BICYCLIC HETEROCYCLES AS MRGPRX2 ANTAGONISTS

Information

  • Patent Application
  • 20250084087
  • Publication Number
    20250084087
  • Date Filed
    August 16, 2024
    9 months ago
  • Date Published
    March 13, 2025
    2 months ago
Abstract
The present disclosure relates to bicyclic heterocycles of Formula (I), and pharmaceutical compositions of the same, that are modulators, antagonists, or inhibitors of the G protein-coupled receptor MRGPRX2 and are useful in the treatment of MRGPRX2 dependent conditions such as inflammatory diseases.
Description
FIELD

The present disclosure relates to bicyclic heterocycles, and pharmaceutical compositions of the same, that are modulators, antagonists or inhibitors of the G protein-coupled receptor MRGPRX2 and are useful in the treatment of MRGPRX2 dependent conditions such as inflammatory diseases.


BACKGROUND

Mas-related G protein-coupled receptor X2 (MRGPRX2) is an orphan, seven transmembrane G protein-coupled receptor that is almost exclusively expressed on connective tissue mast cells. MRGPRX2 belongs to a G protein-coupled receptor subfamily X, comprised of four members X1-X4, specific to humans and primates. MRGPRX2 is a low affinity promiscuous receptor for cyclic and polybasic structure ligands that mediates mast cell degranulation in response to multiple endogenous and exogenous stimuli.


Mast cells constitute an integral part of the human immune system. They are important modulators of inflammatory and physiological processes. MRGPRX2 receptor plays a pivotal role in itch, allergy and inflammation. Activation of the receptor by neuropeptides, antimicrobial host defense peptides as well as numerous FDA-approved drugs leads to mast cell degranulation and release of inflammatory mediators through immunoglobulin-independent pathway (M. Thapaliya, et al., Curr. Allergy Asthma Rep., 2021, 21(1), 3).


Activation of MRGPRX2 receptor drives non-histaminergic itch in chronic refractory pruritus and MRGPRX2 has also been implicated in senile itch (A. He, et al. Biomed. Res. Int., 2017, 4790810; J. Meixiong, et al., Immunity, 2019, 50(5), 1163-71). There is an increased MRGPRX2 gene expression on mast cells in the skin of patients with severe chronic urticaria (hives) (H. Ali, J. Immunobiol., 2016, 1(4), 115; D. Fujisawa, et al., J. Allergy Clin. Immunol. 2014, 134(3), 622-33). Additionally, activation of MRGPRX2 by elevated levels of proadrenomedullin N-terminal 20 peptide (PAMP1-20) in the skin of patients with allergic contact dermatitis (ACD) leads to intensely itchy eczematous skin rash (J. Meixiong, et al., Immunity, 2019, 50(5):1163-71). MRGPRX2 is also involved in the pathogenesis of acne rosacea where dysregulation of the host defense mechanism due to excessive LL-37 antimicrobial peptide production leads to enhanced mast cell activation through MRGPRX2 (H. Ali, Adv. Immunol., 2017, 136, 123-62). MRGPRX2 is also implicated in systemic mastocytosis and in neurogenic inflammation, pain and itch. Substance P released from nerve endings and directly from mast cells in sickle cell anemia patients activates mast cells via MRGPRX2 causing painful crisis (H. Subramanian, et al., J. Allergy Clin. Immunol., 2016, 138(3), 700-10).


The role of MRGPRX2 in mast cell biology is further supported by the fact that naturally occurring missense MRGPRX2 variants: G165E, D184H, W243R, and H259Y inhibit mast cell degranulation in response to endogenous neuropeptides and drugs (I. Alkanfari, et al., J. Immuol., 2018, 201(2), 343-49).


Taken together, these findings suggest that MRGPRX2 plays a critical role in itch, pain, and inflammation. Potential disease indications for MRGPRX2 antagonist encompass chronic urticaria and pruritus (hives/itch), acne rosacea, and systemic mastocytosis. These clinical indications present high unmet medical need, particularly in antihistamine-refractory patients.


Therefore, targeting of MRGPRX2 receptor can be useful in the clinical treatment of mast-cell mediated diseases.


Compounds that modulate MRGPRX2 are discussed in WO2006066599A2, WO2008052072A2, WO2020223255A1, WO2021092240A1, WO2021092262A1, WO2021092264A1, WO2022067094A1, WO2022073904A1, WO2022073905A1, WO2022087083A1, WO2022111473A1, WO2022125636A1, WO2022140520A1, WO2022152852A1, WO2022152853A1, and WO2023039448A1.


There remains a need for new compounds that are effective as modulators, antagonists or inhibitors of MRGPRX2.


SUMMARY

The present disclosure is directed to compounds having Formula (I):




embedded image


or pharmaceutically acceptable salts thereof, wherein constituent variables are defined herein.


The present disclosure is further directed to pharmaceutical compositions comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.


The present disclosure is further directed to methods of modulating such as by antagonizing or inhibiting MRGPRX2 protein comprising contacting the protein with a compound of Formula (I), or a pharmaceutically acceptable salt thereof.


The present disclosure is further directed to a method of treating MRGPRX2 dependent conditions, comprising administering a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a patient in need thereof. The present disclosure is further directed to the use of compounds of Formula (I) and pharmaceutically acceptable salts thereof in the preparation of a medicament for use in therapy. The present disclosure is further directed to compounds of Formula (I) and pharmaceutically acceptable salts thereof for use in therapy.







DETAILED DESCRIPTION
I. Definitions

For the terms “e.g.” and “such as,” and grammatical equivalents thereof, the phrase “and without limitation” is understood to follow unless explicitly stated otherwise.


The singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise.


The term “about” means “approximately” (e.g., plus or minus approximately 10% of the indicated value).


The following abbreviations may be used herein: AcOH (acetic acid); aq. (aqueous); Boc (t-butoxycarbonyl); br (broad); BSA (bovine serum albumin); BTK (Bruton tyrosine kinase); CHO-K1 (Chinese hamster ovary); CSA (cyclosporine A); CTMCs (connective tissue-type mast cells); CYP (cytochrome P450); d (doublet); DBU (1,8-diazabicyclo[5.4.0]undec-7-ene); dd (doublet of doublets); DCM (dichloromethane); DIPEA (N,N-diisopropylethylamine); DMAC (dimethylacetamide); DMEM (Dulbecco's Modified Eagle Medium); DMF (N,N-dimethylformamide); DMSO (dimethylsulfoxide); EDTA (ethylenediaminetetraacetic acid); Et (ethyl); EtOAc (ethyl acetate); Et2O (diethyl ether); EtOH (ethanol); FBS (fetal bovine serum); FcεR1 (high-affinity IgE receptor); FCC (flash column chromatography); FLIPR (Fluorescence Imaging Plate Reader); g (gram(s)); h (hour(s)); GPCR (G protein-coupled receptor); HATU (N,N,N′,N′-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate); HBSS (Hanks' Balanced Salt Solution); HCl (hydrochloric acid); HEPES (N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid); Hex (hexanes); HLM (human liver microsome); HPLC (high performance liquid chromatography); hsMC (human skin mast cells); HTRF (Homogeneous Time Resolved Fluorescence); Hz (hertz); IgE (immunoglobulin E); IV (intravenous); J (coupling constant); JAK (janus kinase); IP1 (myo-Inositol 1 phosphate); K2CO3 (potassium carbonate); KOt-Bu (potassium tert-butoxide); KOH (potassium hydroxide); LCMS (liquid chromatography-mass spectrometry); LiHMDS (lithium bis(trimethylsilyl)amide); m (multiplet); M (molar); mm (millimeter(s)); MS (Mass spectrometry); Me (methyl); MeCN (acetonitrile); MeOH (methanol); MgSO4 (magnesium sulfate); NaBH4 (sodium borohydride); NADPH (nicotinamide adenine dinucleotide phosphate); NaHCO3 (sodium bicarbonate); Na2CO3 (sodium carbonate); NaOt-Bu (sodium tert-butoxide); Na2SO3 (sodium sulfite); Na2SO4 (sodium sulfate); Na2S2O3 (sodium thiosulfate); NFAT (Nuclear factor of activated T cells); NH4Cl (ammonium chloride); nm (nanometer(s)); PBS (phosphate-buffered saline); Ph (phenyl); prep. (preparative); PK (pharmacokinetic); PO (oral); rpm (revolutions per minute); r.t. (room temperature); RFU (Relative Fluorescence Unit); Ruphos Pd G4 ([dicyclohexyl(2′,6′-diisopropoxy-2-biphenylyl)phosphine-κP](methanesulfonatato-xO)[2′-(methylamino-κN)-2-biphenylyl-κC2]palladium); s (singlet); sat. (saturated); SCF (stem cell factor); t (triplet or tertiary); TDI (time dependent inhibition); TEER (transepithelial electrical resistance); tert (tertiary); Tf2O (trifluoromethanesulfonic anhydride); tt (triplet of triplets); t-Bu (tert-butyl); TEA (triethylamine); TFA (trifluoroacetic acid); THE (tetrahydrofuran); v/v (volume per volume); wt % (weight percent); w/v (weight in volume), μg (microgram(s)); μL (microliter(s)); μm (micrometer(s)).


Additional definitions are provided elsewhere in the present disclosure.


II. Compounds

The present disclosure provides a compound having Formula (I):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • X1 is N or CR1;
    • X2 is N or CR2;
    • X3 is N or CR3;
    • X4 is N or CR4;
    • provided that at least one of X3 and X4 is N;
    • X5 is N or CR5;
    • X6 is N or CR6;
    • R1, R2, R3, R4, R5, and R6 are each independently selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, or P(O)(NH2)ORa1; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R1, R2, R3, R4, R5, and R6 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10A; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R1, R2, R3, R4, R5, and R6 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B.
    • R7 and R8 are each independently selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl, wherein the C3-10 cycloalkyl and 4-10 membered heterocycloalkyl forming R7 and R8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R20A; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R7 and R8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R20B;
    • A is a group of the Formula (A);




embedded image




    • wherein custom-character represents the point of attachment of A to the remainder of the molecule;

    • R9A and R9B, together with the carbon atom to which they are both attached, form a C3-12 cycloalkane or 4-12 membered heterocycloalkane ring, wherein the C3-12 cycloalkane or 4-12 membered heterocycloalkane ring formed by R9A and R9B is optionally substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from RA;

    • Cy is a ring selected from C6-10 aryl and 5-10 membered heteroaryl; wherein the C6-10 aryl and 5-10 membered heteroaryl forming Cy are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from RA;

    • each RA is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRa1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)2NRc1Rd1, S(O)(═NRe1)Rb1, P(O)(ORa1)(ORa1), P(O)(ORa1)Rb1, P(O)(NH2)Rb1, P(O)(NH2)(ORa1), and B(ORa1)(ORa1); wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2;

    • each RA1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and RA2; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA1 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2; each RA2 is independently selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, OR3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)OR3, NRc3Rd3, NRc3C(O)Rb3NRc3C(O)ORa3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, S(O)2NRc3Rd3, S(O)(═NRe3)Rb3, P(O)(ORa3)(ORa3), P(O)(ORa3)Rb3, P(O)(NH2)Rb3, P(O)(NH2)(ORa3), and B(ORa3)(ORa3); wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RA2 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA3;

    • each RA3 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa8, SRa8, C(O)Rb8, C(O)NRc8Rd8, C(O)ORa8, OC(O)Rb8, OC(O)NRc8Rd8, NRc8Rd8, NRc8C(O)Rb8, NRc8C(O)ORa8, NRc8C(O)NRc8Rd8, C(═NRe8)Rb8, C(═NORa8)Rb8, C(═NRe8)NRc8Rd8, NRc8C(═NRe8)NRc8Rd8, NRc8S(O)Rb8, NRc8S(O)2Rb8, NRc8S(O)2NRc8Rd8, S(O)Rb8, S(O)NRc8Rd8, S(O)2Rb8, and S(O)2NRc8Rd8; wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RA3 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA3 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • each RCy is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NORa2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRe2Rd2; wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RCy are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RCy are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy2;

    • each RCy1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and RCy2; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RCy1 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy2; each RCy2 is independently selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)ORa4, NRc4S(O)Rb4, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4, S(O)Rb4, S(O)NRc4Rd4, S(O)2Rb4, and S(O)2NRc4Rd4; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RCy2 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • each R10A is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and R10B; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R10A are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R10B;

    • each R10B is independently selected from C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa5, SRa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, OC(O)Rb5, OC(O)NRc5Rd5, NRc5Rd5, NRc5C(O)Rb5, NRc5C(O)ORa5, NRa5C(O)NRc5Rd5, C(═NRc5)Rb5, C(═NORa5)Rb5, C(═NRe5)NRc5Rd5, NRc5C(═NRe5)NRc5Rd5, NRc5S(O)Rb5, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5 S(O)Rb5, S(O)NRc5Rd5, S(O)2Rb5, and S(O)2NRc5Rd5; wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R10B are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R11;

    • each R11 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, ORa6, SR a, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R11 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R11 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • each R20A is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, and C1-6 haloalkyl; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R20A are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R20B;

    • each R20B is independently selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)ORa7, NRc7S(O)Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, and S(O)2NRc7Rd7; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R20B are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • each Ra1, Rc1, Rd1, Ra2, Rc2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra5, Rc5, Rd5, Ra6, Rc6, Rd6, Ra7, Rc7, Rd7, Ra8, Rc8, and Rd8 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl forming Ra1, Rc1, Rd1, Ra2, Re2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra5, Rc5, Rd5, Ra6, Rc6, Rd6, Ra7, Rc7, Rd7, Ra8, Rc8, and Rd8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Ra1, Rc1, Rd1, Ra2, Re2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra5, Rc5, Rd5, Ra6, Rc6, Rd6, Ra7, Rc7, Rd7, Ra8, Rc8, and Rd8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • or any Rc1 and Rd1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc2 and Rd2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc4 and Rd4 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc5 and Rd5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc6 and Rd6 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc7 and Rd7 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Re8 and Rd8 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • each Rb1, Rb2, Rb3, Rb4, Rb5, Rb6, Rb7, and Rb8 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl forming Rb1, Rb2, Rb3, Rb4, Rb5, Rb6, Rb7, and Rb8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Rb1, Rb2, Rb3, Rb4, Rb5, Rb6, Rb7, and Rb8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • each Re1, Re2, Re3, Re5, and Re8 is independently selected from H, CN, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkylthio, C1-6 alkylsulfonyl, C1-6 alkylcarbonyl, C1-6 alkylaminosulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, aminosulfonyl, C1-6 alkylaminosulfonyl and di(C1-6 alkyl)aminosulfonyl;

    • each Rg1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and Rg2; and

    • each Rg2 is independently selected from D, OH, NO2, CN, halo, C3-6 cycloalkyl, HO—C3-6 cycloalkyl, C3-6 cycloalkyl-C1-2 alkylene, 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkyl-C1-2 alkylene, C1-6 alkoxy, C1-6 haloalkoxy, C1-3 alkoxy-C1-3 alkyl, C1-3 alkoxy-C1-3 alkoxy, HO—C1-3 alkoxy, HO—C1-3 alkyl, cyano-C1-3 alkyl, H2N—C1-3 alkyl, amino, C1-6 alkylamino, di(C1-6 alkyl)amino, thio, C1-6 alkylthio, C1-6 alkylsulfinyl, C1-6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, carboxy, C1-6 alkylcarbonyl, C1-6 alkoxycarbonyl, C1-6 alkylcarbonylamino, C1-6 alkylsulfonylamino, aminosulfonyl, C1-6 alkylaminosulfonyl, di(C1-6 alkyl)aminosulfonyl, aminosulfonylamino, C1-6 alkylaminosulfonylamino, di(C1-6 alkyl)aminosulfonylamino, aminocarbonylamino, C1-6 alkylaminocarbonylamino, and di(C1-6 alkyl)aminocarbonylamino;

    • wherein at each occurrence, a heterocycloalkyl group has at least one ring-forming carbon atom and 1, 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; the N and S ring-forming heteroatoms of the heterocycloalkyl group are optionally oxidized; and a ring-forming carbon atom of the heterocycloalkyl group is optionally substituted by oxo to form a carbonyl group;

    • at each occurrence, a heterocycloalkane ring has at least one ring-forming carbon atom and 1, 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; the N and S ring-forming heteroatoms of the heterocycloalkane ring are optionally oxidized; and a ring-forming carbon atom of the heterocycloalkane ring is optionally substituted by oxo to form a carbonyl group; and

    • at each occurrence, a heteroaryl group has at least one ring-forming carbon atom and 1, 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; the N and S ring-forming heteroatoms of the heteroaryl group are optionally oxidized; and a ring-forming carbon atom of the heteroaryl group is optionally substituted by oxo to form a carbonyl group





The present disclosure further provides a compound having Formula (I):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • X1 is N or CR1;
    • X2 is N or CR2;
    • X3 is N or CR3;
    • X4 is N or CR4;
    • provided that at least one of X3 and X4 is N;
    • X5 is N or CR5;
    • X6 is N or CR6;
    • R1, R2, R3, R4, R5, and R6 are each independently selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C310 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R1, R2, R3, R4, R5, and R6 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10A; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R1, R2, R3, R4, R5, and R6 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B;
    • R7 and R8 are each independently selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl, wherein the C3-10 cycloalkyl and 4-10 membered heterocycloalkyl forming R7 and R8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R20A; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R7 and R8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R20B;
    • A is a group of the Formula (A);




embedded image




    • wherein custom-character represents the point of attachment of A to the remainder of the molecule;

    • R9A and R9B, together with the carbon atom to which they are both attached, form a C3-12 cycloalkane or 4-12 membered heterocycloalkane ring, wherein the C3-12 cycloalkane or 4-12 membered heterocycloalkane ring formed by R9A and R9B is optionally substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from RA;

    • Cy is a ring selected from C6-10 aryl and 5-10 membered heteroaryl; wherein the C6-10 aryl and 5-10 membered heteroaryl forming Cy are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Ry;

    • each RA is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRc1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)2NRc1Rd1, S(O)(═NRe1)Rb1, P(O)(ORa1)(ORa1), P(O)(ORa1)Rb1, P(O)(NH2)Rb1, P(O)(NH2)(ORa1), and B(ORa1)(ORa1); wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2;

    • each RA1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and RA2; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA1 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2;

    • each RA2 is independently selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, OR3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)OR3, NRc3Rd3, NRc3C(O)Rb3NRc3C(O)ORa3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, S(O)2NRc3Rd3, S(O)(═NRe3)Rb3, P(O)(ORa3)(ORa3), P(O)(ORa3)Rb3, P(O)(NH2)Rb3, P(O)(NH2)(ORa3), and B(ORa3)(ORa3); wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RA2 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA3;

    • each RA3 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa8, SRa8, C(O)Rb8, C(O)NRc8Rd8, C(O)ORa8, OC(O)Rb8, OC(O)NRc8Rd8, NRc8Rd8, NRc8C(O)Rd8, NRc8C(O)ORa8, NRc8C(O)NRc8Rd8, C(═NRe8)Rb8, C(═NORa8)Rb8, C(═NRe8)NRc8Rd8, NRc8C(═NRe8)NRc8Rd8, NRc8S(O)Rb8, NRc8S(O)2Rd8, NRc8S(O)2NRc8Rd8, S(O)Rd8, S(O)NRc8Rd8, S(O)2Rb8, and S(O)2NRc8Rd8; wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RA3 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA3 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • each RCy is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRe2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NORa2)Rb2, C(═NRe2)NRe2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRe2S(O)2Rb2, NRe2S(O)2NRe2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRe2Rd2; wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RCy are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RCy are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy2;

    • each RCy1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and RCy2; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RCy1 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy2; each RCy2 is independently selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)ORa4, NRc4S(O)Rb4, 5, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4, S(O)Rb4, S(O)NRc4Rd4, S(O)2Rb4, and S(O)2NRc4Rd4; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RCy2 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • each R10A is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and R10B; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R10A are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R10B;

    • each R10B is independently selected from C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa5, SRa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, OC(O)Rb5, OC(O)NRc5Rd5, NRc5Rd5, NRc5C(O)Rb5, NRc5C(O)ORa5, NRc5C(O)NRc5Rd5, C(═NRc5)Rb5, C(═NORa5)Rb5, C(═NRe5)NRc5Rd5, NRc5C(═NRe5)NRc5Rd5, NRc5S(O)Rb5, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5 S(O)Rb5, S(O)NRc5Rd5, S(O)2Rb5, and S(O)2NRc5Rd5; wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R10B are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R11;

    • each R11 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, ORa6, SR a, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R11 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R11 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • each R20A is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, and C1-6 haloalkyl; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R20A are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R20B;

    • each R20B is independently selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)ORa7, NRc7S(O)Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, and S(O)2NRc7Rd7; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R20B are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • each Ra1, Rc1, Rd1, Ra2, Re2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra8, Rc5, Rd5, Rd6, Rc6, Rd6, Ra7, Rc7, Rd7, Ra8, Rc8, and Rd8 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl forming Ra1, Rc1, Rd1, Ra2, Re2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra8, Rc5, Rd5, Ra6, R6, Rd6, Ra7, Rc7, Rd7, Ra8, Rc8, and Rd8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Ra1, Rc1, Rd1, Ra2, Re2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra8, Rc5, Rd5, Ra6, Rc6, Rd6, Ra7, Rc7, Rd7, Ra8, Rc8, and Rd8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • or any Rc1 and Rd1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc2 and Rd2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc4 and Rd4 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc5 and Rd5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc6 and Rd6 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc7 and Rd7 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Re8 and Rd8 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • each Rb1, Rb2, Rb3, Rb4, Rb5, Rb6, Rb7, and Rb8 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl forming Rb1, Rb2, Rb3, Rb4, Rb5, Rb6, Rb7, and Rb8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Rb1, Rb2, Rb3, Rb4, Rb5, Rb6, Rb7, and Rb8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • each Re1, Re2, Re3, Re5, and Re8 is independently selected from H, CN, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkylthio, C1-6 alkylsulfonyl, C1-6 alkylcarbonyl, C1-6 alkylaminosulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, aminosulfonyl, C1-6 alkylaminosulfonyl and di(C1-6 alkyl)aminosulfonyl;

    • each Rg1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and Rg2; and

    • each Rg2 is independently selected from D, OH, NO2, CN, halo, C3-6 cycloalkyl, C3-6 cycloalkyl-C1-2 alkylene, 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkyl-C1-2 alkylene, C1-6 alkoxy, C1-6 haloalkoxy, C1-3 alkoxy-C1-3 alkyl, C1-3 alkoxy-C1-3 alkoxy, HO—C1-3 alkoxy, HO—C1-3 alkyl, cyano-C1-3 alkyl, H2N—C1-3 alkyl, amino, C1-6 alkylamino, di(C1-6 alkyl)amino, thio, C1-6 alkylthio, C1-6 alkylsulfinyl, C1-6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, carboxy, C1-6 alkylcarbonyl, C1-6 alkoxycarbonyl, C1-6 alkylcarbonylamino, C1-6 alkylsulfonylamino, aminosulfonyl, C1-6 alkylaminosulfonyl, di(C1-6 alkyl)aminosulfonyl, aminosulfonylamino, C1-6 alkylaminosulfonylamino, di(C1-6 alkyl)aminosulfonylamino, aminocarbonylamino, C1-6 alkylaminocarbonylamino, and di(C1-6 alkyl)aminocarbonylamino;

    • wherein at each occurrence, a heterocycloalkyl group has at least one ring-forming carbon atom and 1, 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; the N and S ring-forming heteroatoms of the heterocycloalkyl group are optionally oxidized; and a ring-forming carbon atom of the heterocycloalkyl group is optionally substituted by oxo to form a carbonyl group;

    • at each occurrence, a heterocycloalkane ring has at least one ring-forming carbon atom and 1, 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; the N and S ring-forming heteroatoms of the heterocycloalkane ring are optionally oxidized; and a ring-forming carbon atom of the heterocycloalkane ring is optionally substituted by oxo to form a carbonyl group; and

    • at each occurrence, a heteroaryl group has at least one ring-forming carbon atom and 1, 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; the N and S ring-forming heteroatoms of the heteroaryl group are optionally oxidized; and a ring-forming carbon atom of the heteroaryl group is optionally substituted by oxo to form a carbonyl group.





The present disclosure further provides a compound having Formula (I):




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • X1 is N or CR1;
    • X2 is N or CR2;
    • X3 is N or CR3;
    • X4 is N or CR4;
    • provided that at least one of X3 and X4 is N;
    • X5 is N or CR5;
    • X6 is N or CR6;
    • R1, R2, R3, R4, R5, and R6 are each independently selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRc1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R1, R2, R3, R4, R5, and R6 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10A; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R1, R2, R3, R4, R5, and R6 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B;
    • R7 and R8 are each independently selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl, wherein the C3-10 cycloalkyl and 4-10 membered heterocycloalkyl forming R7 and R8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R20A; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R7 and R8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R20B;
    • A is a group of the Formula (A);




embedded image




    • wherein custom-character represents the point of attachment of A to the remainder of the molecule;

    • R9A and R9B, together with the carbon atom to which they are both attached, form a C3-12 cycloalkane or 4-12 membered heterocycloalkane ring, wherein the C3-12 cycloalkane or 4-12 membered heterocycloalkane ring formed by R9A and R9B is optionally substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from RA;

    • Cy is a ring selected from C6-10 aryl and 5-10 membered heteroaryl; wherein the C6-10 aryl and 5-10 membered heteroaryl forming Cy are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from RCy;

    • each RA is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRc1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)2NRc1Rd1, S(O)(═NRe1)Rb1, P(O)(ORa1)(ORa1), P(O)(ORa1)Rb1, P(O)(NH2)Rb1, P(O)(NH2)(ORa1), and B(ORa1)(ORa1); wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2;

    • each RA1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and RA2; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA1 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2;

    • each RA2 is independently selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, OR3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)OR3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)ORa3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, S(O)2NRc3Rd3, S(O)(═NRe3)Rb3, P(O)(ORa3)(ORa3), P(O)(ORa3)Rb3, P(O)(NH2)Rb3, P(O)(NH2)(ORa3), and B(ORa3)(ORa3); wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RA2 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA3;

    • each RA3 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa8, SRa8, C(O)Rb8, C(O)NRc8Rd8, C(O)ORa8, OC(O)Rb8, OC(O)NRc8Rd8, NRc8Rd8, NRc8C(O)Rb8, NRc8C(O)ORa8, NRc8C(O)NRc8Rd8, C(═NRe8)Rb8, C(═NORa8)Rb8, C(═NRe8)NRc8Rd8, NRc8C(═NRe8)NRc8Rd8, NRc8S(O)Rb5, NRc8S(O)2Rb8, NRc8S(O)2NRc8Rd8, S(O)Rb8, S(O)NRc8Rd8, S(O)2Rb8, and S(O)2NRc8Rd8; wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RA3 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA3 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • each RCy is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NORa2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRe2Rd2; wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RCy are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RCy are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy2;

    • each RCy1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and RCy2; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RCy1 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy2; each RCy2 is independently selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)ORa4, NRc4S(O)Rb4, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4, S(O)Rb4, S(O)NRc4Rd4, S(O)2Rb4, and S(O)2NRc4Rd4; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming RCy2 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • each R10A is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and R10B; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R10A are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R10B;

    • each R10B is independently selected from C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, NO2, ORa5, SRa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORas, OC(O)Rb5, OC(O)NRc5Rd5, NRc5Rd5, NRc5C(O)Rb5, NRc5C(O)ORa5, NRc5C(O)NRc5Rd5, C(═NRc5)Rb5, C(═NORa5)Rb5, C(═NRe5)NRc5Rd5, NRc5C(═NRe5)NRc5Rd5, NRc5S(O)Rb5, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5 S(O)Rb5, S(O)NRc5Rd5, S(O)2Rb5, and S(O)2NRc5Rd5; wherein the C3-10 cycloalkyl, 4-12 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-12 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R10B are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R11;

    • each R11 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, ORa6, SR a, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R11 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R11 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • each R20A is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, and C1-6 haloalkyl; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R20A are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from R20B;

    • each R20B is independently selected from C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene, 5-10 membered heteroaryl-C1-3 alkylene, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)ORa7, NRc7S(O)Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, and S(O)2NRc7Rd7; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl-C1-3 alkylene, 4-10 membered heterocycloalkyl-C1-3 alkylene, C6-10 aryl-C1-3 alkylene and 5-10 membered heteroaryl-C1-3 alkylene forming R20B are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • each Ra1, Rc1, Rd1, Ra2, Re2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra5, Rc5, Rd5, Ra6, Rc6, Rd6, Ra7, Rc7, Rd7, Ra8, Rc8, and Rd8 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl forming Ra1, Rc1, Rd1, Ra2, Re2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra5, Rc5, Rd5, Ra6, Rc6, Rd6, Ra7, Rc7, Rd7, Ra8, Rc8, and Rd8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Ra1, Rc1, Rd1, Ra2, Re2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra5, Rc5, Rd5, Ra6, Rc6, Rd6, Ra7, Rc7, Rd7, Ra8, Rc8, and Rd8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • or any Rc1 and Rd1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc2 and Rd2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc4 and Rd4 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc5 and Rd5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc6 and Rd6 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc7 and Rd7 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • or any Rc8 and Rd8 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;

    • each Rb1, Rb2, Rb3, Rb4, Rb5, Rb6, Rb7, and Rb8 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl; wherein the C3-10 cycloalkyl, 4-10 membered heterocycloalkyl, C6-10 aryl and 5-10 membered heteroaryl forming Rb1, Rb2, Rb3, Rb4, Rb5, Rb6, Rb7, and Rb8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Rb1, Rb2, Rb3, Rb4, Rb5, Rb6, Rb7, and Rb8 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;

    • each Re1, Re2, Re3, Re5, and Re8 is independently selected from H, CN, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkylthio, C1-6 alkylsulfonyl, C1-6 alkylcarbonyl, C1-6 alkylaminosulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, aminosulfonyl, C1-6 alkylaminosulfonyl and di(C1-6 alkyl)aminosulfonyl;

    • each Rg1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and Rg2; and

    • each Rg2 is independently selected from D, OH, NO2, CN, halo, C3-6 cycloalkyl, C3-6 cycloalkyl-C1-2 alkylene, C1-6 alkoxy, C1-6 haloalkoxy, C1-3 alkoxy-C1-3 alkyl, C1-3 alkoxy-C1-3 alkoxy, HO—C1-3 alkoxy, HO—C1-3 alkyl, cyano-C1-3 alkyl, H2N—C1-3 alkyl, amino, C1-6 alkylamino, di(C1-6 alkyl)amino, thio, C1-6 alkylthio, C1-6 alkylsulfinyl, C1-6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, carboxy, C1-6 alkylcarbonyl, C1-6 alkoxycarbonyl, C1-6 alkylcarbonylamino, C1-6 alkylsulfonylamino, aminosulfonyl, C1-6 alkylaminosulfonyl, di(C1-6 alkyl)aminosulfonyl, aminosulfonylamino, C1-6 alkylaminosulfonylamino, di(C1-6 alkyl)aminosulfonylamino, aminocarbonylamino, C1-6 alkylaminocarbonylamino, and di(C1-6 alkyl)aminocarbonylamino;

    • wherein at each occurrence, a heterocycloalkyl group has at least one ring-forming carbon atom and 1, 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; the N and S ring-forming heteroatoms of the heterocycloalkyl group are optionally oxidized; and a ring-forming carbon atom of the heterocycloalkyl group is optionally substituted by oxo to form a carbonyl group;

    • at each occurrence, a heterocycloalkane ring has at least one ring-forming carbon atom and 1, 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; the N and S ring-forming heteroatoms of the heterocycloalkane ring are optionally oxidized; and a ring-forming carbon atom of the heterocycloalkane ring is optionally substituted by oxo to form a carbonyl group; and

    • at each occurrence, a heteroaryl group has at least one ring-forming carbon atom and 1, 2, 3, or 4 ring-forming heteroatoms independently selected from N, O, and S; the N and S ring-forming heteroatoms of the heteroaryl group are optionally oxidized; and a ring-forming carbon atom of the heteroaryl group is optionally substituted by oxo to form a carbonyl group.





In some embodiments, the compound is not one of the following compounds:

  • 5-methyl-N-[[tetrahydro-4-(4-methoxyphenyl)-2H-pyran-4-yl]methyl]-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • 5-(methoxymethyl)-N-[(4-phenyl-4-piperidinyl)methyl]-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • N-[[4-(4-fluorophenyl)tetrahydro-2H-pyran-4-yl]methyl]-5-methyl-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • 5-(methoxymethyl)-N-[(tetrahydro-4-phenyl-2H-pyran-4-yl)methyl]-pyrazolo[1,5-a]pyrimidin-7-amine;
  • N-[[4-(3-fluorophenyl)tetrahydro-2H-pyran-4-yl]methyl]-5-propyl-pyrazolo[1,5-a]pyrimidin-7-amine;
  • 5-ethyl-N-[(1-methyl-4-phenyl-4-piperidinyl)methyl]-pyrazolo[1,5-a]pyrimidin-7-amine;
  • 5-ethyl-N-[(tetrahydro-4-phenyl-2H-pyran-4-yl)methyl]-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • 5-methyl-N-[[tetrahydro-4-(4-methoxyphenyl)-2H-pyran-4-yl]methyl]-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • 5-(4-pyridinyl)-N-[(tetrahydro-4-phenyl-2H-pyran-4-yl)methyl]-pyrazolo[1,5-a]pyrimidin-7-amine;
  • N-[[4-(3-fluorophenyl)tetrahydro-2H-pyran-4-yl]methyl]-2,3,5-trimethyl-pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2,5-dimethyl-N-[(1-methyl-4-phenyl-4-piperidinyl)methyl]-pyrazolo[1,5-a]pyrimidin-7-amine;
  • 5-methyl-N-[(1-phenylcyclopentyl)methyl]-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • N-[[1-[4-(difluoromethoxy)-3-methoxyphenyl]cyclopentyl]methyl]-5-methyl-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • 5-(methoxymethyl)-N-[(1-phenylcyclopentyl)methyl]-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • 5-methyl-N-[[1-(3-thienyl)cyclopentyl]methyl]-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • 5-methyl-N-[(1-phenylcyclobutyl)methyl]-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • N2-(5-chloro-2-methoxyphenyl)-5-methyl-N7-[(1-phenylcyclobutyl)methyl]-[1,2,4]triazolo[1,5-a]pyrimidine-2,7-diamine;
  • N2-(5-chloro-2-methoxyphenyl)-5-methyl-N7-[[1-(2-pyridinyl)cyclobutyl]methyl]-[1,2,4]triazolo[1,5-a]pyrimidine-2,7-diamine;
  • 5-methyl-N-[(1-phenylcyclobutyl)methyl]-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • N-[[1-(3-bromophenyl)cyclobutyl]methyl]-5-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine;
  • 4-chloro-2-[[5-methyl-7-[[(1-phenylcyclobutyl)methyl]amino][1,2,4]triazolo[1,5-a]pyrimidin-2-yl]amino]-phenol;
  • N2-[5-chloro-2-[2-(dimethylamino)ethoxy]phenyl]-5-methyl-N7-[(1-phenylcyclobutyl)methyl]-[1,2,4]triazolo[1,5-a]pyrimidine-2,7-diamine;
  • N-[[4-(3-bromophenyl)tetrahydro-2H-pyran-4-yl]methyl]-[1,2,4]triazolo[1,5-a]pyrazin-5-amine; and
  • 3-phenyl-3-[([1,2,4]triazolo[1,5-a]pyrazin-5-ylamino)methyl]-1-(trifluoromethyl)-cyclobutanol.


In some embodiments, the compound is not one of the following compounds:

  • N2-(5-chloro-2-methoxyphenyl)-5-methyl-N7-[(1-phenylcyclobutyl)methyl]-[1,2,4]triazolo[1,5-a]pyrimidine-2,7-diamine; and
  • N2-[5-chloro-2-[2-(dimethylamino)ethoxy]phenyl]-5-methyl-N7-[(1-phenylcyclobutyl)methyl]-[1,2,4]triazolo[1,5-a]pyrimidine-2,7-diamine.


In some embodiments, the compound has Formula (II-A), (II-B), (IT-C), (II-D), (II-E), or (II-F):




embedded image


or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, and A are as defined herein.


In some embodiments, A is a group of the Formula (A-1), (A-2), (A-3), (A-4), (A-5), (A-6), (A-7), (A-8), (A-9), (A-10), (A-11), (A-12), (A-13), (A-14), (A-15), (A-16), (A-17), (A-18), or (A-19):




embedded image


embedded image


embedded image




    • wherein custom-character represents the point of attachment of A to the remainder of the molecule;

    • 0, 1, 2, 3, 4, 5, or 6 of the groups RA10 are independently selected from RA and the remainder the groups RA10 are H; and

    • Cy and RA are as defined herein.





In some embodiments, A is a group of the Formula (A-20), (A-21), (A-22), (A-23), (A-24), (A-25), (A-26), (A-27), (A-28), (A-29), (A-30), (A-31), (A-32), (A-33), (A-34), (A-35), (A-36), (A-37), (A-38), (A-39), or (A-40):




embedded image


embedded image


embedded image




    • wherein custom-character represents the point of attachment of A to the remainder of the molecule;

    • RA10 is H or RA; and

    • Cy and RA are as defined herein.





In some embodiments, A is a group of the Formula (A-41), (A-42), (A-43), (A-44), (A-45), or (A-46):




embedded image




    • wherein custom-character represents the point of attachment of A to the remainder of the molecule; and

    • Rb1, Rc1, Rd1 and Cy are as defined herein.





In some embodiments, the compound is other than a compound of Formula (II-A). In some embodiments, the compound is other than a compound of Formula (II-B). In some embodiments, the compound is other than a compound of Formula (II-C). In some embodiments, the compound is other than a compound of Formula (II-D). In some embodiments, the compound is other than a compound of Formula (II-E): In some embodiments, the compound is other than a compound of Formula (TI-F).


In some embodiments, A is a group other than a group of Formula (A-1). In some embodiments, A is a group other than a group of Formula (A-2). In some embodiments, A is a group other than a group of Formula (A-3). In some embodiments, A is a group other than a group of Formula (A-4). In some embodiments, A is a group other than a group of Formula (A-5). In some embodiments, A is a group other than a group of Formula (A-6). In some embodiments, A is a group other than a group of Formula (A-7). In some embodiments, A is a group other than a group of Formula (A-8). In some embodiments, A is a group other than a group of Formula (A-9). In some embodiments, A is a group other than a group of Formula (A-10). In some embodiments, A is a group other than a group of Formula (A-11). In some embodiments, A is a group other than a group of Formula (A-12). In some embodiments, A is a group other than a group of Formula (A-13). In some embodiments, A is a group other than a group of Formula (A-14). In some embodiments, A is a group other than a group of Formula (A-15). In some embodiments, A is a group other than a group of Formula (A-16). In some embodiments, A is a group other than a group of Formula (A-17). In some embodiments, A is a group other than a group of Formula (A-18). In some embodiments, A is a group other than a group of Formula (A-19). In some embodiments, A is a group other than a group of Formula (A-20). In some embodiments, A is a group other than a group of Formula (A-21). In some embodiments, A is a group other than a group of Formula (A-22). In some embodiments, A is a group other than a group of Formula (A-23). In some embodiments, A is a group other than a group of Formula (A-24). In some embodiments, A is a group other than a group of Formula (A-25). In some embodiments, A is a group other than a group of Formula (A-26). In some embodiments, A is a group other than a group of Formula (A-27). In some embodiments, A is a group other than a group of Formula (A-28). In some embodiments, A is a group other than a group of Formula (A-29). In some embodiments, A is a group other than a group of Formula (A-30). In some embodiments, A is a group other than a group of Formula (A-31). In some embodiments, A is a group other than a group of Formula (A-32). In some embodiments, A is a group other than a group of Formula (A-33). In some embodiments, A is a group other than a group of Formula (A-34). In some embodiments, A is a group other than a group of Formula (A-35). In some embodiments, A is a group other than a group of Formula (A-36). In some embodiments, A is a group other than a group of Formula (A-37). In some embodiments, A is a group other than a group of Formula (A-38). In some embodiments, A is a group other than a group of Formula (A-39). In some embodiments, A is a group other than a group of Formula (A-40). In some embodiments, A is a group other than a group of Formula (A-41). In some embodiments, A is a group other than a group of Formula (A-42). In some embodiments, A is a group other than a group of Formula (A-43). In some embodiments, A is a group other than a group of Formula (A-44). In some embodiments, A is a group other than a group of Formula (A-45). In some embodiments, A is a group other than a group of Formula (A-46).


In some embodiments, X1 is CR1. In some embodiments, X1 is N.


In some embodiments, R1 is selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R2 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B.


In some embodiments, R1 is selected from H, D, C1-6 alkyl, C1-6 haloalkyl, halo, CN, and ORa1; wherein the C1-6 alkyl forming R1 is optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B. In some embodiments, R1 is selected from H, D, C1-6 alkyl (such as methyl or ethyl), C1-6 haloalkyl (such as CF3, CHF2, CF2CF3), CN, and halo (such as F, Cl, or Br). In some embodiments, R1 is H.


In some embodiments, X2 is CR2. In some embodiments, X2 is N.


In some embodiments, R2 is selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R2 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B.


In some embodiments, R2 is C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or halo.


In some embodiments, R2 is H, D, C1-6 alkyl (such as methyl or ethyl), C1-6 haloalkyl (such as CF3, CHF2, CF2CF3), CN, or halo (such as F, Cl, or Br). In some embodiments, R2 is C1-6 alkyl, C1-6 haloalkyl, or halo. In some embodiments, R2 is CH3, CH(CH3)2, CF3, CF2CF3, or Br.


In some embodiments, R2 is CF3. In some embodiments, R2 is CH3, CH(CH3)2, CF3, CF2CF3, CHF2, Br, or Cl. In some embodiments, R2 is H, CH3, CH(CH3)2, CF3, CF2CF3, CHF2, Br, or Cl.


In some embodiments, R2 is CH3, CH(CH3)2, CF3, CF2CF3, CHF2, cyclopropyl, CN, Br, or Cl.


In some embodiments, X3 is CR3. In some embodiments, X3 is N.


In some embodiments, R3 is selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R3 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B.


In some embodiments, R3 is selected from H, D, C1-6 alkyl, C1-6 haloalkyl, halo, CN, and ORa1; wherein the C1-6 alkyl forming R3 is optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B. In some embodiments, R3 is H, D, C1-6 alkyl (such as methyl or ethyl), C1-6 haloalkyl (such as CF3, CHF2, CF2CF3), CN, or halo (such as F, Cl, or Br).


In some embodiments, R3 is H.


In some embodiments, X4 is CR4. In some embodiments, X4 is N.


In some embodiments, R4 is selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R4 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B.


In some embodiments, R4 is selected from H, D, C1-6 alkyl, C1-6 haloalkyl, halo, CN, and ORa1; wherein the C1-6 alkyl forming R4 is optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B. In some embodiments, R4 is H, D, C1-6 alkyl (such as methyl or ethyl), C1-6 haloalkyl (such as CF3, CHF2, CF2CF3), CN, or halo (such as F, Cl, or Br). In some embodiments, R4 is H.


In some embodiments, X5 is CR5. In some embodiments, X5 is N.


In some embodiments, R5 is selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R5 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B.


In some embodiments, R5 is selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, C3-6 cycloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═Re1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R5 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B. In some embodiments, R5 is H, C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, ORa1, or halo.


In some embodiments, R5 is H, D, C1-6 alkyl (such as methyl or ethyl), C1-6 haloalkyl (such as CF3, CHF2, CF2CF3), CN, or halo (such as F, Cl, or Br). In some embodiments, R5 is H, CH3, CF3, or Cl. In some embodiments, R5 is CH3, CF3, or Cl. In some embodiments, R5 is H, C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or halo. In some embodiments, R5 is C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or halo. In some embodiments, R5 is H, CH3, CF3, CHF2, CH2CH3, CD3, cyclopropyl, or Cl. In some embodiments, R5 is CH3, CF3, CHF2, CH2CH3, CD3, cyclopropyl, or Cl. In some embodiments, R5 is H, CH3, CF3, CHF2, CH2CH3, CD3, CD2CD3, cyclopropyl, cyclobutyl, OCH3, N(CH3)2, I, or Cl.


In some embodiments, X6 is CR6. In some embodiments, X6 is N.


In some embodiments, R6 is selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R6 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B.


In some embodiments, R6 is selected from H, D, C1-6 alkyl, C1-6 haloalkyl, halo, CN, and ORa1; wherein the C1-6 alkyl forming R6 is optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B. In some embodiments, R6 is H, D, C1-6 alkyl (such as methyl or ethyl), C1-6 haloalkyl (such as CF3, CHF2, CF2CF3), CN, or halo (such as F, Cl, or Br). In some embodiments, R6 is H.


In some embodiments, R7 and R8 are each selected from H, D, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, and P(O)(NH2)ORa1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming R7 and R8 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B.


In some embodiments, R7 and R8 are each selected from H, D, C1-6 alkyl, C1-6 haloalkyl, halo, CN, and ORa1; wherein the C1-6 alkyl forming R7 and R8 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B. In some embodiments, R7 and R8 are each H, D, C1-6 alkyl (such as methyl or ethyl), C1-6 haloalkyl (such as CF3, CHF2, CF2CF3), or halo (such as F, Cl, or Br). In some embodiments, R7 and R8 are each H.


In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a ring that is substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from RA. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a ring that is substituted with 1, 2, 3, 4, or 5 substituents independently selected from RA. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a ring that is substituted with 1, 2, 3, or 4 substituents independently selected from RA. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a ring that is substituted with 1, 2, or 3 substituents independently selected from RA. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a ring that is substituted with 1 or 2 substituents independently selected from RA. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a ring that is substituted with 1 substituent selected from RA. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a ring that is unsubstituted.


In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a 4-12 membered heterocycloalkane ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a 4-7 membered heterocycloalkane ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form an azetidine ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a pyrrolidine ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a piperidine ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form an oxetane ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a tetrahydrofuran ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a tetrahydro-2H-pyran ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a 1,4-dioxane ring.


In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a C3-6 cycloalkylane ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a cyclopropane ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a cyclobutane ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a cyclopentane ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a cyclohexane ring. In some embodiments, R9A and R9B, together with the carbon atom to which they are both attached, form a cyclopentene ring.


In some embodiments, each RA is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, 4-12 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1; wherein the 4-12 membered heterocycloalkyl and 5-10 membered heteroaryl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2.


In some embodiments, each RA is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2.


In some embodiments, RA is independently selected from 4-12 membered heterocycloalkyl and 5-10 membered heteroaryl, wherein the 4-12 membered heterocycloalkyl and 5-10 membered heteroaryl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA1.


In some embodiments, RA is independently selected from 1,6-dihydropyridazinyl, 1,2,4-triazolyl, piperazinyl, pyrazinyl, 1,2,3-triazolyl, pyridinyl, and 1,2-dihydropyridinyl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA1.


In some embodiments, RA is 1-methyl-6-oxo-1,6-dihydropyridazin-3-yl, 5-amino-1H-1,2,4-triazol-3-yl, 4-methylpiperazin-1-yl, 5-cyanopyrazin-2-yl, 1-methyl-1H-1,2,3-triazol-4-yl, 6-carbamoylpyridin-2-yl, or 1-cyclopropyl-2-oxo-1,2-dihydropyridin-4-yl.


In some embodiments, RA is independently selected from 1,6-dihydropyridazinyl, 1,2,4-triazolyl, piperazinyl, pyrazinyl, 1,2,3-triazolyl, pyridinyl, 1,2-dihydropyridinyl, and 1,2-dihydropyrimidinyl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA1.


In some embodiments, RA is 1-methyl-6-oxo-1,6-dihydropyridazin-3-yl, 5-amino-1H-1,2,4-triazol-3-yl, 4-methylpiperazin-1-yl, 5-cyanopyrazin-2-yl, 1-methyl-iH-1,2,3-triazol-4-yl, 6-carbamoylpyridin-2-yl, 1-cyclopropyl-2-oxo-1,2-dihydropyridin-4-yl, or 1-methyl-2-oxo-1,2-dihydropyrimidin-4-yl.


In some embodiments, each RA is selected from C1-6 alkyl (such as methyl or ethyl), C1-6 haloalkyl (such as CF3, CHF2, CF2CF3), halo (such as F, Cl, or Br), and ORa1 (such as methoxy or ethoxy). In some embodiments, each RA is selected from C1-6 alkyl, C(O)NRc1Rd1, S(O)2NRc1Rd1, C(O)ORa1, and C(O)Rb1, wherein C1-6 alkyl is optionally substituted with RA2. In some embodiments, each RA is selected from methyl, C(O)NH2, C(O)N(H)CH3, C(O)OCH3, S(O)2NH2, C(O)CH3, CH2CH2OH, and C(O)CH2CN. In some embodiments, each RA is selected from methyl, CH2OH, CH2CH2OH, CN, NH2, NHCH3, OCH3, OH, C(O)NH2, C(O)N(H)CH3, C(O)OCH3, C(O)CH3, C(O)CH2CN, C(O)N(H)CD3, C(O)N(H)CH2CF3, C(O)N(H)CH2CHF2, N(H)CH2C(CH3)2OH, N(H)CH2CH2CN, N(CH3)C(O)CH3, and S(O)2NH2.


In some embodiments, each RA is selected from C1-6 alkyl, NRcRd1, C(O)NRc1Rd1S(O)2NRc1Rd1, C(O)ORa1, and C(O)Rb1, wherein C1-6 alkyl is optionally substituted with RA2.


In some embodiments, each RA1 is independently selected from C1-6 alkyl and RA2. In some embodiments, each RA1 is independently selected from CH3 and RA2.


In some embodiments, each RA2 is independently selected from halo, CN, ORa3, SR3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)ORa3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, and S(O)2NRc3Rd3. In some embodiments, RA2 is ORa3.


In some embodiments, each RA2 is independently selected from C3-6 cycloalkyl, halo, CN, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)ORa3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3 S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, and S(O)2NRc3Rd3,


In some embodiments, each RA2 is independently selected from cyclopropyl, CN, ORa3, NRc3Rd3, and C(O)NRc3Rd3.


In some embodiments, Cy is substituted with 1, 2, 3, 4, 5 or 6 substituents independently selected from RCy. In some embodiments, Cy is substituted with 1, 2, 3, 4, or 5 substituents independently selected from RCy. In some embodiments, Cy is substituted with 1, 2, 3, or 4 substituents independently selected from RCy. In some embodiments, Cy is substituted with 1, 2, or 3 substituents independently selected from RCy. In some embodiments, Cy is substituted with 1 or 2 substituents independently selected from RCy. In some embodiments, Cy is substituted with 1 substituent selected from RCy. In some embodiments, Cy is unsubstituted.


In some embodiments, Cy is 5-10 membered heteroaryl substituted with 1, 2, 3, 4, 5 or 6 substituents independently selected from RCy. In some embodiments, Cy is 5-10 membered heteroaryl substituted with 1, 2, 3, 4, or 5 substituents independently selected from RCy. In some embodiments, Cy is 5-10 membered heteroaryl substituted with 1, 2, 3, or 4 substituents independently selected from RCy. In some embodiments, Cy is 5-10 membered heteroaryl substituted with 1, 2, or 3 substituents independently selected from RCy. In some embodiments, Cy is 5-10 membered heteroaryl substituted with 1 or 2 substituents independently selected from RCy. In some embodiments, Cy is 5-10 membered heteroaryl substituted with 1 substituent selected from RCy. In some embodiments, Cy is unsubstituted 5-10 membered heteroaryl.


In some embodiments, Cy is unsubstituted or substituted 2H-pyrazolo[3,4-c]pyridinyl, 1,7-naphthyridinyl, 2,7-naphthyridinyl, 3,7-naphthyridinyl, 4,7-naphthyridinyl, pyrazolyl or pyridinyl. In some embodiments, Cy is unsubstituted or substituted pyrazolyl. In some embodiments, Cy is unsubstituted or substituted pyridinyl. In some embodiments, Cy is unsubstituted or substituted pyrazol-1-yl. In some embodiments, Cy is unsubstituted or substituted pyridin-2-yl. In some embodiments, Cy is unsubstituted or substituted pyridin-3-yl. In some embodiments, Cy is unsubstituted or substituted pyridin-2-yl or pyridin-3-yl.


In some embodiments, Cy is C6-10 aryl substituted with 1, 2, 3, 4, 5 or 6 substituents independently selected from RCy. In some embodiments, Cy is C6-10 aryl substituted with 1, 2, 3, 4, or 5 substituents independently selected from RCy. In some embodiments, Cy is C6-10 aryl substituted with 1, 2, 3, or 4 substituents independently selected from RCy. In some embodiments, Cy is C6-10 aryl substituted with 1, 2, or 3 substituents independently selected from RCy. In some embodiments, Cy is C6-10 aryl substituted with 1 or 2 substituents independently selected from RCy. In some embodiments, Cy is C6-10 aryl substituted with 1 substituent selected from RCy. In some embodiments, Cy is unsubstituted C6-10 aryl.


In some embodiments, Cy is phenyl substituted with 1, 2, 3, 4, or 5 substituents independently selected from RCy. In some embodiments, Cy is phenyl substituted with 1, 2, 3, or 4 substituents independently selected from RCy. In some embodiments, Cy is phenyl substituted with 1, 2, or 3 substituents independently selected from RCy. In some embodiments, Cy is phenyl substituted with 1 or 2 substituents independently selected from RCy. In some embodiments, Cy is phenyl substituted with 1 substituent selected from RCy. In some embodiments, Cy is unsubstituted phenyl.


In some embodiments, Cy is selected from: 4-halophenyl; 3-halopyridin-4-yl; 2,4-dihalophenyl; 3,4-dihalophenyl; 3,5-dihalophenyl; 3,5-dihalopyridin-2-yl; 3-halo-1H-pyrazol-1-yl; 5-halopyridin-2-yl; phenyl; pyridine-2-yl; pyridine-3-yl; pyridine-4-yl; and 1H-pyrazol-1-yl.


In some embodiments, Cy is selected from: 4-bromophenyl; 4-chlorophenyl; 3-chloropyridin-4-yl; 2.4-difluorophenyl; 3,4-difluorophenyl; 3,5-difluorophenyl; 3,5-difluoropyridin-2-yl; 4-fluorophenyl; 3-fluoro-1H-pyrazol-1-yl; 5-fluoropyridin-2-yl; 4-methoxyphenyl; phenyl; pyridine-2-yl; pyridine-3-yl; pyridine-4-yl; and 1H-pyrazol-1-yl.


In some embodiments, Cy is selected from: 4-bromophenyl; 4-chlorophenyl; 3-chloropyridin-4-yl; 2.4-difluorophenyl; 3,4-difluorophenyl; 3,5-difluorophenyl; 3,5-difluoropyridin-2-yl; 4-fluorophenyl; 3-fluoro-1H-pyrazol-1-yl; 5-fluoropyridin-2-yl; 4-methoxyphenyl; phenyl; pyridine-2-yl; pyridine-3-yl; pyridine-4-yl; 1H-pyrazol-1-yl, and 4-fluoro-1H-pyrazol-1-yl.


In some embodiments, Cy is selected from: 4-bromophenyl; 4-chlorophenyl; 3-chloropyridin-4-yl; 2.4-difluorophenyl; 3,4-difluorophenyl; 3,5-difluorophenyl; 3,5-difluoropyridin-2-yl; 4-fluorophenyl; 3-fluoro-1H-pyrazol-1-yl; 5-fluoropyridin-2-yl; 4-methoxyphenyl; phenyl; pyridin-2-yl; pyridin-3-yl; pyridin-4-yl; 1H-pyrazol-1-yl, 4-fluoro-1H-pyrazol-1-yl, 6-methylpyridin-3-yl, and 4-(dimethylcarbamoyl)phenyl.


In some embodiments, each RCy is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRe2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NORa2)Rb2, C(═NRe2)NRe2Rd2, NRe2C(═NRe2)Re2Rd2, NRc2S(O)Rb2, NRe2S(O)2Rb2, NRe2S(O)2NRe2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRe2Rd2; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RCy are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy2.


In some embodiments, each RCy is selected from C1-6 alkyl (such as methyl or ethyl), C1-6 haloalkyl (such as CF3, CHF2, CF2CF3), halo (such as F, Cl, or Br), and ORa2 (such as methoxy or ethoxy). In some embodiments, each RCy is selected from halo and ORa2. In some embodiments, each RCy is selected from C1-6 alkyl, halo, C(O)N(CH3)2, and ORa2. In some embodiments, each RCy is selected from F, Br, and methoxy. In some embodiments, each RCy is selected from methyl, F, Br, and methoxy.


In some embodiments, each Rg2 is independently selected from D, OH, NO2, CN, halo, C3-6 cycloalkyl, C3-6 cycloalkyl-C1-2 alkylene, C1-6 alkoxy, C1-6 haloalkoxy, C1-3 alkoxy-C1-3 alkyl, C1-3 alkoxy-C1-3 alkoxy, HO—C1-3 alkoxy, HO—C1-3 alkyl, cyano-C1-3 alkyl, H2N—C1-3 alkyl, amino, C1-6 alkylamino, di(C1-6 alkyl)amino, thio, C1-6 alkylthio, C1-6 alkylsulfinyl, C1-6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, carboxy, C1-6 alkylcarbonyl, C1-6 alkoxycarbonyl, C1-6 alkylcarbonylamino, C1-6 alkylsulfonylamino, aminosulfonyl, C1-6 alkylaminosulfonyl, di(C1-6 alkyl)aminosulfonyl, aminosulfonylamino, C1-6 alkylaminosulfonylamino, di(C1-6 alkyl)aminosulfonylamino, aminocarbonylamino, C1-6 alkylaminocarbonylamino, and di(C1-6 alkyl)aminocarbonylamino.


Also provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt thereof, wherein:

    • X1 is N or CR1;
    • X2 is N or CR2;
    • X3 is N or CR3;
    • X4 is N or CR4;
    • provided that at least one of X3 and X4 is N;
    • X5 is CR5;
    • X6 is N or CR6;
    • R1, R2, R3, R4, R5, and R6 are each independently selected from H, D, C1-6 alkyl, C3-6 cycloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1NRd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)Rc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, or P(O)(NH2)ORa1; wherein the C1-6 alkyl forming R1, R2, R3, R4, R5, and R6 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B;
    • R7 and R8 are each independently selected from H, D, C1-6 alkyl, and C1-6 haloalkyl;
    • R9A and R9B, together with the carbon atom to which they are both attached, form a C3-12 cycloalkane or 4-12 membered heterocycloalkane ring, wherein the C3-12 cycloalkane or 4-12 membered heterocycloalkane ring formed by R9A and R9B is optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA;
    • Cy is a ring selected from C6-10 aryl and 5-10 membered heteroaryl; wherein the C6-10 aryl and 5-10 membered heteroaryl forming Cy are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Ry;
    • each RA is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, 4-12 membered heterocycloalkyl, 5-10 membered heteroaryl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRc1)NRc1Rd1, NRc1C(═NRc1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRccRd1, S(O)2Rb1, and S(O)2NRc1Rd1; wherein the 4-12 membered heterocycloalkyl and 5-10 membered heteroaryl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA1; and wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2;
    • each RA1 is independently selected from C1-6 alkyl and RA2;
    • each RA2 is independently selected from C3-6 cycloalkyl, halo, CN, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)ORa3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3 S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, and S(O)2NRc3Rd3;
    • each RCy is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NORa2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRc2Rd2; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RCy are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy2;
    • each RCy2 is independently selected from halo, CN, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)ORa4, NRc4S(O)Rb4, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4 S(O)Rb4, S(O)NRc4Rd4, S(O)2Rb4, and S(O)2NRc4Rd4;
    • each R10B is independently selected from halo, CN, NO2, ORa5, SRa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, OC(O)Rb5, OC(O)NRc5Rd5, NRc5Rd5, NRc5C(O)Rb1, NRc5C(O)ORa5, NRc5C(O)NRc5Rd5, C(═NRe5)Rb5, C(═NORa5)Rb5, C(═NRe5)NRc5Rd5, NRc5C(═NRe5)NRc5Rd5, NRc5S(O)Rb1, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5 S(O)Rb5, S(O)NRc5Rd5, S(O)2Rb5, and S(O)2NRc5Rd5;
    • each Ra1, Rc1, Rd1, Ra2, Rc2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra5, Rc5, and Rd5 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, and C1-6 haloalkyl; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Ra1, Rc1, Rd1, Ra2, Rc2, Rd2, Ra3, Rc3, Rd3Ra4, Rc4, Rd4, Ra5, Rc5, and Rd5 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;
    • or any Rc1 and Rd1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • or any Rc2 and Rd2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • or any Rc4 and Rd4 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • or any Rc5 and Rd5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • each Rb1, Rb2, Rb3, Rb4, and Rbs is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Rb1, Rb2, Rb3, Rb4, and Rbs are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;
    • each Re1, Re2, and Re5 is independently selected from H, CN, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkylthio, C1-6 alkylsulfonyl, C1-6 alkylcarbonyl, C1-6 alkylaminosulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, aminosulfonyl, C1-6 alkylaminosulfonyl and di(C1-6 alkyl)aminosulfonyl;
    • each Rg1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and Rg2; and
    • each Rg2 is independently selected from D, OH, NO2, CN, halo, C3-6 cycloalkyl, C3-6 cycloalkyl-C1-2 alkylene, 3-6 membered heterocycloalkyl, 3-6 membered heterocycloalkyl-C1-2 alkylene, C1-6 alkoxy, C1-6 haloalkoxy, C1-3 alkoxy-C1-3 alkyl, C1-3 alkoxy-C1-3 alkoxy, HO—C1-3 alkoxy, HO—C1-3 alkyl, cyano-C1-3 alkyl, H2N—C1-3 alkyl, amino, C1-6 alkylamino, di(C1-6 alkyl)amino, thio, C1-6 alkylthio, C1-6 alkylsulfinyl, C1-6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, carboxy, C1-6 alkylcarbonyl, C1-6 alkoxycarbonyl, C1-6 alkylcarbonylamino, C1-6 alkylsulfonylamino, aminosulfonyl, C1-6 alkylaminosulfonyl, di(C1-6 alkyl)aminosulfonyl, aminosulfonylamino, C1-6 alkylaminosulfonylamino, di(C1-6 alkyl)aminosulfonylamino, aminocarbonylamino, C1-6 alkylaminocarbonylamino, and di(C1-6 alkyl)aminocarbonylamino.


Also provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt thereof, wherein:

    • X1 is N or CR1;
    • X2 is N or CR2;
    • X3 is N or CR3;
    • X4 is N or CR4;
    • provided that at least one of X3 and X4 is N;
    • X5 is CR5;
    • X6 is N or CR6;
    • R1, R2, R3, R4, R5, and R6 are each independently selected from H, D, C1-6 alkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, S(O)(═NH)Rb1, S(O)2NRc1Rd1, P(O)(NH2)Rb1, or P(O)(NH2)ORa1; wherein the C1-6 alkyl forming R1, R2, R3, R4, R5, and R6 are each optionally substituted with 1, 2, 3, or 4 substituents each independently selected from R10B;
    • R7 and R8 are each independently selected from H, D, C1-6 alkyl, and C1-6 haloalkyl;
    • R9A and R9B, together with the carbon atom to which they are both attached, form a C3-12 cycloalkane or 4-12 membered heterocycloalkane ring, wherein the C3-12 cycloalkane or 4-12 membered heterocycloalkane ring formed by R9A and R9B is optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA;
    • Cy is a ring selected from C6-10 aryl and 5-10 membered heteroaryl; wherein the C6-10 aryl and 5-10 membered heteroaryl forming Cy are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from Ry;
    • each RA is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa1, SRa1, C(O)Rb1, C(O)NRcRd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(═NRe1)Rb1, C(═NORa1)Rb1, C(═NRe1)NRc1Rd1, NRc1C(═NRc1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1 S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and S(O)2NRc1Rd1; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RA are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RA2;
    • each RA2 is independently selected from halo, CN, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)ORa3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3 S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, and S(O)2NRc3Rd3;
    • each RCy is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, halo, CN, NO2, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(═NRe2)Rb2, C(═NORa2)Rb2, C(═NRe2)NRc2Rd2, NRc2C(═NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and S(O)2NRc2Rd2; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming RCy are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy2;
    • each RCy2 is independently selected from halo, CN, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)ORa4, NRc4S(O)Rb4, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4, S(O)Rb4, S(O)NRc4Rd4, S(O)2Rb4, and S(O)2NRc4Rd4;
    • each R10B is independently selected from halo, CN, NO2, ORa5, SRa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, OC(O)Rb5, OC(O)NRc5Rd5, NRc5Rd5, NRc5C(O)Rb5, NRc1C(O)ORa5, NRc5C(O)NRc5Rd5, C(═NRe5)Rb5, C(═NORa5)Rb5, C(═NRe5)NRc5Rd5, NRc5C(═NRe5)NRc5Rd5, NRc5S(O)Rb1, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5, S(O)Rb5, S(O)NRc5Rd5, S(O)2Rb5, and S(O)2NRc5Rd5;
    • each Ra1, Rc1, Rd1, Ra2, Rc2, Rd2, Ra3, Rc3, Rd3, Ra4, Rc4, Rd4, Ra5, Rc5, and Rd5 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, and C1-6 haloalkyl; wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Ra1, Rc1, Rd1, Ra2, Rc2, Rd2, Ra3, Rc3, Rd3Ra4, Rc4, Rd4, Ra5, Rc5, and Rd5 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;
    • or any Rc1 and Rd1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • or any Re2 and Rd2 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • or any Rc3 and Rd3 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • or any Rc4 and Rd4 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • or any Rc5 and Rd5 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • each Rb1, Rb2, Rb3, Rb4, and Rbs is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Rb1, Rb2, Rb3, Rb4, and Rbs are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;
    • each Re1, Re2, and Re5 is independently selected from H, CN, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkylthio, C1-6 alkylsulfonyl, C1-6 alkylcarbonyl, C1-6 alkylaminosulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, aminosulfonyl, C1-6 alkylaminosulfonyl and di(C1-6 alkyl)aminosulfonyl;
    • each Rg1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and Rg2; and
    • each Rg2 is independently selected from D, OH, NO2, CN, halo, C3-6 cycloalkyl, C3-6 cycloalkyl-C1-2 alkylene, C1-6 alkoxy, C1-6 haloalkoxy, C1-3 alkoxy-C1-3 alkyl, C1-3 alkoxy-C1-3 alkoxy, HO—C1-3 alkoxy, HO—C1-3 alkyl, cyano-C1-3 alkyl, H2N—C1-3 alkyl, amino, C1-6 alkylamino, di(C1-6 alkyl)amino, thio, C1-6 alkylthio, C1-6 alkylsulfinyl, C1-6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, carboxy, C1-6 alkylcarbonyl, C1-6 alkoxycarbonyl, C1-6 alkylcarbonylamino, C1-6 alkylsulfonylamino, aminosulfonyl, C1-6 alkylaminosulfonyl, di(C1-6 alkyl)aminosulfonyl, aminosulfonylamino, C1-6 alkylaminosulfonylamino, di(C1-6 alkyl)aminosulfonylamino, aminocarbonylamino, C1-6 alkylaminocarbonylamino, and di(C1-6 alkyl)aminocarbonylamino.


Also provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt thereof, wherein:

    • X1 is N or CR1;
    • X2 is N or CR2;
    • X3 is N or CR3;
    • X4 is N or CR4;
    • provided that at least one of X3 and X4 is N;
    • X5 is CR5;
    • X6 is N or CR6;
    • R1 is H;
    • R2 is C1-6 alkyl, C1-6 haloalkyl, or halo;
    • R3 is H;
    • R4 is H;
    • R5 is H, C1-6 alkyl, C1-6 haloalkyl, or halo;
    • R6 is H;
    • R7 and R8 are each H;
    • R9A and R9B, together with the carbon atom to which they are both attached, form a C3-6 cycloalkane or 4-7 membered heterocycloalkane ring, wherein the C3-6 cycloalkane or 4-7 membered heterocycloalkane ring formed by R9A and R9B is optionally substituted with 1, 2, 3, 4, 5, or 6 substituents independently selected from RA;
    • Cy is a ring selected from phenyl and 5-6 membered heteroaryl; wherein the phenyl and 5-6 membered heteroaryl forming Cy are each optionally substituted with 1, 2, 3 or 4 substituents independently selected from Ry;
    • each RA is selected from C1-6 alkyl, C(O)NRc1Rd1, S(O)2NRc1Rd1, C(O)ORa1, and C(O)Rb1, wherein C1-6 alkyl is optionally substituted with RA2;
    • each RA2 is ORa3;
    • each RCy is selected from halo and ORa2;
    • each Ra1, Rc1, Rd1, Ra2, and Ra3 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Ra1, Rc1, Rd1, Ra2, and Ra3 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;
    • or any Rc1 and Rd1 attached to the same N atom, together with the N atom to which they are attached, form a 4-, 5-, 6- or 7-membered heterocycloalkyl group optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg1;
    • each Rb1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, wherein the C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl forming Rb1 are each optionally substituted with 1, 2, 3, or 4 substituents independently selected from Rg2;
    • each Rg1 is independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, and Rg2; and
    • each Rg2 is independently selected from D, OH, NO2, CN, and halo.


In some embodiments, the compound of Formula (I) is selected from:

  • 4-(((7-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide;
  • 4-phenyl-4-(((2-(trifluoromethyl)imidazo[1,2-c]pyrimidin-5-yl)amino)methyl)piperidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-sulfonamide;
  • (R)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-sulfonamide;
  • (S)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-sulfonamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-carboxamide;
  • (R)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-carboxamide;
  • (S)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-carboxamide;
  • 4-(((7-chloro-2-(trifluoromethyl)imidazo[1,2-c]pyrimidin-5-yl)amino)methyl)-4-phenylpiperidine-1-sulfonamide;
  • 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-sulfonamide;
  • 4-(((5-methyl-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide;
  • 2-methyl-N-((4-phenylpiperidin-4-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluoro-1H-pyrazol-1-yl)azetidine-1-sulfonamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluoro-1H-pyrazol-1-yl)azetidine-1-carboxamide;
  • 3-(4-fluoro-1H-pyrazol-1-yl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-sulfonamide;
  • 3-(4-fluoro-1H-pyrazol-1-yl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide;
  • N-methyl-4-phenyl-4-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)piperidine-1-carboxamide;
  • methyl 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxylate;
  • 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-methyl-4-phenylpiperidine-1-carboxamide;
  • 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide;
  • N-((1-methyl-4-phenylpiperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • N-methyl-4-(((5-methyl-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide;
  • (3,3-difluoroazetidin-1-yl)(4-phenyl-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidin-1-yl)methanone;
  • 1-(4-phenyl-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidin-1-yl)ethan-1-one;
  • N-methyl-4-phenyl-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidine-1-carboxamide;
  • 4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxamide;
  • 2-(4-(5-fluoropyridin-2-yl)-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidin-1-yl)ethan-1-ol;
  • 3-(4-(5-fluoropyridin-2-yl)-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidin-1-yl)-3-oxopropanenitrile;
  • 3-(((7-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)-N-methylazetidine-1-carboxamide;
  • N-methyl-4-(((2-(perfluoroethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide;
  • 4-(((7-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-N-methyl-4-phenylpiperidine-1-carboxamide;
  • N-methyl-4-(((7-methyl-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide;
  • 4-(((2-isopropylimidazo[1,2-a]pyridin-5-yl)amino)methyl)-N-methyl-4-phenylpiperidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-sulfonamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide;
  • 3-(4-fluorophenyl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-sulfonamide;
  • 3-(4-fluorophenyl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide;
  • 3-(4-fluorophenyl)-3-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)azetidine-1-sulfonamide;
  • 3-(4-fluorophenyl)-3-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide;
  • N-((1-(4-fluorophenyl)cyclobutyl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine;
  • N-((1-(pyridin-4-yl)cyclobutyl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine;
  • N-((1-(4-bromophenyl)cyclobutyl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine; and
  • N-((4-(4-methoxyphenyl)tetrahydro-2H-pyran-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine;
    • or a pharmaceutically acceptable salt of any of the aforementioned compounds.


In some embodiments, the compound of Formula (I) is selected from:

  • 6-(4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidin-1-yl)-2-methylpyridazin-3(2H)-one;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluoro-1H-pyrazol-1-yl)-N-methylazetidine-1-carboxamide;
  • N-((1-(5-amino-1H-1,2,4-triazol-3-yl)-4-phenylpiperidin-4-yl)methyl)-5-(difluoromethyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-((3-(4-methylpiperazin-1-yl)-1-phenylcyclobutyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-(((1s,3s)-3-(4-methylpiperazin-1-yl)-1-phenylcyclobutyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-(((1r,3r)-3-(4-methylpiperazin-1-yl)-1-phenylcyclobutyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-((3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutyl)amino)ethan-1-ol;
  • 2-(((1s,3s)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutyl)amino)ethan-1-ol;
  • 2-(((1r,3r)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutyl)amino)ethan-1-ol;
  • 5-(3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidin-1-yl)pyrazine-2-carbonitrile;
  • N-((3-(4-fluorophenyl)-1-(1-methyl-1H-1,2,3-triazol-4-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 6-(3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidin-1-yl)picolinamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,3r)-3-hydroxycyclobutyl)azetidine-1-carboxamide; (R)-(4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidin-1-yl)(1-methylazetidin-2-yl)methanone;
  • 4-(3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidin-1-yl)-1-cyclopropylpyridin-2(1H)-one;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1s,3s)-3-hydroxy-3-methylcyclobutyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1R,3R)-3-hydroxycyclopentyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1s,4s)-4-hydroxy-4-(trifluoromethyl)cyclohexyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-(2-hydroxypropan-2-yl)cyclohexyl)azetidine-1-carboxamide;
  • 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)-N-((1R,3R)-3-hydroxycyclopentyl)piperidine-1-carboxamide;
  • 3-(((2-chloro-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide;
  • 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-chlorophenyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)azetidine-1-sulfonamide;
  • 2-(azetidin-1-yl)-1-(4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidin-1-yl)ethan-1-one;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)-N-(methyl-d3)azetidine-1-carboxamide;
  • 3-(((2-(difluoromethyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(methyl-d3)azetidine-1-carboxamide;
  • N-cyclopropyl-3-(((2-(difluoromethyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-(3-cyanobicyclo[1.1.1]pentan-1-yl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-(3-cyanobicyclo[1.1.1]pentan-1-yl)-3-(3,5-difluorophenyl)azetidine-1-carboxamide;
  • 3-(((5-chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide;
  • 3-(4-fluorophenyl)-3-(((5-methyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxamide;
  • 3-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide;
  • 3-(((5-cyclopropyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide;
  • 3-(4-fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxamide; and
  • 3-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(2,2,2-trifluoroethyl)azetidine-1-carboxamide;
    • or a pharmaceutically acceptable salt of any of the aforementioned.


In some embodiments, the compound of Formula (I) is selected from:

  • 1-amino-4-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexane-1-carbonitrile;
  • 1-((3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutyl)amino)-2-methylpropan-2-ol;
  • 1-((4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexyl)amino)-2-methylpropan-2-ol;
  • N-(((1s,3s)-3-methoxy-1-phenylcyclobutyl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 1-amino-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutane-1-carboxamide;
  • 2-methyl-N-((3-phenyltetrahydrofuran-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 5-iodo-2-methyl-N-((3-phenyltetrahydrofuran-3-yl)methyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 3-(((7-chloro-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)-3-phenylazetidine-1-carboxamide;
  • 3-(((2,7-bis(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutan-1-ol;
  • 3-((4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)amino)propanenitrile;
  • 2-bromo-N-((1-phenylcyclopent-3-en-1-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-((4-(methylamino)-1-phenylcyclohexyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol;
  • N-(4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)-N-methylacetamide;
  • 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol;
  • 2-bromo-N-((3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine; (2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol; (2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol;
  • 4-(3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidin-1-yl)-1-methylpyrimidin-2(1H)-one;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,3r)-3-hydroxy-3-methylcyclobutyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(((1r,3r)-3-hydroxycyclobutyl)methyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1S,2S)-2-hydroxycyclobutyl)azetidine-1-carboxamide;
  • 3-(((2-cyclopropyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(1H-pyrazol-1-yl)azetidine-1-sulfonamide;
  • 3-(((2-cyano-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(1H-pyrazol-1-yl)azetidine-1-sulfonamide;
  • 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(6-methylpyridin-3-yl)azetidine-1-sulfonamide;
  • 3-(((2-(difluoromethyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide;
  • 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(3,4-difluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide;
  • 3-(4-fluorophenyl)-N-(methyl-d3)-3-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide;
  • 3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)-3-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide;
  • 3-(4-fluorophenyl)-N-(methyl-d3)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide;
  • 5-chloro-7-(((4-phenylpiperidin-4-yl)methyl)amino)pyrazolo[1,5-a]pyrimidine-2-carbonitrile;
  • 3-(((5-(dimethylamino)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-sulfonamide;
  • 3-(4-fluorophenyl)-3-(((5-methoxy-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-sulfonamide;
  • 3-(((5-cyclopropyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-(dimethylcarbamoyl)phenyl)-N-(methyl-d3)azetidine-1-carboxamide;
  • 3-(((5-cyclobutyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(methyl-d3)azetidine-1-carboxamide;
  • 3-(4-fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-(2,2,2-trifluoroethyl)azetidine-1-carboxamide;
  • N-(2,2-difluoroethyl)-3-(4-fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxamide;
  • 3-(((5-(ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide; and
  • 3-(((5-(ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide;
    • or a pharmaceutically acceptable salt of any of the aforementioned.


In some embodiments, the compound of Formula (I) is selected from:

  • 2-bromo-N-(((1s,3s)-3-(4-methylpiperazin-1-yl)-1-phenylcyclobutyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-(((1r,3r)-3-(4-methylpiperazin-1-yl)-1-phenylcyclobutyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-(((1s,3s)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutyl)amino)ethan-1-ol;
  • 2-(((1r,3r)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutyl)amino)ethan-1-ol;
  • (1s,4s)-1-amino-4-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexane-1-carbonitrile;
  • (1r,4r)-1-amino-4-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexane-1-carbonitrile;
  • 1-(((1r,3r)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutyl)amino)-2-methylpropan-2-ol;
  • 1-(((1s,3s)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutyl)amino)-2-methylpropan-2-ol;
  • 1-(((1r,4r)-4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexyl)amino)-2-methylpropan-2-ol;
  • 1-(((1s,4s)-4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexyl)amino)-2-methylpropan-2-ol;
  • 3-(((1r,4r)-4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)amino)propanenitrile;
  • 3-(((1s,4s)-4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)amino)propanenitrile;
  • 2-bromo-N-(((1r,4r)-4-(methylamino)-1-phenylcyclohexyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-(((1s,4s)-4-(methylamino)-1-phenylcyclohexyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • (1R*,3S*)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol;
  • (1S*,3S*)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol;
  • (1R,3S)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol;
  • (1S,3R)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol;
  • (1S,3S)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol;
  • (1R,3R)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol;
  • N-((1r,4r)-4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)-N-methylacetamide;
  • N-((1s,4s)-4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)-N-methylacetamide;
  • (1R*,3R*)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol;
  • (1R*,3S*)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol;
  • (1R,3R)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol;
  • (1S,3S)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol;
  • (1R,3S)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol;
  • (1S,3R)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol;
  • 2-bromo-N-(((1R*,3R*)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-(((1S*,3R*)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-(((1R,3R)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-(((1S,3S)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-(((1R,3S)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • 2-bromo-N-(((1S,3R)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine;
  • ((1R*,2S*)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol;
  • ((1R*,2R*)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol;
  • ((1R,2S)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol;
  • ((1S,2R)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol;
  • ((1R,2R)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol;
  • ((1S,2S)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol;
  • ((1R*,2R*)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol;
  • ((1R*,2S*)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol;
  • ((1R,2R)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol;
  • ((1S,2S)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol;
  • ((1R,2S)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol;
  • ((1S,2R)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol;
    • or a pharmaceutically acceptable salt of any of the aforementioned.


In other embodiments, the compound of Formula (I) is in the form of a pharmaceutically acceptable salt. In other embodiments, the compound of Formula (I) is in the form of a free base or free acid, or other than in the form of a salt.


In another aspect, provided herein is a pharmaceutical composition comprising a compound of Formula (I), or any of the embodiments thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.


It is further appreciated that certain features of the disclosure, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the disclosure which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.


At various places in the present specification, substituents of compounds of the disclosure are disclosed in groups or in ranges. It is specifically intended that the disclosure include each and every individual subcombination of the members of such groups and ranges. For example, the term “C1-6 alkyl” is specifically intended to individually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and C6 alkyl.


At various places in the present specification various aryl, heteroaryl, cycloalkyl, and heterocycloalkyl rings are described. Unless otherwise specified, these rings can be attached to the rest of the molecule at any ring member as permitted by valency. For example, the term “a pyridine ring” or “pyridinyl” may refer to a pyridin-2-yl, pyridin-3-yl, or pyridin-4-yl ring.


The term “n-membered” where n is an integer typically describes the number of ring-forming atoms in a moiety where the number of ring-forming atoms is n. For example, piperidinyl is an example of a 6-membered heterocycloalkyl ring, pyrazolyl is an example of a 5-membered heteroaryl ring, pyridyl is an example of a 6-membered heteroaryl ring, and 1,2,3,4-tetrahydronaphthalene is an example of a 10-membered cycloalkyl group.


For compounds of the disclosure in which a variable appears more than once, each variable can be a different moiety independently selected from the group defining the variable. For example, where a structure is described having two R groups that are simultaneously present on the same compound, the two R groups can represent different moieties independently selected from the group defined for R.


The phrase “optionally substituted” means unsubstituted or substituted.


The term “substituted” means that an atom or group of atoms formally replaces hydrogen as a “substituent” attached to another group. The term “substituted” refers, unless otherwise indicated, to any level of substitution, e.g., mono-, di-, tri-, tetra- or penta-substitution, where such substitution is permitted. The substituents are independently selected, and substitution may be at any chemically accessible position. It is to be understood that substitution at a given atom is limited by valency. It is to be understood that substitution at a given atom results in a chemically stable molecule. A single divalent substituent, e.g., oxo, can replace two hydrogen atoms.


The term “Cn-m” where n and m are integers is employed in combination with a chemical group to designate a range of the number of carbon atoms in the chemical group, with n and m defining the range. For example, C1-6 alkyl refers to an alkyl group having 1, 2, 3, 4, 5, or 6 carbon atoms. The term is intended to include each and every member in the indicated range. Thus, Cn-m includes each member in the series Cn, Cn+1, . . . Cm−1, and Cm. Examples include C14 (which includes C1, C2, C3, and C4), C1-6 (which includes C1, C2, C3, C4, C5, and C6) and the like.


The term “alkyl,” employed alone or in combination with other terms, refers to a saturated hydrocarbon group that may be straight-chain or branched. The term “Cn-m alkyl,” refers to an alkyl group having n to m carbon atoms. An alkyl group formally corresponds to an alkane with one C—H bond replaced by the point of attachment of the alkyl group to the remainder of the compound. In some embodiments, the alkyl group contains 1 to 6, 1 to 4, or 1 to 3 carbon atoms. Examples of alkyl moieties include, but are not limited to, chemical groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methyl-1-butyl, 3-pentyl, n-hexyl, 1,2,2-trimethylpropyl, and the like. In some embodiments, the alkyl group is methyl, ethyl, or propyl. In some embodiments, the alkyl group is deuterated, for example, CD3, CD2CD3, CH2CD3, and the like.


The term “alkylene,” employed alone or in combination with other terms, refers to a divalent alkyl linking group, which may be straight-chain or branched. An alkylene group formally corresponds to an alkane with two C—H bond replaced by points of attachment of the alkylene group to the remainder of the compound. The term “Cn-m alkylene” refers to an alkylene group having n to m carbon atoms. Examples of alkylene groups include, but are not limited to, methylene, ethan-1,2-diyl, ethan-1,1-diyl, propan-1,3-diyl, propan-1,2-diyl, propan-1,1-diyl, butan-1,4-diyl, butan-1,3-diyl, butan-1,2-diyl, 2-methyl-propan-1,3-diyl and the like.


The term “alkenyl,” employed alone or in combination with other terms, refers to a straight-chain or branched hydrocarbon group corresponding to an alkyl group having one or more carbon-carbon double bonds. The term “Cn-m alkylenyl” refers to an alkenyl group having n to m carbon atoms. An alkenyl group formally corresponds to an alkene with one C—H bond replaced by the point of attachment of the alkenyl group to the remainder of the compound. In some embodiments, the alkenyl moiety contains 2 to 6 or 2 to 4 carbon atoms. Example alkenyl groups include, but are not limited to, ethenyl, n-propenyl, isopropenyl, n-butenyl, sec-butenyl, and the like.


The term “alkynyl,” employed alone or in combination with other terms, refers to a straight-chain or branched hydrocarbon group corresponding to an alkyl group having one or more carbon-carbon triple bonds. The term “Cn-m alkynyl” refers to an alkynyl group having n to m carbon atoms. An alkynyl group formally corresponds to an alkyne with one C—H bond replaced by the point of attachment of the alkyl group to the remainder of the compound. In some embodiments, the alkynyl moiety contains 2 to 6 or 2 to 4 carbon atoms. Example alkynyl groups include, but are not limited to, ethynyl, propyn-1-yl, propyn-2-yl, and the like.


The term “alkoxy,” employed alone or in combination with other terms, refers to a group of formula —O-alkyl. The term “Cn-m alkoxy” refers to an alkoxy group, the alkyl group of which has n to m carbons. Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy and the like. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms. Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, alkoxy is methoxy.


The term “amino,” employed alone or in combination with other terms, refers to NH2.


The term “alkylamino,” employed alone or in combination with other terms, refers to a group of formula —NH(alkyl). In some embodiments, the alkylamino group has 1 to 6 or 1 to 4 carbon atoms. Example alkylamino groups include methylamino, ethylamino, propylamino (e.g., n-propylamino and isopropylamino), and the like.


The term “C1-3 alkoxy-C1-3 alkyl” refers to a group of formula —(C1-3 alkylene)-(C1-3 alkoxy).


The term “C1-3 alkoxy-C1-3 alkoxy” refers to a group of formula —(C1-3 alkoxylene)-(C1-3 alkoxy).


The term “Cn-m alkoxycarbonyl” refers to a group of formula —C(O)O-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylamino” refers to a group of formula —NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylcarbamyl” refers to a group of formula —C(O)—NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylcarbonyl” refers to a group of formula —C(O)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylcarbonylamino” refers to a group of formula —NHC(O)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylsulfonylamino” refers to a group of formula —NHS(O)2-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylaminosulfonyl” refers to a group of formula —S(O)2NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylaminosulfonylamino” refers to a group of formula —NHS(O)2NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylaminocarbonylamino” refers to a group of formula —NHC(O)NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylsulfinyl” refers to a group of formula —S(O)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “Cn-m alkylsulfonyl” refers to a group of formula —S(O)2-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “di(Cn-m alkyl)aminosulfonyl” refers to a group of formula —S(O)2N(alkyl)2, wherein each alkyl group independently has n to m carbon atoms. In some embodiments, each alkyl group has, independently, 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “di(Cn-m alkyl)aminosulfonylamino” refers to a group of formula —NHS(O)2N(alkyl)2, wherein each alkyl group independently has n to m carbon atoms. In some embodiments, each alkyl group has, independently, 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “di(Cn-m alkyl)aminocarbonylamino” refers to a group of formula —NHC(O)N(alkyl)2, wherein each alkyl group independently has n to m carbon atoms. In some embodiments, each alkyl group has, independently, 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “di(Cn-m-alkyl)carbamyl” refers to a group of formula —C(O)N(alkyl)2, wherein the two alkyl groups each has, independently, n to m carbon atoms. In some embodiments, each alkyl group independently has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.


The term “aminosulfonyl” refers to a group of formula —S(O)2NH2.


The term “aminosulfonylamino” refers to a group of formula —NHS(O)2NH2.


The term “aminocarbonylamino”, employed alone or in combination with other terms, refers to a group of formula —NHC(O)NH2.


The term “carbonyl,” employed alone or in combination with other terms, refers to a —C(═O)— group.


The term “carboxy” refers to a group of formula —C(O)OH.


The term “cyano” or “nitrile” refers to a group of formula —C≡N, which also may be written as —CN.


The term “cyano-C1-3 alkyl” refers to a group of formula —(C1-3 alkylene)-CN.


The term “halo” or “halogen”, employed alone or in combination with other terms, refers to fluoro, chloro, bromo, and iodo. In some embodiments, “halo” refers to a halogen atom selected from F, Cl, or Br. In some embodiments, halo is F or Cl. In some embodiments, halo is F.


The term “haloalkyl,” employed alone or in combination with other terms, refers to an alkyl group in which one or more of the hydrogen atoms has been replaced by a halogen atom, having up to the full valency of halogen atom substituents, which may either be the same or different. In some embodiments, the halogen atoms are fluoro atoms. The term “Cn-m haloalkyl” refers to a Cn-m alkyl group having n to m carbon atoms and from at least one up to {2(n to m)+1}halogen atoms, which may either be the same or different. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms. Example haloalkyl groups include CF3, C2F5, CHF2, CCl3, CHCl2, C2Cl5, and the like. In some embodiments, the haloalkyl group is a fluoroalkyl group.


The term “haloalkoxy,” employed alone or in combination with other terms, refers to a group of formula —O-(haloalkyl). The term “Cn-m haloalkoxy” refers to a haloalkoxy group, the haloalkyl group of which has n to m carbons. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms. An example haloalkoxy group is —OCF3.


The term “H2N—C1-3 alkyl” refers to a group of formula —(C1-3 alkylene)-NH2.


The term “HO—C1-3 alkoxy” refers to a group of formula —(C1-3 alkoxylene)-OH.


The term “HO—C1-3 alkyl” refers to a group of formula —(C1-3 alkylene)-OH.


The term “oxo” refers to an oxygen atom as a divalent substituent, forming a carbonyl group when attached to carbon, or attached to a heteroatom forming a sulfoxide or sulfone group, or an N-oxide group. In some embodiments, heterocyclic groups may be optionally substituted by 1 or 2 oxo (═O) substituents.


The term “oxidized” in reference to a ring-forming N atom refers to a ring-forming N-oxide.


The term “oxidized” in reference to a ring-forming S atom refers to a ring-forming sulfonyl or ring-forming sulfinyl.


The term “thio” refers to a group of formula —SH.


The term “alkylthio,” employed alone or in combination with other terms, refers to a group of formula —S-alkyl. The term “Cn-m alkylthio” refers to a group of formula —S-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4 carbon, or 1 to 3 carbon atoms.


The term “aromatic” refers to a carbocycle or heterocycle having one or more polyunsaturated rings having aromatic character (i.e., having (4n+2) delocalized π (pi) electrons where n is an integer).


The term “aryl,” employed alone or in combination with other terms, refers to an aromatic hydrocarbon group, which may be monocyclic or polycyclic (e.g., having 2 fused rings). The term “Cn-m aryl” refers to an aryl group having from n to m ring carbon atoms. Aryl groups include, e.g., phenyl, naphthyl, and the like. In some embodiments, aryl groups have from 6 to about 10 carbon atoms. In some embodiments, aryl groups have 6 carbon atoms. In some embodiments, aryl groups have 10 carbon atoms. In some embodiments, the aryl group is phenyl. In some embodiments, the aryl group is naphthyl.


The term “heteroaryl” or “heteroaromatic” employed alone or in combination with other terms, refers to a monocyclic or polycyclic (e.g., having 2 or 3 fused rings) aromatic hydrocarbon moiety, having one or more heteroatom ring members independently selected from nitrogen, sulfur and oxygen. In some embodiments, the heteroaryl group is a monocyclic or bicyclic group having 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, sulfur and oxygen. Example heteroaryl groups include, but are not limited to, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrryl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, purinyl, carbazolyl, benzimidazolyl, indolinyl, pyrrolyl, azolyl, quinolinyl, isoquinolinyl, benzisoxazolyl, imidazo[1,2-b]thiazolyl, pyridone, or the like. The carbon atoms or heteroatoms in the ring(s) of the heteroaryl group can be oxidized to form a carbonyl, an N-oxide, or a sulfonyl group (or other oxidized linkage) or a nitrogen atom can be quaternized, provided the aromatic nature of the ring is preserved. In some embodiments the heteroaryl group is a 5 to 10 membered heteroaryl group. In another embodiment the heteroaryl group is a 5 to 6 membered heteroaryl group. In some embodiments, the heteroaryl is a 5-6 membered heteroaryl moiety having carbon and 1, 2, or 3 heteroatoms independently selected from N, O and S. In some embodiments, the heteroaryl is a 5-10 membered heteroaryl moiety having carbon and 1, 2, or 3 heteroatoms independently selected from N, O and S. In some embodiments, the heteroaryl has 5-6 ring atoms and 1 or 2 heteroatom ring members independently selected from nitrogen, sulfur and oxygen. In some embodiments, no more than 2 heteroatoms of a 5-membered heteroaryl moiety are N.


A five-membered heteroaryl ring is a heteroaryl group having five ring atoms wherein one or more (e.g., 1, 2 or 3) ring atoms are independently selected from N, O and S. Exemplary five-membered ring heteroaryls include thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1,2,3-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-triazolyl, 1,2,4-thiadiazolyl, 1,2,4-oxadiazolyl, 1,3,4-triazolyl, 1,3,4-thiadiazolyl and 1,3,4-oxadiazolyl.


A six-membered heteroaryl ring is a heteroaryl group having six ring atoms wherein one or more (e.g., 1, 2 or 3) ring atoms are independently selected from N, O and S. Exemplary six-membered ring heteroaryls are pyridyl, pyrazinyl, pyrimidinyl, triazinyl, isoindolyl, and pyridazinyl.


A nine-membered heteroaryl ring is a heteroaryl group having nine ring atoms wherein one or more (e.g., 1, 2 or 3) ring atoms are independently selected from N, O and S. Exemplary nine-membered ring heteroaryls include benzofuran, benzo[b]thiophene, 1H-indole, 1H-benzo[d]imidazolyl, benzo[d]oxazolyl, benzo[d]thiazolyl, 1H-pyrrolo[3,2-b]pyridinyl, 1H-imidazo[4,5-b]pyridinyl, 1H-pyrrolo[3,2-c]pyridinyl, 1H-imidazo[4,5-c]pyridinyl, 1H-pyrrolo[2,3-c]pyridinyl, 3H-imidazo[4,5-c]pyridinyl, 1H-pyrrolo[2,3-b]pyridinyl, 3H-imidazo[4,5-b]pyridinyl, imidazo[1,2-a]pyridinyl,imidazo[1,2-a]pyrimidinyl,pyrazolo[1,5-a]pyrimidinyl,[1,2,4]triazolo[1,5-a]pyrimidinyl,[1,2,4]triazolo[1,5-a]pyridinyl,imidazo[1,2-c]pyrimidinyl,pyrrolo[1,2-a]pyrimidinyl, and 2H-pyrazolo[3,4-c]pyridinyl.


A ten-membered heteroaryl ring is a heteroaryl group having ten ring atoms wherein one or more (e.g., 1, 2 or 3) ring atoms are independently selected from N, O and S. Exemplary ten-membered ring heteroaryls are 1,7-naphthyridinyl, 2,7-naphthyridinyl, 3,7-naphthyridinyl, and 4,7-naphthyridinyl.


The term “cycloalkyl” or “cycloalkane” employed alone or in combination with other terms, refers to a non-aromatic cyclic hydrocarbon including cyclized alkyl and alkenyl groups. The terms “Cn-m cycloalkyl” refers to a cycloalkyl that has from n to m ring member carbon atoms. Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3, or 4 fused, bridged, or spiro rings) ring systems. Also included are moieties that have one or more aromatic rings (e.g., aryl or heteroaryl rings) fused to (i.e., having a bond in common with) the cycloalkyl ring, for example, benzo derivatives of cyclopentane, cyclohexene, cyclohexane, and the like, or pyrido derivatives of cyclopentane or cyclohexane. A cycloalkyl group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring. Ring-forming carbon atoms of a cycloalkyl group can be optionally substituted by oxo. The term “cycloalkyl” also includes bridgehead cycloalkyl groups (e.g., non-aromatic cyclic hydrocarbon moieties containing at least one bridgehead carbon, such as admantan-1-yl) and spirocycloalkyl groups (e.g., non-aromatic hydrocarbon moieties containing at least two rings fused at a single carbon atom, such as spiro[2.5]octane and the like). In some embodiments, the cycloalkyl group has 3 to 10 ring members, or 3 to 7 ring members, or 3 to 6 ring members. In some embodiments, the cycloalkyl group is monocyclic or bicyclic. In some embodiments, the cycloalkyl group is monocyclic. In some embodiments, the cycloalkyl group is a C3-7 monocyclic cycloalkyl group. In some embodiments, the cycloalkyl group is cyclopropyl, cyclobutyl, or cyclohexyl.


The term “heterocycloalkyl,” or “heterocycloalkane” or employed alone or in combination with other terms, refers to a non-aromatic ring or ring system, which has at least one carbon atom ring member and at least one heteroatom ring member independently selected from nitrogen, sulfur, oxygen, and phosphorus, and which has 4-14 ring members, 4-10 ring members, 4-7 ring members, or 4-6 ring members. The ring may contain one or more alkylene, alkenylene or alkynylene groups as part of the ring structure. The term “n-m-membered heterocycloalkyl” where n and m are integers refer to a heterocycloalkyl ring or ring system containing from n to m ring-forming atoms. An n-m-membered heterocycloalkyl include from 1 to m-1 carbon atoms and from 1 to m-1 heteroatoms. The term “n-membered heterocycloalkyl” where n is an integer refers to a heterocycloalkyl ring or ring system containing from n to m ring-forming atoms. Included within the term “heterocycloalkyl” are monocyclic 4-, 5-, 6- and 7-membered heterocycloalkyl. Heterocycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused, bridged, or spiro rings) or spirocyclic ring systems. In some embodiments, the heterocycloalkyl group is a monocyclic or bicyclic group having 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, sulfur and oxygen. Also included in the definition of heterocycloalkyl are moieties that have one or more aromatic rings (e.g., aryl or heteroaryl rings) fused (i.e., having a bond in common with) to the non-aromatic heterocycloalkyl ring, for example, 1,2,3,4-tetrahydro-quinoline and the like. Heterocycloalkyl groups can also include bridgehead heterocycloalkyl groups (e.g., a heterocycloalkyl moiety containing at least one bridgehead atom, such as azaadmantan-1-yl and the like) and spiroheterocycloalkyl groups (e.g., a heterocycloalkyl moiety containing at least two rings fused at a single atom, such as [1,4-dioxa-8-aza-spiro[4.5]decan-N-yl] and the like). In some embodiments, the heterocycloalkyl group has 3 to 10 ring-forming atoms, 4 to 10 ring-forming atoms, or 3 to 8 ring forming atoms. In some embodiments, the heterocycloalkyl group has 1 to 5 heteroatoms, 1 to 4 heteroatoms, 1 to 3 heteroatoms, or 1 to 2 heteroatoms. The carbon atoms or heteroatoms in the ring(s) of the heterocycloalkyl group can be oxidized to form a carbonyl, an N-oxide, or a sulfonyl group (or other oxidized linkage) or a nitrogen atom can be quaternized. In some embodiments, the heterocycloalkyl portion is a C2-7 monocyclic heterocycloalkyl group. In some embodiments, the heterocycloalkyl group is a morpholine ring, pyrrolidine ring, piperazine ring, piperidine ring, dihydropyran ring, tetrahydropyran ring, tetrahyropyridine, azetidine ring, or tetrahydrofuran ring. In some embodiments, the heterocycloalkyl is a 4-7 membered heterocycloalkyl moiety having carbon and 1, 2, or 3 heteroatoms independently selected from N, O and S. In some embodiments, the heterocycloalkyl is 4-10 membered heterocycloalkyl moiety having carbon and 1, 2, or 3 heteroatoms independently selected from N, O and S.


At certain places, the definitions or embodiments refer to specific rings (e.g., an azetidine ring, a pyridine ring, etc.). Unless otherwise indicated, these rings can be attached to any ring member provided that the valency of the atom is not exceeded. For example, an azetidine ring may be attached at any position of the ring, whereas an azetidin-3-yl ring is attached at the 3-position.


The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C═N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.


Resolution of racemic mixtures of compounds can be carried out by methods known in the art. An example method includes fractional recrystallization using a chiral resolving acid which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids. Other resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1,2-diaminocyclohexane, and the like.


Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.


In some embodiments, the compounds of the disclosure have the (R)-configuration. In other embodiments, the compounds have the (S)-configuration. In compounds with more than one chiral center, each of the chiral centers in the compound may be independently (R) or (S), unless otherwise indicated. In compounds with a single chiral center, the stereochemistry of the chiral center can be (R) or (S). In compounds with two chiral centers, the stereochemistry of the chiral centers can each be independently (R) or (S) so the configuration of the chiral centers can be (R) and (R), (R) and (S); (S) and (R), or (S) and (S). In compounds with three chiral centers, the stereochemistry each of the three chiral centers can each be independently (R) or (S) so the configuration of the chiral centers can be (R), (R) and (R); (R), (R) and (S); (R), (S) and (R); (R), (S) and (S); (S), (R) and (R); (S), (R) and (S); (S), (S) and (R); or (S), (S) and (S).


Compounds of the disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Example prototropic tautomers include ketone-enol pairs, amide-imidic acid pairs, lactam-lactim pairs, enamine-imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H-imidazole, 1H-, 2H- and 4H-1,2,4-triazole, 1H- and 2H-isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. Compounds herein identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms unless otherwise specified (e.g., in the case of purine rings, unless otherwise indicated, if a compound name or structure described the 9H tautomer, it would be understood that the 7H tautomer is also encompassed).


The term, “compound,” is intended to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted. The term is also meant to refer to compounds of the disclosure, regardless of how they are prepared, e.g., synthetically, through biological process (e.g., metabolism or enzyme conversion), or a combination thereof.


All compounds, and pharmaceutically acceptable salts thereof, can be found together with other substances such as water and solvents (e.g., in the form of hydrates and solvates) or can be isolated. When in the solid state, the compounds described herein and salts thereof may occur in various forms and may, e.g., take the form of solvates, including hydrates. The compounds may be in any solid state form, such as a polymorph or solvate, so unless clearly indicated otherwise, reference in the specification to compounds and salts thereof should be understood as encompassing any solid state form of the compound.


In some embodiments, the compounds of the disclosure, or salts thereof, are substantially isolated. By “substantially isolated” is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compounds of the disclosure. Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compounds of the disclosure, or salt thereof. Methods for isolating compounds and their salts are routine in the art.


The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.


The present disclosure also includes pharmaceutically acceptable salts of the compounds described herein. The term “pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present disclosure include the non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or MeCN are preferred. Lists of suitable salts are found in A. R. Gennaro (Ed.), Remington's Pharmaceutical Sciences, 17th Ed., (Mack Publishing Company, Easton, 1985), p. 1418, S. M. Berge et al., J. Pharm. Sci., 1977, 66(1), 1-19 and in P. H. Stahl et al., Handbook of Pharmaceutical Salts: Properties, Selection, and Use, 2nd Ed. (Wiley, 2011).


III. Synthesis

As will be appreciated by those skilled in the art, the compounds provided herein, including salts and stereoisomers thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes.


The reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected by the skilled artisan.


Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art. The chemistry of protecting groups is described, e.g., in P. Kocienski, Protecting Groups, 3rd Ed. (Thieme, 2005); J. Robertson, Protecting Group Chemistry, (Oxford University Press, 2000); M. B. Smith et al., March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 8th Ed. (Wiley, 2020); S. Petursson, J. Chem. Educ., 1997, 74(11), 1297-303; and P. G. M. Wuts et al., Greene's Protective Groups in Organic Synthesis, 5th Ed., (Wiley, 2014).


Reactions can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography (TLC).


Compounds can be purified by those skilled in the art by a variety of methods, including high performance liquid chromatography (HPLC) (K. F. Blom, et al., J. Combi. Chem. 2004, 6(6), 874-83) and normal phase silica chromatography.


The Schemes below provide general guidance in connection with preparing the compounds of the invention. One skilled in the art would understand that the preparations shown in the Schemes can be modified or optimized using general knowledge of organic chemistry to prepare various compounds of the invention.




embedded image


Compounds of Formula (I) can be prepared using a process illustrated in Scheme 1. In which, compounds of Formula (1-1) can react with compounds of Formula (1-2) via nucleophilic aromatic substitution reactions (e.g., in the presence of a base, such as DIEPA) or Buchwald-Hartwig C—N coupling conditions (G. Mayoka, et al., J. Med. Chem. 2019, 62(2), 1022-35), resulting in the formation of the compounds of Formula (I). If necessary (e.g., if the ring formed by R9A and R9B contains an NBoc group), final compounds of Formula (I) can be obtained after deprotection (e.g., treatment with HCl or TFA) and corresponding functional groups conversions (e.g., urea/amide/sulfamide formations).




embedded image


For compounds of Formula (1-1) wherein X4═N, X5═CR5, and X6═CR6 can be prepared via a sequence as depicted in Scheme 2. In which, compounds of Formula (2-1) can react with compounds of Formula (2-2) to give the cyclized products of Formula (2-3) under acidic conditions (e.g., AcOH in EtOH or TFA in DMSO) (Tetrahedron 2022, 90, 153611). Compounds of Formula (2-3) can then react with activating reagents (e.g., POCl3 or Tf2O and DIPEA) (P. J. Choi et al., Tetrahedron Lett., 2022, 90, 153611) to afford the compounds of Formula (1-1).




embedded image


Alternatively, as outlined in Scheme 3, when X3═N, X1═CR1, and X2═CR2, compounds of Formula (3-1) can react with compounds of Formula (3-2) via cyclization conditions (e.g., 1-butanol, 100° C.) to afford compounds of Formula (1-1):




embedded image


For compounds of Formula (1-2), when not commercially available, they can be prepared via two different ways shown in Scheme 4. Primarily, compounds of Formula (4-1) can react 5 with NH-containing compounds of Formula (4-2) (e.g., pyrazoles or triazoles) in the presence of catalytic amount of base (e.g., DBU) to afford compounds of Formula (4-4); or they can react with boronic acid compounds of Formula (4-3) under Rh-catalyzed conditions (e.g., Rh(COD)Cl2) (G. Wuitschik, et al., J. Med. Chem. 2010, 53(8), 3227-46) to afford the nitro compounds of Formula (4-4). Compounds of Formula (4-4) can then be converted to compounds of Formula (1-2) under reduction conditions (e.g., B2(OH)4 in MeOH) (H.-C. Du, et al., Org. Lett. 2019, 21(7), 2194-99). Alternatively, compounds of Formula (4-5) can react with compounds of Formula (4-6) via SNAr conditions in the presence of strong base (e.g., LiHMDS) (A. D. Thompson, et al., J. Org. Chem. 2013, 78(2), 762-69) to afford nitrile compounds of Formula (4-7), which can then be converted to compounds of Formula (1-2) under reduction conditions (e.g., NiCl2 and NaBH4 in MeOH.) (S. Caddick, et al., Tetrahedron 2003, 59(29), 5417-23).




embedded image


Alternatively, When X1═CR1, X2═CR2, X3═X4═N, and X5═CR5, compounds of Formula (I) can be synthesized via slightly different sequences as depicted in Scheme 5. In which, compounds of Formula (5-1) can first react with compounds of Formula (1-2) under similar SNAr or Buchwald-Hartwig coupling conditions depicted in Scheme 1 to give compounds of Formula (5-2). These intermediates can then react with compounds of Formula (3-2) under the same cyclization conditions shown in Scheme 3 to afford the final compounds of Formula (I).


For the synthesis of particular compounds, the general schemes described above and specific methods described herein for preparing particular compounds can be modified. For example, the products or intermediates can be modified to introduce particular functional groups. Alternatively, the substituents can be modified at any step of the overall synthesis by methods know to one skilled in the art, e.g., as described by R. C. Larock, et al., Comprehensive Organic Transformations: A Guide to Functional Group Preparations, 3rd Ed. Vols. 1-4 (Wiley, 2018); A. R. Katritzky, et al. (Eds.), Comprehensive Organic Functional Group Transformations, Vols. 1-6 (Pergamon Press, 1995), and A. R. Katritzky et al. (Eds.), Comprehensive Organic Functional Group Transformations II, Vols. 1-6 (Elsevier, 2nd Edition, 2005);


Starting materials, reagents and intermediates whose synthesis is not expressly described herein are either commercially available, known in the literature, or may be prepared by methods known to one skilled in the art.


It will be appreciated by one skilled in the art that the processes described are not the exclusive means by which compounds of the invention may be synthesized and that a broad repertoire of synthetic organic reactions is available to be potentially employed in synthesizing compounds of the invention. The person skilled in the art knows how to select and implement appropriate synthetic routes. Suitable synthetic methods of starting materials, intermediates and products may be identified by reference to the literature, including reference sources such as: Advances in Heterocyclic Chemistry, Vols. 1-114 (Elsevier, 1963-2023); Journal of Heterocyclic Chemistry Vols. 1-60 (Journal of Heterocyclic Chemistry, 1964-2023); E. M. Carreira, et al. (Eds.) Science of Synthesis, Vols. 1-48 (2001-2010) and Knowledge Updates KU2010/1-4; 2011/1-4; 2012/1-4, 2013/1-4; 2014/1-4, 2015/1-2; 2016/1-3, 2017/1-3; 2018/1-4, 2019/1-3; 2020/1-3, 2021/1-3, 2022/1-3, 2023/1 (Thieme, 2001-2023); Houben-Weyl, Methoden der Organischen Chemie, 4th Ed. Vols. 1-67 (Thieme, 1952-1987); Houben-Weyl, Methoden der Organischen Chemie, E-Series. Vols. 1-23 (Thieme, 1982-2003); A. R. Katritzky, et al. (Eds.), Comprehensive Organic Functional Group Transformations, Vols. 1-6 (Pergamon Press, 1995); A. R. Katritzky et al. (Eds.), Comprehensive Organic Functional Group Transformations II, Vols. 1-6 (Elsevier, 2nd Edition, 2005); A. R. Katritzky et al. (Eds.); Comprehensive Heterocyclic Chemistry, Vols. 1-8 (Pergamon Press, 1984); A. R. Katritzky, et al. (Eds.); Comprehensive Heterocyclic Chemistry II, Vols. 1-10 (Pergamon Press, 1996); A. R. Katritzky, et al. (Eds.); Comprehensive Heterocyclic Chemistry III, Vols. 1-14 (Elsevier Science, 2008); D. St. C. Black, et al. (Eds.); Comprehensive Heterocyclic Chemistry IV, Vols. 1-14 (Elsevier Science, 2022); M. B. Smith et al., March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 8th Ed. (Wiley, 2020); B. M. Trost et al. (Ed.), Comprehensive Organic Synthesis, Vols. 1-9 (Pergamon Press, 1991); and Patai's Chemistry of Functional Groups, 100 Vols. (Wiley 1964-2022).


IV. Uses of the Compounds

Compounds of the present disclosure, including the compounds of Formula (I), or any of the embodiments thereof, are useful for therapy as described in further detail below. The present disclosure provides compounds of Formula (I), for use as a medicament, or for use in medicine, as described in further detail below. The present disclosure also provides the use of compounds of Formula (I), or any of the embodiments thereof, as a medicament, or for treating disease, as described in further detail below. The present disclosure also provides the use of compounds of Formula (I), or any of the embodiments thereof, in the manufacture of medicament for treating disease, as described in further detail below.


Compounds of the present disclosure can modulate, antagonize or inhibit the activity of the MRGPRX2 protein. As MRGPRX2 modulators, antagonists or inhibitors, the compounds of the disclosure are useful in the treatment of MRGPRX2 dependent conditions.


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. IgE independent activation of MRGPRX2 is capable of inducing mast cell degranulation and release of inflammatory mediators.


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.


In some embodiments, the MRGPRX2 dependent condition is an itch associated condition, such as chronic itch; senile 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 pruritus; chronic spontaneous urticaria; chronic inducible 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, porphyria; 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, stasis dermatitis; swimmer's itch; thyroid disease; tinea cruris; uremic pruritus; rosacea; cutaneous amyloidosis; scleroderma; acne; wound healing; burn healing; ocular itch; and urticaria.


In some embodiments, the MRGPRX2 dependent condition is 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 (CRPS), reflex sympathetic dystrophy syndrome (RDS), 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 neuralgia, 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, pain crisis in sickle cell disease; Parsonage-Turner syndrome, pelvic pain, periodontitis pain, peripheral neuropathy, phantom limb pain, pinched nerve, polycystic kidney disease, polymyalgia rheumatica, polymyositis, porphyria, post herniorrhaphy pain syndrome, post mastectomy pain, postoperative pain, pain syndrome, post stroke pain, post thoracotomy 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, sarcoidosis, Scheuermann's kyphosis disease, sciatica, scoliosis, shingles (herpes zoster), Sjögren'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.


In some embodiments, the MRGPRX2 dependent condition is a pseudo-allergic reaction, such as pseudo-allergic reactions caused by secretagogues, cationic peptidergic drugs, anionic peptidergic drugs, neutral peptidergic drugs, non-steroidal anti-inflammatory drugs, neuropeptides, antimicrobial peptides, opioids, neuromuscular blocking agents, antidepressant agents, antipsychotic agents, antihistamine agents, antineoplastic agents, fluoroquinolone and non-fluoroquinolone antibiotics and tyrosine-kinase inhibitors. The phrase “pseudo-allergic reaction” refers to an IgE-independent allergic reaction, characterized by release of histamine and cytokines, activation of the complement system, atypical synthesis of eicosanoids, inflammation, skin flushing, headache, edema, hypotension, urticaria (hives), bronchospasm, or any combination thereof. A pseudo-allergic reaction is a hypersensitivity reaction manifested by systemic responses. The symptoms of pseudo-allergic reaction are identical to anaphylaxis, however their mechanism is non-IgE-mediated. 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. In one embodiment, the pseudo-allergic reaction is caused by MCD peptide, substance P, VIP, PACAP, dynorphin, somatostatin, Compound 48/80, cortistatin-14, mastoparan, melittin, 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.


In some embodiments, the MRGPRX2 dependent condition is an autoimmune disorder or inflammatory condition, 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 eosinophilia, 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, schwannomatosis, 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, primary sclerosing cholangitis, primary biliary cholangitis, neuromyelitis optica spectrum 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.


In some embodiments, the compounds of the disclosure are useful 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-Dubé 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-Fraumeni syndrome, lymphoma (Hodgkin and non-Hodgkin), lynch syndrome, mastocytosis, medulloblastoma, meningioma, mesothelioma, multiple endocrine neoplasia, multiple myeloma, MUTYH-associated polyposis, myelodysplastic syndrome, nasal cavity and paranasal sinus cancer, neuroblastoma, neuroendocrine tumors, neurofibromatosis, penile cancer, parathyroid cancer, ovarian fallopian tube and peritoneal cancer, osteosarcoma, pituitary gland tumor, pleuropulmonary blastoma, oral and oropharyngeal, thyroid, uterine, pancreatic, carney complex, brain and spinal cord cancer, cervical cancer, Cowden syndrome, craniopharyngioma, desmoid tumor, desmoplastic 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, leiomyomatosis and renal cell cancer, hereditary pancreatitis, hereditary papillary renal carcinoma, hereditary mixed polyposis syndrome, HIV/AIDS related cancers, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, Kaposi sarcoma, small bowel cancer, stomach cancer, testicular cancer, thymoma and thymic carcinoma, thyroid cancer, vaginal cancer, culver cancer, Wermer's syndrome and xeroderma pigmentosum.


The MRGPRX2 dependent condition may be selected from the group consisting of abdominal aortic aneurysms, acute contact dermatitis, allergic rhinitis, amyotrophic lateral sclerosis, asthma, atopic dermatitis, autism, cancer, chronic inducible urticaria, chronic itch, chronic obstructive pulmonary disease, chronic spontaneous urticaria, cold urticaria, contact urticaria, coronary artery disease, cough, Crohn's disease, deep vein thrombosis, drug-induced anaphylactic reactions, endometriosis, fibromyalgia, geographic atrophy, idiopathic chronic cough, idiopathic pulmonary fibrosis, inflammatory pain, interstitial cystitis, irritable bowel syndrome, mast cell activation syndrome, mastocytosis, metabolic syndrome, migraine, multiple sclerosis, nasal polyps, neurodermatitis, neuropathic itch, neuropathic pain, obesity, oesophageal reflux, osteoarthritis, periodontitis, prurigo nodularis, pruritus, pseudo-anaphylaxis, psoriasis, rheumatoid arthritis, rosacea, seborrheic dermatitis, sickle cell disease, ulcerative colitis, and ulcers.


The MRGPRX2 dependent condition may be selected from the group consisting of autoimmune diseases, arthritis, rheumatoid arthritis, psoriatic arthritis, juvenile idiopathic arthritis, multiple sclerosis, systemic lupus erythematosus (SLE), myasthenia gravis, juvenile onset diabetes, diabetes mellitus type 1, graft-versus-host disease (GvHD), Guillain-Barre syndrome, Hashimoto's encephalitis, Hashimoto's thyroiditis, ankylosing spondylitis, psoriasis, Sjögren's syndrome, vasculitis, glomerulonephritis, auto-immune thyroiditis, Behcet's disease, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, ichthyosis, Graves ophthalmopathy, inflammatory bowel disease, Addison's disease, vitiligo, asthma, allergic asthma, acne vulgaris, celiac disease, chronic prostatitis, inflammatory bowel disease, pelvic inflammatory disease, reperfusion injury, ischemia reperfusion injury, stroke, sarcoidosis, transplant rejection, interstitial cystitis, atherosclerosis, scleroderma, and atopic dermatitis.


The MRGPRX2 dependent condition may be selected from the group consisting of acute disseminated encephalomyelitis (ADEM), acute necrotizing hemorrhagic leukoencephalitis, Addison's disease, agammaglobulinemia, alopecia areata, amyloidosis, ankylosing spondylitis, anti-GBM/Anti-TBM nephritis, antiphospholipid syndrome (APS), autoimmune angioedema, autoimmune aplastic anemia, autoimmune dysautonomia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune immunodeficiency, autoimmune inner ear disease (AIED), autoimmune myocarditis, autoimmune oophoritis, autoimmune pancreatitis, autoimmune retinopathy, autoimmune thrombocytopenic purpura (ATP), autoimmune thyroid disease, autoimmune urticaria, axonal or neuronal neuropathies, Balo disease, Behcet's disease, bullous pemphigoid, cardiomyopathy, Castleman disease, celiac disease, Chagas disease, chronic fatigue syndrome, chronic inflammatory demyelinating polyneuropathy (CIDP), chronic recurrent multifocal ostomyelitis (CRMO), Churg-Strauss syndrome, cicatricial pemphigoid/benign mucosal pemphigoid, Crohn's disease, Cogan's syndrome, cold agglutinin disease, congenital heart block, coxsackie myocarditis, CREST disease, essential mixed cryoglobulinemia, demyelinating neuropathies, dermatitis herpetiformis, dermatomyositis, Devic's disease (neuromyelitis optica), discoid lupus, Dressler's syndrome, endometriosis, eosinophilic esophagitis, eosinophilic fasciitis, erythema nodosum, experimental allergic encephalomyelitis, Evans syndrome, fibromyalgia, fibrosing alveolitis, giant cell arteritis (temporal arteritis), giant cell myocarditis, glomerulonephritis, Goodpasture's syndrome, granulomatosis with polyangiitis (GPA) (formerly called Wegener's granulomatosis), Graves' disease, Guillain-Barre syndrome, Hashimoto's encephalitis, Hashimoto's thyroiditis, hemolytic anemia, Henoch-Schonlein purpura, herpes gestationis, hypogammaglobulinemia, idiopathic thrombocytopenic purpura (ITP), IgA nephropathy, IgG4-related sclerosing disease, Immunoregulatory lipoproteins, inclusion body myositis, interstitial cystitis, juvenile arthritis, juvenile diabetes (type 1 diabetes), juvenile myositis, Kawasaki syndrome, Lambert-Eaton syndrome, leukocytoclastic vasculitis, lichen planus, lichen sclerosus, ligneous conjunctivitis, linear IgA disease (LAD), lupus (SLE), Lyme disease, chronic, Meniere's disease, microscopic polyangiitis, mixed connective tissue disease (MCTD), Mooren's ulcer, Mucha-Habermann disease, multiple sclerosis, myasthenia gravis, myositis, narcolepsy,neuromyelitis optica (Devic's), neutropenia, ocular cicatricial pemphigoid, optic neuritis, palindromic rheumatism, PANDAS (pediatric autoimmune neuropsychiatric disorders associated with streptococcus), paraneoplastic cerebellar degeneration, paroxysmal nocturnal hemoglobinuria (PNH), Parry Romberg syndrome, Parsonnage-Turner syndrome, pars planitis (peripheral uveitis), pemphigus, peripheral neuropathy, perivenous encephalomyelitis, pernicious anemia, POEMS syndrome, polyarteritis nodosa, type I, II, & III autoimmune polyglandular syndromes, polymyalgia rheumatica, polymyositis, postmyocardial infarction syndrome, postpericardiotomy syndrome, progesterone dermatitis, primary biliary cirrhosis, primary sclerosing cholangitis, psoriasis, psoriatic arthritis, idiopathic pulmonary fibrosis, pyoderma gangrenosum, pure red cell aplasia, Raynaud's phenomenon, reactive arthritis, reflex sympathetic dystrophy, Reiter's syndrome, relapsing polychondritis, restless legs syndrome, retroperitoneal fibrosis, rheumatic fever, rheumatoid arthritis, sarcoidosis, Schmidt syndrome, scleritis, scleroderma, Sjögren's syndrome, sperm & testicular autoimmunity, stiff person syndrome, subacute bacterial endocarditis (SBE), Susac's syndrome, sympathetic ophthalmia, takayasu's arteritis, temporal arteritis/giant cell arteritis, thrombocytopenic purpura (TTP), tolosa-hunt syndrome, transverse myelitis, type 1 diabetes, ulcerative colitis, undifferentiated connective tissue disease (UCTD), uveitis, vasculitis, vesiculobullous dermatosis, vitiligo, or Wegener's granulomatosis (i.e., granulomatosis with polyangiitis (GPA)).


The present disclosure provides a method of treating a disease in a patient. The disease can be a MRGPRX2 dependent condition, including any of the MRGPRX2 dependent conditions described herein. The method comprises administering to the patient in need of the treatment a therapeutically effective amount of a compound of Formula (I), or any of the embodiments thereof. The condition treated can be any of the conditions described herein.


The phrase “MRGPRX2 dependent condition” refers to a condition in which 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), neuromuscular blocking agents (atracurium mivacurium), non-steroidal anti-inflammatory drugs, neuropeptides, 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 being bound by theory, it is to be understood that by modulating MRGPRX2, pseudo-allergic reactions, itch, pain, inflammatory and autoimmune disorders can be eased.


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.


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).


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.


The term “cell” is meant to refer to a cell that is in vitro, ex vivo or in vivo. In some embodiments, an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal. In some embodiments, an in vitro cell can be a cell in a cell culture. In some embodiments, an in vivo cell is a cell living in an organism such as a mammal.


The term “contacting” refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, “contacting” the MRGPRX2 with a compound described herein includes the administration of a compound described herein to an individual or patient, such as a human, having MRGPRX2, as well as, for example, introducing a compound described herein into a sample containing a cellular or purified preparation containing the MRGPRX2.


The term “individual” or “patient,” used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.


The phrase “therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent such as an amount of any of the solid forms or salts thereof as disclosed herein that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. An appropriate “effective” amount in any individual case may be determined using techniques known to a person skilled in the art.


The phrase “pharmaceutically acceptable” is used herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, immunogenicity or other problem or complication, commensurate with a reasonable benefit/risk ratio.


The phrase “pharmaceutically acceptable carrier or excipient” refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or encapsulating material. Excipients or carriers are generally safe, non-toxic and neither biologically nor otherwise undesirable and include excipients or carriers that are acceptable for veterinary use as well as human pharmaceutical use. In one embodiment, each component is “pharmaceutically acceptable” as defined herein. See, e.g., Remington: The Science and Practice of Pharmacy, 21st ed.; Lippincott Williams & Wilkins: Philadelphia, Pa., 2005; Handbook of Pharmaceutical Excipients, 6th ed.; Rowe et al., Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed.; Ash and Ash Eds.; Gower Publishing Company: 2007; Pharmaceutical Preformulation andFormulation, 2nd ed.; Gibson Ed.; CRC Press LLC: Boca Raton, Fla., 2009.


The term “treating” or “treatment” refers to inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology) or ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease.


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.


It is appreciated that certain features of the disclosure, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment (while the embodiments are intended to be combined as if written in multiply dependent form). Conversely, various features of the disclosure which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.


V. Combination Therapies

One or more additional pharmaceutical agents or treatment methods can be used in combination with compounds described herein for treatment of MRGPRX2 dependent conditions, as described herein. The agents can be combined with the present compounds in a single dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.


In some embodiments, the additional therapeutic agent is an antihistamine, such as an H1 receptor antagonist or an H2 receptor antagonist. In one embodiment, the additional therapeutic agent is an H1 receptor antagonist antihistamine, such as levocetirizine, loratadine, fexofenadine, cetirizine, desloratadine, olopatadine, diphenhydramine, cyproheptadine, hydroxyzine pamoate or ketotifen. In one embodiment, the additional therapeutic agent is a H2 receptor antagonist, such as cimetidine, nizatidine, ranitidine or famotidine. In one embodiment, the additional 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 additional therapeutic agent is an immunomodulatory agent such as omalizumab or immunoglobulin therapy. In one embodiment, the additional therapeutic agent is a corticosteroid, such as hydrocortisone, cortisone, betamethasone, triamcinolone, prednisone, prednisolone, or fludrocortisone. In one embodiment, the additional therapeutic agent is a tricyclic antidepressant that can relieve itch such as doxepin, amitriptyline or nortriptyline. In one embodiment, the additional therapeutic agent is an anti-inflammatory drug such as dapsone, sulfasalazine, hydroxychloroquine or colchicine. In one embodiment, the additional therapeutic agent is an immunosuppressant such as cyclosporine, methotrexate, mycophenolic acid or tacrolimus.


VI. Pharmaceutical Formulations and Dosage Forms

When employed as pharmaceuticals, compounds described herein can be administered in the form of pharmaceutical compositions which refers to a combination of one or more compounds described herein, and at least one pharmaceutically acceptable carrier or excipient. These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal), ocular, oral or parenteral. Methods for ocular delivery can include topical administration (eye drops), subconjunctival, periocular or intravitreal injection or introduction by balloon catheter or ophthalmic inserts surgically placed in the conjunctival sac. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal, or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.


This disclosure also includes pharmaceutical compositions which contain, as the active ingredient, one or more compounds described herein in combination with one or more pharmaceutically acceptable carriers or excipients. In making the compositions described herein, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders. In some embodiments, the composition is suitable for topical administration.


In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.


The compounds of the disclosure may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types. Finely divided (nanoparticulate) preparations of the compounds of the disclosure can be prepared by processes known in the art see, e.g., WO 2002/000196.


Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions described herein can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.


In some embodiments, the pharmaceutical composition comprises silicified microcrystalline cellulose (SMCC) and at least one compound described herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the silicified microcrystalline cellulose comprises about 98% microcrystalline cellulose and about 2% silicon dioxide w/w.


In some embodiments, the composition is a sustained release composition comprising at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one component selected from microcrystalline cellulose, lactose monohydrate, hydroxypropyl methylcellulose and polyethylene oxide. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystalline cellulose, lactose monohydrate and hydroxypropyl methylcellulose. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystalline cellulose, lactose monohydrate and polyethylene oxide. In some embodiments, the composition further comprises magnesium stearate or silicon dioxide. In some embodiments, the microcrystalline cellulose is Avicel PH102™. In some embodiments, the lactose monohydrate is Fast-flo 316™. In some embodiments, the hydroxypropyl methylcellulose is hydroxypropyl methylcellulose 2208 K4M (e.g., Methocel K4 M Premier™) and/or hydroxypropyl methylcellulose 2208 K100LV (e.g., Methocel K00LV™). In some embodiments, the polyethylene oxide is polyethylene oxide WSR 1105 (e.g., Polyox WSR 1105™).


In some embodiments, a wet granulation process is used to produce the composition. In some embodiments, a dry granulation process is used to produce the composition.


The compositions can be formulated in a unit dosage form, each dosage containing from, for example, about 5 mg to about 1000 mg, about 5 mg to about 100 mg, about 100 mg to about 500 mg, or about 10 to about 30 mg, of the active ingredient. In some embodiments, each dosage contains about 10 mg of the active ingredient. In some embodiments, each dosage contains about 50 mg of the active ingredient. In some embodiments, each dosage contains about 25 mg of the active ingredient. The term “unit dosage forms” refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.


The components used to formulate the pharmaceutical compositions are of high purity and are substantially free of potentially harmful contaminants (e.g., at least National Food grade, generally at least analytical grade, and more typically at least pharmaceutical grade). Particularly for human consumption, the composition is preferably manufactured or formulated under Good Manufacturing Practice standards as defined in the applicable regulations of the U.S. Food and Drug Administration. For example, suitable formulations may be sterile and/or substantially isotonic and/or in full compliance with all Good Manufacturing Practice regulations of the U.S.


Food and Drug Administration.

The active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.


The therapeutic dosage of a compound of the present disclosure can vary according to, e.g., the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound of the disclosure in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, the compounds of the disclosure can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 μg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.


For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid pre-formulation composition containing a homogeneous mixture of one or more compounds described herein. When referring to these pre-formulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid pre-formulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present disclosure.


The tablets or pills of the present disclosure can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.


The liquid forms in which the compounds, or compositions as described herein can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.


Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.


Topical formulations can contain one or more conventional carriers. In some embodiments, ointments can contain water and one or more hydrophobic carriers selected from, e.g., liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like. Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g., glycerinemonostearate, PEG-glycerinemonostearate and cetylstearyl alcohol. Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, e.g., glycerol, hydroxyethyl cellulose, and the like. In some embodiments, topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2 or at least about 5 wt % of the compound of the disclosure. The topical formulations can be suitably packaged in tubes of, e.g., 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.


The amount of compound or composition administered to a patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.


The compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.


The therapeutic dosage of a compound of the present disclosure can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of the compounds in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, compounds of the present disclosure can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 μg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.


Compounds described herein can also be formulated in combination with one or more additional active ingredients, which can include any pharmaceutical agent such as anti-viral agents, vaccines, antibodies, immune enhancers, immune suppressants, anti-inflammatory agents and the like.


VII. Labelled Compounds

Compounds of the disclosure also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers.


The present disclosure further includes isotopically-labelled compounds of the disclosure. An “isotopically-labelled” is a compound of the disclosure where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). A “radio-labelled” compound is an isotopically-labelled compound in which one or more atoms are replaced or substituted by an atom of an isotope that is radioactive.


Suitable isotopes that may be incorporated in compounds of the present disclosure include but are not limited to 2H (also written as D for deuterium), 3H (also written as T for tritium), 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 18F, 35S, 36Cl, 82Br, 75Br, 76Br, 77Br, 123I, 124I, 125I and 131I. For example, one or more hydrogen atoms in a compound of the present disclosure can be replaced by deuterium atoms (e.g., one or more hydrogen atoms of a C1-6 alkyl group of Formula (I) can be optionally substituted with deuterium atoms, such as —CD3 being substituted for —CH3). In some embodiments, alkyl groups in Formula (I) can be perdeuterated. The symbol D included in a chemical formula or as a substituent indicates that deuterium is incorporated in the position labelled at greater than natural abundance, and typically indicates an abundance of equal to or greater than 50%, preferably equal to or greater than 90% or equal to or greater than 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.95%, or 99.99% relative to other forms of hydrogen or relative to all forms of hydrogen.


One or more constituent atoms of the compounds presented herein can be replaced or substituted with isotopes of the atoms in natural or non-natural abundance. In some embodiments, the compound includes at least one deuterium atom. In some embodiments, the compound includes at least one deuterium atom. For example, one or more hydrogen atoms in a compound of the present disclosure can be replaced or substituted by deuterium. In some embodiments, the compound includes two or more deuterium atoms. In some embodiments, the compound includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 deuterium atoms.


Synthetic methods for including isotopes into organic compounds are known in the art (A. F. Thomas, Deuterium Labeling in Organic Chemistry, (Appleton-Century-Crofts, New York, N.Y., 1971); J. Atzrodt, et al., Angew. Chem. Int. Ed., 2007, 7744-65; J. R. Hanson, The Organic Chemistry of Isotopic Labelling, (Royal Society of Chemistry, 2011)). Isotopically labelled compounds can used in various studies such as NMIR spectroscopy, metabolism experiments, and/or assays.


Substitution with heavier isotopes such as deuterium, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances. (A. Kerekes, et al., J. Med. Chem. 2011, 54(1), 201-10; R. Xu et al., J. Label. Compd. Radiopharm. 2015, 58, 308-12).


The radionuclide that is incorporated in the instant radio-labelled compounds will depend on the specific application of that radio-labelled compound. For example, for in vitro adenosine receptor labeling and competition assays, compounds that incorporate 3H, 14C, 82Br, 125I, 131I or 35S can be useful. For radio-imaging applications 11C, 18F, 125I, 123I, 124I, 131I, 75Br, 76Br or 77Br can be useful.


It is understood that a “radio-labelled” or “labelled compound” is a compound that has incorporated at least one radionuclide. In some embodiments, the radionuclide is selected from the group consisting of 3H, 14C, 125I, 35S and 82Br.


The present disclosure can further include synthetic methods for incorporating radio-isotopes into compounds of the disclosure. Synthetic methods for incorporating radio-isotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily recognize the methods applicable for the compounds of disclosure.


VIII. Kits

The present disclosure also includes pharmaceutical kits useful, for example, in the treatment of MRGPRX2 dependent conditions, as described herein, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the disclosure. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.


Examples

The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of non-critical parameters which can be changed or modified to yield essentially the same results.


Experimental procedures for compounds of the disclosure are provided below. Preparatory LCMS purifications of some of the compounds prepared were performed on Waters mass-directed fractionation systems. The basic equipment setup, protocols, and control software for the operation of these systems have been described in detail in the literature. See e.g., K. F. Blom, J. Combi. Chem., 2002, 4(4), 295-301; K. F. Blom, et al., J. Combi. Chem., 2003, 5(5), 670-683; and K. F. Blom, et al., J. Combi. Chem. 2004, 6(6), 874-83. The compounds separated were typically subjected to analytical liquid chromatography mass spectrometry (LCMS) for purity analysis under the following conditions: Instrument; Agilent 1100 series, LC/MSD, Column: Waters SUNFIRE® C18 5 μm, 2.1×50 mm, Buffers: mobile phase A: 0.025% aq. TFA and mobile phase B: MeCN; gradient 2% to 80% of B in 3 min. with flow rate 2.0 mL/min.


Some of the compounds prepared were also separated on a preparative scale by reverse-phase high performance liquid chromatography (RP-HPLC) with MS detector or FCC (silica gel) as indicated in the Examples. Typical preparative reverse-phase high performance liquid chromatography (RP-HPLC) column conditions are as follows:


pH=2 purifications: Waters SUNFIRE® C18 5 μm, 19×100 mm column, eluting with mobile phase A: 0.1% aq. TFA and mobile phase B: MeCN; the flow rate was 30 mL/min., the separating gradient was optimized for each compound using the Compound Specific Method Optimization protocol as described in the literature [see K. F. Blom, et al., J. Combi. Chem. 2004, 6(6), 874-83]. Typically, the flow rate used with the 30×100 mm column was 60 mL/min.


pH=10 purifications: Waters XBRIDGE® C18 5 μm, 19×100 mm column, eluting with mobile phase A: 0.15% aq. NH4OH and mobile phase B: MeCN; the flow rate was 30 mL/min., the separating gradient was optimized for each compound using the Compound Specific Method Optimization protocol as described in the literature [See K. F. Blom, et al., J. Combi. Chem. 2004, 6(6), 874-83]. Typically, the flow rate used with 30×100 mm column was 60 mL/min.


Example 1. 4-(((7-Chloro-2-(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide



embedded image


Step 1. tert-Butyl 4-(((2-amino-6-chloropyrimidin-4-yl)amino)methyl)-4-phenylpiperidine-1-carboxylate



embedded image


A mixture of 4,6-dichloropyrimidin-2-amine (100 mg, 0.61 mmol), tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate (177 mg, 0.61 mmol) and DIPEA (0.39 mL, 2.26 mmol) in DMSO (2 mL) was heated at 125° C. overnight. Upon completion, the reaction mixture was allowed to cool to r.t. and was poured into water (10 mL) with vigorous stirring. The sub-title compound precipitated as pale yellow solid, which was then filtered, washed with water and hexane and dried over air flow. The product was used for the next reaction without further purification. LCMS calc. for C21H29ClN5O2 (M+H)+: m/z=418.2. found: 418.2.


Step 2. 7-Chloro-N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-amine



embedded image


A mixture of tert-butyl 4-(((2-amino-6-chloropyrimidin-4-yl)amino)methyl)-4-phenylpiperidine-1-carboxylate (20 mg, 0.048 mmol) and 1-chloro-3,3-difluorobutan-2-one (7 mg, 0.048 mmol) in 1,4-dioxane (1 mL) was heated at 100° C. overnight. The mixture was allowed to cool to r.t. and TFA (1 mL) was added and stirred for 10 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C19H20ClF3N5 (M+H)+: m/z=410.1. found: 410.1.


Step 3. 4-(((7-Chloro-2-(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide

A mixture of 7-chloro-N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-amine (5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and phenyl carbamate (3 mg, 0.023 mmol) in 1,4-dioxane (0.5 mL) was heated at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C20H21ClF3N6O (M+H)+: m/z=453.1. found: 453.1.


Example 2. 4-Phenyl-4-(((2-(trifluoromethyl)imidazo[1,2-c]pyrimidin-5-yl)amino)methyl)piperidine-1-carboxamide



embedded image


A mixture of N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-c]pyrimidin-5-amine (Example 5, 5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and phenyl carbamate (3 mg, 0.023 mmol) in 1,4-dioxane (0.5 mL) was heated at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C20H22F3N6O (M+H)+: m/z=419.2. found: 419.2.


Example 3. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-sulfonamide



embedded image


A mixture of N-((3-phenylpyrrolidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (Example 4, step 1, 5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and sulfuric diamide (5 mg, 0.056 mmol) in 1,4-dioxane (0.5 mL) was heated at 115° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C19H19F6N6O2S (M+H)+: m/z=509.1. found: 509.1.


The title compound exists as enantiomers that are separable, e.g., by chiral chromatography: (R)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-sulfonamide and (S)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-sulfonamide.


Example 4. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-carboxamide



embedded image


Step 1. N-((3-Phenylpyrrolidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


To a stirred solution of 2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol (100 mg, 0.369 mmol) and DIPEA (193 μL, 1.11 mmol) in DCM (5 mL) was added Tf2O (0.075 mL, 0.44 mmol) dropwise at 0° C. Then, the resulting solution was added into a stirred solution of tert-butyl 3-(aminomethyl)-3-phenylpyrrolidine-1-carboxylate (107 mg, 0.369 mmol) and DIPEA 193 μL, 1.11 mmol) in DCM (5 mL) at 0° C. The resulting mixture was stirred at r.t. for 1 h before TFA (5 mL) was added and stirring was continued for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C19H18F6N5 (M+H)+: m/z=430.1. found: 430.1.


Step 2. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-carboxamide

A mixture of N-((3-phenylpyrrolidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and phenyl carbamate (3 mg, 0.023 mmol) in 1,4-dioxane (0.5 mL) was heated at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C20H19F6N6O (M+H)+: m/z=473.1. found: 473.1.


The title compound exists as enantiomers that are separable, e.g., by chiral chromatography: (R)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-carboxamide and (S)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylpyrrolidine-1-carboxamide.


Example 5. 4-(((7-Chloro-2-(trifluoromethyl)imidazo[1,2-c]pyrimidin-5-yl)amino)methyl)-4-phenylpiperidine-1-sulfonamide



embedded image


Step 1. 7-Chloro-N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-c]pyrimidin-5-amine



embedded image


A mixture of 5,7-dichloro-2-(trifluoromethyl)imidazo[1,2-c]pyrimidine (0.226 mmol), 5 tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate (64 mg, 0.226 mmol) and DIPEA (0.39 mL, 2.26 mmol) in DMSO (1 mL) was heated at 125° C. overnight. The resulting mixture was allowed to cool to r.t. before TFA (5 mL) was added and stirring was continued for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C19H20ClF3N5 (M+H)+: m/z=410.1. found: 410.1.


Step 2. 4-(((7-Chloro-2-(trifluoromethyl)imidazo[1,2-c]pyrimidin-5-yl)amino)methyl)-4-phenylpiperidine-1-sulfonamide

A mixture of 7-chloro-N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-c]pyrimidin-5-amine (5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and sulfuric diamide (5 mg, 0.056 mmol) in 1,4-dioxane (0.5 mL) was heated at 85° C. and stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C19H21ClF3N6O2S (M+H)+: m/z=489.1. found: 489.1.


Example 6. 4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-sulfonamide



embedded image


A mixture of N-((4-phenylpiperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (example 16, step 1, 5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and sulfuric diamide (5 mg, 0.056 mmol) in 1,4-dioxane (0.5 mL) was heated at 115° C. and stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C20H21F6N6O2S (M+H)+: m/z=523.1. found: 523.1.


Example 7. 4-(((5-Methyl-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide



embedded image


Step 1. 5-Methyl-N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine



embedded image


A mixture of 7-chloro-5-methyl-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidine (50 mg, 0.211 mmol), tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate (61 mg, 0.211 mmol) and DIPEA (111 μL, 0.634 mmol) in 1,4-dioxane (2.0 mL) was heated at 100° C. for 2 h. The resulting mixture was allowed to cool to r.t. before TFA (5 mL) was added and stirring was continued for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with DCM (30 mL), washed with sat. aq. NaHCO3 and dried over Na2SO4. The organic layers were filtered and concentrated to afford the sub-title compound, which was subjected to the following reaction without further purification. LCMS calc. for C19H22F3N6 (M+H)+: m/z=391.2. found: 391.2.


Step 2. 4-(((5-Methyl-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide

A mixture of 5-methyl-N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (10 mg, 0.026 mmol), DIPEA (0.022 mL, 0.13 mmol) 5 and isocyanatotrimethylsilane (4.43 mg, 0.038 mmol) in DCM (1.0 mL) was stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C20H23F3N7O (M+H)+: m/z=434.2. found: 434.2.


Example 8. 2-Methyl-N-((4-phenylpiperidin-4-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


A mixture of 7-chloro-2-methyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (50 mg, 0.21 mmol), tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate (62 mg, 0.21 mmol) and DIPEA (110 μL, 0.64 mmol) in 1,4-dioxane (2.0 mL) was heated at 100° C. for 2 h. The resulting mixture was allowed to cool to r.t. before TFA (5 mL) was added and stirring was continued for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C20H23F3N5 (M+H)+: m/z=390.2. found: 390.2.


Example 9. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluoro-1H-pyrazol-1-yl)azetidine-1-sulfonamide



embedded image


A mixture of N-((3-(4-fluoro-1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (example 10, step 2, 5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and sulfuric diamide (5 mg, 0.056 mmol) in 1,4-dioxane (0.5 mL) was heated at 85° C. and stirred for 30 min. Upon completion, the reaction mixture was 5 diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C15H14F7N8O2S (M+H)+: m/z=503.1. found: 503.1.


Example 10. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluoro-1H-pyrazol-1-yl)azetidine-1-carboxamide



embedded image


Step 1. tert-Butyl 3-(aminomethyl)-3-(4-fluoro-1H-pyrazol-1-yl)azetidine-1-carboxylate



embedded image


To a solution of tert-butyl 3-(nitromethylene)azetidine-1-carboxylate (1.0 g, 4.67 mmol) and 4-fluoro-1H-pyrazole (402 mg, 4.67 mmol) in 20 mL MeCN was added DBU (70 mg, 0.47 mmol) and the resulting mixture was stirred at r.t. overnight. The solution was then cooled to 0° C. and MeOH (5 mL), 4,4′-bipyridine (73 mg, 0.47 mmol) and hypodiboric acid (1.25 g, 14.0 mmol) were added. Upon completion of the reaction, the reaction mixture was concentrated in vacuo. The residue was dissolved in DCM and washed with sat. aq. NaHCO3. The organic layers were concentrated in vacuo. The crude product (1.0 g) was used without further purification. LCMS calc. for C12H20FN4O2 (M+H)+: m/z=271.2. found: 271.2.


Step 2. N-((3-(4-Fluoro-1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


To a stirred solution of 2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol (100 mg, 0.369 mmol), and DIPEA (193 μL, 1.11 mmol) in DCM (5 mL), Tf2O (0.075 mL, 0.44 mmol) was added dropwise at 0° C. The resulting solution was added into a stirred solution of tert-butyl 3-(aminomethyl)-3-(4-fluoro-1H-pyrazol-1-yl)azetidine-1-carboxylate (120 mg, 0.444 mmol) and DIPEA (193 μL, 1.11 mmol) in DCM (5 mL) at 0° C. The resulting mixture was stirred at r.t. for 1 h before TFA (5 mL) was added and stirring was continued at r.t. for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C15H13F7N7 (M+H)+: m/z=424.1.1. found: 424.1.


Step 3. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluoro-1H-pyrazol-1-yl)azetidine-1-carboxamide

A mixture of N-((3-(4-fluoro-1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and phenyl carbamate (3 mg, 0.023 mmol) in 1,4-dioxane (0.5 mL) was heated and stirred at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C16H14F7N8O (M+H)+: m/z=467.1. found: 467.1.


Example 11. 3-(4-Fluoro-1H-pyrazol-1-yl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-sulfonamide



embedded image


A mixture of N-((3-(4-fluoro-1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine (example 12, step 2, 5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and sulfuric diamide (5 mg, 0.056 mmol) in 1,4-dioxane (0.5 mL) was heated and stirred at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C14H15F4N8O2S (M+H)+: m/z=435.1. found: 435.1.


Example 12. 3-(4-Fluoro-1H-pyrazol-1-yl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide



embedded image


Step 1. N-((3-(4-Fluoro-1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine



embedded image


A mixture of 5-chloro-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridine (50 mg, 0.226 mmol), tert-butyl 3-(aminomethyl)-3-(4-fluoro-1H-pyrazol-1-yl)azetidine-1-carboxylate (60 mg, 0.222 mmol) and DIPEA (0.39 mL, 2.26 mmol) in DMSO (1 mL) was heated at 125° C. overnight. The resulting mixture was allowed to cool to r.t. before TFA (5 mL) was added and stirring was continued for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C14H14F4N7 (M+H)+: m/z=356.1. found: 356.1.


Step 2. 3-(4-Fluoro-1H-pyrazol-1-yl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide

A mixture of N-((3-(4-fluoro-1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine (5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and phenyl carbamate (3 mg, 0.023 mmol) in 1,4-dioxane (0.5 mL) was heated and stirred at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C15H15F4N8O (M+H)+: m/z=399.1. found: 399.1.


Example 13. N-Methyl-4-phenyl-4-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)piperidine-1-carboxamide



embedded image


Step 1. N-((4-Phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine



embedded image


A mixture of 5-chloro-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridine (50 mg, 0.226 mmol), tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate (64 mg, 0.226 mmol) and DIPEA (0.39 mL, 2.26 mmol) in DMSO (1 mL) was heated at 125° C. overnight. The resulting mixture was allowed to cool to r.t. before TFA (5 mL) was added and 5 stirring was continued for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C19H21F3N5 (M+H)+: m/z=376.2. found: 376.2.


Step 2. N-Methyl-4-phenyl-4-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)piperidine-1-carboxamide

A mixture of N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine (5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and methylcarbamic chloride (1.6 mg, 0.017 mmol) in DCM (1.0 mL) was stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C21H24F3N6O (M+H)+: m/z=433.2. found: 433.2.


Example 14. Methyl 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxylate



embedded image


A mixture of N-((4-phenylpiperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (example 16, step 1, 5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and methyl carbonochloridate (1.6 mg, 0.017 mmol) in DCM (1.0 mL) was stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C22H22F6N5O2 (M+H)+: m/z=502.2. found: 502.2.


Example 15. 4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-methyl-4-phenylpiperidine-1-carboxamide



embedded image


A mixture of N-((4-phenylpiperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (example 16, step 1, 5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and methylcarbamic chloride (1.6 mg, 0.017 mmol) in DCM (1.0 mL) was stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C22H23F6N6O (M+H)+: m/z=501.2. found: 501.2.


Example 16. 4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide



embedded image


Step 1. N-((4-Phenylpiperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


To a stirred solution of 2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol (100 mg, 0.369 mmol), and DIPEA (193 μL, 1.11 mmol) in DCM (5 mL), Tf2O (0.075 mL, 0.44 mmol) was added dropwise at 0° C. Then, the resulting solution was added into a stirred solution of tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate (107 mg, 0.369 mmol) and DIPEA (193 μL, 1.11 mmol) in DCM (5 mL) at 0° C. The resulting mixture was stirred at r.t. for 1 h before TFA (5 mL) was added and stirring was continued at r.t. for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C20H20F6N5 (M+H)+: m/z=444.2. found: 444.2.


Step 2. 4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide

A mixture of N-((4-phenylpiperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (5 mg, 0.011 mmol), DIPEA (0.020 mL, 0.11 mmol) and phenyl carbamate (3 mg, 0.023 mmol) in 1,4-dioxane (0.5 mL) was heated and stirred at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C21H21F6N6O (M+H)+: m/z=487.2. found: 487.2.


Example 17. N-((1-Methyl-4-phenylpiperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


Step 1. 2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol



embedded image


A solution of 3-(trifluoromethyl)-1H-pyrazol-5-amine (2.0 g, 13.24 mmol) and ethyl (Z)-4,4,4-trifluoro-3-hydroxybut-2-enoate (2.4 g, 13.24 mmol) in EtOH (20 mL) was stirred at 100° C. for 12 h. Upon completion, the reaction mixture was cooled and concentrated in vacuo, the crude product was then recrystallized in MeOH to afford the sub-title compound as a colorless solid. LCMS calc. for C8H4F6N3O (M+H)+: m/z=272.0. found: 272.0.


Step 2. N-((1-Methyl-4-phenylpiperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine

To a stirred solution of 2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol (25 mg, 0.092 mmol), and DIPEA (48.3 μL, 0.277 mmol) in DCM (1 mL), Tf2O (0.019 mL, 0.111 mmol) was added dropwise at 0° C. Then, the resulting solution was added into a stirred solution of (1-methyl-4-phenylpiperidin-4-yl)methanamine (18.8 mg, 0.092 mmol) and DIPEA (48.3 μL, 0.277 mmol) in DCM (1 mL) at 0° C. After 30 min., the reaction mixture was concentrated and diluted with 4 mL MeOH, and the resulting solution was then purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C21H22F6N5 (M+H)+: m/z=458.2. found: 458.2.


Example 18. N-Methyl-4-(((5-methyl-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide



embedded image


A mixture of 5-methyl-N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (example 7, step 1, 10 mg, 0.026 mmol), DIPEA (0.022 mL, 0.13 mmol) and methylcarbamic chloride (3.6 mg, 0.038 mmol) in DCM (1.0 mL) was stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C21H25F3N7O (M+H)+: m/z=448.2. found: 448.2.


Example 19. (3,3-Difluoroazetidin-1-yl)(4-phenyl-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidin-1-yl)methanone



embedded image


A mixture of N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine (example 21, step 1, 10 mg, 0.027 mmol), DIPEA (0.014 mL, 0.08 mmol) and a 3,3-difluoroazetidine-1-carbonyl chloride (6 mg, 0.040 mmol) in DCM (1.0 mL) was stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep.


LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C24H25F5N5O (M+H)+: m/z=494.2. found: 494.2.


Example 20. 1-(4-Phenyl-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidin-1-yl)ethan-1-one



embedded image


A mixture of N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine (example 21, step 1, 10 mg, 0.027 mmol), DIPEA (0.014 mL, 0.08 mmol) and acetyl chloride (3 mg, 0.040 mmol) in DCM (1.0 mL) was stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C22H24F3N4O (M+H)+: m/z=417.2. found: 417.2.


Example 21. N-Methyl-4-phenyl-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidine-1-carboxamide



embedded image


Step 1. N-((4-Phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine



embedded image


A mixture of 5-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine (80 mg, 0.36 mmol), tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate (100 mg, 0.36 mmol), Ruphos Pd G4 (30 mg, 0.036 mmol) and KOt-Bu (120 mg, 1.07 mmol) in 1,4-dioxane (2 mL) was heated at 100° C. for 2 h. The resulting mixture was allowed to cool to r.t. before TFA (5 mL) was added and the mixture was stirred for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with DCM (30 mL) and washed with sat. NaHCO3 (aq.) and dried over Na2SO4. The organic layers were filtered and concentrated to afford the crude sub-title compound, which was used in next step without further purification. LCMS calc. for C20H22F3N4 (M+H)+: m/z=375.1. found: 375.1.


Step 2. N-Methyl-4-phenyl-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidine-1-carboxamide

A mixture of N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine (10 mg, 0.027 mmol), DIPEA (0.014 mL, 0.08 mmol) and methylcarbamic chloride (4 mg, 0.040 mmol) in DCM (1.0 mL) was stirred for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C22H25F3N5O (M+H)+: m/z=432.2. found: 432.2.


Example 22. 4-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxamide



embedded image


Step 1. 2-Bromo-5,7-dichloropyrazolo[1,5-a]pyrimidine



embedded image


A mixture of 3-bromo-1H-pyrazol-5-amine (3 g, 18.52 mmol) and sodium ethoxide (3.8 g, 55.6 mmol) in diethyl malonate (30 mL, 197 mmol) was heated at 120° C. After 20 h, the mixture was allowed to cool to r.t. and the precipitated solid was filtered, washed with Et2O and air-dried. The obtained solid was then taken up in phenylphosphonic dichloride (25 mL, 178 mmol) and heated at 120° C. After 20 h, the mixture was allowed to cool to r.t. and slowly quenched with sat. aq. NaHCO3 in an ice bath until pH=7. The mixture was then diluted with DCM and the organic layer was extracted, washed with brine and concentrated. The crude product was taken up in Et2O and the precipitated solid was filtered. The filtrate containing the product was concentrated and purified by FCC (EtOAc/Hexanes) to obtain 2-bromo-5,7-dichloropyrazolo[1,5-a]pyrimidine (2.4 g, 8.95 mmol, 48.4% yield) as a yellow solid. LCMS calc. for C6H3BrCl2N3 (M+H)+: m/z=267.9. found: 267.9.


Step 2. 2-Bromo-5,7-diiodopyrazolo[1,5-a]pyrimidine



embedded image


To a suspension of 2-bromo-5,7-dichloropyrazolo[1,5-a]pyrimidine (1.00 g, 3.75 mmol) and sodium iodide (5.62 g, 37.5 mmol) in MeCN (25 mL), acetyl chloride (0.346 mL, 4.87 mmol) was added, and the mixture was stirred at 80° C. After 3 h, the mixture was allowed to cool to r.t., then quenched with sat. aq. K2CO3, aq. Na2SO3 and aq. Na2S2O3. The mixture was then diluted with DCM and the organic layer was extracted, washed with brine, dried with Na2SO4, filtered and concentrated. The obtained solid was used directly in the next step. LCMS calc. for C6H3BrI2N3 (M+H)+: m/z=449.8. found: 449.7.


Step 3. tert-Butyl 4-cyano-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate



embedded image


To a solution of tert-butyl 4-cyanopiperidine-1-carboxylate (1.70 g, 8.08 mmol) and 2,5-difluoropyridine (1.14 g, 9.87 mmol) in THE (40 mL) at −78° C., LiHMDS (11.3 mL, 11.3 mmol, 1.0 M in THF) was added, and the resulting solution was stirred at −78° C. for 1 h. The cold bath was then removed and reaction mixture was stirred at r.t. for 20 h. The reaction was then quenched with sat. aq. NH4Cl, diluted with DCM and the organic layer was extracted, washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by FCC (EtOAc/Hexanes) to obtain the sub-title compound (1.48 g, 4.85 mmol, 60% yield) as colorless solid. LCMS calc. for C12H13FN3O2(M-C4H7)+: m/z=250.1. found: 250.2.


Step 4. tert-Butyl 4-(aminomethyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate



embedded image


To a solution of tert-butyl 4-cyano-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate (0.50 g, 1.64 mmol) and nickel(II) chloride hexahydrate (0.389 g, 1.64 mmol) in MeOH (10 mL), NaBH4 (0.310 g, 8.19 mmol) was added portion wise. The resulting solution was stirred at r.t. for 1.5 h. The reaction was then quenched with aq. K2CO3, the mixture was diluted with DCM and the organic layer was separated, washed with brine, dried with Na2SO4, filtered and concentrated. The residue was taken up in MeOH (10 mL) and stirred with SiliaMetS® thiol metal scavenger for 30 min. The mixture was then filtered and concentrated. The product obtained was used directly in the next step. LCMS calc. for C16H25FN3O2 (M+H)+: m/z=310.2. found: 310.2.


Step 5. tert-Butyl 4-(((2-bromo-5-iodopyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate



embedded image


A solution of 2-bromo-5,7-diiodopyrazolo[1,5-a]pyrimidine (1.18 g, 2.62 mmol), tert-butyl 4-(aminomethyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate (770 mg, 2.49 mmol) and DIPEA (1.30 mL, 7.47 mmol) in DMSO (12.4 mL) was heated at 100° C. for 2 h. The reaction mixture was allowed to cool to r.t. and water was added. The precipitated solid was filtered, washed with water and dried to obtain the sub-title compound as brown solid (1.43 g, 2.27 mmol, 91% yield). LCMS calc. for C22H26BrFIN6O2 (M+H)+: m/z=631.0. found: 631.1.


Step 6. tert-Butyl 4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate



embedded image


A solution of tert-butyl 4-(((2-bromo-5-iodopyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate (1.43 g, 2.27 mmol), copper(II) chloride (0.046 g, 0.340 mmol) and methyl 2,2-difluoro-2-(fluorosulfonyl)acetate (0.870 mL, 6.80 mmol) in DMF (11.3 mL) was heated at 110° C. for 5 h. The mixture was allowed to cool to r.t., diluted with EtOAc and water. The organic layer was separated, washed with brine, dried with Na2SO4, filtered, and concentrated. The residue was purified by FCC (EtOAc/hexanes) to obtain the sub-title compound as yellow oil (790 mg, 1.38 mmol, 60.8% yield). LCMS calc. for C23H26BrF4N6O2 (M+H)+: m/z=573.1. found: 573.1.


Step 7. 2-Bromo-N-((4-(5-fluoropyridin-2-yl)piperidin-4-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


tert-Butyl 4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate (790 mg, 1.38 mmol) was taken up in 10 mL DCM and TFA (5 mL) and stirred at r.t. for 1 h. The solvent was then removed to obtain the sub-title compound as yellow oil. LCMS calc. for C18H18BrF4N6 (M+H)+m/z=473.1. found: 473.1.


Step 8. 4-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxamide

A solution of 2-bromo-N-((4-(5-fluoropyridin-2-yl)piperidin-4-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (15 mg, 0.032 mmol), phenyl carbamate (9 mg, 0.063 mmol) and DIPEA (0.055 mL, 0.317 mmol) in 1,4-dioxane (1 mL) was heated at 85° C. for 20 h. The reaction mixture was allowed to cool to r.t. and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to obtain the title compound. LCMS calc. for C19H19BrF4N7O (M+H)+: m/z=516.1. found: 516.1. 1H NMR (500 MHz, DMSO-d6) δ 8.74 (t, J=6.9 Hz, 1H), 8.48 (d, J=2.7 Hz, 1H), 7.56-7.47 (m, 2H), 6.77 (s, 1H), 5.55 (s, 1H), 3.77-3.70 (m, 2H), 3.63 (d, J=6.8 Hz, 2H), 2.74-2.64 (m, 2H), 2.43-2.36 (m, 2H), 1.81-1.71 (m, 2H).


Example 23. 2-(4-(5-Fluoropyridin-2-yl)-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidin-1-yl)ethan-1-ol



embedded image


Step 1. N-((4-(5-fluoropyridin-2-yl)piperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine



embedded image


A mixture of tert-butyl 4-(aminomethyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate (200 mg, 0.646 mmol), 5-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine (214 mg, 0.970 mmol), Ruphos Pd G4 (82 mg, 0.097 mmol), and NaOt-Bu (124 mg, 1.29 mmol) in 1,4-dioxane (3 mL) was heated at 85° C. for 20 h. The reaction mixture was then allowed to cool to r.t., diluted with EtOAc, filtered through diatomaceous earth and concentrated. The residue was purified by FCC (EtOAc/Hex) to obtain tert-butyl 4-(5-fluoropyridin-2-yl)-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidine-1-carboxylate (176 mg, 0.357 mmol, 55.2% yield). The product was taken up in 4 mL DCM and 2 mL TFA was added. The solution was stirred at r.t. for 1 h. The solvent was then removed and the product obtained was used directly in the next step. LCMS calc. for C19H20F4N5 (M+H)+: m/z=394.2. found: 394.2.


Step 2. 2-(4-(5-Fluoropyridin-2-yl)-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidin-1-yl)ethan-1-ol

A mixture of N-((4-(5-fluoropyridin-2-yl)piperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine (15 mg, 0.038 mmol), 2-((tert-butyldimethylsilyl)oxy)acetaldehyde (20 mg, 0.114 mmol) and sodium triacetoxyborohydride (16 mg, 0.076 mmol) in DCM (1 mL) was stirred at r.t. for 20 h. TFA (1 mL) was then added and the resulting solution was stirred at 50° C. for 2 h. The reaction mixture was purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to obtain the title compound. LCMS calc. for C21H24F4N5O (M+H)+: m/z=438.2. found: 438.3.


Example 24. 3-(4-(5-Fluoropyridin-2-yl)-4-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)piperidin-1-yl)-3-oxopropanenitrile



embedded image


A solution of N-((4-(5-fluoropyridin-2-yl)piperidin-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine (10 mg, 0.025 mmol), 2-cyanoacetic acid (4.3 mg, 0.051 mmol), HATU (14.5 mg, 0.038 mmol) and DIPEA (0.013 mL, 0.076 mmol) in DMF (1 mL) was stirred at r.t. for 1 h. The reaction mixture was purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to obtain the title compound. LCMS calc. for C22H21F4N6O (M+H)+: m/z=461.2. found: 461.3. 1H NMR (500 MHz, DMSO-d6) δ 8.56-8.51 (m, 2H), 7.60-7.55 (m, 2H), 7.15 (t, J=8.3 Hz, 1H), 6.83 (d, J=8.8 Hz, 1H), 6.71 (t, J=6.5 Hz, 1H), 5.69 (d, J=7.8 Hz, 1H), 4.13-3.91 (m, 3H), 3.61-3.47 (m, 3H), 2.98-2.89 (m, 1H), 2.74-2.64 (m, 1H), 2.54-2.44 (m, 2H), 1.94-1.76 (m, 2H).


Example 25. 3-(((7-Chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)-N-methylazetidine-1-carboxamide



embedded image


Step 1. 5-Bromo-7-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine



embedded image


A mixture of 3-bromo-1,1,1-trifluoropropan-2-one (230 mg, 1.2 mmol), 6-bromo-4-chloropyridin-2-amine (250 mg, 1.2 mmol), and butan-1-ol (3 mL) in a sealed vial was heated at 100° C. for 4 h. Upon completion, the reaction mixture was allowed to cool to r.t. and washed with DCM and water. The organic layers were combined and concentrated in vacuo. This residue was purified by FCC (hexanes/EtOAc, 0-100% EtOAc) to provide the sub-title compound. LCMS calc. for C8H4BrClF3N2 (M+H)+: m/z=298.9, 300.9. found: 298.9, 300.9.


Step 2. tert-Butyl 3-(((7-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate



embedded image


A vial containing 5-bromo-7-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine (213 mg, 0.7 mmol), tert-butyl 3-(aminomethyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate (200 mg, 0.7 mmol), NaOt-Bu (205 mg, 2.1 mmol), and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (52 mg, 0.07 mmol) was evacuated and backfilled with nitrogen. Subsequently 1,4-dioxane (2 mL) was added and the reaction mixture was heated at 100° C. for 30 min. Upon completion, the mixture was diluted with DCM and filtered through a plug of diatomaceous earth. The filtrate was concentrated in vacuo and purified by FCC (hexanes/EtOAc, 0-100% EtOAc) to provide the sub-title compound. LCMS calc. for C22H23ClF4N5O2 (M+H)+: m/z=500.2. found: 500.2.


Step 3. 7-Chloro-N-((3-(5-fluoropyridin-2-yl)azetidin-3-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine



embedded image


To a solution containing tert-butyl 3-(((7-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate (20 mg, 0.04 mmol) in DCM (400 μL), TFA (100 μL, 1.3 mmol) was added and the mixture was stirred at r.t. for 1 h. Upon completion, the reaction mixture was concentrated in vacuo. The resulting crude product was used in the next step without further purification. LCMS calc. for C17H15ClF4N5 (M+H)+: m/z=400.1. found: 400.1.


Step 4. 3-(((7-Chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)-N-methylazetidine-1-carboxamide

To a solution of 7-chloro-N-((3-(5-fluoropyridin-2-yl)azetidin-3-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine (10 mg, 0.025 mmol), and DIPEA (22 μL, 0.125 mmol), in MeCN (250 μL) methylcarbamic chloride (12 mg, 0.125 mmol) was added and the resulting solution was stirred at r.t. for 10 min. Upon reaction completion, the reaction mixture was diluted with MeCN and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to give the title compound. LCMS calc. for C19H18ClF4N6O (M+H)+: m/z=457.1. found: 457.1.


Example 26. N-Methyl-4-(((2-(perfluoroethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide



embedded image


Step 1. 4-(Aminomethyl)-N-methyl-4-phenylpiperidine-1-carboxamide



embedded image


A solution containing tert-butyl ((4-phenylpiperidin-4-yl)methyl)carbamate (700 mg, 2.4 mmol) and DIPEA (850 μL, 4.8 mmol) in MeCN (6 mL) was cooled to 0° C. and methylcarbamic chloride (450 mg, 4.8 mmol) was added. The resulting mixture was allowed to warm to r.t. and stirred for 30 min. Upon completion, ice cold water was added and the resulting precipitate was collected by filtration. The collected solid was then taken up in HCl (6 mL, 24 mmol, 4 M in 1,4-dioxane) and stirred at r.t. for 1 h. The resulting mixture was concentrated in vacuo, the residue was then neutralized with sat, aq. NaHCO3 and subsequently concentrated again to dryness. The resulting solid was suspended in 20 mL of DCM and 5 mL of MeOH. This suspension was filtered and the filtrate was then concentrated in vacuo and used directly for the next step. LCMS calc. for C14H22N3O (M+H)+: m/z=248.2. found: 248.2.


Step 2. 5-Bromo-2-(perfluoroethyl)imidazo[1,2-a]pyridine



embedded image


A mixture of 1-bromo-3,3,4,4,4-pentafluorobutan-2-one (348 mg, 1.5 mmol), 6-bromopyridin-2-amine (250 mg, 1.5 mmol), and butan-1-ol (3 mL) in a sealed vial was stirred at 100° C. for 16 h. Upon completion, the reaction mixture was concentrated in vacuo and the resulting crude product was used directly for the next step. LCMS calc. for C9H5BrF5N2 (M+H)+: m/z=315.0, 317.0. found: 314.9, 316.9.


Step 2. N-Methyl-4-(((2-(perfluoroethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide

A vial containing 5-bromo-2-(perfluoroethyl)imidazo[1,2-a]pyridine (13 mg, 0.04 mmol), 4-(aminomethyl)-N-methyl-4-phenylpiperidine-1-carboxamide (10 mg, 0.04 mmol), NaOt-Bu (12 mg, 0.12 mmol), and RuPhos Pd G4 (4 mg, 4 μmol) was evacuated and backfilled with nitrogen. 1,4-Dioxane (0.5 mL) was then added and the reaction mixture was heated at 80° C. for 2 h. Upon completion, the solution was diluted with MeCN, filtered through a Silicycle SiliaPrep® Thiol cartridge (Cat. #SPE-R51030B), and the product was purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to give the title compound. LCMS calc. for C23H25F5N5O (M+H)+: m/z=482.2. found: 482.3.


Example 27. 4-(((7-Chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-N-methyl-4-phenylpiperidine-1-carboxamide



embedded image


The title compound was prepared according to the procedures described in Example 26, with 5-bromo-7-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine (Example 25, Step 1) replacing 5-bromo-2-(perfluoroethyl)imidazo[1,2-a]pyridine in Step 2. LCMS calc. for C22H24ClF3N5O (M+H)+: m/z=466.2. found: 466.1.


Example 28. N-Methyl-4-(((7-methyl-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)-4-phenylpiperidine-1-carboxamide



embedded image


The title compound was prepared according to the procedures described in Example 25, with 6-bromo-4-methylpyridin-2-amine replacing 6-bromo-4-chloropyridin-2-amine in Step 1, and tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate replacing tert-butyl 3-(aminomethyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate in Step 2. LCMS calc. for C23H27F3N5O (M+H)+: m/z=446.2. found: 446.2.


Example 29. 4-(((2-Isopropylimidazo[1,2-a]pyridin-5-yl)amino)methyl)-N-methyl-4-phenylpiperidine-1-carboxamide



embedded image


The title compound was prepared according to the procedures described in Example 25, with 1-bromo-3-methylbutan-2-one replacing 3-bromo-1,1,1-trifluoropropan-2-one and 6-bromopyridin-2-amine replacing 6-bromo-4-chloropyridin-2-amine in Step 1. Additionally tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate replaced tert-butyl 3-(aminomethyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate in Step 2. LCMS calc. for C24H32N5O (M+H)+: m/z=406.3. found: 406.3.


Example 30. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-sulfonamide



embedded image


Step 1. tert-Butyl 3-(4-fluorophenyl)-3-(nitromethyl)azetidine-1-carboxylate



embedded image


Aq. KOH (3.1 mL 1.5 M, 4.67 mmol) was added to a solution of chloro(1,5-cyclooctadiene)rhodium(I) dimer (92 mg, 0.187 mmol) in 1,4-dioxane (20 mL) and the resulting mixture was stirred for 5 min. tert-Butyl 3-(nitromethylene)azetidine-1-carboxylate (1.0 g, 4.67 mmol), and (4-fluorophenyl)boronic acid (1.3 g, 9.34 mmol) were added. The mixture was then stirred at r.t. for 12 h. Upon completion, the reaction mixture was quenched with sat, aq. NH4Cl (20 mL) and diluted with EtOAc (100 mL). The organic layer was separated and dried with MgSO4 and concentrated in vacuo. The crude product obtained was used in the next step without further purification. LCMS calc. for C15H20FN2O4 (M+H)+: m/z=311.1. found: 255.1.


Step 2. tert-Butyl 3-(aminomethyl)-3-(4-fluorophenyl)azetidine-1-carboxylate



embedded image


To a solution of tert-butyl 3-(4-fluorophenyl)-3-(nitromethyl)azetidine-1-carboxylate (4.66 mmol) in MeCN (20 mL) and MeOH (5 mL), 4,4′-bipyridine (73 mg, 0.47 mmol) and hypodiboric acid (1.25 g, 14.0 mmol) were added at r.t. The resulting mixture was stirred for 15 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was dissolved in DCM and washed with aq. NaHCO3. The organic layer was concentrated in vacuo. The crude product (1.3 g) obtained was used in the next step without further purification. LCMS calc. for C15H22FN2O2 (M+H)+: m/z=281.2. found: 225.2.


Step 3. N-((3-(4-Fluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


To a solution of 2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol (50 mg, 0.18 mmol) in DCM (1 mL) at 0° C., DIPEA (0.064 mL, 0.37 mmol) and Tf2O (0.22 mL 1 M in DCM, 0.22 mmol) were added. After 15 min., the mixture was transferred to a solution of tert-butyl 3-(aminomethyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (52 mg, 0.184 mmol) in DCM (1 mL) at 0° C. The resulting mixture was stirred at r.t. for 1 h before TFA (5 mL) was added and stirring at r.t. was continued for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C18H15F7N5 (M+H)+: m/z=434.1. found: 434.1.


Step 4. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-sulfonamide

A mixture of N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (5 mg, 0.012 mmol), DIPEA (0.04 mL, 0.23 mmol) and sulfuric diamide (5.5 mg, 0.058 mmol) in 1,4-dioxane (0.5 mL) was heated with stirring at 115° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C18H16F7N6O2S (M+H)+: m/z=513.1. found: 513.1.


Example 31. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide



embedded image


A mixture of N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (example 30, step 3, 5 mg, 0.012 mmol), TEA (0.032 mL, 0.23 mmol) and phenyl carbamate (8 mg, 0.058 mmol) in 1,4-dioxane (0.5 mL) was heated with stirring at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C19H16F7N6O (M+H)+: m/z=477.1. found: 477.1.


Example 32. 3-(4-Fluorophenyl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-sulfonamide



embedded image


Step 1. N-((3-(4-Fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine



embedded image


A mixture of 5-chloro-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridine (50 mg, 0.226 mmol), tert-butyl 3-(aminomethyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (64 mg, 0.226 mmol) and DIPEA (0.39 mL, 2.26 mmol) in DMSO (1 mL) was heated at 125° C. overnight. The resulting mixture was allowed to cool to r.t. before TFA (5 mL) was added and the mixture was stirred for 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C17H16F4N5 (M+H)+: m/z=366.1. found: 366.1.


Step 2. 3-(4-Fluorophenyl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-sulfonamide

A mixture of N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine (5 mg, 0.012 mmol), DIPEA (0.04 mL, 0.23 mmol) and sulfuric diamide (6 mg, 0.058 mmol) in 1,4-dioxane (0.5 mL) was heated with stirring at 115° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C17H17F4N6O2S (M+H)+: m/z=445.1. found: 445.1.


Example 33. 3-(4-Fluorophenyl)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide



embedded image


A mixture of N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine (example 32, step 1, 5 mg, 0.012 mmol), TEA (0.032 mL, 0.23 mmol) and phenyl carbamate (8 mg, 0.058 mmol) in 1,4-dioxane (0.5 mL) was heated with stirring at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C18H17F4N6O (M+H)+: m/z=409.1. found: 409.1.


Example 34. 3-(4-Fluorophenyl)-3-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)azetidine-1-sulfonamide



embedded image


Step 1. N-((3-(4-Fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine



embedded image


A mixture of 5-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine (80 mg, 0.36 mmol), tert-butyl 3-(aminomethyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (100 mg, 0.36 mmol), Ruphos Pd G4 (30 mg, 0.036 mmol) and KOt-Bu (120 mg, 1.07 mmol) in 1,4-dioxane (2 mL) was heated at 100° C. for 2 h. The resulting mixture was allowed to cool to r.t. before TFA (5 mL) was added and stirring was continued 10 min. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the sub-title compound. LCMS calc. for C18H17F4N4 (M+H)+: m/z=365.1. found: 365.1.


Step 2. 3-(4-Fluorophenyl)-3-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)azetidine-1-sulfonamide

A mixture of N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine (5 mg, 0.012 mmol), DIPEA (0.04 mL, 0.23 mmol) and sulfuric diamide (6 mg, 0.058 mmol) in 1,4-dioxane (0.5 mL) was heated with stirring at 115° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C18H18F4N5O2S (M+H)+: m/z=444.1. found: 444.1.


Example 35. 3-(4-Fluorophenyl)-3-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide



embedded image


A mixture of N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine (example 34, step 1, 5 mg, 0.012 mmol), TEA (0.032 mL, 0.23 mmol) and phenyl carbamate (8 mg, 0.058 mmol) in 1,4-dioxane (0.5 mL) was heated with stirring at 85° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound.


LCMS calc. for C19H18F4N5O (M+H)+: m/z=408.1. found: 408.1.


Example 36. N-((1-(4-Fluorophenyl)cyclobutyl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine



embedded image


A mixture of 5-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine (27 mg, 0.12 mmol), (1-(4-fluorophenyl)cyclobutyl)methanamine (33 mg, 0.2 mmol), and DIPEA (0.21 mL, 1.2 mmol) in DMSO (1 mL) was heated at 125° C. overnight. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C19H18F4N3 (M+H)+: m/z=364.1. found: 364.1.


Example 37. N-((1-(Pyridin-4-yl)cyclobutyl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine



embedded image


A mixture of 5-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine (27 mg, 0.12 mmol), (1-(pyridin-4-yl)cyclobutyl)methanamine (32 mg, 0.2 mmol), and DIPEA (0.21 mL, 1.2 mmol) in DMSO (1 mL) was heated at 125° C. overnight. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C18H18F3N4 (M+H)+: m/z=347.1. found: 347.1.


Example 38. N-((1-(4-Bromophenyl)cyclobutyl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine



embedded image


A mixture of 5-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine (27 mg, 0.12 mmol), (1-(4-bromophenyl)cyclobutyl)methanamine (48 mg, 0.2 mmol), and DIPEA (0.21 mL, 1.2 mmol) in DMSO (1 mL) was heated at 125° C. overnight. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C19H18BrF3N3 (M+H)+: m/z=424.1. found: 424.1.


Example 39. N-((4-(4-Methoxyphenyl)tetrahydro-2H-pyran-4-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine



embedded image


A mixture of 5-chloro-2-(trifluoromethyl)imidazo[1,2-a]pyridine (22 mg, 0.1 mmol), (4-(4-methoxyphenyl)tetrahydro-2H-pyran-4-yl)methanamine (44 mg, 0.2 mmol), and DIPEA (0.21 mL, 1.2 mmol) in DMSO (1 mL) was heated at 125° C. overnight. Upon completion, the reaction mixture was diluted with MeOH and purified by prep. LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at a flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C21H23F3N3O2 (M+H)+: m/z=406.2. found: 406.2.


Example 40. 6-(4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidin-1-yl)-2-methylpyridazin-3(2H)-one



embedded image


Step 1. 2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol



embedded image


A mixture of 5 3-(trifluoromethyl)-1H-pyrazol-5-amine (2.0 g, 13.24 mmol), ethyl 4,4,4-trifluoro-3-oxobutanoate (2.4 g, 13.24 mmol) and TFA (1.02 mL, 12.2 mmol) in 15 mL DMSO was stirred at r.t. overnight. The resulting mixture was poured into water while stirring at 0° C. to afford the sub-title compound as a white solid which was filtered and dried. LCMS calc. for C8H4F6N3O (M+H)+: m/z=272. found: 272.


Step 2. 7-Chloro-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidine



embedded image


A mixture of 2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol (1.5 g, 5.5 mmol), POCl3 (5.2 mL, 55 mmol) and DIPEA (1 mL, 55 mmol) in toluene (10 mL) was stirred at 100° C. for 3 h. After the full conversion, the mixture was allowed to cool to r.t. and toluene was removed in vacuo. The resulting solution was poured into ice while stirring to afford the sub-title compound as a white solid, which was filtered and washed with water, cold hexane and dried. LCMS calc. for C8H3ClF6N3 (M+H)+: m/z=290.0. found: 290.0.


Step 3. N-((4-(4-Fluorophenyl)piperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


A mixture of 7-chloro-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (50 mg, 0.17 mmol), tert-butyl 4-(aminomethyl)-4-(4-fluorophenyl)piperidine-1-carboxylate (53 mg, 0.17 mmol) and DIPEA (0.09 mL, 0.51 mmol) in dioxane (5 mL) was stirred at 100° C. for 1 h. After complete conversion, the mixture was allowed to cool to r.t. and TFA 2 mL was added and the mixture was stirred for 1 h at r.t. Then the mixture was diluted with MeOH and purified by prep. HPLC to afford the sub-title compound. LCMS calc. for C20H19F7N5 (M+H)+: m/z=426.2. found: 426.2.


Step 4. 6-(4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidin-1-yl)-2-methylpyridazin-3(2H)-one

A mixture of N-((4-(4-fluorophenyl)piperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (10 mg, 0.022 mmol), 6-chloro-2-methylpyridazin-3(2H)-one (6 mg, 0.043 mmol) and DIPEA (0.011 mL, 0.065 mmol) in NMP 1.0 mL was stirred at 120° C. for 3 h. The mixture was allowed to cool to r.t., diluted with MeOH and purified by prep. HPLC to afford the title compound as a racemic mixture. LCMS calc. for C25H23F7N7O (M+H)+: m/z=570.2. found: 570.2.


Example 41. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluoro-1H-pyrazol-1-yl)-N-methylazetidine-1-carboxamide



embedded image


A mixture of N-((3-(4-fluoro-1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (example 10, step 2, 10 mg, 0.024 mmol), DIPEA (0.012 mL, 0.071 mmol) and methylcarbamic chloride (3.3 mg, 0.035 mmol) in 1.0 mL DCM was stirred for 30 mins at r.t. Upon completion, the reaction mixture was diluted with MeOH and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C17H16F7N8O (M+H)+: m/z=481.1. found: 481.1.


Example 42. N-((1-(5-Amino-1H-1,2,4-triazol-3-yl)-4-phenylpiperidin-4-yl)methyl)-5-(difluoromethyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


Step 1. 5-(Difluoromethyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol



embedded image


A mixture of 5 3-(trifluoromethyl)-1H-pyrazol-5-amine (2.0 g, 13.24 mmol), ethyl 4,4-difluoro-3-hydroxybut-2-enoate (2.2 g, 13 mmol) and TFA (1.0 mL, 12 mmol) in 15 mL DMSO was stirred at r.t. overnight. The resulting mixture was poured into water while stirring at 0° C. to afford the sub-title compound as a white solid which was filtered and dried. LCMS calc. for C8H5F5N3O (M+H)+: m/z=254.0 found: 254.0.


Step 2. 7-Chloro-5-(difluoromethyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine



embedded image


A mixture of 5-(difluoromethyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol (1.5 g, 5.9 mmol), POCl3 (5.5 mL, 59 mmol) and DIPEA (1.0 mL, 5.9 mmol) in toluene (10 mL) was stirred at 100° C. for 3 h. After full conversion, the mixture was allowed to cool to r.t. and toluene was removed in vacuo. The resulting solution was poured into ice while stirring to afford the sub-title compound as a white solid which was filtered, washed with water, cold hexane and dried. LCMS calc. for C8H4ClF5N3 (M+H)+: m/z=272.0. found: 272.0.


Step 3. 5-(Difluoromethyl)-N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


A mixture of 7-chloro-5-(difluoromethyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (50 mg, 0.18 mmol), tert-butyl 4-(aminomethyl)-4-phenylpiperidine-1-carboxylate (53 mg, 0.18 mmol) and DIPEA (0.096 mL, 0.55 mmol) in dioxane (5 mL) was stirred at 100° C. for 1 h. After complete conversion, the mixture was allowed to cool to r.t., TFA 2 mL was added and the reaction mixture was stirred for 1 h at r.t. Then the mixture was diluted with MeOH and purified by prep. HPLC to afford the sub-title compound. LCMS calc. for C20H21F5N5 (M+H)+: m/z=426.2. found: 426.2.


Step 4. N-((1-(5-Amino-1H-1,2,4-triazol-3-yl)-4-phenylpiperidin-4-yl)methyl)-5-(difluoromethyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine

5-(Difluoromethyl)-N-((4-phenylpiperidin-4-yl)methyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (10 mg, 0.024 mmol), diphenyl cyanocarbonimidate (6.5 mg, 0.027 mmol) and DIPEA (11 μL, 0.065 mmol) in 1,4-dioxane (2 mL) stirred at 100° C. for 0.5 h. Then, the mixture was allowed to cool to r.t. and 1 M solution of hydrazine (0.068 mL, 0.068 mmol) in EtOH was added. The mixture was stirred at 60° C. for 30 min. Upon completion, the reaction mixture was diluted with MeOH and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C22H23F5N9 (M+H)+: m/z=508.2. found: 508.2.


Example 43. 2-Bromo-N-((3-(4-methylpiperazin-1-yl)-1-phenylcyclobutyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


Step 1. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutan-1-ol



embedded image


A solution of 2-bromo-7-chloro-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (290 mg, 0.95 mmol) (see Example 60 for preparation), commercially available 3-(aminomethyl)-3-phenylcyclobutan-1-ol (170 mg, 0.95 mmol) and DIPEA (0.84 mL, 4.7 mmol) in DMSO (5 mL) was stirred at 100° C. for 2 h. After cooling to r.t., the mixture was diluted with water (100 mL) and extracted with EtOAc (2×100 mL). The combined organic phases were dried over Na2SO4 and concentrated to give a crude oil which was used directly in the next step. LC-MS calc. for C18H17BrF3N4O (M+H)+: m/z=441.1. found 441.1.


Step 2. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutan-1-one



embedded image


A solution of 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutan-1-ol (400 mg, 0.907 mmol), NaHCO3 (762 mg, 9.07 mmol) and Dess-Martin periodinane (577 mg, 1.36 mmol) in DCM (5 mL) was stirred at 23° C. for 1 h. The mixture was then diluted with more DCM and washed with sat. aq. Na2S2O3 and brine. The organic phase was dried over Na2SO4, concentrated, and purified by FCC (BIOTAGE® ISOLERA™, pure hexane to 20% EtOAc) to give the sub-title compound as a white solid. LC-MS calc. for C18H15BrF3N4O (M+H)+: m/z=439.0. found 439.1.


Step 3. 2-Bromo-N-((3-(4-methylpiperazin-1-yl)-1-phenylcyclobutyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine

To a stirred solution of 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutan-1-one (400 mg, 0.911 mmol) and 1-methylpiperazine (365 mg, 3.64 mmol) in MeOH (10 mL), sodium cyanoborohydride (172 mg, 2.73 mmol) was added. The mixture was stirred at r.t. overnight, and then was diluted with MeCN and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% NH4OH, at flow rate of 60 mL/min., gradient of 50-65% MeCN over 5 min.). The two diastereomers could be separated, with the more potent diastereomer as an inhibitor of MRGPRX2 eluting first (at 3.8 min.). LC-MS calc. for C23H27BrF3N6 (M+H)+: m/z=523.1. found 523.1. 1H NMR (500 MHz, DMSO-d6) δ 8.76 (t, J=6.8 Hz, 1H), 7.14-7.09 (m, 2H), 7.08-7.02 (m, 3H), 6.73 (s, 1H), 5.52 (s, 1H), 3.79 (d, J=6.6 Hz, 2H), 2.89 (p, J=7.7 Hz, 1H), 2.74-2.67 (m, 2H), 2.45-2.15 (m, 8H), 2.13 (s, 3H), 1.98 (td, J=8.5, 2.7 Hz, 2H).


The diastereomers of the title compound that are separable by chromatography are: 2-bromo-N-(((1s,3s)-3-(4-methylpiperazin-1-yl)-1-phenylcyclobutyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine and 2-bromo-N-(((1r,3r)-3-(4-methylpiperazin-1-yl)-1-phenylcyclobutyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine.


Example 44. 2-((3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutyl)amino)ethan-1-ol



embedded image


The title compound was prepared according to the procedure described in Example 43 (Step 3), using ethanolamine instead of 1-methylpiperazine. Diluted crude mixture with MeCN and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% NH4OH, at flow rate of 60 mL/min., gradient of 41.3-59.3% MeCN in 12 min.). The two diastereomers of the title compound could be separated by chromatography, with the diasteroisomer that is more potent as an inhibitor of MRGPRX2 eluting first (at 5 min.). LC-MS calc. for C20H22BrF3N5O (M+H)+: m/z=484.1. found 484.2.


The diastereomers of the title compound that are separable by chromatography are: 2-(((1s,3s)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutyl)amino)ethan-1-ol and 2-(((1r,3r)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutyl)amino)ethan-1-ol.


Example 45. 5-(3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidin-1-yl)pyrazine-2-carbonitrile



embedded image


A mixture of N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (10 mg, 0.023 mmol), 5-chloropyrazine-2-carbonitrile (6.4 mg, 0.046 mmol) and DIPEA (0.020 mL, 0.12 mmol) in DMSO (0.2 mL) was stirred at 120° C. for 2 h. Upon completion, the reaction mixture was diluted with MeOH and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C23H16F7N8 (M+H)+: m/z=537.1. found: 537.1.


Example 46. N-((3-(4-Fluorophenyl)-1-(1-methyl-1H-1,2,3-triazol-4-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


A vial containing N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (10 mg, 0.023 mmol), 4-bromo-1-methyl-1H-1,2,3-triazole (3.7 mg, 0.023 mmol) and sodium trimethylsilanolate (5.2 mg, 0.046 mmol) was degassed and added GPhos Pd G6 TES (2.2 mg, 2.31 μmol) and THE (0.5 mL). The reaction mixture was heated at 85 C for 16 h. Upon completion, the reaction mixture was diluted with MeOH and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C21H18F7N8 (M+H)+: m/z=515.2. found: 515.2. 1H NMR (600 MHz, DMSO-d6) δ 9.22 (t, J=6.8 Hz, 1H), 7.38 (s, 1H), 7.23-7.17 (m, 2H), 7.13 (s, 1H), 7.02-6.95 (m, 2H), 5.89 (s, 1H), 4.35-4.31 (m, 2H), 4.14 (d, J=6.8 Hz, 2H), 4.95-4.92 (m, 2H), 3.91 (s, 3H) ppm.


Example 47. 6-(3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidin-1-yl)picolinamide



embedded image


The title compound was prepared according to the procedure described in Example 45, using 6-fluoropicolinamide instead of 5-chloropyrazine-2-carbonitrile as starting material.


LCMS calc. for C24H19F7N7O (M+H)+: m/z=554.2. found: 554.2.


Example 48. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide



embedded image


Step 1. 4-Nitrophenyl 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate



embedded image


To a solution of N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (4.00 g, 9.23 mmol) and Et3N (5.15 mL, 36.9 mmol) in DCM (80 mL) was added 4-nitrophenyl carbonochloridate (2.79 g, 13.9 mmol) portionwise and the resulting solution was stirred at r.t. for 1 h. The mixture was diluted with water and extracted with DCM. The crude product was purified by FCC (EtOAc/Hex) to obtain the sub-title compound. LCMS calc. for C25H18F7N6O4 (M+H)+: m/z=599.2. found: 599.2.


Step 2. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide

A mixture of 4-nitrophenyl 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (20 mg, 0.033 mmol), (1r,4r)-4-aminocyclohexan-1-ol (15.4 mg, 0.134 mmol) and DIPEA (0.047 mL, 0.27 mmol) in DMSO (0.05 mL) was stirred 130° C. for 1 h. Upon completion, the reaction mixture was diluted with MeOH and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.375% NH4OH, at flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C25H26F7N6O2 (M+H)+: m/z=575.2. found: 575.3. 1H NMR (500 MHz, DMSO-d6) δ 9.10 (s, 1H), 7.21-7.15 (m, 2H), 7.12 (s, 1H), 7.00 (t, J=8.6 Hz, 2H), 6.09 (d, J=8.0 Hz, 1H), 5.88 (s, 1H), 4.47 (d, J=4.4 Hz, 1H), 4.24 (d, J=8.2 Hz, 2H), 4.01-3.94 (m, 4H), 3.34-3.28 (m, 2H), 1.81-1.73 (m, 2H), 1.72-1.62 (m, 2H), 1.24-1.09 (m, 4H) ppm.


Example 49. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,3r)-3-hydroxycyclobutyl)azetidine-1-carboxamide



embedded image


The title compound was prepared according to the procedure described in Example 48, using (1r,3r)-3-aminocyclobutan-1-ol instead of (1r,4r)-4-aminocyclohexan-1-ol as starting material. LCMS calc. for C23H22F7N6O2 (M+H)+: m/z=547.2. found: 547.2. 1H NMR (500 MHz, DMSO) δ 9.12 (s, 1H), 7.21-7.15 (m, 2H), 7.13 (s, 1H), 7.00 (t, J=8.7 Hz, 2H), 6.54 (d, J=7.1 Hz, 1H), 5.85 (s, 1H), 4.87 (d, J=5.2 Hz, 1H), 4.28-4.23 (m, 2H), 4.23-4.18 (m, 1H), 4.16-4.05 (m, 1H), 4.03-3.96 (m, 4H), 2.14-2.06 (m, 2H), 2.06-1.98 (m, 2H) ppm.


Example 50. (R)-(4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidin-1-yl)(1-methylazetidin-2-yl)methanone



embedded image


To a mixture of N-((4-(4-fluorophenyl)piperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (10.0 mg, 0.022 mmol), (R)-1-methylazetidine-2-carboxylic acid (2.5 mg, 0.022 mmol) in DMF (0.1 mL) was added HATU (12.4 mg, 0.033 mmol) and DIPEA (0.015 mL, 0.087 mmol). The mixture was stirred at r.t. for 1 h. Upon completion, the reaction mixture was diluted with MeOH and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C25H26F7N6O (M+H)+: m/z=559.2. found: 559.2.


Example 51. 4-(3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidin-1-yl)-1-cyclopropylpyridin-2(1H)-one



embedded image


The title compound was prepared according to the procedure described in Example 46, using 4-bromo-1-cyclopropylpyridin-2(1H)-one instead of 4-bromo-1-methyl-1H-1,2,3-triazole as starting material. LCMS calc. for C26H22F7N6O (M+H)+: m/z=567.2. found: 567.2.


Example 52. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1s,3s)-3-hydroxy-3-methylcyclobutyl)azetidine-1-carboxamide



embedded image


The title compound was prepared according to the procedure described in Example 48, using (1s,3s)-3-amino-1-methylcyclobutan-1-ol instead of (1r,4r)-4-aminocyclohexan-1-ol as starting material. LCMS calc. for C24H24F7N6O2 (M+H)+: m/z=561.2. found: 561.2.


Example 53. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1R,3R)-3-hydroxycyclopentyl)azetidine-1-carboxamide



embedded image


The title compound was prepared according to the procedure described in Example 48, using (1R,3R)-3-aminocyclopentan-1-ol instead of (1r,4r)-4-aminocyclohexan-1-ol as starting material. LCMS calc. for C24H24F7N6O2 (M+H)+: m/z=561.2. found: 561.2. 1H NMR (600 MHz, DMSO-d6) δ 9.12 (s, 1H), 7.20-7.15 (m, 2H), 7.12 (s, 1H), 7.00 (t, J=8.6 Hz, 2H), 6.23 (d, J=7.7 Hz, 1H), 5.85 (s, 1H), 4.41 (d, J=3.7 Hz, 1H), 4.27-4.23 (m, 2H), 4.16-4.11 (m, 1H), 4.13-4.04 (m, 1H), 4.04-3.96 (m, 4H), 1.95-1.80 (m, 2H), 1.74-1.67 (m, 1H), 1.55-1.48 (m, 1H), 1.42-1.34 (m, 1H), 1.33-1.26 (m, 1H) ppm.


Example 54. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1s,4s)-4-hydroxy-4-(trifluoromethyl)cyclohexyl)azetidine-1-carboxamide



embedded image


The title compound was prepared according to the procedure described in Example 48, using (1s,4s)-4-amino-1-(trifluoromethyl)cyclohexan-1-ol instead of (1r,4r)-4-aminocyclohexan-1-ol as starting material. LCMS calc. for C26H25F10N6O2 (M+H)+: m/z=643.2. found: 643.2.


Example 55. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-(2-hydroxypropan-2-yl)cyclohexyl)azetidine-1-carboxamide



embedded image


The title compound was prepared according to the procedure described in Example 48, using 2-((1r,4r)-4-aminocyclohexyl)propan-2-ol instead of (1r,4r)-4-aminocyclohexan-1-ol as starting material. LCMS calc. for C28H32F7N6O2 (M+H)+: m/z=617.2. found: 617.2.


Example 56. 4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)-N-((1R,3R)-3-hydroxycyclopentyl)piperidine-1-carboxamide



embedded image


Step 1. 4-Nitrophenyl 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidine-1-carboxylate



embedded image


The sub-title compound was prepared according to the procedure described in Example 48 (Step 1), using N-((4-(4-fluorophenyl)piperidin-4-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine instead of N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine as starting material. LCMS calc. for C27H22F7N6O4 (M+H)+: m/z=627.2. found: 627.3.


Step 2. 4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)-N-((1R,3R)-3-hydroxycyclopentyl)piperidine-1-carboxamide

A mixture of 4-nitrophenyl 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidine-1-carboxylate (15 mg, 0.024 mmol), (1R,3R)-3-aminocyclopentan-1-ol (2.4 mg, 0.024 mmol) and DIPEA (0.013 mL, 0.072 mmol) in DMSO (0.3 mL) was heated to 90° C. overnight. Upon completion, the reaction mixture was diluted with MeOH and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.375% NH4OH, at flow rate of 60 mL/min.) to afford the title compound. LCMS calc. for C26H28F7N6O2 (M+H)+: m/z=589.2. found: 589.2.


Example 57. 3-(((2-Chloro-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide



embedded image


Step 1. 2-Chloro-5,7-diiodopyrazolo[1,5-a]pyrimidine



embedded image


The sub-title compound was prepared according to the procedure described in Example 22 (Steps 1 & 2), using 3-chloro-1H-pyrazol-5-amine instead of 3-bromo-1H-pyrazol-5-amine as starting material. LC-MS calc. for C6H3ClI2N3 (M+H)+: m/z=405.8. found 405.8.


Step 2. tert-Butyl 3-(((2-chloro-5-iodopyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate



embedded image


A mixture of 2-chloro-5,7-diiodopyrazolo[1,5-a]pyrimidine (0.23 g, 0.57 mmol), tert-butyl 3-(aminomethyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (0.16 g, 0.57 mmol) and DIPEA (0.30 mL, 1.7 mmol) in DMSO (3 mL) was heated at 100° C. for 2 h. The reaction mixture was then allowed to cool to r.t. and water was added. The precipitated solid was then filtered, rinsed with water, and dried to obtain the sub-title compound (0.28 g, 0.50 mmol, 88% yield). LCMS calc. for C21H23ClFIN5O2 (M+H)+: m/z=558.1. found: 558.0.


Step 3. 2-Chloro-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


A mixture of tert-butyl 3-(((2-chloro-5-iodopyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (0.28 g, 0.50 mmol), methyl 2,2-difluoro-2-(fluorosulfonyl)acetate (0.19 mL, 1.5 mmol) and copper(II) chloride (10 mg, 0.075 mmol) in DMF (2.5 mL) was heated at 110° C. for 3 h. The mixture was then allowed to cool to r.t., diluted with EtOAc and water. The organic layer was extracted, washed with brine, dried with Na2SO4, filtered and concentrated. The crude product was purified by FCC (EtOAc/Hexane) to obtain tert-butyl 3-(((2-chloro-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate. The above material was then taken up in 10 mL DCM and 5 mL TFA and stirred at r.t. for 1 h. Solvent was then removed to obtain the sub-title compound. LCMS calc. for C17H15ClF4N5 (M+H)+: m/z=400.1. found: 400.1.


Step 4. 3-(((2-Chloro-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide

A mixture of 2-chloro-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (10 mg, 0.025 mmol), phenyl carbamate (6.9 mg, 0.050 mmol) and DIPEA (0.044 mL, 0.25 mmol) in dioxane (1 mL) was heated at 85° C. for 20 h. The reaction mixture was allowed to cool to r.t. and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to obtain the title compound. LCMS calc. for C18H16ClF4N6O (M+H)+: m/z=443.1. found: 443.1.


Example 58. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-chlorophenyl)azetidine-1-carboxamide



embedded image


Step 1. tert-Butyl 3-(aminomethyl)-3-(4-chlorophenyl)azetidine-1-carboxylate



embedded image


The sub-title compound was prepared according to the procedure described in Example 30 (Steps 1 & 2), using (4-chlorophenyl)boronic acid instead of (4-fluorophenyl)boronic acid as starting material. LC-MS calc. for C11H14ClN2O2(M-C4H7)+: m/z=241.1. found 241.0.


Step 2. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-chlorophenyl)azetidine-1-carboxamide

The title compound was prepared according to the procedure described in Example 22, using tert-butyl 3-(aminomethyl)-3-(4-chlorophenyl)azetidine-1-carboxylate instead of tert-butyl 4-(aminomethyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate as starting material. LC-MS calc. for C18H16BrClF3N6O (M+H)+: m/z=503.0. found 503.1.


Example 59. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)azetidine-1-sulfonamide



embedded image


Step 1. tert-Butyl 3-cyano-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate



embedded image


To a solution of tert-butyl 3-cyanoazetidine-1-carboxylate (15 g, 82.0 mmol) and 2,5-difluoropyridine (10.4 g, 91.0 mmol) in THE (400 mL) at −78° C. was added lithium bis(trimethylsilyl)amide (66 mL, 99.0 mmol) and the resulting solution was stirred at −78° C. for 1 h. The mixture was then stirred at r.t. for 20 h. The reaction mixture was then quenched with sat. aq. NH4Cl and diluted with EtOAc. The organic layer was separated, washed with brine, dried with Na2SO4, filtered and concentrated. The crude product was purified by FCC (EtOAc/Hexane) to obtain the sub-title compound. LC-MS calc. for C10H9FN3O2(M-C4H7)+: m/z=222.1. found 222.1.


Step 2. tert-Butyl 3-(aminomethyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate



embedded image


To a solution of tert-butyl 3-cyano-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate (9.38 g, 33.8 mmol) and nickel(II) chloride hexahydrate (8.04 g, 33.8 mmol) in MeOH (169 mL) was added NaBH4 (6.40 g, 169 mmol) portion wise at 0° C. over 30 min. under N2. The obtained solution was stirred at r.t. for 2 h. The reaction mixture was then quench with aq. K2CO3, and NH4OH and diluted with DCM. The organic layer was separated, washed with brine, dried with Na2SO4, filtered and concentrated to obtain the sub-title compound. LC-MS calc. for C14H21FN3O2 (M+H)+: m/z=282.2. found 282.2.


Step 3. tert-Butyl 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate



embedded image


A mixture of 7-chloro-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (4.71 g, 16.3 mmol), tert-butyl 3-(aminomethyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate (5.72 g, 20.3 mmol) and DIPEA (7.10 mL, 40.7 mmol) in DMSO (54.2 mL) was heated at 100° C. for 2 h. Reaction mixture was then allowed to cool to r.t. and added dropwise to ice/water. The precipitated solid was filtered and dried. The crude product was purified by FCC (EtOAc/Hexane) to obtain the sub-title compound as yellow foam. LC-MS calc. for C22H22F7N6O2 (M+H)+: m/z=535.2. found 535.1.


Step 4. N-((3-(5-Fluoropyridin-2-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


To a solution of tert-butyl 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate (6.09 g, 11.4 mmol) in DCM (60 mL) was added TFA (30.0 mL) and the resulting solution was stirred at r.t. for 1 h. Solvent was then removed. The obtained crude product was taken up in 30 mL DCM and 30 mL H2O, followed by addition of sat. aq. NaHCO3 (60 mL) slowly. The mixture was stirred at r.t. for 15 min. The organic layer was then extracted with DCM, dried with Na2SO4 and concentrated to obtain the sub-title compound. LC-MS calc. for C17H14F7N6 (M+H)+: m/z=435.1. found 435.2.


Step 5. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)azetidine-1-sulfonamide

A mixture of N-((3-(5-fluoropyridin-2-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (20.5 mg, 0.047 mmol), sulfuric diamide (9.1 mg, 0.094 mmol), and DIPEA (0.041 mL, 0.24 mmol) in dioxane (1 mL) was heated at 85° C. for 20 h. The reaction mixture was allowed to cool to r.t. and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to obtain the title compound. LCMS calc. for C17H15F7N7O2S (M+H)+: m/z=514.1. found: 514.2. 1H NMR (600 MHz, DMSO-d6) δ 9.25 (t, J=6.8 Hz, 1H), 8.42 (d, J=2.9 Hz, 1H), 7.60 (m, 1H), 7.41 (dd, J=8.7, 4.3 Hz, 1H), 7.16 (s, 1H), 6.93 (s, 2H), 5.69 (s, 1H), 4.16-4.10 (m, 4H), 4.07-4.03 (m, 2H) ppm.


Example 60. 2-(Azetidin-1-yl)-1-(4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidin-1-yl)ethan-1-one



embedded image


Step 1. 2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol



embedded image


A mixture of 3-bromo-1H-pyrazol-5-amine (3.00 g, 18.5 mmol), methyl 4,4,4-trifluoro-3-oxobutanoate (4.72 g, 27.8 mmol) and TFA (0.29 mL, 3.70 mmol) in DMSO (14.3 mL) was stirred at r.t. for 4 days. Water was added and the precipitated solid was filtered and dried to obtain the sub-title compound (3.59 g, 12.73 mmol, 68.7% yield). LC-MS calc. for C7H4BrF3N3O (M+H)+: m/z=281.9. found 281.9.


Step 2. 2-Bromo-7-chloro-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine



embedded image


A solution of 2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-ol (3.75 g, 13.3 mmol) in POCl3 (37.5 mL, 402 mmol) was heated to 110° C. After 20 h, the mixture was allowed to cool to r.t. and poured over ice water slowly with stirring. The precipitated solid was filtered, washed with water and dried to give the sub-title compound as white solid (3.5 g, 12 mmol, 88% yield).


Step 3. tert-Butyl 4-cyano-4-(4-fluorophenyl)piperidine-1-carboxylate



embedded image


To a solution of 2-(4-fluorophenyl)MeCN (1.55 g, 11.5 mmol) and tert-butyl bis(2-chloroethyl)carbamate (3.33 g, 13.8 mmol) in DMF (38.2 mL) under N2 at 0° C. was added 5 sodium hydride (1.15 g, 28.7 mmol) portion wise over 30 mins. The mixture was stirred at r.t. for 30 min., then heated to 80° C. for 20 h. The mixture was then allowed to cool to r.t. and poured in ice water. EtOAc was added and organic layer was separated, washed with brine, dried over Na2SO4 and concentrated. The crude product was purified by FCC (EtOAc/Hexane) to obtain the product as yellow oil (2.82 g, 11.5 mmol, 81% yield). LC-MS calc. for C13H14FN2O2(M−C4H7)+: m/z=249.1. found 249.1.


Step 4. tert-Butyl 4-(aminomethyl)-4-(4-fluorophenyl)piperidine-1-carboxylate



embedded image


To a solution of tert-butyl 4-cyano-4-(4-fluorophenyl)piperidine-1-carboxylate (2.82 g, 9.27 mmol) and nickel(II) chloride hexahydrate (2.20 g, 9.27 mmol) in MeOH (46.3 mL) was added NaBH4 (1.75 g, 46.3 mmol) portion wise. The obtained solution was stirred at r.t. for 2 h. The mixture was then quenched with aq. K2CO3 and diluted with DCM. The organic layer was separated, washed with brine, dried over Na2SO4 and concentrated to obtain the sub-title compound (2.74 g, 9.27 mmol, 96% yield). LC-MS calc. for C13H18FN2O2(M-C4H7)+: m/z=253.1. found 253.2.


Step 5. tert-Butyl 4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidine-1-carboxylate



embedded image


A mixture of 2-bromo-7-chloro-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (200 mg, 0.666 mmol), tert-butyl 4-(aminomethyl)-4-(4-fluorophenyl)piperidine-1-carboxylate (246 mg, 0.799 mmol) and DIPEA (230 μL, 1.33 mmol) in DMSO (3 mL) was stirred at 100° C. for 3 h. Water was then added and the precipitated solid was filtered and dried to obtain the sub-title compound. LC-MS calc. for C24H27BrF4N5O2 (M+H)+: m/z=572.1. found 572.1.


Step 6. 2-Bromo-N-((4-(4-fluorophenyl)piperidin-4-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


The obtained product from Step 5 was taken up in 3 mL DCM and 2 mL TFA was added. The mixture was stirred at r.t. for 1 h. The solvent was concentrated to obtain the sub-title compound. LC-MS calc. for C19H19BrF4N5 (M+H)+: m/z=472.1. found 472.1.


Step 7. 2-(Azetidin-1-yl)-1-(4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)piperidin-1-yl)ethan-1-one

A mixture of 2-bromo-N-((4-(4-fluorophenyl)piperidin-4-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (10 mg, 0.021 mmol), 2-(azetidin-1-yl)acetic acid hydrochloride (6.4 mg, 0.042 mmol), HATU (12 mg, 0.032 mmol) and DIPEA (0.015 mL, 0.085 mmol) in DMF (1 mL) was stirred at r.t. for 1 h. The reaction mixture was then purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to obtain the title compound. LCMS calc. for C24H26BrF4N6O (M+H)+: m/z=569.1. found: 569.2.


Example 61. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)-N-(methyl-d3)azetidine-1-carboxamide



embedded image


A mixture of N-((3-(5-fluoropyridin-2-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (38 mg, 0.089 mmol), 4-nitrophenyl (methyl-d3)carbamate (35 mg, 0.18 mmol) and DIPEA (0.078 mL, 0.45 mmol) in dioxane (2 mL) was heated at 85° C. for 2 h. The mixture was then allowed to cool to r.t. and solvent was removed. The crude product was purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to obtain the title compound. LCMS calc. for C19H14D3F7N7O (M+H)+: m/z=495.2. found: 495.2. 1H NMR (600 MHz, DMSO-d6) δ 9.21-9.16 (m, 1H), 8.43 (d, J=3.0 Hz, 1H), 7.59 (m, 1H), 7.43 (dd, J=8.7, 4.3 Hz, 1H), 7.16 (s, 1H), 5.85 (s, 1H), 4.23-4.19 (m, 2H), 4.11-4.06 (m, 4H) ppm.


Example 62. 3-(((2-(Difluoromethyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(methyl-d3)azetidine-1-carboxamide



embedded image


Step 1. tert-Butyl 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate



embedded image


The sub-title compound was prepared according to the procedure described in Example 60 (Step 5), using tert-butyl 3-(aminomethyl)-3-(4-fluorophenyl)azetidine-1-carboxylate instead of tert-butyl 4-(aminomethyl)-4-(4-fluorophenyl)piperidine-1-carboxylate as starting material.


LC-MS calc. for C22H23BrF4N5O2 (M+H)+: m/z=544.1. found 544.0.


Step 2. tert-Butyl 3-(4-fluorophenyl)-3-(((5-(trifluoromethyl)-2-vinylpyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxylate



embedded image


A mixture of tert-butyl 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (485 mg, 0.891 mmol), potassium vinyltrifluoroborate (179 mg, 1.34 mmol), Pd(dppf)Cl2·DCM (73 mg, 0.089 mmol) and Et3N (248 μL, 1.78 mmol) in 1-propanol (4.5 mL) was heated at 90° C. for 3 h. The reaction mixture was then allowed to cool to r.t., filtered through diatomaceous earth and washed with DCM. The crude product was purified by FCC (EtOAc/Hexane) to obtain the sub-title compound (390 mg, 0.79 mmol, 89% yield). LC-MS calc. for C24H26F4N5O2 (M+H)+: m/z=492.2. found 492.2.


Step 3. tert-Butyl 3-(4-fluorophenyl)-3-(((2-formyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxylate



embedded image


To a mixture of tert-butyl 3-(4-fluorophenyl)-3-(((5-(trifluoromethyl)-2-vinylpyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxylate (390 mg, 0.79 mmol) in THE (3 mL), DCM (1 mL) and water (1 mL) was added osmium tetroxide (0.242 mL, 4 wt. % in water, 0.040 mmol) and 4-methylmorpholine N-oxide (0.20 mL, 0.95 mmol). The resulting mixture was stirred at r.t. for 20 h. To the reaction mixture was then added sodium periodate (424 mg, 1.984 mmol) and stirred at r.t. for 3 h. The reaction mixture was diluted with DCM and sat. aq. sodium thiosulfate solution. The product was extracted with DCM, and the organic phase was washed with brine, dried with Na2SO4, filtered and concentrated to obtain the sub-title compound (370 mg, 0.75 mmol, 94% yield). LC-MS calc. for C23H24F4N5O3 (M+H)+: m/z=494.2. found 494.3.


Step 4. 2-(Difluoromethyl)-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


To a solution of tert-butyl 3-(4-fluorophenyl)-3-(((2-formyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxylate (170 mg, 0.35 mmol) in DCM (3 mL) was added DAST (0.14 mL, 1.0 mmol) and the resulting solution was stirred at r.t. for 20 h. The reaction was quenched with sat. aq. NaHCO3 and the organic layer was extracted with DCM, washed with brine and dried over Na2SO4 to obtain tert-butyl 3-(((2-(difluoromethyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate. The above material was then taken up in 2 mL DCM and 2 mL TFA was added. The reaction mixture was stirred at r.t. for 1 h. Solvent was then removed to obtain the sub-title compound. LC-MS calc. for C18H16F6N5 (M+H)+: m/z=416.2. found 416.2.


Step 5. 3-(((2-(Difluoromethyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(methyl-d3)azetidine-1-carboxamide

The title compound was prepared according to the procedure described in Example 61, using 2-(difluoromethyl)-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine instead of N-((3-(5-fluoropyridin-2-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine as starting material. LC-MS calc. for C20H16D3F6N6O (M+H)+: m/z=476.2. found 476.3. 1H NMR (600 MHz, DMSO-d6) δ 9.06 (t, J=6.8 Hz, 1H), 7.28 (s, 1H), 7.21-7.16 (m, 2H), 7.00 (t, J=8.6 Hz, 2H), 6.87 (s, 1H), 5.80 (s, 1H), 4.27-4.23 (m, 2H), 4.02-3.97 (m, 4H) ppm.


Example 63. N-Cyclopropyl-3-(((2-(difluoromethyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide



embedded image


A solution of 2-(difluoromethyl)-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (20 mg, 0.048 mmol), cyclopropylcarbamic chloride (12 mg, 0.096 mmol) and DIPEA (0.084 mL, 0.48 mmol) in dioxane (1 mL) and DCM (1 mL) was stirred at r.t. for 2 h. The solvent was removed and the crude product was purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to obtain the title compound. LC-MS calc. for C22H21F6N6O (M+H)+: m/z=499.2. found 499.2.


Example 64. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-(3-cyanobicyclo[1.1.1]pentan-1-yl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxamide



embedded image


Step 1. 4-Nitrophenyl (3-cyanobicyclo[1.1.1]pentan-1-yl)carbamate



embedded image


A mixture of 4-nitrophenyl carbonochloridate (1.00 g, 5.0 mmol), 3-aminobicyclo[1.1.1]pentane-1-carbonitrile (0.62 g, 5.7 mmol) and sodium carbonate (1.2 g, 11 mmol) in DCM (100 mL) was stirred at r.t. for 4 days. The reaction mixture was then filtered, and the obtained filtrate was concentrated to give the sub-title compound.


Step 2. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-(3-cyanobicyclo[1.1.1]pentan-1-yl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxamide

A mixture of N-((3-(5-fluoropyridin-2-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (100 mg, 0.23 mmol), 4-nitrophenyl (3-cyanobicyclo[1.1.1]pentan-1-yl)carbamate (130 mg, 0.46 mmol) and DIPEA (0.20 mL, 1.2 mmol) in dioxane (3 mL) was heated at 85° C. for 4 h. The solvent was removed and the crude product was purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to obtain the title compound.


LC-MS calc. for C24H20F7N8O (M+H)+. m/z=569.2. found 569.1.


Example 65. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-(3-cyanobicyclo[1.1.1]pentan-1-yl-3-3,5-difluorophenyl)azetidine-1-carboxamide



embedded image


Step 1. tert-Butyl 3-(aminomethyl)-3-(3,5-difluorophenyl)azetidine-1-carboxylate



embedded image


The sub-title compound was prepared according to the procedure described in Example 30, using (3,5-difluorophenyl)boronic acid instead of (4-fluorophenyl)boronic acid as starting material. LC-MS calc. for C11H13F2N2O2(M-C4H7)+: m/z=243.1. found 243.1.


Step 2. N-((3-(3,5-Difluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


The sub-title compound was prepared according to the procedure described in Example 59 (Steps 3 and 4), using tert-butyl 3-(aminomethyl)-3-(3,5-difluorophenyl)azetidine-1-carboxylate instead of tert-butyl 3-(aminomethyl)-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate as starting material. LC-MS calc. for C18H14F8N5 (M+H)+: m/z=452.1. found 452.0.


Step 3. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-(3-cyanobicyclo[1.1.1]pentan-1-yl)-3-(3,5-difluorophenyl)azetidine-1-carboxamide

The title compound was prepared according to the procedure described in Example 64, using N-((3-(3,5-difluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine instead of N-((3-(5-fluoropyridin-2-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine as starting material. LC-MS calc. for C25H20F8N7O (M+H)+: m/z=586.2. found 586.2.


Example 66. 3-(((5-Chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide



embedded image


Step 1. 5,7-Dichloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine



embedded image


A mixture of 3-(trifluoromethyl)-1H-pyrazol-5-amine (1.0 g, 6.6 mmol), diethyl malonate (10 mL, 66.2 mmol), and sodium ethoxide (1.4 g, 20 mmol) was heated to 100° C. and stirred for 16 h. Upon completion the suspension was filtered, and the precipitate was washed with Et2O. This solid was then taken up in POCl3 (10 mL, 110 mmol) and heated to 120° C. for 5 h. The crude reaction mixture was then concentrated in vacuo. The resulting residue was washed with DCM and sat. aq. NaHCO3. The organic layers were then concentrated in vacuo and purified by FCC using BIOTAGE® ISOLERA™ (hexane/EtOAc, 0-100% EtOAc) to provide the sub-title compound. LCMS calc. for C7H3C12F3N3 (M+H)+: m/z=256.0, 258.0. found: 256.0, 257.9.


Step 2. tert-Butyl 3-(((5-chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate



embedded image


To a vial containing 5,7-dichloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (0.5 g, 2.0 mmol) and tert-butyl 3-(aminomethyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (Example 30 Step 2, 0.55 g, 2.0 mmol) in MeCN (20 mL) was added DIPEA (0.7 mL, 3.9 mmol). The vial was sealed and heated to 80° C. for 16 h. Upon completion the crude mixture was concentrated in vacuo and purified by FCC using BIOTAGE® ISOLERA™ (hexane/EtOAc, 0-100% EtOAc) to provide the sub-title compound. LCMS calc. for C22H23ClF4N5O2 (M+H)+: m/z=500.2, 502.1. found: 500.2, 502.2.


Step 3. 5-Chloro-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


A solution of tert-butyl 3-(((5-chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (15 mg, 0.030 mmol) and trifluoroacetic acid (46 μL, 0.60 mmol) in DCM (0.5 mL) stirred at r.t. for 1 h. Upon completion the crude mixture was concentrated in vacuo and used directly for the next step. LCMS calc. for C17H15ClF4N5 (M+H)+: m/z=400.1. found: 400.1.


Step 4. 3-(((5-Chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide

To a solution of 5-chloro-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (15 mg, 0.036 mmol) and phenyl carbamate (10 mg, 0.70 mmol) in MeCN (0.5 mL) was added DIPEA (13 μL, 0.073 mmol) and heated to 80° C. for 1 h. Upon completion the mixture was allowed to cool to r.t., the residue was diluted with MeCN and purified by prep.-LCMS (XBRIDGE® C18 column, eluting with a gradient of MeCN/water containing 0.1% TFA, at flow rate of 60 mL/min.) to afford the title compound.


LCMS calc. for C18H16ClF4N6O (M+H)+: m/z=443.1. found: 443.2.


Example 67. 3-(4-Fluorophenyl)-3-(((5-methyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxamide



embedded image


Step 1. 7-Chloro-5-methyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine



embedded image


A solution of 3-(trifluoromethyl)-1H-pyrazol-5-amine (1.5 g, 9.9 mmol), ethyl 3-oxobutanoate (6.5 g, 50 mmol), in acetic acid (25 mL) was heated to 90° C. and stirred for 16 h. Upon completion the suspension was diluted with Et2O, the precipitate was filtered and collected. This solid was then taken up in POCl3 (10 mL, 107 mmol) and heated to 100° C. for 2 h. The crude reaction mixture was then concentrated in vacuo. The resulting residue was dissolved in DCM and was washed with sat. aq, NaHCO3. The organic layers were then concentrated in vacuo and purified by FCC using (hexane/EtOAc, 0-100% EtOAc) to provide the sub-title compound. LCMS calc. for C8H6ClF3N3 (M+H)+: m/z=236.0. found: 236.0.


Step 2. 3-(4-Fluorophenyl)-3-(((5-methyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxamide

The title compound was prepared according to the procedures described in Example 66, with 7-chloro-5-methyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine replacing 5,7-dichloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine in Step 2. LCMS calc. for C19H19F4N6O (M+H)+: m/z=423.2. found: 423.1. 1H NMR (600 MHz, DMSO) δ 8.83 (s, 1H), 7.29-7.22 (m, 2H), 7.10 (t, J=8.8 Hz, 2H), 6.82 (s, 1H), 5.95 (s, 2H), 5.78 (s, 1H), 4.23 (d, J=8.3 Hz, 2H), 4.00 (d, J=8.3 Hz, 2H), 3.88 (d, J=6.7 Hz, 2H), 2.25 (s, 3H).


Example 68. 3-(((5-Ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide



embedded image


The title compound was prepared according to the procedures described in Example 67, with methyl 3-oxopentanoate replacing ethyl 3-oxobutanoate in Step 1. LCMS calc. for C20H21F4N6O (M+H)+: m/z=437.2. found: 437.2. 1H NMR (500 MHz, DMSO) δ 8.69 (s, 1H), 7.24 (dd, J=8.5, 5.3 Hz, 2H), 7.07 (t, J=8.7 Hz, 2H), 6.80 (s, 1H), 5.94 (s, 2H), 5.67 (s, 1H), 4.24 (d, J=8.3 Hz, 2H), 3.99 (d, J=8.3 Hz, 2H), 3.91 (d, J=6.7 Hz, 2H), 2.50-2.44 (m, 2H), 1.10 (t, J=7.6 Hz, 3H).


Example 69. 3-(((5-Cyclopropyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide



embedded image


Step 1. 4-Nitrophenyl ((1r,4r)-4-hydroxycyclohexyl)carbamate



embedded image


A solution of (1r,4r)-4-aminocyclohexan-1-ol (0.30 g, 2.6 mmol), 4-nitrophenyl chloroformate (0.35 g, 1.7 mmol), and sodium bicarbonate (0.29 g, 3.5 mmol) in DCM (5 mL) was stirred at r.t. for 4 days. Upon completion the suspension was filtered, the filtrate was collected, concentrated in vacuo, and used directly for the next step. LCMS calc. for C13H17N2O5 (M+H)+: m/z=281.1. found: 281.1.


Step 2. 3-(((5-Cyclopropyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide

The title compound was prepared according to the procedures described in Example 67, with ethyl 3-cyclopropyl-3-oxopropanoate replacing ethyl 3-oxobutanoate in Step 1, and 4-nitrophenyl ((1r,4r)-4-hydroxycyclohexyl)carbamate replacing phenyl carbamate in the final step. LCMS calc. for C27H31F4N6O2 (M+H)+: m/z=547.2. found: 547.2. 1H NMR (600 MHz, DMSO) δ 8.25 (d, J=7.0 Hz, 1H), 7.26 (dd, J=8.4, 5.3 Hz, 2H), 7.11 (t, J=8.6 Hz, 2H), 6.69 (s, 1H), 6.08 (d, J=8.0 Hz, 1H), 5.75 (s, 1H), 4.21 (d, J=8.3 Hz, 2H), 3.96 (d, J=8.3 Hz, 2H), 3.83 (d, J=6.7 Hz, 2H), 3.34-3.28 (m, 1H), 3.25 (s, 1H), 1.87-1.80 (m, 1H), 1.80-1.73 (m, 2H), 1.71-1.63 (m, 2H), 1.20-1.11 (m, 4H), 0.97-0.90 (m, 2H), 0.89-0.83 (m, 2H).


Example 70. 3-(4-Fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxamide



embedded image


Step 1. tert-Butyl 3-(4-fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxylate




embedded image


To a vial under N2 atmosphere was added tert-butyl 3-(((5-chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (Example 66, Step 2, 300 mg, 0.60 mmol), ferric acetylacetonate (21 mg, 0.060 mmol), and THE (15 mL). This was cooled to 0° C. and (methyl-d3)magnesium iodide (5 mL, 5 mmol) was added slowly in 3 portions over 10 min. The mixture was then allowed to warm to r.t. and stirred for 2 h. Upon completion this was quenched with sat. aq. NH4Cl and washed with DCM. The organic layer was filtered through diatomaceous earth, concentrated in vacuo and used directly for the next step. LCMS calc. for C23H23D3F4N5O2 (M+H)+: m/z=483.2. found: 483.2.


Step 2. 3-(4-Fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxamide

The title compound was prepared according to the procedures described in Example 66, with tert-butyl 3-(4-fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxylate replacing tert-butyl 3-(((5-chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate in Step 4. LCMS calc. for C19H16D3F4N6O (M+H)+: m/z=426.2. found: 426.2. 1H NMR (500 MHz, DMSO) δ 8.71 (s, 1H), 7.30-7.22 (m, 2H), 7.10 (t, J=8.8 Hz, 2H), 6.80 (s, 1H), 5.93 (s, 2H), 5.77 (s, 1H), 4.23 (d, J=8.3 Hz, 2H), 4.00 (d, J=8.3 Hz, 2H), 3.87 (d, J=6.6 Hz, 2H).


Example 71. 3-(((5-Ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(2,2,2-trifluoroethyl)azetidine-1-carboxamide



embedded image


The title compound was prepared according to the procedures described in Example 67, with methyl 3-oxopentanoate replacing ethyl 3-oxobutanoate in Step 1, and 1,1,1-trifluoro-2-isocyanatoethane replacing phenyl carbamate in the final step. LCMS calc. for C22H22F7N6O (M+H)+: m/z=519.2. found: 519.2. 1H NMR (600 MHz, DMSO) δ 8.73 (s, 1H), 7.26 (dd, J=8.5, 5.3 Hz, 2H), 7.14-7.05 (m, 3H), 6.81 (s, 1H), 5.68 (s, 1H), 4.33 (d, J=8.3 Hz, 2H), 4.06 (d, J=8.4 Hz, 2H), 3.92 (d, J=6.7 Hz, 2H), 3.80-3.71 (m, 2H), 2.51-2.46 (m, 2H), 1.10 (t, J=7.6 Hz, 3H).


Example 72. 1-Amino-4-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexane-1-carbonitrile



embedded image


Step 1. 1-(4-Fluorophenyl)-4-hydroxycyclohexane-1-carbonitrile




embedded image


To a solution of 1-(4-fluorophenyl)-4-oxocyclohexane-1-carbonitrile (5.0 g, 23.02 mmol) in MeOH (115 ml) was added sodium borohydride (0.96 g, 25.3 mmol) at 0° C. and the reaction mixture was stirred for 2 h at rt. The reaction mixture was quenched with water and extracted with EtOAc (2×100 mL). The combined organic phases were dried over Na2SO4 and concentrated to give a crude oil, which was used directly in the next step without further purification. LC-MS calculated for C13H15FNO (M+H)+: m/z=220.1. found 220.1.


Step 2. 4-(Aminomethyl)-4-(4-fluorophenyl)cyclohexan-1-ol



embedded image


This compound was prepared according to the procedure described in Example 60, step 4 using 1-(4-fluorophenyl)-4-hydroxycyclohexane-1-carbonitrile as starting material. LCMS calculated for C13H19FNO (M+H)+: m/z=224.1. found: 224.1.


Step 3. 4-(((5-Ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexan-1-ol



embedded image


This compound was prepared according to the procedure described in Example 43, step 1 using 7-chloro-5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine and 4-(aminomethyl)-4-(4-fluorophenyl)cyclohexan-1-ol as starting material. LCMS calculated for C22H25F4N4O (M+H)+: m/z=437.2. found: 437.3.


Step 4. 4-(((5-Ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexan-1-one



embedded image


This compound was prepared according to the procedure described in Example 43, step 2 using 4-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexan-1-ol as starting material. LCMS calculated for C22H23F4N4O (M+H)+: m/z=435.1. found: 435.2.


Step 5. 1-Amino-4-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexane-1-carbonitrile

A solution of 4-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexan-1-one (25 mg, 0.058 mmol), ammonium chloride (4 mg, 0.069 mmol), ammonia (57.5 μl, 0.115 mmol) in EtOH (60 μl) and water (50 μl) was stirred for 10 min before sodium cyanide (4 mg, 0.075 mmol) was added. The reaction mixture was stirred for 48 h at rt. Upon completion, the reaction mixture was diluted with MeOH and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the desired product. The two diastereomers could be separated, with the desired diastereomer eluting at 4.8 minutes. LCMS calculated for C23H25F4N6 (M+H)+: m/z=461.2. found: 461.3.


The diastereomers of the title compound that are separable by chromatography are: (1s,4s)-1-amino-4-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexane-1-carbonitrile and (1r,4r)-1-amino-4-(((5-ethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexane-1-carbonitrile.


Example 73. 1-((3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutyl)amino)-2-methylpropan-2-ol



embedded image


Step 1. 1-(4-Fluorophenyl)-3-hydroxycyclobutane-1-carbonitrile



embedded image


This compound was prepared according to the procedure described in Example 72, step 1 using 1-(4-fluorophenyl)-3-oxocyclobutane-1-carbonitrile as starting material. LCMS calculated for C11H11FNO (M+H)+: m/z=192.1. found: 192.1.


Step 2. 3-(Aminomethyl)-3-(4-fluorophenyl)cyclobutan-1-ol



embedded image


This compound was prepared according to the procedure described in Example 72, step 2 using 1-(4-fluorophenyl)-4-hydroxycyclohexane-1-carbonitrile as starting material. LCMS calculated for C11H15FNO (M+H)+: m/z=196.1. found: 196.2.


Step 3. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutan-1-ol



embedded image


This compound was prepared according to the procedure described in Example 72, step 3 using 7-chloro-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidine and 3-(aminomethyl)-3-(4-fluorophenyl)cyclobutan-1-ol as starting material. LCMS calculated for C19H16F7N4O (M+H)+: m/z=449.1. found: 449.2.


Step 4. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutan-1-one



embedded image


This compound was prepared according to the procedure described in Example 72, step 4 using 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutan-1-ol as starting material. LCMS calculated for C19H14F7N4O (M+H)+: m/z=447.1. found: 447.2.


Step 5. 1-((3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutyl)amino)-2-methylpropan-2-ol

This compound was prepared according to the procedure described in Example 43, step 3 using 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutan-1-one and 1-amino-2-methylpropan-2-ol as starting material. Upon completion the reaction mixture was diluted with MeOH and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% NH4OH. The two diastereomers could be separated, with the desired diastereomer eluting first (at 3.5 minutes).


LC-MS calculated for C23H25F7N5O (M+H)+: m/z=520.2. found 520.3. 1H NMR (500 MHz, DMSO) δ 9.00 (t, J=6.8 Hz, 1H), 8.53 (d, J=9.0 Hz, 1H), 7.17-7.07 (m, 3H), 6.99 (t, J=8.6 Hz, 2H), 5.71 (s, 1H), 5.12 (s, 1H), 3.86 (m, 3H), 2.92 (m, 2H), 2.83-2.73 (m, 2H), 2.55-2.42 (m, 2H), 1.18 (s, 6H).


The diastereomers of the title compound that are separable by chromatography are: 1-(((1r,3r)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutyl)amino)-2-methylpropan-2-ol and 1-(((1s,3s)-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutyl)amino)-2-methylpropan-2-ol.


Example 74. 1-((4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexyl)amino)-2-methylpropan-2-ol



embedded image


Step 1. 4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexan-1-ol



embedded image


This compound was prepared according to the procedure described in Example 72, step 3 using 7-chloro-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidine and 4-(aminomethyl)-4-(4-fluorophenyl)cyclohexan-1-ol as starting material. LCMS calculated for C21H20F7N4O (M+H)+: m/z=477.1. found: 477.2.


Step 2. 4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexan-1-one



embedded image


This compound was prepared according to the procedure described in Example 72, step 4 using 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexan-1-ol as starting material. LCMS calculated for C21H18F7N4O (M+H)+: m/z=475.1. found: 475.2.


Step 3. 1-((4-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexyl)amino)-2-methylpropan-2-ol

This compound was prepared according to the procedure described in Example 73, step 5 using 4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexan-1-one and 1-amino-2-methylpropan-2-ol as starting materials. Upon completion, the reaction mixture was diluted with MeOH and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the desired product. The two diastereomers could be separated, with the desired diastereomer eluting first (at 8.7 minutes). LC-MS calculated for C25H29F7N5O (M+H)+: m/z=548.2. found 548.3. 1H NMR (500 MHz, DMSO) δ 8.79 (t, J=7.0 Hz, 1H), 7.92 (s, 1H), 7.42 (dd, J=8.7, 5.3 Hz, 2H), 7.12 (s, 1H), 7.05 (t, J=8.7 Hz, 2H), 5.80 (s, 1H), 5.04 (s, 1H), 3.54 (d, J=6.9 Hz, 2H), 3.06 (d, J=13.1 Hz, 1H), 2.84-2.75 (m, 2H), 2.58 (m, 2H), 2.04-1.92 (m, 2H), 1.64-1.52 (m, 2H), 1.28 (m, 2H), 1.13 (s, 6H).


The diastereomers of the title compound that are separable by chromatography are: 1-(((1r,4r)-4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexyl)amino)-2-methylpropan-2-ol and 1-(((1s,4s)-4-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-(4-fluorophenyl)cyclohexyl)amino)-2-methylpropan-2-ol.


Example 75. N-(((1s,3s)-3-Methoxy-1-phenylcyclobutyl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


This compound was prepared according to the procedure described in Example 72, step 3 using 7-chloro-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidine and ((1s,3s)-3-methoxy-1-phenylcyclobutyl)methanamine as starting material. Upon completion, the reaction mixture was diluted with MeOH and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the desired product. LC-MS calculated for C20H19F6N4O (M+H)+: m/z=445.1. found 445.2.


Example 76. 1-Amino-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutane-1-carboxamide



embedded image


Step 1. 2-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(4-fluorophenyl)-5,7-diazaspiro[3.4]octane-6,8-dione



embedded image


To a solution of 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutan-1-one (50 mg, 0.112 mmol) (See example 73 for preparation) in ethanol (75 μl) and water (75 μl) was added NaCN (8 mg, 0.168 mmol) and ammonium carbonate (54 mg, 0.560 mmol). The reaction mixture was then heated at 80° C. for 12 h. The reaction mixture was quenched with water and extracted with EtOAc (2×30 mL). The combined organic phases were dried over Na2SO4 and concentrated to give a crude product, which was used directly in the next step without further purification. LCMS calculated for C21H16F7N6O2 (M+H)+: m/z=517.1. found: 517.2.


Step 2. 1-Amino-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutane-1-carboxylic acid



embedded image


To a solution of the above crude in dioxane (75 μl) was added 1M NaOH (1120 μl, 1.120 mmol) and the reaction mixture was heated at 110° C. for 4 h. The reaction mixture was then quenched with TFA and concentrated to give a crude product, which was used directly in the next step without further purification. LCMS calculated for C20H17F7N5O2 (M+H)+: m/z=492.1. found: 492.2.


Step 3. 1-Amino-3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutane-1-carboxamide

To a solution of the above crude (30 mg) in MeCN (600 μl) were added EDC (18 mg, 0.092 mmol), HOBt (14 mg, 0.092 mmol) and 0.4 M ammonia (1526 μl, 0.611 mmol) in dioxane followed by DIPEA (53 μl, 0.305 mmol). The reaction mixture was then stirred at rt for 12h.


Upon completion, the reaction mixture was diluted with MeCN and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the desired product. LC-MS calculated for C20H18F7N6O (M+H)+: m/z=491.1. found 491.2.


Example 77. 2-Methyl-N-((3-phenyltetrahydrofuran-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


A mixture of 7-chloro-2-methyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (10 mg, 0.042 mmol), (3-phenyltetrahydrofuran-3-yl)methanamine (7.5 mg, 0.042 mmol) and DIPEA (0.015 ml, 0.082 mmol) in dioxane (1 ml) was stirred at 100° C. for 1 h. After complete conversion, the reaction was cooled to rt and diluted with MeOH and purified by prep HPLC to afford the desired product. LCMS calculated for C19H20F3N4O (M+H)+: m/z=377.2. found: 377.2.


Example 78. 5-Iodo-2-methyl-N-((3-phenyltetrahydrofuran-3-yl)methyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


Step 1. 5,7-Diiodo-2-methylpyrazolo[1,5-a]pyrimidine



embedded image


To a magnetically stirred suspension of the 5,7-dichloro-2-methylpyrazolo[1,5-a]pyrimidine (0.10 g, 0.50 mmol) and sodium iodide (0.74 g, 4.95 mmol) in acetonitrile (4 ml) was added acetyl chloride (0.035 ml, 0.495 mmol), maintaining the temperature at 18° C. Then the reaction was heated for 3 hours at 80° C., then cooled down. The reaction mixture was treated with potassium carbonate (3 ml of a 10% w/v aqueous solution), sodium sulfite (3 ml of a 5% w/v aqueous solution), sodium thiosulfate (3 ml of a saturated aqueous solution) and dichloromethane (20 ml). The phases were separated, and the aqueous layer was extracted with dichloromethane (3×10 ml). The combined organic phases were dried over MgSO4. The combined organic phases were filtered and concentrated under reduced pressure. The ensuing residue was purified by flash chromatography. LCMS calculated for C7H6I2N3 (M+H)+: m/z=385.9. found: 385.9.


Step 2. 5-Iodo-2-methyl-N-((3-phenyltetrahydrofuran-3-yl)methyl)pyrazolo[1,5-a]pyrimidin-7-amine

A mixture of 5,7-diiodo-2-methylpyrazolo[1,5-a]pyrimidine (25 mg, 0.065 mmol), (3-phenyltetrahydrofuran-3-yl)methanamine (12 mg, 0.065 mmol) and DIPEA (34 μl, 0.20 mmol) in dioxane (1 ml) was stirred at 100° C. for 1 h. After complete conversion, the reaction was cooled to rt, diluted with MeOH and purified by prep HPLC to afford the desired product. LCMS calculated for C18H20IN4O (M+H)+: m/z=435.1. found: 435.1.


Example 79. 3-(((7-Chloro-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)-3-phenylazetidine-1-carboxamide



embedded image


Step 1. 1,2-Diamino-4,6-dichloropyridin-1-ium 4-methylbenzenesulfonate



embedded image


To a solution of tert-butyl tosylcarbamate (1.6 g, 5.8 mmol) in chloroform (15 mL) was added TFA (5.6 ml, 73 mmol) in small portions at 0° C. The reaction was stirred at 0° C. for 1.5 hours under N2 atmosphere, then poured into crushed ice. The white precipitate was filtered and washed with water. The solid was transferred to a round bottom flask while wet, and immediately dissolved in DCM (100 mL). The organic layer was dried over Na2SO4, filtered and concentrated. The crude material was dissolved in DCM (30 mL) and the solution was then added to a solution of 4,6-dichloropyridin-2-amine (0.70 g, 4.3 mmol) in DCM (6 mL) slowly at 0° C. The reaction mixture was stirred at 25-28° C. for 12 hours. The reaction was monitored by LCMS. After completion of the reaction, the mixture was cooled to 0° C. and the white precipitate was filtered and washed with cold DCM (5 mL). The crude material was dried in vacuo to yield the desired product as a yellow solid, which was used for the next step without further purification.


Step 2. 5,7-Dichloro-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridine



embedded image


To a suspension of 1,2-diamino-4,6-dichloropyridin-1-ium 4-methylbenzenesulfonate (0.77 g, 2.2 mmol) in MeOH (18 ml) at 0° C. was added TEA (0.92 ml, 6.6 mmol), followed by 2,2,2-trifluoroacetic anhydride (0.46 ml, 3.3 mmol) dropwise via syringe. The reaction was maintained at 0° C. for 10 min then stirred at rt for 16 h. The reaction mixture was diluted with EtOAc (20 mL) and saturated aqueous NaHCO3 (20 mL), and then the layers were separated. The aqueous layer was extracted with EtOAc (20 mL×2). The combined organic extracts were washed with brine, dried over Na2SO4 and filtered. The filtrate was concentrated in vacuo and purified by flash chromatography to afford the desired product as a white solid. LCMS calculated for C7H3C12F3N3 (M+H)+: m/z=256.0. found: 256.0.


Step 3. 7-Chloro-N-((3-phenylazetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine



embedded image


A mixture of 5,7-dichloro-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridine (0.050 g, 0.20 mmol), tert-butyl 3-(aminomethyl)-3-phenylazetidine-1-carboxylate (0.055 g, 0.20 mmol) and DIPEA (0.10 ml, 0.59 mmol) in dioxane (2 ml) was heated to 100° C. overnight. The resulting mixture was cooled to rt before TFA (5 mL) was added and stirred for 10 mins. Upon Boc deprotection completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the desired product. LCMS calculated for C17H16ClF3N5 (M+H)+: m/z=382.1. found: 382.1.


Step 4. 3-(((7-Chloro-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)-3-phenylazetidine-1-carboxamide

A mixture of 7-chloro-N-((3-phenylazetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine (10 mg, 0.025 mmol), DIPEA (0.022 ml, 0.13 mmol) and phenyl carbamate (10 mg, 0.075 mmol) in 1,4-dioxane (0.5 mL) was heated to 85° C. and stirred for 30 mins. Upon completion, the reaction mixture was diluted with MeOH and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford desired product. LCMS calculated for C18H17ClF3N6O (M+H)+: m/z=425.1. found: 425.1.


Example 80. 3-(((2,7-Bis(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutan-1-ol



embedded image


Step 1. 2,7-Bis(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-ol



embedded image


A mixture of 4-(trifluoromethyl)-1H-imidazol-2-amine hydrochloride (0.50 g, 2.7 mmol), ethyl (Z)-4,4,4-trifluoro-3-hydroxybut-2-enoate (0.49 g, 2.7 mmol) and TFA (0.21 ml, 2.7 mmol) in DMSO (10 mL) was stirred at rt overnight. The resulting mixture was poured into water while stirring at 0° C. to afford the desired product as a white solid which was filtered and dried. LCMS calculated for C8H4F6N3O (M+H)+: m/z=272.1. found: 272.1.


Step 2. 5-Chloro-2,7-bis(trifluoromethyl)imidazo[1,2-a]pyrimidine



embedded image


A mixture of 2,7-bis(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-ol (0.30 g, 1.1 mmol) and POCl3 (2.0 ml, 21 mmol) was stirred at 100° C. for 5 h. Then the reaction was cooled to rt, and icy water (20 ml) was poured into the reaction to precipitate the desired product, which was filtered and dried. LCMS calculated for C8H3F6N3 (M+H)+: m/z=290.0. found: 290.0.


Step 3. 3-(((2,7-Bis(trifluoromethyl)imidazo[1,2-a]pyrimidin-5-yl)amino)methyl)-3-(4-fluorophenyl)cyclobutan-1-ol

A mixture of 5-chloro-2,7-bis(trifluoromethyl)imidazo[1,2-a]pyrimidine (30 mg, 0.10 mmol), 3-(aminomethyl)-3-(4-fluorophenyl)cyclobutan-1-ol (20 mg, 0.10 mmol) and DIPEA (0.054 ml, 0.31 mmol) in dioxane (2 ml) was heated to 100° C. for 3h. Upon completion, the reaction mixture was concentrated in vacuo. The residue was diluted with MeOH and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the desired product. LCMS calculated for C19H16F7N4O (M+H)+: m/z=449.2. found: 449.2.


Example 81. 3-((4-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)amino)propanenitrile



embedded image


Step 1. 4-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexan-1-ol



embedded image


The procedure and workup in Example 43, Step 1 was followed using commercially available 4-(aminomethyl)-4-phenylcyclohexan-1-ol in place of 3-(aminomethyl)-3-phenylcyclobutan-1-ol. The crude material was used directly in the next step. LCMS calculated for C20H21BrF3N4O (M+H)+: m/z=469.1. found: 469.1.


Step 2. 4-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexan-1-one



embedded image


The procedure and workup in Example 43, Step 2 was followed using 4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexan-1-ol in place of 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutan-1-ol. The crude material was used directly in the next step. LCMS calculated for C20H19BrF3N4O (M+H)+: m/z=467.1. found: 467.1.


Step 3. 3-((4-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)amino)propanenitrile

The procedure in Example 43, Step 3 was followed using 4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexan-1-one in place of 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutan-1-one, and 3-aminopropanenitrile in place of 1-methylpiperazine. After reaction completion, diluted with MeCN and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% NH4OH, at flow rate of 60 mL/min, gradient of 53-66.1% MeCN in 12 minutes). The two diastereomers could be separated, with the more biologically active diastereomer eluting first (at 5.2 minutes). LCMS calculated for C23H25BrF3N6 (M+H)+: m/z=521.1. found 521.1.


The diastereomers of the title compound that are separable by chromatography are: 3-(((1r,4r)-4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)amino)propanenitrile and 3-(((1s,4s)-4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)amino)propanenitrile.


Example 82. 2-Bromo-N-((1-phenylcyclopent-3-en-1-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


The procedure in Example 43, Step 1 was followed using (1-phenylcyclopent-3-en-1-yl)methanamine (for preparation see Org. Lett. 2022, 24, 8791) in place of 3-(aminomethyl)-3-phenylcyclobutan-1-ol. Upon completion, the reaction mixture was diluted with MeCN and purified by prep-LCMS (Waters Sunfire C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min). LCMS calculated for C19H17BrF3N4 (M+H)+: m/z=437.1. found: 437.1.


Example 83. 2-Bromo-N-((4-(methylamino)-1-phenylcyclohexyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


The procedure in Example 81, Step 3 was followed using methylamine hydrochloride in place of 3-aminopropanenitrile. After completion, diluted with MeCN and purified by prep-LCMS (Waters Sunfire C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min, gradient of 23-41% MeCN in 12 minutes). The two diastereomers could be separated, with the more biologically active diastereomer eluting first (at 10.1 minutes). LCMS calculated for C21H24BrF3N5 (M+H)+: m/z=482.1. found 482.1.


The diastereomers of the title compound that are separable by chromatography are: 2-bromo-N-(((1r,4r)-4-(methylamino)-1-phenylcyclohexyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine and 2-bromo-N-(((1s,4s)-4-(methylamino)-1-phenylcyclohexyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine.


Example 84. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol



embedded image


Step 1. 3-(Aminomethyl)-3-phenylcyclopentan-1-ol



embedded image


A mixture of 1-phenylcyclopent-3-ene-1-carbonitrile (375 mg, 2.22 mmol) (for preparation see Org. Lett. 2022, 24, 8791) and borane tetrahydrofuran complex (4.4 ml, 4.4 mmol, 1 M solution) in THE (5 mL) was stirred at RT for 2 hours. After this time, NaOH (7.4 mL, 22.2 mmol, 3 M solution in water) was added followed by H2O2(2.3 mL, 22.2 mmol, 30%). The reaction was left to stir at RT for another 3 hours, and then was diluted with EtOAc and washed with water. The organic phase was dried over sodium sulfate, filtered, concentrated, and purified by chromatography (Biotage Isolera, pure DCM to 10% MeOH) to give the desired product as an oil. LCMS calculated for C12H18NO (M+H)+: m/z=192.1. found 192.1.


Step 2. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol

The procedure in Example 43, Step 1 was followed using 3-(aminomethyl)-3-phenylcyclopentan-1-ol in place of 3-(aminomethyl)-3-phenylcyclobutan-1-ol. Upon completion, the reaction mixture was diluted with MeCN and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% NH4OH, at flow rate of 60 mL/min, gradient of 47.4-65.4% MeCN in 12 minutes). The two diastereomers could be separated, with the more biologically active diastereomer eluting first (at 5.3 minutes). LCMS calculated for C19H19BrF3N4O (M+H)+: m/z=455.1. found 455.1.


The diastereomers of the title compound that are separable by chromatography are: (1R*,3S*)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol and (1S*,3S*)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol.


Each diastereomer was obtained as a racemic mixture and each is separable into its enantiomers (1R,3S)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol and (1S,3R)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol, and (1S,3S)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol and (1R,3R)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclopentan-1-ol, respectively.


Example 85. N-(4-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)-N-methylacetamide



embedded image


To a solution of 2-bromo-N-((4-(methylamino)-1-phenylcyclohexyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (30 mg, 0.062 mmol) (see Example 83 for preparation; mixture of diastereomers was used) and DIPEA (0.03 mL, 0.19 mmol) in DCM (1 mL), acetyl chloride (4.9 μl, 0.07 mmol) in DCM (0.1 mL) was added slowly. The mixture was stirred at RT for 3 hours, and then was concentrated, diluted with MeOH, and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% NH40H, at flow rate of 60 mL/min, gradient of 46-64% MeCN in 12 minutes). The two diastereomers could be separated, with the more biologically active diastereomer eluting first (at 6.5 minutes). LCMS calculated for C23H26BrF3N5O (M+H)+: m/z=524.1. found 524.1.


The diastereomers of the title compound that are separable by chromatography are: N-((1r,4r)-4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)-N-methylacetamide and N-((1s,4s)-4-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-4-phenylcyclohexyl)-N-methylacetamide.


Example 86. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol



embedded image


Step 1. 1-(Pyridin-4-yl)cyclopent-3-ene-1-carbonitrile



embedded image


To a solution of 2-(pyridin-4-yl)acetonitrile hydrochloride (1.79 g, 11.6 mmol) in NMP (12 mL) and THE (12 mL) was added sodium tert-butoxide (3.90 g, 40.6 mmol). After stirring at RT for 30 minutes, (Z)-1,4-dichlorobut-2-ene (2.4 mL, 23.2 mmol) was added slowly and then the solution was stirred at RT overnight. The solution was diluted with DCM and washed with water and brine. The organic phase was dried over sodium sulfate, concentrated, and then purified by chromatography (Biotage Isolera, pure Hex to 20% EtOAc) to give the desired product. LCMS calculated for C11H11N2 (M+H)+: m/z=171.1. found 171.1.


Step 2. 3-(Aminomethyl)-3-(pyridin-4-yl)cyclopentan-1-ol



embedded image


The procedure and workup in Example 84, Step 1 was followed using 1-(pyridin-4-yl)cyclopent-3-ene-1-carbonitrile in place of 1-phenylcyclopent-3-ene-1-carbonitrile. The crude material was purified by chromatography (Biotage Isolera, pure DCM to 10% MeOH) to give the desired product. LCMS calculated for C11H17N2O (M+H)+: m/z=193.1. found 193.1.


Step 3. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol

The procedure in Example 43, Step 1 was followed using 3-(aminomethyl)-3-(pyridin-4-yl)cyclopentan-1-ol in place of 3-(aminomethyl)-3-phenylcyclobutan-1-ol. Upon completion, the reaction mixture was diluted with MeCN and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% NH4OH, at flow rate of 60 mL/min, gradient of 29.9-47.9% MeCN in 12 minutes). The two diastereomers could be separated, with the more biologically active diastereomer eluting first (at 5.3 minutes). LC-MS calculated for C18H18BrF3N5O (M+H)+: m/z=456.1. found 456.1.


The diastereomers of the title compound that are separable by chromatography are: (1R*,3R*)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol and (1R*,3S*)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol.


Each diastereomer was obtained as a racemic mixture and each is separable into its enantiomers (1R,3R)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol and (1S,3S)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol, and (1R,3S)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol and (1S,3R)-3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-4-yl)cyclopentan-1-ol, respectively.


Example 87. 2-Bromo-N-((3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


Step 1. 1-(Pyridin-3-yl)cyclopent-3-ene-1-carbonitrile



embedded image


The procedure and workup in Example 86, Step 1 was followed using 2-(pyridin-3-yl)acetonitrile in place of 2-(pyridin-4-yl)acetonitrile hydrochloride. The crude was purified by chromatography (Biotage Isolera, pure Hex to 20% EtOAc) to give the desired product. LCMS calculated for C11H11N2 (M+H)+: m/z=171.1. found 171.1.


Step 2. 3-(Aminomethyl)-3-(pyridin-3-yl)cyclopentan-1-ol



embedded image


The procedure and workup in Example 84, Step 1 was followed using 1-(pyridin-3-yl)cyclopent-3-ene-1-carbonitrile in place of 1-phenylcyclopent-3-ene-1-carbonitrile. The crude material was purified by chromatography (Biotage Isolera, pure DCM to 10% MeOH) to give the desired product. LCMS calculated for C11H17N2O (M+H)+: m/z=193.1. found 193.1.


Step 3. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-3-yl)cyclopentan-1-ol



embedded image


The procedure in Example 43, Step 1 was followed using 3-(aminomethyl)-3-(pyridin-3-yl)cyclopentan-1-ol in place of 3-(aminomethyl)-3-phenylcyclobutan-1-ol. Upon completion, the reaction mixture was diluted with DCM and washed with water and brine. The organic phase was dried over sodium sulfate, filtered, concentrated, and then used directly in the next step. LCMS calculated for C18H18BrF3N5O (M+H)+: m/z=456.1. found 456.1.


Step 4. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-3-yl)cyclopentan-1-one



embedded image


The procedure and workup in Example 43, Step 2 was followed using 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-3-yl)cyclopentan-1-ol in place of 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutan-1-ol. The crude material was purified by chromatography (Biotage Isolera, pure Hex to 30% EtOAc) to give the desired product. LCMS calculated for C18H16BrF3N5O (M+H)+: m/z=454.0. found 454.0.


Step 5. 2-Bromo-N-((3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine

The procedure in Example 43, Step 3 was followed using 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(pyridin-3-yl)cyclopentan-1-one in place of 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-phenylcyclobutan-1-one, and methylamine hydrochloride in place of 1-methylpiperazine. After reaction completion, diluted with MeCN and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% NH4OH, at flow rate of 60 mL/min, gradient of 42.3-60.3% MeCN in 12 minutes). The diastereomer eluting second (at 4.5 minutes) was collected. LCMS calculated for C19H21BrF3N6 (M+H)+: m/z=469.1. found 469.1.


The diastereomers of the title compound that are separable by chromatography are: 2-bromo-N-(((1R*,3R*)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine and 2-bromo-N-(((1S*,3R*)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine.


Each diastereomer can be obtained as a racemic mixture and each is separable into its enantiomers 2-bromo-N-(((1R,3R)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine and 2-bromo-N-(((1S,3S)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine, and 2-bromo-N-(((1R,3S)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine and 2-bromo-N-(((1S,3R)-3-(methylamino)-1-(pyridin-3-yl)cyclopentyl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine, respectively


Example 88. (2-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol



embedded image


Step 1. 2-(Hydroxymethyl)-1-(pyridin-2-yl)cyclopropane-1-carbonitrile



embedded image


To a solution of 2-(pyridin-2-yl)acetonitrile (500 mg, 4.23 mmol) in THE (10 mL) at −78° C., MeLi (2.65 ml, 4.23 mmol) was added over 30 minutes. The solution was left to stir at −78° C. for another 1 hour and then 2-(bromomethyl)oxirane (0.325 ml, 3.93 mmol) was added over 20 minutes. The solution was left to stir at −78° C. for another 1 hour, and then MeMgBr (1.411 ml, 4.23 mmol) was added over 30 minutes. The mixture was left to stir to RT overnight, and then was quenched with 100 mL of water and extracted with EtOAc. The organic phase was dried over sodium sulfate, filtered, concentrated, and purified by chromatography (Biotage Isolera, pure Hex to 60% EtOAc) to give the desired product mixed with the cyclobutanol isomer. LCMS calculated for C10H11N2O (M+H)+: m/z=175.1. found 175.1.


Step 2. (2-(Aminomethyl)-2-(pyridin-2-yl)cyclopropyl)methanol



embedded image


A solution of 2-(hydroxymethyl)-1-(pyridin-2-yl)cyclopropane-1-carbonitrile (170 mg, 0.98 mmol) (mixed with cyclobutanol isomer) and LiAlH4 (0.59 ml, 1.17 mmol) in THE (5 mL) was heated to 65° C. for 4 hours. The solution was cooled to RT, quenched with 50 mL of water, and extracted with EtOAc. The organic phase was dried over sodium sulfate, filtered, and concentrated to give the crude cyclopropyl product mixed with the cyclobutanol isomer. The crude was used directly in the next step. LCMS calculated for C10H15N2O (M+H)+: m/z=179.1; found 179.1.


Step 3. (2-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol

The procedure in Example 43, Step 1 was followed using (2-(aminomethyl)-2-(pyridin-2-yl)cyclopropyl)methanol (mixed with cyclobutanol isomer) in place of 3-(aminomethyl)-3-phenylcyclobutan-1-ol. Upon completion, the reaction mixture was diluted with MeCN and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% NH4OH, at flow rate of 60 mL/min, gradient of 31.6-49.6% MeCN in 12 minutes). The cyclopropyl product eluted as a single diastereomer (at 8.6 minutes) after the cyclobutyl isomer. LCMS calculated for C17H16BrF3N5O (M+H)+: m/z=442.0. found 442.1. 1H NMR (600 MHz, DMSO-d6) δ 8.44 (d, J=4.8 Hz, 1H), 7.66 (m, 1H), 7.25 (d, J=8.0 Hz, 1H), 7.16 (dd, J=7.4, 4.8 Hz, 1H), 6.81-6.70 (m, 2H), 4.50 (d, J=14.5 Hz, 1H), 4.01 (dd, J=11.9, 5.3 Hz, 1H), 3.80 (d, J=14.6 Hz, 1H), 3.46 (dd, J=11.9, 10.1 Hz, 1H), 1.88-1.81 (m, 1H), 1.35 (dd, J=9.0, 4.4 Hz, 1H), 1.13 (t, J=5.4 Hz, 1H).


The two possible diastereomers of the title compound are: ((1R*,2S*)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol and ((1R*,2R*)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol.


Each diastereomer can be obtained as a racemic mixture and each is separable into its enantiomers ((1R,2S)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol and ((1S,2R)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol, and ((1R,2R)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol and ((1S,2S)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-(pyridin-2-yl)cyclopropyl)methanol, respectively.


Example 89. (2-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol



embedded image


Step 1. 2-(Hydroxymethyl)-1-phenylcyclopropane-1-carbonitrile



embedded image


The procedure and workup in Example 88, Step 1 was followed using 2-phenylacetonitrile in place of 2-(pyridin-2-yl)acetonitrile. The crude mixture was purified by chromatography (Biotage Isolera, pure Hex to 50% EtOAc) to give the desired product mixed with the cyclobutanol isomer. LCMS calculated for C11H12NO (M+H)+: m/z=174.1. found 174.0.


Step 2. (2-(Aminomethyl)-2-phenylcyclopropyl)methanol



embedded image


To a solution of 2-(hydroxymethyl)-1-phenylcyclopropane-1-carbonitrile (1.6 g, 9.24 mmol) (mixed with cyclobutanol isomer) and NiCl2·6H2O (2.2 g, 9.24 mmol) in MeOH (10 mL) at RT, NaBH4 (1.7 g, 46 mmol) was added portionwise. After stirring for 2 hours, the mixture was diluted with aq NaHCO3 (200 mL) and water (100 mL) and then extracted with EtOAc (2×200 mL). The combined organic phases were dried over sodium sulfate, filtered, concentrated, and purified by chromatography (Biotage Isolera, pure DCM to 10% MeOH) to give the desired product mixed with the cyclobutanol isomer. LCMS calculated for C11H16NO (M+H)+: m/z=178.1. found 178.1.


Step 3. 2-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropane-1-carbaldehyde



embedded image


The procedure and workup in Example 43, Step 1 was followed using (((2-(aminomethyl)-2-phenylcyclopropyl)methanol (mixed with cyclobutanol isomer) in place of 3-(aminomethyl)-3-phenylcyclobutan-1-ol. The crude was oxidized using the procedure and workup in Example 43, Step 2 to give a mixture of the desired cyclopropyl isomer and cyclobutanone isomer which were separable by chromatography (Biotage Isolera, pure Hex to 30% EtOAc, the cyclopropyl isomer eluted first). LCMS calculated for C18H15BrF3N4O (M+H)+: m/z=439.0. found 439.0.


Step 4. (2-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol

To a solution of 2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropane-1-carbaldehyde (60 mg, 0.14 mmol) in MeOH (1 mL), NaBH4 (10 mg, 0.27 mmol) was added slowly. The mixture was stirred at RT overnight, and then was diluted with MeOH and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% NH4OH, at flow rate of 60 mL/min) to give the desired product as a single diastereomer. LCMS calculated for C18H17BrF3N4O (M+H)+: m/z=441.1. found 441.1. 1H NMR (400 MHz, CDCl3) δ 7.72 (d, J=8.5 Hz, 1H), 7.40-7.32 (m, 2H), 7.27-7.16 (m, 3H), 6.61 (s, 1H), 5.79 (s, 1H), 4.43 (dd, J=11.6, 5.5 Hz, 1H), 4.03 (dd, J=14.1, 8.8 Hz, 1H), 3.61-3.49 (m, 2H), 1.81-1.71 (m, 1H), 1.28 (dd, J=8.7, 5.2 Hz, 1H), 0.97 (t, J=5.5 Hz, 1H).


The two possible diastereomers of the title compound are: ((1R*,2R*)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol and ((1R*,2S*)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol.


Each diastereomer can be obtained as a racemic mixture and each is separable into its enantiomers ((1R,2R)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol and ((1S,2S)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol, and ((1R,2S)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol and ((1S,2R)-2-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-2-phenylcyclopropyl)methanol, respectively.


Example 90. 4-(3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidin-1-yl)-1-methylpyrimidin-2(1H)-one



embedded image


This compound was prepared according to the procedure described in Example 46, using 4-chloro-1-methylpyrimidin-2(1H)-one instead of 4-bromo-1-methyl-1H-1,2,3-triazole as starting material. LCMS calculated for C23H19F7N7O (M+H)+: m/z=542.2. found: 542.2. 1H NMR (500 MHz, DMSO-d6) δ 9.21 (t, J=6.8 Hz, 1H), 7.96 (d, J=7.5 Hz, 1H), 7.32-7.20 (m, 2H), 7.16 (s, 1H), 7.09-7.02 (m, 2H), 5.96 (d, J=7.5 Hz, 1H), 5.92 (s, 1H), 4.82-4.71 (m, 2H), 4.58-4.43 (m, 2H), 4.13 (d, J=10.0 Hz, 2H), 3.34 (s, 3H) ppm.


Example 91. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,3r)-3-hydroxy-3-methylcyclobutyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedure described in Example 48, using (1r,3r)-3-amino-1-methylcyclobutan-1-ol instead of (1r,4r)-4-aminocyclohexan-1-ol as starting material. LCMS calculated for C24H24F7N6O2 (M+H)+: m/z=561.2. found: 561.2. 1H NMR (500 MHz, DMSO-d6) δ 9.12 (s, 1H), 7.22-7.15 (m, 2H), 7.13 (s, 1H), 7.03-6.97 (m, 2H), 6.49 (d, J=7.5 Hz, 1H), 5.86 (s, 1H), 4.69 (s, 1H), 4.28-4.23 (m, 2H), 4.19-4.09 (m, 1H), 4.02-3.96 (m, 4H), 2.18-2.10 (m, 2H), 1.90-1.83 (m, 2H), 1.22 (s, 3H) ppm.


Example 92. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(((1r,3r)-3-hydroxycyclobutyl)methyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedure described in Example 48, using (1r,3r)-3-(aminomethyl)cyclobutan-1-ol instead of (1r,4r)-4-aminocyclohexan-1-ol as starting material. LCMS calculated for C24H24F7N6O2 (M+H)+: m/z=561.2. found: 561.2. 1H NMR (500 MHz, DMSO-d6) δ 9.14 (s, 1H), 7.21-7.14 (m, 2H), 7.13 (s, 1H), 7.03-6.97 (m, 2H), 6.41 (t, J=5.8 Hz, 1H), 5.85 (s, 1H), 4.86 (d, J=6.1 Hz, 1H), 4.28-4.23 (m, 2H), 4.18-4.08 (m, 1H), 4.03-3.95 (m, 4H), 3.04-2.97 (m, 2H), 2.16-2.09 (m, 1H), 1.98-1.90 (m, 2H), 1.86-1.76 (m, 2H) ppm.


Example 93. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1S,2S)-2-hydroxycyclobutyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedure described in Example 48, using (1S,2S)-2-aminocyclobutan-1-ol instead of (1r,4r)-4-aminocyclohexan-1-ol as starting material.


LCMS calculated for C23H22F7N6O2 (M+H)+: m/z=547.2. found: 547.2. 1H NMR (600 MHz, DMSO-d6) δ 9.17 (s, 1H), 7.21-7.16 (m, 2H), 7.13 (s, 1H), 7.02-6.97 (m, 2H), 6.63 (d, J=8.3 Hz, 1H), 5.83 (s, 1H), 5.09 (d, J=7.2 Hz, 1H), 4.29-4.24 (m, 2H), 4.04-3.97 (m, 4H), 3.88-3.74 (m, 2H), 1.86 (q, J=8.8 Hz, 1H), 1.78 (q, J=9.2 Hz, 1H), 1.35-1.25 (m, 1H), 1.19 (m, 1H) ppm.


Example 94. 3-(((2-Cyclopropyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(1H-pyrazol-1-yl)azetidine-1-sulfonamide



embedded image


Step 1. N-((3-(1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


This compound was prepared according to the procedure described in Example 22 (Steps 5-7), using tert-butyl 3-(aminomethyl)-3-(1H-pyrazol-1-yl)azetidine-1-carboxylate instead of tert-butyl 4-(aminomethyl)-4-(5-fluoropyridin-2-yl)piperidine-1-carboxylate as starting material. LCMS calculated for C14H14BrF3N7 (M+H)+: m/z=416.0. found: 416.0.


Step 2. N-((3-(1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-cyclopropyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


A mixture of N-((3-(1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (50.0 mg, 0.12 mmol), potassium cyclopropyltrifluoroborate (53.3 mg, 0.36 mmol), cesium carbonate (78.0 mg, 0.24 mmol), XPhos Pd G2 (18.9 mg, 0.024 mmol) in dioxane (1 mL) and water (0.2 mL) was heated at 100° C. for 20 hrs. The reaction mixture was then cooled to r.t., diluted with EtOAc and filtered through celite. The solvent was removed and the obtained product was used directly in the next step. LCMS calculated for C17H19F3N7 (M+H)+: m/z=378.2. found: 378.2.


Step 3. 3-(((2-Cyclopropyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(1H-pyrazol-1-yl)azetidine-1-sulfonamide

A mixture of N-((3-(1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-cyclopropyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (40.0 mg, 0.106 mmol), sulfuric diamide (20.4 mg, 0.212 mmol), N,N-diisopropylethylamine (0.19 ml, 1.06 mmol) in dioxane (1 mL) was heated at 85° C. for 20 hrs. The reaction mixture was cooled to r.t. and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to obtain desired product. LCMS calculated for C17H20F3N8O2S (M+H)+: m/z=457.1. found: 457.2.


Example 95. 3-(((2-Cyano-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(1H-pyrazol-1-yl)azetidine-1-sulfonamide



embedded image


Step 1. 7-(((3-(1H-pyrazol-1-yl)azetidin-3-yl)methyl)amino)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine-2-carbonitrile



embedded image


A mixture of N-((3-(1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (50.0 mg, 0.12 mmol), potassium ferrocyanide(II) hydrate (60.9 mg, 0.144 mmol), potassium acetate (4.7 mg, 0.048 mmol), t-BuXphos Pd G3 (19.1 mg, 0.024 mmol) in dioxane (1 mL) and water (1 mL) was heated at 120° C. in microwave for 1 hr. The reaction mixture was then cooled to r.t., diluted with EtOAc and filtered through celite. The solvent was removed and the obtained product was used directly in the next step. LCMS calculated for C15H14F3N8 (M+H)+: m/z=363.1. found: 363.2.


Step 2. 3-(((2-Cyano-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(H-pyrazol-1-yl)azetidine-1-sulfonamide

This compound was prepared according to the procedure described in Example 94 (Step 3), using 7-(((3-(1H-pyrazol-1-yl)azetidin-3-yl)methyl)amino)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine-2-carbonitrile instead of N-((3-(1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-cyclopropyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine as starting material. LCMS calculated for C15H15F3N9O2S (M+H)+: m/z=442.1. found: 442.1.


Example 96. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide



embedded image


Step 1. 2-Bromo-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


To a solution of tert-butyl 3-(((2-bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (600 mg, 1.102 mmol) in 5 mL DCM was added 3 mL TFA and the resulting solution was stirred at r.t. for 1 hr. Solvent was then removed to obtain desired product. LCMS calculated for C17H15BrF4N5 (M+H)+: m/z=444.0. found: 444.0.


Step 2. 3-(((2-Bromo-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide

A solution of 2-bromo-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine (70.0 mg, 0.158 mmol), 4-nitrophenyl ((1r,4r)-4-hydroxycyclohexyl)carbamate (66.2 mg, 0.236 mmol) and N,N-diisopropylethylamine (0.14 ml, 0.79 mmol) in 3 mL dioxane was heated at 85° C. for 1 hr. The reaction mixture was cooled to r.t. and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to obtain desired product. LCMS calculated for C24H26BrF4N6O2 (M+H)+: m/z=585.1. found: 585.1. 1H NMR (500 MHz, DMSO-d6) δ 8.99 (t, J=6.8 Hz, 1H), 7.20-7.15 (m, 2H), 7.03-6.96 (m, 2H), 6.78 (s, 1H), 6.09 (d, J=7.9 Hz, 1H), 5.75 (s, 1H), 4.25-4.19 (m, 2H), 3.99-3.92 (m, 4H), 3.36-3.23 (m, 2H), 1.82-1.75 (m, 2H), 1.72-1.65 (m, 2H), 1.22-1.11 (m, 4H) ppm.


Example 97. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(6-methylpyridin-3-yl)azetidine-1-sulfonamide



embedded image


Step 1. tert-Butyl 3-cyano-3-(6-methylpyridin-3-yl)azetidine-1-carboxylate



embedded image


To a solution of tert-butyl 3-cyanoazetidine-1-carboxylate (400 mg, 2.195 mmol), 5-bromo-2-methylpyridine (566 mg, 3.29 mmol), Pd2(dba)3 (201 mg, 0.22 mmol) and N-Xantphos (242 mg, 0.439 mmol) in 10 mL THF was added a solution of LiHMDS (3.29 mL, 3.29 mmol, 1M in THF). The resulting solution was stirred at r.t. for 10 min then heated to 80° C. for 20 hrs. The reaction mixture was then cooled to r.t., diluted with EtOAc and filtered through celite. Solvent was removed and the crude was purified by Combi-Flash (EtOAc/Hexane) to obtain desired product. LC-MS calculated for C15H20N3O2 (M+H)+: m/z=274.2. found 274.1.


Step 2. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(6-methylpyridin-3-yl)azetidine-1-sulfonamide

This compound was prepared according to the procedure described in Example 59 (Steps 2-5), using tert-butyl 3-cyano-3-(6-methylpyridin-3-yl)azetidine-1-carboxylate instead of tert-butyl 3-cyano-3-(5-fluoropyridin-2-yl)azetidine-1-carboxylate as starting material. LCMS calculated for C18H18F6N7O2S (M+H)+: m/z=510.1. found: 510.1.


Example 98. 3-(((2-(Difluoromethyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedure described in Example 62, using 4-nitrophenyl ((1r,4r)-4-hydroxycyclohexyl)carbamate instead of 4-nitrophenyl (methyl-d3)carbamate as starting material. LCMS calculated for C25H27F6N6O2 (M+H)+: m/z=557.2. found: 557.3. 1H NMR (600 MHz, DMSO-d6) δ 9.03 (s, 1H), 7.27 (s, 1H), 7.21-7.15 (m, 2H), 7.03-6.98 (m, 2H), 6.87 (s, 1H), 6.11 (d, J=8.0 Hz, 1H), 5.81 (s, 1H), 4.49 (d, J=4.4 Hz, 1H), 4.27-4.20 (m, 2H), 4.00-3.95 (m, 4H), 3.35-3.25 (m, 2H), 1.81-1.75 (m, 2H), 1.71-1.65 (m, 2H), 1.22-1.11 (m, 4H) ppm.


Example 99. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(5-fluoropyridin-2-yl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedure described in Example 61, using 4-nitrophenyl ((1r,4r)-4-hydroxycyclohexyl)carbamate instead of 4-nitrophenyl (methyl-d3)carbamate as starting material. LCMS calculated for C24H25F7N7O2 (M+H)+: m/z=576.2. found: 576.2. 1H NMR (500 MHz, DMSO-d6) δ 9.15 (t, J=6.7 Hz, 1H), 8.43 (d, J=2.9 Hz, 1H), 7.59 (m, 1H), 7.42 (dd, J=8.8, 4.3 Hz, 1H), 7.15 (s, 1H), 6.10 (d, J=7.9 Hz, 1H), 5.87 (s, 1H), 4.21-4.17 (m, 2H), 4.10-4.03 (m, 4H), 3.26-3.22 (m, 2H), 1.81-1.74 (m, 2H), 1.71-1.64 (m, 2H), 1.22-1.11 (m, 4H) ppm.


Example 100. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(3,4-difluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide



embedded image


Step 1. N-((3-(3,4-difluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


This compound was prepared according to the procedure described in Example 30 (Steps 1-3), using (3,4-difluorophenyl)boronic acid instead of (4-fluorophenyl)boronic acid as starting material. LCMS calculated for C18H14F8N5 (M+H)+: m/z=452.1. found: 452.0.


Step 2. 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(3,4-difluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide

This compound was prepared according to the procedure described in Example 98, using N-((3-(3,4-difluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine instead of 2-(difluoromethyl)-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine as starting material. LCMS calculated for C25H25F8N6O2 (M+H)+: m/z=593.2. found: 593.3. 1H NMR (600 MHz, DMSO-d6) δ 9.14 (s, 1H), 7.39-7.33 (m, 1H), 7.22-7.11 (m, 2H), 6.89 (m, 1H), 6.12 (d, J=7.9 Hz, 1H), 5.98 (s, 1H), 4.52-4.48 (m, 1H), 4.22 (d, J=8.3 Hz, 2H), 4.02 (s, 2H), 3.96 (d, J=8.4 Hz, 2H), 3.40-3.31 (m, 2H), 1.81-1.74 (m, 2H), 1.71-1.63 (m, 2H), 1.22-1.09 (m, 4H) ppm.


Example 101. 3-(4-Fluorophenyl)-N-(methyl-d3)-3-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedure described in Example 61, using N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine instead of N-((3-(5-fluoropyridin-2-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine as starting material. LCMS calculated for C20H17D3F4N5O (M+H)+: m/z=425.2. found: 425.1.


Example 102. 3-(4-Fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)-3-(((2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedure described in Example 98, using N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)imidazo[1,2-a]pyridin-5-amine instead of 2-(difluoromethyl)-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine as starting material. LCMS calculated for C25H28F4N5O2 (M+H)+: m/z=506.2. found: 506.1.


Example 103. 3-(4-Fluorophenyl)-N-(methyl-d3)-3-(((2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-yl)amino)methyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedure described in Example 61, using N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine instead of N-((3-(5-fluoropyridin-2-yl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine as starting material. LCMS calculated for C19H16D3F4N6O (M+H)+: m/z=426.2. found: 426.4.


Example 104. 5-Chloro-7-(((4-phenylpiperidin-4-yl)methyl)amino)pyrazolo[1,5-a]pyrimidine-2-carbonitrile



embedded image


Step 1. 2-Bromo-5,7-dichloropyrazolo[1,5-a]pyrimidine



embedded image


A mixture of 3-bromo-1H-pyrazol-5-amine (1.5 g, 9.3 mmol), diethyl malonate (15 mL, 98 mmol), and sodium ethoxide (1.9 g, 28 mmol) was heated to 100° C. and stirred for 16h. Upon completion the suspension was filtered, and the precipitate was washed with Et2O. This solid was then taken up in phenylphosphonic dichloride (10 mL, 71 mmol) and heated to 120° C. for 20 h. The crude reaction mixture was then concentrated in vacuo. The resulting residue was washed with DCM and saturated aqueous NaHCO3. The organic layers were then concentrated in vacuo and purified by Biotage Isolara (hexane/EtOAc, 0-100% EtOAc) to provide the desired product. LCMS calculated for C6H3BrCl2N3 (M+H)+: m/z=265.9, 267.9, 269.9. found: 265.9, 267.9, 269.9.


Step 2. 4-(((2-Bromo-5-chloropyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-methyl-4-phenylpiperidine-1-carboxamide



embedded image


This compound was prepared according to the procedures described in Example 13, with 2-bromo-5,7-dichloropyrazolo[1,5-a]pyrimidine replacing 5-chloro-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridine in Step 1, LCMS calculated for C20H23BrClN6O (M+H)+: m/z=477.1, 479.1. found: 477.1, 479.1


Step 3. 5-Chloro-7-(((4-phenylpiperidin-4-yl)methyl)amino)pyrazolo[1,5-a]pyrimidine-2-carbonitrile

A solution of 4-(((2-bromo-5-chloropyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-methyl-4-phenylpiperidine-1-carboxamide (20 mg, 0.042 mmol) and copper (I) cyanide (7.5 mg, 0.84 mmol) in NMP (0.4 mL) was heated to 120° C. for 18 h. Upon completion (loss of urea also occurs) the reaction was cooled to rt, the residue was diluted with acetonitrile and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the desired product. LCMS calculated for C19H20ClN6 (M+H)+: m/z=367.1, 369.1. found: 367.2, 369.2.


Example 105. 3-(((5-(Dimethylamino)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-sulfonamide



embedded image


Step 1. N7-((3-(4-Fluorophenyl)azetidin-3-yl)methyl)-N5,N5-dimethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine-5,7-diamine



embedded image


A mixture of tert-butyl 3-(((5-chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (Example 66, Step 2) (10 mg, 0.2 mmol), dimethylamine (2M in THF, 100 μL, 0.2 mmol), and cesium carbonate (20 mg, 0.06 mmol) was heated to 80° C. and stirred for 16 h. Upon completion the suspension was filtered, and the filtrate concentrated in vacuo, then taken up in 0.5 mL of TFA and 1.5 mL DCM and stirred at rt for 1 h. Upon Boc deprotection the crude mixture was concentrated in vacuo and used directly for the next step. LCMS calculated for C19H21F4N6 (M+H)+: m/z=409.2. found: 409.2.


Step 2. 3-(((5-(Dimethylamino)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-sulfonamide

This compound was prepared according to the procedures described in Example 11, with N7-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-N′,N′-dimethyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine-5,7-diamine replacing N-((3-(4-fluoro-1H-pyrazol-1-yl)azetidin-3-yl)methyl)-2-(trifluoromethyl)-[1,2,4]triazolo[1,5-a]pyridin-5-amine, LCMS calculated for C19H22F4N7O2S (M+H)+: m/z=488.2. found: 488.2


Example 106. 3-(4-Fluorophenyl)-3-(((5-methoxy-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-sulfonamide



embedded image


This compound was prepared according to the procedures described in Example 105, with methanol replacing dimethylamine in step 1, LCMS calculated for C18H19F4N6O3S (M+H)+: m/z=475.1. found: 475.2


Example 107. 3-(((5-Cyclopropyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-(dimethylcarbamoyl)phenyl)-N-(methyl-d3)azetidine-1-carboxamide



embedded image


Step 1. tert-Butyl 3-(aminomethyl)-3-(4-(dimethylcarbamoyl)phenyl)azetidine-1-carboxylate



embedded image


This compound was prepared according to the procedures described in Example 30, Steps 1 and 2 with (4-(dimethylcarbamoyl)phenyl)boronic acid replacing (4-fluorophenyl)boronic acid in Step 1, LCMS calculated for C14H20N3O3 (M-Boc+H)+: m/z=278.2. found: 278.1


Step 2. 3-(((5-Cyclopropyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-(dimethylcarbamoyl)phenyl)-N-(methyl-d3)azetidine-1-carboxamide

This compound was prepared according to the procedures described in Example 69, with methyl-d3-amine replacing (1r,4r)-4-aminocyclohexan-1-ol in Step 1, and tert-butyl 3-(aminomethyl)-3-(4-(dimethylcarbamoyl)phenyl)azetidine-1-carboxylate replacing tert-butyl 3-(aminomethyl)-3-(4-fluorophenyl)azetidine-1-carboxylate. LCMS calculated for C25H26D3F3N7O2 (M+H)+: m/z=519.3. found: 519.3.


Example 108. 3-(((5-Cyclobutyl-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(methyl-d3)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedures described in Example 69, with methyl-d3-amine replacing (1r,4r)-4-aminocyclohexan-1-ol in Step 1, and ethyl 3-cyclobutyl-3-oxopropanoate replacing ethyl 3-oxobutanoate. LCMS calculated for C23H22D3F4N6O (M+H)+: m/z=480.2. found: 480.3.


Example 109. 3-(4-Fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-N-(2,2,2-trifluoroethyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedures described in Example 66, with tert-butyl 3-(4-fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxylate (Example 70, Step 2) replacing tert-butyl 3-(((5-chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate in Step 3, and 1,1,1-trifluoro-2-isocyanatoethane replacing phenyl carbamate in the final step. LCMS calculated for C21H17D3F7N6O (M+H)+: m/z=508.2. found: 508.1 1H NMR (600 MHz, DMSO) δ 8.43 (s, 1H), 7.27 (dd, J=8.4, 5.3 Hz, 2H), 7.10 (q, J=7.7 Hz, 3H), 6.75 (s, 1H), 5.70 (s, 1H), 4.31 (s, 1H), 4.05 (d, J=8.4 Hz, 2H), 3.84 (d, J=6.7 Hz, 2H), 3.79-3.70 (m, 2H).


Example 110. N-(2,2-Difluoroethyl)-3-(4-fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxamide



embedded image


Step 1. 4-Nitrophenyl (2,2-difluoroethyl)carbamate



embedded image


This compound was prepared according to the procedures in Example 69, Step 1 with 2,2-difluoroethan-1-amine replacing (1r,4r)-4-aminocyclohexan-1-ol.


Step 2. N-(2,2-Difluoroethyl)-3-(4-fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxamide

This compound was prepared according to the procedures described in Example 66, with tert-butyl 3-(4-fluorophenyl)-3-(((5-(methyl-d3)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)azetidine-1-carboxylate (Example 70, Step 2) replacing tert-butyl 3-(((5-chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate in Step 3, and 4-nitrophenyl (2,2-difluoroethyl)carbamate replacing phenyl carbamate in the final step. LCMS calculated for C21H18D3F6N6O (M+H)+: m/z=490.2. found: 490.2. 1H NMR (600 MHz, DMSO) δ 8.69 (s, 1H), 7.29-7.24 (m, 2H), 7.11 (t, J=8.9 Hz, 2H), 6.84 (t, J=6.0 Hz, 1H), 6.80 (s, 1H), 6.02-5.72 (m, 2H), 4.29 (d, J=8.4 Hz, 2H), 4.04 (d, J=8.4 Hz, 2H), 3.86 (d, J=6.7 Hz, 2H), 3.34 (m, 2H).


Example 111. 3-(((5-(Ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide



embedded image


Step 1. (Ethyl-d5)magnesium bromide



embedded image


To a dry round bottom flask equipped with an air condenser was added magnesium turnings (66 mg, 2.7 mmol), one small crystal of iodine, and 2 mL of THF. This suspension was stirred vigorously at rt and 1-bromoethane-1,1,2,2,2-d5 (228 mg, 2 mmol) was added dropwise in several portions over 10 min. This was allowed to stir at ambient temperature for 1 hr. Upon completion the solution was used directly for the next step.


Step 2. tert-Butyl 3-(((5-(ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate



embedded image


This compound was prepared according to the procedures described in Example 70 Step 1 with (ethyl-d5)magnesium bromide replacing (methyl-d3)magnesium iodide. LCMS calculated for C24H23D5F4N5O2 (M+H)+: m/z=499.3. found: 499.3.


Step 3. 3-(((5-(Ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxamide

This compound was prepared according to the procedures described in Example 66, with tert-butyl 3-(((5-(ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate replacing tert-butyl 3-(((5-chloro-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate in Step 3. LCMS calculated for C20H16D5F4N6O (M+H)+: m/z=442.2. found: 442.2. 1H NMR (500 MHz, DMSO) δ 8.59 (s, 1H), 7.28-7.20 (m, 2H), 7.07 (t, J=8.8 Hz, 2H), 6.78 (s, 1H), 5.92 (s, 2H), 5.65 (s, 1H), 4.24 (d, J=8.3 Hz, 2H), 4.00 (d, J=8.3 Hz, 2H), 3.90 (d, J=6.7 Hz, 2H).


Example 112. 3-(((5-(Ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide



embedded image


Step 1. 5-(Ethyl-d5)-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine



embedded image


A solution of tert-butyl 3-(((5-(ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (Example 111, Step 2) (50 mg, 0.10 mmol) and trifluoroacetic acid (150 μl, 2 mmol) in DCM (0.5 mL) stirred at rt for 1 h. Upon completion the crude mixture was concentrated in vacuo and used directly for the next step. LCMS calculated for C19H15D5F4N5 (M+H)+: m/z=399.2. found: 399.1.


Step 2. 4-Nitrophenyl 3-(((5-(ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate



embedded image


This compound was prepared according to the procedures described in Example 49, Step 2 with 5-(ethyl-d5)-N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine replacing N-((3-(4-fluorophenyl)azetidin-3-yl)methyl)-2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-amine LCMS calculated for C26H18D5F4N6O4 (M+H)+: m/z=564.2. found: 564.2.


Step 3. 3-(((5-(Ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide

A solution of 4-nitrophenyl 3-(((5-(ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-carboxylate (40 mg, 0.071 mmol) and (1r,4r)-4-aminocyclohexan-1-ol (17 mg, 0.14 mmol) in NMP (0.7 mL) was heated to 80° C. for 18 h. Upon completion the reaction was cooled to rt, the residue was diluted with acetonitrile and purified by prep-LCMS (XBridge C18 column, eluting with a gradient of acetonitrile/water containing 0.1% TFA, at flow rate of 60 mL/min) to afford the desired product. LCMS calculated for C26H26D5F4N6O2 (M+H)+: m/z=540.3. found: 540.3. 1H NMR (600 MHz, DMSO) δ 8.58 (s, 1H), 7.26-7.20 (m, 2H), 7.08 (dd, J=10.0, 7.6 Hz, 2H), 6.79 (s, 1H), 6.08 (d, J=8.0 Hz, 1H), 5.70 (s, 1H), 4.22 (d, J=8.3 Hz, 2H), 3.96 (d, J=8.3 Hz, 2H), 3.87 (d, J=6.7 Hz, 2H), 3.44-3.22 (m, 2H), 1.76 (dd, J=9.9, 4.1 Hz, 2H), 1.67-1.61 (m, 2H), 1.15 (m, 4H).


Example 113. 3-(((5-(Ethyl-d5)-2-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1R,3R)-3-hydroxycyclopentyl)azetidine-1-carboxamide



embedded image


This compound was prepared according to the procedures described in Example 112, with (1R,3R)-3-aminocyclopentan-1-ol replacing (1r,4r)-4-aminocyclohexan-1-ol in Step 3. LCMS calculated for C25H24D5F4N6O2 (M+H)+: m/z=526.3. found: 526.3. 1H NMR (500 MHz, DMSO) δ 8.50 (s, 1H), 7.27-7.19 (m, 2H), 7.12-7.04 (m, 2H), 6.78 (s, 1H), 6.20 (d, J=7.7 Hz, 1H), 5.65 (s, 1H), 4.22 (dd, J=8.3, 1.8 Hz, 2H), 4.13 (m, 1H), 4.04 (m, 1H), 3.97 (d, J=8.3 Hz, 2H), 3.88 (d, J=6.7 Hz, 2H), 1.95-1.78 (m, 2H), 1.70 m, 1H), 1.50 (m, 1H), 1.43-1.23 (m, 2H).


Example A. MRGPRX2 FLIPR Calcium Mobilization Assay

Chinese hamster ovary (CHO-K1) cells stably expressing human MRGPRX2 were purchased from Genescript (Piscataway, NJ). The cells were maintained in culture medium (Ham's F-12K) containing 10% (v/v) FBS, 200 μg/mL Zeocin, 100 units/mL penicillin G and 100 μg/mL streptomycin (Life Technologies, Carlsbad, CA). For the assay, the cells were harvested and resuspended with culture medium without Zeocin before plating at 8000 cells per well in 20 μL in 384-well black clear bottom cell culture plates (VWR, Radnor, PA). After 24 hour culture at 37° C. and 5% CO2, the cells were loaded with 20 μL/well of calcium dye (FLIPR Calcium 6 Assay Kit, Molecular Devices, San Jose, CA) diluted in loading buffer (1× Hank's Balanced Salt Solution (HBSS), 5 mM probenecid, 20 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES), pH 7.4, VWR, Radnor, PA) followed with 45 min. incubation at 37° C. and 40 min. at r.t. in the dark. Cells were then treated with the addition of 10 uL/well test compounds with a range of increasing concentrations diluted in the assay buffer (1×HBSS, 20 mM HEPES, pH 7.4, VWR, Radnor, PA) with FLIPR Penta (Molecular Devices. San Jose, CA) and incubated for 30 min. at r.t. Ca2+ release was measured with FLIPR Penta with 10 seconds basal fluorescence measurement, then 12.5 μL of 5× agonist Cortistatin-14 (Tocris, Minneapolis, MN) at final concentration corresponding to the EC80 were added followed with continued fluorescence signal monitoring for an additional 110 seconds. The base line adjusted (median of first 10s base line) max value of the Relative Fluorescence Unit (RFU) was plotted against compound concentrations. Wells with no compound were served as the positive controls, and wells with high concentration of reference antagonist were used as negative controls. IC50 curves were globally fitted with 3- or 4-parameter Hill equation in a Genedata Screener (Genedata Basel, Switzerland).


Example B: MRGPRX2 IP1 HTRF Assay

This assay measures compound inhibition of myo-Inositol 1 phosphate (IP1) accumulation in CHO-K1 cells. Chinese hamster ovary (CHO-K1) cells stably expressing human MRGPRX2 were purchased from GenScript (Piscataway, NJ).


For the HTRF IP1 determination, an IP-One GqKit (Perkin Elmer Cisbio, Waltham, MA), which includes all reagents and buffers in this protocol, was used. The cells were maintained in culture medium (Ham's F-12K) containing 10% (v/v) FBS, and 200 μg/mL Zeocin. For the assay, the cells were harvested and resuspended in culture medium with 2% FBS and without Zeocin, then were passed through a 40 μm filter. Cells were added at 20000 cells in a 5 μL/well to a 384-well white small volume cell culture plate (Greiner VWR, Radnor, PA) which contained 50 nL/well of test compound serially diluted at a selected concentration range in DMSO. 5 μL/well of prepared 2× Stimulation Buffer2 was then added to plates and was incubated at 37° C. with 5% CO2 for 1 h. 5 μL/well of agonist Cortistatin 14 (Bio-TechneR&D Systems, Minneapolis, MN) in 1× Stimulation Buffer2, for a 1 μM final concentration, was then added to plates and incubated at 37° C. with 5% CO2 for 1 h. 6 μL/well of 1:20 diluted Detection Reagent Mix (d2 and Cryptate) was added to the plates and incubated for 1 h in the dark. Plates were read on the PHERAstar microplate reader (BMG Labtech Cary, NC) to determine the HTRF ratio of the acceptor and donor emission signals. The IP1 concentrations were calculated from a standard curve following the IP-One GQ kit instructions. Wells with DMSO only and wells with high concentration of reference antagonist were used as controls for normalization. Compound IC50 curves were globally fitted with 3- or 4-parameter Hill equation in a Genedata Screener (Genedata Basel, Switzerland).


Example C: MRGPRX2 P-Arrestin Assay

This assay measures compound inhibition of P-Arrestin recruitment, which is part of the G protein-independent pathway that results from the ligand-activated GPCR phosphorylation by specific GPCR kinases. The PathHunter CHO-K1 MRPGRX2 B-Arrestin Cell line stably expressing ProLink tagged MRGPRX2 and Enzyme Acceptor Tagged B-Arrestin was purchased from Eurofins DiscoverX, (Fremont, CA).


The cells were maintained in culture medium from the Europhins DiscoverX Cell Culture Kit-107 which includes FBS, hygromycin B and G418. For the assay, the cells were harvested and were resuspended with Cell Plating Reagent 2 (Eurofins DiscoverX). Cells were added 10000 cells in 25 μL/well to a 384-well black cell culture plate (Greiner VWR, Radnor, PA) which contained 125 nL/well of compound at a selected serially diluted concentration range or DMSO. The plate was incubated at 37° C. with 5% CO2 for 1 h. 2 μL/well of agonist Cortistatin 14 was diluted in Protein Dilution Buffer, (Eurofins DiscoverX) for a final concentration of 0.25 μM, was added and the plate was incubated at 37° C. with 5% CO2 for 90 min. 14 μL/well of Detection Reagent Mix (PathHunter Detection Kit, Eurofins DiscoverX) was added to the plates and further incubated for 1 h in the dark. Plates were read on the PHERAstar microplate reader (BMG Labtech Cary, NC) measuring luminescence 0.1 to 1 second per well. Data was normalized using DMSO only wells and wells with high concentration of reference antagonist as controls. Compound IC50 curves were globally fitted with 3- or 4-parameter Hill equation in a Genedata Screener (Genedata Basel, Switzerland).


Table of Assay Data for the Assays of Examples A-C

Data obtained for the compounds of the Examples in the assays of Examples A (MRGPRX2 FLIPR), B (“IP1 HTRF”) and C (“MRGPRX2 P-Arrestin”) are shown in Table A. The symbol “++++” indicates an IC50 value of <0.1 nM; “+++” indicates an IC50 value of >0.1 nM but ≤100 nM; the symbol “++” indicates an IC50 value of >100 nM but ≤1000 nM; the symbol “+” indicates an IC50 value of >1000 nM. ND indicates no data.














TABLE A








MRGPRX2
MRGPRX2
MRGPRX2



Example
FLIPR
IP1 HTRF
β-Arrestin





















1
ND
++
ND



2
ND
++
ND



3
+++
+++
++



4
+++
+++
+++



5
+++
+++
ND



6
+++
+++
+++



7
+++
++
ND



8
+++
++
+++



9
+++
+++
+++



10
+++
+++
ND



11
+++
+++
ND



12
+++
++
ND



13
+++
+++
ND



14
+++
++
ND



15
+++
+++
ND



16
+++
+++
ND



17
+++
+++
ND



18
+++
+++
+++



19
+++
+++
ND



20
+++
+++
+++



21
+++
+++
+++



22
+++
+++
ND



23
++
++
ND



24
+++
++
ND



25
ND
+++
+++



26
+++
+++
ND



27
+++
+++
ND



28
+++
+++
ND



29
+++
++
ND



30
+++
+++
+++



31
+++
+++
+++



32
+++
+++
+++



33
+++
+++
+++



34
+++
+++
ND



35
+++
+++
ND



36
+++
+++
+++



37
+++
+++
+++



38
+++
+++
ND



39
+++
+++
+++



40
+++
ND
+++



41
ND
+++
+++



42
ND
+++
+++



43
+++
+++
+++



44
+++
+++
+++



45
+++
+++
+++



46
+++
ND
+++



47
+++
ND
+++



48
+++
+++
+++



49
+++
+++
+++



50
+++
ND
+++



51
ND
ND
+++



52
ND
+++
+++



53
ND
+++
+++



54
ND
+++
+++



55
ND
+++
+++



56
ND
+++
+++



57
+++
+++
+++



58
+++
+++
+++



59
+++
+++
+++



60
+++
ND
+++



61
+++
+++
+++



62
+++
+++
+++



63
+++
ND
+++



64
ND
ND
+++



65
ND
+++
+++



66
+++
+++
+++



67
+++
+++
+++



68
+++
+++
+++



69
ND
+++
+++



70
ND
ND
+++



71
ND
+++
+++



72
ND
ND
+++



73
ND
+++
+++



74
ND
+++
+++



75
ND
ND
+++



76
ND
ND
+++



77
+++
++
ND



78
++
ND
ND



79
ND
ND
+++



80
ND
ND
+++



81
+++
ND
+++



82
+++
ND
+++



83
+++
ND
+++



84
+++
ND
+++



85
+++
ND
+++



86
ND
ND
+++



87
ND
ND
+++



88
ND
ND
+



89
ND
ND
++



90
ND
+++
+++



91
ND
+++
+++



92
ND
+++
+++



93
ND
+++
+++



94
+++
++
++



95
+++
ND
ND



96
+++
+++
+++



97
ND
ND
++



98
ND
+++
+++



99
ND
+++
+++



100
ND
+++
+++



101
ND
+++
+++



102
ND
+++
+++



103
ND
+++
+++



104
++
+
ND



105
+++
ND
ND



106
+++
+++
+++



107
ND
ND
+



108
ND
ND
++



109
ND
+++
+++



110
ND
+++
+++



111
ND
+++
+++



112
ND
+++
+++



113
ND
+++
+++










Example D: Caco2 Assay

Caco-2 cells are grown at 37° C. in an atmosphere of 500 CO2 in DMEM growth medium supplemented with 1000 (v/v) fetal bovine serum, 10% (v/v) nonessential amino acids, penicillin (100 U/*mL), and streptomycin (100 g/*mL). Confluent cell monolayers are subcultured every 7 days or 4 days for Caco-2 by treatment with 0.050% trypsin containing 1 M EDTA. Caco-2 cells are seeded in 96-well Transwell plates. The seeding density for Caco-2 cells is 14,000 cells/well. DMEM growth medium is replaced every other day after seeding. Cell monolayers are used for transport assays between 22 and 25 days for Caco-2 cells.


Cell culture medium is removed and replaced with HBSS. To measure the TEER, the HBSS is added into the donor compartment (apical side) and receiver compartment (basolateral side). The TEER is measured by using a RIMS Autosampler to ensure the integrity of the cell monolayers. Caco-2 cell monolayers with TEER values ≤300 Ω·cm2 are used for transport experiments. To determine the Papp in the absorptive direction (A-B), solution of test compound (50 M) in HBSS is added to the donor compartment (apical side), while HBSS solution with 4% BSA is added to the receiver compartment (basolateral side). The apical volume was 0.075 mL, and the basolateral volume is 0.25 mL. The incubation period is 120 min. at 37° C. in an atmosphere of 5% CO2. At the end of the incubation period, 5 samples from the donor and receiver sides are removed and an equal volume of MeCN is added for protein precipitation. The supernatants are collected after centrifugation (3000 rpm, Allegra X-14R Centrifuge from Beckman Coulter, Indianapolis, IN) for LCMS analysis. The permeability value is determined according to the equation:









P
app

(

cm
/
s

)

=


(

F
*
VD

)

/

(

SA
*
MD

)



,




where the flux rate (F, mass/time) is calculated from the slope of cumulative amounts of compound of interest on the receiver side, SA is the surface area of the cell membrane, VD is the donor volume, and MD is the initial amount of the solution in the donor chamber.


Example E: Human Whole Blood Stability

The whole blood stability of the exemplified compounds is determined by LC-MS/MS. The 96-Well Flexi-Tier™ Block (Analytical Sales & Services, Inc, Flanders, NJ) is used for the incubation plate containing 1.0 mL glass vials with 0.5 mL of blood per vial (pooled gender, human whole blood sourced from BIOIVT, Hicksville, NY or similar). Blood is pre-warmed in water bath to 37° C. for 30 min. 96-deep well analysis plate is prepared with the addition of 100 μL ultrapure water/well. 50 μL chilled ultrapure water/well is added to 96-deep well sample collection plate and covered with a sealing mat. 1 μL of 0.5 mM compound working solution (DMSO:water) is added to the blood in incubation plate to reach final concentrations of 1 μM, mixed by pipetting thoroughly and 50 μL is transferred 50 into the T=0 wells of the sample collection plate. Blood is allowed to sit in the water for 2 min. and then 400 μL stop solution/well is added (MeCN containing an internal standard). The incubation plate is placed in the Incu-Shaker CO2 Mini incubator (Benchmark Scientific, Sayreville, NJ) at 37° C. with shaking at 150 rpm. At 1, 2 and 4-h, the blood samples are mixed thoroughly by pipetting and 50 μL is transferred into the corresponding wells of the sample collection plate. Blood is allowed to sit in the water for 2 min. and then 400 μL of stop solution/well is added. The collection plate is sealed and vortexed at 1700 rpm for 3 min. (VX-2500 Multi-Tube Vortexer, VWR International, Radnor, PA), and samples are then centrifuged in the collection plate at 3500 rpm for 10 min. (Allegra X-14R Centrifuge Beckman Coulter, Indianapolis, IN). 100 μL of supernatant/well is transferred from the sample collection plate into the corresponding wells of the analysis plate. The final plate is vortexed at 1700 rpm for 1 min. and analyze samples by LC-MS/MS. The peak area ratio of the 1, 2, and 4 h samples relative to T=0 is used to determine the percent remaining. The natural log of the percent remaining versus time is used determine a slope to calculate the compounds half-life in blood (t1/2=0.693/slope).


Example F: In Vitro Intrinsic Clearance Protocol

For in vitro metabolic stability experiments, test compounds are incubated with human liver microsomes at 37° C. The incubation mixture contains test compounds (1 M), NADPH (2 mM), and human liver microsomes (0.5 mg protein/*mL) in 100 mM phosphate buffer (pH 7.4). The mixture is pre-incubated for 2 min. at 37° C. before the addition of NADPH. Reactions are commenced upon the addition of NADPH and quenched with ice-cold MeOH at 0, 10, 20, and 30 min. Terminated incubation mixtures are analyzed using LC-MS/MS system. The analytical system consisted of a Shimadzu LC-30AD binary pump system and SIL-30AC autosampler (Shimadzu Scientific Instruments, Columbia, MD) coupled with a Sciex Triple Quad 6500+ mass spectrometer from Applied Biosystems (Foster City, CA). Chromatographic separation of test compounds and internal standard is achieved using a Hypersil Gold C18 column (50×2.1 mm, 5 μM, 175 Å) from ThermoFisher Scientific (Waltham, MA). Mobile phase A consists of 0.1% formic acid in water, and mobile phase B consists of 0.1% formic acid in MeCN. The total LC-MS/MS runtime can be 2.75 min. with a flow rate of 0.75 mL/min. Peak area integrations and peak area ratio calculations are performed using Analyst software (version 1.6.3) from Applied Biosystems.


The in vitro intrinsic clearance, CLint, in vitro, is calculated from the t1/2 of test compound disappearance as CLint, in vitro=(0.693/t1/2)×(1/Cprotein), where Cprotein is the protein concentration during the incubation, and t1/2 is determined by the slope (k) of the log-linear regression analysis of the concentration versus time profiles; thus, t1/2=ln2/k. The CLint, in vitro values are scaled to the in vivo values for human by using physiologically based scaling factors, hepatic microsomal protein concentrations (45 mg protein/g liver), and liver weights (21 g/kg body weight). The equation CLint=CLint, in vitro×(*mg protein/g liver weight)×(g liver weight/kg body weight) is used. The in vivo hepatic clearance (CLH) is then calculated by using CLint and hepatic blood flow, Q (20 mL·min.−1·kg−1 in humans) in the well-stirred liver model disregarding all binding from CLH=(Q×CLint)/(Q+CLint). The hepatic extraction ratio is calculated as CLH divided by Q.


Example G: In Vivo Pharmacokinetics Protocol

For in vivo pharmacokinetic experiments, test compounds are administered to male Sprague Dawley rats or male and female Cynomolgus monkeys intravenously or via oral gavage. For intravenous (IV) dosing, test compounds are dosed at 0.5 to 1 mg/kg using a formulation of 10% dimethylacetamide (DMAC) in acidified saline via IV bolus for rat and 5 min. or 10 min. IV infusion for monkey. For oral (PO) dosing, test compounds are dosed at 1.0 to 3.0 mg/kg using 5% DMAC in 0.5% methylcellulose in citrate buffer (pH 2.5). Blood samples are collected at predose and various time points up to 24 h postdose. All blood samples are collected using EDTA as the anticoagulant and centrifuged to obtain plasma samples. The plasma concentrations of test compounds are determined by LC-MS methods. The measured plasma concentrations are used to calculate PK parameters by standard noncompartmental methods using Phoenix® WinNonlin software program (version 8.0, Pharsight Corporation).


In rats and monkeys, cassette dosing of test compounds are conducted to obtain preliminary PK parameters.


In vivo pharmacokinetic experiments with male beagle dogs may be performed under the conditions described above.


Example H: Time Dependent Inhibition (TDI) of CYP Protocol

This assay is designed to characterize an increase in CYP inhibition as a test compounds is metabolized over time. Potential mechanisms for this include the formation of a tight-binding, quasi-irreversible inhibitory metabolite complex or the inactivation of P450 enzymes by covalent adduct formation of metabolites. While this experiment employs a 10-fold dilution to diminish metabolite concentrations and therefore effects of reversible inhibition, it is possible (but not common) that a metabolite that is an extremely potent CYP inhibitor could result in a positive result.


The results are from a cocktail of CYP specific probe substrates at 4 times their Km concentrations for CYP2C9, 2C19, 2D6 and 3A4 (midazolam) using human liver microsomes (HLM). The HLMs can be pre-incubated with test compounds at a concentration 10 μM for 30 min. in the presence (+N) or absence (−N) of a NADPH regenerating system, diluted 10-fold, and incubated for 8 min. in the presence of the substrate cocktail with the addition of a fresh aliquot of NADPH regenerating system. A calibration curve of metabolite standards can be used to quantitatively measure the enzyme activity using LC-MS/MS. In addition, incubations with known time dependent inhibitors, tienilic aicd (CYP2C9), ticlopidine (CYP2C19), paroxetine (CYP2D6), and troleandomycin (CYP3A4), used as positive controls are pre-incubated 30 min. with or without a NADPH regenerating system.


The analytical system consists of a Shimadzu LC-30AD binary pump system and SIL-30AC autosampler (Shimadzu Scientific Instruments, Columbia, MD) coupled with a Sciex Triple Quad 6500+ mass spectrometer from Applied Biosystems (Foster City, CA). Chromatographic separation of test compounds and internal standard can be achieved using an ACQUITY UPLC BEH 130A, 2.1×50 mm, 1.7 m HPLC column (Waters Corp, Milford, MA). Mobile phase A consists of 0.1% formic acid in water, and mobile phase B consists of 0.1% formic acid in MeCN. The total LC-MS/MS runtime will be 2.50 min. with a flow rate of 0.9 mL/min. Peak area integrations and peak area ratio calculations are performed using Analyst software (version 1.6.3) from Applied Biosystems.


The percentage of control CYP2C9, CYP2C19, CYP2D6, and CYP3A4 activity remaining following preincubation of the compounds with NADPH is corrected for the corresponding control vehicle activity and then calculated based on 0 min. as 100%. A linear regression plot of the natural log of % activity remaining versus time for each isozyme is used to calculate the slope. The −slope is equal to the rate of enzyme loss, or the Kobs.


Example I: In Vitro Degranulation Assay-β-Hexosaminidase Release Assay

The assay is performed essentially as described by Hermans, et al., Front. Immunol., 2021, 12, 625284. Mature mast cells are cultured in StemPro-34 medium with StemPro-34 nutrient supplement (Gibco), 2 mM L-glutamine (Gibco), 100 U/mL penicillin/100 μg/mL streptomycin (Gibco), 100 ng/mL recombinant human stem cell factor (PeproTech) and 100 ng/mL recombinant human interleukin 6 (Peprotech). The cells are washed in HEPES-buffered Tyrode's solution (Thermo Fisher) containing 0.1% (w/v) bovine serum albumine (further called “releasing medium”) and seeded at 1×105 cells per well in 96-well plate in the releasing medium. Selected wells are pretreated with predefined concentrations of MRGPRX2 antagonist for 1 h at 37° C. with 5% CO2. Then, about 10 μg/mL compound 48/80 (poly-p-methoxyphenethylmethylamine) or about 30 μM of substance P is added to treated wells and equal amount of releasing medium is added into negative control wells. Following 1 h incubation, the plate is centrifuged for 5 min. and 50 μL of supernatant is collected into a new 96-well plate. In a positive control group, remaining supernatant is removed and 100 μL 0.1% Triton X-100 (v/v) is added to lyse the cell at 300 g for 5 min. After that, the plate is centrifuged and 50 μL of the lysate is also transferred into the new plate. Supernatant and lysate samples are then incubated with 50 μL of p-nitrophenyl N-acetyle-B-D-glucosaminide in 0.1 M citrate buffer pH 4-4.5 (Thermo Fisher) for 1 h at 37° C. The enzymatic reaction is stopped by the addition of 100 μL/well of 0.1 M Na2CO3 buffer pH 10 (Thermo Fisher). The absorbance is read at 405 nm. Net β-hexosaminidase release (%) is calculated as =[(stimulated release-spontaneous release)/total content in lysed cells]×100.


Example J: Evaluation of the MRGPRX2 Antagonist Effects on In Vitro-Derived Human Mast Cells

This assay evaluates the effects of example compound on preventing mast cell degranulation via the MRGPRX2 receptor. The effects of example compounds on mast cell degranulation induced by Substance P (MRGPRX2 pathway), or IgE/anti-IgE is investigated by measuring β-hexosaminidase and mast cell specific cytokine/chemokine release into the culture media. The addition of IgE/anti-IgE serves as a differentiation for the mechanism of action of the test compound.


Mature connective tissue-type mast cells (CTMCs) are plated at 50,000 cells by well in 50 μL and stimulated as follows.


IgE Sensitization (Day 0)

All CTMCs are pre-treated with 100 μM IgE before treatment to coat the FcεR1 receptors with IgE to resemble in vitro mast cells. In this study, CTMCs are sensitized 48 h prior to pre-treatment with example compounds and degranulation (Day 2).


Pre-Treatment (Day 0)

Omalizumab is added 4 h after IgE sensitization (IgE containing media is removed after 4 h and replaced with culture media containing omalizumab and kept for 48 h) and for the following 48 h before degranulation induction.


Pre-Treatment (Day 2)

Test compounds and vehicles are added 48 h post IgE sensitization/omalizumab treatment for 30 min. at Day 2 of culture.


Degranulation (Day 2+30 Mins)

The CTMCs are stimulated with Substance P, Compound 48/80, anti-IgE, or Tyrode's buffer on Day 2 of culture.


β-Hexosaminidase Assay to Assess Mast-Cell Degranulation

For the β-hexosaminidase assay, 50 μL of cell supernatant is collected in each well 45 min. after degranulation treatment. Supernatant is collected on n=3 wells per condition.


β-Hexosaminidase is a potent inflammatory mediator stored in mast cells and is released by activated mast cells. The determination of p-hexosaminidase is used to evaluate the level of mast cell degranulation. The assay is a colorimetric assay measuring the 4-nitrophenol production using a multimode plate reader. See J. Karhausen, et al., J. Clin. Invest., 2016, 126(10), 3981-98.


Cytokine Release Assay

For the cytokine release assay, 60 μL of cell supernatant is collected in each well 8 h after degranulation treatment, an anti-protease cocktail added, and frozen at −80° C. until used. Supernatant is collected on n=3 wells per condition.


Cytokine release into the culture medium is measured using a custom V-PLEX Plus Human Cytokine Kit (MesoScale Discovery): TNF-α, IL-13, GM-CSF, VEGF-A, MCP-1, IL-6, IL-4, IL-5, IL-10, and IL-8. Culture media is diluted as follows:


Plate 1: Chemokine Panel 1 (human) kit-ref K15047 (panel MCP-1)-1:10;


Plate 2: Proinflammatory Panel 1 (human) kit-ref K15049 (panel IL-4, IL-6, IL-8, IL-10, IL-13, TNF-α)-1:5;


Plate 3: Cytokine Panel 1 (human) kit-ref K15050 (panel GM-CSF, IL-5, VEGF)-1:5.


Example K: Determining the Effect of Example Compounds on Cytokine/Chemokine Release from HypoSkin Models after Subcutaneous Injection of Cetrorelix or Cortistatin-14

This assay evaluates the effect of the test compounds on cytokine/chemokine release (potentially from mast cells) upon systematic pre-treatment and treatment of HypoSkin models prior to subcutaneous injection of drugs known to degranulate mast cells and cause injection site reactions (namely, Cortistatin-14 and Cetrorelix). 74 HypoSkin models of 20 mm in diameter and 10 mm total thickness with 15/20 mm diameter silicon rings are produced from 2 donors (37 models per donor) according to standard procedures and cultured with 2 mL standard HypoSkin medium. Models are maintained in standard cell culture conditions for the whole culture duration at 37° C., 5% CO2 and water saturation, with culture medium renewed every day except during weekends.


Systematic Treatment

Vehicle control and test compounds are added to the culture media daily, from Day 0 to Day 2. The compounds remain in the culture media upon subcutaneous injections.


Subcutaneous Injection

100 μL of Cetrorelix, Cortistatin-14, or PBS are subcutaneously injected in to the models on Day 2.


End of Culture and Sampling

Culture media is collected on Days 2+8 h and Day 3 (24 h post-injection) and frozen at −80° C. Sampling of HypoSkin models is performed on Day 0 and Day 3 (24 h post-injection) and processed as follows:


½ are fixed in 10% buffered formalin and processed for paraffin wax embedding; and


½ are snap frozen and stored at −80° C.


Hematoxylin and Eosin Staining

Hematoxylin and Eosin staining is performed on one 5 μm thick skin cross section for each sample. Representative images of both the epidermis/dermis and hypodermis are taken at 40× magnification to analyze skin structure integrity and viability.


MSD Immunoassay

Cytokine release in the culture medium is measuring using the V-PLEX Plus Human Cytokine 36-Plex Kit (K15089G, MesoScale Discovery): Eotaxin, Eotaxin-3, GM-CSF, IFN-7, IL-1α, IL-1β, Il-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-8 (HA), IL-10, IL12/IL-23p40, IL-23p70, IL-13, IL-15, IL-16, IL-17a, IL-21, IL-22, IL-23, IL-27, IL-31, IP-10, MCP-1, MCP-4, MDC, MIP-1α, MIP-1β, MIP-3α, TARC, TNF-α, TNF-β, and VEGF-A.


Example L: Characterization of JAK Inhibitors MRGPRX2 Antagonists on Mast Cell Activation

The effect of the janus kinase inhibitors Povorcitinib, Ruxolitinib, and MRGPRX2 antagonists disclosed herein on the activation of mast cells in vitro is compared and characterized under different activating conditions, mimicking the in vivo disease situation. This assay determines the effect of JAK inhibition and MRGPRX2 antagonism on interference different mast cell activation pathways; the effect of JAK inhibition and MRGPRX2 antagonism in mast cells to prevent the release of mediators involved in the activation of primary human blood eosinophils and T cells.


Primary human skin mast cells (hsMC) are used to study how Povorcitinib, Ruxolitinib, and MRGPRX2 antagonists interfere with different prototype mast cell activators. The cells are obtained from individuals undergoing circumcision or breast reduction surgery by isolation from skin explants in a multi-step protocol. In vitro dose response studies using different concentration of the compounds to identify IC50 values are conducted. For these studies, hsMCs are pre-incubated in the presence or absence of the compounds for 20 min., followed by stimulation with anti-IgE (FcεRI stimulation) or cortistatin-14 (MRGPRX2 agonist). Degranulation responses are measured by determination of p-hexosaminidase after 1 h at 37° C.


Next, three different concentrations of the compounds according to the determined IC50 are employed to assess the spectrum of mast cell activating pathways inhibited by each compound. Therefore, hsMCs are pre-incubated in the presence or absence of the individual compounds for 20 min., followed by stimulation with five distinct activators: anti-IgE, corstatin-14 (CST), stem cell factor (SCF), complement peptides C3a, and C5a. In a parallel setup, hsMCs are pre-treated with various concentrations of cyclosporine A (CSA), to assess difference in efficacy to the test compounds. CSA, inhibiting the translocation of the transcription factor NFAT into the nucleus in MCs and other immune cells, is widely used to treat urticaria. As an alternative to CSA, another JAK or BTK inhibitor can be used. Degranulation responses are measured by determination of the 0-hexosaminidase after 1 h at 37° C. In addition, cell viability and surface expression of FcεRI, MRGPRX2, cKitm C3aR/C5aR are measured by flow cytometry. Mast cells from three individual mast cell preparations are used.


To assess if treatment of mast cells with Povorcitinib, Ruxolitinib, or MRGPRX2 antagonists alters their mediator release profile and thereby their ability to activate eosinophils or T cells, hsMCs are pre-treated with the compounds for 20 min., followed by stimulation with anti-IgE, CST, SCF, C3a, or C5a for 1 h, 4 h, 8 h, and 24 h. Culture supernatants are collected and added to freshly isolated human peripheral blood eosinophils and total T cells. To achieve complete T cell activation, simultaneous activation of the TCR and costimulatory receptor with antibodies targeting CD3 and CD28 together with the mast cell supernatant is performed.


Cell activation is assessed using flow cytometry by measuring upregulation of CD69 and CD63 on eosinophils, as well as level of CD69, CD154, CD25, or CD62L on T cells. In addition, cytokines secreted by the mast cells are measured using bead-based cytokine multiplex assays (45 plex). Three individual experiments are performed using mast cells and eosinophils from three individual donors.


Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference, including all patent, patent applications, and publications, cited in the present application is incorporated herein by reference in its entirety.

Claims
  • 1-169. (canceled)
  • 170. A compound, which is 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(4-hydroxycyclohexyl)azetidine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
  • 171. The compound of claim 170, which is 3-(((2,5-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(4-hydroxycyclohexyl)azetidine-1-carboxamide.
  • 172. A pharmaceutical composition comprising the compound of claim 170, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • 173. A pharmaceutical composition comprising the compound of claim 171 and a pharmaceutically acceptable carrier.
  • 174. A method of treating an MRGPRX2 dependent condition in a patient comprising administering to the patient a therapeutically effective amount of a compound or pharmaceutically acceptable salt thereof of claim 170.
  • 175. The method of claim 174, wherein the MRGPRX2 dependent condition is selected from chronic itch, senile 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 pruritus, chronic spontaneous urticaria, chronic inducible 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, urticaria, chronic inflammation, mast cell activation syndrome, multiple sclerosis, Steven Johnson's syndrome, toxic epidermal necrolysis, appendicitis, bursitis, cutaneous lupus, colitis, cystitis, phlebitis, reflex sympathetic dystrophy/complex regional pain syndrome (RSD/CRPS), rhinitis, tendonitis, tonsillitis, acne vulgaris, sinusitis, 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 eosinophilia, 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, schwannomatosis, 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 cancers, alcoholic liver disease, polycystic liver disease, liver cholangiocarcinoma, primary sclerosing cholangitis, primary biliary cholangitis, neuromyelitis optica spectrum disorder, cardiovascular disease, inflammation induced by bacterial or viral infection, inflammation associated with SARS-COV-2 infection or its variants or 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, and vasculitis.
  • 176. A compound, which is 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
  • 177. The compound of claim 176, which is 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,4r)-4-hydroxycyclohexyl)azetidine-1-carboxamide.
  • 178. A pharmaceutical composition comprising the compound of claim 176, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • 179. A pharmaceutical composition comprising the compound of claim 177 and a pharmaceutically acceptable carrier.
  • 180. A method of treating an MRGPRX2 dependent condition in a patient comprising administering to the patient a therapeutically effective amount of a compound or pharmaceutically acceptable salt thereof of claim 176.
  • 181. The method of claim 180, wherein the MRGPRX2 dependent condition is selected from chronic itch, senile 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 pruritus, chronic spontaneous urticaria, chronic inducible 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, urticaria, chronic inflammation, mast cell activation syndrome, multiple sclerosis, Steven Johnson's syndrome, toxic epidermal necrolysis, appendicitis, bursitis, cutaneous lupus, colitis, cystitis, phlebitis, reflex sympathetic dystrophy/complex regional pain syndrome (RSD/CRPS), rhinitis, tendonitis, tonsillitis, acne vulgaris, sinusitis, 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 eosinophilia, 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, schwannomatosis, 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 cancers, alcoholic liver disease, polycystic liver disease, liver cholangiocarcinoma, primary sclerosing cholangitis, primary biliary cholangitis, neuromyelitis optica spectrum disorder, cardiovascular disease, inflammation induced by bacterial or viral infection, inflammation associated with SARS-COV-2 infection or its variants or 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, and vasculitis.
  • 182. A compound, which is 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(3-hydroxycyclobutyl)azetidine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
  • 183. The compound of claim 182, which is 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-(3-hydroxycyclobutyl)azetidine-1-carboxamide.
  • 184. A pharmaceutical composition comprising the compound of claim 182, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • 185. A pharmaceutical composition comprising the compound of claim 183 and a pharmaceutically acceptable carrier.
  • 186. A method of treating an MRGPRX2 dependent condition in a patient comprising administering to the patient a therapeutically effective amount of a compound or pharmaceutically acceptable salt thereof of claim 182.
  • 187. The method of claim 186, wherein the MRGPRX2 dependent condition is selected from chronic itch, senile 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 pruritus, chronic spontaneous urticaria, chronic inducible 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, urticaria, chronic inflammation, mast cell activation syndrome, multiple sclerosis, Steven Johnson's syndrome, toxic epidermal necrolysis, appendicitis, bursitis, cutaneous lupus, colitis, cystitis, phlebitis, reflex sympathetic dystrophy/complex regional pain syndrome (RSD/CRPS), rhinitis, tendonitis, tonsillitis, acne vulgaris, sinusitis, 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 eosinophilia, 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, schwannomatosis, 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 cancers, alcoholic liver disease, polycystic liver disease, liver cholangiocarcinoma, primary sclerosing cholangitis, primary biliary cholangitis, neuromyelitis optica spectrum disorder, cardiovascular disease, inflammation induced by bacterial or viral infection, inflammation associated with SARS-COV-2 infection or its variants or 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, and vasculitis.
  • 188. A compound, which is 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,3r)-3-hydroxycyclobutyl)azetidine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
  • 189. The compound of claim 188, which is 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)-N-((1r,3r)-3-hydroxycyclobutyl)azetidine-1-carboxamide.
  • 190. A pharmaceutical composition comprising the compound of claim 188, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • 191. A pharmaceutical composition comprising the compound of claim 189 and a pharmaceutically acceptable carrier.
  • 192. A method of treating an MRGPRX2 dependent condition in a patient comprising administering to the patient a therapeutically effective amount of a compound or pharmaceutically acceptable salt thereof of claim 188.
  • 193. The method of claim 192, wherein the MRGPRX2 dependent condition is selected from chronic itch, senile 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 pruritus, chronic spontaneous urticaria, chronic inducible 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, urticaria, chronic inflammation, mast cell activation syndrome, multiple sclerosis, Steven Johnson's syndrome, toxic epidermal necrolysis, appendicitis, bursitis, cutaneous lupus, colitis, cystitis, phlebitis, reflex sympathetic dystrophy/complex regional pain syndrome (RSD/CRPS), rhinitis, tendonitis, tonsillitis, acne vulgaris, sinusitis, 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 eosinophilia, 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, schwannomatosis, 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 cancers, alcoholic liver disease, polycystic liver disease, liver cholangiocarcinoma, primary sclerosing cholangitis, primary biliary cholangitis, neuromyelitis optica spectrum disorder, cardiovascular disease, inflammation induced by bacterial or viral infection, inflammation associated with SARS-COV-2 infection or its variants or 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, and vasculitis.
  • 194. A compound, which is 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-sulfonamide, or a pharmaceutically acceptable salt thereof.
  • 195. The compound of claim 194, which is 3-(((2,5-Bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)methyl)-3-(4-fluorophenyl)azetidine-1-sulfonamide.
  • 196. A pharmaceutical composition comprising the compound of claim 194, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • 197. A pharmaceutical composition comprising the compound of claim 195 and a pharmaceutically acceptable carrier.
  • 198. A method of treating an MRGPRX2 dependent condition in a patient comprising administering to the patient a therapeutically effective amount of a compound or pharmaceutically acceptable salt thereof of claim 194.
  • 199. The method of claim 198, wherein the MRGPRX2 dependent condition is selected from chronic itch, senile 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 pruritus, chronic spontaneous urticaria, chronic inducible 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, urticaria, chronic inflammation, mast cell activation syndrome, multiple sclerosis, Steven Johnson's syndrome, toxic epidermal necrolysis, appendicitis, bursitis, cutaneous lupus, colitis, cystitis, phlebitis, reflex sympathetic dystrophy/complex regional pain syndrome (RSD/CRPS), rhinitis, tendonitis, tonsillitis, acne vulgaris, sinusitis, 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 eosinophilia, 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, schwannomatosis, 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 cancers, alcoholic liver disease, polycystic liver disease, liver cholangiocarcinoma, primary sclerosing cholangitis, primary biliary cholangitis, neuromyelitis optica spectrum disorder, cardiovascular disease, inflammation induced by bacterial or viral infection, inflammation associated with SARS-COV-2 infection or its variants or 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, and vasculitis.
Provisional Applications (2)
Number Date Country
63533447 Aug 2023 US
63570350 Mar 2024 US