groups were analyzed using one-way analysis of variance and post-hoc Newman-Keuls multiple comparisons test. P<0.05 was taken as statistically significant.

Figures 8A and 8C show original LC/MS data and linear standard curves in plasma and urine in which known amounts of PDSin h -COl were added to plasma and urine from untreated mice. Figures 8B and 8D summarize PDS lnh -C01 concentrations in plasma and urine following bolus intraperitoneal (ip) administration of 10 mg/kg PDSin h -COl, showing predicted therapeutic concentrations for several hours.

EXAMPLE 9

PENDR1N INHIBITION ALONE DOES NOT AFFECT RENAL FUNCTION PDSm h -COl was administered to mice by ip injection as done in the pharmacokinetics measurements. Figure 9A shows similar 3-h urine volume and osmolality in two different strains of mice (CD- I and C57B/6) treated with vehicle or PDSin h -COl . Even at a very high dose of 50 mg/kg, PDSin h -COl administration did not significantly change urine pH (Figure 9B) or blood gas values (Figure 9C), nor did it affect 3-h urinary salt excretion (Figure 9D). Similar results were obtained using PDSin h -AOl (data not shown).

EXAMPLE 10

PENDRIN INHIBITION POTENTIATES THE ACUTE DIURETIC ACTION OF FUROSEMIDE

Although pendrin inhibitors alone did not produce a diuretic response in mice, pendrin inhibition was shown to augment the diuretic response to acute administration of furosemide, a loop-diuretic that increases salt deliver}' to the pendrin- expressing CNT and CCD.

In acute studies, mice (both CD-I and C57B1/6 strains) were injected ip with 10 or 50 mg/kg PDSm h -COl . In some experiments mice were treated with 10 mg/kg PDSm h -COl together with furosemide (5, 10, 20 or 50 mg/kg, ip). The mice were placed in individual metabolic cages and spontaneously voided urine was collected for 3 h for measurement of volume and osmolality (freezing point depression osmometry, Micro-osmometer; Precision Systems, Natick, MA, USA). Mice were administered furosemide and PDSinh-COl (or vehicle) ip at zero time, and urine was collected for the next 3 hours. Figure 10A shows that PDSinh- COl (10 mg/kg) significantly increased urine volume by -30% at each dose of furosemide tested, without effect on urine osmolality. The diuretic effect was significantly greater than that produced by maximal furosemide (50 mg/kg). Increasing PDSinh-COl dose to 50 mg/kg did not further potentiate the furosemide effect. PDSinh- C01, when given with 20 mg/kg furosemide, did not affect urine pH (Figure 1 GB), but produced a compensated metabolic alkalosis (Figure IOC). PDSinh-COl increased 3-h urinary Na+ excretion, with no significant effect on K+ excretion (Figure 10D). Similar results were obtained using PDSinh-COl (data not shown). To rale out an inhibitory effect of furosemide on pendrin activity that could confound the physiological data, in vitro measurements showed no effect of furosemide on pendrin activity (Figure 10E).

EXAMPLE 11

PENDRIN INHIBITION POTENTIATES THE CHRONICAL DIURETIC ACTION OF FUROSEMIDE

Because chronic loop diuretic treatment upregulates renal pendrin expression, which might potentiate the diuretic efficacy of pendrin inhibition, the action of PDSinh-COl in a chronic furosemide treatment model.

In chronic studies, mice were injected with 20 mg/kg furosemide (ip) twice a day for 8 days and then administered PDSinh-COl (10 mg/kg) or vehicle at the time of the final furosemide dose. See Figure 11 A. Urine was collected for 3 h as described above.

After 8 days of furosemide treatment, PDSinh-COl further potentiated the furosemide effect. Figure 1 IB shows a -60% increase in urine volume following PDSinh-COl in the chronically furosemide-treated mice, without effect on urine osmolality. PDS in h-C01 significantly increased urinary Na ÷ and K ÷ excretion (Figure 1 IC), Similar results were obtained using PDSmh-A01 (data not shown).

The various embodiments described above can be combined to provide further embodiments. All U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications, and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modifi ed, if necessary to employ concepts of the various patents, applications, and publications to provide yet further embodiments.

These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible

embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.