NLRP3 ACTIVATORS FOR USE IN THE TREATMENT OF INFECTIOUS DISEASES OR CANCER BY ACTIVATING NLRP3 INFLAMMASOME

Abstract
The present invention relates to a compound or pharmaceutically acceptable salts thereof, that modulates NLRP3 in that the NLRP3 inflammasome is activated. This invention further relates to the compounds and/or compositions for use in the prevention and treatment of a condition, disease or a disorder by activating the NLRP3 inflammasome, wherein the disease is selected from a group comprising infectious diseases, or cancer. Moreover, the invention relates to the use of the present compounds for analyzing a the activity of NLRP3 activation.
Description
TECHNICAL FIELD

The present invention relates to compounds or pharmaceutically acceptable salts thereof, that activate the NLRP3 inflammasome. Moreover, TLR7 and/or TLR8 is not activated by these compounds. Moreover, the mitochondrial respiratory chain is not inhibited by these compounds. This invention further relates to the compounds and/or compositions for use in the prevention and treatment of a condition, a disease or a disorder by activating the NLRP3 inflammasome, wherein the disease is selected from a group comprising infectious diseases or cancer. Moreover, the invention relates to the use of the present compounds as an adjuvant for vaccination. Moreover, the invention relates to the use of the present compounds for analyzing the mechanisms and consequences of NLRP3 activation.


BACKGROUND OF THE INVENTION

The present invention provides compounds useful as modulators of the NLRP3 inflammasome. NLRP (Nucleotide-binding oligomerization domain, Leucine rich Repeat and Pyrin domain containing) proteins, are a subfamily of NLRs (NOD-like receptor), which play a role in the formation of inflammasomes.


Inflammasomes are intracellular multiprotein oligomers of the innate immune system responsible for the activation of inflammatory responses. Moreover, inflammasomes control the bioactivity of potent pro-inflammatory cytokines of the interleukin-1 (IL-1) family members such as IL-1β and induce a form of lytic, proinflammatory cell death known as pyroptosis. These effects of inflammasome activity are mediated by one of its components, the protease caspase-1. It directly mediates maturation of inactive pro-IL-1β and controls IL-1 release and subsequent pyroptosis by its pore-forming substrate Gasdermin-D (GSDMD). The inflammasome/IL-1 axis promotes protective inflammatory responses in the context of infection, but its dysregulation can lead to pathological inflammation. The different inflammasomes respond to a variety of endogenous and exogenous danger signals and thus contribute significantly to the defense against pathogens. Most inflammasomes are triggered by the presence of specific pathogen components within the cytoplasm, either by direct binding or by detecting their activity. Instead, the NLRP3 inflammasome appears to respond to cellular stress signals triggered by structurally diverse pathogens, endogenous danger signals, and environmental irritants.


NLRP3 is expressed predominantly in certain immune cells such as in macrophages as well as in epithelial cells. NLRP3 is a cytoplasmic sensor protein responding to sterile endogenous danger signals that are typically products of damaged cells such as extracellular ATP and crystalline uric acid. Upon activation, it nucleates an inflammasome by connecting to the protease caspase-1 via the adaptor protein ASC (encoded by PYCARD), thereby triggering an immune response. While other inflammasome nucleating receptors mainly react to pathogen-derived signals, NLRP3 is the main contributor to sterile inflammation.


A targeted activation of NLRP3 can induce an inflammasome-dependent anti-cancer immune response, e.g. by increasing the activity of cytotoxic cells against tumor cells. Thus, NLRP3 activators may be used for the treatment of malignancies.


Moreover, many of the currently known NLRP3 activators have additional unspecific or unrelated activities that are unfavorable. For example, bacterial ionophore toxins such as nigericin that allow potassium efflux thought to be involved in NLRP3 activation integrate into cellular membranes of any cell type. Furthermore, known small molecule NLRP3 activators such as the imidazoquinoline compounds imiquimod (also known as R837) or CL097 also activate toll-like receptor-7 (TLR7) and/or TLR8. TLR7/8 activation mechanistically explains the production of interferons and other proinflammatory cytokines like TNF and IL-6 and the antiviral activity triggered by this class of molecules, and it is classically thought to account for their anti-tumor effect. It was discovered that imiquimod and the similar molecule CL097 are not only NLRP3 activators and TLR7/8 ligands, but that they also inhibit cellular respiration by inhibiting the mitochondrial electron transport chain (Christina J. Groß, Ritu Mishra, Katharina S. Schneider, Guillaume Médard, Jennifer Wettmarshausen, Daniela C. Dittlein, Hexin Shi, Oliver Gorka, Paul-Albert Koenig, Stephan Fromm, Giovanni Magnani, Tamara Ćiković, Lara Hartjes, Joachim Smollich, Avril A. B. Robertson, Matthew A. Cooper, Marc Schmidt-Supprian, Michael Schuster, Kate Schroder, Petr Broz, Claudia Traidl-Hoffmann, Bruce Beutler, Bernhard Kuster, Jürgen Ruland, Sabine Schneider, Fabiana Perocchi and Olaf Groß, “K+Efflux-Independent NLRP3 Inflammasome Activation by Small Molecules Targeting Mitochondria”, Immunity, volume 45, issue 4, Oct. 18, 2016, pp. 761-773). Both TLR7/8 activation and electron transport chain inhibition by imidazoquinolines might contribute to their antiviral and anti-tumor activity, but are also known to have adverse effects. Thus, there is still a need to provide selective activators for the NLRP3 inflammasome.


In summary, there is a need for NLRP3 activators, which are more selective than previous NLRP3 activators and can boost immune responses and trigger tumoricidal activity of the immune system.


In addition, new small molecule NLRP3 activators are useful to study the biology of the NLRP3 inflammasome and thus make an important contribution to the fundamental understanding of the activation mechanisms of the NLRP3 inflammasome and its function in health and disease and enable the targeted development of specific drugs.


The present invention addresses these needs. In particular, the present invention provides compounds and compositions comprising these compounds, which can activate NLRP3 without activating TLR7 and/or TLR8 and/or the mitochondrial electron transport chain. Therefore, these compounds are useful in the prevention or treatment of diseases such as cancer as well as infectious diseases or in the context of vaccination to boost specific immune responses as an adjuvant by activating NLRP3. Moreover, these compounds are useful in methods for analyzing the biology of NLRP3 activation and signaling mechanisms involved.


SUMMARY OF THE INVENTION

According to the first aspect, the present invention relates to a compound for use in the prevention or treatment of a disease by activating the NLRP3 inflammasome, wherein the disease is selected from a group comprising infectious diseases or cancer, and wherein the compound is a compound of Formula (I), Formula (I′) or Formula (I”)




embedded image


or a pharmaceutically acceceptable salt thereof, wherein:


A and A′ are independently selected from each other an optionally substituted C6-C18 aryl; optionally substituted 5-20 membered heteroaryl; optionally substituted C3-C20 cycloalkyl; optionally substituted 3-20 membered cycloheteroalkyl, preferably an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; an optionally substituted C3-C10 cycloalkyl; or an optionally substituted 3-10 membered cycloheteroalkyl, more preferably an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl. More preferably, A and A′ are as defined herein below


B is O or S, preferably O.


X and X′ are each independently present or not, wherein X and X′ are each independently an optionally substituted C1-C10 alkyl group; preferably a C1-C5 alkyl group, or a C1-C5 alkanol group; more preferably selected from —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


R6 and R7 are each independently hydrogen, an optionally substituted C1-C10 alkyl, preferably a C1-C5 alkyl, more preferably —H, —CH3, —C2H5, —C3H7, or —C3H9, or optionally


R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


preferably selected from




embedded image


or optionally,


R7, N and X when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


In one embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 1, 0 to 2, 0 to 3 or 0 to 4, or C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, wherein preferably only one of X and X′ is present and is selected from a C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—; and wherein


R6 and R7 are each independently hydrogen, a C1-C10 alkyl, preferably a C1-C5 alkyl, C1-C4 alkyl, C1-C3 alkyl, C1-C2 alkyl, more preferably hydrogen or CH3, even more preferably preferably wherein R6 and R7 are each hydrogen, and wherein A and A′ are preferably as defined herein below.


In one further embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 1, 0 to 2, 0 to 3 or 0 to 4, or C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, preferably only one of X and X′ is present and is a C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—; and wherein


R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


preferably selected from




embedded image


and wherein A and A′ are preferably as defined herein below.


In one further embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 1, 0 to 2, 0 to 3 or 0 to 4, or C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, preferably only one of X and X′ is present and is a C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—; and wherein R7, N and X when taken together form an optionally substituted five- or six-membered heterocyclic


ring, preferably selected from




embedded image


and wherein A and A′ are preferably as herein below.


In one embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 2, preferably 0 or 1, more preferably 0, wherein A and A′ are preferably as defined herein below.


In one embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 2, preferably 0 or 1, more preferably 0, and R6 and R7 are each independently hydrogen, wherein A and A′ are preferably as defined herein below.


In one further embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises none of X and X′; and R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


preferably selected from




embedded image


and wherein A is preferably as defined herein below.


In one further embodiment, in the compound of Formula (I), (I′) or (I″) of the invention R7, N and X when taken together form an optionally substituted five- or six-membered heterocyclic


ring, preferably selected from




embedded image


and wherein and A′ are as herein below.


In one preferred embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently a C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, and


preferably only one of X and X′ is present and is a C1-C3 alkyl, a C1-C2 alkyl, preferably selected from the group consisting of, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, and


A and A′ are each independently optionally substituted isoquinoline, quinoline, benzene, pyridine, 1,5-naphthyridine, isothiazole, or oxadiazole. A and A′ may be identical to each other or different to each other. In one preferred embodiment, A and A′ are different to each other. A and A′ are preferably as defined herein below.


In one embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises A and A′, wherein A and A′ are each independently optionally substituted benzene, pyrrolidine, pyridine, pyrazole, pyridazine, pyrimidine, pyrrole, pyrrolizine, furan, 2-benzofuran, 1,3-dihydro-2-benzofuran, diazole, triazole, oxazole, oxadiazole, 1,2,4-oxadiazole, thiazole, thiophene, thiadiazole, imidazole, indazole, indole, indoline, isoindole, isoindoline, dihydroindoline, quinoline, isoquinoline, isothiazole, isoxazole, naphthyridine, 1,5-naphthyridine, 1,6-naphthyridine, 1,8-naphthyridine, phthalazine, purine, quinazoline, quinoxaline, benzimidazole, benzoisoxazole, tetrahydrothiophene, benzoxazole or benzthiazole,


more preferably wherein A and A′ are each independently optionally substituted quinoline, isoquinoline, indole, benzene, pyridine, pyrazole, pyrrol, furan, thiophene, naphthyridine, 1,5-naphthyridine, 1,8-naphthyridine, isothiazole, or oxadiazole,


even more preferably wherein A and A′ are each independently optionally substituted isoquinoline, quinoline, benzene, pyridine, naphthyridine, 1,5-naphthyridine, isothiazole, oxadiazole, or thiophene,


most preferably wherein A and A′ are each independently optionally substituted isoquinoline, quinoline, benzene, pyridine, 1,5-naphthyridine, isothiazole, or oxadiazole as defined in the following.


A and A′ may be identical to each other or different to each other. In one embodiment, A and A′ are different to each other.


Preferably, A and A′ are each independently optionally substituted quinoline, isoquinoline, benzene, pyridine, naphthyridine, 1,5-naphthyridine, 1,6-naphthyridine, 1,8-naphthyridine, isothiazole, oxadiazole, or thiophene, preferably selected from groups as defined in the following in any of sections a) to i):

    • a) an optionally substituted benzene group of the following structure:




embedded image


wherein R1-R5 are each independently selected from the group consisting of: hydrogen; —OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —OCH(CH3)2; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, —CH3, —CH2F, —CHF2, —CF3, —CH2Cl, —CHCl2, —CCl3, CH(CH3)2; CHCH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —NH—CH(CH3)2, —NH-cyclopropyl, —CHF2, —C(O)—C2H5; —C(O)—CH3; —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl, optionally substituted C1-C10 cycloalkyl, optionally substituted C1-C10 cycloheteroalkyl,




embedded image


an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; or


NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH—CH(CH3)2, —NH-cyclopropyl, C1-C5 alkyl, —CH2F, —CHF2, —CF3, —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; or


—C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; —CH2NHS(O)2CH3; or —NHR43, wherein R43 is selected from




embedded image


wherein R60 is selected from hydrogen or —CH3; or




embedded image


wherein


R53 is preferably selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CH2F, —CHF2, —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO,




embedded image


with X=0 or 1, preferably 1;




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CF3, —CH2Cl; —CHCl2; —CCl3; OCH3, —F, —Cl, —Br, NH2, NO2, and x is 0, 1 or 2; or


R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2CF, —CF3, —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —C, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above;


and optionally, wherein R1 and R2, R2 and R3, R3 and R4, or R4 and R5 together, preferably R2 and R3 together, form an optionally substituted five- or six-membered cyclic or heterocyclic ring, preferably selected from




embedded image


Preferably X is not present in Formulae (I), (I′) or (I″) in case A or A′ is an optionally substituted benzene group as defined under a). Likewise, one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Likewise preferably either all of R1 to R5 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R1 to R5 are different from hydrogen and selected from substitutions defined or presented herein above under a) for R1 to R5, and remaining substituents of R1 to R5 are hydrogen.

    • b) an optionally substituted pyridine group of one of the following structures:




embedded image


wherein R44-R47 are each independently selected from the group consisting of:


hydrogen, —OH, —OCH3, CH3, F, Cl, Br, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CN; NO2; NH2; —CH3, CH(CH3)2; —CH2CH3; —CH2OH; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3;




embedded image


—NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH-cyclopropyl, —CHF2, —CH2F; —CHF2; —CF3, —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine; an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; optionally substituted C1-C10 cycloalkyl, optionally substituted C1-C10 cycloheteroalkyl,


—C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; —CH2NHS(O)2CH3; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl,




embedded image


or


—NHR43, wherein R43 is selected from




embedded image


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted pyridine group as defined under b) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Likewise preferably either all of R44 to R47 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R45 to R47 are different from hydrogen and selected from substitutions defined or presented herein above under b) for R44 to R47, and remaining substituents of R44 to R47 are hydrogen.

    • c) an optionally substituted quinoline or an isoquinoline group, preferably of one of the the following structures:




embedded image


embedded image


wherein


R21 to R26 are each independently selected from the group consisting of:


hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —C2H—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine; —C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; —CH2NHS(O)2CH3; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl,




embedded image


an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; optionally substituted C1-C10 cycloalkyl, optionally substituted C1-C10 cycloheteroalkyl; or


—NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH-cyclopropyl, —CH2F, —CHF2, —CF3, —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; or


—NHR43, wherein R43 is selected from




embedded image


or more preferably wherein R21 to R26 are each hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3;


or even more preferably wherein R21 to R26 are each hydrogen, or —F; Cl, Br.


Preferably in case A and/or A′ is an optionally substituted quinoline or an isoquinoline group in Formula (I), (I′) or (I″) as defined in c) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Likewise preferably either all of R21 to R26 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R21 to R26 are different from hydrogen and selected from substitutions defined or presented herein above under c) for R21 to R26 and remaining substituents of R21 to R26 are hydrogen;

    • d) a naphthyridine, preferably a 1,5 naphthyridine group, a 1,6 naphthyridin group or a 1,8 naphthyridin group of any of the following structures:




embedded image


embedded image


wherein


R48 to R52 are each independently selected from the group consisting of:


hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO,


—NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH-cyclopropyl, —CHF2, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine;


—C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; —CH2NHS(O)2CH3; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl,




embedded image


an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; optionally substituted C1-C10 cycloalkyl, optionally substituted C1-C10 cycloheteroalkyl;


—NHR43, wherein R43 is selected from




embedded image


or more preferably wherein R48 to R52 are each hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3;


or even more preferably wherein R48 to R52 are each hydrogen, or —F; Cl, Br.


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted 1,5-naphthyridine group, 1,6 naphthyridine group or a 1,8 naphthyridine group as defined under d) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Likewise preferably either all of R48 to R52 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R48 to R52 are different from hydrogen and selected from substitutions defined or presented herein above under d) for R48 to R52 and remaining substituents of R48 to R52 are hydrogen;

    • e) an oxadiazole group, preferably selected from one of the following structures:




embedded image


wherein


R53 is preferably selected from the group consisting of:


hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CHF2, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; an optionally substituted C1-C10 cycloalkyl, an optionally substituted C1-C10 cycloheteroalkyl,




embedded image


with X=0 or 1, preferably 1;




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NH2, NO2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, OCF3; —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above, and x is 0, 1 or 2;


and wherein preferably either all of R54 to R58 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R54 to R58 are different from hydrogen and selected from substitutions defined or presented herein above under e) for R54 to R58 and remaining positions of R54 to R58 are hydrogen;


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted oxadiazole group as defined under e) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • f) a thiadiazole group, preferably selected from one of the following structures:




embedded image


wherein R53 is preferably as defined above in section e);


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted thiadiazole group as defined under f) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • g) an isothiazole group, preferably selected from one of the following structures:




embedded image


wherein R66 and R67 are each selected independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CHF2, —F, —Cl, —Br, preferably wherein either of R66 or R67 is hydrogen and the other residue is selected from —CH3, —C2H5, —CF3, —CHF2, —F, —Cl, —Br, or both of R66 and R67 is hydrogen;


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted isothiazole group as defined under g) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • h) a thiophene group, preferably selected from one of the following structures:




embedded image


wherein R68, R69 and R70 are each selected independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —C, —Br, preferably wherein either of R68, R69 or R70 is hydrogen and the other residue is selected from —CH3, —C2H5, —CF3, —CHF2, —F, —C, —Br, or each of R68, R69 and R70 is hydrogen.


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted thiophene group as defined under g) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • i) A is NH2 in Formula (I), (I′) or (I″), wherein A′ is different to A and is selected from any of groups a) to h) as defined above.


In one embodiment, the compound of Formula (I), (I′) or (I″) of the invention is a compound of any one of Formulae (Ic), (Ic1), (Ic2), (Ic3), or (Ic4), or a pharmaceutically acceptable salt thereof:




embedded image


wherein B=O or S, preferably O;


wherein R6 and R7 are independently each hydrogen, C1-3 alkyl, preferably hydrogen, CH3, more preferably, either R6 is hydrogen and R7 is CH3, or R6 and R7 are both hydrogen; and


R21 to R37 are each independently selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —O—N═O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2 (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, or C(NR11)R12, —NHC(O)R11, and —NHC(O)NHR11, and wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C5 alkyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, optionally substituted 2-20 membered heteroalkyl, —CH2F; —CHF2, —CF3, —CH2Cl; —CHCl2, and —CCl3; or


preferably wherein R21 to R37 are each independently selected from the group consisting of hydrogen, C1-C6 alkoxy, C1-C5 alkyl, C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, —CF3, —F, —Cl, and —Br; or


wherein preferably either all of R21 to R37 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R21 to R37 are different from hydrogen and selected from substitutions presented above for R21 to R37 and remaining substituents of R21 to R37 are hydrogen; or more preferably wherein R21 to R37 are each hydrogen.


In one more preferred embodiment, the compound of Formula (I), (I′) or (I″) of the invention is a compound any one of Formulae (Ic5), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13), or (Ic14) or a pharmaceutically acceptable salt thereof:




embedded image


embedded image


wherein R21 to R37 are each independently selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F; —Cl; —Br; —CN; NO2; NH2; C1-C5 alkoxy, C1-C5 alkyl, preferably —CH3, CH(CH3)2; —CH2CH3; or C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, or


even more preferably R21 to R37 are each hydrogen, or 1, 2 or 3, preferably 1 or 2 of R21 to R37 are different from hydrogen and selected from substitutions presented above for R21 to R37 and remaining substituents of R21 to R37 are hydrogen.


In a second aspect the compound of Formula (I) of the invention is a compound of Formula (Ia) or (Ia1),




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


X is either present or not, and X, if present, is preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—; and


R6 and R7 are each independently hydrogen, —CH3, —C2H5, —C3H7, or —C3H9, or optionally,


R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


or optionally,


R7, N and X when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


and


wherein R1-R5 are each independently preferably selected as defined above, more preferably selected from the group consisting of hydrogen; —OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —OCH(CH3)2; —F; —Cl; —Br; —CN; NO2; NH2; —CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; CH(CH3)2; —CH2CH3;


—C(O)OR42, wherein R42 is selected from —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3;




embedded image


wherein R60 is selected from hydrogen or —CH3;


optionally, wherein R1 and R2, R2 and R3, R3 and R4, or R4 and R5 together, preferably R1 and R2 or R2 and R3 together, form an optionally substituted five- or six-membered cyclic or heterocyclic ring, preferably selected from




embedded image


A in any of Formulae (Ia) or (Ia1) is preferably selected from one of the following substituents as defined above for A or A′, more preferably from A as defined below in sections a) to e):


A may be preferably selected for any of Formulae (Ia) or (Ia1) from the following groups a) to e):

    • a) a benzene group of the following structure:




embedded image


wherein R61-R65 are each independently selected from the group consisting of:


hydrogen; —OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —OCH(CH3)2; —F; —Cl; —Br; —CN; NO2; NH2; —CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —C(O)—C2H5; —C(O)—CH3; —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine; —CH2NHS(O)2CH3; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl,




embedded image


or


—NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH—CH(CH3)2, —NH-cyclopropyl, —CHF2, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; or


—C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; or


—NHR43, wherein R43 is selected from




embedded image


wherein R60 is selected from hydrogen or —CH3; or




embedded image


wherein R53 is preferably as defined above selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CHF2, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —




embedded image


with X=0 or 1, preferably 1;




embedded image


or R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; or


or R53 is




embedded image


wherein R59 is




embedded image


and


wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above, and x is 0, 1 or 2;


and


optionally, wherein R61 and R62, R62 and R63, R63 and R64, or R64 and R65 together, preferably R62 and R63 together, form an optionally substituted five- or six-membered cyclic or heterocyclic ring, preferably selected from




embedded image


wherein preferably either all of R1 to R5 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R1 to R5 are different from hydrogen and selected from substitutions defined above for R1 to R5 and remaining substituents of R1 to R5 are hydrogen;


Likewise preferably, X is then not present in Formula (Ia) and (Ia1).

    • b) an oxadiazole group of one of the following structures:




embedded image


wherein


R53 is preferably selected as above from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —C, —Br, NO2, NH2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above, and x is 0, 1 or 2;


and wherein preferably either all of R54 to R58 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R54 to R58 are different from hydrogen and selected from substitutions presented above and remaining positions of R54 to R58 are selected from hydrogen;


Preferably, X is then selected in Formula (Ia) or (Ia1), from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • c) a thiadiazole group of one of the following structures:




embedded image


wherein R53 is preferably as defined herein above.


Preferably, X is then selected in formula (Ia) or (Ia1), from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • d) an isothiazole group of the following structure:




embedded image


wherein R66 and R67 are each selected independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —C, —Br.


Preferably, X is then selected in Formula (Ia) or (Ia1), from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • e) a thiophene group, preferably selected from one of




embedded image


wherein R68, R69 and R70 are each selected independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —Cl, —Br, preferably wherein either of R68, R69 or R70 is hydrogen and the other residue is selected from —CH3, —C2H5, —CF3, —CHF2, —F, —Cl, —Br, and preferably X is then selected in Formula (Ia) or (Ia1), from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


According to one particular aspect, in Formula (Ia) or Formula (Ia1) A is preferably an optionally substituted C6-C18 aryl as defined above, and more preferably the following optionally substituted aryl/benzene group:




embedded image


and X is present or not present, preferably not present;


or A is an optionally substituted five- or six-membered heterocyclic ring as defined above, and more preferably A is an oxadiazole group of one of the following structures:




embedded image


wherein R53 is preferably as defined herein,


wherein X is preferably selected in formula (Ia) or (Ia1), from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—,


and, in either case R1, R2, R3, R4, R5, R3 and R9 are each independently selected from hydrogen, —F, —Cl, —Br, —NO2, —O—N═O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkoxy, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 cycloalkyl C1-C10 alkyl, optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, optionally substituted C6-C18 aryl, optionally substituted C6-C18 aryl C1-C10 alkyl, optionally substituted 5-20 membered heteroaryl, optionally substituted 5-20 membered heteroaryl C1-C10 alkyl, (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, optionally substituted 2-20 membered heteroalkyl, —CH2F; —CHF2, —CF3, —CH2Cl; —CHCl2, and —CCl3; or


R1 and R2, R2 and R3, R3 and R4, R4 and R5, or R3 and R9, when taken together form an optionally substituted five- or six-membered cycloalkyl, optionally substituted five- or six-membered cycloheteroalkyl, optionally substituted five- or six-membered aryl, or optionally substituted five- or six-membered heteroaryl, and


R6 and R7 are each independently hydrogen, an optionally substituted C1-C10 alkyl or R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


According to one further aspect, in Formula (Ia) or Formula (Ia1) R1, R2, R3, R4 and R5 are each hydrogen and/or A is




embedded image


and R8 and R9 are each independently hydrogen, substituted C1-C10 alkyl, substituted 2-10 membered heteroalkyl, substituted C2-C14 aryl, substituted 5-20 membered heteroaryl, substituted five- or six-membered cycloheteroalkyl, —S(O)2CH3, —F, —Cl, —Br, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 is as defined above selected from C1-C5 alkyl, C3-C6 cycloalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; or five-membered heterocyclic ring; —OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —OCH(CH3)2; —CN; NO2; NH2; —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —NH—CH(CH3)2, —NH-cyclopropyl, CH2CH3, CHCH(CH3)2; —C(O)—C2H5; —C(O)—CH3; —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine; —C(CH3)═NOH; —CH2NHS(O)2CH3; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl,




embedded image


or


NHC(O)—R41, wherein R41 is selected from furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH—CH(CH3)2, —NH-cyclopropyl, C1-C5 alkyl, —CHF2, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; or


—C(O)OR42, wherein R42 is selected from —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; or


—NHR43, wherein R43 is selected from




embedded image


wherein R60 is selected from hydrogen or —CH3; or




embedded image


wherein


R53 is selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO,




embedded image


with X=0 or 1, preferably 1;




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —C, —Br, NH2, NO2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above;


with the optional proviso that R8 is not identical to R9, or optionally


R8 and R9, when taken together form an optionally substituted five- or six-membered cycloalkyl, optionally substituted five- or six-membered cycloheteroalkyl or optionally substituted five or six-membered aryl, optionally substituted five- or six-membered heteroaryl, preferably the heterocyclic ring is selected from:




embedded image


According to one further aspect, in the compound according to Formulae (Ia) and (Ia1) R8 and R9 are each independently defined as above, with the proviso that if one of R8 or R9 is —F, —Cl, or —Br, the other R8 or R9 is neither —F, —Cl, —Br nor hydrogen.


According to one further aspect, in the compound according to Formulae (Ia) and (Ia1) R8 and R9 are each independently defined as above, with the proviso that R8 or R9 are not identical to each other.


Preferably in another aspect according to Formulae (Ia) and (Ia1), in the compound according to Formulae (Ia) and (Ia1), R9 is selected from a C1-C3 alkyl, a five-membered heterocyclic ring, —OCH3, —S(O)2CH3, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 is selected from C2-C5 alkyl, C3-C6 cycloalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CHCl2, —CCl3; or five-membered heterocyclic ring; and/or


R8 is —S(O)2CH3, —NHC(O)R11, or —NHC(O)NHR11, and wherein R11 is selected from C2-C5 alkyl, C3-C5 cycloalkyl, —CHF2; or five-membered heterocyclic ring;


or R8 and R9, when taken together form a heterocyclic ring selected from:




embedded image


According one specific aspect the —X-A group in any of Formulae (Ia) or (Ia1) is




embedded image


wherein R10 is hydrogen or X is a C1-C3 alkyl, preferably —CH(CH3)2—CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, or —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, or —(CH2)n-A, wherein n is an integer from 1-3, preferably 1, and/or A is




embedded image


preferably forming Formula (Ib) or (Ib1) as shown below):


According one specific aspect of the invention the compound according to Formulae (Ia) and (Ia1) is a compound according to Formula (Ib) or (Ib1):




embedded image


wherein X is preferably a C1-C3 alkyl, preferably —CH(CH3)2—CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, or —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—,


wherein R53 is preferably hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO,




embedded image


with X=0 or 1, preferably 1;




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NH2, NO2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and


wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above, and x is 0, 1 or 2;


and wherein preferably either all of R54 to R58 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R54 to R58 are different from hydrogen and selected from substitutions presented above and remaining positions of R54 to R58 are selected from hydrogen.


According to a further aspect of the present invention the compound according to Formulae (Ia) and (Ia1) is a compound according Formula (Id):




embedded image


wherein preferably


B is O or S, preferably O,


R1, R2, R4 and R5 are each selected separately from hydrogen, —CH3, —C2H5, —CH2CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, preferably wherein two or three of R1, R2, R4 and R5 are hydrogen and the remaining of R1, R2, R4 and R5 are not hydrogen; and


R66 and R67 are each selected separately from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, preferably wherein one of R66 and R67 is hydrogen and the other is one of —CH3, —C2H5, —CF3, —CHF2, —OCH3, —F, —Cl, —Br, NO2, NH2.


According to a third aspect, the present invention relates to a compound of Formula (II),





R21-D-CO—R22  Formula (II)


or a pharmaceutically acceptable salt thereof, wherein:


D is an optionally substituted heteroaromatic bicycle selected from the group comprising 2-benzofuran, 2-benzothiophene, isoindol, indol, indolizine, and


wherein D is preferably indolizine




embedded image


R71 is an optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, and


R72 is optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, —OR73, —NR73, and wherein


R73 is hydrogen, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl.


According to one particular aspect, the present invention relates to the present compounds for use as a medicament.


According to one aspect, the present invention relates to the present compounds for use in the prevention or treatment of a disease by activating the NLRP3 inflammasome, wherein the disease is selected from a group comprising infectious diseases or cancer.


According to one aspect, the present invention relates to a pharmaceutical composition comprising a compound of the present invention and one or more pharmaceutically acceptable excipients.


According to one aspect, the present invention relates to a method for modulating NLRP3 activity, the method comprising engaging NLRP3 with a compound of the present invention.


According to one aspect, the present invention relates to a method of treating cancer, comprising administering to a subject in need of such treatment an effective amount of a compound of the present invention, or a pharmaceutical composition of the present invention


According to one aspect, the present invention relates to a method for analyzing the mechanisms of NLRP3 activation.


According to one aspect, the present invention relates to the use of the compounds of the present invention for boosting immune responses or breaking immune evasion in cancer diseases in a subject.


According to one aspect, the present invention relates to the use of the compounds of the present invention for boosting immune responses as an adjuvant in a vaccine in a subject. Preferably, the compounds of the present invention may be used as an adjuvant in the context of vaccination to boost specific immune responses by activating the NLRP3 inflammasome.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1: Line diagram of the high-throughput screening of about 50 000 low molecular weight compounds. The diagram shows a partial data set for 39 440 compounds tested at 25 μM of the compounds after about 20 hours of incubation in LPS-primed murine bone marrow-derived dendritic cells (BMDCs). Primary identification of cell death inducing compounds.



FIG. 2: Dot diagram of the selection of NLRP3 activators for which cell death is dependent on the inflammasome adapter ASC (Pycard−/−) in LPS-primed BMDCs after 16 hours incubation at 50 μM of the compounds, the framed dots represent the respective replicates of the tested compounds EN21, EN22 and EN54.



FIG. 3: Dot diagram of the selection of activators, which lead to the release of IL-1p as an indicator of inflammasome activation (from BMDCs after 20 hours incubation at 50 μM of the compounds). The cells were each pretreated for 3 hours with 50 ng ml-LPS to prime for inflammasome activation.



FIG. 4A: Line diagram of the dose dependent cytotoxicity of compound EN21 in wild-type and inflammasome knockout (Pycard−/−) BMDCs. Cytotoxicity was assessed by quantification of cellular ATP levels.



FIG. 4B: Line diagram of the dose dependent cytotoxicity of compound EN22 in wild-type and inflammasome knockout (Pycard−/−) BMDCs. Cytotoxicity was assessed by quantification of cellular ATP levels.



FIG. 4C: Line diagram of the dose dependent cytotoxicity of compound EN54 in wild-type and inflammasome knockout (Pycard−/−) BMDCs. Cytotoxicity was assessed by quantification of cellular ATP levels.



FIG. 4D: Line diagram of the EC50 for induction of IL-1p release from BMDCs for compounds EN21 and EN22 in comparison to the imidazoquinoline NLRP3 activators imiquimod and CL097. The presence of IL-1p in the cell culture supernatant was determined by enzyme-linked immunosorbent assay (ELISA) using a kit selective for the mature form of the cytokine.



FIG. 5A: Immunoblot analysis of the effects of compound EN21. Caspase-1 activation and release of the biologically active form of IL-1β in BMDCs after treatment with the present compounds at 50 μM for 3 hours. The cells were pretreated with 50 ng ml−1 LPS.



FIG. 5B: Immunoblot analysis of the effects of compound EN22. Caspase-1 activation and release of the biologically active form of IL-1β in BMDCs after treatment with the present compounds at 50 μM for 3 hours. The cells were pretreated with 50 ng ml−1 LPS.



FIG. 6: Images of ASC speck formation in BMDCs of ASCcitrine mice expressing a fluorescent fusion protein of the inflammasome adapter ASC. ASC speck formation can be used as a readout for inflammasome activation. ASC 1 (green/bright grey, ASC specks green/bright grey spots), nuclei 2 (blue/dark grey, bigger round structure), CD45 3 (red/dark grey, small spots). The cells were first treated with 50 ng ml−1 LPS for 3 hours, then with 25 μM of the activators for 2 hours.


The present compounds induce typical characteristics of inflammasome activation, including the activation of caspase-1 and the release of the biologically active forms of leaderless proinflammatory cytokines of the IL-1 family such as IL-1β (FIGS. 5A and B). Moreover, inflammasome-dependent lytic cell death (pyroptosis, FIG. 4) and ASC speck formation are induced by the present compounds (FIG. 6). For the induction a concentration dependence can be observed. An advantage of the present compounds is that they are active in vitro at approximately 10-fold more lower concentrations than established imidazoquinolines.



FIG. 7: Bar diagram of IL-1β and lactate dehydrogenase release (as marker for lytic cell death and pyroptosis) from BMDCs from wild-type or NLRP3 knockout mice. The cells were each pretreated with 50 ng ml−1 LPS and stimulated with 50 μM EN21, EN22 or control activators.



FIG. 8: Bar diagram of IL-1β and lactate dehydrogenase release from BMDCs from wild-type mice with or without pre-incubation with the NLRP3-specific inhibitor MCC950. The cells were each pretreated with 50 ng ml-LPS and stimulated with 50 μM EN21, EN22 or control activators.


Without wishing to be bound to a theory, it is assumed that the compounds EN21 and EN22 activate the NLRP3 inflammasome, since the observed characteristics of inflammasome activation do not occur in cells isolated from NLRP3 knockout mice (FIG. 7) and, in addition, the NLRP3-specific inhibitor MCC950 completely suppresses inflammasome activation (FIG. 8).


A further advantage of the present compounds is that the present compounds seem to activate NLRP3 rapidly. Especially with EN22, the inflammasome activation in BMDCs from wild-type mice without inhibitor is almost maximum after only 30 minutes.



FIG. 9: Bar diagram of TNF and IL-6 release of BMDCs as readout for toll-like receptor activation. The cells were stimulated with 50 μM EN21, EN22 or control activators.


In contrast to the small molecule the NLRP3 inflammasome activators imiquimod (patent US 2019/0127368 A1), the present compounds are more specific, since no simultaneous activation of Toll-like receptors (TLR7/TLR8) occurs and thus the present compounds do not induce the release of further proinflammatory cytokines such as TNF or IL-6.



FIG. 10: Line diagram of extracellular flux analysis in BMDCs of inflammasome knockout mice. Upper panel: Oxygen consumption rate (OCR) from the medium, which shows the mitochondrial respiration over time. Lower panel: Acidification of the medium, which shows glycolytic activity of the cells over time (extracellular acidification rate, ECAR). The cells were treated with 50 μM EN21, EN22 or 10 μM Piericidin as a control. The mitochondrial uncoupler FCCP was used to prove responsiveness of the cells and therefore functionality of the assay.


In contrast to other known small molecule NLRP3 activators, the present compounds do not inhibit mitochondrial respiration and are therefore less susceptible to unspecific undesired effects.



FIG. 11: Bar diagram of IL-1β release from BMDCs of wild-type mice either left without inhibitor or pretreated with 50 mM extracellular KCl to inhibit potassium efflux from the cells, or with 30 μM ebselen used as a ROS scavenger. The cells were pretreated with 50 ng ml−1 LPS and stimulated with 50 μM EN21, EN22 or control activators.


Most of the known NLRP3 activators lead to substantial potassium efflux from the cell via different mechanisms, which precedes inflammasome activation. Without wishing to be bound to a theory, it is assumed that the present compounds such as EN21 and EN22 activate the NLRP3 inflammasome independently of potassium efflux from the cell. It is assumed that reactive oxygen species (ROS) are involved in the mechanism of action for NLRP3 activation of the present compounds, since the ROS scavenger ebselen can inhibit inflammasome activation. It might be also possible that ROS influences a further mechanism that is required for NLRP3 activation in parallel to the mechanism activated by EN21 and/or EN22.



FIG. 12: Bar diagram of IL-1β release from LPS-primed BMDCs from wild-type mice either left without inhibitor or pretreated with the endocytosis inhibitor cytochalasin D. The cells were stimulated with either 50 μM EN21, EN22 or control activators.


The endocytosis inhibitor cytochalasin D prevents MSU-induced NLRP3 activation, but has no effect on NLRP3 activation by EN21 and EN22. Without wishing to be bound to a theory, it is assumed that contrary to particulate NLRP3 activators such as uric acid crystals (mono sodium urate, MSU) or aluminum hydroxide, the present compounds are not endocyted by immune cells as a prerequisite for the NLRP3 inflammasome activation.



FIG. 13: Bar diagram of IL-1β and lactate dehydrogenase release (as marker for lytic cell death and pyroptosis) from BMDCs from wild-type or NLRP3 knockout mice The cells were each pretreated with 50 ng ml−1 LPS and stimulated with 5 μM, 50 μM, 100 μM BRM/BRG1 ATP Inhibitor-1 (concentrations increasing from left to right, indicated by the wedge below). or control activators.



FIG. 14: Bar diagram of IL-1β (lower figure) and lactate dehydrogenase (upper figure) release of BMDCs from wild-type and NLRP3 knockout mice. The cells were primed with 50 ng ml−1 LPS for 3 hours and then stimulated with the indicated compounds for 3 hours as follows: EN21 12.5 μM, EN22 25 μM, EN54 50 μM, EN23 25 μM, 50 μM, 100 μM, 200 μM (concentrations increasing from left to right, indicated by the wedge below).



FIG. 15: Bar diagram of IL-1β and lactate dehydrogenase release (as marker for lytic cell death and pyroptosis) from BMDCs from wild-type mice. The cells were each pretreated with 50 ng ml−1 LPS and stimulated with 25 μM, 50 μM, 75 μM AH4 (concentrations increasing from left to right, indicated by the wedge below) or 10 μM EN22.



FIG. 16: Bar diagram of IL-1β (lower figure) and lactate dehydrogenase (upper figure) release of BMDCs from wild-type and NLRP3 knockout mice. The cells were primed with 50 ng ml−1 LPS for 3 hours and then stimulated with the indicated compounds for 3 hours as follows: EN22 20 μM, JB010 20 μM, 40 μM, 80 μM (concentrations increasing from left to right, indicated by the wedge below).


Without wishing to be bound to a theory, it is assumed that the compounds BRM/BRG1 ATP Inhibitor-1, EN23, AH4 and JB010 act similarly as EN21 and EN22 activates the NLRP3 inflammasome, since the observed characteristics of inflammasome activation do not occur in cells isolated from NLRP3 knockout mice (FIGS. 7, 13, 14 and 16).



FIG. 17: Bar diagram of IL-1β release of wild-type BMDCs. The cells were primed with 50 ng ml−1 LPS for 3 hours, incubated with 50 mM extracellular KCl to prevent potassium efflux from the cells for 30 minutes and then stimulated with 12.5 μM EN23 for 3 hours.


Without wishing to be bound to a theory, it is assumed that the present compounds such as EN23 activate the NLRP3 inflammasome independently of potassium efflux from the cell. (FIG. 17) FIG. 18: Bar diagram of IL-1β and lactate dehydrogenase release of BMDCs from wild-type and ASC knockout (Pycard−/−) mice. The cells were primed with 50 ng ml−1 LPS for 3 hours and then stimulated with the indicated compounds for 3 hours as follows: nigericin 5 μM, EN54 10 μM, 20 μM, 40 μM, 80 μM (concentrations increasing from left to right, indicated by the wedge below).


Without wishing to be bound to a theory, it is assumed that the compound EN54 acts similarly as EN21 and EN22 activates the NLRP3 inflammasome, since the observed characteristics of inflammasome activation do not occur in cells isolated from NLRP3 knockout mice (FIG. 18).



FIG. 19: Bar diagram of TNF and IL-6 release of BMDCs as readout for toll-like receptor activation.





DETAILED DESCRIPTION OF THE INVENTION

The terms “a” and “an” do not denote a limitation of quantity, but rather denote the presence of at least one of the referenced item. The term “or” or “and/or” is used as a function word to indicate that two words or expressions are to be taken together or individually. The terms “comprising”, “having”, “including”, and “containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to”). The endpoints of all ranges directed to the same component or property are inclusive and independently combinable.


The term “present compound(s)” or “compound(s) of the present invention” refers to compounds encompassed by a structural formula disclosed herein, such as Formulae (I), (I′), (I″), (Ia), (Ia1), (Ib), (Ib1), (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic5), (Ic6), (Ic7), (Ic8), (Ic9), (Ic10), (Ic11), (Ic12), (Ic13), (Ic14), (Id), (II), (IIa), and (IIb) and includes any subgenus and specific compounds within the formula whose structure is disclosed herein. Compounds may be identified either by their chemical structure and/or chemical name. When the chemical structure and chemical name conflict, the chemical structure is determinative of the identity of the compound. The compounds described herein may contain one or more chiral centers and/or double bonds and therefore, may exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), enantiomers or diastereomers. Accordingly, the chemical structures depicted herein encompass all possible enantiomers and stereoisomers of the illustrated compounds including the stereoisomerically pure form (e.g., geometrically pure, enantiomerically pure or diastereomerically pure) and enantiomeric and stereoisomeric mixtures. Enantiomeric and stereoisomeric mixtures can be resolved into their component enantiomers or stereoisomers using separation techniques or chiral synthesis techniques well known to the skilled artisan. The compounds may also exist in several tautomeric forms including the enol form, the keto form and mixtures thereof. Accordingly, the chemical structures depicted herein encompass all possible tautomeric forms of the illustrated compounds. The compounds described also include isotopically labeled compounds where one or more atoms have an atomic mass different from the atomic mass conventionally found in nature. Examples of isotopes that may be incorporated into the compounds of the invention include, but are not limited to, 2H, 3H, 13C, 14C, 15N, 18O, 17O, etc. Compounds may exist in unsolvated forms as well as solvated forms, including hydrated forms and as N-oxides. In general, compounds may be hydrated, solvated or N-oxides. Certain compounds may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated herein and are intended to be within the scope of the present invention. Further, it should be understood, when partial structures of the compounds are illustrated, that brackets indicate the point of attachment of the partial structure to the rest of the molecule. The term “tautomer” as used herein refers to isomers that change into one another with great ease so that they can exist together in equilibrium.


“Alkyl,” by itself or as part of another substituent, refers to a saturated branched, or straight-chain hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane. Typical alkyl groups include, but are not limited to, methyl; ethyl; propyls such as propan-1-yl, propan-2-yl (isopropyl), etc.; butanyls such as butan-1-yl, butan-2-yl (sec-butyl), 2-methyl-propan-1-yl (isobutyl), 2-methyl-propan-2-yl (t-butyl); and the like. In some embodiments, an alkyl group comprises from 1 to 20 carbon atoms (C1-C20 alkyl). In other embodiments, an alkyl group comprises from 1 to 10 carbon atoms (C1-C10 alkyl). In still other embodiments, an alkyl group comprises from 1 to 6 carbon atoms (C1-C6 alkyl) or 1 to 4 carbon atoms (C1-C4 alkyl). In still other embodiments, an alkyl group comprises from 1 to 3 carbon atoms (C1-C3 alkyl). C1-C8 alkyl is also known as “lower alkyl”.


“Cycloalkyl,” by itself or as part of another substituent, refers to a saturated cycloalkyl hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent cycloalkane. Cycloalkyl further includes cycloalkenyls as defined herein. Typical cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl; cyclobutadienyl, cyclohexadienyl; and the like. In some embodiments, a cycloalkyl group comprises from 3 to 20 carbon atoms (C3-C20 cycloalkyl). In other embodiments, a cycloalkyl group comprises from 3 to 10 carbon atoms (C3-C10 cycloalkyl). In a preferred embodiment, a cycloalkyl group comprises from 3 to 6 carbon atoms (C3-C6 cycloalkyl). In a more preferred embodiment, a cycloalkyl group comprises from 3 to 5 carbon atoms (C3-C5 cycloalkyl).


“Cycloalkyl alkyl,” by itself or as part of another substituent, refers to an acyclic alkyl group in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a cycloalkyl group as defined herein. That is, cycloalkyl alkyl can also be considered as an alkyl substituted by cycloalkyl. Typical cycloalkyl alkyl groups include, but are not limited to, cyclopropylmethyl, 2-cyclopropylethan-1-yl, 2-cyclopropylethen-1-yl, pentylmethyl, 2-pentylethan-1-yl, 2-pentylethen-1-yl, and the like. Where specific alkyl moieties are intended, the nomenclature cycloalkyl alkanyl, cycloalkyl alkenyl and/or cycloalkyl alkynyl is used. In some embodiments, a cycloalkyl alkyl group is (C3-C30) cycloalkyl alkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the cycloalkyl alkyl group is (C1-C10) alkyl and the cycloalkyl moiety is (C3-C20) cycloalkyl. In other embodiments, a cycloalkyl alkyl group is (C4-C20) cycloalkyl alkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the cycloalkyl alkyl group is (C1-C8) alkyl and the cycloalkyl moiety is (C3-C12) cycloalkyl. In still other embodiments, a cycloalkyl alkyl group is (C4-C10) cycloalkyl alkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the cycloalkyl alkyl group is (C1-C4) alkyl and the cycloalkyl moiety is (C3-C6) cycloalkyl.


It is noted that when an alkyl group is further connected to another atom, it becomes an “alkylene” group. In other words, the term “alkylene” refers to a divalent alkyl. For example, —CH2CH3 is an ethyl, while —CH2CH2— is an ethylene. That is, “Alkylene,” by itself or as part of another substituent, refers to a saturated or unsaturated, branched, straight-chain or cyclic divalent hydrocarbon radical derived by the removal of two hydrogen atoms from a single carbon atom or two different carbon atoms of a parent alkane, alkene or alkyne. The term “alkylene” is specifically intended to include groups having any degree or level of saturation, i.e., groups having exclusively single carbon-carbon bonds, groups having one or more double carbon-carbon bonds, groups having one or more triple carbon-carbon bonds and groups having mixtures of single, double and triple carbon-carbon bonds. In some embodiments, an alkylene group comprises from 1 to 20 carbon atoms (C1-C20 alkylene). In other embodiments, an alkylene group comprises from 1 to 10 carbon atoms (C1-C10 alkylene). In still other embodiments, an alkylene group comprises from 1 to 6 carbon atoms (C1-C6 alkylene).


“Alkenyl,” by itself or as part of another substituent, refers to an unsaturated branched or straight-chain monovalent hydrocarbon radical having at least one carbon-carbon double bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkene. The group may be in either the cis or trans conformation about the double bond(s). Typical alkenyl groups include, but are not limited to, ethenyl; propenyls such as prop-1-en-1-yl, prop-1-en-2-yl, prop-2-en-1-yl (allyl), prop-2-en-2-yl, butenyls such as but-1-en-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-1-yl, but-2-en-1-yl, but-2-en-2-yl, buta-1,3-dien-1-yl, buta-1,3-dien-2-yl, etc.; and the like. In some embodiments, an alkenyl group comprises from 2 to 20 carbon atoms (C2-C20 alkenyl). In other embodiments, an alkenyl group comprises from 2 to 10 carbon atoms (C2-C10 alkenyl). In still other embodiments, an alkenyl group comprises from 2 to 6 carbon atoms (C2-C6 alkenyl) or 1 to 4 carbon atoms (C2-C4 alkenyl). C2-C6 alkenyl is also known as “lower alkenyl”.


“Cycloalkenyl,” by itself or as part of another substituent, refers to an unsaturated cycloalkenyl hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent cycloalkene. Typical cycloalkenyl groups include, but are not limited to, cyclopropenyls such as clycloprop-1-en-1-yl, cycloprop-2-en-1-yl, cyclobutenyls such as cyclobut-1-en-1-yl, cyclobut-1-en-3-yl, cyclobuta-1,3-dien-1-yl, etc.; and the like. In some embodiments, a cycloalkenyl group comprises from 3 to 20 carbon atoms (C3-C20 cycloalkenyl). In other embodiments, a cycloalkenyl group comprises from 3 to 10 carbon atoms (C3-C10 cycloalkenyl). In a preferred embodiment, a cycloalkenyl group comprises from 3 to 6 carbon atoms (C3-C6 cycloalkenyl). In a more preferred embodiment, a cycloalkenyl group comprises from 3 to 5 carbon atoms (C3-C5 cycloalkenyl).


“Cycloalkenyl alkyl,” by itself or as part of another substituent, refers to an acyclic alkyl group in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a cycloalkenyl group as defined herein. That is, cycloalkenyl alkyl can also be considered as an alkyl substituted by cycloalkenyl. Typical cycloalkenyl alkyl groups include, but are not limited to, cyclopropenylmethyl, 2-cyclopropenylethan-1-yl, 2-cyclopropenylethen-1-yl, pentylmethyl, 2-pentenylethan-1-yl, 2-pentenylethen-1-yl, and the like. Where specific alkyl moieties are intended, the nomenclature cycloalkenyl alkanyl, cycloalkenyl alkenyl and/or cycloalkenyl alkynyl is used. In some embodiments, a cycloalkenyl alkyl group is (C3-C30) cycloalkenyl alkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the cycloalkenyl alkyl group is (C1-C10) alkyl and the cycloalkenyl moiety is (C3-C20) cycloalkenyl. In other embodiments, a cycloalkenyl alkyl group is (C4-C20) cycloalkenyl alkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the cycloalkenyl alkyl group is (C1-C8) alkyl and the cycloalkenyl moiety is (C3-C12) cycloalkenyl. In still other embodiments, a cycloalkenyl alkyl group is (C4-C10) cycloalkenyl alkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the cycloalkenyl alkyl group is (C1-C4) alkyl and the cycloalkenyl moiety is (C3-C6) cycloalkenyl.


“Alkynyl,” by itself or as part of another substituent refers to an unsaturated branched, or straight-chain monovalent hydrocarbon radical having at least one carbon-carbon triple bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkyne. Typical alkynyl groups include, but are not limited to, ethynyl; propynyls such as prop-1-yn-1-yl, prop-2-yn-1-yl, etc.; butynyls such as but-1-yn-1-yl, but-1-yn-3-yl, but-3-yn-1-yl, etc.; and the like. In some embodiments, an alkynyl group comprises from 1 to 20 carbon atoms (C2-C20 alkynyl). In other embodiments, an alkynyl group comprises from 1 to 10 carbon atoms (C2-C10 alkynyl). In still other embodiments, an alkynyl group comprises from 1 to 6 carbon atoms (C2-C6 alkynyl) or 1 to 4 carbon atoms (C2-C4 alkynyl). C2-C6 alkynyl is also known as “lower alkynyl”.


“Alkoxy,” by itself or as part of another substituent, refers to a radical of the formula —O—R199, where R199 is alkyl or substituted alkyl as defined herein. In some embodiments, R199 is a C1-C3 alkyl substituent. In some embodiments, R199 is a C1-C2 alkyl substituent. In some embodiments, R199 is a C1 alkyl substituent.


“Acyl” by itself or as part of another substituent refers to a radical —C(O)R200, where R200 is hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroalkyl, substituted heteroalkyl, heteroarylalkyl or substituted heteroarylalkyl as defined herein. Representative examples include, but are not limited to formyl, acetyl, cyclohexylcarbonyl, cyclohexylmethylcarbonyl, benzoyl, benzylcarbonyl and the like.


“Aryl,” by itself or as part of another substituent, refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system, as defined herein. Typical aryl groups include, but are not limited to, groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene and the like. In some embodiments, an aryl group comprises from 6 to 18 carbon atoms (C6-C18 aryl). In other embodiments, an aryl group comprises from 6 to 15 carbon atoms (C6-C15 aryl). In still other embodiments, an aryl group comprises from 6 to 10 carbon atoms (C6-C10 aryl).


“Arylalkyl,” by itself or as part of another substituent, refers to an acyclic alkyl group in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl group, as defined herein. That is, arylalkyl can also be considered as an alkyl substituted by aryl. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, 2-phenylethen-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, 2-naphthylethen-1-yl, naphthobenzyl, 2-naphthophenylethan-1-yl and the like. Where specific alkyl moieties are intended, the nomenclature arylalkanyl, arylalkenyl and/or arylalkynyl is used. In some embodiments, an arylalkyl group is (C6-C30) arylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is (C1-C10) alkyl and the aryl moiety is (C6-C18) aryl. In other embodiments, an arylalkyl group is (C6-C18) arylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is (C1-C8) alkyl and the aryl moiety is (C6-C12) aryl. In still other embodiments, an arylalkyl group is (C6-C14) arylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is (C1-C5) alkyl and the aryl moiety is (C6-C10) aryl.


“Carbocyclic,” or “Carbocyclyl,” by itself or as part of another substituent, refers to a saturated or partially saturated, aromatic, cyclic monovalent hydrocarbon radical, including cycloalkyl, cycloalkenyl, and cycloalkynyl as defined herein. Typical carbocyclyl groups include, but are not limited to, groups derived from cyclopropane, cyclobutane, cyclopentane, cyclohexane, and the like. In some embodiments, the cycloalkyl group comprises from 3 to 10 ring atoms (C3-C10 cycloalkyl). In other embodiments, the cycloalkyl group comprises from 3 to 7 ring atoms (C3-C7 cycloalkyl). The carbocyclyl may be further substituted by one or more heteroatoms including, but not limited to, N, P, O, S, and Si, which attach to the carbon atoms of the cycloalkyl via monovalent or multivalent bond.


“Heteroalkyl,” by themselves or as part of other substituents, refer to alkyl groups, in which one or more of the carbon atoms, are each, independently of one another, replaced with the same or different heteroatoms or heteroatomic groups. Each atom in the linear heteroalkyl forms a member of the heteroalkyl. For example —CH2—O—CH2—CH2—O—CH3 is a six-membered heteroalkyl. In some embodiments, the heteroalkyl group comprises from 2 to 20 members. In other embodiments, the heteroalkyl group comprises from 2 to 15 members. In some embodiments, the heteroalkyl group comprises from 2 to 10 members. In other embodiments, the heteroalkyl group comprises from 2 to 5 members. Typical heteroatoms or heteroatomic groups, which can replace the carbon atoms include, but are not limited to, —O—, —S—, —N—, —Si—, —NH—, —S(O)—, —S(O)2—, —S(O)NH—, —S(O)2NH— and the like and combinations thereof. The heteroatoms or heteroatomic groups may be placed at any interior position of the alkyl group. Typical heteroatomic groups which can be included in these groups include, but are not limited to, —O—, —S—, —O—O—, —S—S—, —O—S—, —NR201R202—, =N—N=, —N═N—, —N═N—NR203R204, —PR205—, —P(O)2—, —POR206—, —O—P(O)2—, —SO—, —SO2—, —SnR207R2 08— and the like, where R201, R202, R203, R204, R205, R206, R207 and R208 are independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, cycloalkyl, substituted cycloalkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl or substituted heteroarylalkyl. Preferably the heteroatoms, which can replace the carbon atoms include —O—, —S—, —N—, or —NH—.


“Heterocyclic ring” or “Heterocyclyl” by itself or as part of another substituent, refers to a carbocyclic radical in which one or more carbon atoms are independently replaced with the same or different heteroatom. The heterocyclyl may be a cycloheteroaryl or a cycloheteroalkyl. The heterocyclyl may be further substituted by one or more heteroatoms including, but not limited to, N, P, O, S, and Si, which attach to the carbon atoms of the heterocyclyl via monovalent or multivalent bond. Typical heteroatoms to replace the carbon atom(s) include, but are not limited to, N, P, O, S, Si, etc. Typical heterocyclyl groups include, but are not limited to, groups derived from epoxides, azirines, thiiranes, imidazolidine, morpholine, piperazine, piperidine, pyrazolidine, pyrrolidone, quinuclidine, and the like. In preferred embodiments, heterocyclyl groups include, but are not limited to tetrahydrofuran, furan, pyrrolidine, pyridine, pyrazol, indolizine, quinoline, thiadiazol, oxadiazol and 1,2,4-oxadiazol. In some embodiments, the heterocyclyl group comprises from 3 to 20 ring atoms (3-20 membered heterocyclyl). In preferred embodiments, the heterocyclyl group comprises from 3 to 14 ring atoms (3-14 membered heterocyclyl). In more preferred embodiments, the heterocyclyl group comprises from 3 to 10 ring atoms (3-10 membered heterocyclyl). In still more preferred embodiments, the heterocyclyl group comprise from 5 to 7 ring atoms (5-7 membered heterocyclyl). A cycloheteroalkyl group may be substituted at a heteroatom, for example, a nitrogen atom, with a (C1-C6) alkyl group. As specific examples, N-methyl-imidazolidinyl, N-methyl-morpholinyl, N-methyl-piperazinyl, N-methyl-piperidinyl, N-methyl-pyrazolidinyl and N-methyl-pyrrolidinyl are included within the definition of “heterocyclyl.” A heterocyclyl group may be attached to the remainder of the molecule via a ring carbon atom or a ring heteroatom.


“Halo,” by itself or as part of another substituent refers to a radical —F, —Cl, —Br or —I.


“Heteroaryl,” by itself or as part of another substituent, refers to a monovalent heteroaromatic radical derived by the removal of one hydrogen atom from a single atom of a parent heteroaromatic ring systems, as defined herein. Typical heteroaryl groups include, but are not limited to, groups derived from acridine, β-carboline, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, dihydroindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, 1,5-naphthyridine, 1,6-naphthyridine, 1,8-naphthyridine, oxadiazole, oxazole, perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine, furan, 2-benzofuran, 1,3-dihydro-2-benzofuran, quinazoline, quinoline, quinolizine, quinoxaline, tetrazole, thiadiazole, thiazole, thiophene, triazole, xanthene, and the like. In some embodiments, the heteroaryl group comprises from 5 to 20 ring atoms (5-20 membered heteroaryl). In other embodiments, the heteroaryl group comprises from 5 to 10 ring atoms (5-10 membered heteroaryl). Exemplary heteroaryl groups include those derived from furan, thiophene, pyrrole, benzothiophene, benzofuran, benzimidazole, indole, pyridine, pyrazole, quinoline, imidazole, oxazole, isoxazole and pyrazine.


“Heteroarylalkyl” by itself or as part of another substituent refers to an acyclic alkyl group in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heteroaryl group. Where specific alkyl moieties are intended, the nomenclature heteroarylalkanyl, heteroarylakenyl and/or heteroarylalkynyl is used. In some embodiments, the heteroarylalkyl group is a 4-30 membered heteroarylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the heteroarylalkyl is (C1-C10) alkyl and the heteroaryl moiety is a 5-20-membered heteroaryl. In other embodiments, the heteroarylalkyl is a 5-15 membered heteroarylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety is (C1-C5) alkyl and the heteroaryl moiety is a 5-10 membered heteroaryl.


“Salt” refers to a salt of a compound, which possesses the desired pharmacological activity of the parent compound. Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine and the like.


“Solvate” means a compound formed by solvation (the combination of solvent molecules with molecules or ions of the solute), or an aggregate that consists of a solute ion or molecule, i.e., a compound of the present invention, with one or more solvent molecules. When water is the solvent, the corresponding solvate is “hydrate”. In one embodiment, each compound of the present invention is a solvate.


“Substituted,” when used to modify a specified group or radical, means that one or more hydrogen atoms of the specified group or radical are each, independently of one another, replaced with the same or different substituent(s). Substituent groups useful for substituting saturated carbon atoms in the specified group or radical include, but are not limited to —Ra, halo, —O—, ═O, —ORb, —SRb, —S—, ═S, —NRcRc, =NRb, =N—ORb, trihalomethyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN, —OCN, —SCN, —NO, —NO2, ═N2, —N3, —S(O)2Rb, —S(O)2NRb, —S(O)2O—, —S(O)2ORb, —OS(O)2Rb, —OS(O)2O—, —OS(O)2ORb, —P(O)(O)2, —P(O)(ORb)(O), —P(O)(ORb)(ORb), —C(O)Rb, —C(S)Rb, —C(NRb)Rb, —C(O)O—, —C(O)ORb, —C(S)ORb, —C(O)NRcRc, —C(NRb)NRcRc, —OC(O)Rb, —OC(S)Rb, —OC(O)O, —OC(O)ORb, —OC(S) ORb, —NRbC(O)Rb, —NRbC(S)Rb, —NRbC(O)O—, —NRbC(O)ORb, —NRbC(S)ORb, —NRbC(O)NRcRc, —NRbC(NRb)Rb and —NRbC(NRb)NRcRc, where Ra is selected from the group consisting of alkyl, cycloalkyl, heteroalkyl, cycloheteroalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl; each Rb is independently hydrogen or Ra; and each Rc is independently Rb or alternatively, the two Rcs may be taken together with the nitrogen atom to which they are bonded to form a 4-, 5-, 6- or 7-membered cycloheteroalkyl which may optionally include from 1 to 4 of the same or different additional heteroatoms selected from the group consisting of O, N and S. As specific examples, —NRcRc is meant to include —NH2, —NH-alkyl, N-pyrrolidinyl and N-morpholinyl. As another specific example, a substituted alkyl is meant to include -alkylene-O-alkyl, -alkylene-heteroaryl, -alkylene-cycloheteroalkyl, -alkylene-C(O)ORb, -alkylene-C(O)NRbRb, and —CH2—CH2—C(O)—CH3. The one or more substituent groups, taken together with the atoms to which they are bonded, may form a cyclic ring including cycloalkyl and cycloheteroalkyl.


Similarly, substituent groups useful for substituting unsaturated carbon atoms in the specified group or radical include, but are not limited to, —Ra, halo, —O, —ORb, —SRb, —S—, —NRcRc, trihalomethyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN, —OCN, —SCN, —NO, —NO2, —N3, —S(O)2Rb, —S(O)2O, —S(O)2ORb, —OS(O)2Rb, —OS(O)2 O, —OS(O)2ORb, —P(O)(O)2, —P(O)(ORb)(O), —P(O)(ORb)(ORb), —C(O)Rb, —C(S)Rb, —C(NRb)Rb, —C(O)O, —C(O)ORb, —C(S)ORb, —C(O)NRcRc, —C(NRb)NRcRc, —OC(O)Rb, —OC(S)Rb, —OC(O)O, —OC(O)ORb, —OC(S)ORb, —NRbC(O)Rb, —NRbC(S)Rb, —NRbC(O)O, —NRbC(O)ORb, —NRbC(S)O Rb, —NRbC(O)NRcRc, —NRbC(NRb)Rb and —NRbC(NRb)NRcRc, where Ra, Rb and Rc are as previously defined.


Substituent groups useful for substituting nitrogen atoms in heteroalkyl and cycloheteroalkyl groups include, but are not limited to, —Ra, —O, —ORb, —SRb, —S, —NRcRc, trihalomethyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN, —NO, —NO2, —S(O)2Rb, —S(O)2O, —S(O)2ORb, —OS(O)2Rb, —OS(O)2O, —OS(O)2ORb, —P(O)(O)2, —P(O)(ORb)(O), —P(O)(ORb)(ORb), —C(O)Rb, —C(S)Rb, —C(NRb)Rb, —C(O)ORb, —C(S)O Rb, —C(O)NRcRc, —C(NRb)NRcRc, —OC(O)Rb, —OC(S)Rb, —OC(O)ORb, —OC(S)ORb, —NRbC(O)Rb, —NRbC(S)Rb, —NRbC(O)ORb, —NRbC(S)ORb, —NRbC(O)NRcRc, —NRbC(NRb)Rb and —NRbC(NRb)NRcRc, where Ra, Rb and Rc are as previously defined.


Substituent groups from the above lists useful for substituting other specified groups or atoms will be apparent to those of skill in the art.


The term “substituted” specifically envisions and allows for one or more substitutions that are common in the art. However, it is generally understood by those skilled in the art that the substituents should be selected so as to not adversely affect the useful characteristics of the compound or adversely interfere with its function. Suitable substituents may include, for example, halogen groups, perfluoroalkyl groups, perfluoroalkoxy groups, alkyl groups, alkenyl groups, alkynyl groups, hydroxy groups, oxo groups, mercapto groups, alkylthio groups, alkoxy groups, aryl or heteroaryl groups, aryloxy or heteroaryloxy groups, arylalkyl or heteroarylalkyl groups, arylalkoxy or heteroarylalkoxy groups, amino groups, alkyl- and dialkylamino groups, carbamoyl groups, alkylcarbonyl groups, carboxyl groups, alkoxycarbonyl groups, alkylaminocarbonyl groups, dialkylamino carbonyl groups, arylcarbonyl groups, aryloxycarbonyl groups, alkylsulfonyl groups, arylsulfonyl groups, cycloalkyl groups, cyano groups, C1-C6 alkylthio groups, arylthio groups, nitro groups, keto groups, acyl groups, boronate or boronyl groups, phosphate or phosphonyl groups, sulfamyl groups, sulfonyl groups, sulfinyl groups, and combinations thereof. In the case of substituted combinations, such as “substituted arylalkyl,” either the aryl or the alkyl group may be substituted, or both the aryl and the alkyl groups may be substituted with one or more substituents. Additionally, in some cases, suitable substituents may combine to form one or more rings as known to those of skill in the art.


In a preferred embodiment, a “substituted” cycloalkyl, heterocyclic ring or aryl group includes the substitution with one or more substituent each substituent independently selected from the group comprising —F, —Cl, —Br, —NO2, —O—N═O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkoxy, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 cycloalkyl C1-C10 alkyl, optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, optionally substituted C6-C18 aryl, optionally substituted C6-C18 aryl C1-C10 alkyl, optionally substituted 5-20 membered heteroaryl, optionally substituted 5-20 membered heteroaryl C1-C10 alkyl, (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, optionally substituted 2-20 membered heteroalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; and —CCl3.


More preferably, a “substituted” alkyl, heteroalkyl, cycloalkyl, heterocyclic ring or aryl group includes the substitution with one or more substituent each substituent independently selected from the group comprising —F, —Cl, —Br, —NO2, —O—N═O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, C1-C4 alkyl, C1-C4 alkoxy, C2-C4 alkenyl, C3-C10 cycloalkyl, C3-C10 cycloalkyl C1-C2 alkyl, 3-10 membered cycloheteroalkyl or 3-10 membered cycloheteroalkyl C1-C2 alkyl, C6-C10 aryl, C6-C10 aryl C1-C2 alkyl, 5-10 membered heteroaryl, 5-10 membered heteroaryl C1-C2 alkyl, (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of C1-C4 alkyl, C2-C4 alkenyl, C3-C10 cycloalkyl, C6-C10 aryl, 5-10 membered heteroaryl, 2-10 membered heteroalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; and —CCl3.


Preferably, a “substituted” cycloalkyl, heterocyclic ring or aryl group includes the substitution with one, two or three substituents, still more preferably with one or two substituents.


The term “optionally substituted” denotes the presence or absence of the substituent group(s). That is, it means “substituted or unsubstituted”. For example, optionally substituted alkyl includes both unsubstituted alkyl and substituted alkyl. The substituents used to substitute a specified group can be further substituted, typically with one or more of the same or different groups selected from the various groups specified above.


“Carrier” refers to a diluent, excipient or vehicle with which a compound is administered.


“NLRP3” is meant to include, without limitation, nucleic acids, polynucleotides, oligonucleotides, sense and antisense polynucleotide strands, complementary sequences, peptides, polypeptides, proteins, homologous and/or orthologous NLRP3 molecules, isoforms, precursors, mutants, variants, derivatives, splice variants, alleles, different species, and active fragments thereof.


A “modulator” herein refers to a compound that can regulate the activity of NLRP3. Such regulation includes activating NLRP3, blocking NLRP3, or potentiating/reducing the activation of NLRP3. That is, the modulators include agonists, antagonists, enhancers, etc.


An “activator” or an “agonist” of NLRP3 herein refers to a compound that can (positively) modulate the activity of NLRP3. Such modulation includes activating NLRP3, or increasing the activation of NLRP3. An “activator” or an “agonist” includes compounds that, at the protein level, directly bind or modify NLRP3 such that an activity of NLRP3 is increased, e.g., by activation, stabilization, altered distribution, or otherwise.


“Treating” or “treatment” of any condition, disease or disorder refers to ameliorating the condition, disease or disorder (i.e., arresting or reducing the development of the condition, disease or disorder or at least one of the clinical symptoms thereof). In other embodiments “treating” or “treatment” refers to ameliorating at least one physical parameter, which may not be discernible by the patient. In yet other embodiments, “treating” or “treatment” refers to inhibiting the condition, disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter) or both. In yet other embodiments, “preventing,” “prevention,” “treating” or “treatment” refers to delaying the onset of the condition, disease or disorder.


“Therapeutically effective amount” means the amount of the present compound, a pharmaceutically acceptable salt and/or hydrate and/or cocrystal thereof; e.g., a compound, such as niclosamide or a pharmaceutically acceptable salt and/or hydrate and/or cocrystal thereof that, when administered to a subject for treating a condition, disease or disorder, is sufficient to effect such treatment for the condition, disease or disorder. The “therapeutically effective amount” will vary depending on the compound, the condition, disease or disorder and its severity and the age, weight, etc., of the subject to be treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.


“Subject” refers to an animal, including, but not limited to, a primate (e.g., human), monkey, cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms “subject” and “patient” are used interchangeably herein in reference, for example, to a mammalian subject, such as a human.


“Pharmaceutical composition” refers to a mixture of a compound described herein with other chemical components (referred to collectively herein as “excipients”), such as carriers, stabilizers, diluents, dispersing agents, suspending agents, and/or thickening agents. The pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to: rectal, oral, intravenous, aerosol, parenteral, ophthalmic, pulmonary, and topical administration. A further technique of administering is the intratumoral injection.


“Excipient” or “pharmaceutically acceptable excipient” means a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, carrier, solvent, or encapsulating material. In one embodiment, each component is “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, e.g., Remington: The Science and Practice of Pharmacy, 21st ed.; Lippincott Williams & Wilkins: Philadelphia, P A, 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 and Formulation, 2nd ed.; Gibson Ed.; CRC Press LLC: Boca Raton, F L, 2009.


Embodiments of the Compounds

According to the first aspect, the present invention relates to a compound for use in the prevention or treatment of a disease by activating the NLRP3 inflammasome, wherein the disease is selected from a group comprising infectious diseases or cancer, and


wherein the compound is a compound of Formula (I), Formula (I′) or Formula (I″)




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


A and A′ are independently selected from each other an optionally substituted C6-C18 aryl; optionally substituted 5-20 membered heteroaryl; optionally substituted C3-C20 cycloalkyl; optionally substituted 3-20 membered cycloheteroalkyl, preferably an optionally substituted C1-C10 aryl; an optionally substituted 5-10 membered heteroaryl; an optionally substituted C3-C10 cycloalkyl; or an optionally substituted 3-10 membered cycloheteroalkyl, more preferably an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl. A and A′ are more preferably as defined herein below.


B is O or S, preferably O;


and X and X′ are each independently present or not, wherein X and X′ are each independently an optionally substituted C1-C10 alkyl group; preferably an C1-C5 alkyl group, or an C1-C5 alkanol group; more preferably selected from —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


R6 and R7 are each independently hydrogen, an optionally substituted C1-C10 alkyl or R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


or optionally,


R7, N and X when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


wherein A is as herein above and below.


X and X′ are each independently a linker group which bonds the nitrogen of the urea to the A or A′ respectively. If X or X′ is not present, the nitrogen of the urea is directly bound to the A or A′, respectively.


In one embodiment, the compound of Formula (I), (I′) or I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 1, 0 to 2, 0 to 3 or 0 to 4, or C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, preferably only one of X and X′ is present and is a C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—; and wherein


R6 and R7 are each independently hydrogen, a C1-C10 alkyl, preferably a C1-C5 alkyl, C1-C4 alkyl, C1-C3 alkyl, C1-C2 alkyl, more preferably hydrogen or CH3, even more preferably wherein R6 and R7 are each hydrogen.


In one further embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 1, 0 to 2, 0 to 3 or 0 to 4, or C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, preferably only one of X and X′ is present and is a C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—; and wherein


R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


preferably selected from




embedded image


and


wherein A and A′ are preferably as defined herein below.


In one further embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 1, 0 to 2, 0 to 3 or 0 to 4, or C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, preferably only one of X and X′ is present and is a C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—; and wherein R7, N and X when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


and wherein A and A′ are preferably as herein below.


In one embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 2, preferably 0 or 1, more preferably 0.


In one embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 2, preferably 0 or 1, more preferably 0, and R6 and R7 are each independently hydrogen, wherein A and A′ are preferably as defined herein below.


In one further embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises none of X and X′; and wherein R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


preferably selected from




embedded image


and wherein A is preferably as defined herein below.


In one further embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises none of X and X′, and wherein R7, N and X when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


and wherein A is as herein above and below.


In one preferred embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently a C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—,


preferably only one of X and X′ is present and is a C1-C3 alkyl, a C1-C2 alkyl, preferably selected from the group consisting of, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, and A and A′ are each independently optionally substituted isoquinoline, quinoline, benzene, pyridine, 1,5-naphthyridine, isothiazole, or oxadiazole. A and A′ may be identical to each other or different to each other. In one preferred embodiment, A and A′ are different to each other. A and A′ are preferably as defined herein below.


In one preferred embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises A and A′, wherein A and A′ are each independently optionally substituted benzene, imidazolidine, morpholine, piperazine, piperidine, pyrazolidine, pyrrolidone, quinuclidine, tetrahydrofuran, furan, pyrrolidine, pyridine, pyrazol, indolizine, quinoline, thiadiazol, oxadiazol, 1,2,4-oxadiazol, acridine, β-carboline, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, dihydroindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, 1,5-naphthyridine, 1,6-naphthyridine, 1,8-naphthyridine, oxadiazole, oxazole, perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine, furan, 2-benzofuran, 1,3-dihydro-2-benzofuran, quinazoline, quinoline, quinolizine, quinoxaline, tetrazole, thiadiazole, thiazole, thiophene, triazole, xanthene, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl; cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl; cyclobutadienyl, cyclohexadienyl, carbazole, benzimidazole, imidazopyridine, benzoisoxazole, tetrahydrothiophene, benzofuran, benzoxazole, or benzthiozole,


more preferably wherein A and A′ are each independently optionally substituted benzene, pyrrolidine, pyridine, pyrazole, pyridazine, pyrimidine, pyrrole, pyrrolizine, furan, 2-benzofuran, 1,3-dihydro-2-benzofuran, diazole, triazole, oxazole, oxadiazole, 1,2,4-oxadiazole, thiazole, thiophene, thiadiazole, imidazole, indazole, indole, indoline, isoindole, isoindoline, dihydroindoline, quinoline, isoquinoline, isothiazole, isoxazole, naphthyridine, 1,5-naphthyridine, 1,6-naphthyridine, 1,8-naphthyridine, phthalazine, purine, quinazoline, quinoxaline, benzimidazole, benzoisoxazole, tetrahydrothiophene, benzoxazole or benzthiazole,


even more preferably wherein A and A′ are each independently optionally substituted quinoline, isoquinoline, indole, benzene, pyridine, pyrazole, pyrrol, furan, thiophene; naphthyridine,1,5-naphthyridine, 1,6-naphthyridine, 1,8-naphthyridine, isothiazole, or oxadiazole,


likewise more preferably wherein A and A′ are each independently optionally substituted quinoline, benzene, pyridine, naphthyridine, 1,5-naphthyridine, isothiazole, oxadiazole, or thiophene,


most preferably wherein A and A′ are each independently optionally substituted isoquinoline, quinoline, benzene, pyridine, 1,5-naphthyridine, isothiazole, or oxadiazole.


A and A′ may be identical to each other or different to each other. In one preferred embodiment, A and A′ are different to each other.


In one embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently —(CH2)m—, wherein each m is independently an integer from 0 to 2, preferably 0 or 1, more preferably 0.


In one preferred embodiment, the compound of Formula (I), (I′) or (I″) of the invention comprises X and X′, wherein X and X′ are each independently a C1-C3 alkyl, a C1-C2 alkyl, preferably, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—,


preferably only one of X and X′ is present and is a C1-C3 alkyl, a C1-C2 alkyl, preferably selected from the group consisting of, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, and A and A′ are each independently optionally substituted isoquinoline, quinoline, benzene, pyridine, 1,5-naphthyridine, 1,6-naphthyridine, 1,8-naphthyridine, isothiazole, or oxadiazole. A and A′ may be identical to each other or different to each other. In one preferred embodiment, A and A′ are different to each other.


Preferably, according to the preferred embodiment, A and A′ are each independently optionally substituted quinoline, isoquinoline, benzene, pyridine, naphthyridine, 1,5-naphthyridine, 1,6-naphthyridine, 1,8-naphthyridine, isothiazole, oxadiazole, or thiophene, preferably selected from groups as defined in the following in any of sections a) to h):

    • a) an optionally substituted benzene group of the following structure:




embedded image


wherein R1-R5 are each independently selected from the group consisting of:


hydrogen; —OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —OCH(CH3)2; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, —CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —NH—CH(CH3)2, —NH-cyclopropyl, —CH3, CH2CH3, CHCH(CH3)2; —C(O)—C2H5; —C(O)—CH3; —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl, optionally substituted C1-C10 cycloalkyl, optionally substituted C1-C10 cycloheteroalkyl,




embedded image


an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl;


NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH—CH(CH3)2, —NH-cyclopropyl, C1-C5 alkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2;


—C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; —CH2NHS(O)2CH3;


—NHR43, wherein R43 is selected from




embedded image


wherein R60 is selected from hydrogen or —CH3;




embedded image


wherein


R53 is preferably selected from the group consisting of hydrogen, an optionally substituted C1-C10 alkyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted 3-20 membered cycloheteroalkyl, optionally substituted C6-C18 aryl or optionally substituted 5-20 membered heteroaryl, preferably an optionally substituted C6-C14 aryl or 5-20 membered heteroaryl; more preferably from hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CHF2, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO,




embedded image


with X=0 or 1, preferably 1;




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NH2, NO2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above;


and


optionally, wherein R1 and R2, R2 and R3, R3 and R4, or R4 and R5 together, preferably R2 and R3 together, form an optionally substituted five- or six-membered cyclic or heterocyclic ring, preferably selected from




embedded image


Preferably X is not present in Formulae (I), (I′) or (I″) in case A or A′ is an optionally substituted benzene group as defined under a). Likewise preferably, one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Preferably either all of R1 to R5 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R1 to R5 are different from hydrogen and selected from substitutions defined or presented herein above under a) for R1 to R5, and remaining substituents of R1 to R5 are hydrogen.

    • b) an optionally substituted pyridine group of one of the following structures:




embedded image


wherein R44-R47 are each independently selected from the group consisting of:


hydrogen, —OH, —OCH3, CH3, F, Cl, Br, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CN; NO2; NH2; —CH3, CH(CH3)2; —CH2CH3; —CH2OH; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3;




embedded image


—NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH-cyclopropyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine; an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; optionally substituted C1-C10 cycloalkyl, optionally substituted C1-C10 cycloheteroalkyl,


—C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; —CH2NHS(O)2CH3; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl,




embedded image


or


—NHR43, wherein R43 is selected from




embedded image


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted pyridine group as defined under b) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Preferably either all of R44 to R47 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R45 to R47 are different from hydrogen and selected from substitutions defined or presented herein above under b) for R44 to R47, and remaining substituents of R44 to R47 are hydrogen.

    • c) an optionally substituted quinoline or an isoquinoline group, preferably of one of the the following structures:




embedded image


embedded image


wherein


R21 to R26 are each independently selected from the group consisting of:


hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine; —C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; —CH2NHS(O)2CH3; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl,




embedded image


an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; optionally substituted C1-C10 cycloalkyl, optionally substituted C1-C10 cycloheteroalkyl; —NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH-cyclopropyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; —NHR43, wherein R43 is selected from




embedded image


or more preferably wherein R21 to R26 are each hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3;


or even more preferably wherein R21 to R26 are each hydrogen, or —F; Cl, Br.


Preferably in case A and/or A′ is an optionally substituted quinoline or an isoquinoline group optionally substituted quinoline or an isoquinoline group in Formula (I), (I′) or (I″) as defined in c) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Preferably either all of R21 to R26 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R21 to R26 are different from hydrogen and selected from substitutions defined or presented herein above under c) for R21 to R26 and remaining substituents of R21 to R26 are hydrogen;

    • d) a naphthyridine, preferably a 1,5 naphthyridine group, a 1,6 naphthyridin group or a 1,8 naphthyridin group of any of the following structures:




embedded image


embedded image


wherein


R48 to R52 are each independently selected from the group consisting of:


hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO,


—NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH-cyclopropyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine;


—C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; —CH2NHS(O)2CH3; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl,




embedded image


an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; optionally substituted C1-C10 cycloalkyl, optionally substituted C1-C10 cycloheteroalkyl;


—NHR43, wherein R43 is selected from




embedded image


or more preferably wherein R48 to R52 are each hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3;


or even more preferably wherein R48 to R52 are each hydrogen, or —F; Cl, Br.


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted 1,5-naphthyridine group as defined under d) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Preferably either all of R48 to R52 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R48 to R52 are different from hydrogen and selected from substitutions defined or presented herein above under d) for R48 to R52 and remaining substituents R48 to R52 are hydrogen;

    • e) an oxadiazole group of one of the following formulae:




embedded image


wherein


R53 is preferably selected from the group consisting of:


hydrogen, an optionally substituted C1-C10 alkyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted 3-20 membered cycloheteroalkyl, optionally substituted C6-C18 aryl or optionally substituted 5-20 membered heteroaryl, preferably an optionally substituted C6-C14 aryl or 5-20 membered heteroaryl; more preferably from


hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, an optionally substituted C6-C10 aryl; an optionally substituted 5-10 membered heteroaryl; an optionally substituted C1-C10 cycloalkyl, an optionally substituted C1-C10 cycloheteroalkyl,




embedded image


with X=0 or 1, preferably 1;




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NH2, NO2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above, and x is 0, 1 or 2;


and wherein preferably either all of R54 to R58 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R54 to R58 are different from hydrogen and selected from substitutions defined or presented herein above under e) for R54 to R58 and remaining positions of R54 to R58 are hydrogen;

    • f) a thiadiazole group, preferably selected of one of the following formulae:




embedded image


wherein R53 is preferably as defined above in section e);


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted thiadiazole group as defined under f) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • g) an isothiazole group of the following formula:




embedded image


wherein R66 and R67 are each selected independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —Cl, —Br, preferably wherein either of R66 or R67 is hydrogen and the other residue is selected from —CH3, —C2H5, —CF3, —CHF2, —F, —Cl, —Br;


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted isothiazole group as defined under g) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • h) a thiophene group, preferably selected of any of the following formulae:




embedded image


wherein R68, R69 and R70 are each preferably selected independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —C, —Br, preferably wherein either of R68, R69 or R70 is hydrogen and the other residue is selected from —CH3, —C2H5, —CF3, —CHF2, —F, —Cl, —Br


Preferably in Formula (I), (I′) or (I″) in case A and/or A′ is an optionally substituted thiophene group as defined under g) X and/or X′ are either not present, only one of X or X′ is present or both of X and X′ are present, and X and X′ or are each preferably selected from —CH2—, —C2H4—C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


In one alternatively preferred embodiment, the compound of Formula (I), (I′) or (I″) of the invention is a compound of any one of Formulae (Ic), (Ic1), (Ic2), (Ic3), or (Ic4), or a pharmaceutically acceptable salt thereof:




embedded image


wherein B=O or S, preferably O;


wherein R6 and R7 are independently each hydrogen, an optionally substituted C1-C10 alkyl, preferably a C1-C5 alkyl, or C1-3 alkyl, more preferably —H, —CH3, —C2H5, —C3H7, or —C3H9, even more preferably hydrogen or CH3, more preferably, either R6 is hydrogen and R7 is CH3, —C2H5, —C3H7, or —C3H9, or more preferably, either R7 is hydrogen and R6 is CH3, —C2H5, —C3H7, or —C3H9, or R6 and R7 are independently each hydrogen, or optionally


R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


preferably selected from




embedded image


In the compound of any one of Formulae (Ic), (Ic1), (Ic2), (Ic3), or (Ic4), R21 to R37 are preferably each independently selected from the group consisting of hydrogen, —OH, —F, —C, —Br, —NO2, —O—N═O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F; —C; —Br; —CN; NO2; NH2; —O—N═O, —N═O, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3 CH═NH, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkoxy, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 cycloalkyl C1-C10 alkyl, optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, optionally substituted C6-C18 aryl, optionally substituted C6-C18 aryl C1-C10 alkyl, optionally substituted 5-20 membered heteroaryl, optionally substituted 5-20 membered heteroaryl C1-C10 alkyl, e.g. —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, and —NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, and optionally substituted 2-20 membered heteroalkyl, or


more preferably wherein R21 to R37 are each independently selected from the group consisting of hydrogen, C1-C6 alkoxy, C1-C5 alkyl, —(C═O)CH3, —CN, C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —Cl, and —Br; or even


more preferably wherein R21 to R37 are each hydrogen.


Even more preferably, in a compound Formula (Ic), (Ic1), (Ic2), (Ic3), or (Ic4), R6 and R7 are independently each hydrogen, C1-3 alkyl, preferably hydrogen, CH3, more preferably, either R6 is hydrogen and R7 is CH3, or R6 and R7 are both hydrogen; and


R21 to R37 are each independently selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —O—N═O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2 (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, or C(NR11)R12, —NHC(O)R11, and —NHC(O)NHR11, and wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C5 alkyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, and optionally substituted 2-20 membered heteroalkyl; or


preferably wherein R21 to R37 are each independently selected from the group consisting of hydrogen, C1-C6 alkoxy, C1-C5 alkyl, C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3, —F, —Cl, and —Br; or


wherein preferably either all of R21 to R37 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R21 to R37 are different from hydrogen and selected from substitutions presented above for R21 to R37 and remaining substituents of R21 to R37 are hydrogen; or


more preferably wherein R21 to R37 are each hydrogen.


In one further alternatively preferred embodiment, the compound of Formula (I), (I′) or (I″) of the invention is a compound of any one of Formulae (Ic5), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13), or (Ic14) or a pharmaceutically acceptable salt thereof:




embedded image


embedded image


wherein R6 and R7 are independently each hydrogen, an optionally substituted C1-C10 alkyl, preferably a C1-C5 alkyl, or C1-3 alkyl, more preferably —H, —CH3, —C2H5, —C3H7, or —C3H9, even more preferably hydrogen or CH3, more preferably, either R6 is hydrogen and R7 is CH3, —C2H5, —C3H7, or —C3H9, or more preferably, either R7 is hydrogen and R6 is CH3, —C2H5, —C3H7, or —C3H9, or R6 and R7 are independently each hydrogen, or optionally


R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


preferably selected from




embedded image


In the compound of any one of Formulae (Ic5), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13), or (Ic14) R21 to R37 are preferably each independently selected from the group consisting of hydrogen, —OH, —F, —C, —Br, —NO2, —O—N═O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; CH═NH, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkoxy, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 cycloalkyl C1-C10 alkyl, optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, optionally substituted C6-C18 aryl, optionally substituted C6-C18 aryl C1-C10 alkyl, optionally substituted 5-20 membered heteroaryl, optionally substituted 5-20 membered heteroaryl C1-C10 alkyl, e.g. —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, and —NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, and optionally substituted 2-20 membered heteroalkyl; or


more preferably wherein R21 to R37 are each independently selected from the group consisting of hydrogen, C1-C6 alkoxy, C1-C5 alkyl, —(C═O)CH3, —CN, C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —Cl, and —Br; or


even more preferably wherein R21 to R37 are each hydrogen. In an even more preferred embodiment, in the compounds of any of Formulae (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic5), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13) or (Ic14), R21 to R37 are each independently selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F; —Cl; —Br; —CN; NO2; NH2; C1-C6 alkoxy, C1-C5 alkyl, preferably —CH3, CH(CH3)2; —CH2CH3; or C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3.


In a preferred embodiment, the compound of any of Formulae (Ic), (Ic5) and (Ic10) is as defined above, wherein R21, R26, R27 and R32 is preferably hydrogen.


In a preferred embodiment, the compound of any of Formulae (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13) or (Ic14), is as defined above, wherein R21, R26, R33 and R37 is preferably hydrogen.


In a preferred embodiment, the compound of any of Formulae (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic5), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13) or (Ic14), is as defined above, wherein R24 is C1-C6 alkoxy, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; C1-C5 alkyl, preferably —CH3, CH(CH3)2; —CH2CH3; —(C═O)CH3, —CN, C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —Cl, or —Br, and wherein R21 to R23 and R25 to R37 are each hydrogen.


In a preferred embodiment, the compound of any of Formulae (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic5), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13) or (Ic14), is as defined above, wherein R23 is C1-C6 alkoxy, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; C1-C5 alkyl, preferably —CH3, CH(CH3)2; —CH2CH3; —(C═O)CH3, —CN, C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, —CF3, —F, —C, or —Br, and wherein R21, R22 and R24 to R37 are each hydrogen.


In a preferred embodiment, the compound of Formula (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13) or (Ic14), is as defined above, wherein R35 is C1-C6 alkoxy, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; C1-C5 alkyl, preferably —CH3, CH(CH3)2; —CH2CH3; —(C═O)CH3, —CN, C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, —CF3, —F, —Cl, or —Br, and wherein R21 to R26, R33, R34 and R36, R37 are each hydrogen.


In a preferred embodiment, the compound of any of Formula (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13) or (Ic14), is as defined above, wherein R36 is C1-C6 alkoxy, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; C1-C5 alkyl, preferably —CH3, CH(CH3)2; —CH2CH3; —(C═O)CH3, —CN, C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, —CF3, —F, —Cl, or —Br, and wherein R21 to R26, R33, R34 and R35, R37 are each hydrogen.


In a preferred embodiment, the compound of any of Formula (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic5), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13) or (Ic14), is as defined above, wherein R22 is C1-C6 alkoxy, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; C1-C5 alkyl, preferably —CH3, CH(CH3)2; —CH2CH3; —(C═O)CH3, —CN, C1-C5 alkyl(SO2)C1-C5 alkyl, preferably —CH2(SO2)CH3, —CH2(SO2)CH2CH3, —CH2CH2(SO2)CH2CH3; —(SO2)C1-C5 alkyl, preferably —(SO2)CH3, —(SO2)CH2CH3, —CF3, —F, —Cl, or —Br, and wherein R21 and R23 to R37 are each hydrogen.


In a preferred embodiment, the compound of any of Formulae (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic5), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13) or (Ic14) is as defined above, wherein R21 to R7 are each hydrogen.


In a preferred embodiment, the compound of any of Formulae (Ic), (Ic1), (Ic2), (Ic3), (Ic4), (Ic5), (Ic6), (Ic7), (Ic8), (Ic9) (Ic10), (Ic11), (Ic12), (Ic13) or (Ic14) is one of the following structures:




embedded image


embedded image


In a second aspect, the compound of Formula (I) of the invention is a compound of Formula (Ia) or (Ia1),




embedded image


or a pharmaceutically acceptable salt thereof, wherein:


X either present or not, and wherein X, if present, is selected from a C1-C10 alkyl group, a C1-C5 alkyl group, e.g. —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or a C1-C5 alkanol group, e.g. —CH(CH2OH)—; and


R1 and R2, R2 and R3, R3 and R4, R4 and R5, when taken together form an optionally substituted five- or six-membered cycloalkyl, optionally substituted five- or six-membered cycloheteroalkyl, optionally substituted five- or six-membered aryl, or optionally substituted five- or six-membered heteroaryl, and


R6 and R7 are each independently hydrogen, an optionally substituted C1-C10 alkyl, preferably R6 and R7 are each independently hydrogen, —CH3, —C2H5, —C3H7, or —C3H9, or R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


Alternatively, R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


or optionally,


R7, N and X when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


According to one embodiment of formulae (Ia) or (Ia1), R1, R2, R3, R4, and R5 are each independently selected from the group consisting hydrogen, —F, —Cl, —Br, —NO2, —O—N═O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —OCH(CH3)2; —CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; CH(CH3)2; —CH2CH3, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkoxy, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 cycloalkyl C1-C10 alkyl, optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, optionally substituted C6-C18 aryl, optionally substituted C6-C18 aryl C1-C10 alkyl, optionally substituted 5-20 membered heteroaryl, optionally substituted 5-20 membered heteroaryl, C1-C10 alkyl, (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, or—NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, and optionally substituted 2-20 membered heteroalkyl;


Preferably, R1-R5 are each independently selected from the group consisting of hydrogen; —OH, —OCH3, —OCF3, —OCH2OH; —OCH2CH3; —OCH(CH3)2; —F; —Cl; —Br; —CN; NO2; NH2; —CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; CH(CH3)2; —CH2CH3;


—C(O)OR42, wherein R42 is selected from —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3;




embedded image


wherein R60 is selected from hydrogen or —CH3;


optionally, wherein R1 and R2, R2 and R3, R3 and R4, or R4 and R5 together, preferably R1 and R2 or R2 and R3 together, form an optionally substituted five- or six-membered cyclic or heterocyclic ring, preferably selected from




embedded image


In a compound of Formula (Ia) or (Ia1), A is preferably an optionally substituted C6-C18 aryl, an optionally substituted 5-10 membered heteroaryl, optionally substituted 5-10 membered heteroaryl, preferably an optionally substituted benzene, an optionally substituted oxadiazole, an optionally substituted thiadiazole, an optionally substituted isothiazole, or an optionally substituted thiophene.


Preferably, A in a compound of Formula (Ia) or (Ia1) is selected from an optionally substituted benzene, an optionally substituted oxadiazole, an optionally substituted thiadiazole, an optionally substituted isothiazole, or an optionally substituted thiophene as defined in the following under sections a) to e):

    • a) a benzene group of the following structure:




embedded image


wherein R61-R65 are each independently selected from the group consisting of:


hydrogen; —OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —OCH(CH3)2; —F; —Cl; —Br; —CN; NO2; NH2; —CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —C(O)—C2H5; —C(O)—CH3; —C(O)-aziridine; —C(O)-azetidine; —C(O)-pyrrolidine; —CH2NHS(O)2CH3; —C(CH3)═NOH; —S(O)2-cyclopentyl; —S(O)2—CH3; —S(O)2-cyclopentyl; pyrrolidinyl,




embedded image


or


—NHC(O)—R41, wherein R41 is selected from C1-C10 cycloalkyl, C1-C10 cycloheteroalkyl, furan, tetrahydrofuran, cyclopentyl, cyclobutyl, cyclopropyl, —NH—CH(CH3)2, —NH-cyclopropyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —CH3, CH2CH3, CHCH(CH3)2; or


—C(O)OR42, wherein R42 is selected from C1-C5 alkyl, including —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; or


—NHR43, wherein R43 is selected from




embedded image


wherein R60 is selected from hydrogen or —CH3; or




embedded image


wherein R53 is preferably selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —




embedded image


with X=0 or 1, preferably 1;




embedded image


or R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —C, —Br, NO2, NH2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above, and x is 0, 1 or 2;


and


optionally, wherein R61 and R62, R62 and R63, R63 and R64, or R64 and R65 together, preferably R62 and R63 together, form an optionally substituted five- or six-membered cyclic or


heterocyclic ring, preferably selected from




embedded image


wherein preferably either all of R1 to R5 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R1 to R5 are different from hydrogen and selected from substitutions defined above for R1 to R5 and remaining substituents of R1 to R5 are hydrogen;


Likewise, preferably either all of R61 to R65 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R1 to R5 are different from hydrogen and selected from substitutions defined above for R61 to R65 and remaining substituents of R61 to R65 are hydrogen;


Likewise preferably, X is then not present in Formula (Ia) and (Ia1), or may be selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—. More preferably, X is not present.

    • b) an oxadiazole group, preferably selected from one of the following structures:




embedded image


wherein


R53 is preferably selected from the group consisting of is hydrogen, an optionally substituted C1-C10 alkyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted 3-20 membered cycloheteroalkyl, optionally substituted C6-C18 aryl or optionally substituted 5-20 membered heteroaryl, preferably an optionally substituted C6-C14 aryl or 5-20 membered heteroaryl; more preferably from hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above, and x is 0, 1 or 2;


and wherein preferably either all of R54 to R58 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R54 to R58 are different from hydrogen and selected from substitutions presented above and remaining positions of R54 to R58 are selected from hydrogen;


Preferably, X is then selected in Formula (Ia) or (Ia1), from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Alternatively, X may not be present.

    • c) a thiadiazole group, preferably selected from one of the following structures:




embedded image


wherein R is preferably as defined herein above.


Preferably, X is then selected in formula (Ia) or (Ia1), from —CH2-, -C2H4-, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Alternatively, X may not be present.

    • d) an isothiazole group, preferably selected from of the following structures:




embedded image


wherein R66 and R67 are each selected independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —Cl, —Br, preferably wherein either of R66 or R67 is hydrogen and the other residue is selected from —CH3, —C2H5, —CF3, —CHF2, —F, —Cl, —Br.


Preferably, X is then selected in Formula (Ia) or (Ia1), from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.

    • e) a thiophene group, preferably selected from of the following structures




embedded image


wherein R68, R69 and R70 are each selected independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —F, —C, —Br, preferably wherein either of R68, R69 or R70 is hydrogen and the other residue is selected from —CH3, —C2H5, —CF3, —CHF2, —F, —C, —Br.


Preferably, X is then selected in Formula (Ia) or (Ia1), from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—.


Alternatively, X may not be present.


According to one particularly preferred embodiment, the present invention relates to a compound of Formula (Ia) or (Ia1),




embedded image


or a pharmaceutically acceptable salt thereof, wherein A is the following optionally substituted aryl group:




embedded image


and X is preferably a C1-C10 alkyl group, e.g. is selected from —CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, or —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, or X is not present; and wherein


preferably R1, R2, R3, R4, R5, R3 and R9 are each independently hydrogen, —F, —Cl, —OH, —NH2, —S(O)2CH3, —(C═O)OH, —NH(C═O)NH2, —(C═O)NH2, —CHO, —OCHO, —NCHO optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C6 alkenyl, optionally substituted C3-C7 cycloalkyl, optionally substituted C3-C7 cycloalkyl C1-C5 alkyl, optionally substituted C2-C1 cycloheteroalkyl or C2-C14 cycloheteroalkyl C1-C5 alkyl, optionally substituted C6-C14 aryl, optionally substituted C6-C14 aryl C1-C5 alkyl, optionally substituted C2-C14 heteroaryl, optionally substituted C2-C14 heteroaryl C1-C5 alkyl, (C═O)R11, (C═O)OR”, (C═O)NR11R12, (SO2)R11, C(NR11)R12, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C1 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, optionally substituted 2-20 membered heteroalkyl; —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —OCH(CH3)2; —F; —Cl; —Br; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; and —CCl3; —CN; NO2; NH2; —CH3, CH(CH3)2; —CH2CH3; or


—C(O)OR42, wherein R42 is selected from —CH3; —CH2CH3, —CH2CH2CH3; —CH(CH3)2; —CH2CH2CH2CH3; or




embedded image


wherein R60 is selected from hydrogen or —CH3, or, alternatively,


R1 and R2, R2 and R3, R3 and R4, R4 and R5, or R8 and R9, preferably R1 and R2 or R2 and R3 together, more preferably R2 and R3 together, when taken together form an optionally substituted five- or six-membered cycloalkyl, optionally substituted five- or six-membered cycloheteroalkyl, optionally substituted five- or six-membered aryl, or optionally substituted five- or six-membered heteroaryl, preferably selected from




embedded image


or


R8 and R9, when taken together form a optionally such a substituted five- or six-membered cycloalkyl, optionally substituted five- or six-membered cycloheteroalkyl, optionally substituted five- or six-membered aryl, or optionally substituted five- or six-membered heteroaryl, and


R6 and R7 are each independently hydrogen, an optionally substituted C1-C10 alkyl, preferably hydrogen, —CH3, —C2H5, —C3H7, or —C3H9, or R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


or optionally,


R7, N and X when taken together form an optionally substituted five- or six-membered heterocyclic ring, preferably selected from




embedded image


R3 or R9 may be further selected from




embedded image


wherein R53 is preferably selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above, and x is 0, 1 or 2;


and wherein preferably either all of R54 to R58 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R54 to R58 are different from hydrogen and selected from substitutions presented above and remaining positions of R54 to R58 are selected from hydrogen;


preferably with X=0 or 1, preferably 1, or R8 and R9, when taken together form a optionally substituted five- or six-membered cycloalkyl, optionally substituted five- or six-membered cycloheteroalkyl, optionally substituted five- or six-membered aryl, or optionally substituted five- or six-membered heteroaryl.


In a compound of the invention according to Formula (Ia) or (Ia1) as defined above comprising as a moiety A the following optionally substituted aryl group:




embedded image


as defined above, preferably R8 to R9 are selected as follows


R8 is hydrogen, or pyrrolidinyl,




embedded image


methyl, C(O)-azetidine, —F, —Cl, —CH(CH3)2, —CN, —OCHF2, —NH2,




embedded image


—C(CH3)═NOH, —NO2, —CH(CH3)—NHC(O)CH3, or CH2N(CH3)2,


R9 is hydrogen,-methyl, —OCH3, —OCF3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —S(O)2CH3, —CH2NHS(O)2CH3, —C(O)2CH3, or —NHC(O)—R41, wherein R41 is methyl, —NH—CH(CH3)2, furan, tetrahydrofuran, cyclopropyl, —NH-cyclopropyl, cyclopentyl, —NH-tetrahydropyran, —CHF2, or C(O)2CH3, or




embedded image


preferably with R53 as defined above;


or R8 and R9 together, form a six-membered heterocyclic ring selected from




embedded image


Preferably the proviso applies that R8 is not identical to R9.


Particularly preferable in a compound of the invention according to Formula (Ia) or (Ia1) as defined above comprising as a moiety A the following optionally substituted aryl group:




embedded image


preferably R1 to R5 are selected as follows:


R1 is hydrogen, —C1-C2 alkoxy, —C(O)O—C1-C5 alkyl, —OCH3, —C1-C3 alkyl, methyl, or ethyl


R2 is hydrogen, methyl, —F, —Cl, —Br, —CN, —OCH3, or —C(O)O—C1-C5 alkyl;


R3 is hydrogen, methyl, —CH(CH3)2, —CHF2, —F, —Cl, —Br, —CN, —OCH3, —OCF3, NO2, or —C(O)O—(C1-C5 alkyl);


R4 is hydrogen or —F, —Cl, —Br, or —CF3; and/or


R5 is hydrogen, methyl, ethyl or —F, —Cl, —Br, or —CN, or —CH2-phenyl.


Preferably the proviso applies that R8 is not identical to R9.


Likewise preferably, R8 and R9 are defined as indicated above.


In view of the above, in one embodiment the present invention therefore also relates to a compound of Formula (Ia) or (Ia1), wherein the compound is a compound of Formula (Ia2) or (Ia3), wherein X is not present:




embedded image


Typically, R1, R2, R3, R4, R5, R8 and R9 are preferably as defined above for Formula (Ia) or (Ia1),


In one embodiment, the present invention relates to a compound of Formula (Ia) or (Ia1), wherein the compound is a compound of Formula (Ia2) or (Ia3)


wherein R1, R2, R3, R4, R5, and R8 are each independently hydrogen, —F, —Cl, —Br, —NO2, —O—N=O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkoxy, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 cycloalkyl C1-C10 alkyl, optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, optionally substituted C6-C18 aryl, optionally substituted C6-C18 aryl C1-C10 alkyl, optionally substituted 5-20 membered heteroaryl, optionally substituted 5-20 membered heteroaryl C1-C10 alkyl, (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, or—NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, optionally substituted 2-20 membered heteroalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; and —CCl3; and


wherein R9 is selected from C1-C3 alkyl, five-membered heterocyclic ring, —OCH3, —S(O)2CH3, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 is selected from C2-C5 alkyl, C3-C6 cycloalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; or five-membered heterocyclic ring.


In a preferred embodiment, R9 is selected from C1-C3 alkyl, pyrrolidine, —OCH3, —S(O)2CH3, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 is selected from C2-C5 alkyl, C3-C6 cycloalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; or tetrahydrofuran or furan.


In more preferred embodiment, R9 is selected from C, alkyl, C3 alkyl, pyrrolidine, —OCH3, —S(O)2CH3, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 is selected from C5 cycloalkyl, —CHF2, or tetrahydrofuran or furan.


In one embodiment, the present invention relates to a compound of Formula (Ia) or (Ia1), wherein the compound is a compound of Formula (Ia1)


wherein R1, R2, R3, R4, R5, and R9 are each independently hydrogen, —F, —Cl, —Br, —NO2, —O—N=O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkoxy, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 cycloalkyl C1-C10 alkyl, optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, optionally substituted C6-C18 aryl, optionally substituted C6-C18 aryl C1-C10 alkyl, optionally substituted 5-20 membered heteroaryl, optionally substituted 5-20 membered heteroaryl C1-C10 alkyl, (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, or —NHC(O)NHR1,


wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, optionally substituted 2-20 membered heteroalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; and —CCl3; and


R3 is —S(O)2CH3, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 is selected from C2-C5 alkyl, C3-C5cycloalkyl, —CHF2; or five-membered heterocyclic ring.


In a preferred embodiment R8 is —S(O)2CH3, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 is selected from C3 alkyl, C3 cycloalkyl, —CHF2.


In one embodiment, the present invention relates to a compound of Formula (Ia) or (Ia1), wherein the compound is a compound of Formula (Ia2) or (Ia3),


wherein R1, R2, R3, R4, and R5, are each independently hydrogen, —F, —Cl, —Br, —NO2, —O—N═O, —N=O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkoxy, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 cycloalkyl C1-C10 alkyl, optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, optionally substituted C6-C18 aryl, optionally substituted C6-C18 aryl C1-C10 alkyl, optionally substituted 5-20 membered heteroaryl, optionally substituted 5-20 membered heteroaryl C1-C10 alkyl, (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, or—NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, optionally substituted 2-20 membered heteroalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; and —CCl3; and,


wherein R8 and R9, when taken together form a heterocyclic ring is selected from:




embedded image


Alternatively, in one further embodiment, the present invention relates to a compound of Formula (Ia) or (Ia1), wherein the —X-A group is




embedded image


wherein R10 is hydrogen or C1-C3 alkyl, preferably —CH2—, —C2H4—, —C3H6—, —CH(CH3)2, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, or —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—,


or —(CH2)n-A, wherein n is an integer from 1-3 or in one embodiment, n is 2; in another embodiment, n is 1;


and wherein A is




embedded image


wherein R53 is as defined above.


Accordingly, the invention is also directed to a compound related to Formula (Ia) or (Ia1) that is further defined according to any of Formulae (Ib), (Ib1), (Ib2) or (Ib3):




embedded image


wherein X is selected from the group consisting of a C1-C3 alkyl, —CH(CH3)2—CH2—, —C2H4—, —C3H6—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, or —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—, or is not present,


wherein R53 is hydrogen, an optionally substituted C1-C10 alkyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or optionally substituted 5-20 membered heteroaryl, preferably an optionally substituted C6-C14 aryl or 5-20 membered heteroaryl. Preferably, R53 is as defined above for oxadiazoles, more preferably, R53 is hydrogen, an optionally substituted C1-C10 alkyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or optionally substituted 5-20 membered heteroaryl, preferably an optionally substituted C6-C14 aryl or 5-20 membered heteroaryl, more preferably wherein R53 is an optionally substituted C6-C14 aryl or 5-10 membered heteroaryl, preferably a pyridyl or substituted C aryl, more preferably 3-pyridyl or para substituted phenyl, wherein the substituent is selected from the group consisting of —F, —Cl, and —CF3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; or


R53 is preferably selected from the group consisting of hydrogen, OH, —OCH3, —OCF3, —OCHF2, —OCH2OH; —OCH2CH3; —F; —Cl; —Br; —CN; NO2; NH2; optionally substituted C1-C5 alkyl, optionally substituted C1-C5 alkoxy, optionally substituted C2-C5 alkenyl, preferably —CH3, CH(CH3)2; —CH2CH3; —CH2—CH2—CH(CH3)2; —CH2—N(CH3)2; —CH(CH3)—NHC(O)CH3; —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —O—N═O, —N═O, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO,




embedded image


with X=0 or 1, preferably 1;




embedded image


or


R53 is




embedded image


wherein R54, R55, R56, R57 or R58 are selected each independently from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —C, —Br, NH2, NO2, and x is 0, 1 or 2;


or R53 is




embedded image


wherein R59 is




embedded image


and wherein R54, R55, R56, R57 or R58 are selected separately from the above from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, and x is 0, 1 or 2; as defined above, and x is 0, 1 or 2;


and wherein preferably either all of R54 to R58 are hydrogen or 1, 2 or 3, preferably 1 or 2 of R54 to R58 are different from hydrogen and selected from substitutions presented above and remaining positions of R54 to R58 are selected from hydrogen.


R1, R2, R3, R4, and R5 in Formula (Ib), (Ib1), (Ib2) or (Ib3) are preferably each independently hydrogen, —F, —Cl, —Br, —NO2, —O—N═O, —N═O, —OH, —NH2, —CH═NH, —N3, —SH, —SO3H, —SO2H, —S(O)2CH3—CN, —NC, —OCN, —NCO, —SCN, —NCS, —(P=O)(OH)2, —O(P=O)(OH)2, —(C═O)OH, —(C═O)NHOH, —NH(C═O)NH2, —NH(C═S)NH2, —(C═O)NH2, —CHO, —CHS, —OCHO, —NCHO, —B(OH)2, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 alkoxy, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 cycloalkyl C1-C10 alkyl, optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, optionally substituted C6-C18 aryl, optionally substituted C6-C18 aryl C1-C10 alkyl, optionally substituted 5-20 membered heteroaryl, optionally substituted 5-20 membered heteroaryl C1-C10 alkyl, (C═O)R11, (C═O)OR11, (C═O)SR11, (C═O)NR11R12, (SO2)R11, (SNO)R11, (P=O)R11R12, (P=O)OR11OR12 or C(NR11)R12, —NHC(O)R11, or —NHC(O)NHR11, wherein R11 and R12 are independently selected from the group consisting of optionally substituted C1-C10 alkyl, optionally substituted C2-C10 alkenyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C6-C18 aryl, optionally substituted 5-20 membered heteroaryl, optionally substituted 2-20 membered heteroalkyl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; and —CCl3; or


R6 and R7 are each independently hydrogen, an optionally substituted C1-C10 alkyl or R6 and R7 when taken together form an optionally substituted five- or six-membered heterocyclic ring.


According to a particularly preferred embodiment in any of Formulae (Ia), (Ia1), (Ia2), (Ia3), (Ib), (Ib1), (Ib2) or (Ib3) or (Id) defined herein, preferably,


R1 is hydrogen, —C1-C2 alkoxy, —C(O)O—C1-C5 alkyl, —OCH3, —C1-C3 alkyl, methyl, or ethyl


R2 is hydrogen, methyl, —F, —Cl, —Br, —CN, —OCH3, or —C(O)O—C1-C5 alkyl;


R3 if present, is hydrogen, methyl, —CH(CH3)2, —CHF2, —F, —Cl, —Br, —CN, —OCH3, —OCF3, NO2, or —C(O)O—C1-C5 alkyl;


R4 is hydrogen or —F, —Cl, —Br, or —CF3; and/or


R5 is hydrogen, methyl, ethyl or —F, —Cl, —Br, —CN, or —CH2-phenyl.


Preferably, R2 and R3 together, form an optionally substituted five- or six-membered cyclic or heterocyclic ring, preferably selected from




embedded image


In one particular embodiment of the invention, in Formula (Ia) or (Ia1), A is an optionally substituted oxadiazole. In one embodiment, A is




embedded image


wherein R53 is an optionally substituted C6-C14 aryl or 5-10 membered heteroaryl, preferably R53 is pyridyl or substituted C aryl, more preferably 3-pyridyl or para substituted phenyl, wherein the substituent is selected from the group consisting of —F, —Cl, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; and —CCl3;


In one embodiment, the present invention relates to a compound of any one of Formulae (Ia), (Ia1), (Ia2), (Ia3), (Ib), (Ib1), (Ib2) or (Ib3), or (Id), wherein R2 and R5 are preferably each hydrogen,


R1 is hydrogen or methoxy,


R3, if present, is hydrogen or methyl and


R4 is hydrogen or —F, wherein at least one of R1 to R5 is not hydrogen, and preferably R53 is as defined above.


In one embodiment, the present invention relates to a compound of Formula (Ia) or (Ia1), wherein the compound is a compound of Formula (Id):




embedded image


wherein preferably


B is O or S, preferably O,


X is preferably selected from C1-C5-alkyl, —CH2—, —C2H4—, —C3H6—, —CH(CH3)2—, —CHCH(CH3)2—, —CHC(CH3)3—, —CH(CH(CH3)2)—, —CH(CH2CH(CH3)2)—, or —CH(CH2OH)—; or is not present;


R1, R2, R4 and R5 are preferably selected separately from hydrogen, —CH3, —C2H5, —CH2CH3, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, preferably wherein two or three of R1, R2, R4 and R5 are hydrogen and the remaining are not hydrogen; and


R54 and R55 are each preferably selected separately from hydrogen, —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —Cl, —Br, NO2, NH2, preferably wherein one of R54 and R55 is hydrogen and the other is one of —CH3, —C2H5, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; —OCH3, —F, —C, —Br, NO2, NH2.


In one further embodiment, the present invention relates to a compound of the following structure:




embedded image


In one embodiment, the present invention relates to a compound wherein the compound is selected from the compounds delineated in Table 1.












TABLE 1





No.
Code
Structure
IUPAC-Name







 1
EN22


embedded image


1-phenyl-3-(quinolin-6- yl)urea





 2
EN21


embedded image


1-(2-methoxyphenyl)-3- ((3-(4-(trifluoromethyl)- phenyl)-1,2,4-oxadiazol- 5-yl)methyl)urea





 3
EN-A (EN23)


embedded image


1,3-di(quinolin-6-yl)urea





 4
AH4


embedded image


3-phenyl-1-(quinolin-6- yl)thiourea





 5
JB010


embedded image


1-(4-methoxyphenyl)-3- (quinolin-6-yl)urea





 6
BRM/BR G1 ATP Inhibitor-1


embedded image


1-[3-(difluoromethyl)- 1,2-thiazol-5-yl]-3-[2- fluoro-5-(hydroxymeth- yl)pyridin-4-yl]urea





 7
JB011


embedded image


1-(4-cyanophenyl)-3- (quinolin-6-yl)urea





 8
PM07


embedded image


3-{4-[5-({1-[(3-fluoro- 2-methylphenyl)meth- yl]piperidin-3-yl}meth- yl)-1,2,4-oxadiazol-3- yl]phenyl}-1-(4-meth- oxyphenyl)urea





 9
PM16


embedded image


3-{4-[5-({1-[(3-fluoro- 2-methylphenyl)meth- yl]piperidin-3-yl}meth- yl)-1,2,4-oxadiazol-3- yl]phenyl}-1-phenylurea





10
JB014


embedded image


1-(quinolin-6-yl)-3-[4- ({5-[4-(trifluoromethyl)- phenyl]-1,2,4-oxadiazol- 3-yl}methyl)phenyl]urea





11
AH1


embedded image


3-(2-chloro-4-nitrophen- yl)-1-(3,5-dichlorophen- yl)urea





12
AH3


embedded image


1-(3-chlorophenyl)-3-(3- fluoro-2-methylphenyl)- urea





13
JB002


embedded image


1-(1,5-naphthyridin-2- yl)-3-phenylurea





14
JB004


embedded image


1-(3-bromoquinolin-6- yl)-3-phenylurea





15
EN11-16


embedded image


1-benzyl-3-(quinolin-6- yl)urea





16
EN11-13


embedded image


1-(3-phenylpropyl)-3- (quinolin-6-yl)urea





17
L01.110


embedded image


3-(2-methyl-3-oxo-3,4- dihydro-2H-1,4-benzox- azin-6-yl)-1-phenylurea





18
L01.041


embedded image


3-(4-methyl-3-[(propan- 2-yl)carbamoylamino}- phenyl)-1-phenylurea





19
L01.004


embedded image


3-phenyl-1-[4-(pyrrol- idin-1-yl)phenyl]urea





20
L01.007


embedded image


1-(5-fluoro-2-methoxy- phenyl)-3-{2-methyl-1- [3-(pyridin-3-yl)-1,2,4- oxadiazol-5-yl]propyl}- urea





21
L01.104


embedded image


N-{4-[(phenylcarbamo- yl)amino]phenyl}cyclo- pentanecarboxamide





22
L01.116


embedded image


2,2-difluoro-N-{2-meth- yl-5-[(phenylcarbamo- yl)amino]phenyl}acet- amide





23
L01.113


embedded image


1-(3-chloro-2,6-diethyl- phenyl)-3-{2-methyl-1- [3-(pyridin-3-yl)-1,2,4- oxadiazol-5-yl]propyl}- urea





24
L01.101


embedded image


N-{4-[(phenylcarbamo- yl)amino]phenyl}oxo- lane-3-carboxamide





25
L01.092


embedded image


N-{4-[(phenylcarbamo- yl)amino]phenyl}furan- 3-carboxamide





26
L01.124


embedded image


3-(3-fluoro-4-methyl- phenyl)-1-phenylurea





27
TCMDC- 143175


embedded image


3-(2-chloro-4-nitrophen- yl)-1-(3-chlorophenyl)- urea





28
A.C2


embedded image


3-(2-chloro-4-nitrophen- yl)-1-(2-chlorophenyl)- urea





29
AH2


embedded image


3-(2-chloro-4-nitrophen- yl)-1-(3-chlorophenyl)- thiourea





30
EN11-07


embedded image


1-(2-phenylethyl)-3- (quinolin-6-yl)urea





31
EN11-02


embedded image


1-cyclohexyl-3-(quin- olin-6-yl)urea





32
EN11-11


embedded image


3-benzyl-1-[(quinolin-6- yl)methyl]urea





33
L01.016


embedded image


2,2-difluoro-N-{4- [(phenylcarbamoyl)- amino]phenyl}acet- amide





34
L01.028


embedded image


3-cyclopropyl-1-{2- methyl-5-[(phenyl- carbamoyl)amino]- phenyl}urea





35
L01.047


embedded image


1-phenyl-3-[4-(propan-2- yl)phenyl]urea





36
L01.026


embedded image


3-(3-chloro-4-methoxy- phenyl)-1-phenylurea





37
L01.127


embedded image


3-{[3-(4-methylphenyl)- 1,2,4-oxadiazol-5-yl]- methyl}-1-phenylimid- azolidine-2,4-dione





38
L01.061


embedded image


1-phenyl-3-[4-(trifluoro- methoxy)phenyl]urea





39
L01.128


embedded image


3-(2-methyl-1,3-dioxo- 2,3-dihydro-1H-iso- indol-5-yl)-1-phenyl- urea





40
L01.025


embedded image


3-[3-methyl-4-(pyrrol- idin-1-yl)phenyl]-1- phenylurea





41
L01.008


embedded image


1-[4-(azetidine-1-carbon- yl)phenyl]-3-phenylurea





42
L01.089


embedded image


1-phenyl-3-{[3-(thio- phen-2-yl)-1,2,4- oxadiazol-5-yl]meth- yl}imidazolidine-2,4- dione





43
L01.130


embedded image


3-{[3-(4-fluorophenyl)- 1,2,4-oxadiazol-5-yl]- methyl}-5-methyl-1-(4- methylphenyl)imidazol- idine-2,4-dione





44
L01.049


embedded image


3-{[3-(4-chlorophenyl)- 1,2,4-oxadiazol-5-yl]- methyl}-1-phenylimid- azolidine-2,4-dione





45
L01.066


embedded image


3-(2,3-dihydro-1H- inden-5-yl)-1-phenyl- urea





46
L01.043


embedded image


1-(2-methoxyphenyl)-3- {[3-(pyridin-3-yl)-1,2,4- oxadiazol-5-yl]methyl}- urea





47
L01.087


embedded image


1-phenyl-3-{[3-(thio- phen-2-yl)-1,2,4- oxadiazol-5-yl]meth- yl}urea





48
L01.020


embedded image


3-(3-bromophenyl)- 1-phenylurea





49
L01.035


embedded image


3-(4-chloro-3-methane- sulfonylphenyl)-1- phenylurea





50
L01.120


embedded image


3-[3-(difluoromethoxy)- phenyl]-1-phenylurea





51
L01.033


embedded image


3-{[3-(furan-2-yl)-1,2,4- oxadiazol-5-yl]methyl}- 1-phenylimidazolidine- 2,4-dione





52
L01.002


embedded image


1-[4-(cyanomethoxy)- phenyl]-3-phenylurea





53
L01.027


embedded image


N-{3-[(phenylcarbamo- yl)amino]phenyl}furan- 3-carboxamide





54
L01.003


embedded image


3-methyl-N-{3-[(phen- ylcarbamoyl)amino]- phenyl}but-2-enamide





55
L01.038


embedded image


1-(4-cyanophenyl)- 3-phenylurea





56
L01.069


embedded image


3-{[3-(3-methylphenyl)- 1,2,4-oxadiazol-5-yl]- methyl}-1-phenylimid- azolidine-2,4-dione





57
L01.097


embedded image


1-(4-chlorophenyl)-3- [2-methyl-1-(3-phenyl- 1,2,4-oxadiazol-5-yl)- propyl]urea





58
L01.095


embedded image


3-{4-[1-(hydroxyimino)- ethyl]phenyl}-1-phenyl- urea





59
L01.013


embedded image


1-(3,4-diethoxyphenyl)- 3-{[3-(3-methoxyphen- yl)-1,2,4-oxadiazol-5- yl]methyl}urea





60
L01.103


embedded image


N-{3-[(phenylcarbamo- yl)amino]phenyl}cyclo- pentanecarboxamide





61
L01.071


embedded image


1-(4-methylphenyl)-3- {[3-(thiophen-3-yl)- 1,2,4-oxadiazol-5-yl]- methyl}imidazolidine- 2,4,5-trione





62
L01.115


embedded image


1-(3,5-dichlorophenyl)- 3-{[3-(thiophen-3-yl)- 1,2,4-oxadiazol-5-yl]- methyl}urea





63
L01.045


embedded image


1-[3-(2-oxo-1,3-oxazol- idin-3-yl)phenyl]-3- phenylurea





64
L01.077


embedded image


3-(3-methylphenyl)-1- phenylurea





65
L01.084


embedded image


N-{3-[(phenylcarbamo- yl)amino]phenyl}cyclo- propanecarboxamide





66
L01.109


embedded image


N-{4-[(phenylcarbamo- yl)amino]phenyl}acet- amide





67
L01.122


embedded image


3-cyclopropyl-1-{3- [(phenylcarbamoyl)- amino]phenyl}urea





68
L01.083


embedded image


3-[(3-cyclopropyl- 1,2,4-oxadiazol-5-yl)- methyl]-1-[2-(propan- 2-yloxy)phenyl]urea





69
L01.062


embedded image


1-(isoquinolin-5-yl)-3- {[3-(pyridin-3-yl)- 1,2,4-oxadiazol-5-yl]- methyl}urea





70
L01.081


embedded image


3-[(3-cyclopropyl-1,2,4- oxadiazol-5-yl)methyl]- 1-(3-fluorophenyl)urea





71
L01.114


embedded image


3-(1,3-dihydro-2-benzo- furan-5-yl)-1-{2-meth- yl-1-[3-(pyridin-3-yl)- 1,2,4-oxadiazol-5-yl]- propyl}urea





72
L01.090


embedded image


N-(3-chlorophenyl)-3- [3-(thiophen-2-yl)-1,2,4- oxadiazol-5-yl]morph- oline-4-carboxamide





73
L01.136


embedded image


1-(3-methoxyphenyl)-3- methyl-3-{[3-(pyridin- 3-yl)-1,2,4-oxadiazol- 5-yl]methyl}urea





74
L01.023


embedded image


2-methyl-N-{3-[(phenyl- carbamoyl)amino]phen- yl}propanamide





75
L01.064


embedded image


N-{4-[(phenylcarbamo- yl)amino]phenyl}furan- 2-carboxamide





76
L01.093


embedded image


3-{3-methyl-1-[3-(4H- 1,2,4-triazol-3-yl)-1,2,4- oxadiazol-5-yl]butyl}-1- [3-(4-methylpiperidin-1- yl)phenyl]urea





77
L01.125


embedded image


3-[(3-benzyl-1,2,4- oxadiazol-5-yl)methyl]- 1-(4-methoxyphenyl)- urea





78
L01.005


embedded image


3-{2-methyl-1-[3- (pyridin-3-yl)-1,2,4- oxadiazol-5-yl]propyl}- 1-[3-(trifluoromethyl)- phenyl]urea





79
L01.135


embedded image


butyl 4-[{{3-methyl-1- [3-(4H-1,2,4-triazol-3- yl)-1,2,4-oxadiazol-5-yl]- butyl}carbamoyl)amino]- benzoate





80
L01.108


embedded image


3-(3-nitrophenyl)-1- phenylurea





81
L01.112


embedded image


1-(4-cyanophenyl)-3-[2- hydroxy-1-(3-phenyl- 1,2,4-oxadiazol-5-yl)- ethyl]urea





82
L01.054


embedded image


1-[4-fluoro-2-(methoxy- methyl)phenyl]-3-{2- methyl-1-[3-(pyridin-3- yl)-1,2,4-oxadiazol-5- yl]propyl}urea





83
L01.067


embedded image


3-[(3-benzyl-1,2,4- oxadiazol-5-yl)methyl]- 1-(3-methoxyphenyl)- urea





84
L01.099


embedded image


N-[3-(cyclopentane- sulfonyl)phenyl]-2-(3- phenyl-1,2,4-oxadiazol- 5-yl)piperidine-1- carboxamide





85
L01.065


embedded image


1-{4-[(oxan-4-l)amino]- phenyl}-3-phenylurea





86
L01.132


embedded image


3-(3-{[(methylcarbamo- yl)amino]methyl}phen- yl)-1-phenylurea





87
L01.121


embedded image


3-{4-[(dimethylamino)- methyl]phenyl}-1-phen- ylurea





88
L01.056


embedded image


1-(2-methoxy-6-methyl- phenyl)-3-{2-methyl-1- [3-(pyridin-3-yl)-1,2,4- oxadiazol-5-yl]propyl}- urea





89
L01.088


embedded image


3-[(3-benzyl-1,2,4- oxadiazol-5-yl)methyl]- 1-(4-methylphenyl)urea





90
L01.042


embedded image


1-(2-chloro-4-nitrophen- yl)-3-{[3-(pyridin-3-yl)- 1,2,4-oxadiazol-5-yl]- methyl}urea





91
L01.010


embedded image


3-{3-methyl-1-[3-(4H- 1,2,4-triazol-3-yl)-1,2,4- oxadiazol-5-yl]butyl}-1- [3-(2-oxoimidazolidin- 1-yl)phenyl]urea









Particularly preferably, the present invention relates to a compound of Formula (I) selected from any of compounds as defined above numbered as No. 1-91, 1-75 or 1-26, more preferably from any of compounds numbered as No. 1-75 or 1-26, and most preferably from any of compounds numbered as No. 1-6 or 1-26, e.g. 1-6.


According to one further aspect, the present invention relates to a compound of Formula (II),





R71-D-CO—R72

    • or a pharmaceutically acceceptable salt thereof, wherein:


D is an optionally substituted heteroaromatic bicycle selected from the group comprising 2-benzofuran, 2-benzothiophene, isoindol, indol, indolizine and purine and


R71 is an optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, and


R72 is optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, —OR73, —NR73, and wherein


R73 is hydrogen, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl.


In one embodiment of Formula II, D is isoindol, indol, or preferably indolizine. In one embodiment, the present invention relates to a compound of Formula (II), wherein the compound is a compound of Formula (IIa):




embedded image


In one embodiment, the present invention relates to a compound of Formula (II) or (IIa), wherein R72 is —OR73, —NR73, wherein R72 is preferably —NR73 and wherein


R73 is hydrogen, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl.


In one embodiment, the present invention relates to a compound of Formula (IIa), wherein Formula (IIa) is represented by Formula (IIb)




embedded image


wherein R72 is optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, —OR73, —NR73, and


R73 is hydrogen, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or C1-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl and


wherein R74 is hydrogen, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, and wherein R74 is preferably optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl.


In one embodiment, the present invention relates to a compound of Formula (IIb), wherein R72 is —OR73, —NR73, wherein R72 is preferably —NR73 and wherein


R73 is, optionally substituted C2-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted 2-20 membered heteroalkyl, optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, and


wherein R74 is hydrogen, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted C6-C18 aryl or C6-C18 aryl C1-C10 alkyl; optionally substituted 5-20 membered heteroaryl or 5-20 membered heteroaryl C1-C10 alkyl; optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl; optionally substituted 3-20 membered cycloheteroalkyl or 3-20 membered cycloheteroalkyl C1-C10 alkyl, and wherein R74 is preferably optionally substituted C3-C20 cycloalkyl or C3-C20 cycloalkyl C1-C10 alkyl.


In one embodiment, the present invention relates to a compound of Formula (IIb), wherein the compound is




embedded image


In one embodiment, the present invention relates to the present compounds, wherein the present compound induces the NLRP3 inflammasome activation, thereby increasing the release of IL-1β of 0.1×106 BMDCs upon administering the compound in a concentration of 50 μM of the compound and incubation of 2 hours. The increase of the release of the amount of IL-1β corresponds preferably to at least a twofold increase, at least a threefold increase and an even more preferably at least a tenfold increase of the amounts of IL-1β in comparison to BMDCs that are not administered the active compound. In one embodiment, the present invention relates to the present compounds, wherein the present compound induces the NLRP3 inflammasome activation, thereby preferably increasing the release of IL-1β of 0.1×106 BMDCs upon administering the compound in a concentration of 50 μM of the compound and incubation of 2 hours, wherein the increase is at least 0.1 ng ml−1, 0.5 ng ml-1, preferably 2 ng ml−1, 10 ng ml−1, 25 ng ml−1, 50 ng ml−1 and more preferably 4 ng ml−1 or more. The increase of the release of IL-1β of 0.1×106 BMDCs can be measured with the method described in Example 1.8.


Pharmaceutical Compositions and Administration
General

In some embodiments, a compound of the present invention (e.g., a compound that modulates e.g. agonizes) NLRP3, or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination thereof is administered as a pharmaceutical composition that includes the compound of the invention and one or more pharmaceutically acceptable excipients, and optionally one or more additional therapeutic agents as described herein.


In some embodiments, the present compounds can be administered in combination with one or more conventional pharmaceutical excipients. Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, and wool fat. Cyclodextrins such as α-, β, and γ-cyclodextrin, or chemically modified derivatives such as hydroxyalkyl cyclodextrins, including 2- and 3-hydroxypropyl-P-cyclodextrins, or other solubilized derivatives can also be used to enhance delivery of compounds described herein. Dosage forms or compositions containing a compound of the invention as described herein in the range of 0.005% to 100% with the balance made up from non-toxic excipient may be prepared. The contemplated compositions may contain 0.001%-100% of a compound of the invention provided herein, in one embodiment 0.1-95%, in another embodiment 75-85%, in a further embodiment 20-80%. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 22nd Edition (Pharmaceutical Press, London, U K. 2012).


Routes of Administration and Composition Components

In some embodiments, the present compounds described herein or a pharmaceutical composition thereof can be administered to subject in need thereof by any accepted route of administration. Acceptable routes of administration include, but are not limited to, buccal, cutaneous, endocervical, endosinusial, endotracheal, enteral, epidural, interstitial, intra-abdominal, intra-arterial, intrabronchial, intrabursal, intracerebral, intracisternal, intracoronary, intradermal, intraductal, intraduodenal, intradural, intraepidermal, intraesophageal, intragastric, intragingival, intraileal, intralymphatic, intramedullary, intrameningeal, intramuscular, intraovarian, intraperitoneal, intraprostatic, intrapulmonary, intrasinal, intraspinal, intrasynovial, intratesticular, intrathecal, intratubular, intratumoral, intrauterine, intravascular, intravenous, nasal, nasogastric, oral, parenteral, percutaneous, peridural, rectal, respiratory (inhalation), subcutaneous, sublingual, submucosal, topical, transdermal, transmucosal, transtracheal, ureteral, urethral and vaginal. In certain embodiments, a preferred route of administration is parenteral (e.g., intratumoral). In certain embodiments, a preferred route of administration is systemic.


Compositions can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes. Typically, such compositions can be prepared as injectables, either as liquid solutions, emulsions or suspensions; solid forms suitable for use to prepare solutions, emulsions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified. The preparation of such formulations will be known to those of skill in the art in light of the present disclosure.


The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil, or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.


The carrier also can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques, which yield a powder of the active ingredient, plus any additional desired ingredient from a previously sterile-filtered solution thereof. Intratumoral injections are discussed, e.g., in Lammers, et al., “Effect of Intratumoral Injection on the Biodistribution and the Therapeutic Potential of HPMA Copolymer-Based Drug Delivery Systems” Neoplasia. 2006, 10, 788-795.


Pharmacologically acceptable excipients usable in the rectal composition as a gel, cream, enema, or rectal suppository, include, without limitation, any one or more of cocoa butter glycerides, synthetic polymers such as polyvinylpyrrolidone, PEG (like PEG ointments), glycerine, glycerinated gelatin, hydrogenated vegetable oils, poloxamers, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol Vaseline, anhydrous lanolin, shark liver oil, sodium saccharinate, menthol, sweet almond oil, sorbitol, sodium benzoate, anoxid SBN, vanilla essential oil, aerosol, parabens in phenoxyethanol, sodium methyl p-oxybenzoate, sodium propyl p-oxybenzoate, diethylamine, carbomers, carbopol, methyloxybenzoate, macrogol cetostearyl ether, cocoyl caprylocaprate, isopropyl alcohol, propylene glycol, liquid paraffin, xanthan gum, carboxy-metabisulfite, sodium edetate, sodium benzoate, potassium metabi sulfite, grapefruit seed extract, methyl sulfonyl methane (MSM), lactic acid, glycine, vitamins, such as vitamin A and E and potassium acetate.


In certain embodiments, suppositories can be prepared by mixing the present compound described herein with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum and release the active compound. In other embodiments, compositions for rectal administration are in the form of an enema.


In other embodiments, the compounds described herein or a pharmaceutical composition thereof are suitable for local delivery to the digestive or GI tract by way of oral administration (e.g., solid or liquid dosage forms.).


Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the a compound of the invention is mixed with one or more pharmaceutically acceptable excipients, such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.


In one embodiment, the compositions will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with a compound of the invention provided herein, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like. In another solid dosage form, a powder, marume, solution or suspension (e.g., in propylene carbonate, vegetable oils, PEG's, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule). Unit dosage forms in which one or more of the present compounds provided herein or additional active agents are physically separated are also contemplated; e.g., capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two-compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated.


Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives that are particularly useful for preventing the growth or action of microorganisms. Various preservatives are well known and include, for example, phenol and ascorbic acid. In certain embodiments the excipients are sterile and generally free of undesirable matter. These compositions can be sterilized by conventional, well-known sterilization techniques. For various oral dosage form excipients such as tablets and capsules sterility is not required. The USP/NF standard is usually sufficient.


In certain embodiments, solid oral dosage forms can further include one or more components that chemically and/or structurally predispose the composition for delivery of the compound of the invention to the stomach or the lower GI; e.g., the ascending colon and/or transverse colon and/or distal colon and/or small bowel. Exemplary formulation techniques are described in, e.g., Filipski, K. J., et al., Current Topics in Medicinal Chemistry, 2013, 13, 776-802, which is incorporated herein by reference in its entirety.


Examples include upper-GI targeting techniques, e.g., Accordion Pill (Intec Pharma), floating capsules, and materials capable of adhering to mucosal walls.


Other examples include lower-GI targeting techniques. For targeting various regions in the intestinal tract, several enteric/pH-responsive coatings and excipients are available. These materials are typically polymers that are designed to dissolve or erode at specific pH ranges, selected based upon the GI region of desired drug release. These materials also function to protect acid labile drugs from gastric fluid or limit exposure in cases where the active ingredient may be irritating to the upper GI (e.g., hydroxypropyl methylcellulose phthalate series, Coateric (polyvinyl acetate phthalate), cellulose acetate phthalate, hydroxypropyl methylcellulose acetate succinate, Eudragit series (methacrylic acid-methyl methacrylate copolymers), and Marcoat). Other techniques include dosage forms that respond to local flora in the GI tract, Pressure-controlled colon delivery capsule, and Pulsincap. Ocular compositions can include, without limitation, one or more of any of the following: viscogens (e.g., Carboxymethylcellulose, Glycerin, Polyvinylpyrrolidone, Polyethylene glycol); Stabilizers (e.g., Pluronic (triblock copolymers), Cyclodextrins); Preservatives (e.g., Benzalkonium chloride, EDTA, SofZia (boric acid, propylene glycol, sorbitol, and zinc chloride; Alcon Laboratories, Inc.), Purite (stabilized oxychloro complex; Allergan, Inc.)). Topical compositions can include ointments and creams. Ointments are semisolid preparations that are typically based on petrolatum or other petroleum derivatives. Creams containing the selected active agent are typically viscous liquid or semisolid emulsions, often either oil-in-water or water-in-oil. Cream bases are typically water-washable, and contain an oil phase, an emulsifier and an aqueous phase. The oil phase, also sometimes called the “internal” phase, is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation is generally a nonionic, anionic, cationic or amphoteric surfactant. As with other carriers or vehicles, an ointment base should be inert, stable, nonirritating and non-sensitizing.


In any of the foregoing embodiments, pharmaceutical compositions described herein can include one or more one or more of the following: lipids, interbilayer crosslinked multilamellar vesicles, biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles, and nanoporous particle-supported lipid bilayers.


Dosages

The dosages may be varied depending on the requirement of the patient, the severity of the condition being treating and the particular compound being employed. Determination of the proper dosage for a particular situation can be determined by one skilled in the medical arts. The total daily dosage may be divided and administered in portions throughout the day or by means providing continuous delivery.


In some embodiments, the compounds described herein are administered at a dosage of from about 0.0001 mg kg−1 to about 1000 mg kg−1 (e.g., from about 0.001 mg kg−1 to about 200 mg kg−1; from about 0.01 mg kg−1 to about 200 mg kg−1; from about 0.01 mg kg−1 to about 150 mg kg−1; from about 0.01 mg kg−1 to about 100 mg kg−1; from about 0.01 mg kg−1 to about 50 mg kg−1; from about 0.01 mg kg−1 to about 10 mg kg−1; from about 0.01 mg kg−1 to about 5 mg kg−1; from about 0.01 mg kg−1 to about 1 mg kg−1; from about 0.01 mg kg−1 to about 0.5 mg kg−1; from about 0.01 mg kg−1 to about 0.1 mg kg−1; from about 0.1 mg kg−1 to about 200 mg kg−1; from about 0.1 mg kg−1 to about 150 mg kg−1; from about 0.1 mg kg−1 to about 100 mg kg−1; from about 0.1 mg kg−1 to about 50 mg kg−1; from about 0.1 mg kg−1 to about 10 mg kg−1; from about 0.1 mg kg−1 to about 5 mg kg−1; from about 0.1 mg kg−1 to about 1 mg kg−1; from about 0.1 mg kg−1 to about 0.5 mg kg−1).


Regimens

The foregoing dosages can be administered on a daily basis (e.g., as a single dose or as two or more divided doses) or non-daily basis (e.g., every other day, every two days, every three days, once weekly, twice weeks, once every two weeks, once a month).


In some embodiments, the period of administration of a compound described herein is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 1 1 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. In an embodiment, a therapeutic compound is administered to an individual for a period of time followed by a separate period of time. In another embodiment, a therapeutic compound is administered for a first period and a second period following the first period, with administration stopped during the second period, followed by a third period where administration of the therapeutic compound is started and then a fourth period following the third period where administration is stopped. In an aspect of this embodiment, the period of administration of a therapeutic compound followed by a period where administration is stopped is repeated for a determined or undetermined period of time. In a further embodiment, a period of administration is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.


Methods of Treatment

In some embodiments, methods for treating a subject having condition, disease or disorder in which a decrease or increase in NLRP3 activity (e.g., a decrease, e.g., repressed or impaired NLRP3 signaling) contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) are provided.


Indications

In any of the methods described herein, the subject can have a cancer. In some examples of any of the methods described herein, the mammal has been identified as having a cancer, or has been diagnosed as having a cancer.


Non-limiting examples of cancer include: acute myeloid leukemia, adrenocortical carcinoma, Kaposi sarcoma, lymphoma, anal cancer, appendix cancer, teratoid/rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain cancer, breast cancer, bronchial tumor, carcinoid tumor, cardiac tumor, cervical cancer, chordoma, chronic lymphocytic leukemia, chronic myeloproliferative neoplasm, colon cancer, colorectal cancer, craniopharyngioma, bile duct cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, Ewing sarcoma, eye cancer, fallopian tube cancer, gallbladder cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, germ cell tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, hypopharngeal cancer, pancreatic cancer, kidney cancer, laryngeal cancer, chronic myelogenous leukemia, lip and oral cavity cancer, lung cancer, melanoma, Merkel cell carcinoma, mesothelioma, mouth cancer, oral cancer, osteosarcoma, ovarian cancer, penile cancer, pharyngeal cancer, prostate cancer, rectal cancer, salivary gland cancer, skin cancer, small intestine cancer, soft tissue sarcoma, testicular cancer, throat cancer, thyroid cancer, urethral cancer, uterine cancer, vaginal cancer, and vulvar cancer.


Methods for diagnosing a subject as having a cancer or identifying a mammal as having a cancer are well known in the art. For example, a medical professional (e.g., a physician, a physician's assistant, or a technician) can diagnose cancer in a mammal by observing one or more symptoms of cancer in a mammal. Non-limiting examples of symptoms of cancer include: fatigue, lump or area of thickening felt under the skin, weight change, jaundice, darkening or redness of the skin, sores that won't heal, changes to existing moles, changes in bowel or bladder habits, persistent cough or trouble breathing, difficulty swallowing, hoarseness, persistent indigestion or discomfort after eating, persistent, unexplained muscle or joint pain, persistent, unexplained fevers or night sweats, and unexplained bleeding or bruising. Methods of diagnosing a subject as having a cancer or identifying a subject as having a cancer can further include performing one or more diagnostic tests (e.g., performing one or more diagnostic tests on a biopsy or a blood sample).


In some examples of any of the methods described herein, a subject can be a subject having a cancer, a subject diagnosed as having a cancer, or a subject identified as having a cancer that has been unresponsive to a previously administered treatment for cancer. Diagnostic tests for diagnosing a subject as having a cancer or identifying a mammal as having a cancer are known in the art.


In any of the methods described herein, the subject can have an infectious disease. In some examples of any of the methods described herein, the subject has been identified as having an infectious disease, or has been diagnosed as having an infectious disease. For example, an infectious disease can be caused by a bacterium, virus, fungus, or a parasite.


Non-limiting examples of infectious disease include: Acinetoobacter infection, actinomycosis, African sleeping sickness, acquired immunodeficiency syndrome, amebiasis, anaplasmosis, anthrax, Arcanobacterium haemolyticum infection, Argentine hemorrhagic fever, ascariasis, aspergillosis, astrovirus infection, babesiosis, Bacillus cereus infection, bacterial pneumonia, bacterial vaginosis, Bacteroides infection, balantidiasis, Baylisascaris infection, BK virus infection, black piedra, Blastocystic hominis infection, blastomycosis, Bolivian hemorrhagic fever, botulism, Brazilian hemorrhagic fever, brucellosis, bubonic plaque, Burkholderi infection, Buruli ulcer, Calicivirus infection, camptobacteriosis, candidiasis, cat-scratch disease, cellulitis, Chagas disease, chancroid, chickenpox, chikungunya, chlamydia, Chlamydophila pneumoniae infection, cholera, chromoblastomycosis, clonorchiasis, Clostridium difficile infection, coccidioidomycosis, Colorado tick fever, common cold, Creutzfeldt-Jakob disease, Crimean-Congo hemorrhagic fever, crytococcosis, cryptosporidiosis, cutaneous larva migrans, cyclosporiasis, cysticercosis, cytomegalovirus infection, dengue fever, Desmodesmus infection, deintamoebiasis, diphtheria, diphyllobothriasis, dracunculiasis, ebola hemorrhagic fever, echinococcosis, ehrlichiosis, enterobiasis, Enterococcus infection, Enterovirus infection, epidemic typhus, erythema infection, exanthema subitum, fasciolopsiasis, fasciolosis, fatal familial insomnia, filariasis, food poisoning by Clostridium myonecrosis, free-living amebic infection, Fusobacterium infection, gas gangrene, geotrichosis, Gerstmann-Straussler-Scheinker syndrome, giardiasis, glanders, gnathostomiasis, gonorrhea, granuloma inguinale, Group A streptococcal infection, Group B streptococcal infection, Haemophilus influenzae infection, hand foot and mouth disease, hantavirus pulmonary syndrome, Heartland virus disease, Heliobacter pylori infection, hemolytic-uremic syndrome, hemorrhagic fever with renal syndrome, hepatitis A, hepatitis B, hepatitis C, hepatitis D, hepatitis E, herpes simplex, histoplasmosis, hookworm infection, human bocavirus infection, human ewingii ehrlichiosis, human granulocyte anaplasmosis, human metapneuomovirus infection, human monocytic ehrlichiosis, human papillomavirus infection, human parainfluenza virus infection, hymenolepiasis, Epstein-Barr virus infectious mononucleosis, influenza, isosporiasis, Kawasaki disease, keratitis, Kingella kingae infection, kuru, lassa fever, Legionnaires' disease, Pontiac fever, leishmaniasis, leprosy, leptospirosis, listeriosis, lyme disease, lymphatic filariasis, lymphocytic choriomeningitis, malaria, Marburg hemorrhagic fever, measles, Middle East respiratory syndrome, melioidosis, meningitis, meningococcal disease, metagonimiasis, microsporidiosis, molluscum contagiosum, monkeypox, mumps, murine typhus, mycoplasma pneumonia, mycetoma, myiasis, neonatal conjunctivitis, variant Creutzfeldt-Jakob disease, nocardiosis, onchocerciasis, paracoccidioidomycosis, paragonimiasis, pasteurellosis, pediculosis capitis, pediculosis corporis, pediculosis pubis, pelvic inflammatory disease, pertussis, plague, pneumonia, poliomyelitis, Prevotella infection, primary amoebic meningoencephalitis, progressive multifocal leukoencephalopathy, psittacosis, Q fever, rabies, relapsing fever, respiratory syncytial virus infection, rhinosporidiosis, rhinovirus infection, rickettsial infection, rickettsialpox, Rift Valley Fever, Rocky Mountain spotted fever, rotavirus infection, rubella, salmonellosis, severe acute respiratory syndrome, scabies, schistosomiasis, sepsis, shigellosis, shingles, smallpox, sporothrichosis, staphylococcal food poisoning, staphylococcal infection, staphylococcal infection, strongyloidiasis, subacute sclerosing panencephalitis, syphilis, taeniasis, tetanus, tinea barabe, tinea capitis, tinea corporis, tinea cruris, tinea manum, tinea nigra, tinea pedis, tinea unguium, tinea versicolor, toxocariasis, trachoma, toxoplasmosis, trichinosis, trichomoniasis, trichuriasis, tuberculosis, tularemia, typhoid fever, Ureaplasma urealyticum infection, valley fever, Venezuelan hemorrhagic fever, viral pneumonia, West Nile fever, white piedra, Yersinia psuedotuberculosis infection, yersiniosis, yellow fever, and zygomycosis.


Methods for diagnosing a subject as having an infectious disease, or identifying a subject as having an infectious disease are well known in the art. For example, a medical professional (e.g., a physician, a physician's assistant, or a technician) can diagnose infectious disease in a subject by observing one or more symptoms of infectious disease in a subject. Non-limiting examples of symptoms of infectious disease include: fever, diarrhea, fatigue, and muscle aches. Methods of diagnosing a mammal as having an infectious disease or identifying a subject as having an infectious disease can further include performing one or more diagnostic tests (e.g., performing one or more diagnostic tests on a biopsy or a blood sample). Diagnostic tests for diagnosing a subject as having an infectious disease or identifying a subject as having an infectious disease are known in the art. With regard to the combination therapy, this disclosure contemplates both monotherapy regimens as well as combination therapy regimens.


In some embodiments, the methods described herein can further include administering one or more additional therapies (e.g., one or more additional therapeutic agents and/or one or more therapeutic regimens) in combination with administration of the compounds described herein.


In certain embodiments, the methods described herein can further include administering one or more additional cancer therapies.


The one or more additional cancer therapies can include, without limitation, surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy, cancer vaccines (e.g., HPV vaccine, hepatitis B vaccine, Oncophage, Provenge) and gene therapy, as well as combinations thereof. Immunotherapy, including, without limitation, adoptive cell therapy, the derivation of stem cells and/or dendritic cells, blood transfusions, lavages, and/or other treatments, including, without limitation, freezing a tumor.


In some embodiments, the one or more additional cancer therapies is chemotherapy, which can include administering one or more additional chemotherapeutic agents. In certain embodiments, the additional chemotherapeutic agent is an immunomodulatory moiety, e.g., an immune checkpoint inhibitor. In certain of these embodiments, the immune checkpoint inhibitor targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1-PD-L1, PD-1-PD-L2, T cell immunoglobulin and mucin 3 (TEVI3 or HAVCR2), Galectin 9-TEVI3, Phosphatidylserine-TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II, -LAG3, 4-1BB-4-1BB ligand, OX40-OX40 ligand, GITR, GITR ligand-GITR, CD27, CD70-CD27, T FRSF25, TNFRSF25-TL1 A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM-BTLA, HVEM-CD 160, HVEM-LIGHT, HVEM-BTLA-CD160, CD80, CD80-PD-L1, PD-L2-CD80, CD244, CD48-CD244, CD244, ICOS, ICOS-ICOS ligand, B7-H3, B7-H4, VISTA, TMIGD2, HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, K Rs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86-CD28, CD86-CTLA, CD80-CD28, Phosphatidylserine, TIM3, Phosphatidylserine-TIM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PD-1 or PD-L1) and other immunomodulatory agents, such as interleukin-2 (IL-2), indoleamine 2,3-dioxygenase (IDO), IL-10, transforming growth factor-β (TGFp), CD39, CD73, Adenosine-CD39-CD73, and CXCR4-CXCL 12. See, e.g., Postow, M. J. Clin. Oncol. 2015, 33, 1. In certain of these embodiments, the immune checkpoint inhibitor is selected from the group consisting of: Urelumab, PF-05082566, MED16469, TRX518, Varlilumab, CP-870893, Pembrolizumab (PD-1), Nivolumab (PD-1), Atezolizumab (formerly MPDL3280A) (PD-L1), MED14736 (PD-L1), Avelumab (PD-L1), PDR001 (PD-1), BMS-986016, MGA271, Lirilumab, IPH2201, Emactuzumab, INCB024360, Galunisertib, Ulocuplumab, BKT140, Bavituximab, CC-90002, Bevacizumab, and MNRP1685A, and MGA271.


In certain embodiments, the additional chemotherapeutic agent is a STING agonist. For example, the STING agonist can include cyclic di-nucleotides, such as cAMP, cGMP, and cGAMP as well as modified cyclic di-nucleotides that include one or more of the following modification features (2′-0/3′-0 linkage, phosphorothioate linkage, adenine and/or guanine analogue, 2′-OH modification (e.g., —OCH3 or replacement, e.g., —F or N3). See, e.g., WO 2014/189805. In certain embodiments, the additional chemotherapeutic agent is an alkylating agent. Alkylating agents are so named because of their ability to alkylate many nucleophilic functional groups under conditions present in cells, including, but not limited to cancer cells. In a further embodiment, an alkylating agent includes, but is not limited to, Cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin. In an embodiment, alkylating agents can function by impairing cell function by forming covalent bonds with the amino, carboxyl, sulfhydryl, and phosphate groups in biologically important molecules or they can work by modifying a cell's DNA. In a further embodiment an alkylating agent is a synthetic, semisynthetic or derivative.


In certain embodiments, the additional chemotherapeutic agent is an anti-metabolite. Antimetabolites masquerade as purines or pyrimidines, the building-blocks of DNA and in general, prevent these substances from becoming incorporated in to DNA during the “S” phase (of the cell cycle), stopping normal development and division. Antimetabolites can also affect RNA synthesis. In an embodiment, an antimetabolite includes, but is not limited to azathioprine and/or mercaptopurine. In a further embodiment an anti-metabolite is a synthetic, semisynthetic or derivative.


In certain embodiments, the additional chemotherapeutic agent is a plant alkaloid and/or terpenoid. These alkaloids are derived from plants and block cell division by, in general, preventing microtubule function. In an embodiment, a plant alkaloid and/or terpenoid is a vinca alkaloid, a podophyllotoxin and/or a taxane. Vinca alkaloids, in general, bind to specific sites on tubulin, inhibiting the assembly of tubulin into microtubules, generally during the M phase of the cell cycle. In an embodiment, a vinca alkaloid is derived, without limitation, from the Madagascar periwinkle, Catharanthus roseus (formerly known as Vinca rosea). In an embodiment, a vinca alkaloid includes, without limitation, Vincristine, Vinblastine, Vinorelbine and/or Vindesine. In an embodiment, a taxane includes, but is not limited, to Taxol, Paclitaxel and/or Docetaxel. In a further embodiment a plant alkaloid or terpernoid is a synthetic, semisynthetic or derivative. In a further embodiment, a podophyllotoxin is, without limitation, an etoposide and/or teniposide. In an embodiment, a taxane is, without limitation, docetaxel and/or ortataxel. In an embodiment, a cancer therapeutic is a topoisomerase. Topoisomerases are essential enzymes that maintain the topology of DNA. Inhibition of type I or type II topoisom erases interferes with both transcription and replication of DNA by upsetting proper DNA supercoiling. In a further embodiment, a topoisomerase is, without limitation, a type I topoisomerase inhibitor or a type II topoisomerase inhibitor. In an embodiment a type I topoisomerase inhibitor is, without limitation, a camptothecin. In another embodiment, a camptothecin is, without limitation, exatecan, irinotecan, lurtotecan, topotecan, BNP 1350, CKD 602, DB 67 (AR67) and/or ST 1481. In an embodiment, a type II topoisomerase inhibitor is, without limitation, epipodophyllotoxin. In a further embodiment an epipodophyllotoxin is, without limitation, an amsacrine, etoposid, etoposide phosphate and/or teniposide. In a further embodiment a topoisomerase is a synthetic, semisynthetic or derivative, including those found in nature such as, without limitation, epipodophyllotoxins, substances naturally occurring in the root of American Mayapple (Podophyllum peltatum).


In certain embodiments, the additional chemotherapeutic agent is a stilbenoid. In a further embodiment, a stilbenoid includes, but is not limited to, Resveratrol, Piceatannol, Pinosylvin, Pterostilbene, Alpha-Viniferin, Ampelopsin A, Ampelopsin E, Diptoindonesin C, Diptoindonesin F, Epsilon-Vinferin, Flexuosol A, Gnetin H, Hemsleyanol D, Hopeaphenol, Trans-Diptoindonesin B, Astringin, Piceid and Diptoindonesin A. In a further embodiment a stilbenoid is a synthetic, semisynthetic or derivative.


In certain embodiments, the additional chemotherapeutic agent is a cytotoxic antibiotic. In an embodiment, a cytotoxic antibiotic is, without limitation, an actinomycin, an anthracenedione, an anthracycline, thalidomide, dichloroacetic acid, nicotinic acid, 2-deoxyglucose and/or chlofazimine. In an embodiment, an actinomycin is, without limitation, actinomycin D, bacitracin, colistin (polymyxin E) and/or polymyxin B. In another embodiment, an antracenedione is, without limitation, mitoxantrone and/or pixantrone. In a further embodiment, an anthracycline is, without limitation, bleomycin, doxorubicin (Adriamycin), daunorubicin (daunomycin), epirubicin, idarubicin, mitomycin, plicamycin and/or valrubicin. In a further embodiment a cytotoxic antibiotic is a synthetic, semisynthetic or derivative.


In certain embodiments, the additional chemotherapeutic agent is selected from endostatin, angiogenin, angiostatin, chemokines, angioarrestin, angiostatin (plasminogen fragment), basement-membrane collagen-derived anti-angiogenic factors (tumstatin, canstatin, or arrestin), anti-angiogenic antithrombin Ill, signal transduction inhibitors, cartilage-derived inhibitor (CDI), CD59 complement fragment, fibronectin fragment, gro-beta, heparinases, heparin hexasaccharide fragment, human chorionic gonadotropin (hCG), interferon alpha/beta/gamma, interferon inducible protein (IP-10), interleukin-12, kringle 5 (plasminogen fragment), metalloproteinase inhibitors (TIMPs), 2-methoxyestradiol, placental ribonuclease inhibitor, plasminogen activator inhibitor, platelet factor-4 (PF4), prolactin 16 kD fragment, proliferin-related protein (PRP), various retinoids, tetrahydrocortisol-S, thrombospondin-1 (TSP-1), transforming growth factor-beta (TGF-β), vasculostatin, vasostatin (calreticulin fragment) and the like.


In certain embodiments, the additional chemotherapeutic agent is selected from abiraterone acetate, altretamine, anhydrovinblastine, auristatin, bexarotene, bicalutamide, BMS 184476, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, bleomycin, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-proly-l-Lproline-t-butylamide, cachectin, cemadotin, chlorambucil, cyclophosphamide, 3′,4′-didehydro-4′-deoxy-8′-norvin-caleukoblastine, docetaxol, doxetaxel, cyclophosphamide, carboplatin, carmustine, cisplatin, cryptophycin, cyclophosphamide, cytarabine, dacarbazine (DTIC), dactinomycin, daunorubicin, decitabine dolastatin, doxorubicin (adriamycin), etoposide, 5-fluorouracil, finasteride, flutamide, hydroxyurea and hydroxyureataxanes, ifosfamide, liarozole, lonidamine, lomustine (CCNU), MDV3100, mechlorethamine (nitrogen mustard), melphalan, mivobulin isethionate, rhizoxin, sertenef, streptozocin, mitomycin, methotrexate, taxanes, nilutamide, onapristone, paclitaxel, prednimustine, procarbazine, RPR109881, stramustine phosphate, tamoxifen, tasonermin, taxol, tretinoin, vinblastine, vincristine, vindesine sulfate, and vinflunine. In certain embodiments, the additional chemotherapeutic agent is platinum, cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, azathioprine, mercaptopurine, vincristine, vinblastine, vinorelbine, vindesine, etoposide and teniposide, paclitaxel, docetaxel, irinotecan, topotecan, amsacrine, etoposide, etoposide phosphate, teniposide, 5-fluorouracil, leucovorin, methotrexate, gemcitabine, taxane, leucovorin, mitomycin C, tegafur-uracil, idarubicin, fludarabine, mitoxantrone, ifosfamide and doxorubicin. Additional agents include inhibitors of mTOR (mammalian target of rapamycin), including but not limited to rapamycin, everolimus, temsirolimus and deforolimus.


In still other embodiments, the additional chemotherapeutic agent can be selected from those delineated in U.S. Pat. No. 7,927,613, which is incorporated herein by reference in its entirety. In yet another embodiment, the methods can further include administering one or both of: (i) one or more anti-fungal agents (e.g., selected from the group of bifonazole, butoconazole, clotrimazole, econazole, ketoconazole, luliconazole, miconazole, omoconazole, oxiconazole, sertaconazole, sulconazole, tioconazole, albaconazole, efinaconazole, epoziconazole, fluconazole, isavuconazole, itraconazole, posaconazole, propiconazole, ravusconazole, terconazole, voriconazole, abafungin, amorolfin, butenafine, naftifine, terbinafine, anidulafungin, caspofungin, micafungin, benzoic acid, ciclopirox, flucytosine, 5-fluorocytosine, griseofulvin, haloprogin, tolnaflate, undecylenic acid, and balsam of peru) and (ii) one or more antibiotics (e.g., selected from the group of amikacin, gentamicin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem, cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalothin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, linezolid, posizolid, radezolid, torezolid, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin G, penicillin V, piperacillin, penicillin G, temocillin, ticarcillin, amoxicillin, calvulanate, ampicillin, subbactam, piperacillin, tazobactam, ticarcillin, clavulanate, bacitracin, colistin, polymyxin B, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethoxazole, sulfanamide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole, sulfonamideochrysoidine, demeclocycline, minocycline, oytetracycline, tetracycline, clofazimine, dapsone, dapreomycin, cycloserine, ethambutol, ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin, dalopristin, thiamphenicol, tigecycyline, tinidazole, trimethoprim, and teixobactin).


In certain embodiments, the second therapeutic agent or regimen is administered to the subject prior to contacting with or administering the compound of the invention (e.g., about one hour prior, or about 6 hours prior, or about 12 hours prior, or about 24 hours prior, or about 48 hours prior, or about 1 week prior, or about 1 month prior).


In other embodiments, the second therapeutic agent or regimen is administered to the subject at about the same time as contacting with or administering the compound of the invention. By way of example, the second therapeutic agent or regimen and the compound of the invention are provided to the subject simultaneously in the same dosage form. As another example, the second therapeutic agent or regimen and the compound of the invention are provided to the subject concurrently in separate dosage forms.


In still other embodiments, the second therapeutic agent or regimen is administered to the subject after contacting with or administering the compound of the invention (e.g., about one hour after, or about 6 hours after, or about 12 hours after, or about 24 hours after, or about 48 hours after, or about 1 week after, or about 1 month after).


In some embodiments, the methods described herein further include the step of identifying a subject (e.g., a patient) in need of such treatment (e.g., by way of biopsy, endoscopy, or other conventional method known in the art). In certain embodiments, the NLRP3 protein can serve as a biomarker for certain types of cancer.


In some embodiments, the present compounds, methods, and compositions described herein can be administered to certain treatment-resistant patient populations (e.g., patients resistant to checkpoint inhibitors).


Uses of the Present Compounds

The compounds of the present invention, or a salt or solvate thereof, can be used as modulators, e.g., agonists, of the NLRP3 receptor in medicaments as well as in the development of biological or diagnostic assays.


In addition, new small molecule NLRP3 activators are useful to study the biology of inflammasome and thus make an important contribution to the fundamental understanding of the activation mechanisms of the NLRP3 inflammasome and enable the targeted development of specific drugs.


The present compounds have been shown to induce typical characteristics of inflammasome activation, including the activation of caspase-1, the release of the biologically active forms of proinflammatory cytokines such as IL-1β (FIGS. 5A and 5B). Moreover, inflammasome-dependent lytic cell death (pyroptosis) and ASC speck formation are induced by the present compounds (FIGS. 6, 7, 13-16 and 18). For the induction of the lytic cell death, a concentration dependence can be observed (FIGS. 4A-C, 13-16 and 18). FIG. 4D shows the EC50 for induction of IL-1β in BMDCs for compounds EN21 and EN22 in comparison to the imidazoquinoline NLRP3 activators imiquimod and CL097, wherein EC50 for EN21 and EN22 is surprisingly 10 times lower than for imiquimod and CL097.


Without wishing to be bound to a theory, it is assumed that the compounds EN21, EN22, BRM/BRG1 ATP Inhibitor-1, EN23, AH4, JB010 and EN54 activate the NLRP3 inflammasome, since the observed characteristics of inflammasome activation do not occur in cells isolated from NLRP3 knockout mice (FIGS. 7, 13-16 and 18) and, in addition, the NLRP3-specific inhibitor MCC950 completely suppresses inflammasome activation (FIG. 8).


A further advantage of the present compounds is that the present compounds seem to activate NLRP3 rapidly. Especially with EN22, the inflammasome activation in BMDCs from wild-type mice without inhibitor is almost maximum after only 30 minutes. Thus, a rapid effectiveness of a medicament comprising the compounds of the invention is assumed.


It has further been shown that contrary to other small molecule NLRP3 inflammasome activators like the known activator imiquimod, the present compounds are more specific, since no simultaneous activation of Toll-like receptors (TLR7/TLR8) occurs and thus the present compounds do not induce the release of further proinflammatory cytokines such as TNF or IL-6 (FIGS. 9 and 19).


In contrast to other known low molecular weight NLRP3 activators, the present compounds do not inhibit mitochondrial respiration and are therefore less susceptible to unspecific undesired effects (FIG. 10).


Moreover, many known NLRP3 activators lead to massive potassium efflux from the cell via different mechanisms, which precedes inflammasome activation. Without wishing to be bound to a theory, it is assumed that the present compounds such as EN21 and EN22 activate the NLRP3 inflammasome independently of potassium efflux from the cell (FIGS. 11 and 17). It is assumed that reactive oxygen species (ROS) are involved in the mechanism of action of the present compounds, since the ROS scavenger ebselen can inhibit the activation of the inflammasome (FIG. 11).


It has further been shown that in contrary to particulate NLRP3 activators such as uric acid crystals (mono sodium urate, MSU) or aluminum hydroxide, the present compounds are not endocyted by immune cells, followed by endolysosomal rupture to induce inflammasome activation. The endocytosis inhibitor cytochalasin D can prevent particle-induced NLRP3 activation, but cannot prevent NLRP3 activation by EN21 and EN22 (FIG. 12).


Thus, the present compounds are important to the pharmaceutical industry because they can increase or rapidly induce in a selective manner the NLRP3 inflammasome.


According to the one aspect, the present invention relates to the present compounds for use as a medicament. In one embodiment, the present compounds for use as a medicament are used in a therapeutically effective amount.


The medicament can be in any physical form, such as a solid, semi-solid, plaster, solution, suspension, lotion, cream, foam, gel, paste, emulsion, or a combination thereof. Examples of the medicament include, but are not limited to, a pharmaceutical composition, an ingestible composition, a personal care composition, and a combination thereof.


According to the one aspect, the present invention relates to the present compounds for use in the prevention or treatment of a disease by activating the NLRP3 inflammasome, wherein the disease is selected from a group comprising infectious diseases or cancer.


In one embodiment, the present compounds may be used in the prevention or treatment of a bacterial infection.


In one embodiment, the present compounds may be used in the prevention or treatment of a viral infection. The viral infection may be selected from the group comprising genital warts, cutaneous warts and COVID-19.


In one embodiment, the present compounds may be used in the prevention of an infectious disease by boosting vaccination-mediated immunity against pathogens.


In one embodiment, the present compounds may be used in the prevention or treatment of a cancer. In one embodiment, the present compounds may be used in breaking the immune evasion in cancer diseases. The term “Immune evasion of cancer” or “cancer immune evasion” is the ability of cancer to persist in a host having a functioning immune system. There are a number of factors that contribute to tumor persistence despite having a normal host immune system. Immune editing is one of the key aspects why tumors evade surveillance causing the tumors to lie dormant in patients for years through “equilibrium” and “senescence” before re-emerging.


In one embodiment, the compounds of the present invention may be used as an adjuvant.


In one embodiment, the compounds of the present invention may be used to boost the immune response. The boost of the immune response by a compound of the present invention may be suitable during the prevention or treatment of cancer, an infectious disease or during a vaccination process. Preferably, the compounds of the present invention may be used as an adjuvant in the context of vaccination to boost specific immune responses by activating the NLRP3 inflammasome.


In one embodiment, the present compounds may be used in the prevention or treatment of cancer, wherein the cancer is selected from the group comprising solid tumors, skin cancer, wherein skin cancer is selected from the group comprising superficial basal cell carcinoma, genital warts and actinic keratosis, acute myeloid leukemia, adrenocortical carcinoma, Kaposi sarcoma, lymphoma, anal cancer, appendix cancer, teratoid/rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain cancer, breast cancer, bronchial tumor, carcinoid tumor, cardiac tumor, cervical cancer, chordoma, chronic lymphocytic leukemia, chronic myeloproliferative neoplasm, colon cancer, colorectal cancer, craniopharyngioma, bile duct cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, Ewing sarcoma, eye cancer, fallopian tube cancer, gallbladder cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, germ cell tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, hypopharngeal cancer, pancreatic cancer, kidney cancer, laryngeal cancer, chronic myelogenous leukemia, lip and oral cavity cancer, lung cancer, melanoma, Merkel cell carcinoma, mesothelioma, mouth cancer, oral cancer, osteosarcoma, ovarian cancer, penile cancer, pharyngeal cancer, prostate cancer, rectal cancer, salivary gland cancer, skin cancer, small intestine cancer, soft tissue sarcoma, testicular cancer, throat cancer, thyroid cancer, urethral cancer, uterine cancer, vaginal cancer, and vulvar cancer.


In one embodiment, the present compounds may be used in the prevention or treatment of disease, wherein the compound activates the NLRP3 inflammasome and is preferably administered to a subject having a disease in which activation of the NLRP3 inflammasome contributes to the treatment of the pathology and/or symptoms of the disease and/or stops or decreases progression of the disease.


In one embodiment of the invention, a subject to be treated is a human or an animal, preferably a human.


In one embodiment of the invention, the present compounds are used in the activation of the NLRP3 inflammasome and TLR 7 and/or TLR 8 is not activated.


In one embodiment of the invention, the present compounds are administered in an amount of 0.0005 mg kg−1 to 1000 mg kg−1, 0.005 mg kg−1 to 500 mg kg−1, 0.05 mg kg−1 to 100 mg kg−1, 0.5 mg kg−1 to 100 mg kg−1, 5 mg kg−1 to 75 mg kg−1, preferably about 50 mg kg−1 in a mouse or a human.


In one embodiment of the invention, the present compounds are administered topically, orally or systemically. In one embodiment of the invention, the present compounds are administered topically. In one embodiment of the invention, the present compounds are administered systemically.


According to the one aspect, the present invention relates to a pharmaceutical composition comprising a compound of the present invention and one or more pharmaceutically acceptable excipients.


In one embodiment of the invention, the pharmaceutical composition can be in any physical form, such as a solid, semi-solid, plaster, solution, suspension, lotion, cream, foam, gel, paste, emulsion, or a combination thereof. In one embodiment of the invention, the pharmaceutical composition is in form of a tablet, spray, ointment or gel, preferably an ointment.


In one embodiment of the present invention, the pharmaceutical composition comprises the present compound in a concentration ranging from about 0.0001 ppm to 100,000 ppm. In another embodiment of the composition, the present compound is in a concentration ranging from about 0.001 ppm to 10,000 ppm. In another embodiment of the composition, the present compound is in a concentration ranging from about 0.01 ppm to 1,000 ppm. In another embodiment of the composition, the present compound is in a concentration ranging from about 0.1 ppm to 500 ppm. In another embodiment of the composition, the present compound is in a concentration ranging from about 1 ppm to 500 ppm. In another embodiment of the composition, the present compound is in a concentration ranging from about 10 ppm to 500 ppm. In another embodiment of the composition, the present compound is in a concentration ranging from about 1 ppm to 400 ppm.


According to the one aspect, the present invention relates to the use of the compounds of the present invention for boosting immune responses or breaking immune evasion in cancer diseases in a subject.


According to the one aspect, the present invention relates to a method for modulating NLRP3 activity, the method comprising contacting NLRP3 with a compound of the present invention.


In one embodiment of the present invention, the method for modulating NLRP3 comprises increasing the activity of NLRP3.


In one embodiment of the present invention, the method for modulating NLRP3 comprises that the increase of activity of the NLRP3 inflammasome can be measured by the increase of the released amount of IL-1β of 0.25×106 BMDCs, upon administering the compound in a concentration of 50 μM of the compound and incubation of after 22 h, wherein the increase is at least 0.1 ng ml−1, 0.5 ng ml−1, 0.75 ng ml−1, 1 ng ml−1 2 ng ml−1, 10 ng ml−1, 25 ng ml−1, 50 ng ml−1 and more preferably 4 ng ml−1 or more, and still more preferably 6 ng ml−1.


In one embodiment of the present invention, the method for modulating NLRP3 is carried out in vitro or in vivo.


According to the one aspect, the present invention relates to a method of treating cancer, comprising administering to a subject in need of such treatment an effective amount of a compound of the present invention, or a pharmaceutical composition of the present invention.


In one embodiment of the present invention, the method of treating cancer comprises that the compound is administered in combination with one or more additional cancer therapies.


In one embodiment of the present invention, the method of treating cancer comprises the one or more additional cancer therapies, which comprises surgery, radiotherapy, chemotherapy, toxin therapy, immunotherapy, cryotherapy or gene therapy, or a combination thereof.


In one embodiment of the present invention, the method of treating cancer comprises chemotherapy, which comprises administering one or more additional chemotherapeutic agents.


In one embodiment of the present invention, the method of treating cancer comprises chemotherapy, which comprises administering one or more additional chemotherapeutic agents, signal transduction inhibitors (kinase inhibitors) and immunomodulators (checkpoint inhibitors and STING agonists).


In one embodiment of the present invention, the method of treating cancer comprises chemotherapy, which comprises administering one or more additional chemotherapeutic agents, wherein the one or more additional chemotherapeutic agents is selected from an alkylating agent (e.g., cisplatin, carboplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide and/or oxaliplatin); an anti-metabolite (e.g., azathioprine and/or mercaptopurine); a terpenoid (e.g., a vinca alkaloid and/or a taxane; e.g., Vincristine, Vinblastine, Vinorelbine and/or Vindesine Taxol, Pacllitaxel and/or Docetaxel); a topoisomerase (e.g., a type I topoisomerase and/or a type 2 topoisomerase; e.g., camptothecins, such as irinotecan and/or topotecan; amsacrine, etoposide, etoposide phosphate and/or teniposide); a cytotoxic antibiotic (e.g., actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and/or mitomycin); a hormone (e.g., a lutenizing hormone releasing hormone agonist; e.g., leuprolidine, goserelin, triptorelin, histrelin, bicalutamide, flutamide and/or nilutamide); an antibody (e.g., Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Murom onab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and/or Trastuzumab); an anti-angiogenic agent; a cytokine; a thrombotic agent; a growth inhibitory agent; an antihelminthic agent; and an immune checkpoint inhibitor that targets an immune checkpoint receptor selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-1-PD-L1, PD-1-PD-L2, T cell immunoglobulin and mucin 3 (TFM3 or HAVCR2), Galectin 9 TIM3, Phosphatidylserine-TIM3, lymphocyte activation gene 3 protein (LAG3), MHC class II, -LAG3, 4-1BB-4-1BlE igand, OX40-OX40 ligand, GITR, GITR ligand-GITR, CD27, CD70-CD27, TNFRSF25, TNFRSF25-TL1 A, CD40L, CD40-CD40 ligand, HVEM-LIGHT-LTA, HVEM, HVEM-BTLA, HVEM-CD 160, HVEM-LIGHT, HVEM-BTLA-CD160, CD80, CD80 PD-L1, PD-L2-CD80, CD244, CD48-CD244, CD244, ICOS, ICOS-ICOS ligand, B7-H3, B7-H4, VISTA, TMIGD2, HHLA2-TMIGD2, Butyrophilins, including BTNL2, Siglec family, TIGIT and PVR family members, K Rs, ILTs and LIRs, NKG2D and NKG2A, MICA and MICB, CD244, CD28, CD86-CD28, CD86-CTLA, CD80-CD28, Phosphatidylserine, TFM3, Phosphatidylserine-TFM3, SIRPA-CD47, VEGF, Neuropilin, CD160, CD30, and CD155 (e.g., CTLA-4 or PD-1 or PD-L1) and other immunomodulatory agents, such as interleukin-2 (IL-2), indoleamine, 2,3-dioxygenase (IDO), IL-10, transforming growth factor-p (TGFp), CD39, CD73 Adenosine-CD39-CD73, and CXCR4-CXCL12.


Additional chemotherapeutic agents might be immune checkpoint inhibitors.


Preferred additional chemotherapeutic agents and/or immune checkpoint inhibitors are selected from the group consisting of Urelumab, PF-05082566, MED16469, TRX518, Varlilumab, CP-870893, Pembrolizumab (PD-1), Nivolumab (PD1), Atezolizumab (PD-L1), PDR001 (PD-1), BMS-986016, MGA271, Lirlumab, IPH2201, Emactuzumab, INCB024360, Galunisertib, Ulocuplumab, BKT140, Bavituximab, CC-90002, Bevacizumab, 1VINRP1685A, and MGA271.


According to the one aspect, the present invention relates to a method for analyzing the activity of NLRP3 activation.


The NLRP3 inflammasome activation may be tested in different cell types like for examples primary murine bone marrow-derived dendritic cells (BMDCs) and macrophages (BMDMs), THP-1 cells, human peripheral blood mononuclear cells (PBMCs). Cells are typically primed with 50 ng ml−1 LPS for 3 hours before inflammasome activation. Markers of inflammasome activation are investigated by measuring release of the cytosolic enzyme lactate dehydrogenase (LDH) from the cells as a readout for lytic cell death and pyroptosis, detecting release of proinflammatory cytokines such as IL-1β into the cell supernatants by enzyme-linked immunosorbent assay (ELISA), looking at caspase 1 activation and cleavage of Gasdermin D and pro IL-1β via Immunoblotting and following ASC speck formation via immunofluorescence imaging.


According to one embodiment, the present invention relates to the method for analyzing the NLRP3 activation, which comprises the steps of: (i) priming cells with LPS, (ii) contacting the cells with present compounds (iii) detecting the release of IL-1β into the cell supernatant by enzyme-linked immunosorbent assay (ELISA) and analyzing the cells by immunoblotting and immunofluorescence imaging.


According to the one aspect, the present invention relates to the use of the present compounds for analyzing the activity of NLRP3 activation in vitro.


EXAMPLES
1. Biological Assays

The present compounds are useful as modulators of NLRP3. In a high-throughput screen using a screening method developed by us, new, specific NLRP3 activators out of about 50.000 small molecule compounds have been identified, which differ in their structure and several properties of NLRP3 activation from all previously known and protected NLRP3 activators and are novel and unique in them.


1.1 In Vitro Preliminary Data and Properties of the Activators

All NLRP3 activators presented herein are sufficiently soluble and cell-permeable inflammasome activators: The activators showed sufficient solubility in solvents such as DMSO and have the ability to pass the cell membrane, as only then the detection by the high-throughput screening is possible. Moreover, the present compounds showed good activity in vitro. This was tested in cell culture in several different in vitro assays classically used to investigate inflammasome activation. These include measuring inflammasome-dependent lytic cell death (pyroptosis) by detecting release of the cytosolic enzyme lactate dehydrogenase (LDH) using a commercial colorimetric assay and release of proinflammatory cytokines like IL-1β by enzyme-linked immunosorbent assay (ELISA), analyzing cleavage and activation of caspase-1 and IL-1β by immunoblotting and immunofluorescence imaging of ASC speck formation. Lytic cell death was determined via LDH release. Assay results for compounds are illustrated in the Tables below, wherein the concentration of the compounds tested had been 50 μM or as indicated in the table, and the concentration of the known activators has been tested with 5 μM of nigericin, 100 μM of each R837 and CL097. For classification of the activity of a given molecule, release of IL-1β was quantified as % of IL-1% of a respective control activator used for the experiment (highlighted in tables with bold letters). The compounds listes in Table 1 were classified into 4 groups depending on their ability to induce the NLRP3 inflammasome activation, assessed by % IL-1β release as compared to the positive control.


The activity thresholds were assigned as follows:


(++++)>75% IL-1β (% positive control)


(+++) 10-75% IL-1β (% positive control)


(++) 1-10% IL-1β (% positive control)


(+) 0.8-1% IL-1β (% positive control)


1. In Vitro Data and Properties of the Activators

1.1. EN28, EN22, L87.xxx



















Average LDH
Average





release
(% positive


No.
Compound
LDH release (% of max.)
(% of max.)
control)

























DMSO
8.053
7.414
8.022
8.032
10.176
6.889
8.098
11.069



Nigericin 5 μM
61.933
78.845
69.982
78.413
75.764
74.003
73.157
100.000


2
EN21
36.356
47.704
43.019
50.628
60.994
63.141
50.307
68.766


1
EN22
59.410
74.624
67.018
74.225
71.324
71.181
69.630
95.180



R837 100 μM
12.734
15.684
15.495
20.399
34.109
57.377
25.966
35.494



CL097 100 μM
56.511
69.145
62.611
76.051
68.018
76.242
68.096
93.083


17
L01.110
34.845
34.294
31.850



33.663
46.015


18
L01.041
24.371
24.119
23.567



24.019
32.832


19
L01.004
15.163
16.970
18.507



16.880
23.074


20
L01.007
17.823
23.721
19.371



20.305
27.756


21
L01.104
17.190
17.671
19.727



18.196
24.873


22
L01.116
17.956
18.328
17.892



18.058
24.685


23
L01.113
37.377
36.390
34.335



36.034
49.256


24
L01.101
19.384
19.564
20.661



19.870
27.161


25
L01.092
16.396
14.952
13.605



14.985
20.483


26
L01.124
14.119
13.508
14.589



14.072
19.235


33
L01.016
11.582
12.903
13.299



12.595
17.216


34
L01.028
12.612
15.129
15.770



14.503
19.825


35
L01.047
12.065
12.727
11.451



12.081
16.514


36
L01.026
9.228
9.969
13.232



10.810
14.776


37
L01.127
10.995
10.459
9.932



10.462
14.301


38
L01.061
15.898
13.336
13.232



14.155
19.349


39
L01.128
12.143
11.691
11.469



11.768
16.086


40
L01.025
15.039
12.654
15.391



14.362
19.631


41
L01.008
10.960
9.763
7.910



9.544
13.046


42
L01.089
8.669
6.519
7.642



7.610
10.402


43
L01.130
9.387
8.722
7.450



8.520
11.646


44
L01.049
12.648
10.774
11.897



11.773
16.093


45
L01.066
15.611
15.078
12.310



14.333
19.592


46
L01.043
8.072
7.620
8.535



8.076
11.039


47
L01.087
6.788
7.261
6.936



6.995
9.562


48
L01.020
11.587
11.294
9.407



10.763
14.712


49
L01.035
8.461
8.303
7.473



8.079
11.044


50
L01.120
24.981
25.196
23.234



24.470
33.449


51
L01.033
7.999
9.134
7.247



8.127
11.108


52
L01.002
8.068
10.017
11.898



9.994
13.662


53
L01.027
5.203
7.373
8.025



6.867
9.387


54
L01.003
6.283
8.927
8.221



7.810
10.676


55
L01.038
12.838
11.939
11.078



11.952
16.337


56
L01.069
9.197
8.968
9.736



9.301
12.713


57
L01.097
39.470
35.627
37.427



37.508
51.271


58
L01.095
5.207
7.088
5.172



5.822
7.959


59
L01.013
8.752
7.323
6.537



7.537
10.303


60
L01.103
6.576
7.017
10.089



7.894
10.791


61
L01.071
9.014
9.438
7.518



8.657
11.833


62
L01.115
23.646
22.026
20.929



22.200
30.346


63
L01.045
5.976
5.813
3.780



5.189
7.094


64
L01.077
5.283
6.127
6.102



5.838
7.980


65
L01.084
6.005
6.209
5.461



5.892
8.054


66
L01.109
8.457
7.795
7.697



7.983
10.912


67
L01.122
9.165
8.128
6.967



8.087
11.054


68
L01.083
6.798
6.504
5.680



6.327
8.649


69
L01.062
13.450
13.307
13.003



13.253
18.117


70
L01.081
6.870
5.589
6.539



6.332
8.656


71
L01.114
9.462
9.932
8.701



9.365
12.801


72
L01.090
15.329
15.756
14.149



15.078
20.610


73
L01.136
6.954
7.428
6.391



6.924
9.465


74
L01.023
5.103
5.764
5.837



5.568
7.611


75
L01.064
8.962
8.659
8.624



8.748
11.958


76
L01.093
6.676
7.891
9.879



8.149
11.139


77
L01.125
7.721
6.471
6.431



6.874
9.397


78
L01.005
9.311
16.790
18.315



14.805
20.238


79
L01.135
8.895
7.690
7.743



8.109
11.085


80
L01.108
10.902
10.269
10.350



10.507
14.362


81
L01.112
7.964
6.971
7.035



7.323
10.010


82
L01.054
7.787
8.452
8.848



8.362
11.431


83
L01.067
6.481
6.962
7.048



6.830
9.336


84
L01.099
10.559
9.299
9.572



9.810
13.410


85
L01.065
8.406
9.014
9.788



9.069
12.397


86
L01.132
6.462
6.201
6.631



6.431
8.791


87
L01.121
7.801
9.369
8.962



8.711
11.907


88
L01.056
9.335
10.160
9.599



9.698
13.256


89
L01.088
5.685
4.709
5.350



5.248
7.174


90
L01.042
7.095
8.482
6.354



7.310
9.993


91
L01.010
5.639
6.006
5.521



5.722
7.822


























Average
Average





IL-1β
(% positive


No.
Compound
IL-1β (ng/ml)
(ng/ml)
control)

























DMSO
0.042
0.054
0.055
0.067
0.057
0.056
0.055
0.769



Nigericin 5 μM
6.834
6.400
7.872
6.518
7.890
7.496
7.168
100.000


2
EN21
8.582
7.428
8.023
6.613
8.352
7.696
7.782
108.566


1
EN22
8.679
9.014
9.589
8.363
10.000
9.819
9.244
128.957



R837 100 μM
5.472
4.151
1.552
1.212
0.958
0.911
2.376
33.144



CL097 100 μM
7.355
8.020
8.660
8.522
9.540
9.324
8.570
119.560


17
L01.110
2.505
2.943
2.423



2.624
36.599


18
L01.041
1.508
1.847
1.769



1.708
23.826


19
L01.004
1.192
1.485
1.567



1.415
19.737


20
L01.007
0.859
1.572
1.101



1.177
16.422


21
L01.104
1.103
1.142
1.246



1.163
16.230


22
L01.116
1.107
1.192
1.163



1.154
16.099


23
L01.113
1.145
1.165
1.064



1.125
15.691


24
L01.101
1.160
1.071
1.041



1.091
15.216


25
L01.092
1.253
1.060
0.927



1.080
15.066


26
L01.124
0.928
0.902
1.024



0.951
13.268


33
L01.016
0.755
0.723
0.857



0.778
10.857


34
L01.028
0.686
0.757
0.705



0.716
9.988


35
L01.047
0.505
0.513
0.512



0.510
7.117


36
L01.026
0.452
0.473
0.562



0.495
6.911


37
L01.127
0.490
0.475
0.441



0.469
6.538


38
L01.061
0.508
0.439
0.420



0.456
6.360


39
L01.128
0.456
0.394
0.402



0.417
5.819


40
L01.025
0.333
0.280
0.490



0.368
5.130


41
L01.008
0.361
0.324
0.313



0.332
4.638


42
L01.089
0.313
0.316
0.344



0.324
4.526


43
L01.130
0.404
0.317
0.215



0.312
4.350


44
L01.049
0.330
0.251
0.319



0.300
4.189


45
L01.066
0.346
0.256
0.193



0.265
3.698


46
L01.043
0.282
0.272
0.242



0.265
3.697


47
L01.087
0.237
0.239
0.296



0.257
3.589


48
L01.020
0.236
0.231
0.233



0.233
3.256


49
L01.035
0.321
0.189
0.181



0.230
3.211


50
L01.120
0.207
0.200
0.196



0.201
2.800


51
L01.033
0.197
0.197
0.186



0.193
2.698


52
L01.002
0.144
0.249
0.164



0.186
2.590


53
L01.027
0.149
0.197
0.181



0.176
2.451


54
L01.003
0.167
0.153
0.175



0.165
2.301


55
L01.038
0.166
0.127
0.128



0.140
1.956


56
L01.069
0.131
0.138
0.149



0.139
1.941


57
L01.097
0.121
0.143
0.116



0.126
1.765


58
L01.095
0.137
0.128
0.102



0.122
1.703


59
L01.013
0.118
0.126
0.117



0.120
1.677


60
L01.103
0.060
0.049
0.240



0.116
1.624


61
L01.071
0.116
0.097
0.131



0.115
1.598


62
L01.115
0.122
0.108
0.111



0.114
1.584


63
L01.045
0.103
0.116
0.118



0.112
1.568


64
L01.077
0.067
0.165
0.062



0.098
1.368


65
L01.084
0.083
0.111
0.099



0.098
1.362


66
L01.109
0.120
0.083
0.070



0.091
1.269


67
L01.122
0.072
0.093
0.095



0.087
1.214


68
L01.083
0.080
0.077
0.097



0.085
1.180


69
L01.062
0.084
0.087
0.074



0.082
1.141


70
L01.081
0.075
0.072
0.088



0.078
1.093


71
L01.114
0.073
0.081
0.072



0.075
1.049


72
L01.090
0.079
0.077
0.069



0.075
1.044


73
L01.136
0.064
0.073
0.083



0.073
1.025


74
L01.023
0.065
0.075
0.080



0.073
1.021


75
L01.064
0.065
0.054
0.100



0.073
1.014


76
L01.093
0.056
0.050
0.108



0.072
0.998


77
L01.125
0.080
0.071
0.061



0.071
0.988


78
L01.005
0.053
0.074
0.084



0.070
0.980


79
L01.135
0.079
0.073
0.057



0.070
0.975


80
L01.108
0.076
0.061
0.072



0.070
0.973


81
L01.112
0.075
0.069
0.061



0.068
0.955


82
L01.054
0.067
0.062
0.074



0.068
0.947


83
L01.067
0.064
0.069
0.070



0.068
0.942


84
L01.099
0.074
0.067
0.059



0.067
0.932


85
L01.065
0.052
0.070
0.077



0.067
0.928


86
L01.132
0.062
0.066
0.069



0.066
0.920


87
L01.121
0.066
0.068
0.060



0.065
0.906


88
L01.056
0.060
0.073
0.062



0.065
0.905


89
L01.088
0.069
0.058
0.059



0.062
0.866


90
L01.042
0.060
0.069
0.048



0.059
0.826


91
L01.010
0.065
0.059
0.053



0.059
0.822









1.2. EN23



















Average LDH
Average


No.
Compound
LDH release (% of max.)
release (% of max.)
(% positive control)






















Medium
1.952
1.418
1.059
1.476
4.927


1
EN22 25 μM
29.575
29.703
30.593
29.957
100.000


3
EN-A 25 μM
33.731
33.093
32.555
33.126
110.581


3
EN-A 50 μM
32.141
32.628
32.392
32.387
108.112


3
EN-A 100 μM
34.821
33.136
31.042
33.000
110.157


























Average IL-1β
Average


No.
Compound
IL-1β (ng/ml)
(ng/ml)
(% positive control)






















Medium
0.026
0.023
0.023
0.024
0.472


1
EN22 25 μM
4.821
5.196
5.062
5.026
100.000


3
EN-A 25 μM
5.256
4.576
4.256
4.696
93.430


3
EN-A 50 μM
4.819
5.538
4.722
5.026
100.007


3
EN-A 100 μM
5.256
5.328
4.669
5.084
101.159









1.3. BRM/BRG1 ATP-Inhibitor-1
















Average
Average



LDH
(%













LDH release
release
positive


No.
Compound
(% of max.)
(% of max.)
control)

















Medium
5.371
6.529
3.826
5.242
9.764



Nigericin
50.151
43.167
67.752
53.690
100.000



5 μM


6
BRM/BRG1
14.128
16.194
19.482
16.601
30.921



ATP Inhibi-



tor-1 5 μM


6
BRM/BRG1
38.464
36.463
31.314
35.414
65.959



ATP Inhibi-



tor-1 50 μM


6
BRM/BRG1
26.966
18.468
28.907
24.780
46.154



ATP Inhibi-



tor-1 100 μM


























Average
Average





IL-1β
(% positive


No.
Compound
IL-1β (ng/ml)
(ng/ml)
control)






















Medium
0
0.027929
0
0.009
0.29548556



Nigericin
3.007
3.127
3.318
3.151
100.000



5 μM


6
BRM/BRG1
3.556
3.899
3.537
3.664
116.295



ATP Inhibi-



tor-1 5 μM


6
BRM/BRG1
4.561
6.642
6.737
5.980
189.7925



ATP Inhibi-



tor-1 50 μM


6
BRM/BRG1
7.150
3.831
4.087
5.023
159.415



ATP Inhibi-



tor-1 100 μM









1.4. PM07, PM16, TCMDC-143175, A.C2



















Average LDH
Average





release
(% positive


No.
Compound
LDH release (% of max.)
(% of max.)
control)






















Medium
7.050
7.244
6.625
6.973
9.774


1
EN22 18.75 μM
71.367
72.290
70.369
71.342
100.000


8
PM07 18.75 μM
50.410
51.715
50.881
51.002
71.489


8
PM07 37.5 μM
67.709
69.716
70.399
69.274
97.102


8
PM07 75 μM
85.105
85.806
86.028
85.647
120.051


9
PM16 18.75 μM
72.042
74.209
72.166
72.806
102.052


9
PM16 37.5 μM
88.994
89.909
91.152
90.019
126.179


9
PM16 75 μM
100.397
95.086
96.871
97.451
136.598


27
TCMDC-143175
35.255
34.424
36.807
35.495
49.754



18.75 μM


27
TCMDC-143175
72.817
79.023
72.133
74.658
104.647



37.5 μM


27
TCMDC-143175
91.472
93.190
89.533
91.399
128.113



75 μM


28
A.C2 18.75 μM
18.410
18.465
16.442
17.772
24.912


28
A.C2 37.5 μM
46.587
45.313
45.110
45.670
64.016


28
A.C2 75 μM
77.148
73.140
77.102
75.797
106.244


























Average
Average





IL-1β
(% positive


No.
Compound
IL-1β (ng/ml)
(ng/ml)
control)






















Medium
0.040
0.036
0.054
0.043
0.684


1
EN22 18.75 μM
7.071
5.864
6.030
6.322
100.000


8
PM07 18.75 μM
1.117
1.248
1.641
1.335
21.124


8
PM07 37.5 μM
1.916
1.571
1.295
1.594
25.211


8
PM07 75 μM
1.202
1.130
1.320
1.217
19.256


9
PM16 18.75 μM
0.764
0.830
0.736
0.777
12.288


9
PM16 37.5 μM
0.895
0.795
0.848
0.846
13.380


9
PM16 75 μM
0.876
0.848
0.948
0.890
14.083


27
TCMDC-143175
0.102
0.104
0.110
0.105
1.666



18.75 μM


27
TCMDC-143175
0.309
0.275
0.290
0.291
4.609



37.5 μM


27
TCMDC-143175
0.423
0.381
0.446
0.417
6.592



75 μM


28
A.C2 18.75 μM
0.128
0.116
0.111
0.118
1.870


28
A.C2 37.5 μM
0.377
0.353
0.353
0.361
5.712


28
A.C2 75 μM
0.459
0.546
0.571
0.525
8.308









1.5. AH4, AH1, AH3, AH2



















Average
Average





LDH
(%





release
positive


No.
Compound
LDH release (% of max.)
(% of max.)
control)






















Medium
8.589
9.236
8.871
8.898
11.586


1
EN22 10 μM
78.095
74.908
77.404
76.802
100.000


4
AH4 25 μM
19.952
17.994
18.179
18.709
24.359


4
AH4 50 μM
57.619
55.868
55.036
56.175
73.142


4
AH4 75 μM
67.857
68.689
68.988
68.511
89.204


11
AH1 25 μM
42.075
40.911
40.281
41.089
53.499


11
AH1 50 μM
55.721
59.555
56.124
57.133
74.390


11
AH1 75 μM
60.680
59.098
60.723
60.167
78.340


12
AH3 25 μM
12.835
13.259
12.460
12.851
16.733


12
AH3 50 μM
22.186
22.899
22.877
22.654
29.496


12
AH3 75 μM
34.626
33.827
33.778
34.077
44.370


29
AH2 25 μM
23.861
22.415
22.110
22.795
29.680


29
AH2 50 μM
22.752
21.681
21.969
22.134
28.819


29
AH2 75 μM
25.084
25.530
26.645
25.753
33.532


























Average
Average





IL-1β
(% positive


No.
Compound
IL-1β (ng/ml)
(ng/ml)
control)






















Medium
0.059
0.053
0.061
0.058
0.578


1
EN22 10 μM
10.000
10.000
10.000
10.000
100.000


4
AH4 25 μM
2.340
2.250
1.863
2.151
21.507


4
AH4 50 μM
10.000
10.000
10.000
10.000
100.000


4
AH4 75 μM
9.988
10.000
10.000
9.996
99.960


11
AH1 25 μM
1.140
1.156
1.234
1.177
11.766


11
AH1 50 μM
1.533
1.609
1.592
1.578
15.776


11
AH1 75 μM
1.619
1.382
1.598
1.533
15.328


12
AH3 25 μM
0.540
0.630
0.593
0.588
5.875


12
AH3 50 μM
1.267
1.615
1.675
1.519
15.193


12
AH3 75 μM
2.288
2.351
2.032
2.224
22.240


29
AH2 25 μM
0.197
0.186
0.200
0.194
1.942


29
AH2 25 μM
0.280
0.232
0.243
0.251
2.514


29
AH2 75 μM
0.176
0.193
0.193
0.187
1.872









1.6. JB010, JB011, JB014, JB002, JB004



















Average LDH
Average





release
(% positive


No.
Compound
LDH release (% of max.)
(% of max.)
control)






















Medium
10.425
9.244
8.425
9.365
17.580


1
EN22 20 μM
49.359
55.225
55.225
53.269
100.000


5
JB010 20 μM
24.368
23.784
23.266
23.806
44.689


5
JB010 40 μM
31.421
32.791
35.906
33.373
62.649


5
JB010 80 μM
38.624
41.749
43.361
41.245
77.427


7
JB011 20 μM
11.537
12.278
11.156
11.657
21.883


7
JB011 40 μM
12.611
13.286
13.733
13.210
24.799


7
JB011 80 μM
16.367
16.371
17.276
16.671
31.296


10
JB014 20 μM
9.465
7.643
8.614
8.574
16.095


10
JB014 40 μM
9.739
9.811
9.418
9.656
18.126


10
JB014 80 μM
25.331
33.166
30.920
29.806
55.953


13
JB002 20 μM
7.716
6.991
7.485
7.397
13.887


13
JB002 40 μM
9.376
9.412
9.594
9.461
17.760


13
JB002 80 μM
13.963
16.997
17.117
16.026
30.084


14
JB004 20 μM
30.957
16.589
15.720
21.089
39.589


14
JB004 40 μM
25.621
30.949
26.130
27.567
51.749


14
JB004 80 μM
34.302
36.552
38.206
36.353
68.244


























Average
Average





IL-1β
(% positive


No.
Compound
IL-1β (ng/ml)
(ng/ml)
control)






















Medium
0.000
0.0157
0.0316
0.024
0.806


1
EN22
1.632
2.095
2.146
1.957
100.000


5
JB010 20 μM
2.200
3.078
1.934
2.404
122.814


5
JB010 40 μM
2.727
2.742
2.498
2.655
135.661


5
JB010 80 μM
2.757
3.613
5.527
3.966
202.590


7
JB011 20 μM
0.562
0.612
0.553
0.576
29.413


7
JB011 40 μM
0.538
0.564
0.711
0.604
30.877


7
JB011 80 μM
0.912
0.737
0.607
0.752
38.413


10
JB014 20 μM
0.028
0.035
0.029
0.031
1.565


10
JB014 40 μM
0.240
0.245
0.1944
0.227
11.579


10
JB014 80 μM
1.080
1.344
1.097
1.174
59.959


13
JB002 20 μM
0.038
0.050
0.047
0.045
2.292


13
JB002 40 μM
0.190
0.197
0.19017
0.192
9.827


13
JB002 80 μM
0.464
0.505
0.540
0.503
25.697


14
JB004 20 μM
0.966
0.765
1.182
0.971
49.601


14
JB004 40 μM
1.350
1.479
1.691
1.507
76.971


14
JB004 80 μM
1.684
1.413
1.876
1.657
84.675









1.7. EN11-16, EN11-13, EN11-07, EN11-02, EN11-11



















Average LDH
Average





release
(% positive


No.
Compound
LDH release (% of max.)
(% of max.)
control)






















Medium
9.336
12.148
11.930
11.138
21.906


1
EN22 25 μM
51.852
49.199
51.489
50.847
100.000


15
EN11-16 25 μM
7.869
8.212
7.138
7.740
15.222


15
EN11-16 50 μM
8.330
8.767
7.683
8.260
16.245


15
EN11-16 100 μM
14.248
14.125
9.163
12.512
24.607


16
EN11-13 25 μM
7.549
7.580
7.769
7.633
15.011


16
EN11-13 50 μM
10.381
9.817
10.629
10.276
20.209


16
EN11-13 100 μM
19.337
15.515
13.905
16.252
31.963


30
EN11-07 25 μM
13.382
14.369
13.866
13.873
27.283


30
EN11-07 50 μM
14.413
15.441
13.545
14.466
28.451


30
EN11-07 100 μM
11.406
14.721
14.340
13.489
26.529


31
EN11-02 25 μM
10.764
11.389
9.725
10.626
20.897


31
EN11-02 50 μM
10.395
10.847
10.953
10.731
21.105


31
EN11-02 100 μM
10.119
10.610
9.486
10.072
19.808


32
EN11-11 25 μM
6.552
5.506
1.276
4.445
8.741


32
EN11-11 50 μM
6.570
5.610
0.416
4.199
8.257


32
EN11-11 100 μM
5.907
6.309
4.141
5.452
10.723


























Average
Average





IL-1β
(% positive


No.
Compound
IL-1β (ng/ml)
(ng/ml)
control)






















Medium
0.013
0.000
0.011
0.008
0.536


1
EN22 25 μM
1.387
1.391
1.646
1.474
100.000


15
EN11-16 25 μM
0.010
0.014
0.000
0.008
0.555


15
EN11-16 50 μM
0.068
0.054
0.058
0.060
4.074


15
EN11-16 100 μM
0.334
0.324
0.315
0.325
22.016


16
EN11-13 25 μM
0.000
0.011
0.023
0.011
0.764


16
EN11-13 50 μM
0.067
0.067
0.067
0.067
4.560


16
EN11-13 100 μM
0.243
0.331
0.316
0.297
20.130


30
EN11-07 25 μM
0.000
0.000
0.000
0.000
0.000


30
EN11-07 50 μM
0.000
0.000
0.000
0.000
0.000


30
EN11-07 100 μM
0.059
0.052
0.049
0.053
3.621


31
EN11-02 25 μM
0.015

0.015
0.015
1.019


31
EN11-02 50 μM
0.040
0.050
0.036
0.042
2.832


31
EN11-02 100 μM
0.053
0.052
0.046
0.050
3.424


32
EN11-11 25 μM
0.000
0.000
0.000
0.000
0.000


32
EN11-11 50 μM
0.021
0.022
0.000
0.014
0.975


32
EN11-11 100 μM
0.012
0.021
0.000
0.011
0.761









1.8. BMDC Isolation and Differentiation

Mouse bone marrow-derived dendritic cells (BMDCs) were isolated from femurs and tibias previously described (Gross, Olaf, 2012, “Measuring the inflammasome”, Methods in molecular biology, Clifton, N.J., 844, pp. 199-222.). In brief, tibias and femurs were carefully removed and put to sterile 70% ethanol for 1 minute. Using a 22-G needle attached to a 20 ml-syringe filled with sterile flushing medium, the bone marrow was pressed through the mesh of a 100 μM cell strainer. The cells were spun down by centrifugation at 400×g, 4° C. for 5 minutes and red blood cells lysed by resuspension and incubation for 5 minutes at room temperature in RBC lysis buffer. 9 ml of flushing medium were added and cells were again spun down by centrifugation at 400×g, 4° C. for 5 minutes. Cells were then resuspended in BMDC medium (RPMI, 10% fetal bovine serum, 1% penicillin-streptomycin100 U/ml, 20 ng/ml recombinant murine GM-CSF), counted and plated at a density of 0.5×106 cells ml−1 in 10 ml medium and incubated at 37° C., 5% CO2 in a humidified incubator. After 2 days, 10 ml of fresh medium were added to the cells and after 4 days, the medium was fully exchanged to 20 ml of fresh medium. The cells were allowed to differentiate for 6 to 8 days before usage.


1.9. Harvesting of BMDCs and BMDMs

BMDCs after 6 to 8 days of differentiation were harvested by transferring floating cells to 50 ml Falcon tubes and dislodging adherent cells with HBSS containing 5 mM EDTA.


1.10. High-Throughput Screening for Inflammasome Activators

To screen for new small molecule inflammasome activators, a 2-step semi-automated screening strategy was devised and performed. Small molecules from a Novartis public compound library were tested and access to high-throughput screening equipment was provided via the Novartis Facilitated Access to Screening Technologies (FAST) Lab.


For primary screening (see FIG. 1), 200 nl of the small molecule stock solutions (10 mM or 5 mM in DMSO) were pre-dispensed to white 384 well flat-bottom microplates with an Echo® 555 Liquid Handler (Labcyte Inc.). The plates were sealed with a PlateLoc Thermal Microplate Sealer (Agilent Technologies Inc.) and stored at 4-8° C. until further use. Plates were brought to ambient temperature before usage.


BMDCs from wild-type mice after 7 days of differentiation were harvested as described above. Depending on the volume of cell suspension needed for the experiment, the cells were resuspended in an appropriate volume of BMDC and counted with a Countess™ II FL Automated Cell Counter (Thermo Fisher Scientific). The concentration was adjusted to 0.25×106 cells ml−1 and the cells were primed with 50 ng ml−1 E. coli K12 ultra-pure LPS. Immediately after addition of LPS, 40 μl per well cell suspension were plated to the microplates already containing the small molecules to be tested using a Multidrop™ Combi Reagent Dispenser (Thermo Fisher Scientific) resulting in a final small molecule concentration of 50 μM or 25 μM. The cell suspension was mixed on a regular basis to ensure a consistent plating density. 8 wells per plate were filled with nigericin at a final concentration of 5 μM, DMSO at a final concentration of 0.5% or imiquimod and CL097 as references at a final concentration of 100 μM. It was then incubated at 37° C., 5% CO2 in a humidified incubator for about 20 hours. After the incubation, the plates were removed from the incubator and 10 μl per well CellTiter-Glo® Reagent (Promega Corporation) were added with the Multidrop™ Combi Reagent Dispenser. After at least 15 minutes of incubation at room temperature, the luminescence was determined with an EnVision 2105 Multimode Plate Reader (PerkinElmer Inc.) equipped with an EnVision stacker for semi-automated plate loading. Luminescence signals were normalized to nigericin as a positive control for inflammasome activation and DMSO as a negative control. Small molecules were considered active and chosen for subsequent counter screening when they induced a reduction in the luminescence signal that was at least 50% (25% in some cases) of the reduction induced by 5 μM nigericin (see FIG. 1).


For counter screening (see FIGS. 2 and 3), two independent strategies were pursued. To test for inflammasome dependency of cell viability reduction, BMDCs from either wild-type or Pycard−/− mice (Mariathasan, S., Newton, K., Monack, D. et al. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature 430, 213-218 (2004). https://doi.org/10.1038/natureO2664) were used and the same steps were performed as described above for primary screening, whereas the incubation time was reduced to about 16 hours. Three independent replicates of each plate were prepared and measured and the small molecules were either tested at a final concentration of 50 μM or at a dose response of 4 or 8 concentrations per compound going down with a dilution factor of 1/3.16 starting from 50 μM. Small molecules, that induced a reduction in the luminescence signal in the wild-type but not in the Pycard−/− BMDCs were considered as hits for further evaluation (see FIGS. 2 and 4A-C).


To functionally counter-screen (see FIG. 3) for induction of IL-1β release, BMDCs from wild-type mice were harvested, counted and the concentration adjusted to 0.25×106 cells ml−1. The cells were primed with 50 ng ml−1 E. coli K12 ultra-pure LPS directly before plating 40 μl per well cell suspension to white 384 well flat-bottom microplates and it was incubated for 3 hours at 37° C., 5% CO2 in a humidified incubator. 200 nl or 400 nl of the small molecule stock solutions from the libraries that had before been transferred to mother plates for the Echo® 555 Liquid Handler were then added to the microplates with the primed BMDCs to a final compound concentration of 50 μM. After 20 hours of incubation at 37° C., 5% CO2, IL-β release from the cells was then determined by using HTRF® technology developed by Cisbio from cell-free supernatants. HTRF (Homogeneous Time Resolved Fluorescence) is the most frequently used generic assay technology to measure analytes in a homogenous format, which is the ideal platform used for drug target studies in high-throughput screening. This technology combines fluorescence resonance energy transfer technology (FRET) with time-resolved measurement. The measurements were carried out as described in the manufacturer's manual.


1.11. Inflammasome Stimulation

For canonical inflammasome activation BMDCs after 7-9 days of differentiation were harvested and counted with a Neubauer Chamber (Hecht Assistant) and the concentration was set to 1 Mio cells ml−1. 100 μl cell suspension per well were seeded in 96-well plates. The cells were primed with 50 ng ml−1 E. coli K12 ultra-pure LPS for 3 hours and subsequently treated with canonical inflammasome activators or experimental compounds for 3 hours if not indicated differently (incubation time FIG. 4D, 5, 7, 8, 11, 13-18: 3 h, FIG. 12: 4 h). Concentrations of the stimuli were as follows if not indicated differently: 5 μM nigericin, 100 μM imiquimod (also known as R837), 100 μM CL097, 100 μM R848, 1 pg ml−1 poly(dA:dT), 300 μg ml−1 MSU. If an inhibitor was used before stimuli treatment, the inhibitor was added after 2.5 hours of priming and it was incubated for 30 minutes before inflammasome activation. Inhibitors were used as follows: 3 μM MCC950, 45 mM KCl, 30 μM ebselen (2-Phenyl-1,2-benzoselenazol-3-one), 5 μM cytochalasin D (see FIGS. 8, 11, 12 and 17).


All inflammasome stimulations were performed in three technical replicates for each condition. Lytic cell death was determined via LDH release (see FIGS. 8, 13-16 and 18), cytokines quantified in cell-free supernatants via ELISA (see FIGS. 4D, 7-9, 11-19) and cell lysates and supernatants analyzed via immunoblotting (see FIGS. 5A and 5B) as previously described in detail (Gross, Olaf, 2012 “Measuring the inflammasome”, Methods in molecular biology, Clifton, N.J., 844, pp. 199-222. Schneider, Katharina S.; Thomas, Christina J.; Groß, Olaf, 2013, “Inflammasome activation and inhibition in primary murine bone marrow-derived cells, and assays for IL-1α, IL-1β, and caspase-1”, Methods in molecular biology, Clifton, N.J., 1040, pp. 117-135).


1.12. Toll-Like Receptor Stimulation

To test for secretion of inflammasome independent cytokines (TNF and IL-6), BMDCs after 7-9 days of stimulation were left unprimed and then stimulated as described in Example 1.4. Cell-free supernatants were analyzed via ELISA (see FIGS. 9 and 18).


1.13. Determination of Cell Viability
1.13.1 LDH Release

To assess lytic cell death, release of the stable cytosolic enzyme lactate dehydrogenase (LDH) was determined using the CytoTox 96© Non-Radioactive Cytotoxicity kit (Promega Corporation) according to the manufacturer's instructions. The volumes of cell-free supernatants and reagent used were reduced to 35 μl. Background signal that was determined from medium without cells was subtracted from all values and all results were depicted as % of cells that had been lysed with 0.8% Triton® X-100 45 minutes before performing the assay. Absorbance at 490 nm was determined using a Tecan Infinite M200 plate reader (Tecan Life Sciences) (see FIGS. 7, 8, 13-16 and 18).


1.13.2 Cellular ATP

Cellular ATP levels as were determined with the CellTiter-Glo® Luminescent Cell Viability kit (Promega Corporation) according to the protocol provided by the manufacturer except that one fourth of the recommended amount of reagent was used. Luminescence intensities were normalized as % of the highest signals obtained, usually in the untreated control conditions.


1.14. Cytokine Quantification
1.14.1. ELISA

Cytokines were quantified in cell-free supernatants by using enzyme-linked immunosorbent assay (ELISA) kits for murine IL-1β, TNF and IL-6 (Thermo Fisher Scientific). The volumes of cell-free supernatants, antibodies and reagents used were reduced to 50 μl. Each condition was measured as 3 independent technical replicates and data was depicted as mean±SD. Absorbance at 450 nm and 570 nm was determined using a Tecan Infinite M200 plate reader (Tecan Life Sciences). Absolute values were interpolated from a standard curve that was generated with the Magellan™ Data Analysis Software. Depending on the expected cytokine quantity cell-free supernatants were diluted so that the detection values were within the linear range of the standard curve (see FIGS. 4D, 7-9, and 11-19).


1.15. Immunoblotting

Cell lysates for immunoblotting were generated by lysing the cells with 1× reducing SDS-PAGE loading buffer followed by 1 freeze-thaw cycle. Cell-free supernatants were diluted with 3× reducing SDS-PAGE loading buffer. Samples from 3 independent technical replicates were pooled and proteins were separated via SDS-PAGE (sodium dodecyl sulfate polyacrylamide gel electrophoresis) using 12% polyacrylamide gels and applying 80-120 V for 1-2.5 hours. Afterwards, proteins were transferred to nitrocellulose membranes by application of 100 V for 100 minutes. Membranes were rinsed with PBS-tween and success of protein transfer checked via Ponceau S staining. The membranes were then washed with PBS-tween again until Ponceau was completely removed and blocked with 2% skim milk powder in PBS-tween for 1 hour at room temperature. Membranes were rinsed with PBS-tween again and primary antibodies applied overnight at 4° C. (diluted in PBS-tween, 2% skim milk powder, 0.05% sodium azide). Before adding HRP-conjugated secondary antibodies (diluted 1:5,000 in PBS-tween, 2% skim milk powder) for 1 hour at room temperature, membranes were washed with PBS-tween for 2 hours, replacing with fresh PBS-tween every 15 minutes. Enhanced chemiluminescence substrate was applied before imaging with a cooled charge-coupled device. Depending on the signal intensity (16 bit, 64K grayscale) it was exposed for 2-15 minutes.


1.16. Immunofluorescence Imaging for ASC Specks

BMDCs from either wild-type or ASC-citrine mice expressing a ASC fluorescent protein were seeded in chambered coverslips for cell imaging (p-Slide, Ibidi) and primed with 50 ng ml−1 LPS for 3 hours. The cells were then incubated with the inflammasome activator of interest at the respective conditions. After the treatment, the cells were washed with PBS, fixed with 4% paraformaldehyde for 10 minutes and permeabilized with 0.1% (v/v) Triton™ X-100) for 5 minutes. The cells were then stained with polyclonal anti-ASC antibody derived from rabbit (Adipogen) and FITC anti-mouse CD45 antibody (Biolegend) overnight at 4° C. The next day, the cells were washed with blocking buffer and stained with a fluorescence labelled secondary antibody specific for rabbit for 30 minutes at room temperature, washed and subsequently resuspended in Vectashield® Antifade Mounting Medium with DAPI (H-1200) (Vector Laboratories) diluted 1:10 in blocking buffer for at least 10 minutes. Confocal microscopy of the immunostained cells was then performed. Immunostaining for ASC is dispensable if BMDCs from ASC-citrine mice are used.


1.17. Metabolic Analysis (Extracellular Flux Analysis)

Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured by extracellular flux analysis with a Seahorse XFe96 Analyzer. One day before the experiment, BMDCs were plated to XFe cell culture microplates at a density of 80,000 cells per well and a XFe sensor cartridge was rehydrated using XF Calibrant and put to 37° C. in a CO2-free incubator. On the day of the experiment, the cells were first primed with 50 ng ml−1 E. coli K12 ultra-pure LPS for 2-3 hours. The medium was then exchanged to bicarbonate- and phenol red-free seahorse base medium supplemented with 2 mM L-glutamine, 10 mM glucose and 5 mM HEPES, pH 7.4 and the cells were incubated at 37° C. in a CO2-free incubator for 1 hour. In the meantime, 10× dilutions of compounds and inhibitors to be tested were prepared in seahorse base medium and loaded to the injection ports of the rehydrated XFe sensor cartridge. Small molecules were used as indicated and respiratory chain inhibitors (piericidin, antimycin A) and mitochondrial uncouplers (FCCP) were used as follows after an initial titration: 10 μM piericidin, 2 μM antimycin A, 1 μM FCCP.


1.18. Materials





    • 1×PBS (Gibco™, Thermo Fisher Scientific)

    • 3× reducing SDS-PAGE loading buffer: 37.5 ml 1M Tris-HCl pH 7.0 (=187.5 mM), 12 g SDS (=6% w/v), 60 mg phenol red (=0.03% w/v) (Sigma Aldrich), 69 g glycerol (=30% w/v); adjust pH with 0.1N HCl to pH 6.8, 66.7 mM DTT (Carl Roth)

    • 4% Paraformaldehyde solution (Sigma Aldrich)

    • Blocking buffer: PBS (Gibco™, Thermo Fisher Scientific), 5% FCS (Gibco™, Thermo Fisher Scientific), 0.1% Triton™ X-100 (Sigma Aldrich)

    • BMDC medium: RPMI (Gibco™, Thermo Fisher Scientific), 10% FCS (Gibco™, Thermo Fisher Scientific), 100 units ml-1 penicillin/streptomycin (Gibco™, Thermo Fisher Scientific), 50 ng ml-1 recombinant murine GM-CSF (Immunotools)

    • CellTiter-Glo® Luminescent Cell Viability Assay (Promega Corporation)

    • CytoTox 96@ Non-Radioactive Cytotoxicity Assay (Promega Corporation)


    • E. coli K12 ultra-pure LPS (Invivogen)

    • IL-1β, TNF and IL-6 Mouse Uncoated ELISA Kit (Thermo Fisher Scientific)

    • PBS-tween: 4 g KH2PO4 (Potassium phosphate monobasic, Sigma Aldrich), 4 g KCl (Potassium chloride, Sigma Aldrich), 160 g NaCl (>99.8%, Carl Roth), 28.4 g Na2HPO4×2 H2O pH 7.2-7.4 (di-Natriumhydrogenphosphat Dihydrat, >99.5%, Carl Roth), 20 ml Tween-20 (Sigma Aldrich) in 20 I H2O.

    • Primary antibodies for immunoblotting and immunostaining:
      • Anti-Asc, pAb (AL177) (Adipogen)
      • Anti-Caspase-1 (p20) (mouse), mAb (Casper-1) (Adipogen)
      • Mouse IL-1β/IL-1F2 Antibody (R&D)

    • XFe flux analysis consumables and reagents (Agilent Technologies)

    • Nigericin sodium salt (Sigma Aldrich)

    • Imiquimod (R837) (Invivogen)

    • CL097 (Invivogen)

    • R848 (Invivogen)





1.19. Devices





    • Countess™ II FL Automated Cell Counter (Thermo Fisher Scientific)

    • Echo® 555 Liquid Handler (Labcyte Inc.)

    • EnVision 2105 Multimode Plate Reader (PerkinElmer Inc.)

    • Humidified CO2 incubator (Heracell™ 150, Thermo Fisher Scientific)

    • Intas Chemostar ECL Imager (Intas Science Imaging)

    • Leica SP8 confocal microscope equipped with a 63×/1.40 and 40×/1.25 oil objectives (Leica Microsystems)

    • Multidrop™ Combi Reagent Dispenser (Thermo Fisher Scientific)

    • Neubauer chamber (Hecht Assistent)

    • Seahorse XFe96 Flux Analyzer (Agilent Technologies)

    • Tecan Infinite M200 plate reader (Tecan Life Sciences)





2. Synthesis
2.1. General Synthesis Procedure A

An Aniline derivative (1 eq.) was dissolved in an aprotic solvent such as dry DCM or dry THF.


Isocyanatobenzene, thioisocyanatobenzene, 1-isocyanato-4-methoxybenzene or 4-isocyanato-benzonitrile (1 eq.) dissolved in an aprotic solvent such as dry DCM or dry THF was added dropwise to the reaction with a syringe over 5 min. The reaction was stirred at 25° C. for 4 h or until completion of the reaction and stopped by evaporation of the solvents. The crude solid was purified by filtration with cyclohexane/DCM and/or by flash column chromatography with a cyclohexane/ethyl acetate or DCM/methanol of varying eluent composition.


2.2. General Synthesis Procedure B

Triphosgen (0.4 eq.) was dissolved in dry DCM or dry THF and a solution of a primary or secondary aromatic amine (1 eq.) and triethylamine (3 eq.) in dry DCM or dry THF was added dropwise with a syringe over 10 min. The reaction was stirred at 25° C. for 2 h. The solvent was removed by a rotary evaporator under the hood. The crude was filtrated with cyclohexane and the filtrate collected and evaporated. The resulting solid containing the isocyanate was used without further purification in following reactions. No NMR spectra were recorded.


2.3. General Synthesis Procedure C

An isocyanate (1.2 eq.) or carbamic chloride (1.2 eq.) synthesized via general synthesis procedure B was dissolved in an aprotic solvent such as dry DCM or dry THF and added dropwise with a syringe over 5 min to a solution of an aniline (1 eq.) or primary amine (1 eq.) dissolved in an aprotic solvent such as dry DCM or dry THF and stirred at 25° C. for 4 h or until completion of the reaction. The crude solid was purified by filtration with cyclohexane/DCM and/or by flash column chromatography on SiO2 gel with a cyclohexane/ethyl acetate or DCM/MeOH of varying eluent composition or HPLC.


2.4. Preparation of Compound EN21



embedded image


Step A:

To a stirred suspension of 4-(trifluoromethyl)benzonitrile (1) (15 g, 87.7 mmol, 1 eq.) and; sodium hydrogencarbonate (8.8 g, 105.2 mmol, 1.2 eq.) in methanol (150 ml), hydroxylamine hydrochloride (7.3 g, 105.2 mmol, 1.2 eq.) was added portion wise and stirred overnight. After completion of the reaction (TLC data), the mixture was filtrated and the filtrate was carefully evaporated in vacuo at 35° C. The resulting residue was suspended in water (200 mL), stirred for 15 min and then filtrated to afford N′-hydroxy-4-(trifluoromethyl)benzimidamide (2) (16.2 g, 91% yield) as white solid.


Step B:

To a stirred solution of 2-((tert-butoxycarbonyl)amino)acetic acid (15.3 g, 87.3 mmol, 1.1 eq.) in dry 1,4-dioxane (180 mL), 1,1′-carbonyldiimidazole (CDI) (12.9 g, 79.4 mmol, 1 eq.) was added by portion at 60° C. and the resulting mixture was stirred for 30 min at 60° C. N′-hydroxy-4-(trifluoromethyl)benzimidamide (2) (16.2 g, 79.4 mmol, 1 eq.) was added at once and the resulting mixture was stirred at 60° C. until full consumption of 4-(trifluoromethyl)benzimidamide (2) was being achieved (TLC data, approximately 30 min for this scale). Then the reaction mixture was heated at 90° C. for 12 h, cooled to r.t. and evaporated in vacuo. The resulting residue was diluted with 30% aq. solution of potassium carbonate (200 mL) and ethyl acetate (200 mL), the water layer was separated and extracted with ethyl acetate (2×100 mL). The combined organic layers were dried over anhydrous sodium sulfate and evaporated in vacuo. The crude material was purified by column chromatography (chloroform:ethyl acetate, v/v=10:1, rf=0.8) to afford tert-butyl((3-(4-(trifluoromethyl)phenyl)-1,2,4-oxadiazol-5-yl)methyl)carbamae (3) (16.1 g, 59% yield) as white solid.


Step C, D:

To a stirred trifluoroacetic acid (10 mL), tert-butyl ((3-(4-(trifluoromethyl)phenyl)-1,2,4-oxadiazol-5-yl)methyl)carbamate (3) (2 g, 5.8 mmol) was added by portion at r.t. and the resulting mixture was stirred for 30 min. The solvent was removed in vacuo and the residue was diluted with dichloromethane and threated with excess of DIPEA (3.1 mL, 17.5 mmol, 3 eq.). The resulting mixture was stirred at r.t. for 5 min and then, 1-isocyanato-2-methoxybenzene (0.9 g, 5.8 mmol, 1 eq.) was added at once. The resulting mixture was stirred for additional 30 min. The resulting precipitate was filtrated and washed with dichloromethane (2×10 mL) to afford 1-(2-methoxyphenyl)-3-((3-(4-(trifluoromethyl)phenyl)-1,2,4-oxadiazol-5-yl)methyl)urea (EN21, also called EN02-01) (1.1 g, 48% summary yield) as white solid.


EN21: 1H NMR (400 MHz, dmso) δ 3.86 (s, 3H), 4.72 (d, 2H), 6.83 (t, 1H), 6.91 (t, 1H), 6.99 (d, 1H), 7.67 (t, 1H), 7.94 (d, 2H), 8.02 (d, 1H), 8.22 (d, 2H), 8.28 (s, 1H).


2.5. Preparation of compound EN22




embedded image


Step E:

To a solution of amine 6 (3.29 g, 22.8 mmol) in dry dichloromethane (50 mL) was added isocyanate 5 (2.72 g, 22.8 mmol). The reaction mixture was stirred for 2 h at r.t. and the precipitated solid was collected, washed with dichloromethane and hexane, and dried in vacuum to afford 5.15 g of compound EN22 (also called EN02-02) (85% yield).


EN22: 1H NMR (400 MHz, dmso+ccl4) δ 6.99 (t, 1H), 7.30 (t, 2H), 7.45 (dd, 3.9 Hz, 1H), 7.50 (d, 2H), 7.70 (d, 1H), 7.94 (d, 1H), 8.18 (s, 1H), 8.25 (d, 1H), 8.74 (d, 1H), 8.79 (s, 1H), 9.03 (s, 1H).


2.6. Preparation of Compound EN54



embedded image


Step A:

To a stirred solution of acetyl chloride (4.7 g, 60 mmol) in dichloromethane (60 ml) aluminum chloride (8.0 g, 60 mmol) was added. The mixture was cooled to 0° C. and methyl 1-indolizinecarboxylate (1) (7.0 g, 40 mmol) in dichloromethane (40 ml) was added dropwise. The resulting mixture was stirred overnight at room temperature, poured in ice water and diluted with HCl. The organic phase was separated, the water phase was extracted with dichloromethane twice, and the combined organic phase was washed with saturated aqueous sodium bicarbonate and dried over potassium sulphate. The solvent was removed under reduced pressure to give methyl 3-acetyl-1-indolizinecarboxylate (2) without further purification as yellow solid. Yield 7.2 g, 84%.


Step B:

A mixture of methyl 3-acetyl-1-indolizinecarboxylate (2) (7.2 g, 33.6 mmol) and DMF-DMA (8.0 g, 67.2 mmol) in dimethylformamide (10 ml) was heated to 110° C. overnight. The solution was cooled to room temperature, and the formed solid product was collected by filtration and washed with hexane and dried to obtain compound 3. Yield 7.0 g, 78%.


Step C:

A mixture of methyl 3-[3-(dimethylamino)acryloyl]indolizine-1-carboxylate (3) (7.0 g, 26 mmol) and hydrazine hydrate (6.5 g, 130 mmol) in methanol (50 ml) was refluxed for 3 h. The solution was cooled to room temperature and diluted with water (100 ml). The solid product was collected by filtration and dried to obtain compound 4. Yield 4.2 g, 67%.


Step D:

To a stirred solution of methyl 3-(1H-pyrazol-3-yl)indolizine-1-carboxylate (4) (4.2 g, 17.4 mmol) in DMF (40 ml) sodium hydride (0.85 g, 20.9 mmol as suspension in mineral oil) was added. The mixture was stirred for 30 min., cooled to 0° C. and bromocyclopenthane (3.9 g, 26.1 mmol) in DMF (10 ml) was added dropwise. The resulting mixture was stirred overnight at room temperature and poured in ice water. The solid product was collected by filtration and dried to obtain compound 5. Yield 4.8 g, 89%.


Step E:

To a stirred suspension of ethyl 3-(1-cyclopentyl-1H-pyrazol-3-yl)indolizine-1-carboxylate (5) (4.8 g, 15.5 mmol) in methanol (50 ml) potassium hydroxide (8.7 g, 155 mmol) in water (40 ml) was added and the resulting mixture was refluxed for 24 h. The clear solution was cooled to room temperature, diluted with water (50 ml) and diluted HCl was added. The solid product was collected by filtration and dried to obtain compound 6. Yield 4.2 g, 67%.


Step F:

To a stirred suspension of 3-(1-cyclopentyl-1H-pyrazol-3-yl)indolizine-1-carboxylic acid (6) (3.2 g, 10.8 mmol) in dichloromethane (40 ml) thionyl chloride (2.6 g, 21.7 mmol) was added and the resulting mixture was stirred at room temperature to obtain of the clear dark green solution (6-8 h). The solvent and the excess of thionyl chloride were removed under reduced pressure, and the residue was dissolved in dichloromethane (40 ml) and added dropwise to a stirred solution of 2-methoxyethylamine (1.2 g, 16.3 mmol) and DIPEA (2.8 g, 21.7 mmol) in dichloromethane (40 ml) at room temperature. The resulting mixture was stirred overnight, washed with diluted HCl, saturated aqueous sodium bicarbonate and dried over potassium sulphate. The solvent was removed under reduced pressure to give 3.1 g (82%) of the title product. The analytical sample of compound EN54 (also called EN05-04) was obtained by column chromatography on silica gel using ethyl acetate as an eluent.


EN54: 1H NMR (400 MHz, dmso) δ 1.68 (m, 2H), 1.83 (m, 2H), 2.02 (m, 2H), 2.16 (m, 2H), 3.28 (s, 3H), 3.46 (m, 4H), 4.8 (m, 1H), 6.57 (d, 1H), 6.92 (t, 1H), 7.07 (t, 1H), 7.73 (s, 1H), 7.90 (d, 1H), 8.01 (t, 1H), 8.33 (d, 1H), 9.40 (d, 1H).


The measurements of the NMR spectra were done with Bruker AVANCE DRX 500 Mhz and Bruker AVANCE III 400 Mhz.


2.7. Preparation of Compound AH4

1-phenyl-3-(quinolin-6-yl) thiourea. To a stirred solution of quinolin-6-amine (144.18 mg, 1 mmol, 1.0 equiv.) in dichloromethane (5 mL), 1-isothiocyanatobenzene (135.18 mg, 1 mmol, 1.0 equiv.) was added drop wise. The mixture was stirred at room temperature for 5 h and concentrated. The residue was purified on silica gel column chromatography eluting with 1-10% methanol in dichloromethane to afford the desired product as yellow solid (174.02 mg, 0.623 mmol, 62.3%).


AH4: 1H NMR (400 MHz, DMSO-d6) δ 10.12 (s, 1H), 9.99 (s, 1H), 8.83 (dd, J=4.2, 1.7 Hz, 1H), 8.31 (dd, J=8.4, 1.7 Hz, 1H), 8.09 (d, J=2.4 Hz, 1H), 7.98 (d, J=9.1 Hz, 1H), 7.86 (dd, J=9.0, 2.4 Hz, 1H), 7.61-7.41 (m, 3H: 2H, CH, benzyl, 1H), 7.41-7.27 (m, 2H), 7.20-7.11 (m, 1H).


2.8. Preparation of Compound JB010

1-(4-methoxyphenyl)-3-(quinolin-6-yl)urea was synthesized according to general synthesis procedure A. The crude solid was washed and filtered with DCM/cyclohexane (50/50) to gain a white solid (303.2 mg, Yield: 90.0%); rf=0.2 (cyclohexane/ethyl acetate 20/80).


JB010: 1H NMR (400 MHz, DMSO-d6) δ 8.96 (s, 1H, quinoline-NHurea), 8.74 (dd, J=4.2, 1.7 Hz, 1H, quinoline H2), 8.62 (s, 1H, methoxyphenyl-NHUrea), 8.25 (dd, J=8.7, 1.0 Hz, 1H, quinoline H4), 8.17 (d, J=2.4 Hz, 1H, quinoline H5), 7.94 (d, J=9.1 Hz, 1H, quinoline H8), 7.70 (dd, J=9.1, 2.4 Hz, 1H, quinoline H7), 7.46 (dd, J=8.3, 4.2 Hz, 1H, quinoline H3), 7.43-7.38 (m, 2H, methoxyphenyl H2.6), 6.93-6.87 (m, 2H, methoxyphenyl H3.5), 3.73 (s, 3H, phenyl-OCH3).


2.9. Preparation of Compound JB011

1-(4-cyanophenyl)-3-(quinolin-6-yl)urea was synthesized according to general synthesis procedure A. The crude solid was washed and filtered with DCM/cyclohexane (50/50) to gain a white solid (267.8 mg, Yield: 90.2%); rf=0.1 (cyclohexane/ethyl acetate 20/80).


JB011: 1H NMR (400 MHz, DMSO-d6) δ 9.34 (s, 1H, quinoline-NHUrea), 9.24 (s, 1H, cyanophenyl-NHUrea), 8.77 (dd, J=4.2, 1.7 Hz, 1H, quinoline H2), 8.28 (dd, J=8.6, 0.9 Hz, 1H, quinoline H4), 8.19 (d, J=2.3 Hz, 1H, quinoline H5), 7.97 (d, J=9.0 Hz, 1H, quinoline H8), 7.77 (dt, J=8.9, 2.1 Hz, 2H, phenyl H2,6), 7.73 (dd, J=9.1, 2.5 Hz, 1H, quinoline H7), 7.69 (dt, J=9.0, 2.1 Hz, 2H, phenyl H3,5), 7.48 (dd, J=8.3, 4.2 Hz, 1H, quinoline H3).


2.10. Preparation of Compound PMO7 and PM16
Synthesis of ethyl 2-(1-(3-fluoro-2-methyl benzyl) piperidin-3-yl) acetate

The synthesis was performed according to the procedure reported in literature:

  • D. Monaldi, D. Rotili, J. Lancelot, M. Marek, N. Wössner, A. Lucidi, D. Tomaselli, E. Ramos-Morales, C. Romier, R. J. Pierce, A. Mai, M. Jung; J. Med. Chem. 62 (2019) 8733-8759.


Synthesis of Tert-butyl (4-(N′-hydroxycarbamimidoyl) phenyl]carbamate

To a suspension of tert-butyl(4-cyanophenyl) carbamate (1.0 equiv.) and hydroxylamine hydrochloride (1.5 equiv.) in EtOH (25 mL), NaHCO3 (1.5 equiv) was added. The mixture was stirred under reflux for 6 h. and eventually diluted with cold water (50 mL). the aqueous phase was extracted with ethyl acetate (3×50 mL). the combined organic phases were dried over Na2SO4, filtrated and concentrated. The residue was then purified on silica gel column chromatography eluting with 1-10% methanol in dichloromethane to afford the desired product as white solid (1.12 g, 4.46 mmol, 97%).


1H NMR (400 MHz. DMSO-d6): δ 9.48 (s, 1H), 9.45 (s, 1H), 7.55 (d, J=8.1 Hz, 2H), 7.43 (d, J=8.1 Hz, 2H), 5.71 (s, 2H), 1.47 (s, 9H).


Synthesis of N-Tert-butyl (4-(5-((1-(3-fluoro-2-methylbenzyl) piperidin-3-yl) methyl)-1,2,4-oxadiazol-3-yl) phenyl)carbamate

To a solution of ethyl 2-(1-(3-fluoro-2-methyl benzyl) piperidin-3-yl) acetate (1.5 equiv.) and tert-butyl (4-(N′-hydroxycarbamimidoyl) phenyl] carbamate (1.0 equiv.) in DMSO (3.9 mL) NaOH (1.6 equiv.) was rapidly added. The mixture was stirred at room temperature for 4 h and afterwards diluted with cold water (10 mL). The aqueous phase was extracted with cyclohexane (5×6 mL). The combined organic phases were dried over Na2SO4, filtrated and concentrated. The residue was then purified on silica gel column chromatography eluting with 1-10% methanol in dichloromethane to afford the desired product as colourless oil (1.01 g, 2.11 mmol, 54%).


1H NMR (400 MHz, DMSO-d6): δ 9.72 (s, 1H), 7.83 (d, J=8.3 Hz, 2H), 7.62 (d, J=8.3 Hz, 2H), 7.15-6.98 (m, 3H), 3.42 (s, 2H), 2.98-2.86 (m, 2H), 2.77-2.72 (m, 1H), 2.60-2.55 (m, 1H), 2.21 (s, 3H), 2.10-2.03 (m, 2H), 1.96-1.89 (m, 1H), 1.71-1.58 (m, 2H), 1.49 (s, 9H), 1.48-1.46 (m, 1H), 1.18-1.08 (m, 1H).


Synthesis of 4-(5-((1-(3-fluoro-2-methylbenzyl) piperidin-3-yl) methyl)-1,2,4-oxadiazol-3-yl) aniline

To a stirred solution of N-Tert-butyl (4-(5-((1-(3-fluoro-2-methylbenzyl) piperidin-3-yl) methyl)-1,2,4-oxadiazol-3-yl) phenyl) carbamate (1.0 equiv.) in dichloromethane (1.8 mL) trifluoroacetic acid (6 equiv.) was added at 0° C. and stirred for 2 h at 25° C. After completion of the mixture was saturated with sodium bicarbonate solution. The aqueous phase was extracted with ethyl acetate (3×6 mL) and the combined organic phases were dried over Na2SO4, filtrated and concentrated. The residue was then purified on silica gel column chromatography eluting with 1-10% methanol in dichloromethane to afford the desired product as yellow solid (183.6 mg, 0.483 mmol, 69%).


1H NMR (400 MHz, DMSO-d6): δ 7.60 (d, J=8.1 Hz, 2H), 7.12-7.01 (m, 3H), 6.63 (d, J=8.3 Hz, 2H), 5.75 (s, 2H), 3.51-3.39 (m, 2H), 2.97-2.83 (m, 2H), 2.79-2.71 (m, 1H), 2.67-2.56 (m, 1H), 2.21 (s, 3H), 2.11-2.02 (m, 2H), 1.95-1.86 (m, 1H), 1.75-1.55 (m, 2H), 1.50-1.38 (m, 1H), 1.14-1.08 (m, 1H).


1.0 equivalent of 4-(5-((1-(3-fluoro-2-methylbenzyl) piperidin-3-yl) methyl)-1,2,4-oxadiazol-3-yl) aniline was dissolved in DCM (1.3 mL). 1.2 equivalent of the corresponding isocyanate was added dropwise to the stirred solution. The mixture was stirred at room temperature for 4 h and concentrated.


PM07: 1-[4-(5-((1-(3-fluoro-2-methylbenzyl) piperidin-3-yl) methyl)-1,2,4-oxadiazol-3-yl) phenyl]-3-[(4-methoxy) phenyl] urea. The crude was purified on silica gel column chromatography eluting 5-10% methanol in dichloromethane to afford the desired product as white solid (53.1 mg, 0.101 mmol, 39%).


PM07: 1 H NMR (400 MHz, DMSO-d6): δ 8.96 (s, 1H), 8.60 (s, 1H), 7.88-7.85 (d, J=8.3 Hz, 2H), 7.64-7.61 (d, J=8.3 Hz, 2H), 7.39-7.37 (d, J=8.1 Hz, 2H), 7.17-6.99 (m, 3H), 6.90-6.86 (d, J=8.1 Hz, 2H), 3.72 (s, 3H), 3.42 (s, 2H), 3.00-2.88 (m, 2H), 2.77-2.75 (m, 1H), 2.65-2.56 (m, 1H), 2.22 (s, 3H), 2.13-2.04 (m, 2H), 1.97-1.91 (m, 1H), 1.74-1.59 (m, 2H), 1.51-1.41 (m, 1H), 1.19-1.11 (m, 1H).


PM16: 1-(4-(5-((1-(3-fluoro-2-methylbenzyl) piperidin-3-yl) methyl)-1,2,4-oxadiazol-3-yl) phenyl)-3-phenylurea. The crude was purified on silica gel column chromatography eluting 1-5% methanol in dichloromethane to afford the desired product as white solid (46.5 mg, 0.09 mmol, 36%).


PM16: 1H NMR (400 MHz, DMSO-d6): δ 9.07 (s, 1H), 8.82 (s, 1H), 7.88-7.85 (d, J=8.3 Hz, 1H), 7.64-7.61 (d, J=8.3 Hz, 1H), 7.48-7.46 (dd, J=8.3 Hz, 1.8 Hz), 7.29 (dd, J=8.3 Hz), 7.17-7.10 (m, 1H), 7.08-6.98 (m, 3H), 3.42 (s, 2H), 3.00-2.87 (m, 2H), 2.76-2.74 (m, 1H), 2.63-2.56 (m, 1H), 2.22-2.21 (s, 3H), 2.11-2.05 (m, 2H), 1.98-1.89 (m, 1H), 1.73-1.58 (m, 2H), 1.52-1.39 (m, 1H), 1.20-1.07 (m, 1H).


2.11. Preparation of Compound JB014:

1-(quinolin-6-yl)-3-((5-(4-(trifluoromethyl)phenyl)-1,2,4-oxadiazol-3-yl)methyl)urea was synthesized according to general synthesis procedure C. The crude solid was washed and filtrated with DCM/cyclohexane (50/50) and purified by flash column chromatography with DCM/MeOH (eluent composition: 100/0→90/10) and cyclohexane/ethyl acetate (100/0→0/100) to gain a white solid (71.6 mg, Yield: 31.9%); rf=0.4 (DCM/MeOH 90/10).


JB014: 1H NMR (400 MHz, DMSO-d6) δ 9.37 (s, 1H, quinolin-NHUrea), 8.71 (dd, J=4.2, 1.7 Hz, 1H, quinoline H2), 8.22 (d, J=8.1 Hz, 2H, phenyl H2,6), 8.25-8.17 (m, 3H, phenyl H2,6, quinoline H4), 8.13 (d, J=2.4 Hz, 1H, quinoline H5), 7.94 (d, J=8.2 Hz, 2H; phenyl H3,5), 7.91 (d, J=9.1 Hz, 1H, quinoline H8), 7.68 (dd, J=9.1, 2.4 Hz, 1H, quinoline H7), 7.43 (dd, J=8.3, 4.2 Hz, 1H, quinoline H3), 7.12 (t, J=5.8 Hz, 1H, methylene-NHUrea), 4.75 (d, J=5.8 Hz, 2H, oxadiazol-CH2-Urea).


2.12. Preparation of Compound AH1:

1-(2-chloro-4-nitrophenyl)-3-(3,5-dichlorophenyl) urea. To a stirred solution of 3, 5-dichloroanilin (162 mg, 1 mmol, 1.0 equiv.) in dichloromethane (5 mL), 2-chloro-1-isocyanato-4-nitrobenzene (200.7 mg, 1 mmol, 1.0 equiv.) was added drop wise. The mixture was stirred at room temperature for 4 h and concentrated. The residue was purified on silica gel column chromatography eluting with 30-100% ethylacetate in cyclohexane to afford the desired product as yellow solid (184.8 mg, 0.513 mmol, 53%).


AH1: 1H NMR (400 MHz, DMSO-d6) δ 10.04 (s, 1H), 8.91 (s, 1H), 8.51 (d, J=9.4 Hz, 1H), 8.36 (d, J=2.7 Hz, 1H), 8.22 (dd, J=9.3, 2.7 Hz, 1H), 7.53 (d, J=1.9 Hz, 2H), 7.26 (t, J=1.8 Hz, 1H).


2.13. Preparation of Compound AH3:

1-(3-chlorophenyl)-3-(3-fluoro-2-methylphenyl) urea. To a stirred solution of 3-chloroanilin (127.57 mg, 1 mmol, 1.0 equiv.) in dichloromethane (5 mL), 1-fluoro-3-isocyanato-2-methylbenzene (151.14 mg, 1 mmol, 1.0 equiv.) was added drop wise. The mixture was stirred at room temperature for 4.5 h and concentrated. The residue was purified on silica gel column chromatography eluting with 1-10% methanol in dichloromethane to afford the desired product as white solid (89.8 mg, 0.322 mmol, 32%).


AH3: 1H NMR (400 MHz, DMSO-d6) δ 9.25 (s, 1H), 8.18 (s, 1H), 7.75 (t, J=2.1 Hz, 1H), 7.66 (d, J=8.2 Hz, 1H), 7.32 (t, J=8.0 Hz, 1H), 7.29-7.13 (m, 2H), 7.03 (ddd, J=7.9, 2.1, 1.1 Hz, 1H), 6.89 (t, J=8.9 Hz, 1H), 2.15 (d, J=2.1 Hz, 3H).


2.14. Preparation of Compound JB002:

1-(1,5-naphthyridin-2-yl)-3-phenylurea was synthesized according to general synthesis procedure A. The precipitated solid was washed and filtered with DCM/cyclohexane (50/50) to gain an orange solid (184.8 mg, Yield: 93.3%); rf=0.32 (Cyclohexane/ethyl acetate 90/10).


JB002: 1H NMR (400 MHz, DMSO-d6) δ 11.67 (s, 1H, naphthyridine-NHUrea), 10.29-10.16 (m, 1H, phenyl-NHUrea), 8.83 (dd, J=4.2, 1.6 Hz, 1H, H2), 8.40-8.33 (m, 2H, H6/8 or 7), 7.75-7.72 (q, J=4.2 Hz, 1H, naphthyridine H7), 7.72-7-68 (m, 2H, phenyl H2,6), 7.67 (d, J=9.2 Hz, 1H, H7 or 8), 7.41-7.33 (tt, J=7.4, 2.0 Hz, 2H, phenyl H3,5), 7.09 (tt, J=7.4, 1.2 Hz, 1H, phenyl H4).


2.15. Preparation of Compound JB004:

1-(3-bromoquinolin-6-yl)-3-phenylurea was synthesized according to general synthesis procedure A. The precipitated solid was washed and filtered with DCM/cyclohexane (50/50) to gain a yellow solid (147.7 mg, Yield: 73.9%); rf=0.1 (Cyclohexane/ethyl acetate 40/60);


JB004: 1H NMR (400 MHz, DMSO-d6) δ 9.15 (s, 1H, bromoquinoline-NHUrea), 8.82 (s, 1H, phenyl-NHUrea), 8.77 (d, J=2.3 Hz, 1H, bromoquinoline H2), 8.63 (d, J=2.4 Hz, 1H, bromoquinoline H4), 8.15 (d, J=2.5 Hz, 1H, bromoquinoline H5), 7.96 (d, J=9.0 Hz, 1H, bromoquinoline H8), 7.78 (dd, J=9.1, 2.4 Hz, 1H, bromoquinoline H7), 7.51 (d, J=7.5 Hz, 2H, phenyl H2,6), 7.32 (t, J=8.0 Hz, 2H, phenyl H3,5), 7.01 (t, J=7.4 Hz, 1H, phenyl H4).


2.16. Preparation of Compound TCMDC-143175:

TCMDC-143175:_1H NMR (400 MHz, DMSO-d6) δ 9.93 (s, 1H), 8.87 (s, 1H), 8.56-8.53 (d, J=12 Hz, 1H), 8.36 (d, J=2.7 Hz, 1H), 8.22-8.20 (dd, J=12 Hz, 2.7 Hz, 1H), 7.76 (t, J=2.7 Hz, 1H), 7.36 (t, J=12 Hz, 1H), 7.28-7.26 (dd, J=10 Hz, 2.9 Hz 1H), 7.12-7.10 (dd, J=10 Hz, 2.9 Hz 1H).


2.17. Preparation of Compound A.C2:

A.C2: 1H NMR (400 MHz, DMSO-d6) δ 9.50 (s, 1H), 9.42 (s, 1H), 8.52-8.49 (d, J=12 Hz, 1H), 8.36 (d, J=2.7 Hz, 1H), 8.23-8.21 (dd, J=12 Hz, 2.7 Hz, 1H), 8.05-8.03 (d, J=12 Hz, 1H), 7.52-7.50 (d, J=12 Hz, 1H), 7.35 (t, J=12 Hz, 1H), 7.13 (t, J=12 Hz, 1H).


2.18. Preparation of Compound AH2:

1-(2-chloro-4-nitrophenyl)-3-(3-chlorophenyl) thiourea. To a stirred solution of 3-chloroanilin (127.57 mg, 1 mmol, 1.0 equiv.) in dichloromethane (5 mL), 2-chloro-1-isothiocyanato-4-nitrobenzene (215 mg, 1 mmol, 1.0 equiv.) was added drop wise. The mixture was stirred at room temperature for 4.5 h and concentrated. The residue was purified on silica gel column chromatography eluting with 40-100% ethylacetate in cyclohexane to afford the desired product as yellow solid (132.8 mg, 0.388 mmol, 39%).


AH2: 1H NMR (400 MHz, DMSO-d6) δ 10.50 (s, 1H), 9.93 (s, 1H), 8.39 (s, 1H), 8.21 (d, J=9.0 Hz, 1H), 8.12 (d, J=9.1 Hz, 1H), 7.81 (s, 1H), 7.47 (d, J=8.1 Hz, 1H), 7.41 (t, J=8.1 Hz, 1H), 7.25 (d, J=7.8 Hz, 1H).

Claims
  • 1.-21. (canceled)
  • 22. A compound for use in the prevention or treatment of a disease by activating NLRP3 inflammasome, wherein the disease is selected from a group comprising infectious diseases or cancer, and (I) wherein the compound is a compound of Formula (I)
  • 23. The compound for use according to claim 22, wherein the compound is a compound of Formula (Ic5), a compound of Formula (Ic6), a compound of Formula (Ic7), a compound of Formula (Ic8), a compound of Formula (Ic9) or a compound of Formula (Ic10), a compound of Formula (Ic11), a compound of Formula (Ic12), or a compound of Formula (Ic13), or a pharmaceutically acceptable salt thereof:
  • 24. The compound for use according to claim 22, wherein the compound is a compound of one of Formulae (Ia) or (Ia1)
  • 25. The compound for use according to claim 24, wherein R1, R2, R3, R4 and R5 are each independently hydrogen, —OCH3, —OCF3, —OCH2CH3, —OCH(CH3)2, —CH3, —C2H5, —Cl, —Br, —CN, —CH2F; —CHF2; —CF3; —CH2Cl; —CHCl2; —CCl3; NO2, or
  • 26. The compound for use according to claim 24, wherein R6 and R7 are each independently hydrogen, or R6 and R7 when taken together form a heterocyclic ring selected from:
  • 27. The compound for use according to claim 24, wherein R7, N and X when taken together form an six-membered heterocyclic ring, selected from
  • 28. The compound for use according to claim 24, wherein R1, R2, R3, R4 and R5 are each hydrogen and/or wherein A is
  • 29. The compound for use according to claim 28, with the proviso that R8 or R9 are not identical to each other.
  • 30. The compound for use according to claim 24, wherein R1 to R5 are selected as follows: R1 is hydrogen, —C1-C2 alkoxy, —C(O)O—C1-C5 alkyl, —OCH3, —C1-C3 alkyl, methyl, or ethyl;R2 is hydrogen, methyl, —F, —Cl, —Br, —CN, —OCH3, or —C(O)O—C1-C5 alkyl;R3 is hydrogen, methyl, —CH(CH3)2, —CHF2, —F, —Cl, —Br, —CN, —OCH3, —OCF3, NO2, or C(O)O—C1-C5 alkyl,R4 is hydrogen or —F, —Cl, —Br, or —CF3; and/orR5 is hydrogen, methyl, ethyl or —F, —Cl, —Br, or —CN, —CH2-phenyl.
  • 31. The compound for use according to claim 24, wherein the compound is a compound according to any of Formulae (Ib), (Ib1), (Ib2) or (Ib3):
  • 32. The compound for use according to claim 22, wherein the compound is a compound of Formula (Id):
  • 33. The compound for use according to claim 24, wherein the compound is a compound according to any of Formulae (Ib), (Ib1), (Ib2), (Ib3) or (Id) and R1 to R5 are selected as follows: R1 is hydrogen, —C1-C2 alkoxy, —C(O)O—C1-C5 alkyl, —OCH3, —C1-C3 alkyl, methyl, or ethylR2 is hydrogen, methyl, —F, —Cl, —Br, —CN, —OCH3, or —C(O)O—C1-C5 alkyl;R3, if present, is hydrogen, methyl, —CH(CH3)2, —CHF2, —F, —Cl, —Br, —CN, —OCH3, —OCF3, NO2, or —C(O)O—(C1-C5 alkyl);R4 is hydrogen or —F, —Cl, —Br, or —CF3; and/orR5 is hydrogen, methyl, ethyl or —F, —Cl, —Br, —CN, or —CH2-phenyl.
  • 34. The compound for use according to claim 22, wherein the compound is selected from the compounds delineated in Table 1.
  • 35. A compound of claim 22 for use in the prevention or treatment of a disease by activating NLRP3 inflammasome, wherein the disease is selected from a group comprising infectious diseases or cancer, or for use for boosting immune responses or breaking immune evasion in cancer diseases in a subject, or for use as an adjuvant in the context of vaccination to boost specific immune responses by activating NLRP3 inflammasome.
  • 36. Use of the compounds as defined in claim 34 for analyzing the activity of NLRP3 activation in vitro.
Priority Claims (1)
Number Date Country Kind
20202297.6 Oct 2020 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2021/078623 10/15/2021 WO