Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MODULATORS OF MAS-RELATED G-PROTEIN RECEPTOR X2 AND RELATED PRODUCTS AND METHODS
Document Type and Number:
WIPO Patent Application WO/2022/140520
Kind Code:
A1
Abstract:
Methods are provided for modulating MRGPRX2 generally, or for treating a MRGPRX2 dependent condition more specifically, by contacting the MRGPRX2 or administering to a subject in need thereof, respectively, an effective amount of a compound having structure (I): or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein A, B, C, D, E, G, W, Y, Z, R1, R2, R3, R4, R5, R6, R7 and R8 are as defined herein. Pharmaceutical compositions containing such compounds, as well as the compounds themselves, are also provided.

Inventors:
LANIER MARION (US)
SAINZ MARCOS (US)
BOEHM MARCUS (US)
HUANG LIMING (US)
MARTINBOROUGH ESTHER (US)
SELFRIDGE BRANDON (US)
YEAGER ADAM (US)
Application Number:
PCT/US2021/064817
Publication Date:
June 30, 2022
Filing Date:
December 22, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ESCIENT PHARMACEUTICALS INC (US)
International Classes:
C07D209/42; A61K31/381; A61K31/416; A61K31/4184; A61K31/423; A61K31/437; A61P35/00; C07D231/56; C07D263/56; C07D277/64; C07D333/58; C07D487/04
Domestic Patent References:
WO2017143291A12017-08-24
WO2010027500A12010-03-11
Foreign References:
US202562631305P
Other References:
"Uniprot", Database accession no. Q96LB1
LIT ET AL.: "Salt Selection for Basic Drugs", INT. J. PHARM., vol. 33, 1986, pages 201 - 217
"Remington: The Science and Practice of Pharmacy", 2005, LIPPENCOTT WILLIAMS & WILKINS
Attorney, Agent or Firm:
KHANNA, Hemant et al. (US)
Download PDF:
Claims:
CLAIMS

1. A compound having structure (I): or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

R1 is cycloalkyl, aryl, heterocyclyl, -(CH2)nQ, -CHQR, or -CQ(R)2, wherein R1 is optionally substituted with one or more Rql;

Q is Ci-6 alkyl, aryl, cycloalkyl, heterocyclyl, -CH2C(O)OR, -C(O)OR, -C(O)NHR, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, -N(R)C(O)OR, or -N(R)S(O)2R, wherein Q is optionally substituted with one or more Rq2;

Rql and Rq2 are independently H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -O(CH2)nR, haloalkoxy, -C(O)OR, -C(O)R, - OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, -N(R)S(O)2R, S(O)2R, -C(H)Q’R, or - (CH2)nQ’ where Q’ is selected from Ci-6 alkyl, aryl, cycloalkyl, heterocyclyl, OR’, - C(O)OR’, -OC(O)R’, haloalkyl, -CN, -N(R’)2, -N(R’)C(O)R’, and -N(R’)S(O)2R’;

A is N or CRa;

B is N or CRb;

C is N or CRC;

D is CRd;

E is N or CRe;

G is N or CRS;

W is CRW; Y is N, S or CRy;

Z is N, CRZ, S or O; each R is independently H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, alkylamino, -

(CH2)nR’, halo, aryl, cycloalkyl, heteroaryl or heterocyclyl, or two R groups taken together with the atom to which they are attached form a carbocyle or heterocycle;

R’ is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, haloalkyl, aryl, cycloalkyl, heteroaryl, or heterocyclyl;

Ra is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, - N(R)S(O)2R, or S(O)2R;

Rb is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, - N(R)S(O)2R, or S(O)2R;

Rc is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, - N(R)S(O)2R, or S(O)2R;

Rd is either absent, or when present, H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, -N(R)S(O)2R, or S(O)2R;

Re is either absent, or when present, H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, -N(R)S(O)2R, or S(O)2R;

Rs is either absent, or when present, H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, -N(R)S(O)2R, or S(O)2R;

Rw is either absent, or when present, H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, -N(R)S(O)2R, or S(O)2R;

Ry is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, - N(R)S(O)2R, or S(O)2R; Rz is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, - N(R)S(O)2R, or S(O)2R; n is independently 0, 1, 2, 3, 4 and 5;

R6 is C1.4 alkyl, phenyl, -C(O)R, -(CH2)nOR, -CN, F, Cl, Br, CF3, CF2H or CFH2, with the proviso that R is not H; each R8 is independently H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, -N(R)S(O)2R, or S(O)2R;

R2, R3, R4, R5 and R7 are the same or different and either absent or, when present, independently H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, -CN, -N(R)2, -N(R)C(O)R, -N(R)S(O)2R, or S(O)2R; wherein: adjacent atoms on either the “a” ring or “b” ring may be joined together by single bonds to form a 9 atom carbocyclic or heterocyclic ring structure; or atoms 1 and 2, 3 and 4, 5 and 6 and 8 and 9 on rings “a” ring and “b” ring may be joined together by double bonds to form an aromatic 9 atom carbocyclic or heterocyclic ring structure; or atoms 1 and 2, 3 and 4, 6 and 7 and 8 and 9 on rings “a” ring and “b” ring may be joined together by double bonds to form an aromatic 9 atom carbocyclic or heterocyclic ring structure; with the provisos that: when Z is S or N, then A and C cannot both be N; and when Z is N, then C and G cannot both be N.

2. The compound of claim 1, wherein the compound has one of the following structures (la) or (la’):

175

or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

3. The compound of claim 1, wherein the compound has one of the following structures (lb), (Ic) or (Id):

(Ib) (Ic) (Id) or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

4. The compound of any one of claims 1-3, wherein R1 is aryl, -(CH2)nQ, - CHQR, or -CQ(R)2, wherein R1 is optionally substituted with one or more Rql;

Q is aryl, -CH2C(O)OR, -C(O)OR, -C(O)NHR, haloalkyl, -CN, -N(R)2, -N(R)C(O)R, -N(R)C(O)OR, or -N(R)S(O)2R, wherein Q is optionally substituted with one or more Rq2.

176

5. The compound of claim 4, wherein R1 is phenyl.

6. The compound of any one of claims 4-5, wherein the aryl is unsubstituted.

7. The compound of any one of claim 4-5, wherein the aryl is substituted with one or more Rql.

8. The compound of claim 7, wherein Rql is H, -OR, -N(R)S(O)2R, -CN, - N(R)C(O)R or -C(H)Q’R.

9. The compound of any one of claims 1-3, wherein R1 is heterocyclyl, - (CH2)nQ, -CHQR, or -CQ(R)2, wherein R1 is optionally substituted with one or more Rql;

Q is heterocyclyl, -CH2C(O)OR, -C(O)OR, -C(O)NHR, haloalkyl, -CN, -N(R)2, - N(R)C(O)R, -N(R)C(O)OR, or -N(R)S(O)2R, wherein Q is optionally substituted with one or more Rq2.

10. The compound of claim 9, wherein the heterocyclyl is unsubstituted.

11. The compound of claim 10, wherein the heterocyclyl is substituted with one or more Rql.

12. The compound of claim 11, wherein Rql is H or Ci-6 alkyl.

13. The compound of any one of claims 1-12, wherein R6 is CF3, CF2H, CFH2, Ci- 6 alkyl, -C(O)R, -(CH2)nOR or -CN.

14. The compound of claim 13, wherein R6 is CF3.

15. The compound of claim 13, wherein R6 is CF2H.

16. The compound of claim 13, wherein R6 is Ci-6 alkyl.

177

17. The compound of claim 13, wherein R6 is -C(O)R.

18. The compound of claim 17, wherein R is Ci-6 alkyl.

19. The compound of claim 13, wherein R6 is -(CH2)nOR.

20. The compound of claim 19, wherein n is 0.

21. The compound of claim 19, wherein n is 1.

22. The compound of claim 21, wherein R is Ci-6 alkyl.

23. The compound of claim 13, wherein R6 is -CN.

24. The compound of any one of claims 1-23, wherein each R8 is H.

25. The compound of any one of claims 1-23, wherein Z is N or S.

26. The compound of any one of claims 1-25, wherein each of A, B, C, and Y are

CH.

27. The compound of any one of claims 1-25, wherein E is N.

28. The compound of any one of claims 1-27, wherein Rw is absent.

29. The compound of any one of claims 1-28, wherein atoms 1 and 2, 3 and 4, 5 and 6 and 8 and 9 on rings “a” ring and “b” ring are joined together by double bonds to form an aromatic 9 atom heterocyclic ring structure.

178

30. The compound of any one of claims 1-28, wherein atoms 1 and 2, 3 and 4, 6 and 7 and 8 and 9 on rings “a” ring and “b” ring are joined together by double bonds to form an aromatic 9 atom heterocyclic ring structure.

31. A compound selected from any one of the compounds listed in Table I, or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

32. A pharmaceutical composition comprising the compound of any one of claims 1-31, or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof and at least one pharmaceutically acceptable excipient.

33. A method of modulating a Mas-Related G-Protein Receptor (MRGPR) X2 or a MRGPRX2 ortholog by contacting MRGPRX2 or MRGPRX2 ortholog with an effective amount of the pharmaceutical composition of claim 32.

34. A method of treating a MRGPRX2 or a MRGPRX2 ortholog dependent condition by administering to a subject in need thereof an effective amount of the pharmaceutical composition of claim 32.

35. The method of claim 34, wherein the MRGPRX2 or MRGPRX2 ortholog dependent condition is a pseudo-allergic reaction, an itch associated condition, a pain associated condition, or an inflammatory or autoimmune disorder.

36. The method of claim 35, wherein the itch associated condition is chronic itch; contact dermatitis; Allergic blepharitis; Anemia; Atopic dermatitis; Bullous pemphigoid; Candidiasis; Chicken pox; end-stage renal failure; hemodialysis; Chronic urticaria; Contact dermatitis, Atopic Dermatitis; Dermatitis herpetiformis; Diabetes; Drug allergy, Dry skin; Dyshidrotic dermatitis; Ectopic eczema; Eosinophilic fasciitis; Epidermolysis bullosa; Erythrasma; Food allergy; Folliculitis; Fungal skin infection; Hemorrhoids; Herpes; HIV infection; Hodgkin's disease; Hyperthyroidism; Iodinated contrast dye allergy; Iron deficiency anemia; Kidney disease; Leukemia, porphyrias; Lymphoma; Malignancy; Mastocystosis; Multiple myeloma; Neurodermatitis; Onchocerciasis; Paget's disease; Pediculosis; Polycythemia rubra vera; Prurigo nodularis; Lichen Planus; Lichen Sclerosis; Pruritus ani; Pseudorabies; Psoriasis; Rectal prolapse; Sarcoidosis granulomas; Scabies; Schistosomiasis; Scleroderma, Severe stress, Stasia dermatitis; Swimmer's itch; Thyroid disease; Tinea cruris; Rosacea; Cutaneous amyloidosis; Scleroderma; Acne; wound healing; bum healing; ocular itch; or Urticaria.

37. The method of claim 36, wherein the itch associated condition is urticaria, pruritus, atopic dermatitis, dry skin, psoriasis, contact dermatitis, or eczema.

38. The method of claim 35, wherein the pain associated condition is Acute Pain, Advanced Prostate Cancer, AIDS-Related Pain, Ankylosing Spondylitis, Arachnoiditis, Arthritis, Arthrofibrosis, Ataxic Cerebral Palsy, Autoimmune Atrophic Gastritis, Avascular Necrosis, Back Pain, Behcet’s Disease (Syndrome), Burning Mouth Syndrome, Bursitis, Cancer Pain, Carpal Tunnel, Cauda Equina Syndrome, Central Pain Syndrome, Cerebral Palsy, Cervical Stenosis, Charcot-Marie-Tooth (CMT) Disease, Chronic Fatigue Syndrome (CFS), Chronic Functional Abdominal Pain (CFAP), Chronic Pain, Chronic Pancreatitis, Chronic Pelvic Pain Syndrome, Collapsed Lung (Pneumothorax), Complex Regional Pain Syndrome (RSD), Corneal Neuropathic Pain, Crohn’s Disease, Degenerative Disc Disease, Dental Pain, Dercum’s Disease, Dermatomyositis, Diabetic Peripheral Neuropathy (DPN), Dystonia, Ehlers-Danlos Syndrome (EDS), Endometriosis, Eosinophilia-Myalgia Syndrome (EMS), Erythromelalgia, Fibromyalgia, Gout, Headaches, Herniated disc, Hydrocephalus, Intercostal Neuraligia, Interstitial Cystitis, Irritable Bowel syndrome (IBS), Juvenile Dermatositis (Dermatomyositis), Knee Injury, Leg Pain, Loin Pain-Haematuria Syndrome, Lupus, Lyme Disease, Medullary Sponge Kidney (MSK), Meralgia Paresthetica, Mesothelioma, Migraine, Musculoskeletal pain, Myofascial Pain, Myositis, Neck Pain, Neuropathic Pain, Occipital Neuralgia, Osteoarthritis, Paget’s Disease, Parsonage Turner Syndrome, Pelvic Pain, Periodontitis Pain, Peripheral Neuropathy, Phantom Limb Pain, Pinched Nerve, Polycystic Kidney Disease, Polymyalgia Rhuematica, Polymyositis, Porphyria, Post Herniorraphy Pain Syndrome, Post Mastectomy, Postoperative Pain, Pain Syndrome, Post Stroke Pain, Post Thorocotomy Pain Syndrome, Postherpetic Neuralgia (Shingles), Post-Polio Syndrome, Primary Lateral Sclerosis, Psoriatic Arthritis, Pudendal Neuralgia, Radiculopathy, Raynaud’s Disease, Rheumatoid Arthritis (RA), Sacroiliac Joint Dysfunction, Sarcoidosi, Scheuemann’s Kyphosis Disease, Sciatica, Scoliosis, Shingles (Herpes Zoster), Sjogren’s Syndrome, Spasmodic Torticollis, Sphincter of Oddi Dysfunction, Spinal Cerebellum Ataxia (SCA Ataxia), Spinal Cord Injury, Spinal Stenosis, Syringomyelia, Tarlov Cysts, Transverse Myelitis, Trigeminal Neuralgia, Neuropathic Pain, Ulcerative Colitis, Vascular Pain or Vulvodynia.

39. The method of claim 35, wherein the inflammatory or autoimmune disorder is chronic inflammation, mast cell activation syndrome, Multiple Sclerosis, Steven Johnson’s Syndrome, Toxic Epidermal Necrolysis, appendicitis, bursitis, cutaneous lupus, colitis, cystitis, dermatitis, phlebitis, reflex sympathetic dystrophy/complex regional pain syndrome (rsd/crps), rhinitis, tendonitis, tonsillitis, acne vulgaris, sinusitis, rosacea, psoriasis, graft- versus-host disease, reactive airway disorder, asthma, airway infection, autoinflammatory disease, celiac disease, chronic prostatitis, diverticulitis, glomerulonephritis, hidradenitis suppurativa, hypersensitivities, intestinal disorder, epithelial intestinal disorder, inflammatory bowel disease, irritable bowel syndrome, Crohn’s Disease, ulcerative colitis, lupus erythematous, interstitial cystitis, otitis, pelvic inflammatory disease, endometrial pain, reperfusion injury, rheumatic fever, rheumatoid arthritis, sarcoidosis, transplant rejection, psoriasis, lung inflammation, chronic obstructive pulmonary disease, cardiovascular disease, or vasculitis.

Description:
MODULATORS OF MAS-RELATED G-PROTEIN RECEPTOR X2 AND RELATED PRODUCTS AND METHODS

BACKGROUND

Technical Field

The invention relates to modulators of the Mas-related G-protein coupled receptor X2, to products containing the same, as well as to methods of their use and preparation.

Description of the Related Art

Mas-related G-protein receptors (MRGPRs) are a group of orphan receptors with limited expression in very specialized tissues. Very little is known about the function of most of these receptors. There are eight related receptors in this class expressed in humans, only four of which have readily identifiable orthologs in other species (/.< ., MRGPR D, E, F and G). Other than humans, members of MRGPR subfamily X receptors (MRGPR XI, X2, X3 and X4) have been detected in primates, including macaques and rhesus monkeys, and more recently dogs and horses, but they do not have a single corresponding ortholog in rodents.

BRIEF SUMMARY

This invention is based, in part, on the identification of MRGPRX2 or MRGPRX2 ortholog modulator compounds. MRGPRX2 corresponds functionally to mouse MRGPRB2 and dog MRGPRX2 in mast cells. MRGPRX2 and its ortholog receptors mediate disorders including pseudo-allergic reactions including pseudo-allergic drug reactions, chronic itch (e.g., pruritus), inflammation disorders, pain disorders, skin disorders, wound healing, cardiovascular disease, and lung inflammation/COPD. In one embodiment, both MRGPRB2 and MRGPRX2 expression is largely restricted to mast cells. Mast cells are innate immune cells that primarily reside at sites exposed to the external environment, such as the skin, oral/gastrointestinal mucosa and respiratory tract. Mast cells express numerous receptors that respond to mechanical and chemical stimuli. Upon activation, classically by IgE, mast cells release pre-formed mediators from granules (e.g., histamine, proteases, and heparin) and newly synthesized mediators (e.g., thromboxane, prostaglandin D2, leukotriene C4, tumor necrosis factor alpha, eosinol chemotactor factor, and platelet-activating factor) that elicit allergic and inflammatory responses. Histamine dilates post-capillary venules, activates the endothelium, and increases blood vessel permeability. This causes local edema, warmth, redness, and chemotaxis of other inflammatory cells to the site of release. Histamine also contributes to neuronal sensitization that leads to pain or itch. MRGPRB2 and MRGPRX2 mediate immunoglobulin E (IgE) independent activation of mast cells. MRGPRB2 and MRGPRX2 are receptors for (or sensitive to activation by) various ligands, including basic secretagogues (small cationic molecules), certain drugs (e.g., cationic peptidergic drugs), neuropeptides, and antimicrobial peptides, and thus are important for non-IgE mediated pseudo-allergic reactions, inflammation, pain, and itch conditions. Mast cells may also contribute to the progression of autoimmune disorders by promoting chronic inflammation in the local tissue microenvironment and ultimately polarizing toward a T h 17 immune response. Thus, modulating MRGPRX2 or MRGPRX2 ortholog allows for treatment of autoimmune diseases, pseudo-allergic drug reactions, pain, itch, and inflammatory disorders such as inflammatory bowel disease, urticaria, sinusitis, asthma, rosacea, endometriosis, and other MRGPRX2 or MRGPRX2 ortholog dependent conditions as explained in more detail below.

In one embodiment, a method is provided for treating a MRGPRX2 or a MRGPRX2 ortholog dependent condition by administering to a subject in need thereof an effective amount of the pharmaceutical composition of the modulator compounds of the present invention.

Accordingly, in an embodiment, is provided a compound having structure (I):

or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein A, B, C, D, E, G, W, Y, Z, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are as defined herein.

In other embodiments, compounds are provided having any of the structures (la), (la’), (lb), (Ic) or (Id) as defined herein, or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

In yet other embodiments, pharmaceutical compositions are provided comprising a carrier or excipient and a compound having structure (I), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

In other embodiments, pharmaceutical compositions are provided comprising substructures of structure (I) with structures (la), (la’), (lb), (Ic) or (Id) as defined herein or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

In another embodiment, methods are provided for treating an MRGPRX2 or MRGPRX2 ortholog dependent condition by administering to a subject in need thereof an effective amount of a compound having structure (I), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

In some embodiments, the MRGPRX2 or MRGPRX2 ortholog dependent condition is one or more of a pseudo-allergic reaction, itch associated condition, a pain associated condition, an inflammation-associated condition, or an autoimmune disorder.

In one embodiment, the methods of treating the MRGPRX2 or MRGPRX2 ortholog dependent condition are provided which comprise administering an effective amount of a compound of structure (I) or any of the structures (la), (la’), (lb), (Ic) or (Id) as defined herein or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

In another embodiment, compounds are provided having one or more of the structures disclosed herein, or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

DETAILED DESCRIPTION

As mentioned above, the invention relates to modulators of the MRGPRX2, to products containing the same, as well as to methods of their use and preparation. This invention is based, in part, on the identification MRGPRX2 modulator compounds. Both MRGPRB2 and MRGPRX2 are expressed in mast cells and dorsal root ganglia. Both MRGPRX2 and MRGPRB2 are receptors for (or sensitive to activation by) a diverse group of ligands, including basic secretagogues, certain drugs, neuropeptides, antimicrobial peptides, and thus are important for pseudo-allergic reactions, itch, pain, or inflammatory disorders upon exposure.

MRGPRs appear to be sensory receptors that recognize their external environment to exogenous or endogenous signals/chemicals. These receptors likely respond to multiple chemical ligands/agonists. For example, MRGPRX2 recognizes Compound 48/80, Substance P, mastoparan, icatibant, ciprofloxacin, and tracurium, as agonist signals. In certain embodiments, molecules of this invention modulate MRGPRX2 by functioning as inverse agonists that are capable of blocking multiple chemical entities, and/or as competitive antagonists that can specifically block individual ligands. In one embodiment, such modulations are selective against other MRGPRs, such as MRGPRX1, X3 and/or X4.

Definitions

As used herein, the following terms have the meaning defined below, unless the context indicates otherwise.

“Modulating” MRGPRX2 means that the compound interacts with the MRGPRX2 in a manner such that it functions as an inverse agonist to the receptor, and/or as a competitive antagonist to the receptor. In one embodiment, such modulation is partially or fully selective against other MRGPRs, such as MRGPRX1, X3 and/or X4.

“MRGPR” refers to one or more of the Mas-related G protein coupled receptors, which are a group of orphan receptors with limited expression in very specialized tissues (e.g., in mast cells and dorsal root ganglia) and barrier tissues. There are eight related receptors in this class expressed in humans, only 4 of which have readily identifiable orthologs in other species (i.e., MRGPR D, E, F and G). The other four receptors (MRGPR XI, X2, X3 and X4) have counterparts in higher species including monkeys, dogs and horses, but they do not have a single corresponding ortholog in rodents.

“MRGPRX2,” also referred to as “MRGX2,” or “MGRG3,” refers to a member of the MRGPR family that is expressed on mast cells and capable of mediating IgE independent activation (e.g., mast cell degranulation) in response to ligand binding. An exemplary human MRGPRX2 amino acid sequence is set forth in Uniprot Q96LB1.

“MRGPRX2 ortholog” refers to genes in non-human species including dogs and horses that have evolved through speciation events. Orthologs of human MRGPRX2 are determined by expression pattern and pharmacology. Studies of non-human species MRGPRX2 show gene clusters containing some human MRGPRX2 members, some with coding genes, and some showing sequence identity to human MRGPRX2.

“Effective amount” refers to a quantity of a specified agent sufficient to achieve a desired effect in a subject being treated with that agent. Ideally, an effective amount of an agent is an amount sufficient to inhibit or treat the disease without causing substantial toxicity in the subject. The effective amount of an agent will be dependent on the subject being treated, the severity of the affliction, and the manner of administration of the pharmaceutical composition. Methods of determining an effective amount of the disclosed compound sufficient to achieve a desired effect in a subject will be understood by those of skill in the art in light of this disclosure.

“Alkyl” means a saturated or unsaturated straight chain or branched alkyl group having from 1 to 8 carbon atoms, in some embodiments from 1 to 6 carbon atoms, in some embodiments from 1 to 4 carbon atoms, and in some embodiments from 1 to 3 carbon atoms. Examples of saturated straight chain alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl-, n-hexyl, n-heptyl, and n-octyl groups. Examples of branched alkyl groups include, but are not limited to, isopropyl, isobutyl, sec-butyl, t-butyl, neopentyl, isopentyl, and 2,2-dimethylpropyl groups. An unsaturated alkyl includes alkenyl and alkynyl as defined below.

“Alkenyl” means a straight chain or branched alkenyl group having from 2 to 8 carbon atoms, in some embodiments from 2 to 6 carbon atoms, in some embodiments from 2 to 4 carbon atoms, and in some embodiments from 2 to 3 carbon atoms. Alkenyl groups are unsaturated hydrocarbons that contain at least one carbon-carbon double bond. Examples of lower alkenyl groups include, but are not limited to, vinyl, propenyl, butenyl, pentenyl, and hexenyl.

“Alkynyl” means a straight chain or branched alkynyl group having from 2 to 8 carbon atoms, in some embodiments from 2 to 6 carbon atoms, in some embodiments from 2 to 4 carbon atoms, and in some embodiments from 2 to 3 carbon atoms. Alkynyl groups are unsaturated hydrocarbons that contain at least one carbon-carbon triple bond. Examples of alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, and hexynyl.

“Halo” or “halogen” refers to fluorine, chlorine, bromine, and iodine.

“Hydroxy” refers to -OH.

“Cyano” refers to -CN.

Amino refers to -NH2, -NHalkyl or N(alkyl)2, wherein alkyl is as defined above. Examples of amino include, but are not limited to - NH2, -NHCH3, -N(CHs)2, and the like.

“Haloalkyl” refers to alkyl as defined above with one or more hydrogen atoms replaced with halogen. Examples of lower haloalkyl groups include, but are not limited to, -CF3, -CHF2, and the like.

“Alkoxy” refers to alkyl as defined above joined by way of an oxygen atom (i.e., -O-alkyl). Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, n-butoxy, isopropoxy, sec-butoxy, tert-butoxy, and the like.

“Haloalkoxy” refers to haloalkyl as defined above joined by way of an oxygen atom (i.e., -O-haloalkyl). Examples of lower haloalkoxy groups include, but are not limited to, -OCF3, and the like. “Cycloalkyl” refers to alkyl groups forming a ring structure, which can be substituted or unsubstituted, wherein the ring is either completely saturated, partially unsaturated, or fully unsaturated, wherein if there is unsaturation, the conjugation of the pi-electrons in the ring do not give rise to aromaticity. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl groups. In some embodiments, the cycloalkyl group has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms range from 3 to 5, 3 to 6, or 3 to 7. Cycloalkyl groups further include polycyclic cycloalkyl groups such as, but not limited to, norbornyl, adamantyl, bornyl, camphenyl, isocamphenyl, and carenyl groups, and fused rings such as, but not limited to, decalinyl, and the like.

“Aryl” groups are cyclic aromatic hydrocarbons that do not contain heteroatoms. Representative aryl groups include, but are not limited to, phenyl, azulenyl, heptalenyl, biphenyl, indacenyl, fluorenyl, phenanthrenyl, triphenylenyl, pyrenyl, naphthacenyl, chrysenyl, biphenylenyl, anthracenyl, and naphthyl groups. In some embodiments, aryl groups contain 6-14 carbons in the ring portions of the groups. The terms “aryl” and “aryl groups” include fused rings wherein at least one ring, but not necessarily all rings, are aromatic, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like). In one embodiment, aryl is phenyl or naphthyl, and in another embodiment aryl is phenyl.

“Carbocycle” refers to alkyl groups forming a ring structure, which can be substituted or unsubstituted, wherein the ring is either completely saturated, partially unsaturated, or fully unsaturated, wherein if there is unsaturation, the conjugation of the pi-electrons in the ring may give rise to aromaticity. In one embodiment, carbocycle includes cycloalkyl as defined above. In another embodiment, carbocycle includes aryl as defined above.

“Heterocycle” refers to aromatic and non-aromatic ring moieties containing 3 or more ring members, of which one or more is a heteroatom such as, but not limited to, N, O, S, or P. In some embodiments, heterocyclyl include 3 to 20 ring members, whereas other such groups have 3 to 15 ring members. At least one ring contains a heteroatom, but every ring in a polycyclic system need not contain a heteroatom. For example, a dioxolanyl ring and a benzdioxolanyl ring system (methylenedi oxyphenyl ring system) are both heterocyclyl groups within the meaning herein.

Heterocyclyl groups also include fused ring species including those having fused aromatic and non-aromatic groups. A heterocyclyl group also includes polycyclic ring systems containing a heteroatom such as, but not limited to, quinuclidyl, and also includes heterocyclyl groups that have substituents, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups, bonded to one of the ring members. A heterocyclyl group as defined herein can be a heteroaryl group or a partially or completely saturated cyclic group including at least one ring heteroatom. Heterocyclyl groups include, but are not limited to, pyrrolidinyl, furanyl, tetrahydrofuranyl, dioxolanyl, piperidinyl, piperazinyl, morpholinyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl, dihydrobenzofuranyl, indolyl, dihydroindolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthal enyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, quinoxalinyl, and quinazolinyl groups.

In one embodiment, heterocyclyl includes heteroaryl.

“Heteroaryl” refers to aromatic ring moieties containing 5 or more ring members, of which, one or more is a heteroatom such as, but not limited to, N, O, and S. Heteroaryl groups include, but are not limited to, groups such as pyrrolyl, pyrazolyl, pyridinyl, pyridazinyl, pyrimidyl, pyrazyl, pyrazinyl, pyrimidinyl, thienyl, triazolyl, tetrazolyl, triazinyl, thiazolyl, thiophenyl, oxazolyl, isoxazolyl, benzothiophenyl, benzofuranyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthal enyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, and quinazolinyl groups. The terms “heteroaryl” and “heteroaryl groups” include fused ring compounds such as wherein at least one ring, but not necessarily all rings, are aromatic, including tetrahydroquinolinyl, tetrahydroisoquinolinyl, indolyl, and 2,3-dihydro indolyl. “Isomer” is used herein to encompass all chiral, diastereomenc or racemic forms of a structure (also referred to as a stereoisomer, as opposed to a structural or positional isomer), unless a particular stereochemistry or isomeric form is specifically indicated. Such compounds can be enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions, at any degree of enrichment. Both racemic and diastereomeric mixtures, as well as the individual optical isomers can be synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these are all within the scope of certain embodiments of the invention. The isomers resulting from the presence of a chiral center comprise a pair of nonsuperimposable-isomers that are called “enantiomers.” Single enantiomers of a pure compound are optically active (i.e., they are capable of rotating the plane of plane polarized light and designated R or S).

“Isolated optical isomer” means a compound which has been substantially purified from the corresponding optical isomer(s) of the same formula. For example, the isolated isomer may be at least about 80%, at least 80% or at least 85% pure by weight. In other embodiments, the isolated isomer is at least 90% pure or at least 98% pure, or at least 99% pure by weight.

“Substantially enantiomerically or diastereomerically” pure means a level of enantiomeric or diastereomeric enrichment of one enantiomer with respect to the other enantiomer or diastereomer of at least about 80%, and more specifically in excess of 80%, 85%, 90%, 95%, 98%, 99%, 99.5% or 99.9%.

The terms “racemate” and “racemic mixture” refer to an equal mixture of two enantiomers. A racemate is labeled “(±)” because it is not optically active (i.e., will not rotate plane-polarized light in either direction since its constituent enantiomers cancel each other out). All compounds with an asterisk (*) adjacent to a tertiary or quaternary carbon are optically active isomers, which may be purified from the respective racemate and/or synthesized by appropriate chiral synthesis.

A “hydrate” is a compound that exists in combination with water molecules. The combination can include water in stoichiometric quantities, such as a monohydrate or a dihydrate, or can include water in random amounts. As the term is used herein a “hydrate” refers to a solid form; that is, a compound in a water solution, while it may be hydrated, is not a hydrate as the term is used herein. A “solvate” is similar to a hydrate except that a solvent other that water is present. For example, methanol or ethanol can form an “alcoholate”, which can again be stoichiometric or non-stoichiometric. As the term is used herein a “solvate” refers to a solid form; that is, a compound in a solvent solution, while it may be solvated, is not a solvate as the term is used herein.

“Isotope” refers to atoms with the same number of protons but a different number of neutrons, and an isotope of a compound of structure (I) includes any such compound wherein one or more atoms are replaced by an isotope of that atom. For example, carbon 12, the most common form of carbon, has six protons and six neutrons, whereas carbon 13 has six protons and seven neutrons, and carbon 14 has six protons and eight neutrons. Hydrogen has two stable isotopes, deuterium (one proton and one neutron) and tritium (one proton and two neutrons). While fluorine has a number of isotopes, fluorine- 19 is longest-lived. Thus, an isotope of a compound having the structure of structure (I) includes, but not limited to, compounds of structure (I) wherein one or more carbon 12 atoms are replaced by carbon-13 and/or carbon-14 atoms, wherein one or more hydrogen atoms are replaced with deuterium and/or tritium, and/or wherein one or more fluorine atoms are replaced by fluorine- 19.

“Salt” generally refers to an organic compound, such as a carboxylic acid or an amine, in ionic form, in combination with a counter ion. For example, salts formed between acids in their anionic form and cations are referred to as “acid addition salts”. Conversely, salts formed between bases in the cationic form and anions are referred to as “base addition salts.”

The term “pharmaceutically acceptable” refers an agent that has been approved for human consumption and is generally non-toxic. For example, the term “pharmaceutically acceptable salt” refers to nontoxic inorganic or organic acid and/or base addition salts (see, e.g., Lit et al., Salt Selection for Basic Drugs, Int. J. Pharm., 33, 201-217, 1986) (incorporated by reference herein).

Pharmaceutically acceptable base addition salts of compounds of the invention include, for example, metallic salts including alkali metal, alkaline earth metal, and transition metal salts such as, for example, calcium, magnesium, potassium, sodium, and zinc salts. Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, 7V,7Vdibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine.

Pharmaceutically acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric, and phosphoric acids. Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, aromatic aliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, hippuric, malonic, oxalic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, panthothenic, trifluoromethanesulfonic, 2-hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, phydroxybutyric, salicylic, galactaric, and galacturonic acid.

The compounds of the disclosure (i.e., compounds of structure (I) and embodiments thereof), or their pharmaceutically acceptable salts may contain one or more centers of geometric asymmetry and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that are defined, in terms of absolute stereochemistry, as (R)- or (5)- or, as (D)- or (L)- for amino acids. Embodiments thus include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also included. Although pharmaceutically unacceptable salts are not generally useful as medicaments, such salts may be useful, for example as intermediates in the synthesis of compounds having structure (I), for example in their purification by recrystallization.

As used herein, the phrase “MRGPRX2 dependent condition” means a condition where the activation, over sensitization, or desensitization of MRGPRX2 by a natural or synthetic ligand initiates, mediates, sustains, or augments a pathological condition. For example, it is known that some cationic peptidergic drugs cause pseudo-allergic reactions in patients MRGPRX2 is sensitive to (or activated by) secretagogues, cationic peptidergic drugs, including icatibant, leuprolide, or ganirelix, neutral and anionic peptidergic drugs (e.g., exenatide, glucagon, liraglutide, enfuviritide, colistimethate), non-steroidal agonist (atracurium mivacurium), non-steroidal antagonist drugs, neuropeptides, and antimicrobial peptides. Moreover, overexpression of MRGPRX2 and/or overactivity of MRGPRX2 may also render mast cells more susceptible to activation by endogenous and/or exogenous ligands. Without limited by theory, it is to be understood that by modulating MRGPRX2, pseudo-allergic reactions, itch, pain, inflammatory or autoimmune disorders can be eased.

In some embodiments, the MRGPRX2 dependent condition is a condition that is caused by IgE independent activation of MRGPRX2. IgE independent activation of MRGPRX2 is capable of inducing mast cell degranulation. For example, IgE independent mast cell activation associated with some cases of chronic urticaria and other mast cell mediated conditions, which are not responsive to current anti-IgE or antihistamine therapies. Thus, the compounds of the present disclosure may be used for treating an MRGPRX2 dependent condition caused by IgE independent activation of MRGPRX2 and that would benefit from modulating MRGPRX2.

In some embodiments, the MRGPRX2 dependent condition is an itch associated condition, a pain associated condition, a pseudo-allergic reaction, an autoimmune or inflammatory disorder, or cancer-associated condition.

As used herein the phrase “pseudo-allergic reaction” refers to an IgE-independent allergic reaction, characterized by histamine release, inflammation, airway contraction, or any combination thereof. A pseudo-allergic reaction may be an analphylactic reaction. A pseudo-allergic reaction may be caused by a range of cationic substances, collectively called basic secretagogues, including inflammatory peptides and drugs associated with allergic-type reactions. Thus, in one embodiment, the method of present invention is provided to treat a pseudo-allergic reaction, such as pseudo-allergic reactions caused by secretagogues, cationic peptidergic drugs, anionic peptidergic drugs, neutral peptidergic drugs, non-steroidal antagonist drugs, neuropeptides, and antimicrobial peptides. In one embodiment, the pseudo-allergic reaction is caused by MCD peptide, Substance P, VIP, PACAP, dynorphin, somatostatin, Compound 48/80, cortistatin-14, mastoparan, melettin, cathelicidin peptides, ciprofloxacin, vancomycin, leuprolide, goserelin, histrelin, triptorelin, cetrorelix, ganirelix, degarelix, octreotide, lanreotide, pasireotide, sermorelin, tesamorelin, icatibant, glatiramer acetate, teriparatide, pramlintide, bleomycin, exenatide, glucagon, liraglutide, enfuvirtide, colistimethate, succinylcholine, tubocurarine, atracurium, mivacurium, and rocuronium.

As used herein, the phrase “itch associated condition” means pruritus (including acute and chronic pruritus) associated with any condition. The itch sensation can originate, e.g., from the peripheral nervous system (e.g., dermal or neuropathic itch) or from the central nervous system (e.g., neuropathic, neurogenic or psychogenic itch). Thus, in one embodiment, the method of present invention is provided to treat an itch associated condition, such as chronic itch; contact dermatitis; Allergic blepharitis; Anaphylaxis; Anaphylactoid drug reactions; Anaphylactic shock; Anemia; Atopic dermatitis; Bullous pemphigoid; Candidiasis; Chicken pox; end-stage renal failure; hemodialysis; Cholestatic pruritis; Chronic urticaria; Contact dermatitis, Dermatitis herpetiformis; Diabetes; Drug allergy, Dry skin; Dyshidrotic dermatitis; Ectopic eczema; Eosinophilic fasciitis; Epidermolysis bullosa; Erythrasma; Food allergy; Folliculitis; Fungal skin infection; Hemorrhoids; Herpes; HIV infection; Hodgkin's disease; Hyperthyroidism; Iodinated contrast dye allergy; Iron deficiency anemia; Kidney disease; Leukemia, porphyrias; Lymphoma; Mast cell activation syndrome, Malignancy; Mastocystosis; Multiple myeloma; Neurodermatitis; Onchocerciasis; Paget's disease; Pediculosis; Polycythemia rubra vera; Prurigo nodularis; Lichen Planus; Lichen Sclerosis; Pruritus ani; Pseudo-allergic reactions; Pseudorabies; Psoriasis; Rectal prolapse; Sarcoidosis granulomas; Scabies; Schistosomiasis; Scleroderma, Severe stress, Stasia dermatitis; Swimmer's itch; Thyroid disease; Tinea cruris; Uremic Pruritus; Rosacea; Cutaneous amyloidosis; Scleroderma; Acne; wound healing; burn healing; ocular itch; and Urticaria.

As used herein, the phrase “pain associated condition” means any pain due to a medical condition. Thus, in one embodiment, the method of present invention is provided to treat a pain associated condition, such as Acute Pain, Advanced Prostate Cancer, AIDS-Related Pain, Ankylosing Spondylitis, Arachnoiditis, Arthritis, Arthrofibrosis, Ataxic Cerebral Palsy, Autoimmune Atrophic Gastritis, Avascular Necrosis, Back Pain, Behcet’s Disease (Syndrome), Burning Mouth Syndrome, Bursitis, Cancer Pain, Carpal Tunnel, Cauda Equina Syndrome, Central Pain Syndrome, Cerebral Palsy, Cervical Stenosis, Charcot-Marie-Tooth (CMT) Disease, Chronic Fatigue Syndrome (CFS), Chronic Functional Abdominal Pain (CFAP), Chronic Pain, Chronic Pancreatitis, Chronic Pelvic Pain Syndrome, Collapsed Lung (Pneumothorax), Complex Regional Pain Syndrome (RSD), Corneal Neuropathic Pain, Crohn’s Disease, Degenerative Disc Disease, Dental Pain, Dercum’s Disease, Dermatomyositis, Diabetic Peripheral Neuropathy (DPN), Dystonia, Ehlers-Danlos Syndrome (EDS), Endometriosis, Eosinophilia-Myalgia Syndrome (EMS), Erythromelalgia, Fibromyalgia, Gout, Headaches, Herniated disc, Hydrocephalus, Intercostal Neuraligia, Interstitial Cystitis, Irritable Bowel syndrome (IBS), Juvenile Dermatositis (Dermatomyositis), Knee Injury, Leg Pain, Loin Pain-Haematuria Syndrome, Lupus, Lyme Disease, Medullary Sponge Kidney (MSK), Meralgia Paresthetica, Mesothelioma, Migraine, Musculoskeletal pain, Myofascial Pain, Myositis, Neck Pain, Neuropathic Pain, Occipital Neuralgia, Osteoarthritis, Paget’s Disease, Parsonage Turner Syndrome, Pelvic Pain, Periodontitis Pain, Peripheral Neuropathy, Phantom Limb Pain, Pinched Nerve, Polycystic Kidney Disease, Polymyalgia Rhuematica, Polymyositis, Porphyria, Post Herniorraphy Pain Syndrome, Post Mastectomy, Postoperative Pain, Pain Syndrome, Post Stroke Pain, Post Thorocotomy Pain Syndrome, Postherpetic Neuralgia (Shingles), Post-Polio Syndrome, Primary Lateral Sclerosis, Psoriatic Arthritis, Pudendal Neuralgia, Radiculopathy, Raynaud’s Disease, Rheumatoid Arthritis (RA), Sacroiliac Joint Dysfunction, Sarcoidosi, Scheuemann’s Kyphosis Disease, Sciatica, Scoliosis, Shingles (Herpes Zoster), Sjogren’s Syndrome, Spasmodic Torticollis, Sphincter of Oddi Dysfunction, Spinal Cerebellum Ataxia (SCA Ataxia), Spinal Cord Injury, Spinal Stenosis, Syringomyelia, Tarlov Cysts, Transverse Myelitis, Trigeminal Neuralgia, Neuropathic Pain, Ulcerative Colitis, Vascular Pain and Vulvodynia.

As used herein, the term “autoimmune disorder”, or “inflammatory disorder” means a disease or disorder arising from and/or directed against an individual’s own tissues or organs, or a co-segregate or manifestation thereof, or resulting condition therefrom. Typically, various clinical and laboratory markers of autoimmune diseases may exist including, but not limited to, hypergammaglobulinemia, high levels of autoantibodies, antigen-antibody complex deposits in tissues, clinical benefit from corticosteroid or immunosuppressive treatments, and lymphoid cell aggregates in affected tissues. Thus, in one embodiment, the method of present invention is provided to treat an autoimmune disorder, such as chronic inflammation, mast cell activation syndrome, Multiple Sclerosis, Steven Johnson’s Syndrome, Toxic Epidermal Necrolysis, appendicitis, bursitis, cutaneous lupus, colitis, cystitis, dermatitis, phlebitis, reflex sympathetic dystrophy/complex regional pain syndrome (rsd/crps), rhinitis, tendonitis, tonsillitis, acne vulgaris, sinusitis, rosacea, psoriasis, graft-versus-host disease, reactive airway disorder, asthma, airway infection, allergic rhinitis, autoinflammatory disease, celiac disease, chronic prostatitis, diverticulitis, glomerulonephritis, hidradenitis suppurativa, hypersensitivities, intestinal disorder, epithelial intestinal disorder, inflammatory bowel disease, irritable bowel syndrome, Crohn’s Disease, ulcerative colitis, lupus erythematous, interstitial cystitis, otitis, pelvic inflammatory disease, endometrial pain, reperfusion injury, rheumatic fever, rheumatoid arthritis, sarcoidosis, transplant rejection, psoriasis, lung inflammation, chronic obstructive pulmonary disease, permanent sputum eosiniophilia, eosinophilic leukemia, eosinophilic esophagitis, eosinophilic gastritis, eosinophilic duodenitis, eosinophilic gastroenteritis, mast cell gastrointestinal disease, hypereosinophilic syndrome, aspirin-exacerbated respiratory disease, nasal polyposis, chronic rhinosinusitis, antibody-dependent cell-mediated cytotoxicity, neurofibromatosis, swannamatoisis, tubulointerstitial nephritis, glomerulonephritis, diabetic nephropathy, allograft rejection, amyloidosis, renovascular ischemia, reflux nephropathy, polycystic kidney disease, liver fibrosis/cirrhosis, autoimmune liver disease, Biliary atresia, acute and chronic Hepatitis B and C virus, Liver tumors and cancer, Alcoholic liver disease, Polycystic liver disease, Liver cholangiocarcinoma, neuromyelitis optica spectum disorder, cardiovascular disease, inflammation induced by bacterial or viral infection, inflammation associated with SARS-CoV-2 infection or its variants and Coronavirus Disease 2019 (COVID-19), acute respiratory distress syndrome, pneumonia, Long/Long-Term/Chronic COVID, PostAcute Sequelae of COVID-19 (PASC), myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS “Brain Fog”) and vasculitis.

As used herein the phrase “cancer associated condition” means any disease arising from the proliferation of malignant cancerous cells. Thus, in one embodiment, the method of present invention is provided to treat a cancer/tumor associated condition, such as adenoid cystic carcinoma, adrenal gland tumor, amyloidosis, anal cancer, appendix cancer, astrocytoma, ataxia-telangiectasia, beckwith-wiedemann syndrome, cholangiocarcinoma, birt-hogg-dube syndrome, bone cancer, brain stem glioma, brain tumor, breast cancer (inflammatory, metastatic, male), prostrate, basal cell, melanoma, colon, colorectal, bladder, kidney cancer, lacrimal gland cancer, laryngeal and hypopharyngeal cancer, lung cancer (non-small cell, small cell), leukemia (acute lymphoblastic, acute lymphocytic, acute myeloid, B cell prolymphocytic, chronic lymphocytic, chronic myeloid, chronic T cell lymphocytic, eosinophilic), Liver Cancer, Li-Fraumei syndrome, lymphoma (Hodgkin and non-hodgkin), lynch syndrome, mastocytosis, medulloblastoma, meningioma, mesothelioma, multiple endocrine neoplasia, multiple myeloma, MUTYH-associated polyposis, myelodyspastic syndrome, nasal cavity and paranasal sinus cancer, neurobastoma, neuroendocrine tyymors, neurofibromatosis, penile cancer, parathyroid cancer, ovarian fallopian tube and peritoneal cancer, osteosarcoma, pituitary gland tumor, pleupulmonary blastoma, oral and oropharyngeal , thyroid, uterine, pancreatic, carney complex, brain and spinal cord cancer, cervical cancer, cowden syndrome, craniopharyngioma, desmoid tumor, desmoplatic infantile ganglioglioma, ependymoma, esophageal cancer, ewing sarcoma, eye cancer, eyelid cancer, familial adenomatous polyposis, familial GIST, familial malignant melanoma, familial pancreatic cancer, gallbladder cancer, gastrointestinal stromal tumor, germ cell tumor, gestational trophoblastic disease, head and neck cancer, hereditary breast and ovarian cancer, hereditary diffuse gastric cancer, hereditary, leiomyomastosis and renal cell cancer, hereditary pancreatitis, herediatary papillary renal carcinoma, hereditary mixed polyposis syndrome, HIV/AIDS related cancers, retinoblastoma, rhabdomyosarcoma, salivary glanc cancer, Kaposi sarcoma, small bowel cancer, stomach cancer, testicular cancer, thymoma and thymic carcinoma, thyroid cancer, vaginal cancer, culver cancer, wemer syndrome and Xeroderma pigmentosum.

As used herein, the term “administration” refers to providing a compound, or a pharmaceutical composition comprising the compound as described herein. The compound or composition can be administered by another person to the subject or it can be self-administered by the subject. Non-limiting examples of routes of administration are oral, parenteral (e.g., intravenous), or topical.

As used herein, the term “treatment” refers to an intervention that ameliorates a sign or symptom of a disease or pathological condition. As used herein, the terms “treatment”, “treat” and “treating,” with reference to a disease, pathological condition or symptom, also refers to any observable beneficial effect of the treatment. The beneficial effect can be evidenced, for example, by a delayed onset of clinical symptoms of the disease in a susceptible subject, a reduction in severity of some or all clinical symptoms of the disease, a slower progression of the disease, a reduction in the number of relapses of the disease, an improvement in the overall health or well-being of the subject, or by other parameters well known in the art that are specific to the particular disease. A prophylactic treatment is a treatment administered to a subject who does not exhibit signs of a disease or exhibits only early signs, for the purpose of decreasing the risk of developing pathology. A therapeutic treatment is a treatment administered to a subject after signs and symptoms of the disease have developed.

As used herein, the term “subject” refers to an animal (e.g., a mammal, such as a human). A subject to be treated according to the methods described herein may be one who has been diagnosed with a MRGPRX2 or MRGPRX2 ortholog B2 dependent condition, such as a pseudo-allergic reaction, an itch associated condition, a pain associated condition, inflammatory or an autoimmune disorder. Diagnosis may be performed by any method or technique known in the art. One skilled in the art will understand that a subject to be treated according to the present disclosure may have been subjected to standard tests or may have been identified, without examination, as one at risk due to the presence of one or more risk factors associated with the disease or condition. The term “patient” may be used interchangeably with the term “subject.” A subject may refer to an adult or pediatric subject.

The Federal Food, Drug, and Cosmetic Act defines “pediatric” as a subject aged 21 or younger at the time of their diagnosis or treatment. Pediatric subpopulations are further characterized as: (i) neonates - from birth through the first 28 days of life; (ii) infants - from 29 days to less than 2 years; (iii) children - 2 years to less than 12 years; and (iv) adolescents - aged 12 through 21. Despite the definition, depending on the susceptible patient population and clinical trial evaluation, an approved regulatory label may include phrasing that specifically modifies the range of a pediatric population, such as, for example, pediatric patients up to 22 years of age.

In another embodiment, the method of treating a subject having a MRGPRX2 dependent condition (e.g., an itch associated condition, a pain associated condition, a pseudo-allergic reaction, or an inflammatory or autoimmune disorder) described herein further comprises administering to the subject a pharmaceutically effective amount of a second therapeutic agent. In one embodiment, the itch associated condition is a pseudo- allergic condition.

In one embodiment, the second therapeutic agent is an antihistamine, such as an Hl receptor antagonist or an H2 receptor antagonist. In one embodiment, the second therapeutic agent is an Hl receptor antagonist antihistamine, such as levocetirizine, loratadine, fexofenadine, cetirizine, desloratadine, olopatadine, diphenhydramine, cyproheptadine or hydroxyzine pamoate. In one embodiment, the second therapeutic agent is a H2 receptor antagonist, such as cimetidine, nizatidine, ranitidine or famotidine. In one embodiment, the second therapeutic agent is a leukotriene receptor antagonist or leukotriene synthesis inhibitor, such as montelukast, zafirlukast, pranlukast, or 5- lipoxygenase inhibitor (e.g., zileuton, hypericum perforatum). In one embodiment, the second therapeutic agent is an immunomodulatory agent such as Omalizumab or immunoglobulin therapy. In one embodiment, the second therapeutic agent is a corticosteroid, such as hydrocortisone, cortisone, ethamethasoneb, triamcinolone, prednisone, prednisolone, or fludrocortisone. In one embodiment, the second therapeutic agent is a tricylic antidepressant that can relieve itch such as doxepin, amitriptyline or nortriptyline. In one embodiment, the second therapeutic agent is an anti-inflammatory drug such as dapsone, sulfasalazine, hydroxycholoroquine or colchicine. In one embodiment, the second therapeutic agent is an immunosuppressant such as cyclosporine, methotrexate, mycophenolic acid or tacromilus.

The second therapeutic agent may be administered simultaneously, separately, or sequentially with the compounds of the present disclosure. If administered simultaneously, the second therapeutic agent and compound of the present disclosure may be administered in separate dosage forms or in the same dosage form.

In another embodiment, a method of treating a subject having an itch associated condition is provided, the method comprising administering to the subject a pharmaceutically effective amount of a compound having structure (I) or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, or a pharmaceutical composition thereof. In one embodiment, the itch associated condition is urticaria, pruritus, atopic dermatitis, dry skin, psoriasis, contact dermatitis, or eczema. In another embodiment, a method of treating a subject having an inflammation or autoimmune associated condition is provided, the method comprising administering to the subject a pharmaceutically effective amount of a compound having structure (I) or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, or a pharmaceutical composition thereof. In one embodiment, the inflammation or autoimmune associated condition is sinusitis, asthma, rosacea, or endometriosis.

In another embodiment, a method of treating a subject having a pain associated condition is provided, the method comprising administering to the subject a pharmaceutically effective amount of a compound having structure (I) or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, or a pharmaceutical composition thereof. In one embodiment, the pain associated condition is chronic pelvic pain syndrome, endometriosis pain, fibromyalgia, migraine or postoperative pain.

Compounds

As detailed above, the present disclosure provides compounds showing significant activity as MRGPRX2 antagonists. Accordingly, one embodiment provides a compound having the following structure (I): or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

R 1 is cycloalkyl, aryl, heterocyclyl, -(CH2) n Q, -CHQR, or -CQ(R)2, wherein R 1 is optionally substituted with one or more R ql ;

Q is Ci-6 alkyl, aryl, cycloalkyl, heterocyclyl, -CH2C(O)OR, -C(O)OR, - C(O)NHR, haloalkyl, -CN, -N(R) 2 , -N(R)C(O)R, -N(R)C(O)OR, or -N(R)S(O) 2 R, wherein Q is optionally substituted with one or more R q2 ;

R ql and R q2 are independently H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -O(CH2) n R, haloalkoxy, -C(O)OR, -C(O)R, - OC(O)R, halo, haloalkyl, -CN, -N(R) 2 , -N(R)C(O)R, -N(R)S(O) 2 R, S(O) 2 R, -C(H)Q’R, or -(CH2) n Q’ where Q’ is selected from Ci-6 alkyl, aryl, cycloalkyl, heterocyclyl, OR’, - C(O)OR’, -OC(O)R’, haloalkyl, -CN, -N(R’) 2 , -N(R’)C(O)R’, and -N(R’)S(O) 2 R’;

A is N or CR a ;

B is N or CR b ;

C is N or CR C ;

D is CR d ;

E is N or CR e ;

G is N or CR S ;

W is CR W ;

Y is N, S or CR y ;

Z is N, CR Z , S or O; each R is independently H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, alkylamino, - (CH2) n R’, halo, aryl, cycloalkyl, heteroaryl or heterocyclyl, or two R groups taken together with the atom to which they are attached form a carbocyle or heterocycle;

R’ is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, haloalkyl, aryl, cycloalkyl, heteroaryl, or heterocyclyl;

R a is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R) 2 , -N(R)C(O)R, - N(R)S(O) 2 R, or S(O) 2 R;

R b is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R) 2 , -N(R)C(O)R, - N(R)S(O) 2 R, or S(O) 2 R;

R c is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R) 2 , -N(R)C(O)R, - N(R)S(O) 2 R, or S(O) 2 R;

R d is either absent, or when present, H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, - CN, -N(R) 2 , -N(R)C(O)R, -N(R)S(O) 2 R, or S(O) 2 R;

R e is either absent, or when present, H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, - CN, -N(R) 2 , -N(R)C(O)R, -N(R)S(O) 2 R, or S(O) 2 R;

R s is either absent, or when present, H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, - CN, -N(R) 2 , -N(R)C(O)R, -N(R)S(O) 2 R, or S(O) 2 R;

R w is either absent, or when present, H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, - CN, -N(R) 2 , -N(R)C(O)R, -N(R)S(O) 2 R, or S(O) 2 R;

R y is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R) 2 , -N(R)C(O)R, - N(R)S(O) 2 R, or S(O) 2 R; R z is H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R) 2 , -N(R)C(O)R, - N(R)S(O) 2 R, or S(O) 2 R; n is independently 0, 1, 2, 3, 4 and 5;

R 6 is C1.4 alkyl, phenyl, -C(O)R, -(CH 2 ) n OR, -CN, F, Cl, Br, CF 3 , CF 2 H or CFH2, with the proviso that R is not H; each R 8 is independently H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, haloalkyl, -CN, -N(R)2, - N(R)C(O)R, -N(R)S(O) 2 R, or S(O) 2 R;

R 2 , R 3 , R 4 , R 5 and R 7 are the same or different and either absent or, when present, independently H, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, -OR, -C(O)OR, -OC(O)R, halo, -CN, -N(R) 2 , -N(R)C(O)R, -N(R)S(O) 2 R, or S(O) 2 R; wherein: adjacent atoms on either the “a” ring or “b” ring may be joined together by single bonds to form a 9 atom carbocyclic or heterocyclic ring structure; or atoms 1 and 2, 3 and 4, 5 and 6 and 8 and 9 on rings “a” ring and “b” ring may be joined together by double bonds to form an aromatic 9 atom carbocyclic or heterocyclic ring structure; or atoms 1 and 2, 3 and 4, 6 and 7 and 8 and 9 on rings “a” ring and “b” ring may be joined together by double bonds to form an aromatic 9 atom carbocyclic or heterocyclic ring structure; with the provisos that: when Z is S or N, then A and C cannot both be N; and when Z is N, then C and G cannot both be N.

In another embodiment, compounds are provided of the following structures (la) or (la’):

or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

In yet another embodiment, compounds are provided of the following structures (lb), (Ic) or (Id):

(lb) (Ic) (Id) or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

In one embodiment R 1 is aryl, -(CH2) n Q, -CHQR, or -CQ(R)2, wherein R 1 is optionally substituted with one or more R ql ;

Q is aryl, -CH 2 C(O)OR, -C(O)OR, -C(O)NHR, haloalkyl, -CN, -N(R) 2 , - N(R)C(O)R, -N(R)C(O)OR, or -N(R)S(O)2R, wherein Q is optionally substituted with one or more R q2 .

In another embodiment, R 1 is phenyl. In some embodiments, the aryl is unsubstituted.

In yet another embodiment, the aryl is substituted with one or more R ql . In yet other embodiments, R ql is H, -OR, -N(R)S(O) 2 R, -CN, -N(R)C(O)R or -C(H)Q’R.

In one embodiment, R 1 is heterocyclyl, -(CH2) n Q, -CHQR, or -CQ(R) 2 , wherein R 1 is optionally substituted with one or more R ql ;

Q is heterocyclyl, -CH 2 C(O)OR, -C(O)OR, -C(O)NHR, haloalkyl, -CN, -N(R) 2 , -N(R)C(O)R, -N(R)C(O)OR, or -N(R)S(O) 2 R, wherein Q is optionally substituted with one or more R q2 .

In some embodiments, the heterocyclyl is unsubstituted.

In yet another embodiment, the heterocyclyl is substituted with one or more R ql . In some embodiments, R ql is H or Ci-6 alkyl.

In one embodiment, R 6 is CF3, CF 2 H, CFH 2 , Ci-6 alkyl, -C(O)R, -(CH 2 ) n OR or - CN.

In another embodiment, R 6 is CF3.

In yet another embodiment, R 6 is CF 2 H.

In some embodiments, R 6 is Ci-6 alkyl.

In one embodiment, R 6 is -C(O)R. In other embodiments, R is Ci-6 alkyl.

In one embodiment, R 6 is -(CH 2 ) n OR. In another embodiment, n is 0. In yet another embodiment, n is 1. In yet other embodiments, R is Ci-6 alkyl.

In one embodiment, R 6 is -CN.

In other embodiments, each R 8 is H.

In one embodiment, Z is N or S.

In some embodiments, each of A, B, C, and Y are CH.

In one embodiment, E is N.

In some embodiments, R w is absent.

In one embodiment, atoms 1 and 2, 3 and 4, 5 and 6 and 8 and 9 on rings “a” ring and “b” ring are joined together by double bonds to form an aromatic 9 atom heterocyclic ring structure.

In other embodiments, atoms 1 and 2, 3 and 4, 6 and 7 and 8 and 9 on rings “a” ring and “b” ring are joined together by double bonds to form an aromatic 9 atom heterocyclic ring structure. In one embodiment, a compound is selected from any one of compounds listed in Table I, or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

Table I. Representative compounds having Structure (I)

Representative compounds of structure (I), as well as structures (la), (la’), (lb), (Ic) or (Id) as applicable, include, but not limited to, any one of the compounds listed below in their IUPAC names as well as a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.

4-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(lR, 3 S)-3 -{ [2-(tri fluoromethyl)- 1,3-benzothi azol-4- yl]amino}cyclohexyl]benzamide;

N-[(lR,3S)-3-{[2-(ethoxymethyl)-l-benzothiophen-4-yl]amin o}cyclohexyl]-4- methoxybenzamide;

N-[(lR,3S)-3-[(2-acetyl-l-benzothiophen-4-yl)amino]cycloh exyl]-4- methoxybenzamide;

4-fluoro-N-[(ls,4s)-4-[(2-acetyl-l-benzothiophen-4- yl)amino]cyclohexyl]benzamide;

4-fluoro-N-[(ls,4s)-4-{[2-(ethoxymethyl)-l-benzothiophen- 4- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(ls,4s)-4-{[l-methyl-2-(trifluoromethyl)-lH- indol-4- yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(lR,3S)-3-{[2-(difhioromethyl)-l-benzothiophen-4-yl]am ino}cyclohexyl]-4- methoxybenzamide;

N-[(lR,3S)-3-[(2-ethyl-l-benzothiophen-4-yl)amino]cyclohe xyl]-4- methoxybenzamide;

4-methoxy-N-[(lR,3S)-3-[(2-methyl-l-benzothiophen-4- yl)amino]cyclohexyl]benzamide;

4-fluoro-N-[(ls,4s)-4-[(2-methyl-l-benzothiophen-4- yl)amino]cyclohexyl]benzamide;

4-fluoro-N-[(ls,4s)-4-[(2-ethyl-l-benzothiophen-4- yl)amino]cyclohexyl]benzamide;

4-fluoro-N-[( 1 s,4s)-4- { [2-(difluorom ethyl)- 1 -benzothiophen-4- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(lR,3S)-3 -[(2 -methoxy- 1-benzothi ophen-4- yl)amino]cyclohexyl]benzamide; 4-nuoro-N-[(l s,4s)-4-[(2 -methoxy- 1-benzothi ophen-4- yl)amino]cyclohexyl]benzamide;

N-[(lR,3S)-3-[(2-cyano- 1-benzothi ophen-4-yl)amino]cyclohexyl]-4- methoxybenzamide;

4-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l,3-benzothi azol-4- yl ] amino } cy cl ohexyl ]b enzami de ;

4-fluoro-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l-benzothioph en-7- yl ] amino } cy cl ohexyl ]b enzami de ;

4-fluoro-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l,3-benzothia zol-7- yl ] amino } cy cl ohexyl ]b enzami de ;

4-fluoro-N-[(ls,4s)-4-[(2-cyano-l-benzothiophen-4- yl)amino]cyclohexyl]benzamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)- 1-benzothi ophen-4- yl]amino}cyclohexyl]pyrazolo[l,5-a]pyridine-2-carboxamide;

3-acetamido-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l-benzothi ophen-4- yl ] amino } cy cl ohexyl ]b enzami de ;

1 -ethyl -N-[(ls,4s)-4-{ [2-(trifluorom ethyl)- 1 -benzothiophen-4- yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide;

1 -methyl -N-[(l s,4s)-4-{ [2-(trifluorom ethyl)- 1 -benzothiophen-4- yl]amino}cyclohexyl]-l,2,3,4-tetrahydroquinoline-6-carboxami de;

3-(l-cyanoethyl)-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l-ben zothiophen-4- yl ] amino } cy cl ohexyl ]b enzami de ;

3-methanesulfonamido-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l -benzothiophen-4- yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)- 1-benzothi ophen-4- yl]amino}cyclohexyl]imidazo[l,2-a]pyridine-6-carboxamide;

4-fluoro-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l-benzothioph en-4- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(lR,3S)-3-{[2-(trifluoromethyl)-l-benzothiop hen-4- yl ] amino } cy cl ohexyl ]b enzami de ; 4-methoxy-N-[(lR,3S)-3-{[2-(tnnuoromethyl)-l-benzothiophen-7 - yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(lR,3S)-3-{[2-(trifluoromethyl)-l,3-benzoxaz ol-4- yl ] amino } cy cl ohexyl ]b enzami de ;

3-cyano-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l-benzothiophe n-4- yl ] amino } cy cl ohexyl ]b enzami de ;

2-amino-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]-l,3-thiazole-4-carboxamide;

1 -methyl -N-[(ls,4s)-4-{ [2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-l,3-benzothiazole-7-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]imidazo[l,2-a]pyridine-6-carboxamide;

4-(N-methylmethanesulfonamido)-N-[(ls,4s)-4-{[2-

(trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohe xyl]benzamide;

3-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-fluoro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-(dimethylamino)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imida zo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-lH-indole-4-carboxamide;

4-(dimethylamino)-2-fluoro-N-[( 1 s,4s)-4-{ [2-(trifluorom ethyl )imidazo[ 1 ,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

4-(morpholin-4-yl)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-(dimethylamino)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imida zo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ; 4-(propane-2-sulfonamido)-N-[( 1 s,4s)-4-{ [2-(tnnuoromethyl)imidazo[ 1 ,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-lH-indole-3-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-4-(lH-l,2,3,4-tetrazol-l-yl)benzamide;

4-methoxy-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(ls,4s)-4-({2-phenylimidazo[l,2-a]pyridin-5- yl } amino)cy cl ohexyl ]b enzami de ;

3-(propane-2-sulfonamido)-N-[(ls,4s)-4-{[2-(trifluorometh yl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5- yl } amino)cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)-2H-indazol-4 - yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl ] amino } cy cl ohexyl ]b enzami de ;

2-chloro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]benzamide;

2-cyano-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2-(methylamino)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo [l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-chloro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]pyridine-2-carboxamide; N-[(ls,4s)-4-{[2-(tnnuoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide;

2-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]pyridine-4-carboxamide;

2-amino-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ] pyrimi dine- 5 -carb oxami de;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-4-sulfamoylbenzamide;

4-methanesulfonyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imida zo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-2H-l,2,3-triazole-4-carboxamide; l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyr idin-5- yl]amino}cyclohexyl]-lH-l,2,3-triazole-4-carboxamide;

4-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)pyrazolo[l,5- a]pyridin-4- yl ] amino } cy cl ohexyl ]b enzami de ;

3-cyano-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-(methylamino)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo [l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-methanesulfonamido-N-[(ls,4s)-4-{[2-(trifluoromethyl)im idazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

4-chloro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-cyano-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-(methylamino)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo [l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methanesulfonamido-N-[(ls,4s)-4-{[2-(trifluoromethyl)im idazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ; N-[(ls,4s)-4-{[2-(tnnuoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]pyridazine-4-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]pyridine-4-carboxamide;

4-(4-methylpiperazin-l-yl)-N-[(ls,4s)-4-{[2-(trifluoromet hyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

2-chloro-4-methanesulfonyl-N-[(ls,4s)-4-{[2-(trifluoromet hyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

3-chloro-4-(propan-2-yloxy)-N-[(ls,4s)-4-{[2-(trifluorome thyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

6-(methylamino)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo [l,2-a]pyridin-5- yl]amino}cyclohexyl]pyridine-2-carboxamide;

3-(l-cyanoethyl)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidaz o[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-acetamido-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2 -a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-ethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2-acetamido-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2 -a]pyridin-5- yl]amino}cyclohexyl]pyridine-4-carboxamide;

4-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-ethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ]b enzami de ; methyl 4-{[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ] carb amoyl } b enzoate ;

4-acetamido-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2 -a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ; 2-ethyl-N-[(ls,4s)-4-{[2-(tnfluoromethyl)imidazo[l,2-a]pyndi n-5- yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-2H-indazole-4-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-2H-indazole-3-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-2H-indazole-7-carboxamide;

2-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-2H-indazole-3-carboxamide;

5-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-l,3-benzodiazole-6-carboxamide;

5,7-dimethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l, 2-a]pyridin-5- yl]amino}cyclohexyl]pyrazolo[l,5-a]pyrimidine-3-carboxamide;

N-(4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl)imidazo[l,5-a]pyridine-7-carboxamide;

2-methyl-N-(4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin- 5- yl]amino}cyclohexyl)pyrazolo[l,5-a]pyrimidine-6-carboxamide;

N-(4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl)imidazo[l,2-a]pyridine-6-carboxamide; l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyr idin-5- yl]amino}cyclohexyl]-lH-imidazole-5-carboxamide;

1 -methyl -N-[(ls,4s)-4-{ [2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide; l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyr idin-5- yl]amino}cyclohexyl]-lH-l,2,3-triazole-5-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-4-(4H-l,2,4-triazol-4-yl)benzamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]imidazo[l,5-a]pyridine-6-carboxamide; N-[(ls,4s)-4-{[2-(tnnuoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]imidazo[l,2-a]pyridine-7-carboxamide;

1-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-l,3-benzodiazole-6-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-[l,2,4]triazolo[4,3-a]pyridine-7-carbox amide;

4-fluoro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-2H-indazole-5-carboxamide;

6-chloro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]imidazo[l,2-b]pyridazine-2-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-4H,5H,6H,7H-pyrazolo[l,5-a]pyridine-2-c arboxamide;

7-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]-l-benzofuran-2-carboxamide;

5-fluoro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-l-benzofuran-2-carboxamide;

N-[(l s,4s)-4-{ [2-(trifluoromethyl)imidazo[ 1 ,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-imidazo[4,5-b]pyridine-7-carboxamide ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-7H-pyrrolo[2,3-d]pyrimidine-4-carboxami de;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-lH-pyrrolo[2,3-b]pyridine-4-carboxamide ;

4-fluoro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-2H-indazole-3-carboxamide;

2,3-dioxo-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]-2,3-dihydro-lH-indole-7-carboxamide;

2-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-l,3-benzothiazole-7-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-l,3-benzothiazole-4-carboxamide; 4-nuoro-N-[(ls,4s)-4-{[2-(tnfluoromethyl)imidazo[l,2-a]pyndi n-5- yl]amino}cyclohexyl]-lH-indole-3-carboxamide; l,5-dimethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

1 -ethyl -N-[(ls,4s)-4-{ [2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide; l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyr idin-5- yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-3 -carb oxami de;

1 -ethyl -N-[(ls,4s)-4-{ [2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide;

3-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ] - 1 , 2-oxazol e- 5 -carb oxami de;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-l-(2,2,2-trifluoroethyl)-lH-pyrazole-5- carboxamide;

1-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-pyrrole-2-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-l-(2,2,2-trifluoroethyl)-lH-pyrazole-4- carboxamide;

2-hydroxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]pyridine-4-carboxamide;

2-(dimethylamino)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imida zo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-l,3-benzodiazole-2-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]imidazo[l,2-a]pyrazine-2-carboxamide;

6-chloro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]quinoline-8-carboxamide; 6-chloro-N-[(ls,4s)-4-{[2-(tnnuoromethyl)imidazo[l,2-a]pyrid in-5- yl]amino}cyclohexyl]-2H-chromene-3-carboxamide;

5-fluoro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-pyrrolo[2,3-b]pyridine-2-carboxamide ;

6-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-l-benzothiophene-2-carboxamide;

3-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-l-benzofuran-2-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]pyrazolo[l,5-a]pyridine-2-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-[l,2,4]triazolo[l,5-a]pyridine-6-carbox amide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-[l,2,4]triazolo[4,3-a]pyridine-6-carbox amide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]imidazo[l,2-a]pyrimidine-6-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-lH-pyrrolo[3,2-c]pyridine-4-carboxamide ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-2H-pyrazolo[4,3-b]pyridine-7-carboxamid e;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]imidazo[l,2-a]pyrimidine-3-carboxamide; l,3-dimethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide; l-(propan-2-yl)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l, 2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide; l-phenyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyr idin-5- yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide; N-[(ls,4s)-4-{[2-(tnnuoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-2H-pyrazolo[4,3-c]pyridine-7-carboxamid e;

4-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l,3-benzothi azol-7- yl ] amino } cy cl ohexyl ]b enzami de ;

2-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide;

6-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide;

2,6-dimethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l, 2-a]pyridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide;

4-methoxy-N-[(ls,4s)-4-{[7-methyl-2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)-l,3-benzoxaz ol-7- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(ls,4s)-4-{[3-chloro-2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(ls,4s)-4-{[7-chloro-2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-lH-imidazole-5-carboxamide;

3-methoxy-l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide;

3-methoxy-l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

3-cyano-l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidaz o[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

5-amino-l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidaz o[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

3-(difluoromethyl)-l-methyl-N-[(ls,4s)-4-{[2-(trifluorome thyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de; 5-chloro-N-[(ls,4s)-4-{[2-(tnnuoromethyl)imidazo[l,2-a]pyrid in-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-5-(trifluoromethyl)-lH-pyrazole-4-carbo xamide;

3-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-pyrrolo[2,3-b]pyridine-4-carboxamide ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-lH-pyrrolo[2,3-b]pyridine-2-carboxamide ;

3-[(2,2-difluoroethyl)amino]-N-[(ls,4s)-4-{[2-(trifluorom ethyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

3-[(2,2-difluoroethyl)(methyl)amino]-N-[(ls,4s)-4-{[2-

(trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohe xyl]benzamide;

4-(difluoromethyl)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2-ethoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]pyridine-4-carboxamide;

2-(propan-2-yloxy)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl]amino}cyclohexyl]pyridine-4-carboxamide;

4-[(2,2-difluoroethyl)amino]-N-[(ls,4s)-4-{[2-(trifluorom ethyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

4-[(2,2-difluoroethyl)(methyl)amino]-N-[(ls,4s)-4-{[2-

(trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohe xyl]benzamide;

2-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-pyrrolo[2,3-b]pyridine-4-carboxamide ;

1 -methyl -N-[(ls,4s)-4-{ [2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrrolo[2,3-b]pyridine-4-carboxamide ;

3-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-2H-indazole-7-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-2H-pyrazolo[3,4-b]pyridine-4-carboxamid e; 4-chloro-3-methyl-N-[(ls,4s)-4-{[2-(tnfluoromethyl)imidazo[l ,2-a]pyndin-5- yl] amino } cyclohexyl ]benzami de;

5-(methylamino)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo [l,2-a]pyridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide;

2-[(propan-2-yl)amino]-N-[(ls,4s)-4-{[2-(trifluoromethyl) imidazo[l,2- a]pyridin-5-yl]amino}cyclohexyl]pyridine-4-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-lH-pyrrolo[2,3-c]pyridine-4-carboxamide ;

2-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl ] amino } cy cl ohexyl ] pyrimi dine- 5 -carb oxami de; l,2-dimethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]-lH-imidazole-5-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl ] amino } cy cl ohexyl ] - 1 , 2-oxazol e- 5 -carb oxami de;

4-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ] - 1 , 2-oxazol e- 5 -carb oxami de; l-ethyl-3-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l ,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide; l-ethyl-3-(propan-2-yl)-N-[(ls,4s)-4-{[2-(trifluoromethyl)im idazo[l,2- a]pyridin-5-yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide; methyl l-ethyl-5-{[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyr idin-5- yl]amino}cyclohexyl]carbamoyl}-lH-pyrazole-3-carboxylate; methyl 1 -ethyl-3 -{ [( 1 s,4s)-4- { [2-(trifluoromethyl)imidazo[ 1 ,2-a]pyridin-5- yl ] amino } cy cl ohexyl ] carb amoyl } - 1 H-pyrazol e- 5 -carb oxyl ate; l-(2,2-difluoroethyl)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide; l-(propan-2-yl)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l, 2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

5-(difluoromethyl)-l-methyl-N-[(ls,4s)-4-{[2-(trifluorome thyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de; 5-ethyl-N-[(ls,4s)-4-{[2-(tnfluoromethyl)imidazo[l,2-a]pyndi n-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide; l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyr idin-5- yl]amino}cyclohexyl]-3-(trifluoromethyl)-lH-pyrazole-4-carbo xamide;

5-cyclopropyl- 1 -methyl -N-[( 1 s,4s)-4- { [2-(trifluoromethyl)imidazo[ 1 ,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

1-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-5-(trifluoromethyl)-lH-pyrazole-4-carbo xamide;

3-chloro-l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imida zo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

5-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

3-amino-l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidaz o[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

3-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-l,2-thiazole-4-carboxamide;

5-amino-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

5-cyano-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

3-ethyl-l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidaz o[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

4-methoxy-N-[(ls,4s)-4-{[6-chloro-2-(trifluoromethyl)imid azo[l,2-a]pyri din-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2-[(2,2-difluoroethyl)amino]-N-[(ls,4s)-4-{[2-(trifluorom ethyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-2-[(2,2,2-trifluoroethyl)amino]benzamid e;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-3-[(2,2,2-trifluoroethyl)amino]benzamid e; N-[(l s,4s)-4-{ [2-(tnnuoromethyl)imidazo[ 1 ,2-a]pyndin-5- yl]amino}cyclohexyl]-4-[(2,2,2-trifluoroethyl)amino]benzamid e;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-l,3-benzoxazole-7-carboxamide;

2-(ethylamino)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[ l,2-a]pyridin-5- yl]amino}cyclohexyl]pyridine-4-carboxamide; l,3-diethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide; l,5-diethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-3 -carb oxami de;

5-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

4-methoxy-N-[(ls,4s)-4-{[8-methyl-2-(trifluoromethyl)imid azo[l,2-a]pyri din-5- yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-l,2-benzothiazole-7-carboxamide;

3-(difluoromethyl)-l-methyl-N-[(ls,4s)-4-{[2-(trifluorome thyl)imidazo[l,2- a]pyridin-5-yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide;

5-cyclopropyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l ,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

4-methoxy-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyrimidin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(lR,3S)-3-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(lR,3S)-3-{[7-methyl-2-(trifluoromethyl)imid azo[l,2-a]pyridin- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(lR,3S)-3-{[6-chloro-2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ] -4-methoxyb enzami de;

N-[(lR,3S)-3-{[6-fluoro-2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ] -4-methoxyb enzami de; 4-methoxy-N-[(lR,3S)-3-{[8-methyl-2-(trinuoromethyl)imidazo[ l,2-a]pyndin- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(lR,3S)-3-{[7-chloro-2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ] -4-methoxyb enzami de;

N-[(lR,3S)-3-{[8-chloro-2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ] -4-methoxyb enzami de;

N-[(lR,3S)-3-{[8-fluoro-2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ] -4-methoxyb enzami de;

6-[(2,2-difluoroethyl)(methyl)amino]-N-[(ls,4s)-4-{[2-

(trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohe xyl]pyridine-3- carboxamide;

3-amino-l-(2,2-difluoroethyl)-N-[(ls,4s)-4-{[2-(trifluoro methyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

3-amino-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]-l-(2,2,2-trifluoroethyl)-lH-pyrazole-4- carboxamide;

N-[(lR,3S)-3-{[7-fluoro-2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ] -4-methoxyb enzami de; l-(2,2-difluoroethyl)-3-(difluoromethyl)-N-[(ls,4s)-4-{[2-

(trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohe xyl]-lH-pyrazole-4- carboxamide;

3-(difluoromethyl)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-l-(2,2,2-trifluoroethyl)-lH-pyrazole-4- carboxamide; l-(2,2-difluoroethyl)-5-(difluoromethyl)-N-[(ls,4s)-4-{[2-

(trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohe xyl]-lH-pyrazole-4- carboxamide;

3 -chloro- 1 -(2-fluoroethyl)-N-[(l s,4s)-4- { [2-(trifluorom ethyl )imidazo[ 1 ,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

3-chloro-l-(2,2-difluoroethyl)-N-[(ls,4s)-4-{[2-(trifluor omethyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

3-chloro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-l-(2,2,2-trifluoroethyl)-lH-pyrazole-4- carboxamide; 4-methoxy-N-[(ls,4s)-4-{[7-fluoro-2-(trmuoromethyl)imidazo[l ,2-a]pyndin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methoxy-N-[(ls,4s)-4-{[3-fluoro-2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-cyclopropyl-l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl) imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

2-chloro-4-(difluoromethyl)-N-[(ls,4s)-4-{[2-(trifluorome thyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

3-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-2H-pyrazolo[4,3-b]pyridine-7-carboxamid e;

3-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-methoxy-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a] pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-(dimethylamino)-N-[(ls,4s)-4-{[2-(difluoromethyl)imidaz o[l,2-a]pyridin-5- yl]amino}cyclohexyl]benzamide;

4-[(2,2-difluoroethyl)(methyl)amino]-N-[(ls,4s)-4-{[2-

(difluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohex yl]benzamide;

4-(difluoromethyl)-N-[(ls,4s)-4-{[2-(difluoromethyl)imida zo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]-l,3-benzothiazole-7-carboxamide;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-2H-indazole-7-carboxamide;

3-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]-2H-indazole-7-carboxamide;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrrolo[2,3-b]pyridine-4-carboxamide ; l,3-dimethyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide; 2,4-dichloro-N-[(ls,4s)-4-{[2-(dinuoromethyl)imidazo[l,2-a]p yndin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2-[(2,2-difluoroethyl)amino]-N-[(ls,4s)-4-{[2-(difluorome thyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-2-[(2,2,2-trifluoroethyl)amino]benzamid e;

2-(methylamino)-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[ l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3-chloro-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-chloro-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]benzamide; l-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyri din-5- yl]amino}cyclohexyl]-lH-pyrrolo[2,3-b]pyridine-4-carboxamide ;

5-fluoro-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]-l-benzofuran-2-carboxamide;

3-chloro-l-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidaz o[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

3-(difluoromethyl)-l-methyl-N-[(ls,4s)-4-{[2-(difluoromet hyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de; l-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyri din-5- yl]amino}cyclohexyl]-3-(trifluoromethyl)-lH-pyrazole-4-carbo xamide;

3-ethyl-l-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo [l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

3-[(2,2-difluoroethyl)amino]-l-methyl-N-[(ls,4s)-4-{[2-

(difluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohex yl]-lH-pyrazole-4- carboxamide; l-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyri din-5- yl]amino}cyclohexyl]-lH-pyrrole-3-carboxamide; 1-methyl-N-[(ls,4s)-4-{[2-(dinuoromethyl)imidazo[l,2-a]pyndi n-5- yl]amino}cyclohexyl]-lH-pyrazole-5-carboxamide;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-l,3-thiazole-5-carboxamide;

6-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide;

2-amino-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]py ridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide;

6-ethyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]py ridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide;

6-methoxy-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide;

4-(2-fluoroethoxy)-N-[(ls,4s)-4-{[2-(difluoromethyl)imida zo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2-chloro-4-methoxy-N-[(ls,4s)-4-{[2-(difluoromethyl)imida zo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2-chloro-4-fluoro-N-[(ls,4s)-4-{[2-(difluoromethyl)imidaz o[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-fluoro-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-2H-pyrazolo[4,3-b]pyridine-7-carboxamid e;

5-[(2,2-difluoroethyl)amino]-N-[(ls,4s)-4-{[2-(difluorome thyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

6-fluoro-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]pyridine-3-carboxamide; l-propyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyri din-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide; 1-(propan-2-yl)-N-[(ls,4s)-4-{[2-(dinuoromethyl)imidazo[l,2- a]pyndin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

4-methoxy-3-(methylamino)-N-[(ls,4s)-4-{[2-(difluoromethy l)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

3-chloro-l-(2-fluoro-2-methylpropyl)-N-[(ls,4s)-4-{[2-

(difluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohex yl]-lH-pyrazole-4- carboxamide;

4-(dimethylamino)-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyr idin-5- yl } amino)cy cl ohexyl ]b enzami de ;

4-(methylamino)-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyrid in-5- yl } amino)cy cl ohexyl ]b enzami de ;

3-methoxy-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5- yl } amino)cy cl ohexyl ]b enzami de ;

4-fluoro-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5- yl } amino)cy cl ohexyl ]b enzami de ;

2-(methylamino)-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyrid in-5- yl } amino)cy cl ohexyl ]b enzami de ;

4-methyl-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5- yl } amino)cy cl ohexyl ]b enzami de ;

3-chloro-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5- yl } amino)cy cl ohexyl ]b enzami de ;

4-chloro-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5- yl } amino)cy cl ohexyl ]b enzami de ;

3-methyl-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5- yl } amino)cy cl ohexyl ]b enzami de ;

3-methyl-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5- yl}amino)cyclohexyl]-2H-indazole-7-carboxamide;

5-fluoro-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5-yl }amino)cyclohexyl]- 1 -benzofuran-2-carboxamide; l,3-dimethyl-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyridin-5- yl}amino)cyclohexyl]-lH-pyrazole-4-carboxamide; l-(2,2-dmuoroethyl)-3-fluoro-N-[(ls,4s)-4-{[2-(tnnuoromethyl )imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

6-[(2,2-difluoroethyl)(methyl)amino]-N-[(ls,4s)-4-{[2-

(difluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohex yl]pyridine-3- carboxamide;

4-(methylamino)-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[ l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ; l,4-dimethyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-pyrrole-3-carboxamide; l,3-bis(difluoromethyl)-N-[(ls,4s)-4-{[2-(difluoromethyl)imi dazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

3-chloro-l-(2-hydroxy-2-methylpropyl)-N-[(ls,4s)-4-{[2-

(difluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohex yl]-lH-pyrazole-4- carboxamide;

3-chloro-l-methyl-N-[(ls,4s)-4-({2-cyanoimidazo[l,2-a]pyr idin-5- yl}amino)cyclohexyl]-lH-pyrazole-4-carboxamide;

3 -chi oro-1 -(difluoromethyl)-N-[( Is, 4s)-4-{[2-(difluorom ethyl )imidazo[ 1,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

5-fluoro-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]pyrazolo[l,5-a]pyridine-2-carboxamide;

1-(difluoromethyl)-5-methyl-N-[(ls,4s)-4-{[2-(difluoromet hyl)imidazo[l,2- a]pyridin-5-yl]amino}cyclohexyl]-lH-pyrrole-3-carboxamide;

2-[(2-fluoroethyl)amino]-N-[(ls,4s)-4-{[2-(difluoromethyl )imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

3-[(2-fluoroethyl)amino]-N-[(ls,4s)-4-{[2-(difluoromethyl )imidazo[l,2- a]pyridin-5-yl]amino}cyclohexyl]benzamide;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-4-[(2,2,2-trifluoroethyl)amino]benzamid e;

4-[(2,2-difluoroethyl)amino]-N-[(ls,4s)-4-{[2-(difluorome thyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ; 6-[(2,2-dmuoroethyl)amino]-N-[(ls,4s)-4-{[2-(dmuoromethyl)im idazo[l,2- a]pyridin-5-yl]amino}cyclohexyl]pyridine-2-carboxamide;

2-[(2,2-difluoroethyl)amino]-N-[(ls,4s)-4-{[2-(difluorome thyl)imidazo[l,2- a]pyridin-5-yl]amino}cyclohexyl]pyridine-4-carboxamide;

2-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino}cyclohexyl]-lH-imidazo[4,5-b]pyridine-7-carboxamide ;

3-cyano-l-methyl-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo [l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]imidazo[l,2-a]pyridine-6-carboxamide;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]pyrazolo[l,5-a]pyridine-2-carboxamide;

3-fluoro-4-methoxy-N-[(ls,4s)-4-{[2-(difluoromethyl)imida zo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

2.4-dimethoxy-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l, 2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

3.4-dimethoxy-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l, 2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-[(2-fluoroethyl)amino]-N-[(ls,4s)-4-{[2-(difluoromethyl )imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

3-[(2,2-difluoroethyl)amino]-N-[(ls,4s)-4-{[2-(difluorome thyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ]b enzami de ;

2-(difluoromethyl)-N-[(ls,4s)-4-{[2-(difluoromethyl)imida zo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-l,3-benzodiazole-7-carboxamide;

3-[(2,2-difluoroethyl)amino]-N-[(ls,4s)-4-{[2-(difluorome thyl)imidazo[l,2- a]pyridin-5-yl]amino}cyclohexyl]pyrazolo[l,5-a]pyridine-2-ca rboxamide; l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyr idin-5- yl]amino}cyclohexyl]-lH-indole-3-carboxamide;

5-[(2,2-difluoroethyl)amino]-l-methyl-N-[(ls,4s)-4-{[2- (difluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohexyl] -lH-pyrazole-4- carboxamide; 2-nuoro-4-methoxy-N-[(ls,4s)-4-{[2-(dmuoromethyl)imidazo[l,2 -a]pyndin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

1.2-dimethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l, 2-a]pyridin-5- yl]amino}cyclohexyl]-lH-indole-3-carboxamide;

2-chloro-4-[(2,2-difluoroethyl)(methyl)amino]-N-[(ls,4s)- 4-{[2- (trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohexyl ]benzamide;

2-chloro-4-[methyl(2,2,2-trifluoroethyl)amino]-N-[(ls,4s) -4-{[2- (trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino}cyclohexyl ]benzamide;

4-methoxy-N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-2-[(2,2,2-trifluoroethyl)amino]benzamid e;

N-[(ls,4s)-4-{[2-(difluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]quinoline-8-carboxamide;

3-(3,3-difluoropropyl)-l-methyl-N-[(ls,4s)-4-{[2-(difluor omethyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

1.3-bis(difluoromethyl)-N-[(ls,4s)-4-{[2-(trifluoromethyl )imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

3-cyano-l-(difluoromethyl)-N-[(ls,4s)-4-{[2-(trifluoromet hyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

2.2.3.3-tetramethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imi dazo[l,2-a]pyridin-5- yl]amino}cyclohexyl]cyclopropane-l-carboxamide;

2-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-2H-indazole-7-carboxamide; l-methyl-N-(4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino}cyclohexyl)-lH-indole-3-carboxamide;

N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5 - yl]amino}cyclohexyl]-l,2-benzoxazole-3-carboxamide; l,2-dimethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]-lH-indole-4-carboxamide; l,2-dimethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]-lH-l,3-benzodiazole-4-carboxamide; 3-chloro-N-[(ls,4s)-4-{[2-(dmuoromethyl)imidazo[l,2-a]pyndin -5- yl]amino}cyclohexyl]-l-(trifluoromethyl)-lH-pyrazole-4-carbo xamide;

3-chloro-l-(difluoromethyl)-N-[(ls,4s)-4-{[2-(trifluorome thyl)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

4-methoxy-N-[(ls,4s)-4-{[8-fluoro-2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ;

4-chloro-l-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imida zo[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-indole-3-carboxamide;

5-chloro-l-ethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidaz o[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

3-chloro-l-ethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidaz o[l,2-a]pyridin-5- yl]amino}cyclohexyl]-lH-pyrazole-4-carboxamide;

3-chloro-l-(propan-2-yl)-N-[(ls,4s)-4-{[2-(trifluoromethy l)imidazo[l,2- a] pyri din- 5 -yl ] amino } cy cl ohexyl ] - 1 H-pyrazol e-4 -carb oxami de;

2-methyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-lH-l,3-benzodiazole-7-carboxamide;

3-fluoro-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a] pyridin-5- yl]amino}cyclohexyl]-2H-indazole-7-carboxamide;

4-(2-fluoroethoxy)-N-[(ls,4s)-4-{[2-(trifluoromethyl)imid azo[l,2-a]pyridin-5- yl ] amino } cy cl ohexyl ]b enzami de ; and l,5-dimethyl-N-[(ls,4s)-4-{[2-(trifluoromethyl)imidazo[l,2-a ]pyridin-5- yl]amino}cyclohexyl]-lH-pyrrolo[3,2-b]pyridine-3-carboxamide .

Pharmaceutical Compositions

In certain embodiments, the invention provides a pharmaceutical composition comprising a compound of structure (I) or any of the structures (la), (la’), (lb), (Ic) or (Id), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, together with at least one pharmaceutically acceptable carrier, diluent, or excipient. For example, the active compound will usually be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier which can be in the form of an ampoule, capsule, sachet, paper, or other container. When the active compound is mixed with a carrier, or when the carrier serves as a diluent, it can be solid, semi-solid, or liquid material that acts as a vehicle, excipient, or medium for the active compound. The active compound can be adsorbed on a granular solid carrier, for example contained in a sachet. Some examples of suitable carriers are water, salt solutions, alcohols, polyethylene glycols, polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatin, lactose, terra alba, sucrose, dextrin, magnesium carbonate, sugar, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid, or lower alkyl ethers of cellulose, silicic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose, and polyvinylpyrrolidone. Similarly, the carrier or diluent can include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax.

As used herein, the term “pharmaceutical composition” refers to a composition containing one or more of the compounds described herein, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, formulated with a pharmaceutically acceptable carrier, which can also include other additives, and manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal. Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); for administration to a pediatric subject e.g., solution, syrup, suspension, elixir, powder for reconstitution as suspension or solution, dispersible/effervescent tablet, chewable tablet, lollipop, freezer pops, troches, oral thin strips, orally disintegrating tablet, orally disintegrating strip, and sprinkle oral powder or granules); or in any other formulation described herein. Conventional procedures and ingredients for the selection and preparation of suitable formulations are described, for example, in Remington: The Science and Practice of Pharmacy, 21st Ed., Gennaro, Ed., Lippencott Williams & Wilkins (2005) and in The United States Pharmacopeia: The National Formulary (USP 36 NF31 ), publi shed in 2013. In some embodiments, the pharmaceutical composition comprising a compound of structure (I) or any of the structures (la), (la’), (lb), (Ic) or (Id), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, with at least one pharmaceutically acceptable carrier, diluent, or excipient further comprises a second therapeutic agent.

In one embodiment, the second therapeutic agent is an antihistamine, such as an Hl receptor antagonist or an H2 receptor antagonist. In one embodiment, the second therapeutic agent is an Hl receptor antagonist antihistamine, such as levocetirizine, loratadine, fexofenadine, cetirizine, desloratadine, olopatadine, diphenhydramine, cyproheptadine or hydroxyzine pamoate. In one embodiment, the second therapeutic agent is a H2 receptor antagonist, such as cimetidine, nizatidine, ranitidine or famotidine. In one embodiment, the second therapeutic agent is a leukotriene receptor antagonist or leukotriene synthesis inhibitor, such as montelukast, zafirlukast, pranlukast, or 5- lipoxygenase inhibitor (e.g., zileuton, hypericum perforatum). In one embodiment, the second therapeutic agent is an immunomodulatory agent such as Omalizumab or immunoglobulin therapy. In one embodiment, the second therapeutic agent is a corticosteroid, such as hydrocortisone, cortisone, ethamethasoneb, triamcinolone, prednisone, prednisolone, or fludrocortisone. In one embodiment, the second therapeutic agent is a tricylic antidepressant that can relieve itch such as doxepin, amitriptyline or nortriptyline. In one embodiment, the second therapeutic agent is an anti-inflammatory drug such as dapsone, sulfasalazine, hydroxycholoroquine or colchicine. In one embodiment, the second therapeutic agent is an immunosuppressant such as cyclosporine, methotrexate, mycophenolic acid or tacromilus.

In one embodiment, the second therapeutic agent is an Hl receptor antagonist antihistamine, such as levocetirizine, loratadine, fexofenadine, cetirizine, desloratadine, olopatadine, diphenhydramine, cyproheptadine or hydroxyzine pamoate. In one embodiment, the second therapeutic agent is a H2 receptor antagonist, such as cimetidine, nizatidine, ranitidine or famotidine. In one embodiment, the second therapeutic agent is a leukotriene receptor antagonist or leukotriene synthesis inhibitor, such as montelukast, zafirlukast, pranlukast, or 5 -lipoxygenase inhibitor (e.g., zileuton, hypericum perforatum). In one embodiment, the second therapeutic agent is an immunomodulatory agent such as Omalizumab or immunoglobulin therapy. In one embodiment, the second therapeutic agent is a corticosteroid, such as hydrocortisone, cortisone, ethamethasoneb, triamcinolone, prednisone, prednisolone, or fludrocortisone. In one embodiment, the second therapeutic agent is a tricylic antidepressant that can relieve itch such as doxepin, amitriptyline or nortriptyline. In one embodiment, the second therapeutic agent is an anti-inflammatory drug such as dapsone, sulfasalazine, hydroxycholoroquine or colchicine. In one embodiment, the second therapeutic agent is an immunosuppressant such as cyclosporine, methotrexate, mycophenolic acid or tacromilus.

As used herein, the term “pharmaceutically acceptable carrier” refers to any ingredient other than the disclosed compounds, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof (e.g., a carrier capable of suspending or dissolving the active compound) and having the properties of being nontoxic and noninflammatory in a patient. Excipients may include, for example: anti adherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspending or dispersing agents, sweeteners, or waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.

The formulations can be mixed with auxiliary agents which do not deleteriously react with the active compounds. Such additives can include wetting agents, emulsifying and suspending agents, salt for influencing osmotic pressure, buffers and/or coloring substances, preserving agents, sweetening agents, or flavoring agents. The compositions can also be sterilized if desired. The route of administration can be any route which effectively transports the active compound of the invention to the appropriate or desired site of action, such as oral, nasal, pulmonary, buccal, subdermal, intradermal, transdermal, or parenteral, including intravenous, subcutaneous and/or intramuscular. In one embodiment, the route of administration is oral. In another embodiment, the route of administration is topical.

Dosage forms can be administered once a day, or more than once a day, such as twice or thrice daily. Alternatively, dosage forms can be administered less frequently than daily, such as every other day, or weekly, if found to be advisable by a prescribing physician or drug’s prescribing information. Dosing regimens include, for example, dose titration to the extent necessary or useful for the indication to be treated, thus allowing the patient’s body to adapt to the treatment, to minimize or avoid unwanted side effects associated with the treatment, and/or to maximize the therapeutic effect of the present compounds. Other dosage forms include delayed or controlled-release forms. Suitable dosage regimens and/or forms include those set out, for example, in the latest edition of the Physicians' Desk Reference, incorporated herein by reference.

Proper dosages for pediatric patients can be determined using known methods, including weight, age, body surface area, and models such as Simcyp® Pediatric Simulation modeling (CERTARA, Princeton, N.J.) which can be used to establish a pharmacokinetic approach for dosing that takes into account patient age, ontogeny of the clearance pathways to eliminate a compound of any one of structures (la), (la’), (lb), (Ic) or (Id) or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, and body surface area (BSA). In one embodiment, the dosage form is formulated to provide a pediatric dose from about 30% to about 100% of an adult dose, or about 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of an adult dose.

In one embodiment, the invention provides an oral pharmaceutical composition comprising a compound of structure (I) or any of the structures (la), (la’), (lb), (Ic) or (Id), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, together with at least one pharmaceutically acceptable oral carrier, diluent, or excipient. In another embodiment, the invention provides a topical pharmaceutical composition comprising a compound of structure (I) or any of the structures (la), (la’), (lb), (Ic) or (Id), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, together with at least one pharmaceutically acceptable topical carrier, diluent, or excipient. For example, the oral pharmaceutical composition is provided to treat cholestatic pruritus, wherein the dosage regimen is, for example, once a day. In one embodiment, the topical pharmaceutical composition is provided to treat atopic dermatitis.

In another embodiment, there are provided methods of making a composition of a compound described herein including formulating a compound of the invention with a pharmaceutically acceptable carrier or diluent. In some embodiments, the pharmaceutically acceptable carrier or diluent is suitable for oral administration. In some such embodiments, the methods can further include the step of formulating the composition into a tablet or capsule. In other embodiments, the pharmaceutically acceptable carrier or diluent is suitable for parenteral administration. In some such embodiments, the methods further include the step of lyophilizing the composition to form a lyophilized preparation. In some embodiments, the composition is formulated into a pediatric dosage form suitable for treating a pediatric subject.

In certain embodiments, the invention provides a compound of structure (I) or any of the structures (la), (la’), (lb), (Ic) or (Id), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof. Such compounds can be synthesized using standard synthetic techniques known to those skilled in the art. For example, compounds of the present invention can be synthesized using appropriately modified synthetic procedures set forth in the following Examples and Reaction Schemes.

To this end, the reactions, processes, and synthetic methods described herein are not limited to the specific conditions described in the following experimental section, but rather are intended as a guide to one with suitable skill in this field. For example, reactions may be carried out in any suitable solvent, or other reagents to perform the transformationfs] necessary. Generally, suitable solvents are protic or aprotic solvents which are substantially non-reactive with the reactants, the intermediates or products at the temperatures at which the reactions are carried out (i.e., temperatures which may range from the freezing to boiling temperatures). A given reaction may be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction, suitable solvents for a particular work-up following the reaction may be employed.

All reagents, for which the synthesis is not described in the experimental part, are either commercially available, or are known compounds or may be formed from known compounds by known methods by a person skilled in the art. The compounds and intermediates produced according to the methods of the invention may require purification. Purification of organic compounds is well known to a person skilled in the art and there may be several ways of purifying the same compound. In some cases, no purification may be necessary. In some cases, the compounds may be purified by crystallization. In some cases, impurities may be stirred out using a suitable solvent. In some cases, the compounds may be purified by chromatography, particularly flash column chromatography, using purpose-made or prepacked silica gel cartridges and eluents such as gradients of solvents such as heptane, ether, ethyl acetate, acetonitrile, ethanol and the like. In some cases, the compounds may be purified by preparative HPLC using methods as described.

Purification methods as described herein may provide compounds of the present invention which possess a sufficiently basic or acidic functionality in the form of a salt, such as, in the case of a compound of the present invention which is sufficiently basic, a trifluoroacetate or formate salt, or, in the case of a compound of the present invention which is sufficiently acidic, an ammonium salt. A salt of this type can either be transformed into its free base or free acid form, respectively, by various methods known to a person skilled in the art, or be used as salts in subsequent biological assays. It is to be understood that the specific form of a compound of the present invention as isolated and as described herein is not necessarily the only form in which said compound can be applied to a biological assay in order to quantify the specific biological activity.

Chemical names were generated using the ChemDraw naming software (Version 17.0.0.206) by PerkinElmer Informatics, Inc. In some cases, generally accepted names of commercially available reagents were used in place of names generated by the naming software. EXAMPLES

General Methods

1 H NMR (400 MHz) spectra were obtained in solution of deuterochloroform (CDCL), deuteromethanol (CD3OD) or dimethyl sulfoxide - D6 (DMSO). HPLC retention times, purities, and mass spectra (LCMS) were obtained using one of the following methods:

Method 1 : Agilent 1260 Infinity II System equipped with an Agilent Poroshell 120 EC-18, 2.7 pm, 4.6 x 100 mm column at 30 °C, using H2O with 0.1% formic acid as the mobile phase A, and MeCN with 0.1% formic acid as the mobile phase B. An ESI detector in positive mode was used. The gradient was 5-95% mobile phase B over 12 min then held at 95% for 1.8 min, then return to 10% mobile phase B over 0.2 min. The flow rate was 1 mL/min.

Method 2: SHIMADZU LCMS-2020 equipped with Kinetex® EVO Cl 8 2.1x30mm 5um column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.15 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.04 min. The flow rate was 2 mL/min. An ESI detector in positive mode was used.

Method 3: SHIMADZU LCMS-2020 equipped with Kinetex® EVO Cl 8 2.1x30mm 5um column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.6 min, held at 95% for 0.18 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.01 min. The flow rate was 2 mL/min. An ESI detector in positive mode was used.

Method 4: SHIMADZU LCMS-2020 equipped with Kinetex® EVO Cl 8 2.1x20mm 2.6um column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.15 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.04 min. The flow rate was 2 mL/min. An ESI detector in positive mode was used. Method 5 : Shimadzu SCL-10A system equipped with Agilent Eclipse XDB- C18, 5 uM, 4.6 x 150 mm column and PE Sciex API 150 EX, using water with 0.1% trifluoroacetic acid as the mobile phase A, and methanol with 0.1% trifluoroacetic acid as the mobile phase B. The gradient was 5-95% mobile phase B over 12 min then held at 95% mobile phase B for 3 min, then return to 5% mobile phase B for 1 min. The flow rate was 1 mL/min.

Method 6: SHIMADZU LCMS-2020 equipped with Kinetex® EVO Cl 8 2.1x30mm 5um column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.29 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.1 min. The flow rate was 1.5 mL/min. An ESI detector in positive mode was used.

Method 7: SHIMADZU LCMS-2020 equipped with Kinetex® EVO Cl 8 2.1x30mm 5um column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.4 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.34 min. The flow rate was 1.5 mL/min. An ESI detector in positive mode was used. Method 8: Agilent 1200\G6110A equipped with Kinetex® 5um EVO C18 30*2.1mm column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.4 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.29 min. The flow rate was 1.5 mL/min. An ESI detector in positive mode was used.

Method 9: Agilent 1200\G6110A equipped with Chromolith Flash RP-18e 25*2.0 mm column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.4 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.29 min. The flow rate was 1.5 mL/min. An ESI detector in positive mode was used.

Method 10: Agilent 1200\G1956A equipped with Kinetex EVO C18 30*2.1mm, 5um column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.4 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.29 min. The flow rate was 1.5 mL/min. An ESI detector in positive mode was used.

Method 11 : SHIMADZU LCMS-2020 equipped with Kinetex® EVO Cl 8 2.1x20mm 2.6um column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.15 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.04 min. The flow rate was 2 mL/min. An ESI detector in positive mode was used.

Method 12: Agilent 1200 System equipped with a Kinetex C18 50 x 2.1mm (5um particles), using H2O with 0.037% Trifluoroacetic Acid as the mobile phase A, and MeCN with 0.018% Trifluoroacetic Acid as the mobile phase B. An ESI detector in positive mode was used. The gradient was 5% B at 0. OOmin and 5-90% B at 0.00-0.80 min, 90-95% B at 0.80-0.12 min, and then 95-5%B in O.Olmin, hold on 5% B for 0.34min, the flow rate was 1.5 ml/min.

Method 13: Agilent 1260 Infinity II System equipped with an Agilent Poroshell 120 EC-18, 2.7 pm, 4.6 x 100 mm column at 30 °C, using H2O with 0.1% formic acid as the mobile phase A, and MeCN with 0.1% formic acid as the mobile phase B. An ESI detector in positive mode was used. The gradient was 5-95% mobile phase B over 5 min then held at 95% for 1.8 min, then return to 20% mobile phase B over 0.2 min. The flow rate was 1 mL/min.

The pyridine, dichloromethane (DCM), tetrahydrofuran (THF), and toluene used in the procedures were from Aldrich Sure-Seal bottles kept under nitrogen (N2). Other solvents were used as is. All reactions were stirred magnetically, and temperatures are external reaction temperatures. Chromatographies were typically carried out using a Combiflash Rf flash purification system (Teledyne Isco) equipped with Redisep (Teledyne Isco) Rf Gold Normal-Phase silica gel (SiO2) columns or by using a similar system.

Preparative HPLC purifications were typically performed using one of the following systems: 1) Waters System equipped with a Waters 2489 uv/vis detector, an Aquity QDA detector, a Waters xBndge Prep C18 5 pm OBD, 30 x 150 mm column, and eluting with various gradients of H2O/ MeCN (0.1% formic acid) at a 30 mL/min flow rate, 2) Teledyne Isco ACCQPrep® HP150 UV system equipped with a Waters xBridge Prep C18 5 pm OBD, 30 x 150 mm column, and eluting with various gradients of H2O/ MeCN (0.1% formic acid) at a 42.5 mL/min flow rate, or 3) column: Phenomenex Synergi C18 150 x 30 mm- 4pm; mobile phase: [H2O (0.225%formic acid)-MeCN];B%: 55%-85%,12min) and were typically concentrated using a Genevac EZ-2.

The following additional abbreviations are used: ethyl acetate (EA), triethylamine (TEA), water (H2O, sodium chloride (NaCl), Hydrochi oridric acid (HC1), methanol (MeOH), dimethyl sulfoxide (DMSO), silica gel (SiO2), diisobutyl aluminium hydride (DIBAL), trifluoroacetic acid (TFA), 4-dimethylaminopyridine (DMAP), diphenylphosphoryl azide (DPP A), benzoyl peroxide (BPO), 1,1'- bis(diphenylphosphino)ferrocene (dppf), bis(pinacolato)diboron (B2pin2), tetrahydrofuran (THF), l,4-diazabicyclo[2.2.2]octane bis(sulfur dioxide) adduct (DAB SO), hexafluorophosphate azabenzotriazole tetramethyl uronium (HATU), hydroxybenzotriazole (HOBt), A-methyl morpholine (NMM), A-Bromosuccinimide (NBS), diisopropylethyl amine (DIPEA or DIEA), diethyl azodi carboxyl ate (DEAD), diisopropyl azodicarboxylate (DIAD), 2-[2-(dicyclohexylphosphino)phenyl]-A- methylindole (CM-Phos), triflic acid (TfOH), l-ethyl-3 -(3 -dimethylaminopropyl) carbodiimide hydrochloride (EDC), isopropanol (IP A), dimethylformamide (DMF), dimethyl acetamide (DMA), di chloromethane (DCM), 1,2-di chloroethane (DCE), acetonitrile (MeCN or ACN), 1,1’ -thiocarbonyl di imidazole (TCDI), petroleum ether (PE), not determined (ND), retention time (RT), molecular weight (mw), room temperature (rt), hour (hr), and not applicable (N/A). EXAMPLE 1

Scheme 1

Reagents: i. R1COCI, base (DIPEA), solvent (DCM) or R1COOH, peptide coupling reaction conditions, solvent; ii. Acid (HCl), solvent (1,4 -dioxane); in. Base (t-Bu-ONa, TEA, CS2CO3...), Catalyst (Pd(O Ac) 2, BINAP), solvent (1,4-dioxane)

SYNTHESIS OF EXAMPLE 1 Synthesis of tert-butyl N-((lS,4S)-r4-l(4-fluorobenzoyl)amino1cyclohexyl1carbamate

To a mixture of tert-butyl cis-N-(4-aminocyclohexyl)carbamate (1.35 g, 6.31 mmol, 1 eq.) and TEA (1.28 g, 12.61 mmol, 1.76 mL, 2 eq.) in DCM (20 mL) was added 4-fluorobenzoyl chloride (1 g, 6.31 mmol, 757.58 uL, 1 eq.) at 0 °C, then the reaction mixture was stirred at 25 °C for 0.5 hr. The reaction mixture was concentrated to give a residue that was purified by silica on column chromatography (eluent: PE/EtOAc=10: l to 1 :1). Tert-butyl N-[4-[(4-fluorobenzoyl)amino]cyclohexyl]carbamate (2 g, 5.95 mmol, 94.27% yield) was obtained.

Synthesis of cis-N-(4-aminocvclohexyl)-4-fluoro-benzamide.HCl A mixture of tert-butyl N-[4-[(4-fluorobenzoyl)amino]cyclohexyl]carbamate (2 g, 5.95 mmol, 1 eq.) in HClZEtOAc (4 M, 5 mL, 3.36 eq.) was stirred at 25 °C for 1 hr. The reaction mixture was concentrated to give N-(lS,4S)-(4-aminocyclohexyl)-4- fluoro-benzamide.HCl (1.5 g, 5.50 mmol, 92.50% yield). It was used as is in the next step directly. LCMS-ESI (m/z) calculated: 236.3; found 237.4 [M+H] + , RT = 0.237 min

(Method 12)

Synthesis of 4-fluoro-N-((l s,4s)-4-((2-(trifluoromethyl)benzolb1thiophen-4- yl)amino)cyclohexyl) benzamide A mixture of 4-bromo-2-(trifluorom ethyl)- 1 -benzothiophene (103 mg, 1 Eq, 367 pmol), N-(lS,4S)-(4-aminocyclohexyl)-4-fluoro-benzamide.HCl (100 mg, 1 Eq, 367 pmol), cesium carbonate (358 mg, 3 Eq, 1.10 mmol), palladium(II) acetate (4.12 mg, 0.05 Eq, 18.3 pmol), and BINAP (22.8 mg, 0.10 Eq, 36.7 pmol) in 1,4-Dioxane (3 mL) was bubbled with nitrogen for 5 minutes. The vial was capped and the resulting mixture was stirred at 100 °C. After stirring for 19 hours, the reaction mixture was cooled to room temperature. The reaction mixture was filtered, and the filtrate was purified by prep-HPLC to yield 4-fluoro-N-((ls,4s)-4-((2-

(trifluorom ethyl )benzo[b]thiophen-4-yl)amino) cyclohexyl)benzamide (13.7 mg, 31.4 pmol, 8.56 %).

'H NMR (400 MHz, DMSO-d6) 8 8.53 (s, 1H), 8.22 (d, J = 6.6 Hz, 1H), 7.94 (dd, J = 8.5, 5.6 Hz, 2H), 7.34 - 7.20 (m, 4H), 6.56 (d, J = 7.9 Hz, 1H), 6.09 (d, J = 5.5 Hz, 1H), 3.97 - 3.88 (m, 1H), 3.65 - 3.58 (m, 1H), 1.95 - 1.83 (m, 4H), 1.79 - 1.63 (m, 4H).

LCMS-ESI (m/z) calculated: 436.47; found 437.2 [M+H] + , RT = 11.697 min (Method 1)

EXAMPLE 2

SYNTHESIS OF EXAMPLE 2

Synthesis of tert-butyl ((lS,3R)-3-(4-methoxybenzamido)cyclohexyl)carbamate

To a stirring solution of an ice-cold solution of (lS,3R)-3-Amino-l-(Boc- amino)cyclohexane (1.142 g, 1.0 eq., 5.329 mmol) in DCM (30 mL) was added with DIPEA (1.377 g, 1.9 mL, 2.0 eq., 10.66 mmol), followed by slow addition of 4- methoxybenzoyl chloride (954.5 mg, 758 pL, 1.05 eq., 5.595 mmol). The resulting mixture was stirred at room temperature. After stirring for 18 hours, the reaction mixture was filtered, and the filter cake was washed with DCM and dried under high vacuum to yield tert-butyl ((lS,3R)-3-(4-methoxybenzamido)cyclohexyl)carbamate (1.659 g, 4.761 mmol, 89.35 % yield).

'H NMR (400 MHz, DMSO-d6) 8 8.09 (d, J = 7.9 Hz, 1H), 7.82 (d, J = 8.4 Hz, 2H), 6.97 (d, J = 8.4 Hz, 2H), 6.83 (d, J = 8.1 Hz, 1H), 3.80 (s, 3H), 3.79 - 3.71 (m, 1H), 3.31 - 3.22 (m, 1H), 1.93 (d, J = 12.0 Hz, 1H), 1.77 - 1.68 (m, 3H), 1.38 (s, 9H), 1.31 - 1.17 (m, 3H), 1.11 - 1.01 (m, 1H).

LCMS-ESI (m/z) calculated: 348.44; found 349.2 [M+H] + , RT = 4.239 min (Method 13).

Synthesis of N-((lR,3S)-3-aminocvclohexyl)-4-methoxybenzamide.HCl

Methoxybenzamido)cyclohexyl)carbamate (1.654 g, 1 eq., 4.747 mmol) in EtOH (40 mL) was added 1.25M hydrogen chloride in 1,4-dioxane (1.731 g, 37.97 mL, 1.25 molar, 10 eq., 47.47 mmol). The resulting mixture was stirred at 50 °C for 17 hours. The reaction mixture was directly concentrated to yield a white solid, which was further dried under high vacuum to yield N-((lR,3S)-3-aminocyclohexyl)-4-methoxybenzamide hydrochloride (1.331 g, 4.674 mmol, 98.46 % yield). It was used without further purification in the next step.

'H NMR (400 MHz, DMSO-d6) 8 8.26 (d, J = 7.9 Hz, 1H), 8.09 (s, 3H), 7.84 (d, J = 8.4 Hz, 2H), 6.98 (d, J = 8.4 Hz, 2H), 3.91 - 3.81 (m, 1H), 3.80 (s, 3H), 3.13 - 3.05 (m, 1H), 2.12 (d, J = 11.6 Hz, 1H), 1.91 (d, J = 12.0 Hz, 1H), 1.78 (d, J = 12.0 Hz, 2H), 1.44 - 1.21 (m, 4H). LCMS-ESI (m/z) calculated: 248.44; found 249.2 [M+H] + , RT = 1.665 min (Method 13).

Synthesis of 4-methoxy-N-((lR,3 S)-3-((2-(trifluoromethvPbenzolb1thiophen-4- yDamino) cyclohexyDbenzamide

A mixture of 4-bromo-2-(trifluorom ethyl)- 1 -benzothiophene (98.7 mg, 1 Eq, 351 pmol), N-((lR,3S)-3-aminocyclohexyl)-4-methoxybenzamide hydrochloride (100 mg, 1 Eq, 351 pmol), cesium carbonate (343 mg, 3 Eq, 1.05 mmol), palladium(II) acetate (3.94 mg, 0.05 Eq, 17.6 pmol), and BINAP (21.9 mg, 0.10 Eq, 35.1 pmol) in 1,4-Dioxane (3 ml) was bubbled with nitrogen for 5 minutes. The vial was capped and the resulting mixture was stirred at 100 °C. After stirring for 17.5 hours, the reaction mixture was cooled to room temperature. The reaction mixture was filtered, and the filtrate was purified by prep-HPLC to yield 4-methoxy-N-((lR,3S)-3- ((2-(trifluoromethyl)benzo[b]thiophen-4-yl)amino) cyclohexyl)benzamide (14 mg, 31 pmol, 8.9 % yield).

X H NMR (400 MHz, DMSO-t/ 6 ) 8 8.46 (s, 1H), 8.15 (d, J= 7.9 Hz, 1H), 7.81 (d, J= 8.4 Hz, 2H), 7.30 (t, J= 8.0 Hz, 1H), 7.19 (d, J = 8.0 Hz, 1H), 6.97 (d, J= 8.6 Hz, 2H), 6.61 (d, J= 7.9 Hz, 1H), 6.20 (d, J= 7.9 Hz, 1H), 3.99 - 3.90 (m, 1H), 3.79 (s, 3H), 3.58 - 3.49 (m, 1H), 2.22 (d, J= 12.2 Hz, 1H), 2.02 (d, J= 12.5 Hz, 1H), 1.84 (t, J = 16.5 Hz, 2H), 1.53 - 1.30 (m, 3H), 1.28 - 1.21 (m, 1H).

LCMS-ESI (m/z) calculated: 448.5; found 449.2 [M+H] + , RT = 11.163 min (Method 1). EXAMPLE 3

SYNTHESIS OF EXAMPLE 3

Synthesis of 4-methoxy-N-r(lR,3S)-3- (trifluoromethyl)benzothiophen-7-yl1amino1 cyclohexyllbenzamide

To a solution of 7-bromo-2-(trifluoromethyl)benzothiophene (50 mg, 177.88 umol, 1 Eq) , N-[(lR,3S)-3-aminocyclohexyl]-4-methoxy-benzamide (53.00 mg, 213.45 umol, 1.2 Eq), RuPhos Pd G4 (14.88 mg, 17.79 umol, 0.1 Eq) ,Cs 2 CO 3 (173.87 mg, 533.63 umol, 3 Eq) in dioxane (4 mL) under N2. The reaction mixture was stirred at 100 °C for 12 hr. After return to rt, the mixture was filtered. The filtrate was concentrated and purified by reversed-phase HPLC to give desired product (23.1 mg, 50.32 umol, 20.21% yield).

X H NMR (400 MHz, DMSO-d6) 8 = 8.14 (d, J= 7.9 Hz, 1H), 8.02 (d, J= 1.0 Hz, 1H), 7.82 (d, J= 8.9 Hz, 2H), 7.35 - 7.26 (m, 2H), 6.97 (d, J= 8.9 Hz, 2H), 6.80 (d, J= 7.6 Hz, 1H), 5.62 (d, J= 8.1 Hz, 1H), 3.98 - 3.89 (m, 1H), 3.79 (s, 3H), 3.62 - 3.53 (m, 1H), 2.20 (br d, J= 11.8 Hz, 1H), 1.99 (br d, J= 12.4 Hz, 1H), 1.88 - 1.77 (m, 2H), 1.52 - 1.38 (m, 2H), 1.36 - 1.27 (m, 2H)

LCMS-ESI (m/z) calculated: 448.5; found 449.3 [M+H] + , RT = 0.718 min (Method 6). EXAMPLE 4

SYNTHESIS OF EXAMPLE 4

Synthesis of 4-methoxy-N-r(lR,3S)-3- (trifluoromethyl)-L3-benzoxazol-4- yllaminol cyclohexyl] benzamide

To a solution of 4-bromo-2-(trifluoromethyl)-l,3-benzoxazole (70 mg, 263.14 umol, 1 Eq), N-[(lR,3S)-3-aminocyclohexyl]-4-methoxy-benzamide (78.41 mg, 315.77 umol, 1.2 Eq), Pd2(dba)3 (48.19 mg, 52.63 umol, 0.2 Eq) Xantphos (30.45 mg, 52.63 umol, 0.2 Eq) and CS2CO3 (257.21 mg, 789.43 umol, 3 Eq) in toluene (5 mL) under N2. The reaction mixture was stirred at 100 °C for 12 hr. After return to rt, the mixture was filtered through a pad of celite. The filtrate was concentrated under vacuum and the residue was purified by reversed-phase HPLC to yield 4-methoxy-N-[(lR,3S)-3-[[2- (trifluoromethyl)-l,3-benzoxazol-4-yl]amino]cyclohexyl]benza mide (2.8 mg, 6.24 umol, 2.37% yield).

'H NMR (400 MHz, DMSO-d6) 8 = 8.15 (d, J= 7.8 Hz, 1H), 7.84 - 7.80 (m, 2H), 7.35 (t, J= 8.3 Hz, 1H), 6.98 - 6.93 (m, 3H), 6.68 (d, J= 8.3 Hz, 1H), 6.32 (br d, J = 8.8 Hz, 1H), 3.97 - 3.88 (m, 1H), 3.79 (s, 3H), 3.68 (td, J= 3.4, 7.4 Hz, 1H), 2.14 (br d, J= 10.6 Hz, 1H), 1.94 (br d, J= 11.9 Hz, 1H), 1.86 - 1.77 (m, 2H), 1.47 - 1.37 (m, 2H), 1.34 - 1.26 (m, 2H)

LCMS-ESI (m/z) calculated: 433.42; found 434.3 [M+H] + , RT = 0.699 min (Method 9).

The compounds listed in Table 1 were made using the procedures of Scheme 1. Table 1

Scheme 2

Reagents: i. RIX, Base (t-Bu-ONa, TEA, CS2CO3...), Catalyst (Pd(OAc)2, BINAP), solvent (1,4-dioxane); ii. Acid (HCI), solvent (1,4 -dioxane); in. R2COCI, Base (DIPEA), solvent (DCM) or R2COOH, peptide coupling reagent conditions EXAMPLE 5

SYNTHESIS OF EXAMPLE 5

Synthesis of tert-butyl ((lS,4S)-4-((2-(trifluoromethyl)benzolb1thiophen-4- yl)amino)cyclohexyl) carbamate

To a sealed tube containing 4-bromo-2-(trifluoromethyl)benzo[b]thiophene (500 mg, 1.1 Eq, 18 mmol) in toluene (50 mL) was added tert-butyl ((lS,4S)-4- aminocyclohexyl)carbamate (350 mg, 1 Eq, 16.3 mmol), and sodium tert-butoxide (173 mg, 1 Eq., 18 mmol) in THF (5 mL) followed by Pd(dppf)2C12-DCM (147 mg, 0.1 Eq, 1.8 mmol). The tube was purged with N2 three times and then sealed. The reaction mixture was heated to 110 °C for 3 hr. Solvent was removed under vacuo and the resulting crude residue was acidified with TFA and purified by reversed-phase C18 column (10-100% MeOH/FFO with 0.1% TFA) to yield a mixture of tert-butyl ((1 S,4S)-4-((2-(trifluoromethyl)benzo[b]thiophen-4-yl)amino)cyc lohexyl)carbamate (45 mg, 7% yield) and (lS,4S)-Nl-(2-(trifluoromethyl)benzo[b]thiophen-4- yl)cyclohexane-l,4-diamine as TFA salt (26 mg, 4% yield).

LCMS-ESI (m/z) calculated: 414.49; found 415.5 [M+H] + , RT = 5.54 min (Method 5). Synthesis of (lS,4S)-Nl-(2-(trifluoromethyl)benzorb1thiophen-4-yl)cyclohe xane-L4- diamine

To a solution of tert-butyl ((lS,4S)-4-((2-(trifluoromethyl)benzo[b]thiophen-4- yl)amino) cyclohexyl)carbamate (45 mg, 1 Eq, 0.011 mmol) in DCM (2 mL) was added TFA (2 mL). The reaction mixture was stirred for 2 hr and concentrated under vacuo to provide the title compound (lS,4S)-Nl-(2-(trifluoromethyl)benzo[b]thiophen-4- yl)cyclohexane- 1 ,4-diamine.

LCMS-ESI (m/z) calculated: 314.1; found 315.2 [M+H] + , RT = 4.23 min (Method 5).

Synthesis of 3-cyano-N-((l SAS)-4-((2-(trifluoromethyl)benzorb]thiophen-4-yl)amino) cyclohexyDbenzamide

To 3 -cyanobenzoic acid (3.0 mg, 1.1 Eq, 0.020 mmol) in DMF (2 mL), was added HATU (6.5 mg, 1.05 Eq, 0.018 mmol), followed by (lS,4S)-Nl-(2- (trifluoromethyl)benzo[b]thiophen-4-yl)cyclohexane-l,4-diami ne TFA salt (8 mg, 1 Eq, 0.019 mmol). DIE A (9.9 mg, 4 Eq, 0.077 mmol) was added and the reaction mixture was stirred at rt for 16 hr. The reaction mixture was acidified with TFA and purified by RP-C18 ISCO (10-100% MeOH/H 2 O, 0.1% TFA) to yield l-methyl-N-((l S,4S)-4-((2- (trifluoromethyl)benzo[b]thiophen-4-yl)amino)cyclohexyl)- 1,2,3, 4-tetrahydroquinoline- 6-carboxamide as TFA salt (5 mg, 48% yield). LCMS-ESI (m/z) calculated: 443.1; found 444.5 [M+H] + , RT = 13.37 min (Method 5).

EXAMPLE 6

SYNTHESIS OF EXAMPLE 6

Synthesis of 3 -amino- 1 -(2, 2-difhioroethyl)-N-[4- (trifhioromethyl) imidazo[L2- alpyridin-5 yllaminolcyclohexyllpyrazole-4-carboxamide To a solution of 3-amino-N-[4-[[2-(trifluoromethyl) imidazo [1, 2-a] pyridin-5- yl] amino] cyclohexyl]-lH-pyrazole-4-carboxamide (50 mg, 122.73 umol, N/A purity, 1 eq) and CS2CO3 (79.98 mg, 245.46 umol, 2 eq) in MeCN (3 mL) was added 2, 2- difluoroethyl trifluoromethanesulfonate (52.56 mg, 245.46 umol, 2 eq) , the reaction solution was stirred at 60 °C for 1 h. The reaction solution was filtered by filter membrane. The crude product was purified by Prep-HPLC with the following conditions: column: Phenomenex luna C18 150*25mm* lOum; mobile phase: [water (FA)-ACN]; B%: 27%-57%, lOmin. and lyophilized to give 3-amino-l-(2, 2-difluoroethyl)-N-[4-[[2- (trifluoromethyl) imidazo[l,2-a]pyri din-5 yl]amino]cyclohexyl]pyrazole-4-carboxamide (6.3 mg, 12.78 umol, 10.41% yield, 95.6% purity).

LCMS-ESI (m/z) calculated: 471.4; found 472.2 [M+H] + , RT = 0.408 min (Method 4).

'H NMR) (400 MHz, DMSO-d 6 ) 6 = 8.71 (s, 1H), 8.11 (s, 1H), 7.60 (d, J = 6.6 Hz, 1H), 7.42 - 7.24 (m, 1H), 6.95 (d, J = 8.9 Hz, 1H), 6.46 (d, J = 4.8 Hz, 1H), 6.42 (t, J = 3.5 Hz, 1H), 6.10 (d, J = 7.6 Hz, 1H), 5.82 - 5.16 (m, 2H), 4.37 (dt, J = 3.6, 15.1 Hz, 2H), 3.92 - 3.80 (m, 1H), 3.67 (s, 1H), 1.96 - 1.86 (m, 2H), 1.86 - 1.72 (m, 4H), 1.70 - 1.61 (m, 2H).

EXAMPLE 7

SYNTHESIS OF EXAMPLE 7

Synthesis of 3-amino-l-(2,2,2-tnfluoroethyl)-N-r4- (tnfluoromethyl)imidazori,2- a]pyridin-5-yl]amino]cyclohexyl]pyrazole-4-carboxamide

To a solution of 3-amino-N-[4-[[2-(trifluoromethyl) imidazo [1, 2-a] pyridin-5- yl] amino] cyclohexyl]-lH-pyrazole-4-carboxamide (50 mg, 122.73 umol, N/A purity, 1 eq) and Cs2CO3 (79.98 mg, 245.46 umol, 2 eq) in MeCN (3 mL) was added 2,2,2- trifluoroethyl trifluoromethanesulfonate (28.49 mg, 122.73 umol, 1 eq) , the reaction solution was stirred at 50 °C for 1 h. The reaction solution was filtered by filter membrane. The crude product was purified by Prep-HPLC with the following conditions: column: Phenomenex Luna C18 150*25mm*10um; mobile phase: [water(FA)- ACN];B%: 31%- 61%,10min. and lyophilized to give 3-amino-l-(2,2,2-trifluoroethyl)- N-[4-[[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino]c yclohexyl]pyrazole-4- carboxamide (15.5 mg, 25.73% yield) was obtained.

LCMS-ESI (m/z) calculated: 489.4; found 490.0 [M+H] + , RT = 0.471 min (Method 2).

1H NMR (400 MHz, DMSO-d 6 ) 8 = 8.72 (s, 1H), 8.17 (s, 1H), 7.69 (d, J = 6.6 Hz, 1H), 7.35 (dd, J = 7.8, 8.7 Hz, 1H), 6.95 (d, J = 8.9 Hz, 1H), 6.46 (d, J = 4.8 Hz, 1H), 6.10 (d, J = 7.6 Hz, 1H), 5.65 - 5.44 (m, 2H), 4.89 (q, J = 9.1 Hz, 2H), 3.93 - 3.77 (m, 1H), 3.68 (s, 1H), 1.96 - 1.86 (m, 2H), 1.86 - 1.72 (m, 4H), 1.70 - 1.61 (m, 2H). EXAMPLE 8

SYNTHESIS OF EXAMPLE 8

Synthesis of ethyl l-(2-trimethylsilylethoxymethyl)pyrazole-4-carboxylate

To a solution of ethyl lH-pyrazole-4-carboxylate (10 g, 71.36 mmol, 1 eq) in THF (150 mL) was added NaH (3.42 g, 85.63 mmol, 60% purity, 1.2 eq) at 0 °C and stirred for Ih under N2. Then added 2-(chloromethoxy)ethyl-trimethyl-silane (14.28 g, 85.63 mmol, 15.16 mL, 1.2 eq) and stirred for 2 h at 25 °C. The reaction was quenched with saturated NH4CI aqueous (200 mL) and extracted with EtOAc (100 mL*3). The combined organic layers were washed with brine, dried over Na2SC>4, concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=l/O to 80/20). (TLC, PE:EtOAc=5: l, Rf=0.5). ethyl l-(2- trimethylsilylethoxymethyl)pyrazole-4-carboxylate (19 g, 70.27 mmol, 98.47% yield, N/A purity) was obtained as a light yellow oil.

LCMS-ESI (m/z) calculated: 270.1; found 271.2 [M+H] + , RT = 0.511 min (Method 4).

'H NMR (400 MHz, DMSO-d 6 ) 8 = 8.49 (s, IH), 7.91 (s, IH), 5.44 (s, 2H), 4.22 (q, J = 7.1 Hz, 2H), 3.57 - 3.52 (m, 2H), 1.26 (t, J = 7.1 Hz, 3H), 0.85 - 0.80 (m, 2H), - 0.06 (s, 9H). Synthesis of ethyl 5-formyl-l-(2-trimethylsilylethoxymethyl)pyrazole-4-carboxyl ate

To a solution of ethyl l-(2-trimethylsilylethoxymethyl)pyrazole-4-carboxylate (19 g, 70.27 mmol, 1 eq) in THF (200 mL) was added dropwise of LDA (2 M, 52.70 mL, 1.5 eq) at -70 °C over 10 min. After stirred for 30 min, DMF (30.82 g, 421.60 mmol, 32.44 mL, 6 eq) was added. The resulting mixture was stirred at -70 °C for another 20 min. The reaction mixture was slowly poured into saturated NH4CI aqueous (200 mL) and keep the temperature at 0-10 °C, extracted with EtOAc (100 mL*3). The combined organic layers were washed with brine, dried over Na2SO4, concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=l/O to 74/26). (TLC, PE:EtOAc=5: l, Rf=0.65). ethyl 5 -formyl- 1 -(2- trimethylsilylethoxymethyl)pyrazole-4-carboxylate (5.8 g, 19.44 mmol, 27.66% yield) was obtained as light yellow oil.

'H NMR (400 MHz, DMSO-d 6 ) 8 = 10.34 (s, 1H), 8.09 (s, 1H), 5.72 (s, 2H), 4.32 (q, J = 7.1 Hz, 2H), 3.58 - 3.53 (m, 2H), 1.31 (t, J = 7.1 Hz, 3H), 0.83 - 0.79 (m, 2H), -0.08 (s, 9H)

Synthesis of ethyl 5-(difluoromethyl)-l-(2-trimethylsilylethoxymethyl)pyrazole- 4- carboxylate

To a solution of ethyl 5-formyl-l-(2-trimethylsilylethoxymethyl)pyrazole-4- carboxylate (2 g, 6.70 mmol, 1 eq) in DCM (15 mL) was added DAST (1.62 g, 10.05 mmol, 1.33 mL, 1.5 eq) at 0 °C and stirred for 2 h at 25 °C. The reaction mixture concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=l/O to 0/1). (TLC, PE:EtOAc=5: l, Rf=0.8). ethyl 5-(dmuoromethyl)-l-(2- trimethylsilylethoxymethyl)pyrazole-4-carboxylate (1.6 g, 4.99 mmol, 74.51% yield, N/A purity) was obtained as colorless oil.

LCMS-ESI (m/z) calculated: 320.1; found 321.1 [M+H] + , RT = 0.516 min (Method 4).

'H NMR (400 MHz, DMSO-d 6 ) 8 = 8.04 (s, 1H), 7.54 (t, J = 52.2 Hz, 1H), 5.62 (s, 2H), 4.27 (q, J = 7.1 Hz, 2H), 3.61 - 3.55 (m, 2H), 1.29 (t, J = 7.1 Hz, 3H), 0.86 - 0.81 (m, 2H), -0.07 (s, 9H).

Synthesis of 5-(difluoromethyl)-l-(2-trimethylsilylethoxymethyl)pyrazole- 4-carboxylic acid

To a solution of ethyl 5-(difluoromethyl)-l-(2- trimethyl silyl ethoxymethyl )pyrazole-4-carboxylate (200 mg, 624.21 umol, 1 eq) in THF (1.5 mL), H2O (1.5 mL) was added LiOH.H2O (78.58 mg, 1.87 mmol, 3 eq) and stirred for 2 h at 25 °C. Then added LiOH.H2O (78.58 mg, 1.87 mmol, 3 eq) and stirred for 12 h at 25 °C. The reaction mixture was neutralized 5% HC1 to PH 7. The reaction mixture concentrated under reduced pressure to give a residue. The residue was directly used for next step without purification. 5 -(difluoromethyl)- 1 -(2- trimethylsilylethoxymethyl)pyrazole-4-carboxylic acid (180 mg, 615.69 umol, 98.64% yield, N/A purity) was obtained as white solid.

LCMS-ESI (m/z) calculated: 292.1; found 293.2 [M+H] + , RT = 0.532 min (Method 2).

'H NMR (400 MHz, DMSO-d 6 ) 6 = 8.04 (s, 1H), 7.54 (t, J = 52.2 Hz, 1H), 5.62 (s, 2H), 4.27 (q, J = 7.1 Hz, 2H), 3.61 - 3.55 (m, 2H), 1.29 (t, J = 7.1 Hz, 3H), 0.86 - 0.81 (m, 2H), -0.07 (s, 9H). Synthesis of 5- difluoromethyl)-N-r4-rr2- tnfluoromethyl)imidazolL2-a1pyndm-5- yllaminolcyclohexyll-1 -(2 -trimethyl silyl ethoxymethyl)pyrazole-4-carboxamide

To a solution of 5-(difluoromethyl)-l-(2 -trimethylsilyl ethoxymethyl)pyrazole-4- carboxylic acid (160 mg, 547.28 umol, 1 eq), N4-[2-(trifluorom ethyl )imidazo[ 1,2- a]pyridin-5-yl]cyclohexane-l,4-diamine (183.21 mg, 547.28 umol, 1 eq, HC1) in DMF (4 mL) was added HATU (312.14 mg, 820.92 umol, 1.5 eq) DIEA (212.20 mg, 1.64 mmol, 285.98 uL, 3 eq) and stirred for 1.5 h at 25 °C. The mixture was partitioned between EtOAc (10 mL) and water (10 mL). The aqueous phase was extracted with EtOAc (10 mL*2). Organic phase were dried over MgSO4, and concentrated under vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=l/O to 20/1). (TLC, PE:EtOAc=l:l, Rf=0.5). 5-(difluoromethyl)-N- [4-[[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino]cyc lohexyl]-l-(2- trimethylsilylethoxymethyl)pyrazole-4-carboxamide (210 mg, 366.72 umol, 67.01% yield, N/A purity) was obtained as white solid.

LCMS-ESI (m/z) calculated: 572.2; found 573.5 [M+H] + , RT = 0.612 min (Method 2).

'H NMR (400 MHz, DMSO-d 6 ) 8 = 8.77 (s, 1H), 8.33 - 8.22 (m, 2H), 7.76 (t, J = 52.9 Hz, 1H), 7.46 - 7.35 (m, 1H), 7.02 (d, J = 8.9 Hz, 1H), 6.51 (d, J = 4.6 Hz, 1H), 6.17 (d, J = 7.6 Hz, 1H), 5.62 (s, 2H), 4.01 - 3.91 (m, 1H), 3.76 (s, 1H), 3.64 (t, J = 8.1 Hz, 2H), 2.03 - 1.79 (m, 6H), 1.78 - 1.69 (m, 2H), 0.95 - 0.84 (m, 2H), 0.00 (s, 9H). Synthesis of 3-(difluoromethyl)-N-r4- (tnfluoromethyl)imidazorL2-a1pyndin-5- yll aminol cyclohexyll - 1 H-pyrazole-4-Carboxami de

To a solution of 5-(difluoromethyl)-N-[4-[[2-(trifluoromethyl)imidazo[l,2- a]pyridin-5-yl]amino]cyclohexyl]-l -(2 -trimethylsilyl ethoxymethyl)pyrazole-4- carboxamide (200 mg, 349.26 umol, 1 eq) in DCM (2 mL) was added TFA (616.00 mg, 5.40 mmol, 400.00 uL, 15.47 eq) and stirred for 1 h at 25 °C. The reaction mixture concentrated under reduced pressure to give a residue. Then added K2CO3 (241.35 mg, 1.75 mmol, 5 eq), MeOH (2 mL) and stirred for 1 h at 50 °C. The mixture was partitioned between EtOAc (10 mL) and water (lOmL). The aqueous phase was extracted with EtOAc (10 mL*2). Organic phase were dried over MgSO4, and concentrated under vacuum. The residue was directly used for next step without purification. 3-(difhioromethyl)-N-[4-[[2-(trifluoromethyl)imidazo[l,2-a]p yridin-5- yl]amino]cyclohexyl]-lH-pyrazole-4-carboxamide (150 mg, 301.77 umol, 86.40% yield, 89% purity) was obtained.

LCMS-ESI (m/z) calculated: 442.1; found 443.3 [M+H] + , RT = 0.424 min (Method 2).

X H NMR (400 MHz, DMSO-d 6 ) 8 = 13.54 (d, J = 2.0 Hz, 1H), 8.75 (s, 1H), 8.60 - 8.36 (m, 1H), 7.91 (d, J = 6.0 Hz, 1H), 7.57 - 7.24 (m, 2H), 6.99 (d, J = 8.9 Hz, 1H), 6.53 - 6.42 (m, 1H), 6.15 (d, J = 7.6 Hz, 1H), 3.93 (dd, J = 3.7, 6.4 Hz, 1H), 3.73 (s, 1H), 2.00 - 1.69 (m, 8H). Synthesis of l-(2,2-difluoroethyl)-3-(difluoromethyl)-N-14- (trifluoromethyl)imidazori,2-a1pyridin-5-yl1amino1cyclohexyl 1pyrazole-4-carboxamide

Example 8

To a solution of 3-(difluoromethyl)-N-[4-[[2-(trifluoromethyl)imidazo[l,2- a]pyridin-5-yl]amino]cyclohexyl]-lH-pyrazole-4-carboxamide (75 mg, 150.89 umol, 89% purity, 1 eq) 2,2-difluoroethyl trifluoromethanesulfonate (32.31 mg, 150.89 umol, 1 eq) in MeCN (2 mL) was added CS2CO3 (98.32 mg, 301.77 umol, 2 eq) and stirred for 12 h at 50 °C. The mixture was filtered to remove the insoluble. The crude product was purified by reversed-phase HPLC:Column (Phenomenex luna C18 150*25mm* lOum) Condition water((0.225%FA)-ACN) Begin B (41) End B (71) Gradient Time(min) (10). and lyophilized to give desired product l-(2,2-difluoroethyl)-3-(difluoromethyl)-N-[4- [[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5-yl]amino]cycloh exyl]pyrazole-4- carboxamide (6.2 mg, 12.13 umol, 8.04% yield, 99.1% purity).

LCMS-ESI (m/z) calculated: 506.1; found 507.1 [M+H] + , RT = 0.461 min (Method 4).

'H NMR (400 MHz, DMSO-d 6 ) 8 = 8.72 (s, 1H), 8.49 (s, 1H), 8.09 (d, J = 6.8 Hz, 1H), 7.53 - 7.21 (m, 2H), 6.96 (d, J = 8.9 Hz, 1H), 6.63 - 6.25 (m, 2H), 6.11 (d, J = 7.6 Hz, 1H), 4.77 (dt, J = 2.8, 15.6 Hz, 2H), 3.89 (d, J = 3.0 Hz, 1H), 3.69 (s, 1H), 1.96 - 1.74 (m, 6H), 1.72 - 1.63 (m, 2H). EXAMPLE 9

SYNTHESIS OF EXAMPLE 9

Synthesis of 3-(difluoromethyl)-l-(2,2,2-trifluoroethyl)-N-r4- (trifluoromethyl)imidazorL2-a1pyridin-5-yl1amino1cyclohexyl1 pyrazole-4-carboxamide

To a solution of 3-(difluoromethyl)-N-[4-[[2-(trifluoromethyl)imidazo[l,2- a]pyridin-5-yl]amino]cyclohexyl]-lH-pyrazole-4-carboxamide (75 mg, 150.89 umol, 89% purity, 1 eq) 2,2,2-trifluoroethyl trifluoromethanesulfonate (35.02 mg, 150.89 umol, 1 eq) in MeCN (2 mL) was added CS2CO3 (98.32 mg, 301.77 umol, 2 eq) and stirred for 12 h at 50 °C. The mixture was filtered to removed the insoluble. The crude product was purified by reversed-phase HPLC:Column (Phenomenex luna C18 150*25mm* lOum) Condition water((0.225%FA)-ACN) Begin B (39) End B (69) Gradient Time(min) (9) and lyophilized to give 3 -(difluoromethyl)- 1 -(2,2,2- trifluoroethyl)-N-[4-[[2-(trifluoromethyl)imidazo[l,2-a]pyri din-5-yl]amino]cyclohexyl] pyrazole-4-carboxamide (8.5 mg, 16.06 umol, 10.65% yield, 99.1% purity). LCMS-ESI (m/z) calculated: 524.1; found 525.2 [M+H] + , RT = 0.482 min (Method 2).

X H NMR (400 MHz, DMSO-d 6 ) 6 = 8.72 (s, 1H), 8.55 (s, 1H), 8.24 - 8.11 (m, 1H), 7.54 - 7.21 (m, 2H), 6.96 (d, J = 8.9 Hz, 1H), 6.46 (d, J = 4.6 Hz, 1H), 6.11 (d, J = 7.8 Hz, 1H), 5.34 (q, J = 9.1 Hz, 2H), 3.96 - 3.79 (m, 1H), 3.70 (s, 1H), 1.97 - 1.73 (m, 6H), 1.72 - 1.60 (m, 2H).

EXAMPLE 10

SYNTHESIS OF EXAMPLE 10

Synthesis of 3-chloro-l-(2-fluoroethyl)-N-((ls,4s)-4-((2-(trifluoromethyl )imidazorL2- a1pyridin-5-yl)amino)cvclohexyl)-lH-pyrazole-4-carboxamide

Example 10 To a solution of 3-chloro-N-[4-[[2-(tnfluoromethyl)imidazo[l,2-a]pyridm-5- yl]amino]cyclohexyl]-lH-pyrazole-4-carboxamide(50 mg, 117.14 umol, 1 eq) , 1- fluoro-2-iodo-ethane (20.38 mg, 117.14 umol, 1 eq) in MeCN (2 mL) was added Cs2CO3(l 14.50 mg, 351.43 umol, 3 eq). The reaction was stirred at 50 °C for 2 hr. The reaction mixture was filtered through the filter membrane. The filtrate was purified by Prep-HPLC with the following conditions column: Phenomenex luna C18 150*25mm*10um;mobile phase: [water(FA)-ACN];B%: 32%-62%,10.5min. The compound 3-chloro-l-(2-fluoroethyl)-N-[4-[[2-(trifluoromethyl)imidazo [l,2-a]pyridin- 5-yl]amino]cyclohexyl]pyrazole-4-carboxamide (5.3 mg, 11.12 umol, 9.49% yield, 99.2% purity) was obtained as white solid.

LCMS-ESI (m/z) calculated: 472.1; found 472.9 [M+H] + , RT = 0.520 min (Method 2).

'H NMR (400MHz, DMSO-d6) 8 = 8.71 (s, 1 H). 8.36 (s, 1 H) 7.70 (d, J=6.63 Hz, 1 H) 7.35 (t, J=8.25 Hz, 1 H) 6.95 (d, J=8.88 Hz, 1 H) 6.48 (d, J=5.13 Hz, 1 H) 6.11 (d, J=7.63 Hz, 1 H) 4.65 - 4.89 (m, 2 H) 4.35 - 4.52 (m, 2H) 3.91 (s, 1 H) 3.66 (s, 1 H) 1.74 - 1.91 (m, 6 H) 1.69 (dd, J=8.94, 3.81 Hz, 2 H).

EXAMPLE 11

SYNTHESIS OF EXAMPLE 11

I l l Synthesis of 3 -chloro- l-(2,2-difluoroethyl)-N-(( ls,4s)-4-((2-

(trifluoromethyl)imidazori,2-a1pyridin-5-yl)amino)cyclohe xyl)-lH-pyrazole-4- carboxamide

Example 11

To a solution of 3-chloro-N-[4-[[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino]cyclohexyl]-lH-pyrazole-4-carboxamide(50 mg, 117.14 umol, 1 eq), 1,1- difluoro-2-iodo-ethane (22.49 mg, 117.14 umol, 1 eq) in MeCN (2 mL) was added Cs2CO3(l 14.50 mg, 351.43 umol, 3 eq). The reaction was stirred at 50 °C for 14 hr. The reaction mixture was filtered through the filter membrane. The filtrate was purified by Prep-HPLC with the following conditions column: Phenomenex luna Cl 8 150*25mm*10um;mobile phase: [water(FA)-ACN];B%: 34%-64%,10.5min. The compound 3-chloro-l-(2,2-difluoroethyl)-N-[4-[[2-(trifluoromethyl)imi dazo[l,2- a]pyridin-5-yl]amino]cyclohexyl]pyrazole-4-carboxamide (3.9 mg, 7.81 umol, 6.67% yield, 98.3% purity) was obtained as yellow solid.

LCMS-ESI (m/z) calculated: 490.1; found 491.2 [M+H] + , RT = 0.539 min (Method 2).

1 HNMR (400 MHz, DMSO-d 6 ) 8 = 8.71 (s, 1 H) 8.36 (s, 1 H) 7.79 (d, J=6.63 Hz, 1 H) 7.35 (t, J=8.25 Hz, 1 H) 6.95 (d,J=8.88 Hz, 1 H) 6.45 - 6.59 (m, 1 H) 6.22 - 6.43 (m, 1 H) 6.11 (d, J=7.63 Hz, 1 H) 4.65 (td, J=15.26, 3.13 Hz, 2 H) 3.90 (s, 1 H) 3.66 (s, 1 H) 1.74 - 1.96 (m, 6 H) 1.69 (dd, J=8.94, 3.81 Hz, 2 H). EXAMPLE 12

SYNTHESIS OF EXAMPLE 12

Synthesis of 3 -chloro- l-(2,2,2-trifluoroethyl)-N-((ls,4s)-4-((2- (trifluoromethyl)imidazorL2-a1pyridin-5-yl)amino)cyclohexyl) -lH-pyrazole-4- carb oxami de

To a solution of 3-chloro-N-[4-[[2-(trifluoromethyl)imidazo[l,2-a]pyridin-5- yl]amino]cyclohexyl]-lH-pyrazole-4-carboxamide(50 mg, 117.14 umol, 1 eq) , 2,2,2- trifluoroethyl trifluoromethanesulfonate (27.19 mg, 117.14 umol, 1 eq) in MeCN (2 mL) was added CS2CO3 (114.50 mg, 351.43 umol, 3 eq). The reaction was stirred at 50 °C for 2 hr. The reaction mixture was filtered through the filter membrane. The filtrate was purified by Prep-HPLC with the following conditions column: Phenomenex luna C18 150*25mm*10um;mobile phase: [water(FA)-ACN];B%: 37%-67%,10.5min. The compound 3-chloro-l-(2,2,2-trifluoroethyl)-N-[4-[[2-(trifluoromethyl) imidazo[l,2- a]pyridin-5-yl]amino]cyclohexyl]pyrazole-4-carboxamide (16.3 mg, 32.03 umol, 27.34% yield, 100% purity) was obtained.

LCMS-ESI (m/z) calculated: 508.1; found 508.9 [M+H] + , RT = 0.560 min (Method 2). 1HNMR (400 MHz, DMSO-d 6 ) 8 = 8.71 (s, 1 H) 8.40 (s, 1 H) 7.88 (d, J=6.63

Hz, 1 H) 7.35 (t,J=8.25 Hz, 1 H) 6.95 (d, J=8.88 Hz, 1 H) 6.46 (d, J=4.88 Hz, 1 H) 6.11 (d, J=7.63 Hz, 1 H) 5.21 (q, J=9.05 Hz, 2 H) 3.90 (d,J=2.63 Hz, 1 H) 3.67 (s, 1 H) 1.75 - 1.95 (m, 6 H) 1.61 - 1.74 (m, 2 H).

The compounds listed in Table 2 were made using the procedures of Scheme 2.

Table 2

EXAMPLE 13

SYNTHESIS OF EXAMPLE 13 Synthesis of 2-(trifluoromethyl)imidazolL2-a1pyrimidin-5-ol

A solution of 2-aminopyrimidin-4-ol (2 g, 18.00 mmol, 1 eq) and 3-bromo- l,l,l-trifluoro-propan-2-one (3.44 g, 18.00 mmol, 1.87 mL, 1 eq) in EtOH (15 mL). The reaction was stirred at 130 °C for 2 hr under microwave. The reaction mixture was dissolved in MeOH and filtered by a filter membrane. The filtrate was purified by reversed phase HPLC (0.1%FA in water, MeCN) to give a crude product. The crude product was purified by column chromatography on silica gel eluting with (SiCh, Petroleum ether/Ethyl acetate=l/O to 3/2, Rf = 0.60, The TLC (petroleum ether/ethyl acetate=O/l) indicated new spots (Rf=0.70, Rf=0.60, Rf=0.40) formed.). The filtrate was purified by reversed phase HPLC (0.1%FA in water, MeCN) to give a pure product. Compound 2-(trifluoromethyl)imidazo[l,2-a]pyrimidin-5-ol (200 mg, 984.63 umol, 5.47% yield, N/A purity) was obtained as white solid.

LCMS-ESI (m/z) calculated: 203.1; found 204.1 [M+H] + , RT = 0.411 min (Method 11).

X H NMR (400 MHz, DMSO-d 6 ) 8 = 13.26 - 12.88 (m, 1H), 8.17 (d, J = 1.3 Hz, 1H), 8.01 (d, J = 7.5 Hz, 1H), 5.82 (d, J = 7.5 Hz, 1H).

Synthesis of 5-chloro-2-(trifluoromethyl)imidazorL2-a]pyrimidine

To a solution of 2-(trifluoromethyl)imidazo[l,2-a]pyrimidin-5-ol (50 mg, 246.16 umol, 1 eq) in POCL (1 mL) at 25 °C, the reaction solution was heated to 100 °C, the mixture was stirred at 100 °C for 2 hr. The reaction mixture was concentrated under reduced pressure to remove POCL. The residue was purified by prep-TLC (SiCh, Petroleum ether : Ethyl acetate = 1 : 1) collect point of Rf=0.6. Compound 5-chloro-2- (trifhioromethyl)imidazo[l,2-a]pyrimidine (41 mg, 181.34 umol, 73.67% yield, 98% purity) was obtained as a white solid.

LCMS-ESI (m/z) calculated: 221.5; found 222.0 [M+H] + , RT = 0.339 min (Method 11). Synthesis of 4-methoxy-N-r4- (tnfluoromethyl)imidazorL2-a1pynmidin-5- yllaminolcyclohexyllbenzamide

A mixture of 5-chloro-2-(trifluoromethyl)imidazo[l,2-a]pyrimidine (41 mg, 185.05 umol, 1 eq) ,N-(4-aminocyclohexyl)-4-methoxy-benzamide (91.90 mg, 370.09 umol, 2 eq) and TEA (56.17 mg, 555.14 umol, 77.27 uL, 3 eq) in NMP (2 mL) , the mixture was stirred at 25 °C for 12 hr under N2 atmosphere. The reaction solution was filtered by a filter membrane. The residue was purified by prep-HPLC (column: Phenomenex Synergi Polar-RP 100*25mm*4um;mobile phase: [water(0.225%FA)- ACN];B%: 26%-56%,8min). Compound 4-methoxy-N-[4-[[2-

(trifluoromethyl)imidazo[l,2-a]pyrimidin-5-yl]amino]cyclo hexyl]benzamide (23.9 mg, 53.43 umol, 28.88% yield, 96.9% purity) was obtained.

LCMS-ESI (m/z) calculated: 433.4; found 434.2 [M+H] + , RT = 0.446 min (Method 4).

'H NMR(400 MHz, DMSO-d 6 ) 8 = 8.73 (d, J = 0.9 Hz, 1H), 8.31 (d, J = 5.4 Hz, 1H), 7.99 (d, J = 6.0 Hz, 1H), 7.91 - 7.80 (m, 2H), 7.55 (d, J= 6.3 Hz, 1H), 6.99 (d, J = 8.9 Hz, 2H), 6.33 (d, J = 5.6 Hz, 1H), 3.97 - 3.89 (m, 1H), 3.81 (s, 4H), 1.98 - 1.87 (m, 4H), 1.85 -1.76 (m, 2H), 1.74 - 1.63 (m, 2H).

EXAMPLE 14

MRGPRX2 ACTIVITY

CHO cells stably transfected to express human MRGPRX2 were maintained in an incubator at 37° C with 5% CO2 and grown in F12 (HAM) media with 10% fetal bovine serum (FBS), 1% Glutamax, 1% penicillin /streptomycin, 800 pg/mL Geneticin (G418), and 300 pg/mL Hygromycin B. Cells were plated in a 384-well assay plate at 20,000 cells per well in 12 pL of Opti-MEM and kept in an incubator overnight. On the day of the assay, compounds solubilized at 10 mM in DMSO were added as a 10-point curve (30 uM final top concentration with 1 :3 serial dilutions) using a Tecan D300E digital dispenser. Agonists were diluted in assay buffer (final concentrations of 5.7 mM Tris-HCl, 43 mM NaCl, 50 mM LiCl, pH= 8) and 2 pL of the agonist Cortistatin-14 (CPC Scientific, catalog CORT-002) are added to each well. Final concentrations of agonists were 0.3 pM Cortistatin-14. Final concentrations of DMSO were kept consistent across the plate. Plates were incubated in the dark for 1 hr at 37 °C and then for 1 hr at room temperature. IP-1 standards and HTRF detection reagents were added according to the IP-One - Gq Kit purchased from Cisbio (part number 62IPAPEJ) and incubated in the dark for 1 hr at room temperature. The plate was read on a Molecular Devices SpectraMax iD5 plate reader. The HTRF ratio was calculated from the raw data and graphed using GraphPad Prism to calculate an IC50 value for each compound.

Activity data for selected MRGPRX2 antagonists (versus 0.3 uM Corstitatin-14 agonist) are displayed in Table 3. The activity ranges are denoted as follows: “+++++” denotes antagonist activity <50 nM; “++++” denotes antagonist activity between 51 and 100 nM; “+++” denotes activity between 101 and 500 nM; “++” denotes activity between 501 and 1000 nM; and “+” denotes activity >1000 nM

Table 3

EXAMPLE 15

MAST CELL BETA-HEXOSAMINIDASE RELEASE ASSAY

Human LAD2 cells (NIH) were maintained in an incubator at 37°C with 5% CO2 and cultured in StemPro-34 serum-free media (Gibco 10639011) supplemented with 2 mM L-glutamine, 100 U/ml penicillin, 50 mg/ml streptomycin and 100 ng/ml SCF (Invitrogen PEP0860), at a concentration of 2-5 x 10 5 cells/mL, with hemidepletion every 1-2 weeks.

Cells were transferred to SCF-Free medium at 2.5 x 10 5 cells/mL and kept in an incubator overnight. On the day of the assay, cells were washed twice in Assay Buffer (final concentrations 10 mM HEPES, 137 mM NaCl, 5.6 mM D-glucose, 2.7 mM KC1, 1 mM MgCl, 1.8 mM CalCl 2 , 0.4 mM Na 2 HPO 4 .7H 2 O, 0.04% BSA, pH = 7.4) and plated in a 96-well v-bottom plate at 20,000 cells per well in 80 pL Assay Buffer. Antagonist compounds solubilized at 10 mM in DMSO were diluted in Assay Buffer to 10X final desired concentrations as a 10-point curve (10 pM final top concentration with 1 :3 serial dilutions) and 10 pL added per well. Plates were then incubated for 1 hour at 37°C. Agonists were diluted in Assay Buffer to 10X desired concentrations and 10 pL of the appropriate agonist added to each well. The final concentration of Cortistatin-14 (Tocris 3374) used in antagonist assays was 500 nM. Final concentrations of DMSO were kept consistent across the plate. Plates were then incubated in a warm air oven for 30 minutes at 37°C, followed by centrifugation at 4°C for 5 minutes at 450 x g. 50 pL supernatant from each well was then transferred to a 96-well flat bottom plate containing 100 pL substrate solution per well (3.5 mg/mL /?-Nitrophenyl-N-acetyl-P-D-glucosaminide (Sigma 487052) in Citrate Buffer containing a final concentration of 40 mM Citric Acid, 20 mM Na 2 HPO4.7H 2 O, pH = 4.5). To the cell pellets left in the remaining assay buffer, 150 pL 0.1% Triton-X-100 was then added to each well, resuspended by pipetting up and down, and 50 pL cell lysates transferred to a second 96-well flat bottom plate containing 100 pL substrate solution per well. Plates containing transferred supernatant and cell lysates were then incubated in a warm air oven for 90 minutes at 37°C. After incubation, 50 pL of 400 mM Glycine buffer (pH 10.7) was added into each well and the plate was read on a Molecular Devices SpectraMax iD5 plate reader (absorbance at 405 nm with reference filter at 620 nm). After background subtraction, the percentage degranulation (percent beta-hexosaminidase release) was calculated as 100 x (supernatant values)/(supernatant + lysate values), followed by analysis using GraphPad Prism software to calculate an IC50 value for each compound.

Activity data for selected MRGPRX2 antagonists in the Mast Cell Beta- Hexosaminidase Release Assay are displayed in Table 4. The activity ranges are denoted as follows: “+++++” denotes antagonist activity <50 nM; “++++” denotes antagonist activity between 51 and 100 nM; “+++” denotes activity between 101 and 500 nM; “++” denotes activity between 501 and 1000 nM; and “+” denotes activity >1000 nM.

Table 4

The various embodiments described above can be combined to provide further embodiments. All of the 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 modified, 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. This application claims the benefit of priority to U.S. Provisional Application No.

63/130,525, filed December 24, 2020, which application is hereby incorporated by reference in its entirety.