NOVEL AROMATIC COMPOUNDS

- XENIOPRO GMBH

The present invention comprises novel aromatic molecules, which can be used in the treatment of pathological conditions, such as cancer, skin diseases, muscle disorders, and immune system-related disorders such as disorders of the hematopoietic system including the hematologic system in human and veterinary medicine.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

The present invention relates to novel aromatic compounds and their use as therapeutic agents, which can be used in the treatment of pathological conditions, such as cancer, skin disorders, muscle disorders, and immune system-related disorders such as disorders of the hematopoietic system including the hematologic system in human and veterinary medicine.

BACKGROUND

Notch signaling is a fundamental cell-to-cell communication pathway that regulates central processes in embryonic development as well as in the maintenance of adult tissues. The effect of a Notch signal is highly dependent on the signal strength, duration, and most importantly on the cellular context. In this regard, Notch activity leads to numerous cell-type specific responses, which implicate for example cell fate decisions, the induction or inhibition of differentiation, and the regulation of cell proliferation.

If a signaling event is not correctly controlled, a consequent loss of balance in according cellular processes may drive abnormal cellular changes and finally end in diverse disease situations, such as cancer.

Initially, Notch signaling was discovered as an oncogenic pathway. Corresponding pathological conditions are linked to abnormally augmented signaling levels. In these particular cases, the use of Notch inhibiting agents represents a promising strategy for therapeutic intervention and numerous corresponding drugs are currently in development.

Conversely, there is increasing evidence for tumor-suppressor functions of the Notch pathway in other cellular contexts (Lobry et al., J. Exp. Med. 2011, 208, 1931-1935; South et al., Semin. Cell Dev. Biol. 2012, 23, 458-464), most notably concerning organs, in which Notch negatively impacts proliferation or triggers differentiation, such as in the skin or in the neuroendocrine system (Dotto, Oncogene 2008, 27, 5115-5123; Kunnimalaiyaan et al., The Oncologist 2007, 12, 535-542). This finding is not only based on observations that certain tumors display impairments in Notch activity. Additionally, various successful demonstrations confirmed that the artificial activation of Notch signaling has a beneficial impact on according malignant degenerations (Jaskula-Sztul et al, J. Surg. Res. 2011, 171, 23-27; Yu et al., Cancer 2013, 119, 774-781; Ye et al., Sci. Rep. 2016, 6, 26510). Prominent examples comprise non-melanoma skin cancer, neuroendocrine tumors and certain cancers of the hematopoietic system.

In a broader sense, due to the central role of this pathway, the potential use of Notch enhancers is not only limited to the treatment of cancer, but likewise expected to be beneficial in other pathologic conditions that have been shown to be responsive to Notch induction, such as diseases of the skin, muscle or immune system.

To this end, it is highly desirable to develop therapeutic agents that enhance Notch signaling.

Notch Enhancers State of the Art

Current methods to enhance Notch signaling for a potential therapeutic use entail the application of receptor-activating peptides or of small molecules that show Notch-augmenting properties. However, no approved Notch enhancer is available yet in the clinics. Besides, only a small number of according agents is known to date and much less have so far entered a drug development program. Reported small molecule Notch enhancers comprise resveratrol (Pinchot et al., Cancer 2011, 117, 1386-1398; Truong et al., Ann. Surg. Oncol. 2011, 18, 1506-1511; Yu et al., Mol. Cancer Ther. 2013, 12, 1276-1287), valproic acid (Greenblatt et al., Oncologist 2007, 12, 942-951; Platta et al., J. Surg. Res. 2008, 148, 31-37; Mohammed et al., Oncologist 2011, 16, 835-843), hesperetin (Patel et al., Ann. Surg. Oncol. 2014, 21, 497-504), chrysin (Yu et al., Cancer 2013, 119, 774-778), phenethyl isothiocyanate (Kim et al., PLoS One 2011, 6, 10), thiocoraline (Wyche et al., Cancer Gene Ther. 2014, 21, 518-525) and N-methylhemeanthidine chloride (Ye et al., Sci. Rep. 2016, 6, 26510).

A common drawback associated with most of the mentioned compounds is the lack of potency.

Hence, it is absolutely crucial to provide novel Notch enhancers with high therapeutic efficacy.

The screening of a small library of chemical molecules in a Notch-dependent luciferase reporter assay revealed a novel compound family with Notch-augmenting properties (Reinmnller et al., 2015, EPFL Thesis 6887, published in March 2016), the content of which is herein incorporated by reference.

DESCRIPTION OF THE INVENTION

The present invention covers refined structures to the initially discovered limited set of Notch enhancer molecules. These second generation compounds have been designed and are supposed to exhibit increased potency and greater metabolic stability. Alternatively, they present specific modifications of chemical residues, which are supposed to not impair the Notch-augmenting activity, but yet provide novel molecular features that may turn out to beneficially influence pharmacological and physicochemical parameters addressed in the general drug development process.

Thus, the present invention relates to compounds as defined herein that feature Notch enhancing activity, which can be used in the treatment of pathological conditions that are responsive for Notch-regulation, such as cancer, skin diseases, muscle disorders, and immune system-related disorders such as disorders of the hematopoietic system including the hematologic system in human and veterinary medicine.

The biological activity, e.g. the antiproliferative activity of the claimed compounds can be attributed to but may not be limited to Notch signaling enhancing activity. Thus, the present invention also relates to compounds as defined herein that feature antiproliferative activity, which can be used in the treatment of benign and malignant hyperproliferative disorders in human and veterinary medicine. In particular, the present invention relates to compounds as defined herein for the treatment of immune system-related disorders such as disorders of the hematopoietic system including the hematologic system, such as malignancies of the myeloid lineage, malignant and non-malignant disorders of the skin and mucosa, such as squamous and basal cell carcinoma, actinic keratosis, and hyperproliferative disorders of the skin and mucosa, e.g. cornification disorders, malignant and non-malignant disorders of the muscle, including hyperproliferative disorders of the muscle, such as muscle hyperplasia and muscle hypertrophy, disorders of the neuroendocrine system, such as medullary thyroid cancer, and hyperproliferative disorders of the genitourinary tract, e.g. cervical cancer in human and veterinary medicine.

A first aspect of the present invention relates to compounds of formula I and salts and solvates thereof:

wherein X is CH or N,

R1=C1-C12 preferably C1-C6 alkyl, C2-C12 preferably C2-C6 alkenyl, C2-C12 preferably C2-C6 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, C4-C12 bicycloalkyl, C6-C12 bicycloalkenyl, C5-C14 tricycloalkyl,

wherein all alkyl, alkenyl and alkynyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein all cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein all alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues can be perhalogenated, particularly perfluorinated;

and wherein R1 is preferably selected from methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, iso-propyl, tert-butyl, tert-pentyl, 3-pentyl, —CF3, —CF2CF3, —(CF2)2CF3, —(CF2)3CF3, —CH(CF3)2, —CF(CF3)2, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, bicyclo[2.2.2]octyl, adamantyl, and 9-methylbicyclo[3.3.1]nonyl;

R2=H, C1-C6 alkyl, C3-C6 cycloalkyl,

wherein all alkyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein all cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein all alkyl and cycloalkyl residues can be perhalogenated, particularly perfluorinated;

and wherein R2 is preferably selected from H, methyl and ethyl.

In some embodiments, the following compounds shown in Table Ia are explicitly excluded from the scope of the invention:

TABLE Ia Compound R1 R2 X I-A tert-butyl H CH I-B tert-butyl ethyl CH I-C tert-pentyl H CH I-D tert-pentyl ethyl CH I-E cyclo-hexyl H CH I-F cyclo-hexyl ethyl CH I-G adamant-1-yl H CH I-H adamant-1-yl ethyl CH I-I methyl H N I-J methyl ethyl N I-K tert-butyl H N I-L tert-butyl ethyl N I-M tert-pentyl H N I-N tert-pentyl ethyl N I-O cyclo-hexyl H N I-P cyclo-hexyl ethyl N I-Q isopropyl H CH I-R phenyl H CH I-S methyl H CH I-T tert-butyl methyl N I-U methyl methyl N I-V methyl methyl CH I-W methyl ethyl CH I-X n-hexyl H CH I-Y n-octyl H CH I-Z n-dodecyl H CH I-AA iso-propyl H N

Compounds I-A to I-T of Table Ia are known in the art for certain applications in the field of medicine whereas to the best of the inventor's knowledge, compounds I-U to I-AA are not known for any use in medicine. Thus, the invention encompasses any medical use for compounds I-U to I-AA.

Specific examples of compounds falling under the scope of formula I are shown in Table Ib. The compounds in Table Ib are defined by their chemical structure, the indicated nomenclature is only for illustrative purposes.

TABLE Ib X = CH, R2 = H 001 002 003 004 005 006 007 008 009 010 011 012 013 014 015 016 017 018 019 020 021 022 X = CH, R2 = Me 023 024 025 026 027 028 029 030 031 032 033 034 035 036 037 038 039 040 041 042 043 044 045 046 047 X = CH, R2 = Et 048 049 050 051 052 053 054 055 056 057 058 059 060 061 062 063 064 065 066 067 068 069 X = N, R2 = H 070 071 072 073 074 075 076 077 078 079 080 081 082 083 084 085 086 087 088 089 090 X = N, R2 = Me 091 092 093 094 095 096 097 099 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 X = N, R2 = Et 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137

Also included are isomers, e.g. enantiomers or diastereomers or mixtures of isomers, salts, particularly pharmaceutically acceptable salts, and solvates of the compounds listed above.

A second aspect of the present invention relates to compounds of formula II and salts and solvates thereof:

wherein X and R1 are defined as in formula I, including the preferred definition of R1,

R3=H, C1-C6 alkyl, or C3-C6 cycloalkyl,

wherein all alkyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein all cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein all alkyl and cycloalkyl residues can be perhalogenated, particularly perfluorinated;

and wherein R3 is preferably H or methyl;

R4=H, C1-C6 alkyl, C3-C6 cycloalkyl, OH or OC1-C6 alkyl,

wherein all alkyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein all cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein all alkyl and cycloalkyl residues can be perhalogenated, particularly perfluorinated;

and wherein R4 is preferably H, OH or methyl.

In especially preferred embodiments, R3 and R4 are in each case H; H and OH; H and —CH3; or in each case —CH3.

In some embodiments, the following compounds shown in Table IIa are explicitly excluded from the scope of the invention:

TABLE IIa Compound R1 R3 R4 X II-A tert-butyl H H N II-B methyl H methyl CH II-C methyl methyl methyl CH

Compound II-A and II-B of Table IIa are known in the art for certain applications in the field of medicine whereas to the best of the inventor's knowledge, compound II-C is not known for any use in medicine. Thus, the invention encompasses any medical use for compound II-C.

Specific examples of compounds falling under the scope of formula II are shown in Table IIb. The compounds in Table IIb are defined by their chemical structure, the indicated nomenclature is only for illustrative purposes.

TABLE IIb X = CH, R3 = H, R4 = H 138 4-(p-tolyloxy)benzamide 139 4-(4-ethylphenoxy)benzamide 140 4-(4-propylphenoxy)benzamide 141 4-(4-butylphenoxy)benzamide 142 4-(4-pentylphenoxy)benzamide 143 4-(4-hexylphenoxy)benzamide 144 4-(4-isopropylphenoxy)benzamide 145 4-(4-(tert-butyl)phenoxy)benzamide 146 4-(4-(tert-pentyl)phenoxy)benzamide 147 4-(4-(pentan-3-yl)phenoxy)benzamide 148 4-(4-(trifluoromethyl)phenoxy)benzamide 149 4-(4-(perfluoroethyl)phenoxy)benzamide 150 4-(4-(perfluoropropyl)phenoxy)benzamide 151 4-(4-(perfluorobutyl)phenoxy)benzamide 152 4-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)benzamide 153 4-(4-(perifluoropropan-2-yl)phenoxy)benzamide 154 4-(4-cyclopropylphenoxy)benzamide 155 4-(4-cyclobutylphenoxy)benzamide 156 4-(4-cyclopenlylphenoxy)benzamide 157 4-(4-cyclohexylphenoxy)benzamide 158 4-(4-cycloheptylphenoxy)benzamide 159 4-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)benzamide 160 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)benzamide 161 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)benzamide 162 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)benzamide 163 4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9-yl)phenoxy) benzamide X = CH, R3 = H, R4 = OH 164 N-hydroxy-4-(p-tolyloxy)benzamide 165 4-(4-ethylphenoxy)-N-hydroxybenzamide 166 N-hydroxy-4-(4-propylphenoxy)benzamide 167 4-(4-butylphenoxy)-N-hydroxybenzamide 168 M-hydroxy-4-(4-pentylphenoxy)benzamide 169 4-(4-hexylphenoxy)-N-hydroxybenzamide 170 N-hydroxy-4-(4-isopropylphenoxy)benzamide 171 N-hydroxy-4-(4-(tert-pentyl)phenoxy)benzamide 172 N-hydroxy-4-(4-(pentan-3-yl)phenoxy)benzamide 173 N-hydroxy-4-(4-(trifluoromethyl)phenoxy)benzamide 174 N-hydroxy-4-(4-(perfluoroethyl)phenoxy)benzamide 175 N-hydroxy-4-(4-(perfluoropropyl)phenoxy)benzamide 176 N-hydroxy-4-(4-(perfluorobutyl)phenoxy)benzamide 177 4-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N- hydroxybenzamide 178 N-hydroxy-4-(4-(perfluoropropan-2-yl)phenoxy)benzamide 179 4-(4-cyclopropylphenoxy)-N-hydroxybenzamide 180 4-(4-cyclobutyphenoxy)-N-hydroxybenzamide 181 4-(4-cyclopentylphenoxy)-N-hydroxybenzamide 182 4-(4-cyclohexylphenoxy)-N-hydroxybenzamide 183 4-(4-cycloheptylphenoxy)-N-hydroxybenzamide 184 4-(4-((1S,4F)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-N- hydroxybenzamide 185 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-N- hydroxybenzamide 186 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-N-hydroxybenzamide 187 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-N-hydroxybenzamide 188 N-hydroxy-4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)benzamide X = CH, R3 = H, R4 = Me 189 N-methyl-4-(p-tolyloxy)benzamide 190 4-(4-ethylphenoxy)-N-methylbenzamide 191 N-methyl-4-(4-propylphenoxy)benzamide 192 4-(4-butylphenoxy)-N-methylbenzamide 193 N-methyl-4-(4-pentylphenoxy)benzamide 194 4-(4-hexylphenoxy)-N-methylbenzamide 195 4-(4-isopropylphenoxy)-N-methylbenzamide 196 4-(4-(tert-butyl)phenoxy)-N-methylbenzamide 197 N-methyl-4-(4-(tert-pentyl)phenoxy)benzamide 198 N-methyl-4-(4-(pentan-3-yl)phenoxy)benzamide 199 N-methyl-4-(4-(trifluoromethyl)phenoxy)benzamide 200 N-methyl-4-(4-(perfluoroethyl)phenoxy)benzamide 201 N-methyl-4-(4-(perfluoropropyl)phenoxy)benzamide 202 N-methyl-4-(4-(perfluorobutyl)phenoxy)benzamide 203 4-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N- methylbenzamide 204 N-methyl-4-(4-(perfluoropropan-2-yl)phenoxy)benzamide 205 4-(4-cyclopropylphenoxy)-N-methylbenzamide 206 4-(4-cyclobutylphenoxy)-N-methylbenzamide 207 4-(4-cyclopenlylphenoxy)-N-methylbenzamide 208 4-(4-cyclohexylphenoxy)-N-methylbenzamide 209 4-(4-cycloheptylphenoxy)-N-methylbenzamide 210 4-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-N methylbenzamide 211 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-N- methylbenzamide 212 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-N-methylbenzamide 213 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-N-methylbenzamide 214 N-methyl-4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)benzamide X = CH, R3 = Me, R4 = Me 215 N,N-dimethyl-4-(p-tolyloxy)benzamide 216 4-(4-ethylphenoxy)-N,N-dimethylbenzamide 217 N,N-dimethyl-4-(4-propylphenoxy)benzamide 218 4-(4-butylphenoxy)-N,N-dimethylbenzamide 219 N,N-dimethyl-4-(4-pentylphenoxy)benzamide 220 4-(4-hexylphenoxy)-N,N-dimethylbenzamide 221 4-(4-isopropylphenoxy)-N,N-dimethylbenzamide 222 4-(4-(tert-butyl)phenoxy)-N,N-dimethylbenzamide 223 N,N-dimethyl-4-(4-(tert-pentyl)phenoxy)benzamide 224 N,N-dimethyl-4-(4-(pentan-3-yl)phenoxy)benzamide 225 N,N-dimethyl-4-(4-(trifluoromethyl)phenoxy)benzamide 226 N,N-dimethyl-4-(4-(perfluoroethyl)phenoxy)benzamide 227 N,N-dimethyl-4-(4-(perfluoropropyl)phenoxy)benzamide 228 N,N-dimethyl-4-(4-(perfluorobutyl)phenoxy)benzamide 229 4-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N,N- dimethylbenzamide 230 N,N-dimethyl-4-(4-(perfluoropropan-2-yl)phenoxy)benzamide 231 4-(4-cyclopropylphenoxy)-N,N-dimethylbenzamide 232 4-(4-cyclobutylphenoxy)-N,N-dimethylbenzamide 233 4-(4-cyclopentylphenoxy)-N,N-dimethylbenzamide 234 4-(4-cyclohexyphenoxy)-N,N-dimethylbenzamide 235 4-(4-cycloheptylphenoxy)-N,N-dimethylbenzamide 236 4-(4-((1S,4F)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-N,N- dimethylbenzamide 237 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-N,N- dimethylbenzamide 238 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-N,N- dimethylbenzamide 239 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-N,N- dimethylbenzamide 240 N,N-dimethyl-4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)benzamide X = N, R3 = H, R4 = H 241 6-(p-tolyloxy)nicotinamide 242 6-(4-ethylphenoxy)nicotinamide 243 6-(4-propylphenoxy)nicotinamide 244 6-(4-butylphenoxy)nicotinamide 245 6-(4-pentylphenoxy)nicotinamide 246 6-(4-hexylphenoxy)nicotinamide 247 6-(4-isopropylphenoxy)nicotinamide 248 6-(4-(tert-pentyl)phenoxy)nicotinamide 249 6-(4-(pentan-3-yl)phenoxy)nicotinamide 250 6-(4-(trifluoromethyl)phenoxy)nicotinamide 251 6-(4-(perfluoroethyl)phenoxy)nicotinamide 252 6-(4-(perfluoropropy)phenoxy)nicotinamide 253 6-(4-(perfluorobutyl)phenoxy)nicotinamide 254 6-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy) nicotinamide 255 6-(4-(perfluoropropan-2-yl)phenoxy)nicotinamide 256 6-(4-cyclopropylphenoxy)nicotinamide 257 6-(4-cyclobutylphenoxy)nicotinamide 258 6-(4-cyclopentylphenoxy)nicotinamide 259 6-(4-cyclohexylphenoxy)nicotinamide 260 6-(4-cycloheptylphenoxy)nicotinamide 261 6-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)nicotinamide 262 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)nicotinamide 263 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)nicotinamide 264 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)nicotinamide 265 6-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)nicotinamide X = N, R3 = H, R4 = OH 266 N-hydroxy-6-(p-tolyloxy)nicotinamide 267 6-(4-ethylphenoxy)-N-hydroxynicotinamide 268 N-hydroxy-6-(4-propylphenoxy)nicotinamide 269 6-(4-butylphenoxy)-N-hydroxynicotinamide 270 N-hydroxy-6-(4-pentylphenoxy)nicotinamide 271 6-(4-hexylphenoxy)-N-hydroxynicotinamide 272 N-hydroxy-6-(4-isopropylphenoxy)nicotinamide 273 N-hydroxy-6-(4-(tert-pentyl)phenoxy)nicotinamide 274 N-hydroxy-6-(4-(pentan-3-yl)phenoxy)nicotinamide 275 N-hydroxy-6-(4-(trifluoromethyl)phenoxy)nicotinamide 276 N-hydroxy-6-(4-(perfluoroethyl)phenoxy)nicotinamide 277 N-hydroxy-6-(4-(perfluoropropyl)phenoxy)nicotinamide 278 279 6-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N- hydroxynicotinamide 280 N-hydroxy-6-(4-(perfluoropropan-2-yl)phenoxy)nicotinamide 281 6-(4-cyclopropylphenoxy)-N-hydroxynicotinamide 282 6-(4-cyclobutylphenoxy)-N-hydroxynicotinamide 283 6-(4-cyclopentylphenoxy)-N-hydroxynicotinamide 284 6-(4-cyclohexylphenoxy)-N-hydroxynicotinamide 285 6-(4-cycloheptylphenoxy)-N-hydroxynicotinamide 286 6-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-N- hydroxynicotinamide 287 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-N- hydroxynicotinamide 288 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-N- hydroxynicotinamide 289 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-N- hydroxynicotinamide 290 N-hydroxy-6-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)nicotinamide X = N, R3 = H, R4 = Me 291 N-methyl-6-(p-tolyloxy)nicotinamide 292 6-(4-ethylphenoxy)-N-methylnicotinamide 293 N-methyl-6-(4-propylphenoxy)nicotinamide 294 6-(4-butylphenoxy)-N-methylnicotinamide 295 N-methyl-6-(4-pentylphenoxy)nicotinamide 296 6-(4-hexylphenoxy)-N-methylnicotinamide 297 6-(4-isopropylphenoxy)-N-methylnicotinamide 298 N-methyl-6-(4-(tert-pentyl)phenoxy)nicotinamide 299 N-methyl-6-(4-(pentan-3-yl)phenoxy)nicotinamide 300 N-methyl-6-(4-(trifluoromethyl)phenoxy)nicotinamide 301 N-methyl-6-(4-(perfluoroethyl)phenoxy)nicotinamide 302 N-methyl-6-(4-(perfluoropropyl)phenoxy)nicotinamide 303 N-methyl-6-(4-(perfluorobutyl)phenoxy)nicotinamide 304 6-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N- methylnicotinamide 305 N-methyl-6-(4-(perfluoropropan-2-yl)phenoxy) nicotinamide 306 6-(4-cyclopropylphenoxy)-N-methylnicotinamide 307 6-(4-cyclobutylphenoxy)-N-methylnicotinamide 308 6-(4-cyclopentylphenoxy)-N-methylnicotinamide 309 6-(4-cyclohexylphenoxy)-N-methylnicotinamide 310 6-(4-cycloheptylphenoxy)-N-methylnicotinamide 311 6-(4-((1S,4F)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-N- methylnicotinamide 312 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-N- methylnicotinamide 313 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-N- methylnicotinamide 314 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-N- methylnicotinamide 315 N-methyl-6-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9-yl) phenoxy)nicotinamide X = N, R3 = Me, R4 = Me 316 N,N-dimethyl-6-(p-tolyloxy)nicotinamide 317 6-(4-ethylphenoxy)-N,N-dimethylnicotinamide 318 N,N-dimethyl-6-(4-propylphenoxy)nicotinamide 319 6-(4-butylphenoxy)-N,N-dimethylnicotinamide 320 N,N-dimethyl-6-(4-pentylphenoxy)nicotinamide 321 6-(4-hexylphenoxy)-N,N-dimethylnicotinamide 322 6-(4-isopropylphenoxy)-N,N-dimethylnicotinamide 323 N,N-dimethyl-6-(4-(tert-pentyl)phenoxy)nicotinamide 324 N,N-dimethyl-6-(4-(pentan-3-yl)phenoxy)nicotinamide 325 N,N-dimethyl-6-(4-(trifluoromethyl)phenoxy)nicotinamide 326 N,N-dimethyl-6-(4-(perfluoroethyl)phenoxy)nicotinamide 327 N,N-dimethyl-6-(4-(perfluoropropyl)phenoxy)nicotinamide 328 N,N-dimethyl-6-(4-(perfluorobutyl)phenoxy)nicotinamide 329 6-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N,N- dimethylnicotinamide 330 N,N-dimethyl-6-(4-(perfluoropropan-2-yl)phenoxy) nicotinamide 331 6-(4-cyclopropylphenoxy)-N,N-dimethylnicotinamide 332 6-(4-cyclobutylphenoxy)-N,N-dimethylnicotinamide 333 6-(4-cyclopentylphenoxy)-N,N-dimethylnicotinamide 334 6-(4-cyclohexylphenoxy)-N,N-dimethylnicotinamide 335 6-(4-cycloheptylphenoxy)-N,N-dimethylnicotinamide 336 6-(4-((1S,4R)-bicyclo(2.2.1]heptan-2-yl)phenoxy)-N,N- dimethylnicotinamide 337 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-N,N- dimethylnicotinamide 338 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-N,N- dimethylnicotinamide 339 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-N,N- dimethylnicotinamide 340 N,N-dimethyl-6-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)nicotinamide

Also included are isomers, e.g. enantiomers or diastereomers or rotamers or mixtures of isomers, salts, particularly pharmaceutically acceptable salts, and solvates of the compounds listed above.

A third aspect of the present invention relates to compounds of formula III and salts and solvates thereof:

wherein X, R1 and R2 are defined as in formula I, including the preferred definitions of R1 and R2.

In some embodiments, the following compounds shown in Table IIIa are explicitly excluded from the scope of the invention:

TABLE IIIa Compound R1 R2 X III-A tert-butyl H CH III-B tert-butyl ethyl CH III-C phenyl H CH

Specific examples of compounds falling under the scope of formula III are shown in Table IIIb. The compounds in Table IIIb are defined by their chemical structure, the indicated nomenclature is only for illustrative purposes.

TABLE IIIb X = CH, R2 = H 341 3-fluoro-4-(p-tolyloxy)benzoic acid 342 4-(4-ethylphenoxy)-3-fluorobenzoic acid 343 3-fluoro-4-(4-propylphenoxy)benzoic acid 344 4-(4-butylphenoxy)-3-fluorobenzoic acid 345 3-fluoro-4-(4-pentylphenoxy)benzoic acid 346 3-fluoro-4-(4-hexylphenoxy)benzoic acid 347 3-fluoro-4-(4-isopropylphenoxy)benzoic acid 348 3-fluoro-4-(4-(tert-pentyl)phenoxy)benzoic acid 349 3-fluoro-4-(4-(pentan-3-yl)phenoxy)benzoic acid 350 3-fluoro-4-(4-(trifluoromethyl)phenoxy)benzoic acid 351 3-fluoro-4-(4-(perfluoroethyl)phenoxy)benzoic acid 352 3-fluoro-4-(4-(perfluoropropyl)phenoxy)benzoic acid 353 3-fluoro-4-(4-(perfluorobutyl)phenoxy)benzoic acid 354 3-fluoro-4-(4-(1,1,1,3,3,3hexafluoropropan-2-yl)phenoxy) benzoic acid 355 3-fluoro-4-(4-(perfluoropropan-2-yl)phenoxy)benzoic acid 356 4-(4-cyclopropylphenoxy)-3-fluorobenzoic acid 357 4-(4-cyclobutylphenoxy)-3-fluorobenzoic acid 358 4-(4-cyclopentylphenoxy)-3-fluorobenzoic acid 359 4-(4-cyclohexylphenoxy)-3-fluorobenzoic acid 360 4-(4-cycloheptylphenoxy)-3-fluorobenzoic acid 361 4-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-3- fluorobenzoic acid 362 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-3- fluorobenzoic acid 363 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-3- fluorobenzoic acid 364 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-3- fluorobenzoic acid 365 3-fluoro-4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)benzoic acid X = CH, R2 = Me 366 methyl 3-fluoro-4-(p-tolyloxy)benzoate 367 methyl 4-(4-ethylphenoxy)-3-fluorobenzoate 368 methyl 3-fluoro-4-(4-propylphenoxy)benzoate 369 methyl 4-(4-butylphenoxy)-3-fluorobenzoate 370 methyl 3-fluoro-4-(4-pentylphenoxy)benzoate 371 methyl 3-fluoro-4-(4-hexylphenoxy)benzoate 372 methyl 3-fluoro-4-(4-isopropylphenoxy)benzoate 373 methyl 4-(4-(tert-butyl)phenoxy)-3-fluorobenzoate 374 methyl 3-fluoro-4-(4-(tert-pentyl)phenoxy)benzoate 375 methyl 3-fluoro-4-(4-(pentan-3-yl)phenoxy)benzoate 376 methyl 3-fluoro-4-(4-(trifluoromethyl)phenoxy)benzoate 377 methyl 3-fluoro-4-(4-(perfluoroethyl)phenoxy)benzoate 378 methyl 3-fluoro-4-(4-(perfluoropropyl)phenoxy)benzoate 379 methyl 3-fluoro-4-(4-(perfluorobutyl)phenoxy)benzoate 380 methyl 3-fluoro-4-(4-(1,1,1,3,3,3-hexafluoropropan-2- yl)phenoxy)benzoate 381 methyl 3-fluoro-4-(4-(perfluoropropan-2-yl) phenoxy)benzoate 382 methyl 4-(4-cyclopropylphenoxy)-3-fluorobenzoate 383 methyl 4-(4-cyclobutylphenoxy)-3-fluorobenzoate 384 methyl 4-(4-cyclopentylphenoxy)-3-fluorobenzoate 385 methyl 4-(4-cyclohexylphenoxy)-3-fluorobenzoate 386 methyl 4-(4-cycloheptylphenoxy)-3-fluorobenzoate 387 methyl 4-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-3- fluorobenzoate 388 methyl 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-3- fIuorobenzoate 389 methyl 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-3- fluorobenzoate 390 methyl 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-3- fluorobenzoate 391 methyl 3-fluoro-4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9-yl) phenoxy)benzoate X = CH, R2 = Et 392 ethyl 3-fluoro-4-(p-tolyloxy)benzoate 393 ethyl 4-(4-ethylphenoxy)-3-fluorobenzoate 394 ethyl 3-fluoro-4-(4-propylphenoxy)benzoate 395 ethyl 4-(4-butylphenoxy)-3-fluorobenzoate 396 ethyl 3-fluoro-4-(4-pentylphenoxy)benzoate 397 ethyl 3-fluoro-4-(4-hexylphenoxy)benzoate 398 ethyl 3-fluoro-4-(4-isopropylphenoxy)benzoate 399 ethyl 3-fluoro-4-(4-(tert-pentyl)phenoxy)benzoate 400 ethyl 3-fluoro-4-(4-(pentan-3-yl)phenoxy)benzoate 401 ethyl 3-fIuoro-4-(4-(trifluoromethyl)phenoxy)benzoate 402 ethyl 3-fluoro-4-(4-(perfluoroethyl)phenoxy)benzoate 403 ethyl 3-fluoro-4-(4-(perfluoropropyl)phenoxy)benzoate 404 ethyl 3-fluoro-4-(4-(perfluorobutyl)phenoxy)benzoate 405 ethyl 3-fluoro-4-(4-(1,1,1,3,3,3-hexafluoropropan-2- yl)phenoxy)benzoate 406 ethyl 3-fluoro-4-(4-(perfluoropropan-2-yl)phenoxy)benzoate 407 ethyl 4-(4-cyclopropylphenoxy)-3-fluorobenzoate 408 ethyl 4-(4-cyclobutylphenoxy)-3-fluorobenzoate 409 ethyl 4-(4-cyctopentylphenoxy)-3-fluorobenzoate 410 ethyl 4-(4-cyclohexylphenoxy)-3-fluorobenzoate 411 ethyl 4-(4-cycloheptylphenoxy)-3-fluorobenzoate 412 ethyl 4-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-3- fluorobenzoate 413 ethyl 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-3- fluorobenzoate 414 ethyl 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-3-fluorobenzoate 415 ethyl 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-3-fluorobenzoate 416 ethyl 3-fluoro-4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)benzoate X = N, R2 = H 417 5-fluoro-6-(p-tolyloxy)nicotinic acid 418 6-(4-ethylphenoxy)-5-fluoronicotinic acid 419 5-fIuoro-3-(4-propylphenoxy)nicotinic acid 420 6-(4-butylphenoxy)-5-fluoronicotinic acid 421 5-fluoro-6-(4-pentylphenoxy)nicotinic acid 422 5-fluoro-6-(4-hexylphenoxy)nicotinic acid 423 5-fluoro-6-(4-isopropylphenoxy)nicotinic acid 424 6-(4-(tert-butyl)phenoxy)-5-fluoronicotinic acid 425 5-fluoro-6-(4-(tert-pentyl)phenoxy)nicotinic acid 426 5-fluoro-6-(4-(pentan-3-yl)phenoxy)nicotinic acid 427 5-fluoro-6-(4-(trifluoromethyl)phenoxy)nicotinic acid 428 5-fluoro-6-(4-(perfluoroethyl)phenoxy)nicotinic acid 429 5-fluoro-6-(4-(perfluoropropyl)phenoxy)nicotinic acid 430 5-fluoro-6-(4-(perfluorobutyl)phenoxy)nicotinic acid 431 5-fluoro-6-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy) nicotinic acid 432 5-fluoro-6-(4-(perfluoropropan-2-yl)phenoxy)nicotinic acid 433 6-(4-cyclopropylphenoxy)-5-fluoronicotinic acid 434 6-(4-cyclobutylphenoxy)-5-fluoronicotinic acid 435 6-(4-cyclopentylphenoxy)-5-fluoronicotinic acid 436 6-(4-cyclohexylphenoxy)-5-fluoronicotinic acid 437 6-(4-cycloheptylphenoxy)-5-fluoronicotinic acid 438 6-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-5- fluoronicotinic acid 439 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-5- fluoronicotinic acid 440 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-5- fluoronicotinic acid 441 6-(4-((1r,3r,5r,7r)-adamanlan-2-yl)phenoxy)-5- fluoronicotinic acid 442 5-fluoro-6-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)nicotinic acid X = N, R2 = Me 443 methyl 5-fluoro-6-(p-tolyloxy)nicotinate 444 methyl 6-(4-ethylphenoxy)-5-fluoronicotinate 445 methyl 5-fluoro-6-(4-propylphenoxy)nicotinate 446 methyl 6-(4-butylphenoxy)-5-fluoronicotinate 447 methyl 5-fluoro-6-(4-pentylphenoxy)nicotinate 448 methyl 5-fluoro-6-(4-hexylphenoxy)nicotinate 449 methyl 5-fluoro-6-(4-isopropylphenoxy)nicotinate 450 methyl 6-(4-(tert-butyl)phenoxy)-5-fluoronicotinate 451 methyl 5-fluoro-6-(4-(tert-pentyl)phenoxy)nicotinate 452 methyl 5-fluoro-6-(4-(pentan-3-yl)phenoxy)nicotinate 453 methyl 5-fluoro-6-(4-(trifluoromethyl)phenoxy)nicotinate 454 methyl 5-fluoro-6-(4-(perfluoroethyl)phenoxy)nicotinate 455 methyl 5-fluoro-6-(4-(perfluoropropyl)phenoxy)nicotinate 456 methyl 5-fluoro-6-(4-(perfluorobutyl)phenoxy)nicotinate 457 methyl 5-fluoro-6-(4-(1,1,1,3,3,3-hexafluoropropan-2- yl)phenoxy)nicotinate 458 methyl 5-fluoro-6-(4-(perfluoropropan-2-yl)phenoxy) nicotinate 459 methyl 6-(4-cyclopropylphenoxy)-5-fluoronicotinate 460 methyl 6-(4-cyclobutylphenoxy)-5-fluoronicotinate 461 methyl 6-(4-cyclopentylphenoxy)-5-fluoronicotinate 462 methyl 6-(4-cyclohexylphenoxy)-5-fluoronicotinate 463 methyl 6-(4-cycloheptylphenoxy)-5-fluoronicotinate 464 methyl 6-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)- 5-fluoronicotinate 465 methyl 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)- 5-fluoronicotinate 466 methyl 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-5- fluoronicotinate 467 methyl 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-5- fIuoronicotinate 468 methyl 5-fluoro-6-(4-((1R,5S)-9-methylbicyclo[3,3.1] nonan-9-yl)phenoxy)nicotinate X = N, R2 = Et 469 ethyl 5-fluoro-6-(p-tolyloxy)nicotinate 470 ethyl 6-(4-ethylphenoxy)-5-fluoronicotinate 471 ethyl 5-fluoro-6-(4-propylphenoxy)nicotinate 472 ethyl 6-(4-butylphenoxy)-5-fluoronicotinate 473 ethyl 5-fluoro-6-(4-pentylphenoxy)nicotinate 474 ethyl 5-fluoro-6-(4-hexylphenoxy)nicotinate 475 ethyl 5-fluoro-6-(4-isopropylphenoxy)nicotinate 476 ethyl 6-(4-(tert-butyl)phenoxy)-5-fluoronicotinate 477 ethyl 5-fluoro-6-(4-(tert-pentyl)phenoxy)nicotinate 478 ethyl 5-fluoro-6-(4-(pentan-3-yl)phenoxy)nicotinate 479 ethyl 5-fluoro-6-(4-(trifluoromethyl)phenoxy)nicotinate 480 ethyl 5-fluoro-6-(4-(perfluoroethyl)phenoxy)nicotinate 481 ethyl 5-fluoro-6-(4-(perfluoropropyl)phenoxy)nicotinate 482 ethyl 5-fluoro-6-(4-(perfluorobutyl)phenoxy)nicotinate 483 ethyl 5-fluoro-6-(4-(1,1,1,3,3,3-hexafluoropropan-2- yl)phenoxy)nicotinate 484 ethyl 5-fluoro-6-(4-(perfluoropropan-2-yl) phenoxy)nicotinate 485 ethyl 6-(4-cyclopropylphenoxy)-5-fluoronicotinate 486 ethyl 6-(4-cyclobutylphenoxy)-5-fluoronicotinate 487 ethyl 6-(4-cyclopentylphenoxy)-5-fluoronicotinate 488 ethyl 6-(4-cyclohexylphenoxy)-5-fluoronicotinate 489 ethyl 6-(4-cycloheptylphenoxy)-5-fluoronicotinate 490 ethyl 6-(4-((1S,4R)-bicyclo[2,2,1]heptan-2-yl)phenoxy)-5- fluoronicotinate 491 ethyl 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-6- fluoronicotinate 492 ethyl 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-5- fluoronicotinate 493 ethyl 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-5- fluoronicotinate 494 ethyl 5-fluoro-6-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan- 9-yl)phenoxy)nicotinate

Also included are isomers, e.g. enantiomers or diastereomers or mixtures of isomers, salts, particularly pharmaceutically acceptable salts, and solvates of the compounds listed above.

A fourth aspect of the present invention relates to compounds of formula IV and salts and solvates thereof:

wherein X and R1 are defined as in formula I, including the preferred definition of R1,

and R3 and R4 are defined as in formula II, including the preferred definitions of R3 and R4.

Specific examples of compounds falling under the scope of formula IV are shown in Table IV. The compounds in Table IV are defined by their chemical structure, the indicated nomenclature is only for illustrative purposes.

TABLE IV X = CH, R3 = H, R4 = H 495 3-fluoro-4-(p-tolyloxy)benzamide 496 4-(4-ethylphenoxy)-3-fluorobenzamide 497 3-fluoro-4-(4-propylphenoxy)benzamide 498 4-(4-butylphenoxy)-3-fluorobenzamide 499 3-fluoro-4-(4-pentylphenoxy)benzamide 500 3-fluoro-4-(4-hexylphenoxy)benzamide 501 3-fluoro-4-(4-isopropylphenoxy)benzamide 502 4-(4-(tert-butyl)phenoxy)-3-fluorobenzamide 503 3-fluoro-4-(4-(tert-pentyl)phenoxy)benzamide 504 3-fluoro-4-(4-(pentan-3-yl)phenoxy)benzamide 505 3-fluoro-4-(4-(trifluoromethyl)phenoxy)benzamide 506 3-fluoro-4-(4-(perfluoroethyl)phenoxy)benzamide 507 3-fluoro-4-(4-(perfluoropropyl)phenoxy)benzamide 508 3-fluoro-4-(4-(perfluorobutyl)phenoxy)benzamide 509 3-fluoro-4-(4-(1,1,1,3,3,3-hexafluoropropan-2- yl)phenoxy)benzamide 510 3-fluoro-4-(4-(perfluoropropan-2-yl)phenoxy)benzamide 511 4-(4-cyclopropylphenoxy)-3-fluorobenzamide 512 4-(4-cyclobutylphenoxy)-3-fluorobenzamide 513 4-(4-cyclopenlylphenoxy)-3-fluorobenzamide 514 4-(4-cyclohexylphenoxy)-3-fluorobenzamide 515 4-(4-cycloheptylphenoxy)-3-fluorobenzamide 516 4-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-3- fluorobenzamide 517 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-3- fluorobenzamide 518 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-3-fluorobenzamide 519 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-3-fluorobenzamide 520 3-fluoro-4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)benzamide X = CH, R3 = H, R4 = OH 521 3-fluoro-N-hydroxy-4-(p-tolyloxy)benzamide 522 4-(4-ethylphenoxy)-3-fluoro-N-hydroxybenzamide 523 3-fluoro-N-hydroxy-4-(4-propylphenoxy)benzamide 524 4-(4-butylphenoxy)-3-fluoro-N-hydroxybenzamide 525 3-fluoro-N-hydroxy-4-(4-pentylphenoxy)benzamide 526 3-fluoro-4-(4-hexylphenoxy)-N-hydroxybenzamide 527 3-fluoro-N-hydroxy-4-(4-isopropylphenoxy)benzamide 528 4-(4-(tert-butyl)phenoxy)-3-fluoro-N-hydroxybenzamide 529 3-fluoro-N-hydroxy-4-(4-(tert-pentyl)phenoxy)benzamide 530 3-fluoro-N-hydroxy-4-(4-(pentan-3-yl)phenoxy)benzamide 531 3-fluoro-N-hydroxy-4-(4-(trifluoromethyl)phenoxy)benzamide 532 3-fluoro-N-hydroxy-4-(4-(perfluoroethyl)phenoxy)benzamide 533 3-fluoro-N-hydroxy-4-(4-(perfluoropropyl)phenoxy)benzamide 534 3-fluoro-N-hydroxy-4-(4-(perfluorobutyl)phenoxy)benzamide 535 3-fluoro-4-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N- hydroxybenzamide 536 3-fluoro-N-hydroxy-4-(4-(perfluoropropan-2-yl)phenoxy) benzamide 537 4-(4-cyclopropylphenoxy)-3-fluoro-N-hydroxybenzamide 538 4-(4-cyclobutylphenoxy)-3-fluoro-N-hydroxybenzamide 539 4-(4-cyclopentylphenoxy)-3-fluoro-N-hydroxybenzamide 540 4-(4-cyclohexylphenoxy)-3-fluoro-N-hydroxybenzamide 541 4-(4-cycloheptylphenoxy)-3-fluoro-N-hydroxybenzamide 542 4-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-3-fluoro- N-hydroxybenzamide 543 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-3-fluoro- N-hydroxybenzamide 544 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-3-fluoro- N-hydroxybenzamide 545 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-3-fluoro- N-hydroxybenzamide 546 3-fluoro-N-hydroxy-4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan- 9-yl)phenoxy)benzamide X = CH, R3 = H, R4 = Me 547 3-fluoro-N-methyl-4-(p-tolyloxy)benzamide 548 4-(4-ethylphenoxy)-3-fluoro-N-methylbenzamide 549 3-fluoro-N-methyl-4-(4-propylphenoxy)benzamide 550 4-(4-butylphenoxy)-3-fluoro-N-methylbenzamide 551 3-fluoro-N-methyl-4-(4-pentylphenoxy)benzamide 552 3-fluoro-4-(4-hexylphenoxy)-N-methylbenzamide 553 3-fluoro-4-(4-isopropylphenoxy)-N-methylbenzamide 554 4-(4-(tert-butyl)phenoxy)-3-fluoro-N-methylbenzamide 555 3-fluoro-N-methyl-4-(4-(tert-pentyl)phenoxy)benzamide 556 3-fluoro-N-methyl-4-(4-(pentan-3-yl)phenoxy)benzamide 557 3-fluoro-N-methyl-4-(4-(trifluoromethyl)phenoxy) benzamide 558 3-fluoro-N-methyl-4-(4-(perfluoropropyl)phenoxy)benzamide 559 3-fluoro-N-methyl-4-(4-(perfluoroethyl)phenoxy)benzamide 560 3-fluoro-N-methyl-4-(4-(perfluorobutyl)phenoxy)benzamide 561 3-fluoro-4-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N- methylbenzamide 562 3-fluoro-N-methyl-4-(4-(perfluoropropan-2-yl)phenoxy) benzamide 563 4-(4-cyclopropylphenoxy)-3-fluoro-N-methylbenzamide 564 4-(4-cyclobutylphenoxy)-3-fluoro-N-methylbenzamide 565 4-(4-cyclopentylphenoxy)-3-fluoro-N-methylbenzamide 566 4-(4-cyclohexylphenoxy)-3-fluoro-N-methylbenzamide 567 4-(4-cycloheptylphenoxy)-3-fluoro-N-methylbenzamide 568 4-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-3-fluoro-N methylbenzamide 569 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-3-fluoro-N- methylbenzamide 570 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-3-fluoro-N- methylbenzamide 571 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-3-fluoro-N- methylbenzamide 572 3-fluoro-N-methyl-4-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan- 9-yl)phenoxy)benzamide X = CH, R3 = Me, R4 = Me 573 3-fluoro-N,N-dimethyl-4-(p-tolyloxy)benzamide 574 4-(4-ethylphenoxy)-3-fluoro-N,N-dimethylbenzamide 575 3-fluoro-N,N-dimethyl-4-(4-propylphenoxy)benzamide 576 4-(4-butylphenoxy)-3-fluoro-N,N-dimethylbenzamide 577 3-fluoro-N,N-dimethyl-4-(4-pentylphenoxy)benzamide 578 3-fluoro-4-(4-hexylphenoxy)-N,N-dimelhylbenzamide 579 3-fluoro-4-(4-isopropylphenoxy)-N,N-dimethylbenzamide 580 4-(4-(tert-butyl)phenoxy)-3-fluoro-N,N-dimethytbenzamide 581 3-fluoro-N,N-dimethyl-4-(4-(tert-pentyl)phenoxy)benzamide 582 3-fluoro-N,N-dimethyl-4-(4-(pentan-3-yl)phenoxy)benzamide 583 3-fluoro-N,N-dimethyl-4-(4-(trifluoromethyl)phenoxy)benzamide 584 3-fluoro-N,N-dimethyl-4-(4-(perfluoroethyl)phenoxy)benzamide 585 3-fluoro-N,N-dimethyl-4-(4-(perfluoropropyl)phenoxy)benzamide 586 3-fluoro-N,N-dimethyl-4-(4-(perfluorobutyl)phenoxy)benzamide 587 3-fluoro-4-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)- N,N-dimethylbenzamide 588 3-fluoro-N,N-dimethyl-4-(4-(perfluoropropan-2- yl)phenoxy)benzamide 589 4-(4-cyclopropylphenoxy)-3-fluoro-N,N-dimethylbenzamide 590 4-(4-cyclobutylphenoxy)-3-fluoro-N,N-dimethylbenzamide 591 4-(4-cyclopentylphenoxy)-3-fluoro-N,N-dimethylbenzamide 592 4-(4-cyclohexylphenoxy)-3-fluoro-N,N-dimethylbenzamide 593 4-(4-cycloheptylphenoxy)-3-fluoro-N,N-dimethylbenzamide 594 4-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-3-fluoro-N,N- dimethylbenzamide 595 4-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-3-fluoro-N,N- dimethylbenzamide 596 4-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-3-fluoro-N,N- dimethylbenzamide 597 4-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-3-fluoro-N,N- dimethylbenzamide 598 3-fluoro-N,N-dimethyl-4-(4-((1R,5S)-9-methylbicyclo[3.3.l]nonan- 9-yl)phenoxy)benzamide X = N, R3 = H, R4 = H 599 5-fluoro-6-(p-tolyloxy)nicotinamide 600 6-(4-ethylphenoxy)-5-fluoronicotinamide 601 5-fluoro-6-(4-propylphenoxy)nicotinamide 602 6-(4-butylphenoxy)-5-fluoronicotinamide 603 5-fluoro-6-(4-pentylphenoxy)nicotinamide 604 5-fluoro-6-(4-hexylphenoxy)nicotinamide 605 5-fluoro-6-(4-isopropylphenoxy)nicotinamide 606 6-(4-(tert-butyl)phenoxy)-5-fluoronicotinamide 607 5-fluoro-6-(4-(tert-pentyl)phenoxy)nicotinamide 608 5-fluoro-6-(4-(pentan-3-yl)phenoxy)nicotinamide 609 5-fluoro-6-(4-(trifluoromethyl)phenoxy)nicotinamide 610 5-fluoro-6-(4-(perfluoroethyl)phenoxy)nicotinamide 611 5-fluoro-6-(4-(perfluoropropyl)phenoxy)nicotinamide 612 5-fluoro-6-(4-(perfluorobutyl)phenoxy)nicotinamide 613 5-fluoro-6-(4-(1,1,1,3,3,3-hexafluoropropan-2- yl)phenoxy)nicotinamide 614 5-fluoro-6-(4-(perfluoropropan-2-yl)phenoxy)nicotinamide 615 6-(4-cyclopropylphenoxy)-5-fluoronicotinamide 616 6-(4-cyclobutylphenoxy)-5-fluoronicotinamide 617 6-(4-cyclopentylphenoxy)-5-fluoronicotinamide 618 6-(4-cyclohexylphenoxy)-5-fluoronicotinamide 619 6-(4-cycloheptylphenoxy)-5-fluoronicotinamide 620 6-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-5- fluoronicotinamide 621 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-5- fluoronicotinamide 622 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-5-fluoronicotinamide 623 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-5-fluoronicotinamide 624 5-fluoro-6-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)nicotinamide X = N, R3 = H, R4 = OH 625 5-fluoro-N-hydroxy-6-(p-tolyloxy)nicotinamide 626 6-(4-ethylphenoxy)-5-fluoro-N-hydroxynicotinamide 627 5-fluoro-N-hydroxy-6-(4-propylphenoxy)nicotinamide 628 6-(4-butylphenoxy)-5-fluoro-N-hydroxynicotinamide 629 5-fluoro-N-hydroxy-6-(4-pentylphenoxy)nicotinamide 630 5-fluoro-6-(4-hexylphenoxy)-N-hydroxynicotinamide 631 5-fluoro-N-hydroxy-6-(4-isopropylphenoxy)nicotinamide 632 6-(4-(tert-butyl)phenoxy)-5-fluoro-N-hydroxynicotinamide 633 634 5-fluoro-N-hydroxy-6-(4-(pentan-3-yl)phenoxy)nicotinamide 635 5-fluoro-N-hydroxy-6-(4-(trifluoromethyl)phenoxy)nicotinamide 636 5-fluoro-N-hydroxy-6-(4-(perfluoroethyl)phenoxy)nicotinamide 637 5-fluoro-N-hydroxy-6-(4-(perfluoropropyl)phenoxy)nicotinamide 638 5-fluoro-N-hydroxy-6-(4-(perfluorobutyl)phenoxy)nicotinamide 639 5-fluoro-6-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N- hydroxynicotinamide 640 5-fluoro-N-hydroxy-6-(4-(perfluoropropan-2- yl)phenoxy)nicotinamide 641 6-(4-cyclopropylphenoxy)-5-fluoro-N-hydroxynicotinamide 642 6-(4-cyclobutylphenoxy)-5-fluoro-N-hydroxynicotinamide 643 6-(4-cyclopentylphenoxy)-5-fluoro-N-hydroxynicotinamide 644 6-(4-cyclohexylphenoxy)-5-fluoro-N-hydroxynicotinamide 645 6-(4-cycloheptylphenoxy)-5-fluora-N-hydroxynicotinamide 646 6-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-5-fluoro- N-hydroxynicotinamide 647 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-5-fluoro- N-hydroxynicotinamide 648 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-5-fluoro- N-hydroxynicotinamide 649 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-5-fluoro- N-hydroxynicotinamide 650 5-fluoro-N-hydroxy-6-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan- 9-yl)phenoxy)nicotinamide X = N, R3 = H, R4 = Me 651 5-fluoro-N-methyl-6-(p-tolyloxy)nicotinamide 652 6-(4-ethylphenoxy)-5-fluoro-N-methylnicotinamide 653 5-fluoro-N-methyl-6-(4-propylphenoxy)nicotinamide 654 6-(4-butylphenoxy)-5-fluoro-N-methylnicotinamide 655 5-fluoro-N-methyl-6-(4-pentylphenoxy)nicotinamide 656 5-fluoro-6-(4-hexylphenoxy)-N-methylnicotinamide 657 5-fluoro-6-(4-isopropylphenoxy)-N-methylnicotinamide 658 6-(4-(tert-butyl)phenoxy)-5-fluoro-N-methylnicotinamide 659 5-fluoro-N-methyl-6-(4-(tert-pentyl)phenoxy)nicotinamide 660 5-fluoro-N-methyl-6-(4-(pentan-3-yl)phenoxy)nicotinamide 661 5-fluoro-N-methyl-6-(4-(trifluoromethyl)phenoxy)nicotinamide 662 5-fluoro-N-methyl-6-(4-(perfluoroethyl)phenoxy)nicotinamide 663 5-fluoro-N-methyl-6-(4-(perfluoropropyl)phenoxy)nicotinamide 664 5-fluoro-N-methyl-6-(4-(perfluorobutyl)phenoxy)nicotinamide 665 5-fluoro-6-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)-N- methylnicotinamide 666 5-fluoro-N-methyl-6-(4-(perfluoropropan-2-yl)phenoxy) nicotinamide 667 6-(4-cyclopropylphenoxy)-5-fluoro-N-methylnicotinamide 668 6-(4-cyclobutylphenoxy)-5-fluoro-N-methy)nicotinamide 669 6-(4-cyclopentylphenoxy)-5-fluoro-N-methylnicotinamide 670 6-(4-cyclohexylphenoxy)-5-fluoro-N-methylnicotinamide 671 6-(4-cycloheptylphenoxy)-5-fluoro-N-methylnicotinamide 672 6-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-5-fluoro-N- methylnicotinamide 673 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-5-fluoro-N- methylnicotinamide 674 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-5-fluoro-N- methylnicotinamide 675 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-5-fluoro-N- methylnicotinamide 676 5-fluoro-N-methyl-6-(4-((1R,5S)-9-methylbicyclo[3.3.1]nonan-9- yl)phenoxy)nicotinamide X = N, R3 = Me, R4 = Me 677 5-fluoro-N,N-dimethyl-6-(p-tolyloxy)nicotinamide 678 6-(4-ethylphenoxy)-5-fluoro-N,N-dimethylnicotinamide 679 5-fluoro-N,N-dimethyl-6-(4-propylphenoxy)nicotinamide 680 6-(4-butylphenoxy)-5-fluoro-N,N-dimethylnicotinamide 681 5-fluoro-N,N-dimethyl-6-(4-pentylphenoxy)nicotinamide 682 5-fluoro-6-(4-hexylphenoxy)-N,N-dimethylnicotinamide 683 5-fluoro-6-(4-isopropylphenoxy)-N-dimethylnicotinamide 684 6-(4-(tert-bulyl)phenoxy)-5-fluoro-N,N-dimethylnicotinamide 685 5-fluoro-N,N-dimethyl-6-(4-(tert-pentyl)phenoxy)nicotinamide 686 5-fluoro-N,N-dimethyl-6-(4-(pentan-3-yl)phenoxy)nicotinamide 687 5-fluoro-N,N-dimethyl-6-(4-(trifluoromethyl)phenoxy) nicotinamide 688 5-fluoro-N,N-dimethyl-6-(4-(perfluoroethyl)phenoxy) nicotinamide 689 5-fluoro-N,N-dimethyl-6-(4-(perfluoropropyl)phenoxy) nicotinamide 690 5-fluoro-N,N-dimethyl-6-(4-(perfluorobutyl)phenoxy) nicotinamide 691 5-fluoro-6-(4-(1,1,1,3,3,3-hexafluoropropan-2-yl)phenoxy)- N,N-dimethylnicotinamide 692 5-fluoro-N,N-dimethyl-6-(4-(perfluoropropan-2- yl)phenoxy)nicotinamide 693 6-(4-cyclopropylphenoxy)-5-fluoro-N,N-dimethylnicotinamide 694 6-(4-cyclobutylphenoxy)-5-fluoro-N,N-dimethylnicotinamide 695 6-(4-cyclopentylphenoxy)-5-fluoro-N,N-dimethylnicotinamide 696 6-(4-cyclohexylphenoxy)-5-fluoro-N,N-dimethylnicotinamide 697 6-(4-cycloheptylphenoxy)-5-fluoro-N,N-dimethylnicotinamide 698 6-(4-((1S,4R)-bicyclo[2.2.1]heptan-2-yl)phenoxy)-5-fluoro-N,N- dimethylnicotinamide 399 6-(4-((1s,4s)-bicyclo[2.2.2]octan-2-yl)phenoxy)-5-fluoro-N,N- dimethylnicotinamide 700 6-(4-((3r,5r,7r)-adamantan-1-yl)phenoxy)-5-fluoro-N,N- dimethylnicotinamide 701 6-(4-((1r,3r,5r,7r)-adamantan-2-yl)phenoxy)-5-fluoro-N,N- dimethylnicotinamide 702 5-fluoro-N,N-dimethyl-6-(4-((1R,5S)-9-methylbicyclo[3.3.1] nonan-9-yl)phenoxy)nicotinamide

Also included are isomers, e.g. enantiomers or diastereomers or rotamers or mixtures of isomers, salts, particularly pharmaceutically acceptable salts, and solvates of the compounds listed above.

A fifth aspect of the present invention relates to compounds of formula V and salts and solvates thereof:

wherein n 0-5, which comprises cyclopropyl (n=0), cyclobutyl (n=1), cyclopentyl (n=2), cyclohexyl (n=3), cycloheptyl (n=4) and cyclooctyl (n=5),

wherein the said cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl groups can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein the said cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl groups can be perhalogenated, particularly perfluorinated;

and wherein n is preferably 0 as constituting cyclopropyl, particularly as constituting cyclopropyl being unsubstituted;

R5=C1-C12 preferably C1-C6 alkyl, C2-C12 preferably C2-C6 alkenyl, C2-C12 preferably C2-C6 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl,

wherein all alkyl, alkenyl and alkynyl residues can be linear or branched, and are perhalogenated, particularly perfluorinated,

and wherein all cycloalkyl and cycloalkenyl residues are perhalogenated, particularly perfluorinated;

or wherein all alkyl, alkenyl and alkynyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

and wherein all cycloalkyl and cycloalkenyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein R5 is preferably —CF3 or —CF2CF3;

R6-R9 are independently from each other selected from —H, —F, —Cl, —Br, —I, linear or branched C1-C4 alkyl, linear or branched C2-C4 alkenyl, C2-C4 alkynyl, C3-C5 cycloalkyl, and wherein all alkyl, alkenyl, alkynyl and cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

wherein R6-R8 each are preferably H, and R9 is preferably —H, —F, —Cl, or —CH3;

Y=a six-membered aromatic ring selected from benzene, pyridine, pyrimidine, pyridazine or pyrazine;

wherein the benzene ring is not substituted, or it is substituted with one to four of the substituents independently selected from R10-R13,

and wherein the pyridine ring is not substituted, or it is substituted at the carbon positions with one to three of the substituents independently selected from R10-R12, and wherein preferably the N-atom of the pyridine ring is in ortho-position relative to the ether bond,

and wherein the pyrimidine ring is not substituted, or it is substituted at the carbon positions with one or two of the substituents independently selected from R10-R11, and wherein preferably an N-atom of the pyrimidine ring is in ortho-position relative to the ether bond,

and wherein the pyridazine ring is not substituted, or it is substituted at the carbon positions with one or two of the substituents independently selected from R10-R11, and wherein preferably an N-atom of the pyridazine ring is in ortho-position relative to the ether bond,

and wherein the pyrazine ring is not substituted, or it is substituted at the carbon positions with one or two of the substituents independently selected from R10-R11, and wherein preferably an N-atom of the pyrazine ring is in ortho-position relative to the ether bond,

wherein preferably Y=benzene or pyridine being not substituted with any of the residues selected from R10-R13, or being substituted with one of the substituents selected from R10-R13 being F at the carbon atom in ortho-position relative to the ether bond;

R10-R13 are independently from each other selected from —F, —Cl, —Br, —I, linear or branched C1-C4 alkyl, linear or branched C2-C4 alkenyl, C2-C4 alkynyl, C3-C5 cycloalkyl, and wherein all alkyl, alkenyl, alkynyl and cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;

Z=O or S, and preferably Z=O;

R14=OR2 or NR3R4

wherein R2 is defined as in formula I including the preferred definition of R2 as H, methyl or ethyl;

wherein R3 and R4 are defined as in formula II, including the preferred definitions of R3 as H or —CH3 and R4 as H, OH or —CH3;

In a particularly preferred embodiment of the compounds of formula V, the present invention relates to compounds of formula Va and salts and solvates thereof:

wherein n is defined as in formula V, including the preferred definition of n being n=0 as constituting cyclopropyl, particularly as constituting cyclopropyl being unsubstituted,

wherein Z is defined as in formula V, including the preferred definition of Z as Z=O,

wherein R5 is defined as in formula V, including all preferred definitions of R5,

R6-R9 are defined as in formula V, including all preferred definitions of R6-R9,

wherein R14 is defined as in formula V,

wherein X is N or CR13,

and wherein R10-R13 are independently from each other selected from —H, —F, —Cl, —Br, —I, linear or branched C1-C4 alkyl, linear or branched C2-C4 alkenyl, C2-C4 alkynyl, C3-C5 cycloalkyl, and wherein all alkyl, alkenyl, alkynyl and cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from —F, —Cl, —Br, —I; and C1-C3alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated.

Specific examples of compounds falling under the scope of formula V are shown in Table V. The compounds in Table V are defined by their chemical structure, the indicated nomenclature is only for illustrative purposes.

TABLE V 703 4-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy) benzoic acid 704 4-(4-(1-(perfluoroethyl)cyclopropyl)phenoxy) benzoic acid 705 3-fluoro-4-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy) benzoic acid 706 3-fluoro-4-(4-(1-(perfluoroethyl)cyclopropyl)phenoxy) benzoic acid 707 4-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl)phenoxy) benzoic acid 708 4-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl)phenoxy) benzoic acid 709 4-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy) benzoic acid 710 4-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl)phenoxy) benzoic acid 711 4-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy) benzoic acid 712 methyl 4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 713 methyl 4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzoate 714 methyl 3-fluoro-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 715 methyl 3-fluoro-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzoate 716 methyl 4-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 717 methyl 4-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzoate 718 methyl 4-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 719 methyl 4-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzoate 720 methyl 4-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 721 ethyl 4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 722 ethyl 4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzoate 723 ethyl 3-fluoro-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 724 ethyl 3-fluoro-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzoate 725 ethyl 4-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 726 ethyl 4-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzoate 727 ethyl 4-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 728 ethyl 4-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzoate 729 ethyl 4-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzoate 730 6-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy) nicotinic acid 731 6-(4-(1-(perfluoroethyl)cyclopropyl)phenoxy) nicotinic acid 732 5-fluoro-6-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy) nicotinic acid 733 5-fluoro-6-(4-(1-(perfluoroethyl)cyclopropyl)phenoxy) nicotinic acid 734 6-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl)phenoxy) nicotinic acid 735 6-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl)phenoxy) nicotinic acid 736 6-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy) nicotinic acid 737 6-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl)phenoxy) nicotinic acid 738 6-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy) nicotinic acid 739 methyl 6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 740 methyl 6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinate 741 methyl 5-fluoro-6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 742 methyl 5-fluoro-6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinate 743 methyl 6-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 744 methyl 6-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinate 745 methyl 6-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 746 methyl 6-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinate 747 methyl 6-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 748 ethyl 6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 749 ethyl 6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinate 750 ethyl 5-fluoro-6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 751 ethyl 5-fluoro-6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinate 752 ethyl 6-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 753 ethyl 6-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinate 754 ethyl 6-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 755 ethyl 6-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinate 756 ethyl 6-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinate 757 4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 758 4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 759 3-fluoro-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 760 3-fluoro-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 761 4-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 762 4-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 763 4-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 764 4-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 765 4-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 766 N-hydroxy-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 767 N-hydroxy-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 768 3-fluoro-N-hydroxy-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 769 3-fluoro-N-hydroxy-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 770 N-hydroxy-4-(2-methyl-4-(1-(trifluoromethy)cyclopropyl) phenoxy)benzamide 771 N-hydroxy-4-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 772 4-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N-hydroxybenzamide 773 4-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl)phenoxy)- N-hydroxybenzamide 774 4-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N-hydroxybenzamide 775 N-methyl-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 776 N-methyl-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 777 3-fluoro-N-methyl-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 778 3-fluoro-N-methyl-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 779 N-methyl-4-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 780 N-methyl-4-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 781 4-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N-methylbenzamide 782 4-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl)phenoxy)- N-methylbenzamide 783 4-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N-methylbenzamide 784 N,N-dimethyl-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 785 N,N-dimethyl-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 786 3-fluoro-N,N-dimethyl-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 787 3-fluoro-N,N-dimethyl-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 788 N,N-dimethyl-4-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 789 N,N-dimethyl-4-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 790 4-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N,N-dimethylbenzamide 791 4-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl)phenoxy)- N,N-dimethylbenzamide 792 4-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N,N-dimethylbenzamide 793 6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 794 6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 795 5-fluoro-6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 796 5-fluoro-6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 797 6-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 798 6-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 799 6-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 800 6-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 801 6-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 802 N-hydroxy-6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 803 N-hydroxy-6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 804 5-fluoro-N-hydroxy-6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 805 5-fluoro-N-hydroxy-6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 806 N-hydroxy-6-(2-methyl-4-(1-(trifluoromethy))cyclopropyl) phenoxy)nicotinamide 807 N-hydroxy-6-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 808 6-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N-hydroxynicotinamide 809 6-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl)phenoxy)- N-hydroxynicotinamide 810 6-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N-hydroxynicotinamide 811 N-methyl-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 812 N-methyl-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 813 3-fluoro-N-methyl-4-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 814 3-fluoro-N-methyl-4-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 815 N-methyl-4-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)benzamide 816 N-methyl-4-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)benzamide 817 4-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N-methylbenzamide 818 4-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl)phenoxy)- N-methylbenzamide 819 4-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N-methylbenzamide 820 N,N-dimethyl-6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 821 N,N-dimethyl-6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 822 5-fluoro-N,N-dimethyl-6-(4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 823 5-fluoro-N,N-dimethyl-6-(4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 824 N,N-dimethyl-6-(2-methyl-4-(1-(trifluoromethyl)cyclopropyl) phenoxy)nicotinamide 825 N,N-dimethyl-6-(2-methyl-4-(1-(perfluoroethyl)cyclopropyl) phenoxy)nicotinamide 826 6-(2-fluoro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N,N-dimethylnicotinamide 827 6-(2-fluoro-4-(1-(perfluoroethyl)cyclopropyl)phenoxy)- N,N-dimethylnicotinamide 828 6-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)- N,N-dimethylnicotinamide

Also included are isomers, e.g. enantiomers or diastereomers or rotamers or mixtures of isomers, salts, particularly pharmaceutically acceptable salts, and solvates of the compounds listed above.

Further Definitions

The term “C1-C12 alkyl” comprises all isomers of the corresponding saturated aliphatic hydrocarbon groups containing one to twelve carbon atoms; this includes methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, sec-pentyl, 3-pentyl, 2-methylbutyl, iso-pentyl, 2-methylbut-2-yl, 3-methylbut-2-yl, all hexyl-isomers, all heptyl-isomers, all octyl-isomers, all nonyl-isomers, all decyl-isomers, all undecyl-isomers and all dodecyl-isomers.

The term “C2-C12 alkenyl” comprises all isomers of the corresponding unsaturated olefinic hydrocarbon groups containing two to twelve carbon atoms linked by one or more double bonds; this includes vinyl, all propenyl-isomers, all butenyl-isomers, all pentenyl-isomers, all hexenyl-isomers, all heptenyl-isomers, all octenyl-isomers, all nonenyl-isomers, all decenyl-isomers, all undecenyl-isomers and all dodecenyl-isomers.

The term “C2-C12 alkynyl” comprises all isomers of the corresponding unsaturated olefinic hydrocarbon groups containing two to twelve carbon atoms linked by one or more triple bonds; this includes ethynyl, all propynyl-isomers, all butynyl-isomers, all pentynyl-isomers, all hexynyl-isomers, all heptynyl-isomers, all octynyl-isomers, all nonynyl-isomers, all decynyl-isomers, all undecynyl-isomers and all dodecynyl-isomers. The term “alkynyl” also includes compounds having one or more triple bonds and one or more double bonds.

The term “C3-C8 cycloalkyl” comprises the corresponding saturated hydrocarbon groups containing three to eight carbon atoms arranged in a monocyclic ring structure; this includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl.

The term “C3-C8 cycloalkenyl” comprises the corresponding unsaturated non-aromatic, anti-aromatic or aromatic hydrocarbon groups containing three to eight carbon atoms arranged in a monocyclic ring structure and linked by one or more double bonds; this includes cyclopropenyl, all cyclobutenyl-isomers, all cyclopentenyl-isomers, all cyclohexenyl-isomers, all cycloheptenyl-isomers, all cyclooctenyl-isomers.

The term “C4-C12 bicycloalkyl” comprises the corresponding saturated hydrocarbon groups containing four to twelve carbon atoms arranged in a bicyclic ring structure;

The term “C6-C12 bicycloalkenyl” comprises the corresponding unsaturated hydrocarbon groups containing six to twelve carbon atoms arranged in a bicyclic ring structure and linked by one or more double bonds;

The term “C5-C14 tricycloalkyl” comprises the corresponding saturated hydrocarbon groups containing five to fourteen carbon atoms arranged in a tricyclic ring structure;

The term “perhalogenated” relates to the exhaustive halogenation of the carbon scaffold; according residues comprise the corresponding perfluorinated, perchlorinated, perbrominated and periodinated groups. Preferably, the term “perhalogenated” relates to perfluorinated or perchlorinated groups, more preferably to perfluorinated groups.

The following contains definitions of terms used in this specification. The initial definition provided for a group or term herein applies to that group or term throughout the present specification, individually or as part of another group, unless otherwise indicated.

The compounds of the present invention may form salts, which are also within the scope of this invention. Reference to a compound of the invention herein is understood to include reference to salts thereof, unless otherwise indicated. The term “salt(s)”, as employed herein, denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases. Zwitterions (internal or inner salts) are included within the term “salt(s)” as used herein (and may be formed, for example, where the substituents comprise an acid moiety such as a carboxyl group). Also included herein are quaternary ammonium salts such as alkylammonium salts. Salts of the compounds may be formed, for example, by reacting a compound with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.

Exemplary acid addition salts include acetates (such as those formed with acetic acid or trihaloacetic acid, for example, trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, 2-hydroxyethanesulfonates, lactates, maleates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oxalates, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, pivalates, propionates, salicylates, succinates, sulfates (such as those formed with sulfuric acid), sulfonates (such as those mentioned herein), tartrates, thiocyanates, toluenesulfonates such as tosylates, undecanoates, and the like.

Exemplary basic salts (formed, for example, where the substituents comprise an acidic moiety such as a carboxyl group) include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as benzathines, dicyclohexylamines, hydrabamines, N-methyl-D-glucamines, N-methyl-D-glucamides, tert-butyl amines, and salts with amino acids such as arginine, lysine and the like. The basic nitrogen-containing groups may be quaternized with agents such as lower alkyl halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g. decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.

The present invention also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, “pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science 1977, 66 (2), each of which is incorporated herein by reference in its entirety.

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

Furthermore, in the case of the compounds of the invention which contain an asymmetric carbon atom, the invention relates to the D form, the L form and D,L mixtures and also, where more than one asymmetric carbon atom is present, to the diastereomeric forms. Those compounds of the invention which contain asymmetric carbon atoms, and which as a rule accrue as racemates, can be separated into the optically active isomers in a known manner, for example using an optically active acid. However, it is also possible to use an optically active starting substance from the outset, with a corresponding optically active or diastereomeric compound then being obtained as the end product.

Compounds of the invention also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Example prototropic tautomers include ketone-enol pairs, amide-imidic acid pairs, lactam-lactim pairs, amide-imidic acid pairs, enamine-imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H-imidazole, 1H-, 2H- and 4H-1,2,4-triazole, 1H- and 2H-isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.

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

Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium.

Also included are solvates and hydrates of the compounds of the invention and solvates and hydrates of their pharmaceutically acceptable salts.

The term “compound” as used herein is meant to include all stereoisomers, geometric isomers, tautomers, rotamers, and isotopes of the structures depicted, unless otherwise indicated.

In some embodiments, the compound can be provided as a prodrug. The term “prodrug”, as employed herein, denotes a compound, which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of the invention, or a salt and/or solvate thereof.

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

Pharmaceutical Methods

The compounds according to the invention have been found to have pharmacologically important properties, which can be used therapeutically. The compounds of the invention can be used alone, in combination with each other or in combination with other active compounds.

In certain embodiments, compounds of the present invention may be enhancers of Notch signalling.

The communication between cells via Notch signaling (reviewed in Kopan et al., Cell 2009, 137, 216-233; Bray, Nat. Rev. Mol. Cell Biol. 2016, 17, 722-735) is in the first step mediated by two types of transmembrane proteins: The Notch receptors being distributed within the cell membrane of the signal-receiving cell and the Notch ligands covering the membrane of the signal-sending cell. Mechanistically, Notch signaling is activated by receptor-ligand interaction, which leads to the proteolytic release of the intra cellular domain (NICD) of the membrane bound Notch receptor into the inside of the signal receiving cell. Subsequent translocation of NICD into the nucleus in turn leads to the transcriptional activation of certain and cell type specific genes. The Notch-mediated alteration of the previous gene-expression program of a cell is manifested in according cellular changes, which represent the response of the cell to a Notch signal.

The activation level of Notch signaling can be quantified in vitro most reliably by measuring the expression levels of Notch specific target genes. This can be accomplished by the quantification of corresponding mRNA or protein of a particular Notch target gene. Alternatively, cells can be genetically modified to carry a luciferase gene as an artificial Notch target gene, which is expressed in dependence of Notch activity. In this setting, Notch signaling levels can be quantified by measuring the luciferase-derived bioluminescence values.

An according Notch-reporter assay, i.e. a luciferase-based luminescence readout, was used here to quantify the ability of the claimed small molecules to augment Notch signaling in a cellular system. For this purpose, HeLa cells, obtainable from the American Type Culture Collection (ATCC) under the accession number ATCC-CCL-2, were transiently transfected for 24 hours using FuGENE® HD (Promega, #E2311) as transfection reagent with expression vectors of a membrane-tethered form of the constitutively active intracellular domain of the human Notch1 receptor (hNotch1ΔE) to activate the signaling cascade (BPS Bioscience, human analogue to Notch Pathway Reporter Kit #60509 component C), a Firefly luciferase being expressed under the control of a Notch-responsive promoter to monitor Notch signaling (BPS Bioscience, Notch Pathway Reporter Kit #60509, CSL luciferase reporter vector from component A not premixed with Renilla luciferase vector), and a Renilla luciferase being constitutively expressed in a Notch signaling independent manner to include a measure for the cell number per sample (Promega, pRL-SV40, #E2231). HeLa cells were cultivated according to the protocol of the provider in DMEM medium (Fisherscientific, #11584456) containing 10% fetal bovine serum (Fisherscientific, #15517589). The transfection was carried out in a 100 mm-culture dish (StarLab, #CC7682-3394) with cells being properly attached to the plate at a cell confluency of 80-90% in a total volume of 7 mL culture medium. Per dish to be transfected, a transfection mix was prepared by adding to 238 μL Opti-MEM (Fisherscientific, #10149832) 40 μL of the hNotch1ΔE expression vector (100 ng/μL), 80 μL of the CSL luciferase reporter vector (40 ng/L), 4 μL of the pRL-SV40-Renilla luciferase vector (10 ng/L), and in the last step 18.1 μL of FuGENE® HD. After addition of FuGENE® HD the transfection mix was let stand for 15 min at room temperature and hereafter equally distributed into the culture dish. Subsequently, i.e. after 24 hours of transfection, the transfected cells (10.000 cells per well) were incubated with the test-compounds at a final concentration of 10 μM (diluted from 10 mM stock-solutions in DMSO to a final DMSO concentration of 0.1% v/v) or with the empty carrier DMSO at 0.1% v/v as control for 20 hours in 96-well plates suitable for luminescence readouts (CORNING, #3610). Hereafter, the cells were lysed with 30 μL per well of Passive Lysis Buffer (Promega, #E194A, component of Dual-Luciferase® Reporter Assay System, #E1910) and the Firefly as well as Renilla luciferase values were measured with a luminescence reader with applying 15 μL per well each of the corresponding enzyme substrates needed to create the luminescence signals (Promega, Dual-Luciferase® Reporter Assay System, #E1910).

The suitability of the assays for monitoring Notch signaling was controlled by additionally including a generally accepted commercial Notch inhibitor, i.e. DAPT, as negative control, as well as the reported Notch enhancer resveratrol (RES) as positive control (Pinchot et al., Cancer 2011, 117, 1386-1398; Truong et al., Ann. Surg. Oncol. 2011, 18, 1506-1511; Yu et al., Mol. Cancer Ther. 2013, 12, 1276-1287). Both control compounds were likewise tested at 10 μM.

Per single experiment the measurement was performed in six replicates per compound. For every compound, this experiment was repeated in three or more independent replicates. The values of the Notch-reporter luciferase were normalized by division through the corresponding individual Notch-independent Renilla values in order to eliminate the impact of variation in the absolute cell-numbers in between the samples. For every individual plate, a second normalization was performed against the equally weighted arithmetic mean (here abbreviated as AVE) of the six associated Renilla-normalized DMSO-control values within a single experiment in order to obtain the relative values to a baseline level of 1.0. Two independent outlier analyses were performed according to the methods by Peirce and Chauvenet (Ross, Journal of Engineering Technology 2003, 1-12). Outliers confirmed by at least one of the methods were excluded from the calculations but not more than one value out of six per compound within a single experiment. The weighted arithmetic mean (here abbreviated as AVEw) for each compound was calculated from the double-normalized values over all independent replicates of the single experiments comprising the six replicates each. The corresponding standard deviation for the weighted arithmetic mean was calculated according to the method described by Bronstein et al. (Bronstein, Semendjajew, Musiol, Mühlig, Taschenbuch der Mathematik, 5th edition 2001 (German), publisher: Verlag Harri Deutsch, Frankfurt am Main and Thun) and was combined with the Gauß′ error propagation associated with the performed calculation for the normalization. The resulting standard deviation is herein referred to as “combined standard deviation”.

A compound is considered as a Notch augmenting molecule, i.e. an enhancer of Notch signaling, if the weighted arithmetic mean of the luminescence values after subtraction of the corresponding combined standard deviation amounts to 1.1 or higher, in particular to 1.2 or higher, 1.3 or higher, 1.4 or higher, 1.5 or higher, 1.7 or higher, and 2.0 or higher relative to the overall basis level of 1.0. The overall basis level was calculated as the weighted arithmetic mean of all double-normalized values from the DMSO control measurements in analogy to the calculations performed for the test-compounds. The corresponding combined standard deviation for the DMSO values amounts to less than 1·10−2.

According to the method described above, several molecules falling under the scope of the five compound families herein defined in formula I, formula II, formula III, formula IV and formula V have been identified as enhancers of Notch signaling. The so far identified Notch enhancers relate to the compounds listed in Table VI. The entries of Table VI are categorized by the corresponding weighted arithmetic mean of the compounds falling into the activity ranges as indicated.

TABLE VI Notch reporter assay Activity Range Entry Compound Specification AVEw ≥ 2.0 1 030 2 186 3 322 1.7 ≤ AVEw < 2.0 4 003 5 005 6 027 7 043 8 045 9 051 10 114 11 272 12 284 13 288 14 318 1.4 ≤ AVEw < 1.7 15 004 16 026 17 041 18 050 19 052 20 067 21 071 22 072 23 073 24 075 25 117 26 120 27 134 28 216 29 266 30 269 31 291 32 297 33 317 34 336 35 337 36 385 37 394 38 395 39 410 40 544 41 820 1.5 ± 0.0 42 RES Positive control 1.3 ≤ AVEw < 1.4 43 044 44 066 45 122 46 168 47 182 48 184 49 268 50 286 51 292 52 319 53 334 54 344 1.2 ≤ AVEw < 1.3 55 007 56 019 57 025 58 091 59 092 60 133 61 166 62 195 63 217 64 222 65 241 66 242 67 247 68 273 69 275 70 316 71 325 72 363 73 396 74 784 1.1 ≤ AVEw < 1.2 75 086 76 118 77 159 78 170 79 171 80 189 81 215 82 221 83 234 84 267 85 287 86 300 87 323 88 343 89 374 90 399 91 451 92 644 93 703 94 712 95 721 96 730 1.0 ± 0.0 97 DMSO Baseline control 0.1 ± 0.0 98 DAPT Negative control

Several other molecules have not been identified as enhancers of Notch signaling according to the above method.

In the course of the evaluation of molecules falling under formula I, formula II, formula III, formula IV and formula V in further cellular assays, results indicate that compounds of said molecule families exhibit growth inhibiting properties in hyperproliferative processes. In some cases, the growth inhibiting properties correlate with Notch enhancing properties, in other cases the growth inhibiting properties do not correlate with Notch enhancing properties.

The biological activity of the claimed compounds can be attributed to but may not be limited to Notch signaling enhancing activity. The secondary mechanisms of the claimed compounds leading to antiproliferative effects can be used alternatively or in combination with the Notch enhancing properties in medicinal treatments, preferably in the treatment of hyperproliferative disorders including cancer and non-malignant hyperproliferative disorders.

The antiproliferative activities of compounds falling under formula I, formula II, formula III, formula IV and formula V were investigated on cells or cell lines originating from a disorder of the myeloid cell compartment, the neuroendocrine system, the cervix, and the mucosal epithelium, as well as from the skin epithelium. To this end, HL-60 cells, TT cells, HeLa cells, CAL-27 cells and human primary epidermal keratinocytes (HPEK) were seeded into 96-well plates suitable for fluorescence assays (CORNING #3598) at following initial cell numbers: 1000 cells per well for HL-60; 9000 cells per well for TT; 2000 cells per well for HeLa, 2000 cells per well for CAL-27, 2000 cells per well for HPEK. The cells were treated with compounds at indicated final concentrations (diluted from the 1000× stock-solutions in DMSO to a final DMSO concentration of 0.1% v/v) or with the empty carrier DMSO at 0.1% v/v as control for 5 days. At day 5 after starting the treatments the cells were subjected to the alamarBlue® Proliferation Assay (Bio-Rad Serotec GmbH, BUF012B) according to the protocol of the manufacturer. The readout was taken with a multi-well plate-reader in the fluorescence mode with applying a filter for excitation at 560 nm (band width 10 nm) and for emission at 590 nm (band width 10 nm). Resveratrol (RES) treatment was included as control for growth inhibition.

The assays were performed in duplicate or more replicates of independent single experiments each containing a six-fold replicate for every condition. For every individual plate, the measured fluorescence intensity values of the conditions with compound treatment were normalized against the corresponding equally weighted arithmetic mean of the fluorescence intensity values of the six DMSO treated control wells in order to obtain the relative values to a baseline level of 1.0. The statistical calculations were performed in analogy to the luciferase assay as described above. To this end, two independent outlier analyses were performed according to the methods by Peirce and Chauvenet (Ross, Journal of Engineering Technology 2003, 1-12). Outliers confirmed by at least one of the methods were excluded from the calculations but not more than one value out of six per compound within a single experiment. The weighted arithmetic mean AVEw for each compound was calculated from the normalized values over all independent replicates of the single experiments comprising the six replicates each. The corresponding standard deviation for the weighted arithmetic mean was calculated according to the method described by Bronstein et al. (Bronstein, Semendjajew, Musiol, Mühlig, Taschenbuch der Mathematik, 5th edition 2001 (German), publisher: Verlag Harri Deutsch, Frankfurt am Main and Thun) and was combined with the Gauß′ error propagation associated with the performed calculation for the normalization. The resulting standard deviation is herein referred to as “combined standard deviation”.

In certain embodiments, the compounds of the present invention may be growth inhibitors in hyperproliferative processes, including malignant and non-malignant hyperproliferative processes.

In one embodiment, several compounds of the invention were found to inhibit the growth of HL-60 cells (human acute myeloid leukemia cells) obtainable from the Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 3. HL-60 cells were cultivated according to the protocol of the provider in RPMI 1640 medium (Fisherscientific, #11554526) containing 10% fetal bovine serum (Fisherscientific, #15517589).

A compound is considered as a growth inhibitor of HL-60 cells, if—at a reference concentration of 20 μM—the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the corresponding combined standard deviation amounts to 0.9 or lower, in particular to 0.8 or lower, 0.7 or lower, 0.6 or lower, 0.4 or lower, and 0.2 or lower, relative to the overall basis level of 1.0. The overall basis level was calculated as the weighted arithmetic mean of all normalized values from the DMSO control measurements in analogy to the calculations performed for the test-compounds. The corresponding combined standard deviation for the DMSO values amounts to less than 1·10−2.

According to the method described above, several molecules falling under the scope of the five compound families herein defined in formula I, formula II, formula III, formula IV and formula V have been identified as growth inhibitors of HL-60 cells. The so far identified HL-60 growth inhibitors relate to the compounds listed in Table VII. The entries of Table VII are categorized by the corresponding weighted arithmetic means of the compounds falling into the activity ranges as indicated.

TABLE VII Proliferation assay with HL-60 cells at 20 μM Activity Range Entry Compound Specification 1.0 ± 0.0 1 DMSO Baseline control 0.8 < AVEw ≤ 0.9 2 002 3 030 4 054 5 072 6 073 7 075 8 087 9 092 10 165 11 245 12 248 13 298 14 300 15 316 16 317 17 325 18 337 19 374 20 385 21 395 22 399 23 427 24 477 25 592 26 712 27 723 28 731 29 738 30 739 31 740 32 792 33 793 34 811 35 812 36 819 0.7 < AVEw ≤ 0.8 37 041 38 134 39 215 40 223 41 322 42 410 43 440 44 488 45 581 46 674 47 685 48 756 49 785 50 786 51 822 0.6 < AVEw ≤ 0.7 52 067 53 217 54 222 55 334 56 336 57 414 58 492 59 700 60 821 61 828 0.4 < AVEw ≤ 0.6 62 043 63 044 64 045 65 066 66 133 67 159 68 164 69 167 70 218 71 221 72 236 73 238 74 313 75 318 76 319 77 320 78 323 79 389 80 721 81 722 82 729 83 784 84 820 0.4 ± 0.0 85 RES 20 μM Control 0.2 < AVEw ≤ 0.4 86 161 87 210 88 211 89 237 90 596 0.0 ≤ AVEw ≤ 0.2 91 166 92 168 93 170 94 171 95 182 96 184 97 185 98 186 99 216 100 266 101 267 102 268 103 269 104 270 105 272 106 273 107 275 108 284 109 286 110 287 111 288 112 529 113 540 114 544 115 633 116 644 117 648 118 766 119 767 120 768 121 774 122 802 123 803 124 804 125 810

In one embodiment, several compounds of the invention were found to inhibit the growth of CAL-27 cells (human tongue squamous cell carcinoma cells) obtainable from the Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 446. CAL-27 cells were cultivated according to the protocol of the provider (but at 5% instead of 10% CO2) in DMEM medium (Fisherscientific, #11584456) containing 10% fetal bovine serum (Fisherscientific, #15517589).

A compound is considered as a growth inhibitor of CAL-27 cells, if—at a reference concentration of 20 μM—the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the corresponding combined standard deviation amounts to 0.9 or lower, in particular to 0.8 or lower, 0.7 or lower, 0.6 or lower, 0.4 or lower, and 0.2 or lower, relative to the overall basis level of 1.0. The overall basis level was calculated as the weighted arithmetic mean of all normalized values from the DMSO control measurements in analogy to the calculations performed for the test-compounds. The corresponding combined standard deviation for the DMSO values amounts to less than 1·10−2.

According to the method described above, several molecules falling so far under the scope of the three compound families herein defined in formula II, formula IV and formula V have been identified as growth inhibitors of CAL-27 cells. The so far identified CAL-27 growth inhibitors relate to the compounds listed in Table Villa and VIIIb. The entries of Table Villa and VIIIb are categorized by the corresponding weighted arithmetic means of the compounds falling into the activity ranges as indicated.

TABLE VIIIa Proliferation assay with CAL-27 cells at 20 μM Activity Range Entry Compound Specification 1.0 ± 0.0 1 DMSO Baseline control 0.9 ± 0.0 2 RES 20 μM Control 0.8 < AVEw ≤ 0.9 3 236 4 300 5 596 6 820 7 822 0.7 < AVEw ≤ 0.8 8 164 9 210 10 313 11 774 0.6 < AVEw ≤ 0.7 12 167 13 238 0.4 < AVEw ≤ 0.6 14 211 15 237 16 266 0.4 ± 0.0 17 RES 40 μM Control 0.2 < AVEw ≤ 0.4 18 166 19 170 20 182 21 267 22 287 23 288 24 768 0.0 ≤ AVEw ≤ 0.2 25 168 26 171 27 184 28 185 29 186 30 268 31 269 32 270 33 272 34 273 35 275 36 284 37 286 38 529 39 540 40 544 41 633 42 644 43 648 44 766 45 767 46 802 47 803 48 804 49 810

TABLE VIIIb Proliferation assay with CAL-27 cells at 40 μM Activity Range Entry Compound Specification 1.0 ± 0.0 1 DMSO Baseline control 0.9 ± 0.0 2 RES 24M Control 0.8 < AVEw ≤ 0.9 3 300 4 334 0.7 < AVEw ≤ 0.8 5 722 0.6 < AVEw ≤ 0.7 6 159 0.5 ± 0.0 7 RES 40 μM Control 0.2 < AVEw ≤ 0.4 8 161 9 237 10 729 11 768 0.0 ≤ AVEw ≤ 0.2 12 166 13 167 14 210 15 272 16 287

In one embodiment, several compounds of the invention were found to inhibit the growth of TT cells (human medullary thyroid carcinoma cells) obtainable from the American Type Culture Collection (ATCC) under the accession number ATCC-CRL-1803. TT cells were cultivated according to the protocol of the provider in F-12K medium (Fisherscientific, #11580556, or ATCC, #ATCC-30-2004) containing 10% fetal bovine serum (Fisherscientific, #15517589).

A compound is considered as a growth inhibitor of TT cells, if—at a reference concentration of 40 μM—the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the corresponding combined standard deviation amounts to 0.9 or lower, in particular to 0.8 or lower, 0.7 or lower, 0.6 or lower, 0.4 or lower, and 0.2 or lower, relative to the overall basis level of 1.0. The overall basis level was calculated as the weighted arithmetic mean of all normalized values from the DMSO control measurements in analogy to the calculations performed for the test-compounds. The corresponding combined standard deviation for the DMSO values amounts to less than 1·10−2.

According to the method described above, several molecules falling so far under the scope of the three compound families herein defined in formula II, formula IV and formula V have been identified as growth inhibitors of TT cells. The so far identified TT growth inhibitors relate to the compounds listed in Table IX. The entries of Table IX are categorized by the corresponding weighted arithmetic means of the compounds falling into the activity ranges as indicated.

TABLE IX Proliferation assay with TT cells at 40 μM Activity Range Entry Compound Specification 1.0 ± 0.0 1 DMSO Baseline control 0.9 ± 0.0 2 RES 20 μM Control 0.8 < AVEw ≤ 0.9 3 159 4 309 5 334 0.8 ± 0.0 6 RES 40 μM Control 0.7 < AVEw ≤ 0.8 7 748 0.6 < AVEw ≤ 0.7 8 210 0.4 < AVEw ≤ 0.6 9 161 10 237 0.2 < AVEw ≤ 0.4 11 166 12 167 13 171 14 182 15 186 16 287 17 540 18 544 19 644 20 729 0.0 ≤ AVEw ≤ 0.2 21 272 22 284 23 288 24 768

In one embodiment, several compounds of the invention were found to inhibit the growth of HeLa cells (human cervical adenocarcinoma cells) obtainable from the American Type Culture Collection (ATCC) under the accession number ATCC-CCL-2. HeLa cells were cultivated according to the protocol of the provider in DMEM medium (Fisherscientific, #11584456) containing 10% fetal bovine serum (Fisherscientific, #15517589).

A compound is considered as a growth inhibitor of HeLa cells, if—at a reference concentration of 40 μM—the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the corresponding combined standard deviation amounts to 0.9 or lower, in particular to 0.8 or lower, 0.7 or lower, 0.6 or lower, 0.4 or lower, and 0.2 or lower, relative to the overall basis level of 1.0. The overall basis level was calculated as the weighted arithmetic mean of all normalized values from the DMSO control measurements in analogy to the calculations performed for the test-compounds. The corresponding combined standard deviation for the DMSO values amounts to less than 1·10−2.

According to the method described above, several molecules falling so far under the scope of the compound family herein defined in formula II have been identified as growth inhibitors of HeLa cells. The so far identified HeLa growth inhibitors relate to the compounds listed in Table X. The entries of Table X are categorized by the corresponding weighted arithmetic means of the compounds falling into the activity ranges as indicated.

TABLE X Proliferation assay with HeLa cells at 40 μM Activity Range Entry Compound Specification 1.0 ± 0.0 1 DMSO Baseline control 0.9 ± 0.0 2 RES 20 μM Control 0.4 < AVEw ≤ 0.6 3 166 0.4 ± 0.0 4 RES 40 μM Control 0.2 < AVEw ≤ 0.4 5 167 6 287 0.0 ≤ AVEw ≤ 0.2  7 272

In one embodiment, several compounds of the invention were found to inhibit the growth of human epidermal keratinocyte progenitors, (HPEKp, pooled), obtainable from CELLnTEC Advanced Cell Systems AG under the accession number HPEKp. HPEKp cells were cultivated according to the protocol of the provider in CnT-Prime epithelial culture medium (CELLnTEC, #CnT-PR, a fully defined, low calcium formulation, completely free of animal or human-derived components) without addition of further components.

A compound is considered as a growth inhibitor of HPEKp cells, if—at a reference concentration of 10 μM—the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the corresponding combined standard deviation amounts to 0.9 or lower, in particular to 0.8 or lower, 0.7 or lower, 0.6 or lower, 0.4 or lower, and 0.2 or lower, relative to the overall basis level of 1.0. The overall basis level was calculated as the weighted arithmetic mean of all normalized values from the DMSO control measurements in analogy to the calculations performed for the test-compounds. The corresponding combined standard deviation for the DMSO values amounts to less than 1·10−2.

According to the method described above, several molecules falling so far under the scope of the four compound families herein defined in formula II, formula III, formula IV and formula V have been identified as growth inhibitors of HPEKp cells. The so far identified HPEKp growth inhibitors relate to the compounds listed in Table XI. The entries of Table XI are categorized by the corresponding weighted arithmetic means of the compounds falling into the activity ranges as indicated.

TABLE XI Proliferation assay with HPEKp cells at 10 μM Activity Range Entry Compound Specification 1.0 ± 0.0 1 DMSO Baseline control 0.8 < AVEw ≤ 0.9 3 140 4 374 5 731 6 747 7 749 8 801 0.7 < AVEw ≤ 0.8 10 312 11 323 12 424 13 721 14 819 15 828 0.6 < AVEw ≤ 0.7 16 086 17 190 18 334 0.4 < AVEw ≤ 0.6 19 112 20 722 0.4 ± 0.0 21 RES 10 μM Control 0.2 < AVEw ≤ 0.4 22 389 23 440 24 540 0.0 ≤ AVEw ≤ 0.2  25 159 26 182 27 185 28 273 29 287 30 644 31 810

Preliminary results from a single proliferation assay of six replicates per condition using cells derived from murine muscle tissue show that compounds of the invention may exhibit antiproliferative activity on muscle cells. Compounds were tested on C2C12 cells using the alamarBlue® proliferation assay in analogy to the above described method with seeding the cells at an initial number of 2000 cells per 96-well and a duration of treatment with compounds for 3 days.

In one embodiment, two compounds of the invention were found so far to inhibit the growth of C2C12 cells (murine myoblast cells) obtainable from the Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 565. C2C12 cells were cultivated according to the protocol of the provider in RPMI 1640 medium (Fisherscientific, #11554526) containing 10% fetal bovine serum (Fisherscientific, #15517589).

A compound is considered as a growth inhibitor of C2C12 cells, if—at a reference concentration of 40 μM—the equally weighted arithmetic mean (AVE) of the six normalized fluorescence intensity values after addition of the corresponding standard deviation amounts to 0.9 or lower, in particular to 0.8 or lower, 0.7 or lower, 0.6 or lower, 0.4 or lower, and 0.2 or lower, relative to the overall basis level of 1.0. The overall basis level was calculated as the equally weighted arithmetic mean (AVE) of the six normalized values from the DMSO control measurements in analogy to the calculations performed for the test-compounds. The corresponding standard deviations for the tested compounds were calculated including the Gauß′ error propagation associated with the performed calculation for the normalization and amounts for the DMSO values to less than 3·10−2. Outlier analyses were performed as described above.

According to the method described above, molecules falling so far under the scope of the two compound families herein defined in formula II and formula V have been identified as growth inhibitors of C2C12 cells. The so far identified C2C12 growth inhibitors relate to the compounds listed in Table XII. The entries of Table XII are categorized by the corresponding equally weighted arithmetic means of the compounds falling into the activity ranges as indicated.

TABLE XII Proliferation assay with C2C12 cells at 40 μM Activity Range Entry Compound Specification 1.0 ± 0.0 1 DMSO Baseline control 0.8 < AVE ≤ 0.9 2 748 0.3 ± 0.0 3 RES 40 μM Control 0.2 < AVE ≤ 0.4 4 288

Preliminary results from a single proliferation assay of six replicates per condition using squamous cell carcinoma (SCC) cells derived from the human oral mucosa may confirm that compounds of the invention exhibit antiproliferative activity on SCC of the mucosal epithelium. Compounds were tested on BHY cells using the alamarBlue® proliferation assay in analogy to the above described method with seeding the cells at an initial number of 4000 cells per 96-well and a duration of treatment with compounds for 3 days.

In one embodiment, several compounds of the invention were found to inhibit the growth of BHY cells (human oral squamous cell carcinoma cells) obtainable from the Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 404. BHY cells were cultivated according to the protocol of the provider (but at 5% instead of 10% CO2) in DMEM medium (Fisherscientific, #11584456) containing 10% fetal bovine serum (Fisherscientific, #15517589).

A compound is considered as a growth inhibitor of BHY cells, if—at a reference concentration of 40 μM—the equally weighted arithmetic mean (AVE) of the six normalized fluorescence intensity values after addition of the corresponding standard deviation amounts to 0.9 or lower, in particular to 0.8 or lower, 0.7 or lower, 0.6 or lower, 0.4 or lower, and 0.2 or lower, relative to the overall basis level of 1.0. The overall basis level was calculated as the weighted arithmetic mean (AVEw) of all normalized values from the DMSO control measurements. The corresponding combined standard deviation for the DMSO values amounts to less than 1·10−2. The corresponding standard deviations for the tested compounds were calculated including the Gauß′ error propagation associated with the performed calculation for the normalization. The weighted arithmetic mean (AVEw) and the combined standard deviation for RES was calculated in analogy to DMSO. Outlier analyses were performed as described above.

According to the method described above, molecules falling so far under the scope of the two compound families herein defined in formula II and formula IV have been identified as growth inhibitors of BHY cells. The so far identified BHY growth inhibitors relate to the compounds listed in Table XIII. The entries of Table XIII are categorized by the corresponding equally weighted arithmetic means of the compounds falling into the activity ranges as indicated.

TABLE XIII Proliferation assay with BHY cells at 40 μM Activity Range Entry Compound Specification 1.0 ± 0.0 1 DMSO Baseline control 0.6 < AVE ≤ 0.7 2 644 0.6 ± 0.0 3 RES 40 μM Control 0.4 < AVE ≤ 0.6 4 171 0.2 < AVE ≤ 0.4 5 182 6 186 7 272 8 284 9 288 10 544 11 633 0.0 ≤ AVE ≤ 0.2  12 540

In one aspect, the present invention relates to the treatment of skin, skin appendages, mucosa, mucosal appendages, cornea, and all kinds of epithelial tissue. The term “skin” relates to tissue including epidermis and dermis. The term “mucosa” relates to mucous and submucous tissues including oral mucosa, nasal mucosa, ocular mucosa, mucosa of the ear, respiratory mucosa, genital mucosa, urothelial mucosa, anal mucosa and rectal mucosa. The term “appendages” relates to tissue including hair follicles, hair, fingernails, toenails and glands including sebaceous glands, sweat glands, e.g. apocrine or eccrine sweat glands and mammary glands.

In one embodiment, the present invention relates to treatment of non-melanoma skin cancer and pre-cancerous lesions, such as basal cell carcinoma (BCC), squamous cell carcinoma (SCC), e.g. cutaneous SCC, lung SCC, head and neck SCC, oral SCC, esophageal SCC, cervical SCC, periocular SCC, SCC of the thyroid, SCC of the penis, SCC of the vagina, SCC of the prostate, SCC of the bladder, sebaceous gland carcinoma, Merkel cell carcinoma, angiosarcoma, cutaneous B-cell lymphoma, cutaneous T-cell lymphoma, dermatofibrosarcoma, actinic keratosis (AK) or Bowen's disease (BD).

In a further embodiment, the present invention relates to the treatment of skin and mucosal disorders with cornification defects (keratoses) and/or abnormal keratinocyte proliferation, such as Psoriasis, Darier's disease, Lichen planus, Lupus erythematosus, Ichthyosis or Verruca vulgaris (senilis).

In a further embodiment, the invention relates to the treatment of skin and mucosal diseases related to and caused by viral infections, such as warts, HPV-related warts, papillomas, HPV-related papillomas, papillomatoses and HPV-related papillomatoses, e.g. Verruca (plantar warts), Verruca plana (flat warts/plane warts), Verruca filiformis (filiform warts), mosaic warts, periungual warts, subungual warts, oral warts, genital warts, fibroepithelial papilloma, intracanalicular papilloma, intraductal papilloma, inverted papilloma, basal cell papilloma, squamous papilloma, cutaneous papilloma, fibrovasular papilloma, plexus papilloma, nasal papilloma, pharyngeal papilloma, Papillomatosis cutis carcinoides, Papillomatosis cutis lymphostatica, Papillomatosis confluens et reticularis or laryngeal papillomatosis (respiratory papillomatosis), Herpes-related diseases, e.g. Herpes labialis, Herpes genitalis, Herpes zoster, Herpes corneae or Kaposi's sarcoma.

In a further embodiment, the invention relates to the treatment of atopic dermatitis.

In a further embodiment, the invention relates to the treatment of acne.

In a further embodiment, the invention relates to the treatment of wounds of the skin, wherein the process of wound healing is accelerated.

A further aspect of the present invention relates to the treatment of immune system-related disorders. The term “immune system-related” as used herein applies to a pathological condition of the hematopoietic system including the hematologic system, as well as to the intervention into proliferation, differentiation and/or activation of cell lineages of the hematopoietic system including the hematologic system in order to modulate an immune response (immune modulation).

Examples are diseases of the hematopoietic system including the hematologic system, such as malignancies of the myeloid lineage, e.g. chronic myelomonocytic leukemia (CMML) or acute myeloid leukemia (AML), including acute promyelocytic leukemia (APL); malignancies of the lymphoid lineage, e.g. B-cell acute lymphoblastic leukemia (B-ALL), pre-B-cell acute lymphoblastic leukemia (pre-B-ALL), Hodgkin lymphoma or myeloma; or acute lymphoblastic and acute myeloid mixed lineage leukemia with MLL gene translocation.

Furthermore, the compounds of the invention may be used in immunotherapy, alone or together with other immunotherapeutic methods or compounds, or as adjuvant for immunotherapy. The term “immunotherapy” as used herein applies to activation-immunotherapy in patients without immune deficiency or with acquired or congenital immune deficiency, and as immune recovery to enhance the functionality of the immune system in the response against pathogens or pathologically transformed endogenous cells, such as cancer cells.

The term “other immunotherapy methods” as used herein applies to vaccinations, antibody treatment, cytokine therapy, the use of immune checkpoint inhibitors and immune response-stimulating drugs, as well as to autologous transplantations of genetically modified or non-modified immune cells, which may be stimulated with intercellular signals, or signaling molecules, or antigens, or antibodies, i.e. adoptive immune-cell transfer.

Specific examples are activation of peripheral T-lymphocytes in order to amplify an immune response, particularly the stimulation of proliferation and/or cytokine production and/or secretion upon antigen recognition in order to amplify an immune response, such as the activation of B-lymphocytes in order to amplify an immune response, particularly the stimulation of proliferation and/or antibody production and/or secretion, such as the enhancement of an immune response through augmentation of the number of specific immune-cell subtypes, by regulation of differentiation and/or cell fate decision during immune-cell development, as for example to augment the number of marginal zone B-cells, or T-helper (Th) subsets in particular Th1, Th2 and regulatory T-cells; or the use as vaccine adjuvant.

A still further aspect of the invention relates to the treatment of muscular diseases including diseases of skeletal muscle, cardiac muscle and smooth muscle.

In one embodiment, the invention relates to the treatment of muscular dystrophies (MD).

Specific examples are Duchenne MD, Becker MD, congenital MD, Limb-Girdle MD, facioscapulohumeral MD, Emery-Dreifuss MD, distal MD, myotonic MD or oculopharyngeal MD.

In a further embodiment, the invention relates to the treatment of hyperproliferative disorders of the muscle, including myoblastoma, rhabdomyoma, and rhabdomyosarcoma, as well as muscle hyperplasia and muscle hypertrophy.

In a further embodiment, the compounds of the invention may be used for muscle regeneration after pathologic muscle degeneration or atrophy, e.g. caused by traumata, caused by muscle ischemia or caused by inflammation, in aging-related muscle-atrophy or in disease-related muscle atrophy such as myositis and fibromyositis or poliomyelitis.

A still further aspect relates to the treatment of disorders of the neuroendocrine system such as cancer of the neuroendocrine system, comprising neuroendocrine small cell carcinomas, neuroendocrine large cell carcinomas and carcinoid tumors, e.g. of the brain, thyroid, pancreas, gastrointestinal tract, liver, esophagus, and lung, such as neuroendocrine tumor of the pituitary gland, neuroendocrine tumor of the adrenal gland, medullary thyroid cancer (MTC), C-cell hyperplasia, anaplastic thyroid cancer (ATC), parathyroid adenoma, intrathyroidal nodules, insular carcinoma, hyalinizing trabecular neoplasm, paraganglioma, small-cell lung cancer (SCLC), lung carcinoid tumors, neuroblastoma, gastrointestinal carcinoid, Goblet-cell carcinoid, pancreatic carcinoid, gastrinoma, glucagenoma, somatostatinoma, VIPoma, insulinoma, non-functional islet cell tumor, multiple endocrine neoplasia type-1, or pulmonary carcinoid.

A still further aspect relates to the treatment of cancers or precancerous lesions of the brain, pancreas, liver, thyroid, genitourinary tract and endothelial tissue, including glioma, mixed glioma, glioblastoma multiforme, astrocytoma, anaplastic astrocytoma, glioblastoma, oligodendroglioma, anaplastic oligodendroglioma, anaplastic oligoastrocytoma, ependymoma, anaplastic ependymoma, myxopapillary ependymoma, subependymoma, brain stem glioma, optic nerve glioma, and forebrain tumors, pancreatic adenocarcinoma, pancreatic ductal adenocarcinoma, pancreatic acinar cell carcinoma, pancreatic pseudopapillary neoplasm, pancreatic intraductal papillary-mucinous neoplasm, pancreatic mucinous cystadenocarcinoma, pancreatoblastoma and pancreatic intraepithelial neoplesia, hepatocellular carcinoma, fibrolamellar hepatocellular carcinoma, papillary thyroid cancer and follicular thyroid cancer, cervical cancer and angiosarcoma.

As used herein, the term “treating” or “treatment” refers to one or more of (1) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (2) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease. The term “treating” also encompasses post-treatment care.

In some embodiments, administration of a compound of the invention, or pharmaceutically acceptable salt thereof, is effective in preventing the disease; for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease.

The compounds of the invention may be used in human and veterinary medicine, which includes the treatment of companion animals, e.g. horses, dogs, cats, rabbits, guinea pigs, birds, fishes; and livestock, e.g. cattle, poultry, pig, sheep, goat, donkey, yak and camel.

Pharmaceutical Compositions

The present invention further provides pharmaceutical compositions comprising a compound as described herein or a pharmaceutically acceptable salt thereof for use in medicine, e.g. in human or veterinary medicine. In some embodiments, the composition further comprises a pharmaceutically acceptable carrier.

An effective dose of the compounds according to the invention, or their salts, solvates or prodrugs thereof is used, in addition to physiologically acceptable carriers, diluents and/or adjuvants for producing a pharmaceutical composition. The dose of the active compounds can vary depending on the route of administration, the age and weight of the patient, the nature and severity of the diseases to be treated, and similar factors. The daily dose can be given as a single dose, which is to be administered once, or be subdivided into two or more daily doses, and is as a rule 0.001-2000 mg. Particular preference is given to administering daily doses of 0.1-500 mg, e.g. 0.1-100 mg.

Suitable administration forms are topical or systemical including enteral, oral, rectal, and parenteral, as infusion and injection, intravenous, intra-arterial, intraperitoneal, intramuscular, intracardial, epidural, intracerebral, intracerebroventricular, intraosseous, intra-articular, intraocular, intravitreal, intrathecal, intravaginal, intracavernous, intravesical, subcutaneous, intradermal, transdermal, transmucosal, inhalative, intranasal, buccal, sublingual and intralesional preparations. Particular preference is given to using oral, parenteral, e.g. intravenous or intramuscular, intranasal preparations, e.g. dry powder or sublingual, of the compounds according to the invention. The customary galenic preparation forms, such as tablets, sugar-coated tablets, capsules, dispersible powders, granulates, aqueous solutions, alcohol-containing aqueous solutions, aqueous or oily suspensions, gels, hydrogels, ointments, creams, lotions, shampoos, lip balms, mouthwashs, foams, pastes, tinctures, dermal patches and tapes, forms in occlusion or in combination with time release drug delivery systems, with electrophoretic dermal delivery systems including implants and devices, and with jet injectors, liposome and transfersome vesicles, vapors, sprays, syrups, juices or drops and eye drops, can be used.

Solid medicinal forms can comprise inert components and carrier substances, such as calcium carbonate, calcium phosphate, sodium phosphate, lactose, starch, mannitol, alginates, gelatine, guar gum, magnesium stearate, aluminium stearate, methyl cellulose, talc, highly dispersed silicic acids, silicone oil, higher molecular weight fatty acids, (such as stearic acid), gelatine, agar agar or vegetable or animal fats and oils, or solid high molecular weight polymers (such as polyethylene glycol); preparations which are suitable for oral administration can comprise additional flavourings and/or sweetening agents, if desired.

Liquid medicinal forms can be sterilized and/or, where appropriate, comprise auxiliary substances, such as preservatives, stabilizers, wetting agents, penetrating agents, emulsifiers, spreading agents, solubilizers, salts, sugars or sugar alcohols for regulating the osmotic pressure or for buffering, and/or viscosity regulators. Examples of such additives are tartrate and citrate buffers, ethanol and sequestering agents (such as ethylenediaminetetraacetic acid and its non-toxic salts). High molecular weight polymers, such as liquid polyethylene oxides, microcrystalline celluloses, carboxymethyl celluloses, polyvinylpyrrolidones, dextrans or gelatine, are suitable for regulating the viscosity. Examples of solid carrier substances are starch, lactose, mannitol, methyl cellulose, talc, highly dispersed silicic acids, high molecular weight fatty acids (such as stearic acid), gelatine, agar agar, calcium phosphate, magnesium stearate, animal and vegetable fats, and solid high molecular weight polymers, such as polyethylene glycol.

Oily suspensions for parenteral or topical applications can be vegetable, synthetic or semisynthetic oils, such as liquid fatty acid esters having in each case from 8 to 22 C atoms in the fatty acid chains, for example palmitic acid, lauric acid, tridecanoic acid, margaric acid, stearic acid, arachidic acid, myristic acid, behenic acid, pentadecanoic acid, linoleic acid, elaidic acid, brasidic acid, erucic acid or oleic acid, which are esterified with monohydric to trihydric alcohols having from 1 to 6 C atoms, such as methanol, ethanol, propanol, butanol, pentanol or their isomers, glycol or glycerol. Examples of such fatty acid esters are commercially available miglyols, isopropyl myristate, isopropyl palmitate, isopropyl stearate, PEG 6-capric acid, caprylic/capric acid esters of saturated fatty alcohols, polyoxyethylene glycerol trioleates, ethyl oleate, waxy fatty acid esters, such as artificial ducktail gland fat, coconut fatty acid isopropyl ester, oleyl oleate, decyl oleate, ethyl lactate, dibutyl phthalate, diisopropyl adipate, polyol fatty acid esters, inter alia. Silicone oils of differing viscosity, or fatty alcohols, such as isotridecyl alcohol, 2-octyldodecanol, cetylstearyl alcohol or oleyl alcohol, or fatty acids, such as oleic acid, are also suitable. It is furthermore possible to use vegetable oils, such as castor oil, almond oil, olive oil, sesame oil, cotton seed oil, groundnut oil or soybean oil.

Suitable solvents, gelatinizing agents and solubilizers are water or water-miscible solvents. Examples of suitable substances are alcohols, such as ethanol or isopropyl alcohol, benzyl alcohol, 2-octyldodecanol, polyethylene glycols, phthalates, adipates, propylene glycol, glycerol, di- or tripropylene glycol, waxes, methyl cellosolve, cellosolve, esters, morpholines, dioxane, dimethyl sulphoxide, dimethylformamide, tetrahydrofuran, cyclohexanone, etc.

Cellulose ethers which can dissolve or swell both in water or in organic solvents, such as hydroxypropylmethyl cellulose, methyl cellulose or ethyl cellulose, or soluble starches, can be used as film-forming agents.

Mixtures of gelatinizing agents and film-forming agents are also perfectly possible. In this case, use is made, in particular, of ionic macromolecules such as sodium carboxymethyl cellulose, polyacrylic acid, polymethacrylic acid and their salts, sodium amylopectin semiglycolate, alginic acid or propylene glycol alginate as the sodium salt, gum arabic, xanthan gum, guar gum or carrageenan. The following can be used as additional formulation aids: glycerol, paraffin of differing viscosity, triethanolamine, collagen, allantoin and novantisolic acid. Use of surfactants, emulsifiers or wetting agents, for example of Na lauryl sulphate, fatty alcohol ether sulphates, di-Na—N-lauryl-p-iminodipropionate, polyethoxylated castor oil or sorbitan monooleate, sorbitan monostearate, polysorbates (e.g. Tween), cetyl alcohol, lecithin, glycerol monostearate, polyoxyethylene stearate, alkylphenol polyglycol ethers, cetyltrimethylammonium chloride or mono-/dialkylpolyglycol ether orthophosphoric acid monoethanolamine salts can also be required for the formulation. Stabilizers, such as montmorillonites or colloidal silicic acids, for stabilizing emulsions or preventing the breakdown of active substances such as antioxidants, for example tocopherols or butylhydroxyanisole, or preservatives, such as p-hydroxybenzoic acid esters, can likewise be used for preparing the desired formulations.

Preparations for parenteral administration can be present in separate dose unit forms, such as ampoules or vials. Use is preferably made of solutions of the active compound, preferably aqueous solution and, in particular, isotonic solutions and also suspensions. These injection forms can be made available as ready-to-use preparations or only be prepared directly before use, by mixing the active compound, for example the lyophilisate, where appropriate containing other solid carrier substances, with the desired solvent or suspending agent.

Intranasal preparations can be present as aqueous or oily solutions or as aqueous or oily suspensions. They can also be present as lyophilisates which are prepared before use using the suitable solvent or suspending agent.

Inhalable preparations can present as powders, solutions or suspensions. Preferably, inhalable preparations are in the form of powders, e.g. as a mixture of the active ingredient with a suitable formulation aid such as lactose.

The preparations are produced, aliquoted and sealed under the customary antimicrobial and aseptic conditions.

As indicated above, the compounds of the invention may be administered as a combination therapy, as sequence therapy or as simultaneous combination therapy, with further active agents, e.g. therapeutically active compounds useful in the treatment of the above indicated disorders. These therapeutically active compounds may include but are not limited to chemotherapeutic agents such as nucleoside analogs, e.g. Cytarabin, Gemcitabine, Azathioprine, Mercaptopurine, Fluorouracil, Thioguanine, Hydroxyurea, Azacitidine, Capecitabine, Doxifluridine, and Methotrexate; such as platinum-based drugs, e.g. Cisplatin, Oxaliplatin, Carboplatin and Nedaplatin; such as anthracyclines, e.g. Doxorubicin, Epirubicin, Valrubicin, Idarubicin, Daunorubicin, Sabarubicin, Pixantrone and Mitoxantrone; such as peptide antibiotics, e.g. Actinomycin and Bleomycin; such as alkylating agents e.g. Mechlorethamine, Chlorambucil, Melphalan, Nitrosoureas, Dacarbazine, Temozolomide and Cyclophosphamide; such as antimitotic agents including taxanes and vinca alkaloids, e.g. Docetaxel, Paclitaxel, Abraxane, Cabazitaxel, Vinblastine, Vindesine, Vinorelbine and Vincristine; such as topoisomerase inhibitors, e.g. Irinotecan, Topotecan, Teniposide and Etoposide; and targeted therapeutic agents such as kinase inhibitors, regulators i.e. inhibitors and activators of signaling pathways including growth factor signaling, cytokine signaling, NF-kappaB signaling, AP1 signaling, JAK/STAT signaling, EGFR signaling, TGF-beta signaling, Notch signaling, Wnt signaling, Hedgehog signaling, hormone and nuclear receptor signaling, e.g. Erlotinib, Lapatinib, Dasatinib, Imatinib, Afatinib, Vemurafenib, Dabrafenib, Nilotinib, Cetuximab, Trametinib, Palbociclib, Cobimetinib, Cabozantinib, Pegaptanib, Crizotinib, Olaparib, Panitumumab, Cabozantinib, Ponatinib, Regorafenib, Entrectinib, Ranibizumab, Ibrutinib, Trastuzumab, Rituximab, Alemtuzumab, Gefitinib, Bevacizumab, Lenvatinib, Bosutinib, Axitinib, Pazopanib, Everolimus, Temsirolimus, Ruxolitinib, Tofacitinib, Sorafenib, Sunitinib, Aflibercept, Bortezomib, Vandetanib; Vismodegib and Sonidegib; retinoids such as retinol, tretinoin, isotretinoin, alitretinoin, bexarotene, tazarotene, acitretin, adapalene and etretinate; hormone signaling modulators including estrogen receptor modulators, androgen receptor modulators and aromatase inhibitors e.g. Raloxifene, Tamoxifen, Fulvestrant, Lasofoxifene, Toremifene, Bicalutamide, Flutamide, Anastrozole, Letrozole and Exemestane; histone deacetylase inhibitors, e.g. Vorinostat, Romidepsin, Panobinostat, Belinostat and Chidamide; and Ingenol mebutate; and other Notch enhancers not encompassed by the compounds of the present invention, e.g. Valproic acid, Resveratrol, hesperetin, chrysin, phenethyl isothiocyanate, thiocoraline, N-methylhemeanthidine chloride and Notch Signaling-activating peptides or antibodies; and immune response modulating agents e.g. Imiquimod, Ipilimumab, Atezolizumab, Ofatumumab, Rituximab, Nivolumab and Pembrolizumab; and anti-inflammatory agents including glucocorticoids and non-steroidal anti-inflammatory drugs, e.g. cortisol-based preparations, Dexamethason, Betamethason, Prednisone, Prednisolone, Methylprednisolone, Triamcinolon-hexacetonid, Mometasonfuroat, Clobetasolpropionat, acetylsalicylic acid, salicylic acid and other salicylates, Diflunisal, Ibuprofen, Dexibuprofen, Naproxen, Fenoprofen, Ketoprofen, Dexketoprofen, Loxoprofen, Flurbiprofen, Oxaprozin, Indomethacin, Ketorolac, Tolmetin, Diclofenac, Etodolac, Aceclofenac, Nabumetone, Sulindac, Mefenamic acid, Meclofenamic acid, Flufenamic acid, Tolfenamic acid, Celecoxib, Parecoxib, Etoricoxib and Firocoxib; and ACE inhibitors; and beta-blockers; and myostatin inhibitors; and PDE-5 inhibitors; and antihistamines. For a combination therapy, the active ingredients may be formulated as compositions containing several active ingredients in a single dose form and/or as kits containing individual active ingredients in separate dose forms. The active ingredients used in combination therapy may be co-administered or administered separately.

The compounds of the invention may be administered as antibody-drug conjugates.

The compounds of the invention may be administered in combination with surgery, cryotherapy, electrodessication, radiotherapy, photodynamic therapy, laser therapy, chemotherapy, targeted therapy, immunotherapy, gene therapy, antisense therapy, cell-based transplantation therapy, stem cell therapy, physical therapy and occupational therapy.

The compounds of the invention falling under the scope of formula I, formula II, formula III, formula IV and formula V can be synthesized in analogy to the methods described in Reinmuller et al., 2015, EPFL Thesis 6887 by a coupling step to establish the diaryl ether scaffold, which can be prepared by a method of reacting a phenol and an electron-deficient aryl halide in the presence of a base such as potassium carbonate or cesium carbonate in a non-protic organic solvent such as DMSO or DMF at room temperature or at elevated temperature or reflux, preferably at 80° C. or 100° C., with optional assistance of microwave irradiation (Li et al., Org. Lett. 2003, 5, 2169-2171);

or by a method of reacting a phenol and a nitroarene in the presence of a base such as potassium carbonate or cesium carbonate in a non-protic organic solvent such as DMSO or DMF at elevated temperature or reflux, with assistance of microwave irradiation (Sarkate et al., Synlett 2013, 24, 1513-1516);

or by a method of reacting an aryl silyl ether with an electron-deficient aryl halide in the presence of a base such as DBU and trace water in a non-protic organic solvent such as DMSO or DMF at elevated temperature or reflux (Yeom et al., Synlett 2007, 146-150);

or by a method of reacting a phenol with a diaryliodonium triflate or tosylate in the presence of a base such as potassium carbonate or cesium carbonate in a non-protic organic solvent such as acetonitrile at ambient or elevated temperature (Kakinuma et al., Synthesis 2013, 45, 183-184);

or by a method of reacting under Buchwald-Hartwig conditions a phenol with an aryl halide in the presence of a transition metal-based catalyst system such as palladium(II) acetate, an organophosphorus-based ligand such as dppf, a base such as potassium phosphate in an organic solvent such as toluene at elevated temperature or reflux (Burgos et al., Angew. Chem. Int. Ed. 2006, 45, 4321-4326);

or by a method of reacting under Chan-Lam conditions a phenol with an arylboronic acid or ester in the presence of air, a copper-based catalyst system such as copper(II) acetate, a base such as pyridine or triethylamine in a non-protic organic solvent such as DCM, chloroform at ambient temperature (Evans et al., Tetrahedron Letters 1998, 39, 2937-2940);

wherein all said methods of preparation may require a subsequent derivatisation step by standard chemical procedures known to the person skilled in the art, such as saponification, hydrolysis, esterification or amidation to obtain the corresponding carboxylic acids, esters, primary amides, secondary amides, tertiary amides, hydroxamic acids and hydroxamates.

For example, the corresponding carboxylic acids are synthesized by saponification of the corresponding benzoate esters, fluorobenzoate esters, nicotinate esters, or fluoronicotinate esters in the presence of potassium hydroxide or sodium hydroxide in a binary solvent mixture of water and an alcohol, preferably ethanol, or water and tetrahydrofuran at ambient or elevated temperature (Becker et al., Organikum, 22nd edition 2004 (German), pp. 488, publisher: Wiley-VCH Weinheim);

the esters, primary amides, secondary amides, tertiary amides, and hydroxamic acids are synthesized by in situ transformation of the corresponding benzoic acid, fluorobenzoic acid, nicotinic acid, or fluoronicotinic acid to the corresponding acid chlorides in the presence of thionyl chloride and catalytic amounts of DMF in toluene at ambient or elevated temperature, preferably at 80° C., and under inert gas atmosphere, followed by the addition of the respective nucleophile, i.e. alcohol, ammonia, secondary amine, tertiary amine, or hydroxylamine in the presence or absence of a non-nucleophilic base such as triethylamine, at ambient temperature under inert gas atmosphere (Becker et al., Organikum, 22nd edition 2004 (German), pp. 459, publisher: Wiley-VCH Weinheim).

The perfluoroalkylcyclopropyl moiety associated with the compounds of the invention falling under the scope of formula V is synthesized in three steps according to the procedure described in Barnes-Seeman et al., ACS Med. Chem. Lett. 2013, 4, 514-516; first, a bromoperfluoroalkenylbenzene such as 1-bromo-4-(3,3,3-trifluoroprop-1-en-2-yl)benzene or 1-bromo-4-(3,3,4,4,4-pentafluorobut-1-en-2-yl)benzene is obtained by a method of reacting 1-(4-bromophenyl)-2,2,2-trifluoroethan-1-one or 1-(4-bromophenyl)-2,2,3,3,3-pentafluoropropan-1-one, respectively, in the presence of methanesulfonyl chloride and a base such as potassium fluoride in a crown ether such as 18-crown-6 in a non-protic organic solvent such as DMF at elevated temperature, preferably at 80° C.;

second, a bromophenylperfluoroalkyldihydropyrazole such as 3-(4-bromophenyl)-3-(trifluoromethyl)-4,5-dihydro-3H-pyrazole or 3-(4-bromophenyl)-3-(perfluoroethyl)-4,5-dihydro-3H-pyrazole is obtained by a method of reacting a bromoperfluoroalkenylbenzene such as 1-bromo-4-(3,3,3-trifluoroprop-1-en-2-yl)benzene or 1-bromo-4-(3,3,4,4,4-pentafluorobut-1-en-2-yl)benzene, respectively, in the presence of diazomethane in an ether such as diethyl ether or methyl tert-butyl ether at ambient temperature;

and third, the perfluoroalkylcyclopropylarylbromide such as 1-bromo-4-(1-(trifluoromethyl)cyclopropyl)benzene or 1-bromo-4-(1-(perfluoroethyl)cyclopropyl)benzene is obtained by a method of reacting 3-(4-bromophenyl)-3-(trifluoromethyl)-4,5-dihydro-3H-pyrazole or 3-(4-bromophenyl)-3-(perfluoroethyl)-4,5-dihydro-3H-pyrazole, respectively, in an organic solvent such as toluene or xylenes or a mixture thereof.

The obtained perfluoroalkylcyclopropylarylbromide can subsequently be converted into the corresponding phenol for one of the above said coupling reactions with an electron-deficient aryl halide, a nitroarene, a diaryliodonium triflate or tosylate by a method of reaction in the presence of a transition metal-based catalyst system such as Pd2dba3, an organophosphorus-based ligand such as 2-di-tert-butylphosphino-2′,4′,6′-triisopropylbiphenyl (t-Bu XPhos), a base such as potassium hydroxide or sodium hydroxide in a biphasic solvent system such as water/dioxane or water/toluene at elevated temperature or reflux, preferably at 100° C., and under an inert gas atmosphere (Anderson et al., J. Am. Chem. Soc. 2006, 128, 10694-10695);

or by a method of reaction in the presence of a copper-based catalyst system such as CuI, a pyridyl based ligand such as 2-methylquinolin-8-ol or preferably 8-hydroxyquinoline-N-oxide, and tetrabutyl-ammonium hydroxide or preferably cesium hydroxide monohydrate in a non-protic organic solvent such as DMSO or DMF at elevated temperature or reflux, preferably at 110° C., and under an inert gas atmosphere (Paul et al., Synthesis 2010, 4268-4272; Yang et el., Org. Lett. 2011, 13, 4340-4343).

The compounds of the invention falling under the scope of formula I, formula II, formula III, formula IV and formula V, as well as intermediates, can be isolated by column chromatography using silica gel as stationary phase and common organic solvents such as petroleum ether, ethyl acetate, dichloromethane, methanol, or acetic acids as eluent, preferably as binary or tertiary solvent mixtures thereof;

or by crystallization from common organic solvents such as petroleum ether, ethyl acetate, dichloromethane, chloroform, methanol, ethanol, toluene, or tert-butyl methyl ether, and mixtures thereof.

The compounds of the invention falling under the scope of formula I, formula II, formula III, formula IV and formula V, as well as starting materials and intermediates, can be identified by conventional methods such as nuclear magnetic resonance (NMR) spectroscopy, mass spectrometry (MS), or thin layer chromatography (TLC).

Chemical Synthesis

The compounds of the invention falling under the scope of formula I, formula II, formula III, formula IV and formula V can be synthesized and purified by those persons skilled in the art and are preferably synthesized according to the general procedure A, or general procedure B, or general procedure C, or general procedure D, respectively, and according to the detailed synthesis procedures described herein;

Abbreviations

Ac acetyl
BRSM based on recovered starting material (yield)
Bu butyl
δ chemical shift in parts per million (ppm)
dba dibenzylideneacetone
DCE 1,2-dichloroethane
DCM dichloromethane

DMF N,N-dimethylformamide

DMSO dimethyl sulfoxide
Et ethyl
ESI electron spray ionization
M mol/L
Me methyl
Ms methanesulfonyl
PE petroleum ether
TBAF tetrabutylammonium fluoride
THF tetrahydrofuran
TMS trimethylsilyl

General Procedure A: Synthesis of Diaryl Ether Esters

Diaryl ether esters according to formula I, formula III, and formula V can be prepared by nucleophilic aromatic substitution, e.g. by reaction of an alkyl 4-fluorobenzoate, or an alkyl 3,4-difluorobenzoate, or an alkyl 6-chloronicotinate, or an alkyl 6-chloro-5-fluoronicotinate, with a phenol derivative (nucleophile, see Table XIV) in the presence of a base like potassium carbonate in a solvent like dimethyl sulfoxide at a temperature between 80° C. and 150° C. and in an inert atmosphere such as argon.

General Procedure B: Synthesis of Diaryl Ether Acids

Diaryl ether acids according to formula I, formula III, and formula V can be prepared by saponification, e.g. by reaction of the corresponding diaryl ether esters with an aqueous base solution like sodium hydroxide (nucleophile, see Table XIV) in a solvent like ethanol, methanol, tetrahydrofuran or a mixture thereof at a temperature between room temperature and reflux.

General Procedure C: Synthesis of Diaryl Ether Esters

Diaryl ether esters according to formula I, formula III, and formula V can be prepared by esterification via the corresponding acid chloride, e.g by reaction of a diaryl ether acid with thionyl chloride in the presence of catalytic amounts of DMF in a solvent like toluene at a temperature between 50° C. and 100° C. and in an inert atmosphere such as argon. After removal of the volatiles, the such obtained acid chloride intermediate is reacted with the alcohol corresponding to the desired ester (nucleophile, see Table XIV) in the presence of an organic base like triethylamine at a temperature between 0° C. and room temperature and in an inert atmosphere such as argon.

Alternatively, diaryl ether esters according to formula I, formula III, and formula V can be prepared by esterification via the corresponding acid chloride, e.g. by reaction of a diaryl ether acid with thionyl chloride in the presence of the alcohol corresponding to the desired ester (nucleophile, see Table XIV), preferably as the solvent at a temperature between 50° C. and reflux.

General Procedure D: Synthesis of Diaryl Ether Amides

Diaryl ether amides according to formula II, formula IV, and formula V can be prepared by amidation via the corresponding acid chloride, e.g by reaction of a diaryl ether acid with thionyl chloride in the presence of catalytic amounts of DMF in a solvent like toluene at a temperature between 50° C. and 100° C. and in an inert atmosphere such as argon. After removal of the volatiles, the such obtained acid chloride intermediate is reacted with the amine corresponding to the desired amide (nucleophile, see Table XIV) in a solvent like methanol, ethanol, or tetrahydrofuran at a temperature between 0° C. and room temperature and in an inert atmosphere such as argon. The presence of an organic base like triethylamine is needed if the hydrochloride salt of the amine is used.

TABLE XIV List of Synthesized compounds Compound ESI Ion General Nucleophile used Number m/z Type Procedure in the General Procedure 002 241.17 [M − H] B NaOH 003 255.18 [M − H] B NaOH 004 269.18 [M − H] B NaOH 005 283.21 [M − H] B NaOH 019 307.28 [M − H] B NaOH 020 321.34 [M − H] B NaOH 023 243.09 [M + H]+ C methanol 024 257.10 [M + H]+ C methanol 025 271.11 [M + H]+ C methanol 026 285.13 [M + H]+ C methanol 027 299.20 [M + H]+ C methanol 029 271.11 [M + H]+ C methanol 030 299.20 [M + H]+ C methanol 041 311.21 [M + H]+ C methanol 043 323.24 [M + H]+ C methanol 044 337.25 [M + H]+ C methanol 045 362.32 [M + H]+ C methanol 048 257.11 [M + H]+ A 4-methylphenol 049 271.12 [M + H]+ A 4-ethylphenol 050 285.15 [M + H]+ A 4-n-propylphenol 051 299.21 [M + H]+ A 4-n-butylphenol 052 313.26 [M + H]+ A 4-n-pentylphenol 054 285.15 [M + H]+ A 4-isopropylphenol 056 311.17 [M + H]+ A 4-(trifluoromethyl)phenol 066 337.25 [M + H]+ A (±)-4-(bicyclo[2.2.1]heptan-2-yl)phenol (7:1 endo:exo) 067 351.26 [M + H]+ A (±)-4-(bicyclo[2.2.2]octan-2-yl)phenol 070 244.00 [M − H] B NaOH 071 258.09 [M − H] B NaOH 072 272.11 [M − H] B NaOH 073 286.16 [M − H] B NaOH 075 258.10 [M − H] B NaOH 086 308.25 [M − H] B NaOH 087 322.30 [M − H] B NaOH 091 244.07 [M + H]+ C methanol 092 258.09 [M + H]+ C methanol 093 272.11 [M + H]+ C methanol 094 286.15 [M + H]+ C methanol 095 300.19 [M + H]+ C methanol 097 272.11 [M + H]+ C methanol 099 300.19 [M + H]+ C methanol 101 298.09 [M + H]+ C methanol 110 312.19 [M + H]+ C methanol 112 324.22 [M + H]+ C methanol 113 338.24 [M + H]+ C methanol 114 364.29 [M + H]+ C methanol 117 272.12 [M + H]+ A 4-ethylphenol 118 286.16 [M + H]+ A 4-n-propylphenol 119 300.20 [M + H]+ A 4-n-butylphenol 120 314.24 [M + H]+ A 4-n-pentylphenol 122 286.16 [M + H]+ A 4-isopropylphenol 133 338.24 [M + H]+ A (±)-4-(bicyclo[2.2.1]heptan-2-yl)phenol (7:1 endo:exo) 134 352.28 [M + H]+ A (±)-4-(bicyclo[2.2.2]octan-2-yl)phenol 138 228.10 [M + H]+ D ammonia 139 242.10 [M + H]+ D ammonia 140 256.12 [M + H]+ D ammonia 141 270.11 [M + H]+ D ammonia 142 284.16 [M + H]+ D ammonia 144 256.12 [M + H]+ D ammonia 145 270.12 [M + H]+ D ammonia 146 284.16 [M + H]+ D ammonia 157 296.20 [M + H]+ D ammonia 159 308.22 [M + H]+ D ammonia 160 322.26 [M + H]+ D ammonia 161 348.27 [M + H]+ D ammonia 164 244.07 [M + H]+ D hydroxylamine 165 258.09 [M + H]+ D hydroxylamine 166 272.11 [M + H]+ D hydroxylamine 167 286.15 [M + H]+ D hydroxylamine 168 300.19 [M + H]+ D hydroxylamine 170 272.11 [M + H]+ D hydroxylamine 171 300.19 [M + H]+ D hydroxylamine 182 312.20 [M + H]+ D hydroxylamine 184 324.22 [M + H]+ D hydroxylamine 185 338.25 [M + H]+ D hydroxylamine 186 364.36 [M + H]+ D hydroxylamine 190 256.12 [M + H]+ D methylamine 191 270.12 [M + H]+ D methylamine 192 284.15 [M + H]+ D methylamine 193 298.21 [M + H]+ D methylamine 195 270.12 [M + H]+ D methylamine 196 284.15 [M + H]+ D methylamine 197 298.22 [M + H]+ D methylamine 208 310.23 [M + H]+ D methylamine 210 322.26 [M + H]+ D methylamine 211 336.28 [M + H]+ D methylamine 212 362.30 [M + H]+ D methylamine 215 256.12 [M + H]+ D dimethylamine 216 270.12 [M + H]+ D dimethylamine 217 284.16 [M + H]+ D dimethylamine 218 298.21 [M + H]+ D dimethylamine 219 312.23 [M + H]+ D dimethylamine 221 284.15 [M + H]+ D dimethylamine 222 298.21 [M + H]+ D dimethylamine 223 312.24 [M + H]+ D dimethylamine 234 324.27 [M + H]+ D dimethylamine 236 336.28 [M + H]+ D dimethylamine 237 350.30 [M + H]+ D dimethylamine 238 376.33 [M + H]+ D dimethylamine 241 229.09 [M + H]+ D ammonia 242 243.09 [M + H]+ D ammonia 243 257.11 [M + H]+ D ammonia 244 271.12 [M + H]+ D ammonia 245 285.15 [M + H]+ D ammonia 247 257.11 [M + H]+ D ammonia 248 285.16 [M + H]+ D ammonia 250 283.04 [M + H]+ D ammonia 259 297.17 [M + H]+ D ammonia 261 309.21 [M + H]+ D ammonia 262 323.26 [M + H]+ D ammonia 263 349.29 [M + H]+ D ammonia 266 245.07 [M + H]+ D hydroxylamine 267 259.09 [M + H]+ D hydroxylamine 268 273.11 [M + H]+ D hydroxylamine 269 287.14 [M + H]+ D hydroxylamine 270 301.18 [M + H]+ D hydroxylamine 272 273.12 [M + H]+ D hydroxylamine 273 301.19 [M + H]+ D hydroxylamine 275 299.08 [M + H]+ D hydroxylamine 284 313.20 [M + H]+ D hydroxylamine 286 325.21 [M + H]+ D hydroxylamine 287 339.24 [M + H]+ D hydroxylamine 288 365.29 [M + H]+ D hydroxylamine 291 243.10 [M + H]+ D methylamine 292 257.11 [M + H]+ D methylamine 293 271.12 [M + H]+ D methylamine 294 285.15 [M + H]+ D methylamine 295 299.21 [M + H]+ D methylamine 297 271.12 [M + H]+ D methylamine 298 299.21 [M + H]+ D methylamine 300 297.08 [M + H]+ D methylamine 309 311.22 [M + H]+ D methylamine 311 323.25 [M + H]+ D methylamine 312 337.26 [M + H]+ D methylamine 313 363.32 [M + H]+ D methylamine 316 257.11 [M + H]+ D dimethylamine 317 271.12 [M + H]+ D dimethylamine 318 285.15 [M + H]+ D dimethylamine 319 299.21 [M + H]+ D dimethylamine 320 313.24 [M + H]+ D dimethylamine 322 285.16 [M + H]+ D dimethylamine 323 313.25 [M + H]+ D dimethylamine 325 311.13 [M + H]+ D dimethylamine 334 325.26 [M + H]+ D dimethylamine 336 337.26 [M + H]+ D dimethylamine 337 351.28 [M + H]+ D dimethylamine 338 377.32 [M + H]+ D dimethylamine 341 245.13 [M − H] B NaOH 342 259.15 [M − H] B NaOH 343 273.17 [M − H] B NaOH 344 287.18 [M − H] B NaOH 345 301.21 [M − H] B NaOH 347 273.15 [M − H] B NaOH 348 301.24 [M − H] B NaOH 350 299.12 [M − H] B NaOH 359 313.24 [M − H] B NaOH 363 365.39 [M − H] B NaOH 374 317.22 [M + H]+ C methanol 385 329.24 [M + H]+ C methanol 389 381.34 [M + H]+ C methanol 392 275.09 [M + H]+ A 4-methylphenol 393 289.13 [M + H]+ A 4-ethylphenol 394 303.17 [M + H]+ A 4-n-propylphenol 395 317.23 [M + H]+ A 4-n-butylphenol 396 331.24 [M + H]+ A 4-n-pentylphenol 398 303.19 [M + H]+ A 4-isopropylphenol 399 331.25 [M + H]+ A 4-tert-pentylphenol 401 329.17 [M + H]+ A 4-(trifluoromethyl)phenol 410 343.28 [M + H]+ A 4-cyclohexylphenol 414 395.33 [M + H]+ A 4-(1-adamantyl)phenol 417 246.12 [M − H] B NaOH 423 274.15 [M − H] B NaOH 424 288.17 [M − H] B NaOH 425 304.16 [M + H]+ B NaOH 427 300.10 [M − H] B NaOH 436 314.24 [M − H] B NaOH 440 368.29 [M + H]+ B NaOH 451 318.21 [M + H]+ C methanol 462 330.23 [M + H]+ C methanol 466 382.29 [M + H]+ C methanol 469 276.08 [M + H]+ A 4-methylphenol 475 304.15 [M + H]+ A 4-isopropylphenol 476 318.20 [M + H]+ A 4-tert-butylphenol 477 332.24 [M + H]+ A 4-tert-pentylphenol 479 330.14 [M + H]+ A 4-(trifluoromethyl)phenol 488 344.24 [M + H]+ A 4-cyclohexylphenol 492 396.29 [M + H]+ A 4-(1-adamantyl)phenol 503 302.20 [M + H]+ D ammonia 514 314.20 [M + H]+ D ammonia 518 366.29 [M + H]+ D ammonia 529 318.21 [M + H]+ D hydroxylamine 540 330.23 [M + H]+ D hydroxylamine 544 382.30 [M + H]+ D hydroxylamine 555 316.23 [M + H]+ D methylamine 566 328.25 [M + H]+ D methylamine 570 380.31 [M + H]+ D methylamine 581 330.26 [M + H]+ D dimethylamine 592 342.28 [M + H]+ D dimethylamine 596 394.33 [M + H]+ D dimethylamine 607 303.19 [M + H]+ D ammonia 618 315.21 [M + H]+ D ammonia 622 367.36 [M + H]+ D ammonia 633 319.21 [M + H]+ D hydroxylamine 644 331.21 [M + H]+ D hydroxylamine 648 383.27 [M + H]+ D hydroxylamine 659 317.22 [M + H]+ D methylamine 670 329.25 [M + H]+ D methylamine 674 381.31 [M + H]+ D methylamine 685 331.25 [M + H]+ D dimethylamine 696 343.28 [M + H]+ D dimethylamine 700 395.31 [M + H]+ D dimethylamine 703 321.22 [M − H] B NaOH 704 371.30 [M − H] B NaOH 705 339.22 [M − H] B NaOH 711 355.21 [M − H] B NaOH 712 337.16 [M + H]+ C methanol 714 353.31 [M − H] C methanol 721 351.20 [M + H]+ A 4-(1-(trifluoromethyl)cyclopropyl)phenol 722 401.25 [M + H]+ A 4-(1-(perfluoroethyl)cyclopropyl)phenol 723 369.24 [M + H]+ A 4-(1-(trifluoromethyl)cyclopropyl)phenol 729 385.18 [M + H]+ A 2-chloro-4-(1-(trifluoromethyl)cyclo- propyl)phenol 730 322.22 [M − H] B NaOH 731 374.19 [M + H]+ B NaOH 732 340.21 [M − H] B NaOH 738 358.13 [M + H]+ B NaOH 739 338.17 [M + H]+ C methanol 740 388.21 [M + H]+ C methanol 741 356.19 [M + H]+ C methanol 747 372.15 [M + H]+ C methanol 748 352.21 [M + H]+ A 4-(1-(trifluoromethyl)cyclopropyl)phenol 749 402.25 [M + H]+ A 4-(1-(perfluoroethyl)cyclopropyl)phenol 750 370.20 [M + H]+ A 4-(1-(trifluoromethyl)cyclopropyl)phenol 756 386.17 [M + H]+ A 2-chloro-4-(1-(trifluoromethyl)cyclo- propyl)phenol 757 322.18 [M + H]+ D ammonia 759 340.18 [M + H]+ D ammonia 766 338.17 [M + H]+ D hydroxylamine 767 388.22 [M + H]+ D hydroxylamine 768 356.19 [M + H]+ D hydroxylamine 774 372.19 [M + H]+ D hydroxylamine 775 336.20 [M + H]+ D methylamine 777 354.19 [M + H]+ D methylamine 784 350.21 [M + H]+ D dimethylamine 785 400.26 [M + H]+ D dimethylamine 786 368.22 [M + H]+ D dimethylamine 792 384.23 [M + H]+ D dimethylamine 793 323.17 [M + H]+ D ammonia 794 373.21 [M + H]+ D ammonia 795 341.17 [M + H]+ D ammonia 801 357.14 [M + H]+ D ammonia 802 339.17 [M + H]+ D hydroxylamine 803 389.22 [M + H]+ D hydroxylamine 804 357.17 [M + H]+ D hydroxylamine 810 373.17 [M + H]+ D hydroxylamine 811 337.18 [M + H]+ D methylamine 812 387.23 [M + H]+ D methylamine 813 355.20 [M + H]+ D methylamine 819 371.17 [M + H]+ D methylamine 820 351.21 [M + H]+ D dimethylamine 821 401.26 [M + H]+ D dimethylamine 822 369.21 [M + H]+ D dimethylamine 828 385.21 [M + H]+ D dimethylamine

Synthesis of Representative Compounds Compound 005 4-(4-Pentylphenoxy)benzoic acid

Following general procedure B, to a solution of ethyl 4-(4-pentylphenoxy)benzoate (1.69 g, 5.4 mmol) in THF (25 mL) and MeOH (3 mL) was added 2 M aqueous NaOH (10 mL, 20 mmol) and the reaction was stirred at room temperature for 48 hours. The organic solvents were evaporated and the residue acidified with 5 M aqueous HCl to adjust a pH of 1-2. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by recrystallization from hot EtOAc to give the title compound as colorless solid (1.21 g, 79%). 1H NMR (300 MHz, CDCl3) δ 8.12-8.01 (m, 2H), 7.25-7.15 (m, 2H), 7.05-6.94 (m, 4H), 2.68-2.57 (m, 2H), 1.72-1.56 (m, 2H), 1.46-1.33 (m, 2H), 1.38-1.23 (m, 2H), 0.97-0.86 (m, 3H). 13C NMR (75 MHz, CDCl3) δ 171.9, 163.3, 153.2, 139.7, 132.5, 130.0, 123.3, 120.4, 117.0, 35.4, 31.6, 31.4, 22.7, 14.2. HRMS (C18H19O3): expected: 283.1339; found: 283.1326.

Compound 030 Methyl 4-(4-(tert-Pentyl)phenoxy)benzoate

Following general procedure C, to a solution of 4-(4-(tert-pentyl)phenoxy)benzoic acid (122 mg, 0.43 mmol) in MeOH (2 mL) was added SOCl2 (0.1 mL, 1.4 mmol) at 0° C. and the reaction was then stirred at 80° C. in a sealed vessel for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and quenched by the addition of saturated aqueous NaHCO3. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 90% PE-EtOAc gradient to give the title compound as colorless oil (120 mg, 94%). 1H NMR (300 MHz, CDCl3) δ 8.05-7.94 (m, 2H), 7.39-7.28 (m, 2H), 7.04-6.92 (m, 4H), 3.89 (s, 3H), 1.65 (q, J=7.4 Hz, 2H), 1.30 (s, 6H), 0.71 (t, J=7.4 Hz, 3H). 13C NMR (75 MHz, CDCl3) δ 166.8, 162.3, 153.1, 146.0, 131.8, 127.6, 124.3, 119.7, 117.2, 52.1, 37.8, 37.1, 28.7, 9.3. HRMS (C19H23O3+): expected: 299.1642; found: 299.1640.

Compound 044 (±)-Methyl 4-(4-(bicyclo[2.2.2]octan-2-yl)phenoxy)benzoate

Following general procedure C, to a solution of (±)-4-(4-(Bicyclo[2.2.2]octan-2-yl)phenoxy)benzoic acid (30.3 mg, 0.1 mmol) in toluene (1 mL) was added one drop of DMF followed by SOCl2 (0.02 mL, 0.3 mmol) at room temperature and the reaction was then stirred at 80° C. for 3.5 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. A solution of NEt3 (0.2 mL, 1.4 mmol) in MeOH (1 mL) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition of 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 85% PE-EtOAc gradient to give the title compound as colorless oil (29.3 mg, 93%). 1H NMR (300 MHz, CDCl3) δ 8.05-7.94 (m, 2H), 7.35-7.21 (m, 2H), 7.06-6.91 (m, 4H), 3.89 (s, 3H), 3.11-2.94 (m, 1H), 2.01 (dddd, J=12.9, 10.6, 3.9, 1.9 Hz, 1H), 1.85-1.45 (m, 10H), 1.43-1.23 (m, 1H). 13C NMR (75 MHz, CDCl3) δ 166.8, 162.3, 153.3, 143.1, 131.8, 129.3, 124.3, 120.0, 117.1, 52.1, 41.4, 32.6, 31.2, 27.6, 26.1, 25.4, 24.9, 20.6. HRMS (C22H25O3+): expected: 337.1798; found: 337.1778.

Compound 051 Ethyl 4-(4-butylphenoxy)benzoate

Following general procedure A, to 4-butylphenol (1.75 mL, 11.4 mmol) and K2CO3 (1.89 g, 13.7 mmol) in DMSO (18 mL) was added ethyl 4-fluorobenzoate (1.35 mL, 9.2 mmol) and the reaction was then stirred at 120° C. for 3 days in an argon atmosphere. The reaction was cooled to room temperature and quenched by the addition of water. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed first with 1 M aqueous NaOH (1×) then washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 70% PE-DCM gradient to give the title compound as colorless oil (1.72 g, 63%). 1H NMR (300 MHz, CDCl3) δ 8.05-7.94 (m, 2H), 7.24-7.13 (m, 2H), 7.02-6.91 (m, 4H), 4.36 (q, J=7.1 Hz, 2H), 2.67-2.56 (m, 2H), 1.69-1.53 (m, 2H), 1.47-1.23 (m, 5H), 0.95 (t, J=7.3 Hz, 3H). 13C NMR (75 MHz, CDCl3) δ 166.3, 162.3, 153.5, 139.4, 131.7, 130.0, 124.7, 120.1, 117.1, 60.9, 35.1, 33.8, 22.5, 14.5, 14.1. HRMS (C19H23O3+): expected: 299.1642; found: 299.1642.

Compound 071 6-(4-Propylphenoxy)nicotinic acid

Following general procedure B, to a solution of ethyl 6-(4-propylphenoxy)nicotinate (2.11 g, 7.4 mmol) in EtOH (15 mL) was added 2 M aqueous NaOH (10 mL, 20 mmol) and the reaction was stirred at room temperature for 48 hours. The reaction was acidified with 5 M aqueous HCl to adjust a pH of 1-2. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by recrystallization from hot EtOAc to give the title compound as colorless solid (1.26 g, 66%). 1H NMR (300 MHz, CDCl3) δ 11.01 (br, s, 1H), 8.92 (dd, J=2.4, 0.7 Hz, 1H), 8.31 (dd, J=8.7, 2.4 Hz, 1H), 7.28-7.18 (m, 2H), 7.12-7.02 (m, 2H), 6.94 (dd, J=8.7, 0.7 Hz, 1H), 2.61 (dd, J=8.7, 6.7 Hz, 2H), 1.76-1.58 (m, 2H), 0.97 (t, J=7.3 Hz, 3H). 13C NMR (75 MHz, CDCl3) δ 170.5, 167.4, 151.4, 151.2, 141.2, 140.2, 129.9, 121.3, 120.3, 110.9, 37.6, 24.6, 14.0. HRMS (C15H14NO3): expected: 256.0979; found: 256.0979.

Compound 114 Methyl 6-(4-(adamantan-1-yl)phenoxy)nicotinate

Following general procedure C, to a solution of 6-(4-(adamantan-1-yl)phenoxy)nicotinic acid (170 mg, 0.49 mmol) in MeOH (2 mL) was added SOCl2 (0.1 mL, 1.37 mmol) at room temperature and the reaction was then stirred at 80° C. in a sealed vessel for 3.5 hours in an argon atmosphere. The reaction was cooled to room temperature and quenched by the addition of saturated aqueous NaHCO3. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 90% PE-EtOAc gradient to give the title compound as colorless solid (47 mg, 27%). 1H NMR (300 MHz, CDCl3) δ 8.84 (dd, J=2.4, 0.7 Hz, 1H), 8.25 (dd, J=8.6, 2.4 Hz, 1H), 7.46-7.35 (m, 2H), 7.15-7.04 (m, 2H), 6.90 (dd, J=8.6, 0.7 Hz, 1H), 3.91 (s, 3H), 2.11 (p, J=3.0 Hz, 4H), 1.93 (d, J=2.9 Hz, 6H), 1.87-1.68 (m, 6H). 13C NMR (75 MHz, CDCl3) δ 166.7, 165.6, 150.9, 150.5, 148.4, 140.5, 126.3, 121.0, 120.7, 110.7, 52.2, 43.3, 36.8, 36.0, 29.0. HRMS (C23H26NO3+): expected: 364.1907; found: 364.1900.

Compound 117 Ethyl 6-(4-Ethylphenoxy)nicotinate

Following general procedure A, to 4-ethylphenol (1.36 g, 11.1 mmol) and K2CO3 (1.89 g, 13.7 mmol) in DMSO (18 mL) was added ethyl 6-chloronicotinate (1.65 mL, 10.9 mmol) and the reaction was then stirred at 80° C. for 48 hours in an argon atmosphere. The reaction was cooled to room temperature and quenched by the addition of water. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed first with 1 M aqueous NaOH (1×) then washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 60% PE-MeOH gradient to give the title compound as colorless oil (1.78 g, 60%). 1H NMR (300 MHz, CDCl3) δ 8.83 (dd, J=2.4, 0.7 Hz, 1H), 8.25 (dd, J=8.6, 2.4 Hz, 1H), 7.28-7.20 (m, 2H), 7.11-7.00 (m, 2H), 6.90 (dd, J=8.7, 0.8 Hz, 1H), 4.37 (q, J=7.1 Hz, 2H), 2.68 (q, J=7.6 Hz, 2H), 1.38 (t, J=7.1 Hz, 3H), 1.26 (t, J=7.6 Hz, 3H). 13C NMR (75 MHz, CDCl3) δ 166.8, 165.2, 151.3, 150.5, 141.4, 140.6, 129.3, 121.4, 121.3, 110.7, 61.2, 28.4, 15.6, 14.4. HRMS (C16H18NO3+): expected: 272.1281; found: 272.1271.

Compound 159 (±)-4-(4-(Bicyclo[2.2.1]heptan-2-yl)phenoxy)benzamide, mixture of endo and exo

Following general procedure D, to a solution of (±)-4-(4-(bicyclo[2.2.1]heptan-2-yl)phenoxy)benzoic acid (50.2 mg, 0.16 mmol) in toluene (0.8 mL) was added one drop of DMF followed by SOCl2 (0.04 mL, 0.55 mmol) at room temperature and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. 2 M ammonia in MeOH (0.6 mL, 1.3 mmol) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 10% PE-EtOAc gradient to give the title compound as colorless solid (46.6 mg, 93%, 6:1 mixture endo:exo). 1H NMR (300 MHz, CDCl3, mixture of rotamers 0.3:1, and diastereoisomers 6:1) δ 7.82-7.72 (m, 2H), 7.26-7.13 (m, 2H), 7.08-6.91 (m, 4H), 6.05 (s, 2H), 3.20 (major diastereomer, tt, J=14.0, 4.8 Hz, 0.85H), 2.79-2.70 (minor diastereomer, m, 0.15H), 2.46-2.29 (major diastereomer, m, 1.7H), 2.28-2.14 (minor diastereomer, m, 0.3H), 2.12-1.10 (m, 8H). 13C NMR (75 MHz, CDCl3, mixture of rotamers 0.3:1, and diastereoisomers 6:1) δ 169.0, 161.5, 161.4, 153.5, 153.3, 144.1, 140.1, 139.9, 129.8, 129.7, 129.5, 128.6, 127.5, 127.4, 119.9, 119.7, 119.6, 117.6, 117.5, 117.4, 50.3, 46.9, 46.6, 45.6, 43.6, 43.1, 42.7, 42.4, 42.2, 42.1, 41.6, 41.1, 40.7, 39.4, 37.7, 37.7, 37.0, 36.7, 36.4, 36.2, 34.6, 30.7, 30.3, 29.0, 24.7, 24.6, 23.0. HRMS (C20H22NO2+): expected: 308.1645; found: 308.1624.

Compound 186 4-(4-(Adamantan-1-yl)phenoxy)-N-hydroxybenzamide

Following general procedure D, to a solution of 4-(4-(adamantan-1-yl)phenoxy)benzoic acid (200 mg, 0.57 mmol) in toluene (2 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.37 mmol) at room temperature and the reaction was then stirred at 80° C. for 3.5 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. Hydroxylamine hydrochloride (208 mg, 3 mmol) in a solution of NEt3 (1.0 mL, 7.2 mmol) and MeOH (2 mL) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed first with 1 M aqueous HCl (1×) then washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 30% PE-EtOAc gradient to give the title compound as colorless solid (176 mg, 84%). 1H NMR (300 MHz, CDCl3/DMSO-d6) δ 11.10 (s, 1H), 8.87 (s, 1H), 7.80-7.69 (m, 2H), 7.37-7.26 (m, 2H), 6.98-6.86 (m, 4H), 2.09-2.01 (m, 4H), 1.85 (d, J=2.8 Hz, 6H), 1.81-1.63 (m, 6H). 13C NMR (75 MHz, CDCl3/DMSO-d6) δ 163.8, 159.7, 152.9, 146.7, 128.6, 126.7, 126.0, 118.9, 116.7, 42.6, 36.1, 35.3, 28.2. HRMS (C23H24NO3): expected: 362.1761; found: 362.1672.

Compound 195 4-(4-Isopropylphenoxy)-N-methylbenzamide

Following general procedure D, to a solution of 4-(4-isopropylphenoxy)benzoic acid (177 mg, 0.7 mmol) in toluene (2.5 mL) was added two drops of DMF followed by SOCl2 (0.15 mL, 2.1 mmol) at room temperature and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. 33 wt % methylamine in EtOH (2 mL, 16 mmol) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 40% PE-EtOAc gradient to give the title compound as colorless solid (178 mg, 94%). 1H NMR (300 MHz, CDCl3) δ 7.78-7.67 (m, 2H), 7.27-7.16 (m, 2H), 7.01-6.90 (m, 4H), 6.31 (s, 1H), 2.98 (d, J=4.6 Hz, 3H), 2.90 (hept, J=6.9 Hz, 1H), 1.26 (d, J=6.9 Hz, 6H). 13C NMR (75 MHz, CDCl3) δ 167.8, 160.8, 153.8, 145.1, 128.8, 128.8, 127.9, 119.8, 117.5, 33.6, 26.9, 24.2. HRMS (C17H18NO2): expected: 268.1343; found: 268.1384.

Compound 222 4-(4-(tert-Butyl)phenoxy)-N,N-dimethylbenzamide

Following general procedure D, to a solution of 4-(4-(tert-butyl)phenoxy)benzoic acid (154 mg, 0.57 mmol) in toluene (2.5 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.4 mmol) at room temperature and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. 2 M dimethylamine in THE (2.5 mL, 5 mmol) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 40% PE-EtOAc gradient to give the title compound as colorless oil (166 mg, 98%). 1H NMR (300 MHz, CDCl3) δ 7.44-7.31 (m, 4H), 7.03-6.91 (m, 4H), 3.06 (s, 6H), 1.33 (s, 9H). 13C NMR (75 MHz, CDCl3) δ 171.4, 159.1, 153.9, 147.0, 130.6, 129.2, 126.8, 119.2, 117.8, 39.9 (br), 35.6 (br), 34.5, 31.6. HRMS (C19H24NO2+): expected: 298.1802; found: 298.1820.

Compound 241 6-(p-Tolyloxy)nicotinamide

Following general procedure D, to a solution of 6-(p-tolyloxy)nicotinic acid (148 mg, 0.66 mmol) in toluene (2.5 mL) was added two drops of DMF followed by SOCl2 (0.12 mL, 1.6 mmol) at room temperature and the reaction was then stirred at 80° C. for 3.5 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. 2 M ammonia in MeOH (3 mL, 1.3 mmol) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 20% PE-EtOAc gradient to give the title compound as colorless solid (104 mg, 70%). 1H NMR (300 MHz, CDCl3/DMSO-d6, mixture of rotamers 0.3:1) δ 8.89 (d, J=2.5 Hz, 0.25H, minor rotamer), 8.65 (d, J=2.4 Hz, 0.75H, major rotamer), 8.25 (dt, J=8.6, 2.9 Hz, 1H), 7.96 (br, s, 1H), 7.49 (d, J=8.4 Hz, 0.25H, minor rotamer), 7.27 (br, s, 1H), 7.21 (d, J=8.2 Hz, 2H), 7.06-6.97 (m, 2H), 6.93 (d, J=8.6 Hz, 0.75H, major rotamer), 2.35 (s, 3H). 13C NMR (75 MHz, CDCl3/DMSO-d6, mixture of rotamers 0.3:1) S 166.6, 165.9 (minor rotamer), 165.4 (major rotamer), 153.3 (minor rotamer), 151.5 (major rotamer), 149.7 (minor rotamer), 148.0 (major rotamer), 139.5 (major rotamer), 138.9 (minor rotamer), 134.4, 130.3, 129.3 (minor rotamer), 125.2 (major rotamer), 124.1 (minor rotamer), 121.4, 110.5 (major rotamer), 20.9. HRMS (C13H13N2O2+): expected: 229.0972; found: 229.0978.

Compound 275 N-Hydroxy-6-(4-(trifluoromethyl)phenoxy)nicotinamide

Following general procedure D, to a solution of 6-(4-(trifluoromethyl)phenoxy)nicotinic acid (62.9 mg, 0.22 mmol) in toluene (1.5 mL) was added two drops of DMF followed by SOCl2 (0.06 mL, 0.82 mmol) at room temperature and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. Hydroxylamine hydrochloride (94 mg, 1.35 mmol) in a solution of NEt3 (0.5 mL, 3.6 mmol) and MeOH (1 mL) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCL. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 40% PE-EtOAc (+0.2% AcOH) gradient to give the title compound as colorless solid (42.7 mg, 65%). 1H NMR (300 MHz, CDCl3/DMSO-d6) δ 11.29 (s, 1H), 9.07 (s, 1H), 8.52 (d, J=2.4 Hz, 1H), 8.19 (dd, J=8.6, 2.4 Hz, 1H), 7.69 (d, J=8.4 Hz, 2H), 7.29 (d, J=8.4 Hz, 2H), 7.06 (d, J=8.5 Hz, 1H). 13C NMR (75 MHz, CDCl3/DMSO-d6) δ 163.6, 161.9, 156.2 (d, J=1.5 Hz), 146.4, 138.8, 126.6 (q, J=3.8 Hz), 125.6 (q, J=32.4 Hz), 124.2, 123.8 (q, J=273.0 Hz), 121.5, 111.1. HRMS (C13H8F3N2O3): expected: 297.0492; found: 297.0597.

Compound 284 6-(4-Cyclohexylphenoxy)-N-hydroxynicotinamide

Following general procedure D, to a solution of 6-(4-cyclohexylphenoxy)nicotinic acid (150 mg, 0.5 mmol) in toluene (2 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.4 mmol) at room temperature and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. Hydroxylamine hydrochloride (208 mg, 3 mmol) in a solution of NEt3 (1.0 mL, 7.2 mmol) and MeOH (2 mL) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 95% PE-EtOAc gradient to give the title compound as colorless solid (150 mg, 95%). 1H NMR (300 MHz, CDCl3/DMSO-d6) δ 11.27 (s, 1H), 9.05 (s, 1H), 8.54 (s, 1H), 8.20-8.06 (m, 1H), 7.23 (d, J=8.0 Hz, 2H), 7.02 (d, J=8.0 Hz, 2H), 6.95 (d, J=8.6 Hz, 1H), 1.86 (d, J=8.3 Hz, 4H), 1.74 (d, J=12.6 Hz, 1H), 1.43 (q, J=11.3, 10.1 Hz, 4H), 1.35-1.16 (m, 2H). 13C NMR (75 MHz, CDCl3/DMSO-d6) δ 163.4, 160.9, 149.8, 145.2, 142.8, 137.1, 126.2, 122.0, 119.5, 109.0, 41.9, 32.7, 25.0, 24.2. HRMS (C18H21N2O3+): expected: 313.1547; found: 313.1622.

Compound 297 6-(4-Isopropylphenoxy)-N-methylnicotinamide

Following general procedure D, to a solution of 6-(4-isopropylphenoxy)nicotinic acid (151 mg, 0.6 mmol) in toluene (2.5 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.5 mmol) at room temperature and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. 33 wt % methylamine in EtOH (2.5 mL, 20 mmol) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCL. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 20% PE-EtOAc gradient to give the title compound as colorless solid (150 mg, 96%). 1H NMR (300 MHz, CDCl3) δ 8.55 (dd, J=2.5, 0.7 Hz, 1H), 8.11 (dd, J=8.6, 2.5 Hz, 1H), 7.32-7.21 (m, 2H), 7.11-7.00 (m, 2H), 6.90 (dd, J=8.6, 0.7 Hz, 1H), 6.47 (d, J=5.3 Hz, 1H), 2.98 (d, J=4.7 Hz, 3H), 2.94 (hept, J=7.0 Hz, 1H), 1.27 (d, J=6.9 Hz, 6H). 13C NMR (75 MHz, CDCl3) δ 166.1, 165.8, 151.4, 146.6, 145.9, 139.0, 127.8, 125.3, 121.1, 111.0, 33.7, 26.9, 24.1. HRMS (C16H19N2O2+): expected: 271.1441; found: 271.1491.

Compound 322 6-(4-Isopropylphenoxy)-N,N-dimethylnicotinamide

Following general procedure D, to a solution of 6-(4-isopropylphenoxy)nicotinic acid (156 mg, 0.6 mmol) in toluene (2.5 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.5 mmol) at room temperature and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. 2 M dimethylamine in THF (2.5 mL, 5.4 mmol) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 30% PE-EtOAc gradient to give the title compound as colorless oil (169 mg, 98%). 1H NMR (300 MHz, CDCl3) δ 8.28 (dd, J=2.4, 0.8 Hz, 1H), 7.79 (dd, J=8.5, 2.4 Hz, 1H), 7.31-7.20 (m, 2H), 7.11-7.00 (m, 2H), 6.91 (dd, J=8.5, 0.7 Hz, 1H), 3.07 (s, 6H), 2.92 (hept, J=7.0 Hz, 1H), 1.26 (d, J=6.9 Hz, 6H). 13C NMR (75 MHz, CDCl3) δ 168.9, 164.5, 151.4, 146.8, 145.6, 139.2, 127.7, 126.6, 121.0, 111.0, 39.7 (br), 35.6 (br), 33.6, 24.1. HRMS (C17H21N2O2+): expected: 285.1598; found: 285.1643.

Compound 344 4-(4-(Butylphenoxy)-3-fluorobenzoic acid

Following general procedure B, to a solution of ethyl 4-(4-butylphenoxy)-3-fluorobenzoate (1.42 g, 4.5 mmol) in EtOH (9 mL) was added 2 M aqueous NaOH (5 mL, 10 mmol) and the reaction was stirred at room temperature overnight. 1 M aqueous HCl was added to adjust a pH of 1-2. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (2×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by recrystallization from hot EtOAc to give the title compound as colorless solid (0.56 g, 43%). 1H NMR (300 MHz, CDCl3) δ 11.18 (br, s, 1H), 7.91 (dd, J=11.0, 2.0 Hz, 1H), 7.82 (ddd, J=8.6, 2.0, 1.1 Hz, 1H), 7.26-7.15 (m, 2H), 7.04-6.88 (m, 3H), 2.68-2.57 (m, 2H), 1.70-1.54 (m, 2H), 1.47-1.23 (m, 2H), 0.95 (t, J=7.3 Hz, 3H). 13C NMR (75 MHz, CDCl3) δ 171.0 (d, J=2.5 Hz), 153.3, 152.9 (d, J=249.5 Hz), 150.8 (d, J=11.0 Hz), 139.7, 130.1, 127.3 (d, J=3.5 Hz), 124.3 (d, J=6.5 Hz), 119.4, 118.9 (d, J=19.9 Hz), 118.6 (d, J=1.4 Hz), 35.1, 33.8, 22.5, 14.1. HRMS (C17H16FO3): expected: 287.1089; found: 287.1062.

Compound 395 Ethyl 4-(4-butylphenoxy)-3-fluorobenzoate

Following general procedure A, to 4-butylphenol (1.75 mL, 11.4 mmol) and K2CO3 (1.90 g, 13.8 mmol) in DMSO (18 mL) was added ethyl 3,4-difluorobenzoate (1.37 mL, 9 mmol) and the reaction was then stirred at 80° C. for 24 hours in an argon atmosphere. The reaction was cooled to room temperature and quenched by the addition of water. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed first with 1 M aqueous NaOH (1×) then washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 80% PE-DCM gradient to give the title compound as colorless oil (1.80 g, 63%). 1H NMR (300 MHz, CDCl3) δ 7.84 (dd, J=11.2, 2.0 Hz, 1H), 7.75 (ddd, J=8.5, 2.0, 1.2 Hz, 1H), 7.23-7.12 (m, 2H), 7.01-6.88 (m, 3H), 4.37 (q, J=7.1 Hz, 2H), 2.67-2.55 (m, 2H), 1.68-1.52 (m, 2H), 1.46-1.23 (m, 5H), 0.94 (t, J=7.3 Hz, 3H). 13C NMR (75 MHz, CDCl3) δ 165.4 (d, J=2.6 Hz), 153.7, 153.0 (d, J=249.0 Hz), 149.5 (d, J=11.2 Hz), 139.3, 129.9, 126.4 (d, J=3.5 Hz), 126.0 (d, J=6.3 Hz), 119.0, 119.0 (d, J=1.4 Hz), 118.3 (d, J=19.9 Hz), 61.3, 35.1, 33.8, 22.5, 14.4, 14.1. HRMS (C19H22FO3+): expected: 317.1548; found: 317.1549.

Compound 451 Methyl 5-fluoro-6-(4-(tert-pentyl)phenoxy)nicotinate

Following general procedure C, to a solution of 5-fluoro-6-(4-(tert-pentyl)phenoxy)nicotinic acid (100 mg, 0.33 mmol) in MeOH (2 mL) was added SOCl2 (0.1 mL, 1.4 mmol) at 0° C. and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and quenched by the addition of saturated aqueous NaHCO3. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 90% PE-EtOAc gradient to give the title compound as colorless solid (30 mg, 29%). 1H NMR (300 MHz, CDCl3) δ 8.56 (d, J=1.9 Hz, 1H), 8.02 (dd, J=10.0, 1.9 Hz, 1H), 7.43-7.31 (m, 2H), 7.17-7.04 (m, 2H), 3.92 (s, 3H), 1.66 (q, J=7.4 Hz, 2H), 1.31 (s, 6H), 0.72 (t, J=7.4 Hz, 3H). 13C NMR (75 MHz, CDCl3) δ 164.8 (d, J=1.6 Hz), 155.7 (d, J=11.1 Hz), 150.4, 147.1 (d, J=261.8 Hz), 146.9, 144.4 (d, J=6.1 Hz), 127.4, 125.0 (d, J=17.0 Hz), 122.1 (d, J=1.7 Hz), 120.7, 52.6, 37.9, 37.1, 28.6, 9.3. HRMS (C18H21FNO3+): expected: 318.1500; found: 318.1555.

Compound 544 4-(4-(Adamantan-1-yl)phenoxy)-3-fluoro-N-hydroxybenzamide

Following general procedure D, to a solution of 4-(4-(adamantan-1-yl)phenoxy)-3-fluorobenzoic acid (110 mg, 0.3 mmol) in toluene (2 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.4 mmol) at room temperature and the reaction was then stirred at 80° C. for 3.5 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. NEt3 (0.7 mL, 5.1 mmol) and hydroxylamine hydrochloride (148 mg, 2.1 mmol) in MeOH (1.5 mL) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography twice, eluting first with a 100% to 40% PE-EtOAc gradient and then with a 100% to 97% DCM-MeOH gradient to give the title compound as colorless solid (60 mg, 52%). 1H NMR (300 MHz, CDCl3/DMSO-d6) δ 11.23 (s, 1H), 9.00 (s, 1H), 7.68 (dd, J=11.6, 2.0 Hz, 1H), 7.57 (ddd, J=8.5, 2.1, 1.1 Hz, 1H), 7.37-7.25 (m, 2H), 6.97 (t, J=8.4 Hz, 1H), 6.96-6.86 (m, 2H), 2.12-1.97 (m, 4H), 1.84 (d, J=2.9 Hz, 6H), 1.80-1.63 (m, 6H). 13C NMR (75 MHz, CDCl3/DMSO-d6) δ 162.4, 153.2, 152.4 (d, J=247.5 Hz), 146.6, 146.3 (d, J=11.1 Hz), 128.4 (d, J=5.7 Hz), 126.0, 123.5, 119.5, 117.4, 115.5 (d, J=19.6 Hz), 42.6, 36.1, 35.3, 28.2. HRMS (C23H23FNO3): expected: 380.1667; found: 380.1541.

Compound 644 6-(4-Cyclohexylphenoxy)-5-fluoro-N-hydroxynicotinamide

Following general procedure D, to a solution of 6-(4-cyclohexylphenoxy)-5-fluoronicotinic acid (120 mg, 0.4 mmol) in toluene (2.5 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.4 mmol) at room temperature and the reaction was then stirred at 80° C. for 3.5 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. Hydroxylamine hydrochloride (208 mg, 3 mmol) in a solution of NEt3 (1.0 mL, 7.2 mmol) and MeOH (2 mL) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 30% PE-EtOAc gradient to give the title compound as colorless solid (110 mg, 88%). 1H NMR (300 MHz, CDCl3/DMSO-d6) δ 11.32 (s, 1H), 9.12 (s, 1H), 8.28 (d, J=1.9 Hz, 1H), 8.06-7.94 (m, 1H), 7.26-7.15 (m, 2H), 7.08-6.97 (m, 2H), 1.88-1.75 (m, 4H), 1.75-1.64 (m, 1H), 1.48-1.31 (m, 4H), 1.31-1.13 (m, 2H). 13C NMR (75 MHz, CDCl3/DMSO-d6) δ 161.0, 153.2 (d, J=11.3 Hz), 150.4, 146.3 (d, J=259.6 Hz), 144.4, 140.6, 127.4, 124.5, 123.1 (d, J=16.7 Hz), 120.7, 43.2, 34.0, 26.2, 25.5. HRMS (C18H18FN2O3): expected: 329.1307; found: 329.1279.

Compound 703 4-(4-(1-(Trifluoromethyl)cyclopropyl)phenoxy)benzoic acid

Following general procedure B, to a solution of ethyl 4-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy)benzoate (0.86 g, 2.5 mmol) in EtOH (20 mL) was added 2 M aqueous NaOH (10 mL, 20 mmol) and the reaction was stirred at room temperature overnight. 1 M aqueous HCl was added to adjust a pH of 1-2. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (2×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 75% PE-EtOAc (+0.2% AcOH) gradient to give the title compound as slightly yellow solid (0.75 g, 95%). 1H NMR (300 MHz, CDCl3) δ 11.15 (br, s, 1H), 8.15-8.04 (m, 2H), 7.54-7.43 (m, 2H), 7.09-6.98 (m, 4H), 1.38 (dd, J=6.7, 5.1 Hz, 2H), 1.07-1.01 (m, 2H). 13C NMR (75 MHz, CDCl3) δ 171.8, 162.3, 155.7, 133.2, 132.6, 132.5, 126.5 (q, J=273.4 Hz), 124.0, 119.9, 117.8, 27.8 (q, J=33.7 Hz), 10.0 (q, J=2.4 Hz). HRMS (C17H12F3O3): expected: 321.0744; found: 321.0712.

Compound 712 Methyl 4-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy)benzoate

Following general procedure C, to a solution of 4-(4-(1-(Trifluoromethyl)cyclopropyl)phenoxy)benzoic acid (112 mg, 0.35 mmol) in toluene (2 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.4 mmol) at room temperature and the reaction was then stirred at 80° C. for 3.5 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. A solution of NEt3 (0.6 mL, 4.4 mmol) in MeOH (1.2 mL) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCL. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 85% PE-EtOAc gradient to give the title compound as colorless oil (176 mg, 93%). 1H NMR (300 MHz, CDCl3) δ 8.07-7.96 (m, 2H), 7.52-7.41 (m, 2H), 7.07-6.95 (m, 4H), 3.90 (s, 3H), 1.41-1.31 (m, 2H), 1.09-0.97 (m, 2H). 13C NMR (75 MHz, CDCl3) δ 166.7, 161.4, 156.0, 133.1, 132.2, 131.9, 126.4 (q, J=273.0 Hz), 125.0, 119.7, 117.9, 52.2, 27.8 (q, J=33.6 Hz), 10.0 (q, J=2.5 Hz). HRMS (C18H16F3O3+): expected: 337.1046; found: 337.1036.

Compound 729 Ethyl 4-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenoxy)-benzoate

Following general procedure A, to 2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenol (245 mg, 1.5 mmol) and K2CO3 (220 mg, 1.6 mmol) in DMSO (2 mL) was added ethyl 4-fluorobenzoate (0.15 mL, 1.1 mmol) and the reaction was then stirred at 120° C. for 2 days in an argon atmosphere. K2CO3 (220 mg, 1.6 mmol) was added and the reaction was then stirred at 150° C. for 9 hours in an argon atmosphere. The reaction was cooled to room temperature and quenched by the addition of water. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 70% PE-DCM gradient to give the title compound as yellow oil (100 mg, 18%). 1H NMR (300 MHz, CDCl3) δ 8.08-7.97 (m, 2H), 7.58 (d, J=2.1 Hz, 1H), 7.41-7.27 (m, 1H), 7.03 (d, J=8.4 Hz, 1H), 7.03-6.88 (m, 2H), 4.36 (q, J=7.1 Hz, 2H), 1.44-1.33 (m, 5H), 1.11-1.00 (m, 2H). 13C NMR (75 MHz, CDCl3) δ 166.0, 160.7, 151.3, 133.9, 133.8, 131.7, 131.1, 126.2, 126.0 (q, J=273.4 Hz), 125.5, 121.6, 116.8, 60.9, 27.6 (q, J=33.1 Hz), 14.4, 10.0 (q, J=2.3 Hz). HRMS (C19H17ClF3O3+): expected: 385.0813; found: 385.0796.

Compound 730 6-(4-(1-(Trifluoromethyl)cyclopropyl)phenoxy)nicotinic acid

Following general procedure B, to a solution of ethyl 6-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy)nicotinate (1.47 g, 4.2 mmol) in EtOH (20 mL) was added 2 M aqueous NaOH (10 mL, 20 mmol) and the reaction was stirred at room temperature overnight. 1 M aqueous HCl was added to adjust a pH of 1-2. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (2×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 75% PE-EtOAc (+0.2% AcOH) gradient to give the title compound as colorless solid (1.21 g, 90%). 1H NMR (300 MHz, CDCl3/DMSO-d6) δ 12.16 (s, 1H), 8.80 (d, J=2.4 Hz, 1H), 8.30 (dd, J=8.6, 2.3 Hz, 1H), 7.51 (dd, J=8.1, 1.6 Hz, 2H), 7.19-7.08 (m, 2H), 6.97 (d, J=8.6 Hz, 1H), 1.41-1.30 (m, 2H), 1.18-1.02 (m, 2H). 13C NMR (75 MHz, CDCl3/DMSO-d6) δ 166.1, 165.3, 152.9, 149.8, 140.5, 132.2, 132.2, 125.9 (q, J=273.1 Hz), 121.8, 120.7, 110.5, 27.1 (q, J=33.5 Hz), 9.3 (q, J=2.4 Hz). HRMS (C16H13F3NO3+): expected: 324.0842; found: 324.0847.

Compound 749 Ethyl 6-(4-(1-(perfluoroethyl)cyclopropyl)phenoxy)nicotinate

Following general procedure A, to 4-(1-(perfluoroethyl)cyclopropyl)phenol (330 mg, 1.3 mmol) and K2CO3 (305 mg, 2.2 mmol) in DMSO (2.7 mL) was added ethyl 6-chloronicotinate (0.2 mL, 1.3 mmol) and the reaction was then stirred at 80° C. for 3 days in an argon atmosphere. The reaction was cooled to room temperature and quenched by the addition of water. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed first with 1 M aqueous NaOH (1×) then washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 90% PE-EtOAc gradient to give the title compound as yellow oil (445 mg, 88%). 1H NMR (300 MHz, CDCl3) δ 8.83 (dd, J=2.4, 0.7 Hz, 1H), 8.28 (dd, J=8.6, 2.4 Hz, 1H), 7.48 (d, J=8.6 Hz, 2H), 7.14-7.07 (m, 2H), 6.93 (dd, J=8.6, 0.7 Hz, 1H), 4.38 (q, J=7.1 Hz, 2H), 1.43-1.34 (m, 5H), 1.12-1.04 (m, 2H). 13C NMR (75 MHz, CDCl3) δ 166.14, 165.10, 153.39, 150.44, 140.84, 133.21, 133.12, 121.86, 121.20, 111.12, 61.31, 25.88 (t, J=23.9 Hz), 14.42, 10.02 (t, J=4.0 Hz). The two multiplets of the CF2 (tq) and the CF3 (qt) are too weak to be resolved. HRMS (C19H17F5NO3+): expected: 402.1123; found: 402.1124.

Compound 784 N,N-Dimethyl-4-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy)-benzamide

Following general procedure D, to a solution of 4-(4-(1-(Trifluoromethyl)cyclopropyl)phenoxy)benzoic acid (101 mg, 0.3 mmol) in toluene (2 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.4 mmol) at room temperature and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. 2 M dimethylamine in THF (1.2 mL, 2.5 mmol) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 15% PE-EtOAc gradient to give the title compound as colorless oil (110 mg, 100%). 1H NMR (300 MHz, CDCl3) δ 7.48-7.37 (m, 4H), 7.06-6.92 (m, 4H), 3.06 (d, J=9.8 Hz, 6H), 1.39-1.28 (m, 2H), 1.07-0.95 (m, 2H). 13C NMR (75 MHz, CDCl3) δ 171.2, 158.2, 156.7, 133.0, 131.6, 131.4, 129.3, 126.5 (q, J=272.1 Hz), 119.0, 118.6, 39.8 (br), 35.6 (br), 27.7 (q, J=33.7 Hz), 10.0 (q, J=2.5 Hz). HRMS (C19H19F3NO2+): expected: 350.1363; found: 350.1351.

Compound 820 N,N-Dimethyl-6-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy)-nicotinamide

Following general procedure D, to a solution of 6-(4-(1-(trifluoromethyl)cyclopropyl)phenoxy)nicotinic acid (162 mg, 0.5 mmol) in toluene (2 mL) was added two drops of DMF followed by SOCl2 (0.1 mL, 1.4 mmol) at room temperature and the reaction was then stirred at 80° C. for 3 hours in an argon atmosphere. The reaction was cooled to room temperature and the volatiles evaporated on a rotary evaporator. 2 M dimethylamine in THE (2.5 mL, 5 mmol) was added and the reaction was stirred at room temperature overnight in an argon atmosphere. The reaction was quenched by the addition 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 90% to 30% PE-EtOAc gradient to give the title compound as colorless solid (172 mg, 100%). 1H NMR (300 MHz, CDCl3) δ 8.28 (dd, J=2.4, 0.8 Hz, 1H), 7.82 (dd, J=8.5, 2.4 Hz, 1H), 7.54-7.44 (m, 2H), 7.16-7.06 (m, 2H), 6.96 (dd, J=8.5, 0.8 Hz, 1H), 3.18-2.96 (m, 6H), 1.40-1.29 (m, 2H), 1·10-0.98 (m, 2H). 13C NMR (75 MHz, CDCl3) δ 168.8, 163.9, 153.5, 146.7, 139.3, 132.7, 132.7, 127.1, 126.3 (q, J=274.0 Hz), 121.0, 111.4, 39.7 (br), 35.63 (br), 27.7 (q, J=33.6 Hz), 9.8 (q, J=2.5 Hz). HRMS (C18H18F3N2O2+): expected: 351.1315; found: 351.1293.

Synthesis of Intermediates (±)-4-(Bicyclo[2.2.1]heptan-2-yl)phenol, mixture of endo and exo

To a solution of 4-acetoxystyrene (3 mL, 20 mmol) in dicyclopentadiene (3 mL, 22 mmol) was added hydroquinone (10 mg, 0.1 mmol). The reaction vessel was purged with argon and sealed. The reaction mixture was stirred at 160° C. for 24 h. The reaction mixture was filtered through silica and washed with DCM. The solution was concentrated in vacuo and used in the next step without further purification.

The resulting oil was dissolved in EtOAc (40 mL). Under an argon atmosphere, palladium on charcoal (5% Pd, 0.2 g, 0.1 mmol) was added and the reaction vessel was flushed with H2. The reaction was stirred strongly for 22 h at room temperature. The reaction mixture was then purged back with argon, filtered through celite, washed with EtOAc, and concentrated in vacuo. The crude mixture was then filtered on silica (PE/EtOAc), concentrated in vacuo and used in the next step without further purification.

The resulting oil was dissolved in EtOH (40 mL), and 2 M aqueous NaOH (20 mL, 40 mmol) was added. The reaction mixture was stirred for 17 h at room temperature. The reaction was quenched with 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with 100% to 0% PE-DCM gradient. Recrystallization in hot PE afforded the title compound as white needles (1.5 g, 40% over three steps, 7:1 mixture endo:exo)

(±)-4-(Bicyclo[2.2.2]octan-2-yl)phenol

To a solution of 4-acetoxystyrene (3 mL, 20 mmol) in cyclohexadiene (2.1 mL, 22 mmol) was added hydroquinone (10 mg, 0.1 mmol). The reaction vessel was purged with argon and sealed. The reaction mixture was stirred at 160° C. for 24 h. The reaction mixture was filtered through silica and washed with DCM. The solution was concentrated in vacuo and used in the next step without further purification.

The resulting oil was dissolved in EtOAc (40 mL). Under an argon atmosphere, palladium on charcoal (5% Pd, 0.2 g, 0.1 mmol) was added and the reaction vessel was flushed with H2. The reaction was stirred strongly for 22 h. The reaction mixture was then purged back with argon, filtered through celite, washed with EtOAc, and concentrated in vacuo. The crude mixture was then filtered on silica (PE/DCM), concentrated in vacuo and used in the next step without further purification.

The resulting oil was dissolved in EtOH (40 mL), and 2 M aqueous NaOH (20 mL, 40 mmol) was added. The reaction mixture was stirred for 17 h at room temperature. The reaction was quenched with 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with 100% to 0% PE-DCM gradient. Recrystallization in hot PE afforded the title compound as white needles (0.6 g, 15% over three steps)

4-(1-(Trifluoromethyl)cyclopropyl)phenol

Following a procedure from Anderson, K. W. et al., J. Am. Chem. Soc., 2006, 128 (33), 10694-10695, to a solution of KOH (2.6 g, 46.3 mmol), Pd2dba3 (278 mg, 0.30 mmol), and di-tert-butyl-[2-(2,4,6-triisopropylphenyl)phenyl]phosphate (510 mg, 1.20 mmol) in degassed 1,4-dioxane (7.5 mL) and water (7.5 mL) under argon was added 1-bromo-4-(1-(trifluoromethyl)cyclopropyl)benzene (3.98 g, 15.0 mmol). The reaction vessel was then sealed and immerged in a pre-heated oil bath at 100° C. The reaction was stirred for 4-10 h. The reaction was quenched with 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with 100% to 90% PE-EtOAc gradient to give the title compound as a yellow oil (3.0 g, 99%).

2-Chloro-4-(1-(trifluoromethyl)cyclopropyl)phenol

To a solution of 4-(1-(trifluoromethyl)cyclopropyl)phenol (1.03 g, 5.1 mmol) in DCE (25 mL) under argon at 0° C. were added N-chlorosuccinimide (737 mg, 5.52 mmol) and aluminium trichloride (740 mg, 5.55 mmol). The reaction mixture was stirred at 0° C. for 3 h, before being quenched with water. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with 100% to 80% PE-EtOAc gradient to give the title compound as a yellow oil (380 mg, 31%).

6-Chloro-5-fluoronicotinic acid

To a solution of 2-chloro-3-fluoro-5-methylpyridine (512 mg, 3.52 mmol) in pyridine (2.5 mL) and water (2.5 mL) was added one portion of potassium permanganate (1.1 g, 6.9 mmol). The reaction mixture was heated to 100° C. Two more equal portion of potassium permanganate (for a total of 3.3 g, 20.7 mmol) were added after respectively 1 h and 2 h of stirring at 100° C. When needed, the solid accumulated in the condenser were washed down with water and pyridine. After another 1 h of stirring at 100° C., the reaction mixture was cooled down to room temperature. The reaction mixture was quenched with saturated aqueous Na2S2O3 and stirred 30 minutes. The mixture was filtered, then acidified to pH 2 with HCl 5 M. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with 90% to 70% PE-EtOAc gradient to give the title compound as a white solid (300 mg, 49%).

6-Chloro-5-fluoronicotinic acid ethyl ester

To a solution of 6-chloro-5-fluoronicotinic acid (5.1 g, 29.1 mmol) in EtOH (150 mL) at 0° C. was added SOCl2 (4.5 mL, 61.7 mmol). The mixture was heated at reflux for 4 h. The reaction mixture was allowed to cool down to room temperature, and the reaction was quenched with saturated aqueous NaHCO3. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with 100% to 80% PE-EtOAc gradient to give the title compound as a white solid (5.28 mg, 89%).

1-(4-Bromophenyl)-N-cyclohexylethan-1-imine

Following a procedure from Mercadante, M. A., et al., Chemical Science, 2014, 5, 3983-3994, to a solution of 4′-bromoacetophenone (10.0 g, 50.2 mmol) and p-toluenesulfonic acid monohydrate (100 mg, 0.53 mmol) in toluene (70 mL) was added cyclohexylamine (6.1 mL, 53.5 mmol) and the mixture was stirred at reflux with a Dean-Stark for 21 h. The reaction mixture was allowed to cool down to room temperature and PE was added (100 mL). The p-toluenesulfonic acid precipitated and could be filtered off. The solid was washed with PE (2×). The filtrate was concentrate in vacuo to afford crude product that was recrystallized from hot PE to give the title compound as slightly yellow flakes (12.4 g, 88%).

(Iodomethyl)dimethylphenylsilane

Following a procedure from Mercadante, M. A., et al., Chemical Science, 2014, 5, 3983-3994, to a solution of (chloromethyl)dimethylphenylsilane (4.9 mL, 27 mmol) in acetone (30 mL) was added sodium iodide (7.1 g, 47.3 mmol). The reaction mixture was then stirred at reflux for 19 h. The mixture was concentrated in vacuo, filtered over celite, and the solid washed with PE (60 mL). The solution was concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with 100% to 70% PE-DCM gradient to give the title compound as a yellow oil (7.1 g, 95%).

1-(4-Bromophenyl)-3-(dimethyl(phenyl)silyl)propan-1-one

Following a procedure from Mercadante, M. A., et al., Chemical Science, 2014, 5, 3983-3994, to a solution of 1-(4-bromophenyl)-N-cyclohexylethan-1-imine (5.6 g, 20 mmol) in THF (10 mL) at 0° C. was slowly added freshly prepared LDA in THF (approximatively 1.5 M, 15 mL, 22 mmol) dropwise. The mixture was stirred 1 h at 0° C. before adding (iodomethyl)dimethylphenylsilane (6.1 g, 22 mmol). The reaction was stirred for another 1 h at 0° C. before quenching with a buffer aqueous solution of sodium acetate (29.5 g, 360 mmol), acetic acid (10.3 mL, 180 mmol) in water (11 mL). The mixture was stirred for 15 minutes before being diluted with water. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with 100% to 95% PE-EtOAc gradient to give the title compound as a yellow solid (5.24 g, 75%).

Trimethyl(perfluoroethyl)silane

A solution of n-BuLi (2.3 M in cyclohexane, 9 mL, 20.7 mmol) in THF (40 mL) was stirred at −90° C. (Acetone/N2). The system was purged with an atmosphere of pentafluoroethane and the system was kept between −78° C. and −90° C. for 1 h, then slowly warmed to −65° C. and stirred for another 0.5 h. A solution of TMSCl (2.55 mL, 20 mmol) in THF (5 mL) was added and the mixture was allowed to warm-up slowly in the acetone bath and stirred for 15 h at room temperature. The solution was then distilled to obtain the title compound as a solution in THF (65 mL).

4-(1-(Perfluoroethyl)cyclopropyl)phenol

Following a procedure from Mercadante, M. A., et al., Chemical Science, 2014, 5, 3983-3994, to the solution of trimethyl(perfluoroethyl)silane in THF previously obtained (60 mL) at 0° C. was added 1-(4-bromophenyl)-3-(dimethyl(phenyl)silyl)propan-1-one (4.9 g, 14.2 mmol). The mixture was stirred for 10 minutes and TBAF (1 M solution in THF, 0.14 mL, 0.14 mmol) was added and the reaction mixture was stirred at room temperature for 7.5 h. The reaction mixture was cooled down to 0° C., water (1.4 mL) and TBAF (1 M solution in THF, 1.4 mL, 1.4 mmol) were added and the reaction mixture was stirred at room temperature for 14 h. The reaction was quenched with water. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 100% to 95% PE-EtOAc gradient to give 3-(4-bromophenyl)-5-(dimethyl(phenyl)silyl)-1,1,1,2,2-pentafluoropentan-3-ol as a mixture with the starting 1-(4-bromophenyl)-3-(dimethyl(phenyl)silyl)propan-1-one (4.9 g, 1:1 ratio by NMR) due to similar polarity.

To a solution of the previous alcohol/ketone mixture (4.5 g, containing approximatively 5.5 mmol of 3-(4-bromophenyl)-5-(dimethyl(phenyl)silyl)-1,1,1,2,2-pentafluoropentan-3-ol) in THF (25 mL) at 0° C. was added NaH (60 wt % in oil, 565 mg, 14.1 mmol). The mixture was stirred at room temperature for 45 minutes. The reaction was cooled down to 0° C. and MsCl (0.9 mL, 11.6 mmol) was added dropwise. After stirring at room temperature for 2 h, the reaction mixture was cooled down to 0° C. and quenched with water.

The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with water, saturated aqueous NaHCO3, brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. To the resulting oil at 0° C. was added a mixture of pyridine (0.9 mL, 11.2 mmol) and 1,1,1,3,3,3-Hexafluoropropan-2-ol (8 mL). The flask was sealed and the reaction mixture was stirred for 12.5 h. The reaction was quenched with water. The aqueous layer was extracted with PE (3×). The combined organics were washed with aqueous HCl 1 M, water, saturated aqueous NaHCO3 and brine (1×), dried over Na2SO4, filtered and concentrated in vacuo (water bath at 25° C., no lower than 200 mbar, the desired product is volatile). The residue was purified by silica gel flash chromatography eluting with 100% PE to give 1-bromo-4-(1-(perfluoroethyl)cyclopropyl)benzene. As it is a highly volatile product, the PE was not fully removed and the product was directly subjected to the next step. By further eluting the column with 9:1 PE/EtOAc, 1.7 g of the starting 1-(4-bromophenyl)-3-(dimethyl(phenyl)silyl)propan-1-one was recovered.

Following a procedure from Anderson, K. W. et al., J. Am. Chem. Soc., 2006, 128 (33), 10694-10695, to a solution of KOH (900 m16.0 mmol), Pd2dba3 (93 mg, 0.10 mmol), and di-tert-butyl-[2-(2,4,6-triisopropylphenyl)phenyl]phosphate (170 mg, 0.40 mmol) in degassed 1,4-dioxane (2 mL) and water (2 mL) under argon was added 1-bromo-4-(1-(perfluoroethyl)cyclopropyl)benzene (obtained in the previous step) in 1,4-dioxane (0.5 mL) and water (0.5 mL). The reaction vessel was then sealed and immerged in a pre-heated oil bath at 100° C. The reaction was stirred for 4-10 h. The reaction was quenched with 1 M aqueous HCl. The aqueous layer was extracted with EtOAc (3×). The combined organics were washed with brine (1×), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with 100% to 90% PE-EtOAc gradient to give the title compound as a yellow oil (1.04 g, 29% over 4 steps, 44% BRSM).

The compounds listed in Table XIV have been identified by TLC using pre-coated silica TLC sheets and common organic solvents such as petroleum ether, ethyl acetate, dichloromethane, methanol, or acetic acids as eluent, preferably as binary or tertiary solvent mixtures thereof, UV light at a wavelength of 254 or 366 nm, and/or common staining solutions such as phosphomolybdic acid, potassium permanganate, or ninhydrin.

The compounds listed in Table XIV have furthermore been identified by mass spectrometry using formic acid in the mobile phase for detection of positive ions, while no additive was used for negative ions. Ammonium Carbonate was used if the molecule was difficult to ionize. Representative compounds have also been identified by nuclear magnetic resonance spectroscopy. Chemical shifts (S) were reported in parts per million (ppm) relative to residual solvent peaks rounded to the nearest 0.01 ppm for proton and 0.1 ppm for carbon (ref.: CHCl3 [1H: 7.26 ppm, 13C: 77.2 ppm], DMSO [1H: 2.50 ppm, 13C: 39.5 ppm]). Coupling constants (1) were reported in Hz to the nearest 0.1 Hz. Peak multiplicity was indicated as follows: s (singlet), d (doublet), t (triplet), q (quartet), hept (heptet), m (multiplet), and br (broad).

Claims

1. A compound for use in the treatment of immune system-related disorders such as disorders of the hematopoietic system including the hematologic system, such as malignancies of the myeloid lineage, malignant and non-malignant disorders of the skin and mucosa, such as squamous and basal cell carcinoma, actinic keratosis, and hyperproliferative disorders of the skin and mucosa, e.g. cornification disorders, malignant and non-malignant disorders of the muscle, including hyperproliferative disorders of the muscle, such as muscle hyperplasia and muscle hypertrophy, disorders of the neuroendocrine system, such as medullary thyroid cancer, and hyperproliferative disorders of the genitourinary tract, e.g. cervical cancer,

wherein the compound is selected from
(i) a compound of formula I,
wherein X is CH or N,
R1=C1-C12 preferably C1-C6 alkyl, C2-C12 preferably C2-C6 alkenyl, C2-C12 preferably C2-C6 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, C4-C12 bicycloalkyl, C6-C12 bicycloalkenyl, C5-C14 tricycloalkyl,
wherein all alkyl, alkenyl and alkynyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein all cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein all alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues can be perhalogenated, particularly perfluorinated;
and wherein R1 is preferably selected from methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, iso-propyl, tert-butyl, tert-pentyl, 3-pentyl, —CF3, —CF2CF3, —(CF2)2CF3, —(CF2)3CF3, —CH(CF3)2, —CF(CF3)2, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, bicyclo[2.2.2]octyl, adamantyl, and 9-methylbicyclo[3.3.1]nonyl;
R2=H, C1-C6 alkyl, C3-C6 cycloalkyl,
wherein all alkyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein all cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein all alkyl and cycloalkyl residues can be perhalogenated, particularly perfluorinated;
and wherein R2 is preferably selected from H, methyl and ethyl,
or a salt or solvate thereof,
(ii) a compound of formula II,
wherein X and R1 are defined as in formula I, including the preferred definition of R1,
R3=H, C1-C6 alkyl, or C3-C6 cycloalkyl,
wherein all alkyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein all cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein all alkyl and cycloalkyl residues can be perhalogenated, particularly perfluorinated;
and wherein R3 is preferably H or methyl;
R4=H, C1-C6 alkyl, C3-C6 cycloalkyl, OH or OC1-C6 alkyl,
wherein all alkyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein all cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein all alkyl and cycloalkyl residues can be perhalogenated, particularly perfluorinated;
and wherein R4 is preferably H, OH or methyl,
or a salt or solvate thereof,
(iii) a compound of formula III,
wherein X, R1 and R2 are defined as in formula I, including the preferred definitions of R1 and R2,
or a salt or solvate thereof,
(iv) a compound of formula IV,
wherein X and R1 are defined as in formula I, including the preferred definition of R1,
and R3 and R4 are defined as in formula II, including the preferred definitions of R3 and R4,
or a salt or solvate thereof,
(v) a compound of formula V,
wherein n=0-5, which comprises cyclopropyl (n=0), cyclobutyl (n=1), cyclopentyl (n=2), cyclohexyl (n=3), cycloheptyl (n=4) and cyclooctyl (n=5),
wherein the said cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl groups can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein the said cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl groups can be perhalogenated, particularly perfluorinated;
and wherein n is preferably 0 as constituting cyclopropyl, particularly as constituting cyclopropyl being unsubstituted;
R5=C1-C12 preferably C1-C6 alkyl, C2-C12 preferably C2-C6 alkenyl, C2-C12 preferably C2-C6 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl,
wherein all alkyl, alkenyl and alkynyl residues can be linear or branched, and are perhalogenated, particularly perfluorinated,
and wherein all cycloalkyl and cycloalkenyl residues are perhalogenated, particularly perfluorinated;
or wherein all alkyl, alkenyl and alkynyl residues can be linear or branched, and can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
and wherein all cycloalkyl and cycloalkenyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I, —CN, —NCO, —NCS; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein R5 is preferably —CF3 or —CF2CF3;
R6-R9 are independently from each other selected from —H, —F, —Cl, —Br, —I, linear or branched C1-C4 alkyl, linear or branched C2-C4 alkenyl, C2-C4 alkynyl, C3-C5 cycloalkyl, and wherein all alkyl, alkenyl, alkynyl and cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from: —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
wherein R6-R8 each are preferably H, and R9 is preferably —H, —F, —Cl, or —CH3;
Y=a six-membered aromatic ring selected from benzene, pyridine, pyrimidine, pyridazine or pyrazine;
wherein the benzene ring is not substituted, or it is substituted with one to four of the substituents independently selected from R10-R13,
and wherein the pyridine ring is not substituted, or it is substituted at the carbon positions with one to three of the substituents independently selected from R10-R12, and wherein preferably the N-atom of the pyridine ring is in ortho-position relative to the ether bond,
and wherein the pyrimidine ring is not substituted, or it is substituted at the carbon positions with one or two of the substituents independently selected from R10-R11, and wherein preferably an N-atom of the pyrimidine ring is in ortho-position relative to the ether bond,
and wherein the pyridazine ring is not substituted, or it is substituted at the carbon positions with one or two of the substituents independently selected from R10-R11, and wherein preferably an N-atom of the pyridazine ring is in ortho-position relative to the ether bond,
and wherein the pyrazine ring is not substituted, or it is substituted at the carbon positions with one or two of the substituents independently selected from R10-R11, and wherein preferably an N-atom of the pyrazine ring is in ortho-position relative to the ether bond,
wherein preferably Y=benzene or pyridine being not substituted with any of the residues selected from R10-R13, or being substituted with one of the substituents selected from R10-R13 being —F at the carbon atom in ortho-position relative to the ether bond;
R10-R13 are independently from each other selected from —F, —Cl, —Br, —I, linear or branched C1-C4 alkyl, linear or branched C2-C4 alkenyl, C2-C4 alkynyl, C3-C5 cycloalkyl, and wherein all alkyl, alkenyl, alkynyl and cycloalkyl residues can be unsubstituted or substituted with one or more substituents in particular independently selected from —F, —Cl, —Br, —I; and C1-C3 alkyl such as —CH3 optionally halogenated or perhalogenated, particularly perfluorinated such as —CF3; and OC1-C3 alkyl optionally halogenated or perhalogenated, particularly perfluorinated;
Z=O or S, and preferably Z=O;
R14=OR2 or NR3R4
wherein R2 is defined as in formula I including the preferred definition of R2 as H, methyl or ethyl;
wherein R3 and R4 are defined as in formula II, including the preferred definitions of R3 as H or —CH3 and R4 as H, OH or —CH3,
or a salt or solvate thereof.

2. A compound of formula I as defined in claim 1 or a salt or solvate thereof, with the proviso that compounds listed in Table Ia are excluded.

3. The compound of claim 2 as shown in Table Ib, or a salt or solvate thereof.

4. A compound of formula II as defined in claim 1 or a salt or solvate thereof, with the proviso that compounds listed in Table IIa are excluded.

5. The compound of claim 4 as shown in Table IIb, or a salt or solvate thereof.

6. A compound of formula III as defined in claim 1 or a salt or solvate thereof, with the proviso that compounds listed in Table IIIa are excluded.

7. The compound of claim 6 as shown in Table IIIb, or a salt or solvate thereof.

8. A compound of formula IV as defined in claim 1 or a salt or solvate thereof.

9. The compound of claim 8 as shown in Table IV, or a salt or solvate thereof.

10. A compound of formula V as defined in claim 1 or a salt or solvate thereof.

11. The compound of claim 10 as shown in Table V, or a salt or solvate thereof.

12. The compound of any one of claims 2-11 for use in medicine, e.g. in human medicine or veterinary medicine.

13. The compound of any one of claims 2-11 for use in the treatment of disorders associated with, accompanied by and/or caused by dysfunctional Notch signaling.

14. The compound of any one of claims 2-11 for use as an enhancer of Notch signaling.

15. The compound of any one of claims 2-11 for use in the treatment of hyperproliferative disorders, including malignant and non-malignant hyperproliferative disorders.

16. The compound of any one of claims 2-11 for use in the treatment of disorders of the skin, mucosa, skin and mucosal appendages, cornea, and epithelial tissues, including non-melanoma skin cancer and precancerous lesions, skin and/or mucosal disorders with cornification defects and/or abnormal keratinocyte proliferation, skin and/or mucosal diseases associated with, accompanied by and/or caused by viral infections, atopic dermatitis and acne and in the promotion of wound healing of the skin and mucosa.

17. The compound of any one of claims 2-11 for use in immune system-related therapeutic applications including the treatment of disorders of the hematopoietic system including the hematologic system, immunotherapy or as a vaccine adjuvant.

18. The compound of any one of claims 2-11 for use in the treatment of muscular diseases including muscular dystrophies, or in muscle regeneration, or in hyperproliferative disorders of the muscle.

19. The compound of any one of claims 2-11 for use in the treatment of disorders of the neuroendocrine system including small cell carcinomas, large cell carcinomas and carcinoid tumors.

20. The compound of any one of claims 2-11 for use in the treatment of cancers or precancerous lesions of the brain, pancreas, liver, thyroid, genitourinary tract and endothelial tissue.

21. A method of treating a hyperproliferative disorder comprising administering a subject in need thereof, particularly a human subject, a therapeutically effective amount of a compound according to any one of claims 2-11.

22. A method of treating a disorder associated with, accompanied by and/or caused by dysfunctional Notch signaling, comprising administering a subject in need thereof, particularly a human subject, a therapeutically effective amount of a compound according to any one of claims 2-11.

Patent History
Publication number: 20220348535
Type: Application
Filed: Feb 26, 2018
Publication Date: Nov 3, 2022
Patent Grant number: 11591289
Applicant: XENIOPRO GMBH (Kelkheim (Taunus))
Inventor: Viktoria REINMÜLLER (Allschwil)
Application Number: 16/488,458
Classifications
International Classification: C07C 233/65 (20060101); C07C 65/24 (20060101); C07C 69/94 (20060101); C07D 213/80 (20060101);