US Patent Application for COMPOUNDS AND THEIR USES Patent Application (Application No. 20230138480, Issued May 4, 2023) (2023)

BOTTOM

The invention relates to compounds useful for modulating BRG1- or BRM-associated factor (BAF) complexes. In particular, the invention relates to compounds useful in the treatment of disorders associated with the function of the BAF complex.

Chromatin regulation is essential for gene expression, and ATP-dependent chromatin remodeling is one mechanism by which this gene expression occurs. The human chromatin remodeling switch/sucrose non-fermentable complex (SWI/SNF), also known as the BAF complex, has two SWI2-type ATPases known as BRG1 (Brahma-related gene 1) and BRM (Brahma). The transcriptional activator BRG1, also known as the ATP-dependent chromatin remodeler SMARCA4, is encoded by the SMARCA4 gene on chromosome 19. BRG1 is overexpressed in some cancers and is required for cancer cell proliferation. BRM, also known as the putative global transcription activator SNF2L2 and/or the ATP-dependent chromatin remodeler SMARCA2, is encoded by the SMARCA2 gene on chromosome 9 and has been shown to be essential for cancer cell growth in cells characterized by loss of mutations in BRG1 is a function. Inactivation of BRG and/or BRM results in downstream effects on cells, including cell cycle arrest and tumor suppression.

SUMMARY

The present invention relates to compounds useful for modulating a BAF complex. In some embodiments, the compounds are useful in treating disorders associated with an alteration in a BAF complex, such as a disorder associated with an alteration in one or both of the BRG1 and BRM proteins. The compounds according to the invention can be used alone or in combination with other pharmaceutical active substances for the treatment of such diseases.

A compound having the structure of formula I is disclosed herein:

in what

Are you

C optionally substituted2-DO9Heteroaryl or C2-DO9Heterocyclyl;

m is 0, 1, 2, 3 or 4.

R is H

was R1optionally replaced by C1-DO3C optionally substituted alkyl3-DO6C cycloalkyl optionally substituted6-DO10Aryl, optionally C substituted2-DO9Heteroaryl, hydroxy, optionally substituted C1-DO6alkoxy or -NR1aR1b, where Ria and R, respectively1bare independently H or CH3;

each R2is independently halogen, optionally substituted C;1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted1-DO6C alkyl3-DO10Carbocyclyl, optionally C substituted1-DO6Heteroalcohol C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C1-DO6C alkyl2-DO9Heterocyclyl optionally substituted C1-DO6Heteroalcohol C2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted1-DO6C alkyl6-DO10Aryl, optionally C substituted1-DO6Heteroalcohol C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C1-DO6C alkyl2-DO9Heteroaryl optionally substituted C1-DO6Heteroalcohol C2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6optionally substituted heteroalkenyl, hydroxy, thiol or amino.

each R3and R.S5is independently selected from the group consisting of H, optionally substituted C;1-DO6alkyl or optionally substituted C1-DO6Heteroalcohol;

R4is hydrogen, optionally substituted C1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted1-DO6C alkyl3-DO10Carbocyclyl, optionally C substituted1-DO6Heteroalcohol C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C1-DO6C alkyl2-DO9Heterocyclyl optionally substituted C1-DO6Heteroalcohol C2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted1-DO6C alkyl6-DO10Aryl, optionally C substituted1-DO6Heteroalcohol C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C1-DO6C alkyl2-DO9Heteroaryl or optionally substituted C1-DO6Heteroalcohol C2-DO9Heteroaryl; m

this is

where X is N or CRX, in this RXis H, halogen, optionally substituted C;1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C2-DO6alkenyl or optionally substituted C2-DO6Heteroalkenyl;

Y is N or CRY, where RY is H, halogen, optionally substituted C1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C2-DO6alkenyl or optionally substituted C2-DO6Heteroalkenyl;

Z is S, O or NRG, in this RGH, C is optionally substituted1-DO6alkyl or optionally substituted C1-DO6Heteroalcohol;

R6and R.S7are independently H, halogen, optionally C substituted1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted1-DO6optionally substituted C alkyl6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6Heteroalkenyl or optionally substituted amino or two R's7combine with the carbon atoms to which they are attached, forming a 5- or 6-membered ring. is

n is 1, 2, 3 or 4,

or a pharmaceutically acceptable salt thereof,

where if B is

and R.S4is hydrogen, then R6is H, halogen, optionally substituted C;1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6Heteroalkenyl, optionally substituted amino or C6-DO10Aryl optionally substituted with one or more substituents selected from optionally substituted C1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C6-DO10Aryl, optionally C substituted1-DO6Acyl and cyano or two substituents combine with the carbon atoms to which they are attached to form a 5- or 6-membered heterocycle.

In some embodiments, B

In some embodiments, R1is not1aR1b, in which each R1aand R.S1bare independently H or CH3.

In some embodiments, the compound of formula I has the structure of formula II:

was R1aand R.S1bare independently H or CH3or a pharmaceutically acceptable salt thereof.

In some embodiments, m is 0.

In some embodiments, the compound of formula II has the structure of formula IIa:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula II has the structure of formula IIb:

or a pharmaceutically acceptable salt thereof.

In some embodiments, A

In some embodiments, the compound of formula II has the structure of formula IIc:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula II has the structure of formula IId:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula II has the structure of formula IIe:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula II has the structure of formula IIf:

where p is 0, 1, 2, 3, 4 or 5. and each R8independently optionally replaced by C1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C1-DO6Acyl, optionally substituted C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl, cyano, hydroxy, thiol or optionally substituted amino or two R8combine with the carbon atoms to which they are attached to form a 5- or 6-membered heterocycle or a pharmaceutically acceptable salt thereof.

In some embodiments, p is 0.

In some embodiments, the compound of formula II has the structure of formula IIg:

or a pharmaceutically acceptable salt thereof.

In some embodiments, A

In some embodiments, the compound of formula II has the structure of formula IIh:

or a pharmaceutically acceptable salt thereof.

In some embodiments, n is 1. In some embodiments, A is

In some forms R7is

In some embodiments, n is 0.

In some embodiments, the compound of formula II has the structure of formula IIi:

or a pharmaceutically acceptable salt thereof.

In some embodiments, X is N. In some embodiments, X is CH.

In some embodiments, each R1aand R.S1bis chosen independently of the group consisting of H or CH3. In some embodiments, both R1aand R.S1bis H. In some embodiments both are R1aand R.S1bis CH3. In some embodiments, R1ais H and R1bis CH3. In some embodiments, both R3and R.S5is X

In some embodiments, R1optionally replaced by C1-DO3for rent.

In some embodiments, the compound of formula I has the structure of formula III:

was R1optionally replaced by C1-DO3alkyl or a pharmaceutically acceptable salt thereof.

In some embodiments, m is 0.

In some embodiments, the compound of formula III has the structure of formula IIIa:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula III has the structure of formula IIIb:

or a pharmaceutically acceptable salt thereof.

In some embodiments, A is Z

In some embodiments, the compound of formula III has the structure of formula IIIc:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula III has the structure of formula IIId:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula III has the structure of formula IIIe:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula III has the structure of formula IIIf:

where p is 0, 1, 2, 3, 4 or 5. is

each R8independently optionally replaced by C1-DO6C optionally substituted alkyl1-DO6

Heteroalkyl, cyan, optionally C substituted1-DO6Acyl, optionally substituted C6-DO10Aryl, optionally C substituted2-DO9optionally substituted heteroaryl, hydroxy, thiol or amino or a pharmaceutically acceptable salt thereof.

In some embodiments, p is 0.

In some embodiments, the compound of formula III has the structure of formula IIIg:

or a pharmaceutically acceptable salt thereof.

In some embodiments, A

In some embodiments, the compound of formula III has the structure of formula IIIh:

or a pharmaceutically acceptable salt thereof.

In some embodiments, n is 0.

In some embodiments, the compound of formula III has the structure of formula IIIi:

or a pharmaceutically acceptable salt thereof.

In some embodiments, R1is CH3, Isopropyl or Cyclopropyl.

In some embodiments, B is optionally substituted with C2-DO9Heteroaryl or C2-DO9heterocyclyl. In some embodiments, B

In some embodiments, both R3and R.S5is X

In some embodiments, R4is hydrogen,

In some embodiments, R4is

In some embodiments, R4is hydrogen.

In some embodiments, the compound is any of compounds 1-40 or 88-105 in Table 1, or a pharmaceutically acceptable salt thereof.

TABLE 1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105

In another aspect, the invention provides a compound having the structure of formula IV:

was R10is C1-DO6for rent,

C optionally substituted1-DO6Aminoalkyl optionally substituted C1-DO4C alkyl6-DO10Aryl, optionally C substituted3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl or optionally substituted C2-DO9Heteroaryl;

each R9and R.S11is independently selected from the group consisting of H, optionally substituted C;1-DO6alkyl or optionally substituted C1-DO6Heteroalcohol; m

R12is H,C2-DO6C optionally substituted alkyl3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl, hydroxy, thiol, optionally substituted C1-DO6Alkoxy optionally substituted C1-DO6Thioalkoxy or optionally substituted amino or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula IV has the structure of formula IVa:

or a pharmaceutically acceptable salt thereof.

In some embodiments, both R9and R.S11is H. In some embodiments, R is10is

H,

In some embodiments, the compound is any of compounds 41-51 in Table 2, or a pharmaceutically acceptable salt thereof.

MESA 2 41 42 43 44 45 46 47 48 49 50 51

In another aspect, the invention provides a compound having the structure of formula V,

was R14is hydrogen, optionally substituted C1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl or optionally substituted C2-DO9Heteroaryl;

each R13and R.S15is independently selected from the group consisting of H, optionally substituted C;1-DO6alkyl or optionally substituted C1-DO6Heteroalcohol;

R16is H or C1-DO6Rent;

r is 0, 1, 2, 3 or 4.

Rsiis H,C1-DO6Perfluoralquil, C2-DO6C optionally substituted alkyl3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6Heteroalkenyl, optionally substituted acyl, hydroxy, thiol, nitrile, optionally substituted C1-DO6Thioalkoxy, optionally substituted sulfone, optionally substituted sulfonamide, optionally substituted C1-DO6alkylamine or optionally substituted amino.

each RONEit is, however, C2-DO6C optionally substituted alkyl1-DO6Hydroxyalkyl optionally substituted C1-DO6Aminoalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6heteroalkenyl, optionally substituted acyl, hydroxy, thiol, optionally substituted thioalkoxy or optionally substituted amino.

s is 0, 1, 2, 3 or 4.

RHeyH, C is optionally substituted3-DO10Carbocyclyl, optionally C substituted2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6Heteroalkenyl, optionally substituted acyl, hydroxy, thiol or optionally substituted C1-DO6Thioalkyl or RHeyand R.Sdocombine with the carbon atoms to which they are attached to form a 5- or 6-membered heterocycle.

each Rdoit is, however, C2-DO6C optionally substituted alkyl3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6Heteroalkenyl, optionally substituted acyl, hydroxy, thiol, optionally substituted C1-DO6thioalkyl or optionally substituted amino or RHeyand R.Sdocombine with the carbon atoms to which they are attached to form a 5- or 6-membered heterocycle.

if r is 0 then r14is hydrogen, optionally substituted C1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl or optionally substituted C2-DO9Heteroaryl;

see Rsiis H,RHeyis H and s is 0, then r is 1, 2, 3, or 4. is

see Rsiis H,RHeyis H and r is 0, so s is 1, 2, 3 or 4,

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula V has the structure of formula Va:

or a pharmaceutically acceptable salt thereof.

In some embodiments, R16is SH.

In some embodiments, the compound of formula V has the structure of formula Vb:

or a pharmaceutically acceptable salt thereof.

In some embodiments, RHeyis H and s is 0.

In some embodiments, the compound of formula V has the structure of formula Vc:

or a pharmaceutically acceptable salt thereof.

In some embodiments, Rsiis H. In some embodiments, r is 1;

In some embodiments, the compound of formula V has the structure of formula Vd:

or a pharmaceutically acceptable salt thereof.

In some embodiments, RONEoptionally replaced by C1-DO6Hydroxyalkyl optionally substituted C1-DO6Aminoalkyl or optionally substituted acyl. In some embodiments, RONEis -OH, -NH2, -CH3, — CF3, -CH2OH, -CH2NH2,

In some embodiments, Rsiis C2-DO6Alkyl (eg methyl), optionally substituted sulfone (eg -SO).2CH3), C was optionally substituted1-DO6Alkylamine (z. B. -CH2NH2, -CH2N(CH3)2, or -CH2NHC(O) ONLY.3), optionally substituted amino (eg -NH).2, – NHS ONLY3, ou-NHSO2CH3) or optionally substituted acyl (for example -C(O)NHCH3). In some embodiments, Rsiis -OH, -CH3, -E3, — CF3, or -CH2OH.

In some embodiments, the compound is any of compounds 52-87 in Table 3, or a pharmaceutically acceptable salt thereof.

TISCH 3 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87

In another aspect, the invention relates to a compound of formula VI having the structure:

in what

each R16and R.S18is independently selected from the group consisting of H, optionally substituted C;1-DO6alkyl or optionally substituted C1-DO6Heteroalcohol;

R17optionally replaced by C1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl or optionally substituted C2-DO9Heteroaryl;

D is

    • was X1is N or CRX1, in this RX1optionally substituted H or C;1-DO6Rent;
    • Y1is N or CRA'1, in this RA'1H, C is optionally substituted1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6Heteroalkenyl;
    • Z é S, O, C(RZ1a)2, or notZ1b, in this RZ1aH, C is optionally substituted1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6Heteroalkenyl and RZ1bH, C is optionally substituted1-DO6alkyl or optionally substituted C1-DO6Heteroalcohol;
    • q is 0, 1, 2, 3, 4 or 5.
    • each R19is H, halogen, optionally substituted C;1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted2-DO9Heteroaryl optionally substituted C2-DO6Alkenyl optionally substituted C2-DO6heteroalkenyl or optionally substituted amino.
    • each R20is independently halogen, optionally substituted C;1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO10Carbocyclyl, optionally C substituted2-DO9Heterocyclyl optionally substituted C6-DO10Aryl, optionally C substituted2-DO9heteroaryl, hydroxy, thiol, optionally substituted sulfone, optionally substituted sulfonamide, or optionally substituted amino. is

And it is

C optionally substituted2-DO9Heteroaryl or optionally substituted C2-DO9Heterocyclyl,

    • in what
    • r is 0, 1, 2, 3, 4 or 5. is
    • each R21is independently halogen, optionally substituted C;1-DO6C optionally substituted alkyl6-DO10Aryl, optionally C substituted2-DO9Heteroaryl, hydroxy, thiol, optionally substituted sulfone, optionally substituted sulfonamide, or optionally substituted amino, or a pharmaceutically acceptable salt thereof.

In another aspect, the invention relates to a compound having the structure of formula VII:

was R17, R18, R19, and R.S21are independently optionally substituted H or C;1-DO6Rent;

R20is hydrogen, optionally substituted C1-DO6alkyl or optionally substituted C1-DO6Heteroalcohol;

R22optionally replaced by C6-DO10Aryl, optionally C substituted2-DO9Heterocyclyl or optionally substituted C2-DO9Heteroaryl; m

X1and X2are independently N or CH,

or a pharmaceutically acceptable salt thereof.

In some embodiments, R19is hydrogen. In some embodiments, R21is hydrogen.

In some embodiments, X is1is N. In some embodiments, X is1is CH In some embodiments, X is2is n.

In some embodiments, X is2is CH

In some embodiments, R17is hydrogen. In some embodiments, R17optionally replaced by C1-DO6Alkyl (z. B. Methyl).

In some embodiments, R18optionally replaced by C1-DO6Alkyl (z. B. Methyl).

In some embodiments, R20is hydrogen. In some embodiments, R20optionally replaced by C1-DO6Heteroalcohol

In some embodiments, R20optionally replaced by C1-DO6Alkyl (z. B. Methyl).

In some embodiments, R22optionally replaced by C2-DO9Heterocyclyl

where the dashed line represents an optional double bond. and R23optionally replaced by C1-DO6acilo or C2-DO9heteroaryl). In some embodiments, R22is

In some embodiments, R22optionally replaced by C2-DO9Heteroaryl (e.g. optionally substituted C2-DO9bicyclisches Heteroaryl, wie z

In some embodiments, R22optionally replaced by C6-DO10aryl

where n is 0, 1, 2, 3 or 4. R24is hydrogen, optionally substituted C1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO8Carbocyclyl, optionally C substituted6-DO10Aryl, optionally C substituted2-DO9Heterocyclyl optionally substituted C2-DO9Heteroaryl; by R25is cyan, hydroxy, optionally substituted C1-DO6Heteroalkyl, where R is when n is 0;24optionally replaced by C1-DO6C optionally substituted alkyl1-DO6Heteroalkyl optionally substituted C3-DO8Carbocyclyl, optionally C substituted6-DO10Aryl, optionally C substituted2-DO9Heterocyclyl optionally substituted C2-DO9Heteroaril).

In some embodiments, n is 1. In some embodiments, at least one is R25it is blue. In some embodiments, at least one R25is hydroxy. In some embodiments, at least one R25optionally replaced by C1-DO6Heteroalkyl (i.e. Methoxy oder

In some embodiments, n is 0. In some embodiments, R24is hydrogen. In some embodiments, R24optionally replaced by C1-DO6Heteroalcohol

In some embodiments, R24optionally replaced by C1-DO6for rent

In some embodiments, R24optionally replaced by C3-DO8Carbocycline

In some embodiments, R24optionally replaced by C6-DO10Aryl (z. B. Phenyl, 4-Aminomethylphenyl,

In some embodiments, R24optionally replaced by C2-DO9Heterocyclyl

In some embodiments, R24optionally replaced by C2-DO9Heteroaril

In some embodiments, the compound is any of compounds 106-241 in Table 4, or a pharmaceutically acceptable salt thereof.

TABLE 4 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241

In another aspect, the invention relates to a pharmaceutical composition containing any of the above compounds and a pharmaceutically acceptable excipient.

In some embodiments, the pharmaceutical composition comprises a compound of formula I, formula IV, formula V, formula VI or formula VII and a pharmaceutically acceptable excipient.

In some embodiments, the compound is one of compounds 1-241 in Tables 1, 2, 3 and 4.

In another aspect, the invention relates to a method of reducing the activity of a BAF complex in a cell, the method comprising contacting the cell with an effective amount of any of the above compounds or a pharmaceutical composition thereof.

In some embodiments, the cell is a cancer cell.

In another aspect, the invention relates to a method of treating a disorder caused by a BAF complex in a subject in need thereof, the method comprising administering to the subject an effective amount of any of the above compounds (e.g., a compound dual BRM/BRG1 inhibitor ). or a selective BRM compound) or a pharmaceutical composition thereof.

In some embodiments, the disorder caused by the BAF complex is cancer.

In a further aspect, the invention relates to a method for inhibiting BRM, the method comprising contacting a cell with an effective amount of one of the above compounds (e.g., a dual BRM/BRG1 inhibitory compound or a selective BRM compound ) or a pharmaceutical composition thereof.

In some embodiments, the cell is a cancer cell.

In another aspect, the invention relates to a method of inhibiting BRG1, the method comprising contacting the cell with an effective amount of any of the above compounds or a pharmaceutical composition thereof.

In some embodiments, the cell is a cancer cell.

In another aspect, the invention relates to a method of inhibiting BRM and BRG1, the method comprising contacting the cell with an effective amount of any of the above compounds or a pharmaceutical composition thereof.

In some embodiments, the cell is a cancer cell.

In another aspect, the invention provides a method of treating a disorder associated with a BRG1 loss-of-function mutation in a subject in need thereof, the method comprising administering to the subject an effective amount of any of the above compounds (e.g., BRM /BRG1 dual inhibitor compound or a selective BRM compound) or a pharmaceutical composition thereof.

In some embodiments, the disorder associated with a BRG1 loss-of-function mutation is cancer. In other embodiments, the subject is determined to have a BRG1 loss-of-function disorder, e.g. e.g., is determined to have a BRG1-deficient cancer (e.g., the cancer is determined to comprise BRG1-deficient cancer cells).

In another aspect, the invention relates to a method for inducing apoptosis in a cell, the method comprising contacting the cell with an effective amount of one of the above compounds (e.g., a BRM/BRG1 dual inhibitory compound or a selective compound from BRM) includes a pharmaceutical composition thereof.

In some embodiments, the cell is a cancer cell.

In another aspect, the invention provides a method of treating cancer in a patient in need thereof, the method comprising administering to the patient an effective amount of any of the above compounds (e.g., a BRM/BRG1 dual inhibitor compound or selective factor BRM ). compound) or a pharmaceutical composition thereof.

In some embodiments of any of the above methods, the cancer is non-small cell lung cancer, colon cancer, bladder cancer, cancer of unknown origin, glioma, breast cancer, melanoma, non-melanoma skin cancer, endometrial cancer, esophageal cancer. Pancreatic cancer, hepatobiliary cancer, soft tissue sarcoma, ovarian cancer, head and neck cancer, renal cell carcinoma, bone cancer, non-Hodgkin's lymphoma, small cell lung cancer, prostate cancer, embryonal tumor, germ cell tumor, cervical cancer uterus, thyroid cancer, salivary gland cancer, gastrointestinal neuroendocrine tumor, uterine sarcoma, gastrointestinal stromal tumor, CNS - Cancer, thymus tumor, adrenocortical carcinoma, appendectomy cancer, small bowel cancer or penile cancer.

In some embodiments of any of the above methods, the cancer is non-small cell lung cancer, colon cancer, bladder cancer, cancer of unknown origin, glioma, breast cancer, melanoma, non-melanoma skin cancer, endometrial cancer, or penile cancer.

In some embodiments of any of the above methods, the cancer is a drug-resistant cancer or has failed to respond to prior therapy (eg, vemurafenib, dacarbazine, CTLA4 inhibitor, PD1 inhibitor, interferon therapy, BRAF, MEK inhibitor). Radiation therapy, temozolimide, irinotecan, CAR-T therapy, Herceptin, Perjeta, tamoxifen, Xeloda, docetaxel, platinum drugs such as carboplatin, taxanes such as paclitaxel and docetaxel, ALK inhibitors, MET inhibitors, Alimta, Abraxan®, Adriaem Avastin, Halaven, neratinib, PARP inhibitor, RNA810, mTOR inhibitor, topotecan, Gemzar, VEGFR2 inhibitor, folate receptor antagonist, demcizumab, fosbretabulin or PDL1 inhibitor).

In some embodiments of the above methods, the cancer has or has been determined to have BRG1 mutations. In some embodiments of the above methods, the BRG1 mutations are homozygous. In some embodiments of any of the above methods, the cancer does not have, or has been identified as lacking, an epidermal growth factor receptor (EGFR) mutation. In some embodiments of any of the above methods, the cancer lacks or has been found to lack an anaplastic lymphoma kinase (ALK) driver mutation. In some embodiments of any of the above methods, the cancer has, or has been determined to have, a KRAS mutation. In some embodiments of the above methods, the BRG1 mutation is located in the ATPase catalytic domain of the protein. In some embodiments of the above methods, the BRG1 mutation is a C-terminal deletion of BRG1.

In another aspect, the disclosure provides a method of treating a BAF-related disorder (eg, cancer or viral infections) in a patient in need thereof. This method comprises contacting a cell with an effective amount of any of the above compounds (eg, a BRM/BRG1 dual inhibitor compound or a BRM selective compound), or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions . In some embodiments, the disease is a viral infection, an infection with a virus of the Retroviridae family, such as lentiviruses (e.g., human immunodeficiency virus (HIV)) and deltaretoviruses (e.g., human T-cell leukemia virus I (HTLV -I) ), human T-cell leukemia virus II (HTLV-II)), family Hepadnaviridae (eg hepatitis B virus (HBV)), family Flaviviridae (eg hepatitis C virus (HCV)), family Adenoviridae (eg human adenovirus) , Herpes family (eg human cytomegalovirus (HCMV), Epstein-Barr virus, herpes simplex virus 1 (HSV-1), herpes simplex virus 2 (HSV-2), herpes simplex virus human 6 (HHV-6), herpesvirus K*, CMV, varicella-zoster virus), papilloma family (e.g. human papillomavirus (HPV, HPV E1)), family Parvoviridae (e.g. parvovirus B19), family polyomaviruses (eg JC and BK viruses), Paramyxoviridae family (eg Togas virus). family (eg rubella virus). In some embodiments, the condition is Syris coffin, neurofibromatosis (eg, NF-1, NF-2, or schwannomatosis), or multiple meningioma.

In another aspect, the disclosure provides a method of treating a viral infection in a patient in need thereof. This method comprises administering to the subject an effective amount of one of the above compounds (eg, a dual BRM/BRG1 inhibitory compound or a selective BRM compound), or pharmaceutically acceptable salts thereof, or any of the above pharmaceutical compositions. In some embodiments, the viral infection is an infection with a virus of the Retroviridae family, such as lentiviruses (e.g., human immunodeficiency virus (HIV)) and delta retroviruses (e.g., human T-cell leukemia virus I ( HTLV-I ), human T-cell leukemia virus II (HTLV-II), family Hepadnaviridae (eg hepatitis B virus (HBV)), family Flaviviridae (eg hepatitis C virus (HCV)), family Adenoviridae (eg human adenovirus ), herpes family (eg human cytomegalovirus (HCMV), Epstein-Barr virus, herpes simplex virus 1 (HSV-1), herpes simplex virus 2 (HSV-2), human herpesvirus 6 (HHV-6), herpes K. *, CMV, varicella-zoster virus), papilloma family (e.g. human papillomavirus (HPV, HPV E1)), parvovirus family (e.g. parvovirus B19), polyoma family viruses (eg JC virus and BK virus), virus of the paramyxovirus family (eg) or Togaviridae family (eg rubella virus).

In some embodiments of any of the above aspects, the compound is a BRM-selective compound. In some embodiments, the BRM-selective compound inhibits the level and/or activity of BRM at least 10-fold more than the compound inhibits the level and/or activity of BRG1 and/or the compound binds to BRM at least 10-fold more than the compound binds to BRG1. For example, in some embodiments, a selective BRM connector includes an IC50your IP50which is at least ten times smaller than CI50your IP50vs. BRG1. In some embodiments of any of the above aspects, the compound is a BRM/BRG1 dual inhibitor compound. In some embodiments, the BRM/BRG1 dual inhibitor compound has similar activity against BRM and BRG1 (e.g., activity of the compound against BRM and BRG1 within 10-fold (e.g., less than 5-fold, less than 2-fold) In some embodiments, the activity of the dual BRM/BRG1 inhibitor compound is greater against BRM In some embodiments, the activity of the dual BRM/BRG1 inhibitor compound is greater against BRG1 For example, in some embodiments, a dual BRM inhibitor compound /BRG1 to CI50your IP50vs. BRM which is within 10x of the CI50your IP50vs. BRG1.

In another aspect, the invention provides a method of treating melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematologic cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of any of the foregoing compounds or pharmaceutical compositions thereof.

In another aspect, the invention relates to a method of reducing tumor growth of melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematologic cancer in a patient in need thereof, the method comprising administering to the patient a effective amount of any of the above Compounds or pharmaceutical compositions thereof.

In another aspect, the invention relates to a method of suppressing the metastatic progression of melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma or hematologic cancer in a patient, the method comprising administering an effective amount of any of the above compounds or pharmaceutical compositions thereof.

In another aspect, the invention relates to a method of suppressing metastatic colonization of melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematologic cancer in a patient, the method comprising administering an effective amount of any of the above compounds in pharmaceutical compositions thereof.

In another aspect, the invention relates to a method of reducing the level and/or activity of BRG1 and/or BRM in melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma or hematologic cancer cells, the method comprising contacting the cell with an effective amount of any of the above compounds or pharmaceutical compositions thereof.

In some embodiments of any of the above aspects, a subject is a melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or a hematologic cell.

In some embodiments of any of the above aspects, the effective amount of the compound reduces the level and/or activity of BRG1 by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15% ). , 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%) in Compare with reference. In some embodiments, the effective amount of the compound reduces the level and/or activity of BRG1 by at least 50% (e.g., 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% ). %, or 95%) compared to a reference. In some embodiments, the effective amount of the compound reduces the level and/or activity of BRG1 by at least 90% (e.g., 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98). %, or 99%).

In some embodiments, the effective amount of the compound reduces the level and/or activity of BRG1 by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25 % ). , 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%) compared to reference for at least at least 12 hours (eg 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 24 hours, 30 hours, 36 hours, 48 ​​hours, 72 hours or more). In some embodiments, the effective amount of the compound reduces the level and/or activity of BRG1 by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25 % ). %, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%) compared to reference at least 4 days (eg 5 days, 6 days, 7 days, 14 days, 28 days or more).

In some embodiments of any of the above aspects, the effective amount of the compound reduces BRM levels and/or BRM activity by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15% ). 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%) compared by reference. In some embodiments, the effective amount of the compound that reduces BRM levels and/or BRM activity by at least 50% (e.g., 55%, 60%, 65%, 70%, 75%, 80%, 85% , 90% ). or 95%) compared to a reference.

In some embodiments, the effective amount of the compound that reduces BRM levels and/or BRM activity by at least 90% (e.g., 91%, 92%, 93%, 94%, 95%, 96%, 97% , 98% ). or 99%). In some embodiments, the effective amount of the compound reduces BRM level and/or BRM activity by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%). 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%) compared to reference for at least at least 12 hours (eg 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 24 hours, 30 hours, 36 hours, 48 ​​hours, 72 hours or more). In some embodiments, the effective amount of the compound that reduces BRM levels and/or BRM activity by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20% , 25% ). , 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%) compared to reference at least 4 days (eg 5 days, 6 days, 7 days, 14 days, 28 days or more).

In some embodiments, the subject has cancer. In some embodiments, the cancer expresses BRG1 and/or BRM protein and/or the cell or subject has been determined to express BRG1 and/or BRM. In some embodiments, the cancer expresses BRG1 protein and/or the cell or subject has been determined to express BRG1. In some embodiments, the cancer expresses BRM protein and/or the cell or subject has been determined to express BRM. In some embodiments, the cancer is a melanoma (eg, choroidal melanoma, mucosal melanoma, or cutaneous melanoma). In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is a hematologic cancer, for example, multiple myeloma, large cell lymphoma, acute T-cell leukemia, acute myeloid leukemia, myelodysplastic syndrome, immunoglobulin A lambda myeloma, diffuse mixed lymphocytic and histiocytic lymphoma, Lymphoma, acute lymphoblastic leukemia (eg, T-cell acute lymphoblastic leukemia or B-cell acute lymphoblastic leukemia), diffuse large cell lymphoma, or non-Hodgkin's lymphoma. In some embodiments, the cancer is breast cancer (eg, ER-positive breast cancer, ER-negative breast cancer, triple-positive breast cancer, or triple-negative breast cancer). In some embodiments, the cancer is bone cancer (eg, Ewing's sarcoma). In some embodiments, the cancer is renal cell carcinoma (e.g., microphthalmia transcription factor (MITF) family translocation renal cell carcinoma (tRCC)). In some embodiments, the cancer is metastatic (eg, the cancer has spread to the liver). Metastatic cancer may include cells exhibiting migratory cell migration and/or invasion and/or cells exhibiting endothelial recruitment and/or angiogenesis. In other embodiments, the migratory cancer is a migratory cell cancer. In yet other embodiments, the migratory cell cancer is a non-metastatic migratory cell cancer. Metastatic cancer can be cancer that has spread to the surface of the peritoneum, pleura, pericardium, or subarachnoid space with spread. Alternatively, metastatic cancer can be cancer that has spread through the lymphatic system or cancer that has spread through the hematogenous route. In some embodiments, the effective amount of an agent that reduces the level and/or activity of BRG1 and/or BRM is an amount effective to inhibit metastatic colonization of liver cancer.

In some embodiments, the cancer has a mutation in GNAQ. In some embodiments, the cancer has a mutation in GNA11. In some embodiments, the cancer has a mutation in PLCB4. In some embodiments, the cancer has a mutation in CYSLTR2. In some embodiments, the cancer has a mutation in BAP1. In some embodiments, the cancer has a mutation in SF3B1. In some embodiments, the cancer has a mutation in EIF1AX. In some embodiments, the cancer has a TFE3 translocation. In some embodiments, the cancer harbors a TFEB translocation. In some embodiments, the cancer has an M/TF translocation. In some embodiments, the cancer has an EZH2 mutation. In some embodiments, the cancer contains an SUZ12 mutation. In some embodiments, the cancer has an EED mutation.

In some embodiments, the method also comprises administering to the subject or contacting the cell with an anti-cancer treatment, such as a chemotherapeutic or cytotoxic agent, immunotherapy, surgery, radiation therapy, thermotherapy, or photocoagulation. In some embodiments, the cancer treatment is a chemotherapeutic or cytotoxic agent, e.g. B. Antimetabolite, antimitotic, antibiotic antibiotic, specific enzyme enzyme, bisphosphonates, immunoresonator, immunoresonator, immunoresonator. inhibitor, proteasome inhibitor or tyrosine kinase inhibitor.

In some embodiments, the compound of the invention is used in combination with another cancer therapy to treat uveal melanoma, such as surgery, a MEK inhibitor, and/or a PKC inhibitor. For example, in some embodiments, the method further comprises performing a surgical procedure before, after, or simultaneously with the administration of the compound of the invention. In some embodiments, the method also comprises administering a MEK inhibitor and/or a PKC inhibitor before, after, or simultaneously with the administration of the compound of the invention.

In some embodiments, the anticancer therapy and the compound of the invention are administered within 28 days of each other and each in an amount effective together to treat the patient.

In some embodiments, the subject or cancer has been determined to have a BRG1 loss-of-function mutation. In some embodiments, the subject or cancer has been determined to have a BRM loss-of-function mutation.

In some embodiments, the cancer is resistant to one or more chemotherapeutic or cytotoxic agents (e.g., genetic markers have determined that the cancer is resistant to chemotherapeutic or cytotoxic agents or is likely to be resistant to chemotherapeutic or cytotoxic agents, cancers (which failed to respond to a chemotherapeutic or cytotoxic drug).In some embodiments, the cancer has failed to respond to one or more chemotherapeutic or cytotoxic agents. , fotemustine, IMCgp100, a CTLA-4 inhibitor (eg, ipilimumab), a PD inhibitor -1 (eg, nivolumab or pembrolizumab) a PD-L1 inhibitor (eg, MEK) inhibitor (eg, selumetinib, binimetinib, or tametinib) and/or a protein kinase C (PKC) inhibitor (eg ., sotrastaurin or IDE196).

In some embodiments, the cancer is refractory or unresponsive to an administered therapeutic agent previously used to treat uveal melanoma, such as a MEK inhibitor or PKC inhibitor. For example, in some embodiments, the cancer is resistant or unresponsive to a mitogen-activated protein kinase (MEK) inhibitor (e.g., selumetinib, binimetinib, or tametinib) and/or a protein kinase C (PKC) inhibitor. (eg, sorastaurin or IDE196).

chemical terms

The terminology used herein is intended to describe specific modes and is not intended to be limiting.

For each of the following chemical definitions, a number following an atomic symbol indicates the total number of atoms of that element present in a given chemical fraction. It is understood that other atoms, such as H atoms or substituent groups as described herein, may also be present, if desired, to satisfy the valences of the atoms. For example, an unsubstituted C2The alkyl group has the formula -CH2CH3. When used with the groups defined herein, a reference to the number of carbon atoms includes the divalent carbon in acetal and ketal groups, but does not include the carbonyl carbon in acyl, ester, carbonate, or urea groups. Reference to the number of oxygen, nitrogen, or sulfur atoms in a heteroaryl group includes only those atoms that are part of a heterocyclic ring.

The term "acyl" as used herein represents an H or an alkyl group attached to a parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl (ie, a carboxaldehyde group), acetyl, trifluoroacetyl, propionyl, and butanoyl. . Exemplary unsubstituted acyl groups include 1 to 6, 1 to 11 or 1 to 21 carbons.

As used herein, the term "alkyl" refers to a branched or straight chain monovalent saturated aliphatic hydrocarbon having from 1 to 20 carbon atoms (e.g., 1 to 16 carbons, 1 to 10 carbons, carbon, 1 to 6 carbons) . or 1 to 3 carbon atoms).

An alkylene is a divalent alkyl group. The term "alkenyl" as used herein, alone or in combination with other groups, refers to a straight or branched chain hydrocarbon radical having a carbon-carbon double bond and containing from 2 to 20 carbon atoms (e.g., 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 6 carbon atoms, or 2 carbon atoms).

The term "alkynyl" as used herein, alone or in combination with other groups, refers to a straight or branched chain hydrocarbon radical containing a carbon-carbon triple bond and having from 2 to 20 carbon atoms (e.g., 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 6 carbon atoms, or 2 carbon atoms).

The term "amino" as used herein means -N(R).N1)2, in which each RN1are independent H, OH, NO2, N(RN2)2, AFTERWARD2THEN2, AFTERWARD2RN2, SORN2an N-protecting group, alkyl, alkoxy, aryl, arylalkyl, cycloalkyl, acyl (eg, acetyl, trifluoroacetyl or others described herein), each of said RN1Groups can optionally be replaced. or two RsN1combine to form an alkylene or heteroalkylene and wherein each RN2are independently H, alkyl or aryl. Amino groups of the invention may be unsubstituted amino groups (ie -NH).2) or a substituted amino (ie -N(R.)N1)2).

As used herein, the term "aryl" refers to an aromatic mono- or polycarbocyclic radical of 6 to 12 carbon atoms containing at least one aromatic ring. Examples of such groups include, but are not limited to, phenyl, naphthyl, 1,2,3,4-tetrahydronaphthyl, 1,2-dihydronaphthyl, indanyl, and 1H-indenyl.

The term "arylalkyl" as used herein represents an alkyl group replacing an aryl group. Exemplary unsubstituted arylalkyl groups have 7 to 30 carbon atoms (eg 7 to 16 or 7 to 20 carbon atoms as C).1-DO6C alkyl6-DO10arilo, C1-DO10C alkyl6-DO10Arilo or C1-DO20C alkyl6-DO10aryl) such as benzyl and phenethyl. In some embodiments, alkyl and aryl may be further substituted by 1, 2, 3 or 4 substituent groups as defined herein for the respective groups.

The term "azido" used in this document represents a -N3Club.

As used herein, the term "bridged polycycloalkyl" refers to a bridged polycyclic group of 5 to 20 carbon atoms containing 1 to 3 bridges.

The term "cyano" as used herein means a -CN group.

As used herein, the term "carbocyclyl" refers to a non-aromatic C3-DO12monocyclic, bicyclic or tricyclic structure in which the rings are formed by carbon atoms. Carbocyclyl structures include cycloalkyl groups and unsaturated carbocyclyl radicals.

As used herein, the term "cycloalkyl" refers to a monovalent, non-aromatic, saturated mono- or polycarbocyclic radical having from 3 to 10, preferably 3 to 6, carbon atoms. Other examples of this term are radicals such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl and adamantyl.

The term "halogen" as used herein means a fluoro (fluorine), chloro (chloro), bromo (bromine) or iodo (iodine) radical.

The term "heteroalkyl" as used herein refers to an alkyl group, as defined herein, in which one or more of the constituent carbon atoms has been replaced by nitrogen, oxygen or sulfur. In some embodiments, the heteroalkyl group may be further substituted with 1, 2, 3 or 4 substituent groups as described herein for alkyl groups. Examples of heteroalkyl groups are "alkoxy", which herein refers to alkyl-O- (eg, methoxy and ethoxy). A heteroalkylene is a divalent heteroalkyl group. The term "heteroalkenyl" as used herein refers to an alkenyl group, as defined herein, in which one or more of the constituent carbon atoms has been replaced by nitrogen, oxygen or sulfur. In some embodiments, the heteroalkenyl group may be further substituted with 1, 2, 3 or 4 substituent groups as described herein for alkenyl groups. Examples of heteroalkenyl groups are "alkenoxy", which herein refers to alkenyl-O-. A heteroalkenylene is a divalent heteroalkenyl group. The term "heteroalkynyl" as used herein refers to an alkynyl group, as defined herein, in which one or more of the constituent carbon atoms has been replaced by nitrogen, oxygen or sulfur. In some embodiments, the heteroalkynyl group may be further substituted with 1, 2, 3 or 4 substituent groups as described herein for alkynyl groups. Examples of heteroalkynyl groups are "alkynoxy" which, as used herein, refers to alkynyl-O-. A heteroalkynylene is a divalent heteroalkynyl group.

The term "heteroaryl" as used herein refers to a mono- or polycyclic aromatic radical having 5 to 12 members, wherein at least one aromatic ring contains 1, 2, or 3 ring atoms selected from nitrogen, oxygen, and sulfur, wherein the remaining ring contains 1, 2 or 3 ring atoms The atoms are carbon. One or two ring carbon atoms of the heteroaryl group may be replaced by a carbonyl group. Examples of heteroaryl groups are pyridyl, pyrazolyl, benzooxazolyl, benzoimidazolyl, benzothiazolyl, imidazolyl, oxazolyl and thiazolyl.

The term "heteroarylalkyl" as used herein represents an alkyl group substituted with a heteroaryl group. Exemplary unsubstituted heteroarylalkyl groups have 7 to 30 carbons (eg, 7 to 16 or 7 to 20 carbons, such as C).1-DO6C alkyl2-DO9Heteroaril, C1-DO10C alkyl2-DO9Heteroaryl or C1-DO20C alkyl2-DO9heteroaryl). In some embodiments, alkyl and heteroaryl may be further substituted by 1, 2, 3 or 4 substituent groups as defined herein for the respective groups.

The term "heterocyclyl" as used herein refers to a 3- to 12-membered mono- or polycyclic radical containing at least one ring with 1, 2, 3, or 4 ring atoms selected from N, O, or S, in which case no ring is present aromatic. Examples of heterocyclyl groups include morpholinyl, thiomorpholinyl, furyl, piperazinyl, piperidinyl, pyranyl, pyrrolidinyl, tetrahydropyranyl, tetrahydrofuranyl and 1,3-dioxanyl.

The term "heterocyclylalkyl" as used herein represents an alkyl group substituted with a heterocyclyl group. Exemplary unsubstituted heterocyclylalkyl groups have 7 to 30 carbons (eg, 7 to 16 or 7 to 20 carbons, such as C).1-DO6C alkyl2-DO9Heterocyclyl, C1-DO10C alkyl2-DO9Heterocyclyl or C1-DO20C alkyl2-DO9heterocyclyl). In certain embodiments, alkyl and heterocyclyl may each be further substituted by 1, 2, 3 or 4 substituent groups as defined herein for the respective groups.

The term "hydroxyalkyl" as used herein represents the alkyl group substituted with an -OH group.

The term "hydroxyl" as used herein means a -OH group.

The term "N-protecting group" as used herein represents those groups intended to protect an amino group from undesired reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene, "Protective Groups in Organic Synthesis", 3rd Edition (John Wiley & Sons, New York, 1999). N-protecting groups include, but are not limited to, acyl, aryl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalo-phenoxyacetyl, α- chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl and chiral auxiliaries such as D, L or D, protected or unprotected L-amino acids such as alanine, leucine and phenylalanine. sulfonyl containing groups such as benzenesulfonyl and p-toluenesulfonyl. Carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4-benzimethoxycarbonyl, 3,4-benzimethoxycarbonyl. carbonyl, 2,4-20-dimethoxybenzyloxycarbonyl, 4- methoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl, 3,4,5-trimethoxybenzyloxycarbonyl, 1-(p-diphenyl)-1-methylethoxycarbonyl, α,α-dimethyl- hydroxycarbonyl-3,5-dimethyl-hydroxycarbonyl-benzyloxy -butyloxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxycarbonyl, fluoro enyloxycarbonyl, fluorenyloxycarbonyl, fluorenyloxycarbonyl, yl, cyclohexyloxycarbonyl and phenylthiocarbonyl, arylalkyl groups such as benzyl, triphenylmethyl and benzyloxymethyl and silyl groups such as trimethylsilyl. Preferred N-protecting groups are Aloc, formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc) and benzyloxycarbonyl (Cbz).

The term "nitro" used in this document represents a -NO2Club.

The term "thiol" as used herein means a -SH group.

Alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl (eg, cycloalkyl), aryl, heteroaryl, and heterocyclyl groups may be substituted or unsubstituted. When substituted, there are usually 1 to 4 substituents unless otherwise noted. Substituents include, for example: alkyl (e.g., unsubstituted and substituted, wherein the substituents include all groups described herein, e.g., aryl, halo, hydroxy), aryl (e.g., substituted and unsubstituted phenyl), carbocyclyl (eg substituted and unsubstituted). cycloalkyl), halogen (e.g. fluoro), hydroxyl, heteroalkyl (e.g. substituted and unsubstituted methoxy, ethoxy or thioalkoxy), heteroaryl, heterocyclyl, amino (e.g. NH).2or mono- or di-alkylamino), azido, cyano, nitro or thiol. Aryl, carbocyclyl (eg cycloalkyl), heteroaryl and heterocyclyl groups can also be substituted by alkyl (unsubstituted and substituted such as arylalkyl (eg substituted and unsubstituted benzyl)).

Οι ενώσεις σύμφωνα με την εφεύρεση μπορεί να έχουν ένα ή περισσότερα ασύμμετρα άτομα άνθρακα και μπορεί να έχουν τη μορφή οπτικώς καθαρών εναντιομερών, μιγμάτων εναντιομερών όπως ρακεμικών, οπτικώς καθαρών διαστερεοϊσομερών, μιγμάτων διαστερεοϊσομερών, διαστερεοϊσομερών ρακεμικών ρακεμικών ή μιγμάτων διστερεοϊσομερών. Οι οπτικά ενεργές μορφές μπορούν να ληφθούν, για παράδειγμα, με διαχωρισμό ρακεμικού, με ασύμμετρη σύνθεση ή ασύμμετρη χρωματογραφία (χρωματογραφία με χειρόμορφα προσροφητικά ή διαλύματα έκλουσης). Δηλαδή, μερικές από τις αποκαλυπτόμενες ενώσεις μπορεί να υπάρχουν σε διαφορετικές στερεοϊσομερείς μορφές. Τα στερεοϊσομερή είναι ενώσεις που διαφέρουν μόνο στη χωρική τους διάταξη. Τα εναντιομερή είναι ζεύγη στερεοϊσομερών των οποίων οι κατοπτρικές εικόνες δεν είναι υπερτιθέμενες, κυρίως επειδή περιέχουν ένα ασύμμετρα υποκατεστημένο άτομο άνθρακα που δρα ως χειρόμορφο κέντρο. "Εναντιομερές" σημαίνει ένα από ένα ζεύγος μορίων που είναι κατοπτρικά είδωλα το ένα του άλλου και δεν είναι υπερτιθέμενα. Τα διαστερεοϊσομερή είναι στερεοϊσομερή που δεν σχετίζονται με κατοπτρική εικόνα, συνηθέστερα επειδή περιέχουν δύο ή περισσότερα ασύμμετρα υποκατεστημένα άτομα άνθρακα και αντιπροσωπεύουν τη διαμόρφωση των υποκαταστατών γύρω από ένα ή περισσότερα χειρόμορφα άτομα άνθρακα. Τα εναντιομερή μιας ένωσης μπορούν να παρασκευαστούν, για παράδειγμα, με διαχωρισμό ενός εναντιομερούς από ένα ρακεμικό με χρήση μιας ή περισσότερων γνωστών τεχνικών και μεθόδων, όπως χειρόμορφη χρωματογραφία και μέθοδοι διαχωρισμού που βασίζονται σε αυτό. Η κατάλληλη τεχνική και/ή μέθοδος για τον διαχωρισμό ενός εναντιομερούς μιας ένωσης που περιγράφεται εδώ από ένα ρακεμικό μίγμα μπορεί εύκολα να προσδιοριστεί από τους έμπειρους της τέχνης. "ρακεμικό μίγμα" ή "ρακεμικό μίγμα" σημαίνει μια ένωση που περιέχει δύο εναντιομερή, τέτοια μίγματα που δεν παρουσιάζουν οπτική δραστηριότητα. δηλαδή δεν περιστρέφουν το επίπεδο του πολωμένου φωτός. "Γεωμετρικό Ισομερές" σημαίνει ισομερή που διαφέρουν στον προσανατολισμό των ατόμων υποκαταστάτη σε σχέση με έναν διπλό δεσμό άνθρακα-άνθρακα, έναν κυκλοαλκυλικό δακτύλιο ή ένα γεφυρωμένο δικυκλικό σύστημα. Τα άτομα (εκτός από το Η) σε κάθε πλευρά ενός διπλού δεσμού άνθρακα-άνθρακα μπορεί να έχουν διαμόρφωση Ε (οι υποκαταστάτες βρίσκονται στις αντίθετες πλευρές του διπλού δεσμού άνθρακα-άνθρακα) ή διάταξη Ζ (οι υποκαταστάτες είναι ευθυγραμμισμένοι στην ίδια πλευρά). "R", "S", "S*", "R*", "E", "Z", "cis" και "trans" υποδεικνύουν διαμορφώσεις σε σχέση με το μόριο πυρήνα. Μερικές από τις αποκαλυπτόμενες ενώσεις μπορεί να υπάρχουν σε ατροπισομερείς μορφές. Τα ατροπισομερή είναι στερεοϊσομερή που προκύπτουν από την παρεμπόδιση της περιστροφής γύρω από απλούς δεσμούς, όπου το στερικό φράγμα στην περιστροφή είναι αρκετά υψηλό ώστε να επιτρέπει την απομόνωση των διαμορφωτών. Οι ενώσεις σύμφωνα με την εφεύρεση μπορούν να παρασκευαστούν ως μεμονωμένα ισομερή με σύνθεση ειδική για ισομερή ή μπορούν να διαχωριστούν από ένα μείγμα ισομερών. Οι συμβατικές τεχνικές διαχωρισμού περιλαμβάνουν αλατοποίηση της ελεύθερης βάσης κάθε ισομερούς ζεύγους ισομερών χρησιμοποιώντας οπτικά ενεργό οξύ (ακολουθούμενη από κλασματική κρυστάλλωση και αναγέννηση της ελεύθερης βάσης) και αλατοποίηση της όξινης μορφής κάθε ισομερούς ζεύγους ισομερών χρησιμοποιώντας οπτικά ενεργή όξινη αμίνη (ακολουθούμενη από κλασματική κρυστάλλωση και αναγέννηση του ελεύθερου οξέος), σχηματισμός ενός εστέρα ή αμιδίου 35 από κάθε ισομερές ενός ζεύγους ισομερών χρησιμοποιώντας ένα οπτικά καθαρό οξύ, μια αμίνη ή μια αλκοόλη (ακολουθούμενη από χρωματογραφικό διαχωρισμό και απομάκρυνση του βοηθητικού χειρόμορφου). ) ή για διαχωρισμό ενός ισομερούς μίγματος μιας πρώτης ύλης ή ενός τελικού προϊόντος χρησιμοποιώντας διάφορες γνωστές χρωματογραφικές μεθόδους. Όταν η στερεοχημεία μιας αποκαλυπτόμενης ένωσης ονομάζεται ή αντιπροσωπεύεται από τη δομή, το στερεοϊσομερές που ονομάζεται ή αντιπροσωπεύεται είναι τουλάχιστον 60, 70, 80, 90, 99 ή 99,9 τοις εκατό κατά βάρος σε σχέση με τα άλλα στερεοϊσομερή. Όταν ένα μεμονωμένο εναντιομερές ονομάζεται ή αντιπροσωπεύεται από τη δομή, το εναντιομερές που απεικονίζεται ή ονομάζεται είναι τουλάχιστον 60%, 70%, 80%, 90%, 99% ή 99,9% οπτικά καθαρό. Όταν ένα μεμονωμένο διαστερεομερές ονομάζεται ή αντιπροσωπεύεται από τη δομή, το διαστερεομερές που ονομάστηκε ή ονομάστηκε έχει καθαρότητα τουλάχιστον 60%, 70%, 80%, 90%, 99% ή 99,9%. Το ποσοστό οπτικής καθαρότητας είναι η αναλογία του βάρους του εναντιομερούς ή του βάρους του εναντιομερούς συν το βάρος του οπτικού ισομερούς του. Διαστερεομερική καθαρότητα κατά βάρος είναι η αναλογία του βάρους ενός διαστερεομερούς προς το βάρος όλων των διαστερεομερών. Όταν η στερεοχημεία μιας αποκαλυπτόμενης ένωσης ονομαστεί ή δείχνεται από τη δομή, το στερεοϊσομερές που ονομάζεται ή παρουσιάζεται είναι τουλάχιστον 60%, 70%, 80%, 90%, 99% ή 99,9% από καθαρό μοριακό κλάσμα σε σχέση με τα άλλα στερεοϊσομερή. Όταν ένα μεμονωμένο εναντιομερές ονομάζεται ή αντιπροσωπεύεται από τη δομή, τουλάχιστον το 60%, 70%, 80%, 90%, 99% ή 99,9% του εναντιομερούς που αντιπροσωπεύεται ή ονομάζεται αποτελείται από καθαρό μοριακό κλάσμα. Όταν ένα μεμονωμένο διαστερεομερές ονομάζεται ή αντιπροσωπεύεται από τη δομή, τουλάχιστον το 60%, 70%, 80%, 90%, 99% ή 99,9% αποτελεί το διαστερεομερές που ονομάζεται ή ονομάζεται με καθαρό μοριακό κλάσμα. Η εκατοστιαία καθαρότητα ανά μοριακό κλάσμα είναι η αναλογία των γραμμομορίων του εναντιομερούς ή έναντι των γραμμομορίων του εναντιομερούς συν τα γραμμομόρια του οπτικού ισομερούς του. Ομοίως, το ποσοστό καθαρότητας ανά μοριακό κλάσμα είναι η αναλογία γραμμομορίων διαστερεομερούς ή έναντι γραμμομορίων διαστερεομερούς συν γραμμομορίων του ισομερούς του. Όταν μια αποκαλυπτόμενη ένωση ονομάζεται ή αντιπροσωπεύεται από τη δομή της χωρίς να προσδιορίζεται η στερεοχημεία, και η ένωση έχει τουλάχιστον ένα κέντρο χειρομορφίας, το όνομα ή η δομή πρέπει να κατανοηθεί ότι περιλαμβάνει το εναντιομερές της ένωσης απαλλαγμένο από το αντίστοιχο οπτικό ισομερές, ένα ρακεμικό Μίγμα ισομερών της ένωσης ή μιγμάτων εμπλουτισμένων σε ένα εναντιομερές σε σχέση με το αντίστοιχο οπτικό ισομερές. Όταν μια αποκαλυπτόμενη ένωση ονομάζεται ή αντιπροσωπεύεται από τη δομή της χωρίς να προσδιορίζεται η στερεοχημεία και έχει δύο ή περισσότερα κέντρα χειραλικότητας, το όνομα ή η δομή εννοείται ότι περιλαμβάνει ένα διαστερεομερές απαλλαγμένο από άλλα διαστερεομερή, πολλαπλά διαστερεομερή χωρίς άλλα ζεύγη διαστερεομερών. και μίγματα διαστερεομερών, μίγματα ζευγών διαστερεομερών, μίγματα διαστερεομερών στα οποία το ένα διαστερεομερές είναι εμπλουτισμένο σε σχέση με τα άλλα διαστερεομερή, ή μίγματα διαστερεομερών στα οποία ένα ή περισσότερα διαστερεομερή είναι εμπλουτισμένα σε σχέση με τα άλλα διαστερεομερή. Η εφεύρεση περιλαμβάνει όλες αυτές τις μορφές.

Unless otherwise defined, all technical and scientific terms used in this document have the same meaning as is commonly understood by a person skilled in the art to which this invention belongs. Methods and materials for use in the present disclosure are described herein. Other methods and suitable materials known in the art may also be used. Materials, methods and examples are provided for illustrative purposes only and are not intended to be limiting. All publications, patent applications, patents, sequences, database entries and other references cited herein are incorporated by reference in their entirety. In the event of any conflict, this specification, including definitions, controls.

The definition

In this application, unless the context clearly indicates: (i) the term "a" may be taken to mean "at least one"; (ii) the term "or" may be interpreted as "and/or"; and (iii ) the terms "comprising" and "included" may be taken to include separately listed elements or steps, whether presented individually or together with one or more additional elements or steps.

The terms "about" and "about" as used in this document refer to a value that is 10% greater or less than the described value. For example, the term "about 5 nM" indicates a range of 4.5 to 5.5 nM.

As used herein, the term "administration" refers to the administration of a composition (eg, a compound or a preparation containing a compound as described herein) to an individual or system. Administration to an animal (eg, human) can be by any suitable route. In some embodiments, administration can be, for example, bronchial (including bronchial instillation), buccal, enteral, mesodermal, intraarterial, intradermal, intramuscular, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intratumoral, intravenous, nasal, intravenous , oral, rectal, subcutaneous, sublingual, topical, tracheal (including intratracheal instillation), transdermal, vaginal and vitreous.

As used herein, the term "BAF complex" refers to the complex of factors associated with BRG1 or HRBM in a human cell.

As used herein, the term "BAF complex-related disorder" refers to a disorder caused or affected by the level of activity of a BAF complex.

As used herein, the term "BRG1 loss-of-function mutation" refers to a mutation in BRG1 that results in a protein with reduced activity (e.g., at least a 1% reduction in BRG1 activity, e.g., 2%, 5% , 10 %). , 25%, 50% or 100% reduction in BRG1 activity). Exemplary BRG1 loss-of-function mutations include, but are not limited to, a BRG1 homozygous mutation and a BRG1 C-terminal deletion.

As used herein, the term "BRG1 loss-of-function disorder" refers to a disease (eg, cancer) that exhibits a decrease in BRG1 activity (eg, a decrease in BRG1 activity of at least 1%, for example 2%). 5%, 10%, 25%, 50% or 100% reduction in BRG1 activity).

As used herein, the term "BRM-selective compound" refers to a compound that exhibits greater activity against BRM than against BRG1 (eg, the compound inhibits the level and/or activity of BRM at least 2-fold ( at least 5-fold ), at least 10-fold, at least 20-fold, at least 50-fold, at least 100-fold) greater than the compound, inhibits BRG1 level and/or activity, and/or the compound binds at least twice to BRM (e.g. (eg at least 5-fold, at least 10-fold, at least 20-fold, at least 50-fold, at least 100-fold greater than the BRG1-bound compound). For example, in some embodiments, a selective BRM connector includes an IC50your IP50which is at least 2 times (eg at least 5 times, at least 10 times, at least 20 times, at least 50 times, at least 100 times) less than KI50your IP50vs. BRG1.

As used herein, the term "BRM/BRG1 dual inhibitory compound" refers to a compound that is active against both BRM and BRG1. In some embodiments, a BRM/BRG1 dual inhibitor compound has similar activity against BRM and BRG1 (e.g., the activity of the compound against BRM and BRG1 ranges from 10-fold (e.g., less than 5-fold, less than 2 times ).. ). For example, in some embodiments, a BRM/BRG1 dual inhibitor compound has an IC50your IP50vs. BRM which is within 10x of the CI50your IP50vs. BRG1.

The term "cancer" refers to a condition caused by the proliferation of malignant neoplastic cells such as tumors, neoplasia, carcinoma, sarcoma, leukemia and lymphoma.

As used herein, "combination therapy" or "administered in combination" means that two (or more) different drugs or therapies are administered to a subject as part of a defined treatment regimen for a particular disease or condition. The treatment regimen determines the dosage and time of administration of each drug so that the effects of each drug on the patient overlap. In some embodiments, the delivery of two or more active ingredients occurs simultaneously or concurrently, and the active ingredients can be co-formulated. In some embodiments, the two or more active ingredients are not co-formulated and are administered sequentially as part of a predetermined regimen. In some embodiments, the administration of two or more drugs or treatments in combination is such that the reduction in a symptom or other parameter associated with the disorder is greater than if one drug or treatment was administered alone or in the absence of the other. The effect of the two treatments may be partially additive, fully additive, or greater than additive (eg, synergistic). Sequential or substantially simultaneous administration of each therapeutic agent may be by any suitable route, including but not limited to the oral route, the intravenous route, the intramuscular route, and direct absorption through mucosal tissues. The therapeutic agents can be administered by the same or different routes. For example, a first combination therapeutic may be administered by intravenous injection, while a second combination therapeutic may be administered orally.

By "determining the level" of a protein or RNA is meant the direct or indirect detection of a protein or RNA by methods known in the art. "Direct Determination" means performing a procedure (eg, performing an examination or test on a Sample or "sample analysis" as that term is defined herein) to obtain the physical entity or value. "Indirectly Determined" means obtaining the physical item or asset from another party or source (eg, a third-party laboratory that directly acquired the physical item or asset). Methods used to measure protein content generally include, but are not limited to, Western blotting, immunoblotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, immunofluorescence, surface plasmon resonance, chemiluminescence, fluorescence analysis, immunofluorescence, chemical polarization, matrix-assisted laser desorption/ionization mass spectrometry time-of-flight mass spectrometry (MALDI-TOF), liquid chromatography-mass spectrometry (LC), microcytometry, microscopy, fluorescence-activated cell sorting (FACS) and flow cytometry and assays with based property of a protein, including but not limited to enzymatic activity or interaction with other partner proteins. Methods for measuring RNA levels are known in the art and include, but are not limited to, quantitative polymerase chain reaction (qPCR) and Northern blot analysis.

A "decreased level" or an "increased level" of a protein or RNA means a decrease or increase in protein or RNA level, respectively, compared to a reference (eg, about 5% decrease or increase, about 10). %, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500% or more. decrease or increase of more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200% compared to a reference. decrease or increase of less than about 0.01-fold, about 0.02-fold, about 0.1-fold, about 0.3-fold, about 0.5-fold, about 0.8-fold or less. or an increase greater than about 1.2-fold, about 1.4-fold, about 1.5-fold, about 1.8-fold, about 2.0-fold, about 3.0-fold, about 3.5-fold, about 4.5 times, about 5.0 times, about 10 times, about 15 times, about 20 times, about 30 times, about 40 times, about 50 times, about 100 times, about 1000 times or more). The content of a protein can be expressed as a mass/volume (eg g/dl, mg/ml, µg/ml, ng/ml) or as a percentage of the total protein in a sample.

By "reducing the activity of a BAF complex" is meant reducing the level of an activity associated with a BAF complex or a related downstream effect. A non-limiting example of reduced activity of a BAF complex is the activation of Sox2. The level of activity of a BAF complex can be measured using any method known in the art, for example using the methods described in Kadoch et al. methods described. Cell, 2013, 153, 71-85, the methods of which are incorporated herein by reference.

As used herein, the term "inhibition of BRM" refers to blocking or reducing the level or activity of the catalytic binding domain of the ATPase or bromosome of the protein. BRM inhibition can be determined using methods known in the art, such as a BRM-ATPase assay, a nano-DSF assay, or a BRM-luciferase cell assay.

The term "pharmaceutical composition" as used herein means a composition containing a compound described herein formulated with a pharmaceutically acceptable excipient suitable for administration to a mammal, such as a human. Typically, a pharmaceutical composition is manufactured or sold with the approval of a government regulatory agency as part of a therapeutic regimen for the treatment of diseases in mammals. Pharmaceutical compositions can be formulated, for example, for oral administration in unit dose form (for example, tablet, capsule, capsule, gelcap or syrup). for topical administration (eg as a cream, gel, lotion or ointment). for intravenous administration (eg as a sterile solution free of particulate matter and in a solvent system suitable for intravenous use). or in another pharmaceutically acceptable formulation.

A "pharmaceutically acceptable excipient" as used herein refers to any component other than the compounds described herein (eg, a carrier capable of suspending or dissolving the active compound) and has the properties of being substantially non-toxic and non-inflammatory. patient. Examples of excipients may include: release agents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (pigments), plasticizers, emulsifiers, fillers (thinners), film or coating formers, flavors, fragrances, flow aids), lubricants, preservatives , printing inks, Absorbents, suspending or dispersing agents, sweeteners and hydration waters. Examples of excipients include: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, cross-linked polyvinylpyrrolidone, citric acid, crospovidone, cysteine, ethylpylmethylcellulose, hydropyrylcellulose, lactose, stearate genuine, maltitol, mannitol, methionine, methylcellulose , methylparaben, microcrystalline cellulose, polyethylene glycol, polyvinylpyrrolidone, povidone, pregelatinized starch, propylparaben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyltracholylate, sodium carboxymethyltralytecolate, sodium stearate, sodium sorbitol, gastrooxylotoxic sodium aric acid, Sucrose, Talc, Dioxide Titanium, Vitamin A, Vitamin E, Vitamin C and Xylitol.

As used herein, the term "pharmaceutically acceptable salt" means any pharmaceutically acceptable salt of a compound, for example any compound of formula I, formula IV, formula V, formula VI or formula VII. Pharmaceutically acceptable salts of all compounds described herein may be those which, in the context of sound medical judgment, are suitable for use in contact with human and animal tissues without undue toxicity, irritation or allergic reaction and commensurate with a reasonable ratio of benefit /danger. Pharmaceutically acceptable salts are well known in the art. Pharmaceutically acceptable salts are described, for example, in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and Pharmaceutical Salts: Properties, Selection, and Use, (eds. P.H. Stahl and C.G. Wermuth), Wiley -VCH, 2008. Salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reaction of a free base group with an appropriate organic acid.

The compounds of the invention may have ionizable groups which will be prepared as pharmaceutically acceptable salts. These salts may be acid addition salts with inorganic or organic acids or, in the case of acid forms of the compounds of the invention, the salts may be prepared from inorganic or organic bases. Often, the compounds are prepared or used as pharmaceutically acceptable salts, which are prepared as pharmaceutically acceptable acid or base adducts. Suitable pharmaceutically acceptable acids and bases and methods for preparing the corresponding salts are well known in the art. The salts can be prepared from pharmaceutically acceptable, non-toxic acids and bases, including inorganic and organic acids and bases. Representative acid addition salts include acetate, adipic, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, diglyconate, dodecyl sulfate, glycofluorosulfate, ethaneglucosulfate, ethaneglucotenosulfate ionic semisulfate, heptonic, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxyethanesulfonate, lactobionic, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, paleo-sulfonate, pernicotinate, palalomitate, palalate, 3-Phen hylopropionate, phosphate , picranium, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate and valerate. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium and magnesium, as well as non-toxic ammonium, quaternary ammonium and amine cations, including but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine and ethylamine .

By "reference" is meant any useful reference used to compare protein or RNA levels. The reference can be any sample, standard, standard curve or level used for comparison purposes. The reference can be a normal reference sample or a standard or reference level. A "reference sample" can be, for example, a control, such as a predetermined negative control value, such as a "normal control" or a previous sample obtained from the same subject. a sample from a normal healthy person, such as a normal cell or tissue; a sample (eg a cell or tissue) from a person who does not have a disease. a sample from an individual diagnosed with a disease but not previously treated with a compound of the invention. a sample from a subject treated with a compound of the invention. or a sample of purified protein or RNA (eg, any described herein) at a known normal concentration. By "standard or reference level" is meant a value or number derived from a reference sample. A "normal control value" is a predetermined value indicative of a disease-free state, such as a value expected in a healthy control subject. Typically, a normal control value is expressed as a range ("between X and Y"), an upper limit ("not greater than X"), or a lower limit ("not less than X"). An individual with a measured value within the normal control value for a particular biomarker is usually referred to as "within normal limits" for that biomarker. A normal reference value or standard may be a value or number derived from a normal individual not suffering from a disease or disorder (eg, cancer). a subject treated with a compound of the invention. In preferred embodiments, the reference sample, standard, or level is matched to the subject sample based on at least one of the following criteria: age, weight, sex, stage of disease, and general health. A standard curve of concentrations of a purified protein or RNA, such as those described herein, within the normal reference range can also be used as a reference.

As used herein, the term "subject" refers to any organism to which a composition of the invention may be administered, for example, for experimental, diagnostic, prophylactic and/or therapeutic purposes. Typical subjects include all animals (eg, mammals such as mice, rats, rabbits, non-human primates and humans). A subject may seek or need treatment, need treatment, receive treatment, receive treatment in the future, or be a human or animal under the care of a trained professional for a particular disease or condition.

As used herein, the terms "treating", "treating" or "treating" mean therapeutic treatment or any action aimed at delaying (alleviating) an undesirable physiological state, disorder or disease or positive or desired clinical outcomes. Beneficial or desired clinical effects include, but are not limited to, alleviation of symptoms. a decrease in the severity of a disease, disorder, or disease; a stabilized (ie, no worsening) state, disorder, or disease state. Delaying the onset or delaying the condition, disorder or progression of the disease. improvement of the condition, disorder or disease state or remission (partial or complete); an improvement in at least one measurable physical parameter that is not necessarily noticeable to the patient; or worsening or improvement of the condition, disorder or disease. Treatment involves inducing a clinically significant response without undue side effects. Treatment also involves prolonging survival compared to expected survival without treatment. The compounds of the invention may also be used to "prophylactically treat" or "prevent" a disease, for example in a patient at increased risk of developing the disease.

As used herein, the terms "variant" and "derivative" are used interchangeably and refer to naturally occurring synthetic and semi-synthetic analogs of a compound, peptide, protein or other substance described herein. A variant or derivative of a compound, peptide, protein or other substance described herein may retain or enhance the biological activity of the starting material.

Details of one or more embodiments of the invention are set forth in the following description. Further features, details and advantages of the invention will be apparent from the description and claims.

BRIEF DESCRIPTION OF THE DRAWINGS

FIGO.1Figure 12 is a graph illustrating the inhibition of cell proliferation of various cancer cell lines by a BRG1/BRM inhibitor (Compound 87).

FIGO.2Figure 12 is a graph illustrating inhibition of choroidal melanoma cell proliferation by a BRG1/BRM inhibitor (Compound 87), a MEK inhibitor (Selumetinib), and a PKC inhibitor (LXS196).

FIGO.3Figure 12 is a graph illustrating inhibition of cell proliferation of various cancer cell lines by a BRG1/BRM inhibitor (Compound B).

DETAILED DESCRIPTION

The present disclosure provides compounds useful for inhibiting BRG1 and/or BRM. Such compounds can be used to modulate the activity of a BAF complex, for example in the treatment of a BAF-related disease such as cancer. Examples of compounds described herein include compounds having a structure according to formula I, formula IV, formula V or formula VII:

or a pharmaceutically acceptable salt thereof.

In certain embodiments, the compound or pharmaceutically acceptable salt thereof has the structure of any of compounds 1-241 in Tables 1, 2, 3 and 4.

Additional embodiments, as well as exemplary methods for synthesizing and preparing these compounds are described herein.

Pharmaceutical Applications

The compounds described herein are useful in the methods of the invention and, while not limited by theory, are believed to exert their ability to modulate the level, state and/or activity of a BAF complex, i.e. H. to inhibit activity of BRG1 and/or BRM proteins within the BAF complex in a mammal. Disorders associated with the BAF complex include, but are not limited to, disorders associated with BRG1 loss-of-function mutation.

One aspect of the present invention relates to methods of treating disorders associated with BRG1 loss-of-function mutations, such as e.g. B. Cancer (eg, non-small cell lung cancer, colon cancer, bladder cancer, cancer of unknown origin, glioma, breast cancer). , melanoma, non-melanoma skin cancer, endometrial cancer or penile cancer) to a person in need. Another aspect of the invention relates to methods of treating cancer by administering a compound that selectively inhibits BRM. For example, the compound that selectively inhibits BRM is a compound as described herein. In some embodiments, the compound is administered in an amount and for a period of time effective to result in one or more (eg, two or more, three or more, four or more) of: (a) reduction in tumor size; , (b ) reduced tumor growth rate, (c) increased tumor cell death, (d) reduced tumor progression, (e) reduced number of metastases, (f) reduced metastatic rate, (g) reduced tumor recurrence, (h) increased subject survival, (i) increased subject progression-free survival;

Cancer treatment can lead to a decrease in the size or volume of a tumor. For example, after treatment, the size of the tumor is reduced by 5% or more (eg, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more ) relative to pre-treatment size. The size of a tumor can be measured by any reproducible measurement method. For example, the size of a tumor can be measured in terms of the diameter of the tumor.

Cancer treatment can even lead to a reduction in the number of tumors. For example, after treatment, the number of tumors will increase by 5% or more (eg, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) in proportion reduced to the number before treatment. The number of tumors can be measured by any reproducible means of measurement, for example the number of tumors can be measured by counting the volumes that can be seen with the naked eye or at a certain magnification (eg 2×, 3×, 4×, 5) are visible ×, 10× or 50×).

Cancer treatment can lead to a reduction in the number of metastatic nodules in other tissues or organs located far from the site of the primary tumor. For example, after treatment, the number of metastatic nodules is reduced by 5% or more (eg, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more) . ) compared to the number before treatment. The number of metastatic nodules can be measured using any reproducible counting method. For example, the number of metastatic nodules can be measured by counting the metastatic nodules visible to the naked eye or at a certain magnification (eg, 2x, 10x, or 50x).

Cancer treatment can result in an increase in the median survival time of a population of subjects treated in accordance with the present invention compared to a population of subjects not receiving treatment. For example, median survival increases by more than 30 days (over 60 days, 90 days, or 120 days). The increase in the average survival time of a population can be measured by any reproducible means. An increase in the mean survival time of a population can be measured, for example, by calculating the mean survival time for a population after initiation of treatment with the compound of the invention. An increase in the mean survival time of a population can also be measured, for example, by calculating the mean survival time for a population after completion of a first course of treatment with a pharmaceutically acceptable salt of the invention.

Cancer treatment can also cause a reduction in the death rate of a treated population compared to an untreated population. For example, the death rate is reduced by more than 2% (eg, more than 5%, 10%, or 25%). The reduction in the death rate of a population of treated subjects can be measured by any reproducible means, for example by calculating the average number of disease-related deaths per unit time for a population after initiation of treatment with a pharmaceutically acceptable salt of the invention. A reduction in the death rate of a population can also be measured, for example, by calculating the average number of disease-related deaths per unit time for a population after completion of a first course of treatment with a pharmaceutically acceptable salt of the invention.

Examples of cancers that can be treated by the invention include, but are not limited to, non-small cell lung cancer, small cell lung cancer, colon cancer, bladder cancer, glioma, breast cancer, melanoma, non-melanoma skin cancer and endometrial cancer, esophageal cancer, pancreatic cancer, biliary liver cancer, soft tissue sarcoma, ovarian cancer, head and neck cancer, renal cell carcinoma, bone cancer, non-Hodgkin's lymphoma, cancer prostate, embryonal tumor, germ cell tumor, cervical cancer, thyroid cancer, salivary gland cancer, gastrointestinal neuroendocrine tumor, uterine sarcoma, gastrointestinal stromal tumor, CNS cancer, thymic tumor, adrenocortical carcinoma, appendectomy cancer, small bowel cancer, and cancer penis.

Combinatorial compositions and their use

The compounds of the invention can be combined with one or more therapeutically active substances. In particular, the therapeutic agent may be one that treats or prophylactically treats any cancer described herein.

Combined treatments

A compound of the invention may be used alone or in combination with an additional therapeutic agent, for example other agents for the treatment of cancer or related symptoms, or in combination with other types of therapies for the treatment of cancer. In combination therapies, the doses of one or more of the therapeutic compounds may be reduced from the standard doses when administered alone. For example, doses can be determined empirically from combinations and permutations of drugs or derived by isovolographic analysis (eg, Black et al., Neurology 65:S3-S6, 2005). In this case, the doses of the compounds in combination should achieve a therapeutic effect.

Σε ορισμένες πραγματοποιήσεις, ο δεύτερος θεραπευτικός παράγοντας είναι ένας χημειοθεραπευτικός παράγοντας (π.χ., ένας κυτταροτοξικός παράγοντας ή άλλη χημική ένωση χρήσιμη για τη θεραπεία του καρκίνου). Αυτά περιλαμβάνουν αλκυλιωτικούς παράγοντες, αντιμεταβολίτες, ανάλογα φολικού οξέος, ανάλογα πυριμιδίνης, ανάλογα πουρίνης και σχετικούς αναστολείς, αλκαλοειδή βίνκα, επιποδοπυιλοτοξίνες, αντιβιοτικά, L-ασπαραγινάση, αναστολείς τοποϊσομεράσης, ιντερφερόνες, σύμπλοκα συντονισμού διαιθυλενίου και παραγώγων πλατίνας, al κατασταλτικά, αδρενοκορτικοστεροειδή, παράγοντες κύησης, οιστρογόνα, αντιοιστρογόνα, ανδρογόνα, αντιανδρογόνα και ανάλογα ορμόνης απελευθέρωσης γοναδοτροπίνης. Περιλαμβάνονται επίσης 5-φθοροουρακίλη (5-FU), Λευκοβορίνη (LV), Ιρενοτεκάνη, Οξαλιπλατίνη, Καπεσιταβίνη, Πακλιταξέλη και Δοξεταξέλη. Μη περιοριστικά παραδείγματα χημειοθεραπευτικών παραγόντων περιλαμβάνουν αλκυλιωτικούς παράγοντες όπως θειοτέπα και κυκλοφωσφαμίδιο. αλκυλ σουλφονικά όπως βουσουλφάνη, ιμπροσουλφάνη και πιποσουλφάνη. αζιριδίνες όπως benzodopa, carboquon, meturedopa και uredopa. αιθυλενοϊμίνες και μεθυλομελαμίνες, συμπεριλαμβανομένων αλτρεταμίνης, τριαιθυλενομελαμίνης, τριαιθυλενοφωσφοραμιδίου, τριαιθυλενοθειοφωσφοραμιδίου και τριμεθυλολομελαμίνης. ακετογενίνες (ιδιαίτερα bullatacin και bullatacinone). μια καμπτοθεκίνη (συμπεριλαμβανομένου του συνθετικού αναλόγου τοποτεκάνης)· βρυοστατίνη; καλστατίνη? CC-1065 (συμπεριλαμβανομένων των συνθετικών αναλόγων του αδελεσίνη, καρζελεσίνη και μπιζελεσίνη). κρυπτοφυκίνες (ιδιαίτερα κρυπτοφυκίνη 1 και κρυπτοφυκίνη 8). δολαστατίνη; ντουοκαρμυκίνη (συμπεριλαμβανομένων των συνθετικών αναλόγων KW-2189 και CB1-TM1). ελευθεροβίνη; παγκρατιστατίνη; μια σαρκοδικτίνη? σπογγιστατίνη? μουστάρδες αζώτου όπως χλωραμβουκίλη, χλωρναφαζίνη, χολοφωσφαμίδη, οιστραμουστίνη, ιφωσφαμίδη, μεχλωραιθαμίνη, υδροχλωρική οξείδιο μεχλωροαιθαμίνης, μελφαλάνη, νοβεμβικίνη, φενεστερίνη, πρεδνιμουστίνη, τροφοσφαμίδη, τροφοσφαμίδη, νιτροζουρίες όπως καρμουστίνη, χλωροζοτοκίνη, φωτεμουστίνη, λομουστίνη, νιμουστίνη και ρανιμνουστίνη. Αντιβιοτικά όπως αντιβιοτικά ενεδιίνης (π.χ. καλιχεαμυκίνη, ιδιαίτερα καλιχεαμυκίνη γάμμα και καλιχεαμικίνη ωμεγάλη (βλ. π.χ. Agnew, Chem. Intl. Ed Engl. 33:183-186 (1994))· δυναμυκίνη, συμπεριλαμβανομένων των δυναμυκίνης, ασύνδετες, διφωτεινικές, διφωτεινικές, καθώς και χρωμοφόρα νεοκαρζινοστατίνη και χρωμοφόρα σχετιζόμενα με αντιβιοτικά με χρωμοπρωτεΐνη-ενδιίνη), ακλακινομυσίνες, ακτινομυκίνη, αυτραμυκίνη, αζασερίνη, βλεομυκίνη, κακτινομυκίνη, καραβικίνη, καμινομυκίνη, καρσινοφυλλίνη, χρωμομυκινο-νταμυκίνη, 5-Oxo- L-Norleucine, Adriamycin® (δοξορουβικίνη, συμπεριλαμβανομένης της μορφολινο-δοξορουβικίνης, κυανομορφολινο-δοξορουβικίνης, 2-πυρρολινο-δοξορουβικίνης και δεοξυδοξορουβικίνης), επιρουβικίνη, εσορουβικίνη, ιδαρουβικίνη, μαρκελομυκίνη, μιτομυκινομυκίνη cins, πεπλομυκίνη, Ποτφιρομυκίνη, Πουρομυκίνη, Χελαμυκίνη, Ροδορουβικίνη, Στρεπτονιγρίνη, Στρεπτοζοκίνη, Τουμπερκιδίνη, Ουμπενιμέξ, Ζινοστατίνη, Ζορουβικίνη; αντιμεταβολίτες όπως μεθοτρεξάτη και 5-φθοροουρακίλη (5-FU). ανάλογα φολικού οξέος όπως δενοπτερίνη, μεθοτρεξάτη, πτεροπτερίνη, τριμετρεξάτη. ανάλογα πουρίνης όπως fludarabine, 6-mercaptopurine, thiamiprine, thioguanine. ανάλογα πυριμιδίνης όπως ανσιταβίνη, αζακιτιδίνη, 6-αζαουριδίνη, καρμοφούρ, κυταραβίνη, διδεοξυουριδίνη, δοξιφλουριδίνη, ενοσιταβίνη, φλοξουριδίνη. ανδρογόνα όπως καλουστερόνη, προπιονική dromostanolone, epitiostanol, mepitiostan, testolactone. παράγοντες κατά των επινεφριδίων όπως αμινογλουτεθιμίδη, μιτοτάνη, τριλοστάνη. συμπληρώματα φολικού οξέος, όπως φρουλλικό οξύ. ακεγλατόνη; γλυκοσίδιο αλδοφωσφαμιδίου; αμινολεβουλινικό οξύ; ενιλουρακίλη; αμσακρίνη; μπεστραμπουκίλη; Bisantra; edatraxate; defofomin; ντεμεκολκίνη; diaziquon; Ελφομιτίνη; οξικό ελλειπτίνιο; μια εποθιλόνη? αιθογλυκίδιο; νιτρικό γάλλιο; υδροξυουρία; lentinan? λονιδαϊνίνη; Μαϊτανσινοειδή όπως μεϊτανσίνη και ανσαμιτοκίνη. Mitoguazón; μιτοξαντρόνη; μοπιδανμόλη; Νιτροερίνη; πεντοστατίνη; Φαινόμενο; πιραρουβικίνη; λοσοξαντρόνη; ποδοφυλλικό οξύ; 2-αιθυλυδραζίδιο; προκαρβαζίνη; Σύμπλεγμα πολυσακχαριτών PSK® (JHS Natural Products, Eugene, Oregon); Ραζοξάνη; ριζοξίνη; σιζοφουράνιο; σπειρογερμάνιο; Τενουαζονικό οξύ; triaziquon; 2,2',2"-τριχλωροτριαιθυλαμίνη· τριχοθεκένια (ιδιαίτερα τοξίνη Τ-2, βερρακουρίνη Α, ροριδίνη Α και αγγουιδίνη)· ουρεθάνη· βινδεσίνη· δακαρβαζίνη· μαννομουστίνη· μιτοβρονιτόλη· μιτολακτόλη· πιποβρομάνη· γακυτοσίνη· «αραβινοσίδη» (" κυκλοφωσφαμίδη, θειοτέπα, ταξοειδή, π.χ., πακλιταξέλη Taxol® (Bristol-Myers Squibb Oncology, Princeton, N.J.), ABraxane®, σκεύασμα νανοσωματιδίων πακλιταξέλης κατασκευασμένο με αλβουμίνη και απαλλαγμένο από κρεμοφόρες (American ΙΙΙ, Schneautical.) και Taxotere® doxetaxel (Rhône-Poulenc Rorer, Antony, Γαλλία), χλωρανβουκίλη, γεμσιταβίνη Gemzar®, 6-θειογουανίνη, μερκαπτοπουρίνη, μεθοτρεξάτη, σύμπλοκα συντονισμού πλατίνας, όπως σισπλατίνη, οξαλιπλατίνη και καρβοπλατίνη, καρβοπλατίνη -1-6, πλατίνη, 6-θειογουανίνη, μερκαπτοπουρίνη, μεθοτρεξάτη. Ιφοσφαμίδη, μιτοξαντρόνη, βινκριστίνη, Navelbine® βινορελβίνη, νοβαβαντρόνη, τενιποσίδη, εδατρεξάτη, δαουνομυκίνη, αμινοπτερίνη, Xeloda, ιβανδρονάτη, ιρινοτεκάνη (π.χ. CPT-11), αναστολέας τοποϊσομεράσης RFS διφθορινοϊνικό οξύ (ΡΕδνιοφθινοϊνοοειδές 2000) ; καπεσιταβίνη; και φαρμακευτικώς αποδεκτά άλατα, οξέα ή παράγωγα των παραπάνω. Δύο ή περισσότεροι χημειοθεραπευτικοί παράγοντες μπορούν να χρησιμοποιηθούν σε ένα κοκτέιλ που χορηγείται σε συνδυασμό με τον πρώτο θεραπευτικό παράγοντα που περιγράφεται εδώ. Κατάλληλα δοσολογικά σχήματα για συνδυαστικές χημειοθεραπείες είναι γνωστά στην τέχνη και περιγράφονται, για παράδειγμα, στο Saltz et al. περιγράφεται. (1999) Proc ASCO 18:233a και Douillard et αϊ. (2000) Lancet 355:1041-7.

In some embodiments, the second therapeutic agent is a biological therapeutic agent such as a cytokine (eg, interferon or an interleukin (eg, IL-2)) used in the treatment of cancer. In some embodiments, the biologic is an anti-angiogenic agent, such as an anti-VEGF agent, such as bevacizumab (Avastin®). In some embodiments, the biologic is an immunoglobulin-based biologic, e.g. B. a monoclonal antibody (eg, a humanized antibody, a fully human antibody, an Fc fusion protein, or a functional fragment thereof) that targets a target to stimulate an antitumor response or antagonizes an antigen relevant to the cancer. These drugs include Rituxan (rituximab). zenapax (daclizumab); Simulect (vasiliximab); Synagis (palivizumab); Remicade (infliximab); Herceptin (trastuzumab); Mylotarg (gemtuzumab ozogamicin); Campath (alemtuzumab); Zevalin (Ibritumomaabtiuxetane); Humira (adalimumab); Xolair (omalizumab); Bexxar (Tositumomab-1-131); Raptiva (efalizumab); Erbitux (cetuximab); Avastin (bevacizumab); Tysabri (natalizumab); Actemra (tocilizumab); Vectibix (panitumumab); Lucentis (ranibizumab); Soliris (eculizumab); Cimzia (certolizumab pegol); Simponi (golimumab); Ilaris (canakinumab); Stelara (ustekinumab); Arzerra (oftumumab); Prolia (Denosumab); Numax (motavizumab); ABThrax (raxibacumab); Benlysta (belimumab); Yervoy (ipilimumab); Adcetris (brentuximab vedotin); Perjeta (pertuzumab); Kadcyla (ado-trastuzumab emtansine); and Gazyva (obinutuzumab). Antibody-drug conjugates are also included.

The second agent may be a therapeutic agent, which is a non-pharmacological treatment. The second therapeutic means is, for example, radiotherapy, cryotherapy, hyperthermia and/or surgical removal of tumor tissue.

The second active ingredient may be a checkpoint inhibitor. In one embodiment, the checkpoint inhibitor is an inhibitory antibody (eg, a monospecific antibody such as a monoclonal antibody). For example, the antibody may be humanized or fully human. In some embodiments, the checkpoint inhibitor is a fusion protein, such as an Fc receptor fusion protein. In some embodiments, the checkpoint inhibitor is an agent, such as an antibody, that interacts with a checkpoint protein. In some embodiments, the checkpoint inhibitor is an agent, such as an antibody, that interacts with the ligand of a checkpoint protein. In some embodiments, the checkpoint inhibitor is an inhibitor (e.g., an inhibitory antibody or a small molecule inhibitor) of CTLA-4 (e.g., an anti-CTLA4 antibody, such as ipilimumab/yervoy or tremelimumab ). In some embodiments, the checkpoint inhibitor is an inhibitor (e.g., an inhibitory antibody or a small molecule inhibitor) of PD-1 (e.g., Nivolumab/Opdivo®; Pembrolizumab/Keytruda®; Pidilizumab/CT- 011). In some embodiments, the checkpoint inhibitor is an inhibitor (e.g., an inhibitory antibody or a small molecule inhibitor) of PDL1 (e.g., MPDL3280A/RG7446; MEDI4736; MSB0010718C; BMS 55993). In some embodiments, the checkpoint inhibitor is an inhibitor (e.g., an inhibitory antibody or an Fc or small molecule fusion inhibitor) of PDL2 (e.g., a PDL2/Ig fusion protein such as AMP 224). In some embodiments, the checkpoint inhibitor is an inhibitor (e.g., an inhibitory antibody or a small molecule inhibitor) of B7-H3 (e.g., MGA271), B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or a combination thereof.

In any of the combination embodiments described herein, the first and second therapeutic agents are administered simultaneously or sequentially in any order. The first therapeutic agent can be administered immediately, up to 1 hour, up to 2 hours, up to 3 hours, up to 4 hours, up to 5 hours, up to 6 hours, up to 7 hours, up to 8 hours and 9 hours, up to 10 hours, up to 11 hours, up to 12 hours, up to 13 hours, 14 hours, up to 16 hours, up to 17 hours, up to 18 hours, up to 19 hours up to 20 hours, up to 21 hours, up to 22 hours, up to 23 hours -7, 1-14, 1-21 or 1-30 days before or after the second therapeutic agent

pharmaceutical compositions

The compounds of the invention are preferably formulated into pharmaceutical compositions for administration to a mammal, preferably a human, in a biocompatible form suitable for in vivo administration. Accordingly, in one aspect, the present invention provides a pharmaceutical composition comprising a compound of the invention in admixture with a suitable diluent, carrier or excipient.

The compounds of the invention can be used in the form of free bases, in the form of salts, solvates and as prodrugs. All forms are within the scope of the invention. According to the methods of the invention, the described compounds or their salts, solvates or prodrugs can be administered to a patient in a variety of ways, depending on the route of administration chosen, as will be understood by those skilled in the art. The compounds of the invention may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump or transdermal administration and pharmaceutical compositions formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal and topical administration. Parenteral administration can be done by continuous infusion over a selected period of time.

Μια ένωση της εφεύρεσης μπορεί να χορηγηθεί από το στόμα, για παράδειγμα με ένα αδρανές αραιωτικό ή με έναν αφομοιώσιμο βρώσιμο φορέα, ή εγκλεισμένη σε κάψουλες σκληρής ή μαλακής ζελατίνης, ή συμπιεσμένη σε δισκία, ή ενσωματωμένη απευθείας με τροφή διαίτης. Για από του στόματος θεραπευτική χορήγηση, μια ένωση της εφεύρεσης μπορεί να αναμιχθεί με ένα έκδοχο και να χρησιμοποιηθεί με τη μορφή καταπίνων δισκίων, παρειακών δισκίων, παστίλιων, καψουλών, ελιξιρίων, εναιωρημάτων, σιροπιών και γκοφρετών. Μια ένωση της εφεύρεσης μπορεί επίσης να χορηγηθεί παρεντερικά. Διαλύματα μιας ένωσης της εφεύρεσης μπορούν να παρασκευαστούν σε νερό κατάλληλα αναμεμιγμένο με ένα τασιενεργό όπως υδροξυπροπυλοκυτταρίνη. Οι διασπορές μπορούν επίσης να γίνουν σε γλυκερίνη, υγρές πολυαιθυλενογλυκόλες, DMSO και μείγματα αυτών με ή χωρίς αλκοόλη και σε έλαια. Υπό κανονικές συνθήκες αποθήκευσης και χρήσης, αυτά τα παρασκευάσματα μπορεί να περιέχουν ένα συντηρητικό για την πρόληψη της ανάπτυξης μικροοργανισμών. Συμβατικές μέθοδοι και συστατικά για την επιλογή και την παρασκευή κατάλληλων σκευασμάτων περιγράφονται, για παράδειγμα, στο Remington's Pharmaceutical Sciences (2003, 20η έκδοση) και στο The United States Pharmacopeia: The National Formulary (USP 24 NF19), που δημοσιεύτηκε το 1999. Οι κατάλληλες φαρμακευτικές μορφές για ενέσιμη χρήση περιλαμβάνουν στείρα υδατικά διαλύματα ή διασπορές και στείρες σκόνες για την αυτοσχέδια παρασκευή στείρων ενέσιμων διαλυμάτων ή διασπορών. Σε όλες τις περιπτώσεις, το έντυπο πρέπει να είναι αποστειρωμένο και αρκετά ρευστό ώστε να χορηγείται εύκολα με σύριγγα. Οι συνθέσεις για ρινική χορήγηση μπορούν εύκολα να διαμορφωθούν ως αεροζόλ, σταγόνες, πηκτές και σκόνες. Τα σκευάσματα αεροζόλ τυπικά περιλαμβάνουν ένα λεπτό διάλυμα ή εναιώρημα του δραστικού συστατικού σε έναν φυσιολογικά αποδεκτό υδατικό ή μη υδατικό διαλύτη και συνήθως παρουσιάζονται σε αποστειρωμένη μορφή μονάδας ή πολλαπλών δόσεων σε σφραγισμένο δοχείο που μπορεί να έχει τη μορφή φυσιγγίου ή ξαναγέμισμα. για χρήση με νεφελοποιητή. Εναλλακτικά, ο σφραγισμένος περιέκτης μπορεί να είναι μια συσκευή μοναδιαίας διανομής όπως ένας ρινικός εισπνευστήρας μονάδας δόσης ή ένας διανομέας αεροζόλ μετρημένης δόσης που προορίζεται να απορριφθεί μετά τη χρήση. Όταν η μορφή δοσολογίας περιλαμβάνει έναν διανομέα αερολύματος, θα περιέχει ένα προωθητικό το οποίο μπορεί να είναι ένα πεπιεσμένο αέριο, π.χ. συμπιεσμένος αέρας, ή ένα οργανικό προωθητικό, π.χ. χλωροφθοράνθρακας. Οι δοσολογικές μορφές αερολύματος μπορεί επίσης να έχουν τη μορφή ψεκαστήρα αντλίας. Οι συνθέσεις κατάλληλες για στοματική ή υπογλώσσια χορήγηση περιλαμβάνουν δισκία, παστίλιες και παστίλιες στις οποίες το δραστικό συστατικό συνταγοποιείται με έναν φορέα όπως ζάχαρη, ακακία, τραγάκανθος, ζελατίνη και γλυκερίνη. Οι συνθέσεις για πρωκτική χορήγηση είναι βολικά στη μορφή υποθέτων που περιέχουν μια συμβατική βάση υπόθετων όπως το βούτυρο κακάο. Μια ένωση που περιγράφεται εδώ μπορεί να χορηγηθεί ενδοκαρκινικά, για παράδειγμα ως ενδοογκική ένεση. Η ενδοογκική ένεση είναι μια άμεση ένεση στα αγγεία του όγκου και λαμβάνεται υπόψη ειδικά για διακριτούς, συμπαγείς και προσβάσιμους όγκους. Η τοπική, περιφερειακή ή συστημική διαχείριση μπορεί επίσης να είναι κατάλληλη. Μια ένωση όπως περιγράφεται εδώ μπορεί πλεονεκτικά να έρθει σε επαφή με τη χορήγηση μιας ή περισσότερων ενέσεων στον όγκο, π.χ. σε διαστήματα περίπου 1 cm. Στην περίπτωση μιας χειρουργικής διαδικασίας, η παρούσα εφεύρεση μπορεί να χρησιμοποιηθεί προεγχειρητικά για να καταστεί ένας ανεγχείρητος όγκος επιδεκτικός σε εκτομή. Εάν είναι απαραίτητο, μπορεί επίσης να λάβει χώρα συνεχής χορήγηση, για παράδειγμα με εμφύτευση καθετήρα σε όγκο ή αγγειακό σύστημα όγκου.

The compounds of the invention may be administered to an animal, e.g., a human, alone or in combination with pharmaceutically acceptable carriers as indicated herein, the ratio of which is determined by the solubility and chemical nature of the compound, the chosen route of administration , and practice. typical pharmaceutical products.

dosages

The dosage of compounds of the invention and/or compositions comprising a compound of the invention may vary depending on many factors such as: B. the pharmacodynamic properties of the compound. the route of administration; the age, health and weight of the recipient; the nature and extent of symptoms; the frequency of treatment and the type of concomitant treatment, if any; and the rate of clearance of the compound in the treated animal. One skilled in the art can determine the appropriate dosage based on the above factors. The compounds of the invention may be administered initially at an appropriate dose, which may be adjusted as necessary depending on clinical response. In general, satisfactory results can be obtained when the compounds of the invention are administered to a human in a daily dose of, for example, 0.05 mg to 3000 mg (measured as solid form). The dosage range includes, for example, between 10-1000 mg (eg, 50-800 mg). In some embodiments, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 509, are composites.

Alternatively, the dose amount can be calculated based on the patient's body weight. For example, the dose of a compound or pharmaceutical composition thereof administered to a patient may be in the range of 0.1-50 mg/kg (eg, 0.25-25 mg/kg). In non-limiting exemplary embodiments, the dose may range from 0.5-5.0 mg/kg (e.g., 0.5, 1.0, 1.5, 2.0, 2.5, 3.0 , 3.5, 4.0, 4.5, or 5.0 mg/kg or 5.0 mg/kg) 20 mg/kg (eg, 5.5, 6.0, 6.5, 7 ,0, 7, 5, 8.0, 8.5, 9.0, 9.5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or kg).

EXAMPLES Example 1. Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 13)

Step 1: Preparation of tert-butyl N-[2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]carbamate (i-1a)

To a solution of 2-( tert -butoxycarbonylamino)acetic acid (1 g, 5.71 mmol) in dimethylformamide (DMF) (20 ml) was added 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazole . Hexafluorophosphate [4,5-b] 3-pyridinium 3-oxide (HATU) (2.39 g, 6.28 mmol) and N-ethyl-N-(propan-2-yl)propan-2-amine (DIPEA) (2.21 g, 17.13 mmol). ). The solution was stirred for 10 min and then 4-phenylthiazol-2-amine (1.01 g, 5.71 mmol) was added. The solution was stirred at 30°C for 12 hours. The reaction solution was poured into water (200 mL) and then extracted with EtOAc (200 mL). The organic layer was washed with aqueous citric acid (100 mL*5) and brine (200 mL) and then dried over Na2AFTERWARD4. The solution was concentrated in vacuo. The residue was purified by column chromatography (SiO 2 , PE/EtOAc=20/1~3/1) to give intermediate i-1a as a yellow solid.

1H-NMR (400 MHz, CDCl3) δ=10,08 (brs, 1H), 7,83-7,81 (m, 2H), 7,45-7,41 (m, 2H), 7,36- 7,34 (m, 1Η), 7,28 (s, 1Η), 7,17 (s, 1Η), 4,04 (s, 2Η), 1,51 (s, 9Η) ppm.

LCMS (ESI) m/z: [M+H]+=333,9.

Step 2: Preparation of 2-amino-N-(4-phenylthiazol-2-yl)acetamide hydrochloride (i-1b)

To a solution of intermediate i-1a (100 mg, 284.94 μmol) in dioxane (5 ml) was added HCl/dioxane (4M, 5 ml), then the solution was stirred at 30 °C for 1 h. The reaction solution was concentrated in vacuo to give intermediate i-1b (HCl salt) as a white solid, which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=233,9.

Step 3: Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 13)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (75.72 mg, 330.31 μmol) in DCM (10 mL) was added HATU (125.59 mg, 330.31 μmol) and DIPEA (155.24 mg, 1, 20 mmol). The solution was stirred for 10 min and then intermediate i-1b (81 mg, 300.28 μmol) was added. The solution was stirred at 30°C for 12 hours. The reaction solution was poured into water (50 mL) and extracted with EtOAc (100 mL). The organic layer was washed with brine (50 mL), dried (Na2AFTERWARD4) and concentrated in vacuo. MeOH (20 mL) was added to the residue and stirred for 20 min, forming a white solid. The solid was filtered and dried in vacuo to give compound 13 as a white solid.

LCMS (ESI) m/z: [M+H]+=444,9.

1H-NMR (400 MHz, DMSO-d6) δ=12.49 (s, 1H), 9.32 (t, J=5.6 Hz, 1H), 8.27-8.25 (m, 2H) , 7.96-7.90 (m, 3H), 7.81 -7.80 (m, 1H), 7.64 (s, 1H), 7.46-7.42 (m, 2H), 7 , 33-7.30 (m, 1H), 4.23 (d, J=5.6 Hz, 2H), 2.65 (s, 6H) ppm.

Example 2. Preparation of (S)-N-(4-(Methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide (Compound 42)

Step 1: Preparation of (S)-tert-butyl (4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)carbamate (intermediate i-2a)

To a solution of (2S)-2-(tert-butoxycarbonylamino)-4-methylsulfanylbutanoic acid (1 g, 4.01 mmol) in DCM (20 mL) was added 2-ethoxy-1-ethoxycarbonyl-1,2-dihydroquinoline (EEDQ). (EEDQ) 1.24 g, 5.01 mmol) and then 4-phenylthiazol-2-amine (589.05 mg, 3.34 mmol) was added. The mixture was stirred at 28°C for 3 hours. The reaction mixture was diluted with EtOAc (400 mL). The organic layer was washed with brine (40 mL) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2, petroleum ether/ethyl acetate = 50/1 to 8/1) to give intermediate i-2a as a white solid.

1H NMR (400 MHz, METHANOL-d4) δ 7.89 (d, J=7.6 Hz, 2H), 7.43-7.35 (m, 3H), 7.30-7.27 (m, 1H), 4.41-4.32 (m, 1H), 2.62-2.55 (m, 2H), 2.20-2.11 (m, 4H), 2.00-1.94 ( m, 1H), 1.46-1.39 (m, 9H) ppm.

LCMS (ESI) m/z: [M+H]+=408,2.

Step 2: Preparation of (S)-2-amino-4-(methylthio)-N-(4-phenylthiazol-2-yl)butanamide trifluoroacetate (intermediate i-2b)

To a solution of intermediate i-2a (200 mg, 490.74 μmol) in DCM (5 mL) was added TFA (0.25 mL). The mixture was stirred at 28°C for 2 h. To the reaction solution was added a.q NaHCO3to adjust pH to 7, then extract with EtOAc (100mL*2). The combined organic layers were washed with brine (20 mL) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue.

The residue was analyzed by preparative HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 15%-45%, 12 min) purified and lyophilized to give the TFA salt of intermediate i-2b as a white solid.

1H-NMR (400 MHz, METANOL-d4) δ 7,91-7,89 (m, 2Η), 7,46 (s, 1Η), 7,42-7,38 (m, 2Η), 7,32 - 7,31 (m, 1Η), 4,27-4,23 (m, 1Η), 2,67-2,63 (m, 2Η), 2,33-2,22 (m, 2Η), 2 , 14 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=308,1.

Step 3: Preparation of (S)-N-(4-(Methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide (Compound 42)

To a solution of benzoic acid (43.70 mg, 357.80 μmol) in DCM (2 ml) was added EEDQ (120.66 mg, 487.91 μmol) and then intermediate i-2b (100 mg, 325, 27 μmol). The mixture was stirred at 28°C for 3 hours. The reaction mixture was diluted with EA (150 mL), washed with 10% aqueous citric acid (50 mL * 4) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition, column: Phenomenex Synergi C18 150*25*10 µm, mobile phase: [water (0.1% TFA)-ACN]). B%: 45%-75%, 13 minutes cleaning. and lyophilized to give compound 42 as a white solid.

1H-NMR (400 MHz, METANOL-d4) δ 7,91-7,88 (m, 4Η), 7,60-7,51 (m, 1Η), 7,50-7,48 (m, 2Η) , 7,38–7,36 (m, 3Η), 7,29–7,27 (m, 1Η), 4,93–4,89 (m, 1Η), 2,70–2,63 (m, 2Η), 2,28-2,21 (m, 2Η), 2,13 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=412,2.

HPLC-Quirale: AD-3S_3_40_3ML_12MIN_T35.M, 4.866 Min.

Example 3. Preparation of (S)-3-(N,N-dimethylsulfamoyl)-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl )benzamide (Compound 24)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (59.66 mg, 260.22 μmol) in DCM (2 mL) was added EEDQ (96.52 mg, 390.33 μmol) followed by (2S)-2-amino- 4-methylsulfanyl-N- given. (4-Phenylthiazol-2-yl)butanamide (80 mg, 260.22 µmol) was added. The mixture was stirred at 28°C for 2 h. The reaction mixture was diluted with EtOAc (150 mL). The organic layer was washed with brine (50 mL) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 50/1 to 1:1) and lyophilized to give Compound 24 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12.55 (s, 1H), 9.10 (d, J=6.8 Hz, 1H), 8.28-8.26 (m, 2H), 7.92-7.89 (m, 3H), 7.80-7.76 (m, 1H), 7.64 (s, 1H), 7.45-7.41 (m, 2H), 7, 36 -7.30 (m, 1H), 4.83-4.76 (m, 1H), 2.65 (s, 6H), 2.61- 2.55 (m, 2H), 2.21- 2.16 (m, 2H), 2.10 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=519,2.

Example 4. Preparation of (S)-3-(N,N-dimethylsulfamoyl)-N-(1-oxo-1-((4-phenylthiazol-2-yl)amino)propan-2-yl)benzamide (Compound 12 ). )

Step 1: Preparation of (S)-tert-butyl (1-oxo-1-((4-phenylthiazol-2-yl)amino)propan-2-yl)carbamate (intermediate 4a)

To a solution of (2S)-2-(tert-butoxycarbonylamino)propanoic acid (200 mg, 1.06 mmol) in THE (5 mL) was added EEDQ (392.09 mg, 1.59 mmol) followed by 4-phenylthiazol- 2-amine (186.29 mg, 1.06 mmol) was added to the mixture. The mixture was stirred at 25°C for 12 hours. 80 mL (40 x 2) of citric acid (10%) was added and the reaction mixture was extracted with EtOAc (100 mL x 2). The combined organic layers were washed with brine (20 mL) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2, petroleum ether/ethyl acetate = 50/1 to 5:1) to give intermediate i-4a as a white solid.1HNMR (400 MHz, DMSO-d6) δ 12.29 (s, 1H), 7.96-7.84 (m, 2H), 7.63 (s, 1H), 7.49-7.39 (m , 2H), 7.35-7.30 (m, 1H), 7.25 (d, J=6.8 Hz, 1H), 4.31-4.20 (m, 1H), 1.38 ( s, 7H), 1.28 (d, J=7.2 Hz, 4H) ppm.

LCMS (ESI) m/z: [M+H]+=347,9.

SFC-HPLC: AD-3-MeOH(DEA)-5-40-3 mL-35T.I, 1.584 Min.

Step 2: Preparation of (S)-2-amino-N-(4-phenylthiazol-2-yl)propanamide (intermediate i-4b)

To a solution of intermediate i-4a (300 mg, 863.48 μmol) in DCM (5 mL) was added TFA (0.5 mL). The mixture was stirred at 25°C for 2 hours. The reaction solution was poured into water (20 mL) and the pH was adjusted to 7 with NaHCO 33. The reaction mixture was extracted with EA (100 ml*2). The combined organic layers were washed with brine (40 mL) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give intermediate i-4b as a white solid which was used in the next step without further purification.

1HNMR (400 MHz, DMSO-d6) δ 7.93-7.87 (m, 2H), 7.60 (s, 1H), 7.46-7.39 (m, 2H), 7.35-7 .28 (m, 1H), 5.46 (s, 2H), 3.59-3.57 (m, 1H), 3.33 (s, 3H), 1.23 (d, J=6.8 Hz, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=248,0.

SFC-HPLC: OD-3-MeOH(DEA)-5-40-3 mL-35T.I, 1.708 Min.

Step 3: Preparation of (S)-3-(N,N-dimethylsulfamoyl)-N-(1-oxo-1-((4-phenylthiazol-2-yl)amino)propan-2-yl)benzamide (Compound 12 ). )

To a solution of 3-(dimethylsulfamoyl)benzoic acid (92.70 mg, 404.34 μmol) in DCM (3 mL) was added HATU (169.12 mg, 444.78 μmol) and DIPEA (156.77 mg, 1, 21 mmol), then 292 mmol. Intermediate i-4b (100 mg, 404.34 μmol) was added to the mixture. The mixture was stirred at 25°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a solid. To the solid was added MeOH (80 mL) and stirred for 10 min and then filtered, the solid was collected by filtration and dried in vacuo to give compound 12 as a white solid.

1HNMR (400 MHz, DMSO-d6) δ 12.48 (s, 1H), 9.13 (d, J=6.4 Hz, 1H), 8.30-8.23 (m, 2H), 7, 92-7.89 (m, 3H), 7.80-7.76 (m, 1H), 7.64 (s, 1H), 7.46-7.41 (m, 2H), 7.36- 7.29 (m, 1H), 4.76-4.69 (m, 1H), 2.65 (s, 6H), 1.49 (d, J=7.2 Hz, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=459,0.

SFC-HPLC: OD-3-EtOH(DEA)-5-40-3 mL-35T.Ic, 2.455 Min.

Example 5. Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-(3-phenylanilino)ethyl]benzamide (Compound 40)

Step 1: Preparation of tert-butyl N-[2-oxo-2-(3-phenylanilino)ethyl]carbamate (intermediate i-5a)

To a solution of 2-( tert -butoxycarbonylamino)acetic acid (113.87 mg, 650.03 μmol) in DCM (3 mL) was added DIPEA (229.12 mg, 1.77 mmol) and HATU (337.04 mg, 886 .41 μmol). The mixture was stirred at 25°C for 10 min. 3-Phenylaniline (100.00 mg, 590.94 µmol) was added and the mixture was stirred at 25°C for 2 h. The reaction mixture was quenched by addition of water (10 mL) and then extracted with ethyl acetate (10 mL * 2). The combined organic layers were washed with water (10 mL*2) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a crude product. A solvent (EA:PE = 1:5, v/v, 10 ml) was added to the residue and stirred for 10 min, then filtered, the solid was dried under vacuum to obtain intermediate i-5a as a pale yellow solid result. .

LCMS (ESI) m/z: [M+Na]+=349,2.

Step 2: Preparation of 2-amino-N-(3-phenylphenyl)acetamide trifluoroacetate (intermediate i-5b)

To a solution of intermediate i-5a (70.11 mg, 201.48 μmol) in DCM (3 mL) was added TFA (150.00 μL). The mixture was stirred at 25°C for 1 hour. The reaction mixture was evaporated to dryness to give a residue which was taken up with methyl-t-butyl ether (5 ml) and the solid collected by filtration and dried in vacuo to give intermediate i-5b (TFA salt) as an open yellow to give a solid which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=227,2.

Step 3: Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-(3-phenylanilino)ethyl]benzamide (Compound 40)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (45.59 mg, 198.87 μmol) in DCM (3 mL) was added DIPEA (77.11 mg, 596.62 μmol) and HATU (113.43 mg, 298, 31 μmol). The mixture was stirred at 25°C for 10 min, then intermediate i-5b was added and the mixture was stirred at 25°C for 2 h. The reaction mixture was concentrated in vacuo to give a residue. To the residue was added a mixture of methanol:water = 1:1 v/v. (4 ml) was added and stirred for 10 min, then filtered, the filter cake was washed with methanol (2 ml) and then lyophilized to give compound 40 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 10.22 (s, 1H), 9.24 (m, 1H), 8.19-8.32 (m, 2H), 7.90-8.00 (m, 2H), 7.73-7.86 (m, 1H), 7.56-7.65 (m, 3H), 7.32-7.51 (m, 5H), 4.14 (m , 2H), 2.63-2.68 (m, 6H) ppm; LCMS (ESI) m/z: [M+H] + =438.2.

Example 6. Preparation of 3-(methylsulfamoyl)-N-[2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 26)

Step 1: Preparation of 2-amino-N-(4-phenylthiazol-2-yl)acetamide trifluoroacetate (intermediate 6a)

TFA (2.22 ml) was given. The mixture was stirred at 25°C for 1 hour. The reaction mixture was evaporated to dryness. MTBE/PE = (2/1, 30 ml) was added to the residue and stirred for 10 min, then filtered to give a solid which was dried in vacuo to give intermediate i-6a (TFA salt) as a yellow solid for to be delivered.

LCMS (ESI) m/z: [M+H]+=234,2.

Step 2: Preparation of 3-(Methylsulfamoyl)-N-[2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 26)

To a mixture of 3-(methylsulfamoyl)benzoic acid (46.13 mg, 214.33 μmol) in DCM (2 mL) was added HATU (89.64 mg, 235.75 μmol) and DIPEA (138.5 mg, 1, 07 mmol) at 25°C. The mixture was stirred at 25°C for 10 min and then intermediate 6a (50 mg, 143.96 μmol) was added and stirred for 1 h. The reaction mixture was evaporated to dryness to give the crude product. MeOH (30 ml) was added to the residue and stirred for 10 min, then filtered and the filter cake washed with MeOH (30 ml). The solid was dried in vacuo to give compound 26 as a white solid.

1HNMR (400 MHz, DMSO-d6) δ=12,47 (brs, 1Η), 9,25–9,23 (m, 1Η), 8,31 (s, 1Η), 8,16 (d, J= 7,9 Hz, 1H), 7,96 (d, J=7,8 Hz, 1H), 7,90 (m, 2H), 7,76-7,74 (m, 1H), 7,64 ( s, 1Η), 7,00-7,58 (m, 1Η), 7,46-7,40 (m, 2Η), 7,32 -7,29 (m, 1Η), 4,23 (d, J=5,6 Hz, 2Η), 2,43 (m, 3Η) ppm; LCMS (ESI) m/z: [M+H]+=431,1.

Example 7. Preparation of the compound N-(2-Oxo-2-((4-phenylthiazol-2-yl)amino)ethyl)-3-sulfamoylbenzamide (Compound 28)

Step 1: Preparation of N-(2-Oxo-2-((4-phenylthiazol-2-yl)amino)ethyl)-3-sulfamoylbenzamide (Compound 28)

To a solution of 2-amino-N-(4-phenylthiazol-2-yl)acetamide (50 mg, 143.96 μmol) in DCM (2.0 mL) was added 0.96 μmol of 3-sulfamoylbenzoic acid (28.97 mg, 143 μmol) and DIPEA (74.42 mg, 575.85 μmol, 100.30 μL). HATU (54.74 mg, 143.96 μmol) was then added to the reaction. The mixture was stirred at 25°C for 2 hours. Water (80.0 mL) was added and the reaction mixture was extracted with EtOAc (80.0 mL). The combined organic layers were washed with brine (20.0 mL) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. DCM (80.0 mL) was added to the residue, followed by stirring for 10 min. The solid was filtered and dried in vacuo to give compound 28 as an off-white solid.

1HNMR (400 MHz, DMSO-d6) δ=12.47 (s, 1H), 9.21-9.18 (m, 1H), 8.37 (s, 1H), 8.12 (d, J= 7.6 Hz, 1H), 8.01 (d, J=8.0 Hz, 1H), 7.91-7.90 (m, 2H), 7.74-7.72 (m, 1H), 7.64 (s, 1H), 7.48-7.42 (m, 4H), 7.33-7.30 (m, 1H), 4.23 (d, J=5.6 Hz, 2H) ppm.

LCMS (ESI) m/z: [M-t-Bu]+=417,2.

Example 8. Preparation of 3-methylsulfonyl-N-[2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 23)

Step 1: Preparation of 2-amino-N-(4-phenylthiazol-2-yl)acetamide trifluoroacetate (intermediate 8a)

TFA (2.22 ml) was given. The mixture was stirred at 25°C for 1 hour. The reaction mixture was evaporated to dryness. MTBE/PE = (2/1, 30 ml) was added to the residue and stirred for 10 min and then filtered to give a solid which was dried in vacuo to give intermediate i-8a (TFA salt) as yellow to give solid.

LCMS (ESI) m/z: [M+H]+=234,2.

Step 2: Preparation of 3-methylsulfonyl-N-[2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 23)

To a solution of 3-methylsulfonylbenzoic acid (42.91 mg, 214.32 μmol) in DCM (2 mL) was added HATU (89.64 mg, 235.75 μmol) and DIPEA (138.5 mg, 1.07 μmol) given at 25°C. The mixture was stirred for 10 min and then intermediate i-8a (50 mg, 143.96 μmol) was added and the mixture was stirred at 25°C for 1 h. The reaction mixture was evaporated to dryness. To the residue was added 30 mL MeOH and stirred for 10 min, a white solid formed which was filtered and dried in vacuo to give compound 23 as an off-white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12,44 (br s, 1Η), 9,26-9,25 (m, 1Η), 8,44 (s, 1Η), 8,23 (d, J =7,6 Hz, 1H), 8,10 (d, J=7,6 Hz, 1H), 7,91-7,89 (m, 2H), 7,80-7,78 (m, 1H ), 7,63 (s, 1Η), 7,45-7,41 (m, 2Η), 7,32-7,30 (m, 1Η), 4,24 (d, J=6,0 Hz, 2Η), 3,27 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=416,1.

Example 9. Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-[[4-(2-pyridyl)thiazol-2-yl]amino]ethyl]benzamide (Compound 32)

Step 1: Preparation of 2-amino-N-[4-(2-pyridyl)thiazol-2-yl]acetamide trifluoroacetate (intermediate 9a)

For a solution of tert -butyl N -[2-oxo-2-[[4-(2-pyridyl)thiazol-2-yl]amino]ethyl]carbamate (250 mg, 665.39 μmol) in DCM (2 mL) TFA (1.10 ml) was added. The mixture was stirred at 25°C for 1 hour. The reaction mixture was evaporated to dryness. MTBE/PE = (2/1, 30 ml) was added to the residue and stirred for 10 min, then filtered. The solid was dried in vacuo to give intermediate i-9a (TFA salt) as a yellow solid.

LCMS (ESI) m/z: [M+H]+=234,9.

Step 2: Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-[[4-(2-pyridyl)thiazol-2-yl]amino]ethyl]benzamide (Compound 32)

To a mixture of 3-(dimethylsulfamoyl)benzoic acid (98.73 mg, 430.66 μmol) in DCM (2 mL) was added HATU (180.13 mg, 473.73 μmol) and DIPEA (278.30 mg, 2, 15 °C) at 15 °C. The mixture was stirred at 25°C for 10 min, then intermediate 9a (150 mg, 430.66 μmol) was added and stirred for 1 h. The reaction mixture was evaporated to dryness. MeOH (30 ml) was added to the residue and stirred for 10 min, then filtered and the filter cake washed with MeOH (30 ml). The filter cake was evaporated to dryness to give compound 32 as an off-white solid.

1HNMR (400 MHz, DMSO-d6) δ=12,49 (br s, 1Η), 9,30-9,28 (m, 1Η), 8,60 (d, J=4,0 Hz, 1Η), 8,27–8,21 (m, 2H), 7,98–7,92 (m, 2H), 7,92–7,86 (m, 1H), 7,85–7,80 (m, 2H ), 7,36-7,32 (m, 1Η), 4,24 (d, J=5,6 Hz, 2Η), 2,66 (s, 6Η) ppm; LCMS (ESI) m/z: [M+H]+=446,2.

Example 10. Preparation of N-[(1S)-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 45)

Step 1: Preparation of tert-butyl N-[(1S)-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]carbamate (intermediate i-10a)

To a solution of (2S)-2-(tert-butoxycarbonylamino)butanoic acid (200.0 mg, 984.08 μmol) in DCM (5.0 mL) was added EEDQ (331.85 mg, 1.34 mmol) and 4 -phenylthiazole-157.67 ( mg, 894.62 µmol) administered in one dose at 20°C under N2. The mixture was stirred at 20°C for 8 hours. The reaction mixture was diluted with EtOAc (50.0 mL) and washed with citric acid (10%) (50.0 mL*3). The organic phase was washed with saturated brine (20.0 ml * 3) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2, petroleum ether/ethyl acetate = 20/1) to give intermediate i-10a as a white solid.

1H-NMR (400 MHz, CDCl3) δ 9,90 (br s, 1H), 7,94-7,77 (m, 2H), 7,50-7,38 (m, 2H), 7,37- 7,31 (m, 1Η), 7,15 (s, 1Η), 5,10 (s largo, 1Η), 4,36 (s largo, 1Η), 1,50 (s, 9Η), 1,40 - 1,22 (m, 2Η), 1,07-0,97 (m, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=362,2.

Step 2: Preparation of (2S)-2-amino-N-(4-phenylthiazol-2-yl)butanamide trifluoroacetate (intermediate i-10b)

To a solution of intermediate i-10a (300.00 mg, 829.97 μmol) in DCM (10.0 mL) was added TFA (1.08 mL) in one portion at 20 °C under N22. The mixture was stirred at 20°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give intermediate i-10b (TFA salt) as a white solid, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=262,2.

Step 3: Preparation of N-[(1S)-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 45)

To a mixture of HATU (73.95 mg, 194.48 μmol) and benzoic acid (17.42 mg, 142.62 μmol) in DCM (10.0 mL) was added DIPEA (67.03 mg, 518.62 μmol) in one dose at 20 °C2. The mixture was stirred at 20°C for 10 min. Then, intermediate i-10b (50.0 mg, 191.32 μmol) was added and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Boston pH-lex 150*25 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 47%-77%, 10 min) purified and lyophilized, for to give compound 45 as a white solid.

1H RMN (400 MHz, CDCl3) δ=8.00 (d, J=7.2 Hz, 2H), 7.84 (d, J=7.6 Hz, 2H), 7.64-7.54 (m, 1H), 7, 53-7.46 (m, 2H), 7.46-7.40 (m, 2H), 7.40-7.33 (m, 1H), 7.17 (s, 1H), 7.00 ( br d, J=8.0 Hz, 1H), 5.18-5.12 (m, 1H), 2.22-2.17 (m, 1H), 2.05-1.97 (m, 1H ), 1.14-1.11 (m, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=366,0.

HPLC solution: Amycoat-MeOH(DEA)-40-3 mL-35T, 1.417 min.

Example 11. Preparation of N-[(1S)-1-(hydroxymethyl)-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 47)

Step 1: Preparation of tert-butyl N-[(1S)-1-(hydroxymethyl)-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]carbamate (intermediate i-11a)

To a mixture of (2S)-2-(tert-butoxycarbonylamino)-3-hydroxypropanoic acid (200.0 mg, 974.62 μmol) in DCM (5.0 mL) was added EEDQ (328.66 mg, 1.33 mmol) and 4-phenylthiaz 2-amine (156.15 mg, 886.02 μmol) in one dose at 20 °C under N2. The mixture was stirred at 20°C for 3 hours. The reaction mixture was diluted with EtOAc (50.0 mL) and washed with citric acid (10%) (50.0 mL*3). The organic phase was washed with brine (20.0 mL*3) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2, petroleum ether/ethyl acetate = 5/1 to 2:1) to give intermediate i-11a as a colorless oil.

LCMS (ESI) m/z: [M+H]+=364,2.

Step 2: Preparation of ((2S)-2-amino-3-hydroxy-N-(4-phenylthiazol-2-yl)propanamide trifluoroacetate (intermediate i-11b)

To a mixture of intermediate i-11a (130.00 mg, 182.07 μmol) in DCM (10.0 mL) was added trifluoroacetic acid (1.0 mL) in one portion at 20 °C under N22. The mixture was stirred at 20°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give intermediate i-11b (TFA salt) as a white solid, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=264,2.

Step 3: Preparation of N-[(1S)-1-(hydroxymethyl)-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 47)

To a mixture of HATU (47.91 mg, 126.00 μmol) and benzoic acid (11.29 mg, 92.48 μmol) in DCM (2.0 mL) was added DIPEA (43.43 mg, 336.04 μmol) given in a single dose at 20 °C2and the mixture was stirred at 20°C for 10 min. Then, intermediate i-11b (50.0 mg, 132.51 μmol) was added and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 35%-65%, 9 min) purified and lyophilized, for to give compound 47 as a white solid.

1H RMN (400 MHz, CDCl3) δ=7.93 (d, J=8.0 Hz, 2H), 7.84 (d, J=8.0 Hz, 2H), 7.63-7.57 (m, 1H), 7, 52-7.37 (m, 5H), 7.18 (s, 1H), 5.02-4.99 (m, 1H), 4.47-4.43 (m, 1H), 3.96- 3.92 (m, 1 H) ppm.

LCMS (ESI) m/z: [M+H]+=368,0.

HPLC solution: Amycoat-MeOH(DEA)-40-7 min-3 mL, 2.042 min

Example 12. Preparation of N-[(1S)-2-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 50)

Step 1: Preparation of tert-butyl N-[(1S)-2-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]carbamate (intermediate i-12a)

To a solution of (2S)-2-(tert-butoxycarbonylamino)-3-methylbutyric acid (200.0 mg, 920.55 μmol) in DCM (5.0 mL) was added EEDQ (310.42 mg, 1.26 mmol) and 4-phenylthiazole- was added. 2-amine (147.49 mg, 836.86 μmol) in one dose at 20 °C under N2. The mixture was stirred at 20°C for 8 hours. The reaction mixture was diluted with EtOAc (50.0 mL) and washed with citric acid (10%) (50.0 mL*3). The combined organic layers were washed with saturated brine (20.0 mL * 3) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2petroleum ether/ethyl acetate = 20/1) to give intermediate i-12a as a colorless oil.

LCMS (ESI) m/z: [M+H]+=376,2.

Step 2: Preparation of (2S)-2-amino-3-methyl-N-(4-phenylthiazol-2-yl)butanamide trifluoroacetate (intermediate i-12b)

To a mixture of intermediate i-12a (300.00 mg, 629.59 μmol) in DCM (10.0 mL) was added trifluoroacetic acid (1.0 mL) in one portion at 20 °C under N22. The mixture was stirred at 20°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give intermediate i-12b (TFA salt) as a white solid, which was used in the next step without purification.

LCMS (ESI) m/z: [M+H]+=276,2.

Step 3: Preparation of N-[(1S)-2-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 50)

To a mixture of HATU (61.81 mg, 162.56 μmol) and benzoic acid (14.56 mg, 119.23 μmol) in DCM (2.0 mL) was added DIPEA (56.03 mg, 433.53 μmol) in one dose at 20 °C2. The mixture was stirred at 20°C for 10 min. Then, intermediate i-12b (50.0 mg, 128.41 μmol) was added and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Boston pH-lex 150*25 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 50%-80%, 10 min) purified and lyophilized, for to give compound 50 as a white solid.

1H-NMR (400 MHz, CDCl3) δ 10,52 (br s, 1Η), 7,99 (d, J=8,0 Hz, 2H), 7,81 (d, J=8,0 Hz, 2H ), 7,61-7,54 (m, 1Η), 7,52-7,45 (m, 2Η), 7,43-7,36 (m, 2Η), 7,36-7,29 (m , 1Η), 7,16 (s, 1Η), 6,91 (d, J=8,0 Hz, 1Η), 5,24-5,09 (m, 1Η), 2,46-2,38 ( m, 1Η), 1,16-1,13 (m, 6Η).

LCMS (ESI) m/z: [M+H]+=380,0.

HPLC solution: Cellucoat-MeOH(DEA)-5-40-3 mL-35, 1,981 min.

Example 13. Preparation of N-[(1S)-1-[(4-phenylthiazol-2-yl)carbamoyl]butyl]benzamide (Compound 51)

Step 1: Preparation of tert-butyl N-[(1S)-1-[(4-phenylthiazol-2-yl)carbamoyl]butyl]carbamate (intermediate i-13a)

To a mixture of (2S)-2-(tert-butoxycarbonylamino)pentanoic acid (200.0 mg, 920.55 μmol) in DCM (5.0 mL) was added EEDQ (310.42 mg, 1.26 mmol) and 4 -phenylthiazole-2-4749 (mg, 836.86 µmol) administered as a single dose at 20°C under N2. The mixture was stirred at 20°C for 8 hours. The reaction mixture was diluted with EtOAc (50.0 mL) and washed with citric acid (10%) (50.0 mL*3). The combined organic layers were washed with saturated brine (20.0 mL * 3) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2petroleum ether/ethyl acetate = 20/1) to give intermediate i-13a as a colorless oil.

LCMS (ESI) m/z: [M+H]+=376,3.

Step 2: Preparation of (2S)-2-amino-3-methyl-N-(4-phenylthiazol-2-yl)butanamide trifluoroacetate (intermediate i-13b)

To a mixture of intermediate i-13a (300.00 mg, 661.55 μmol) in DCM (10.0 mL) was added trifluoroacetic acid (937.48 μL) in one portion at 20 °C.2. The mixture was stirred at 20°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give intermediate i-13b (TFA salt) as a white solid, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=276,2.

Step 3: Preparation of N-[(1S)-1-[(4-phenylthiazol-2-yl)carbamoyl]butyl]benzamide (Compound 40)

To a mixture of HATU (51.26 mg, 134.83 μmol) and benzoic acid (15.09 mg, 123.59 μmol) in DCM (2.0 mL) was added DIPEA (58.08 mg, 449.42 µmol) in a single dose at 20 °C2. The mixture was stirred at 20°C for 10 min. Then, intermediate i-13b (50.0 mg, 181.57 μmol, TFA salt) was added and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Boston pH-lex 150*25 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 50%-80%, 10 min) purified and lyophilized, for to give compound 51 as a white solid.

1H-NMR (400 MHz, CDCl3) δ 7,95-7,93 (m, 2H), 7,78-7,76 (m, 2H), 7,58-7,51 (m, 1H), 7 , 48-7,41 (m, 5Η), 7,12 (s, 1Η), 7,08-6,99 (m, 1Η), 5,04-4,99 (m, 1Η), 2,09 - 2,03 (m, 1Η), 1,95–1,92 (m, 1Η), 1,56–1,50 (m, 2Η), 1,04–1,00 (m, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=380,0.

HPLC solution: Cellucoat-MeOH(DEA)-5-40-3 mL-3, 2,141 min.

Example 14. Preparation of N-[(1S)-3-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]butyl]benzamide (Compound 48)

Step 1: Preparation of tert-butyl N-[(1S)-3-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]butyl]carbamate (intermediate i-14a)

To a mixture of (2S)-2-(tert-butoxycarbonylamino)-4-methylpentanoic acid hydrate (200.00 mg, 802.24 μmol) in DCM (5.0 mL) was added EEDQ (270.53 mg, 1.09 mmol) and 4-p. -amine (128.53 mg, 729.31 μmol) in one dose at 20°C under N2. The mixture was stirred at 20°C for 3 hours. The reaction mixture was diluted with EA (50.0 mL) and washed with citric acid (10%) (50.0 mL*3). The organic phase was washed with saturated brine (20.0 ml * 3) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 20/1) to give intermediate i-14a as a colorless oil.

LCMS (ESI) m/z: [M+H]+=390,3.

Step 2: Preparation of (2S)-2-amino-3-methyl-N-(4-phenylthiazol-2-yl)butanamide trifluoroacetate (intermediate i-14b)

To a solution of intermediate i-14a (170.00 mg, 392.80 μmol) in DCM (10.0 mL) was added trifluoroacetic acid (742.95 μL) in one portion at 20 °C under N22. The mixture was stirred at 20°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give intermediate i-14b (TFA salt) as a white solid, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=290,2.

Step 3: Preparation of N-[(1S)-3-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]butyl]benzamide (Compound 48)

To a mixture of HATU (69.13 mg, 181.81 μmol) and benzoic acid (16.28 mg, 133.31 μmol) in DCM (2.0 mL) was added DIPEA (62.66 mg, 484.82 μmol) given in a single dose at 20 °C2. The mixture was stirred at 20°C for 10 min. Then, intermediate i-14b (50.0 mg, 172.77 μmol TFA salt) was added and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. Purified by HPLC (TFA condition; Column: Boston pH-lex 150*25 10 µm; Mobile phase: [Water (0.1% TFA)-ACN]; B%: 53%-83%, 10 min) The compound is obtained 48 as a white solid.

1H-NMR (400 MHz, CDCl 3 ) δ 10.19 (br s, 1H), 7.92-7.87 (m, 2H), 7.86-7.81 (m, 2H), 7.61- 7.55 (m, 1H), 7.52-7.46 (m, 2H), 7.45-7.38 (m, 2H), 7.37-7.30 (m, 1H), 7, 16 (s, 1H), 6.59 (br d, J=8.0 Hz, 1H), 5.10-5.00 (m, 1H), 2.02-1.98 (m, 1H), 1.84-1.79 (m, 2H), 1.05-1.02 (m, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=394,1.

HPLC solution: Cellucoat-MeOH(DEA)-5-40-3 mL-3, 1,875 min.

Example 15. Preparation of (1S)-3-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]butyl]benzamide (Compound 41)

Step 1: Preparation of tert-butyl N-[(1S)-1-[(4-phenylthiazol-2-yl)carbamoyl]pentyl]carbamate (intermediate i-15a)

To a solution of (2S)-2-(tert-butoxycarbonylamino)hexanoic acid (200.0 mg, 864.72 μmol) in DCM (5.0 mL) was added EEDQ (291.60 mg, 1.18 mmol) and 4 -phenylthiazole-2-38.54 ( mg, 786.11 µmol) administered in a single dose at 20°C under N2. The mixture was stirred at 20°C for 8 hours. The reaction mixture was diluted with EtOAc (50.0 mL) and washed with citric acid (10%) (50.0 mL*3). The organic phase was washed with saturated brine (20.0 ml * 3) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2petroleum ether/ethyl acetate=20/1). Intermediate i-15a was obtained as a white solid.

LCMS (ESI) m/z: [M+H]+=390,3.

Step 2: Preparation of (2S)-2-amino-N-(4-phenylthiazol-2-yl)hexanamide (intermediate i-15b)

To a mixture of intermediate i-15a (230.00 mg, 590.48 μmol) in DCM (10.0 mL) was added trifluoroacetic acid (1.03 mL) in one portion at 20 °C under N22. The mixture was stirred at 20°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give intermediate i-15b (TFA salt) as a white solid, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=290,2.

Step 3: Preparation of N-[(1S)-3-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]butyl]benzamide (Compound 41)

To a mixture of HATU (67.32 mg, 177.05 μmol) and benzoic acid (15.86 mg, 129.84 μmol) in DCM (5.0 mL) was added DIPEA (61.02 mg, 472.15 μmol) given in a single dose at 20 °C2. The mixture was stirred at 20°C for 10 min. Then, intermediate i-15b (50.0 mg, 123.94 μmol) was added and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Boston pH-lex 150*25 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 53%-83%, 10 min) purified and lyophilized, for to give compound 41 as a white solid.

1H-NMR (400 MHz, CDCl3) δ 7,98-7,96 (m, 2H), 7,83-7,81 (m, 2H), 7,62-7,55 (m, 1H), 7 , 53-7,42 (m, 4Η), 7,41-7,35 (m, 1Η), 7,15 (s, 1Η), 7,04-6,95 (m, 1Η), 5,13 - 5,08 (m, 1Η), 2,15-2,11 (m, 1Η), 1,98-1,94 (m, 1Η), 1,51-1,41 (m, 4Η), 0 , 96-0,92 (m, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=394,0.

HPLC solution: Amycoat-MeOH(DEA)-40-3 mL-35T, 1,431 min

Example 16. Preparation of N-[(1S)-3-phenyl-1-[(4-phenylthiazol-2-)carbamoyl]propyl]benzamide (Compound 49)

Step 1: Preparation of tert-butyl N-[(1S)-3-phenyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]carbamate (intermediate i-16a)

To a mixture of 4-phenylthiazol-2-amine (114.71 mg, 650.91 μmol) in DCM (5 mL) was added EEDQ (241.44 mg, 976.36 μmol) and (2S)-2-(tert- butoxycarbonylamino)-4-p. -butanoic acid (200.0 mg, 716.00 μmol) in one dose at 20°C under N2. The mixture was stirred at 20°C for 8 hours. The reaction mixture was diluted with EtOAc (50.0 mL) and washed with citric acid (10%) (50.0 mL*3). The organic phase was washed with saturated brine (20.0 ml * 3) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2petroleum ether/ethyl acetate=10/1). Intermediate i-16a was obtained as a white solid.

LCMS (ESI) m/z: [M+H]+=438,3.

Step 2: Preparation of (2S)-2-amino-4-phenyl-N-(4-phenylthiazol-2-yl)butanamide (intermediate i-16b)

To a mixture of intermediate i-16a (270.00 mg, 569.67 µmol) in DCM (10.0 mL) was added trifluoroacetic acid (923.19 µL) in one portion at 20°C.2. The mixture was stirred at 20°C for 1 hour. The reaction mixture was concentrated under reduced pressure to give intermediate i-16b (TFA salt) as a yellow oil, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=338,2.

Step 3: Preparation of N-[(1S)-3-phenyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 49)

To a mixture of HATU (57.62 mg, 151.54 µmol) and benzoic acid (13.57 mg, 111.13 µmol) in DCM (2.0 mL) was added DIPEA (52.23 mg, 404.11 µmol) given in one dose at 20°C2. The mixture was stirred at 20°C for 10 min. Then, intermediate i-16b (50.0 mg, 110.75 μmol) was added and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was triturated in MeOH (10.0 mL), the solid collected by filtration and dried in vacuo to give compound 49 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.54 (s, 1H), 8.85 (d, J=7.2 Hz, 1H), 8.02-7.95 (m, 4H) , 7.69 (s, 1H), 7.67-7.61 (m, 1H), 7.61-7.54 (m, 2H), 7.53-7.46 (m, 2H), 7 , 42-7.29 (m, 5H), 7.28-7.22 (m, 1H), 4.80-4.74 (m, 1H), 2.87-2.75 (m, 1H) , 2.55-2.45 (m, 1H), 2.23-2.19 (m, 2H).

LCMS (ESI) m/z: [M+H]+=442,1.

HPLC solution: Amycoat-MeOH(DEA)-40-7 min-3 mL, 3.118 min

Example 17. Preparation of the compound 3-(dimethylsulfamoyl)-N-[2-[[4-(o-tolyl)thiazol-2-yl]amino]-2-oxo-ethyl]benzamide (Compound 16)

Step 1: Preparation of tert-Butyl (2-oxo-2-((4-(o-tolyl)thiazol-2-yl)amino)ethyl)carbamate (intermediate i-17a)

To a solution of 2-( tert -butoxycarbonylamino)acetic acid (184.15 mg, 1.05 mmol) in DCM (2.0 mL) was added DIEA (407.57 mg, 3.15 mmol) and HATU (439.65 mg, 1.16 mmol). After 0.5 h, 4-(o-tolyl)thiazol-2-amine (200.0 mg, 1.05 mmol) was added and the mixture was stirred at 20°C for 1.5 h. The reaction mixture was poured into water (10.0 mL) and extracted with EtOAc (10.0 mL*3). The combined organic layers were washed with brine (10.0 mL) and dried over Na2AFTERWARD4filtered and concentrated. The crude product was triturated with MTBE (10.0 mL). The solid was collected by filtration and dried in vacuo to give intermediate i-17a as a white solid.

1H RMN (400 MHz, CDCl3) δ 10,53-10,01 (m, 1Η), 7,51 (s, 1Η), 7,32-7,30 (m, 6Η), 6,94 (s, 1Η), 5,01 ( s largo, 1H), 3,87 (s largo, 2H), 2,41 (s, 3H), 1,46 (s, 9H) ppm

LCMS (ESI) m/z: [M+H]+=348,1

Step 2: Preparation of 2-amino-N-(4-(o-tolyl)thiazol-2-yl)acetamide (intermediate i-17b)

To a solution of intermediate i-17a (210 mg, 604.44 μmol) in DCM (2.1 ml) was added TFA (0.3 ml) at 20 °C. The mixture was stirred at 20°C for 16 hours. The reaction mixture was poured into sat. pour water3(10.0 mL) and extracted with EtOAc (20.0 mL*3). The combined organic extracts were washed with brine (20.0 mL) and dried over Na2AFTERWARD4filtered and concentrated. The residue was purified by column chromatography (SiO).2, PE:EA=10:1 to 8:1 to 5:1) to give intermediate i-17b as a brown oil.

LCMS (ESI) m/z: [M+H]+=248,1

Step 3: Preparation of -(dimethylsulfamoyl)-N-[2-[[4-(o-tolyl)thiazol-2-yl]amino]-2-oxo-ethyl]benzamide (Compound 16)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (111.24 mg, 485.21 μmol) in DCM (2.0 mL) was added HATU (202.94 mg, 533.73 μmol) and DIEA (188.13 μmol) . mg, 1.46 mmol). The mixture was stirred at 20°C for 0.5 h. Intermediate D (120.0 mg, 485.21 µmol) was added and the mixture was stirred at 20°C for 1.5 h. The reaction mixture was poured into water (10.0 mL) and extracted with EtOAc (10.0 mL*3). The combined organic extract was washed with brine (10.0 mL) and dried over Na2AFTERWARD4filtered and concentrated. The crude product was triturated with MeOH (10.0 mL). The solid was collected by filtration and dried in vacuo to give compound 16 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12,40 (s, 1Η), 9,31-9,28 (m, 1Η), 8,26-8,22 (m, 2Η), 7,95 - 7,93 (m, 1Η), 7,82-7,80 (m, 1Η), 7,59-7,57 (m, 1Η), 7,27-7,25 (m, 4Η), 4 , 22 (d, J=5,6 Hz, 2H), 2,65 (s, 6H), 2,43 (s, 3H) ppm

LCMS (ESI) m/z: [M+H]+=459,0

Example 18. Preparation of the compound 3-(dimethylsulfamoyl)-N-[2-[[4-(m-tolyl)thiazol-2-yl]amino]-2-oxo-ethyl]benzamide (Compound 1)

Step 1: Preparation of 4-(m-tolyl)thiazol-2-amine (intermediate i-18a)

In a mixture of 1-(m-tolyl)ethanone (100.0 mg, 745.30 μmol), thiourea (85.10 mg, 1.12 mmol) and 12 (189.16 mg, 745.30 μmol) in EtOH (1.0 mL) became 6 mg, NaF (30). µmol). The mixture was stirred at 20°C for 3 hours. The reaction mixture was poured into water (8.0 mL) and extracted with EtOAc (10.0 mL*3). The combined organic layers were washed with brine (10.0 mL) and dried over Na2AFTERWARD4filtered and concentrated in vacuo to give intermediate i-18a as a green oil which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=191,1

Step 2: Preparation of tert-Butyl (2-oxo-2-((4-(m-tolyl)thiazol-2-yl)amino)ethyl)carbamate (intermediate i-18b)

To a solution of 2-( tert -butoxycarbonylamino)acetic acid (138.11 mg, 788.38 μmol) in DCM (2.0 mL) was added DIEA (305.68 mg, 2.37 mmol) and HATU (329.74 mg , 867.2 μmol) at 202 μmol C. The mixture was stirred at 20°C for 0.5 h. Intermediate i-18a (150.0 mg, 788.38 μmol) was added and the mixture was stirred at 20°C for 1.5 h. The reaction mixture was poured into water (10.0 mL) and extracted with EtOAc (10.0 mL*3). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4, filtered and concentrated to give intermediate i-18b as a brown oil, which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=348,1

Step 3: Preparation of 2-amino-N-(4-(m-tolyl)thiazol-2-yl)acetamide (intermediate i-18c)

To a solution of intermediate i-18b (75 mg, 215.87 μmol) in DCM (1.0 mL) was added TFA (0.25 mL) at 20 °C. The mixture was stirred at 20°C for 2 hours. The reaction mixture was poured into saturated sodium hydroxide solution2CO3(10.0 mL) and extracted with EtOAc (10.0 mL*3). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4filtered and concentrated. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 15%-45%, 9 min) to give a fraction. The fraction was then concentrated in vacuo and then poured into saturated sodium hydroxide solution2CO3(15.0 mL) and extracted with EtOAc (20.0 mL*3). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4filtered and concentrated to give intermediate i-18c as a yellow oil.

LCMS (ESI) m/z: [M+H]+=248,1

Step 4: Preparation of 3-(dimethylsulfamoyl)-N-[2-[[4-(m-tolyl)thiazol-2-yl]amino]-2-oxo-ethyl]benzamide (Compound 1)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (46.35 mg, 202.17 μmol) in DCM (1.0 mL) was added HATU (84.56 mg, 222.39 μmol) and DIEA (78.39 μmol, 606.51 μmol) in the mixture. stirred at 20°C for 0.5 h. Then, intermediate i-18c (50.0 mg, 202.17 μmol) was added and the mixture was stirred at 20°C for 1.5 h. The reaction mixture was poured into water (5.0 mL) and extracted with EtOAc (10.0 mL*3). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4filtered and concentrated. The residue was purified by preparative HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 42%-72%, 9 min) and lyophilized Compound 1 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.45 (s, 1H), 9.30-9.27 (m, 1H), 8.26-8.22 (m, 2H), 7, 94 (d, J=7.6 Hz, 1H), 7.82 -7.77 (m, 1H), 7.73-7.65 (m, 2H), 7.60 (s, 1H), 7 , 32-7.30 (m, 1H), 7.14 (d, J=7.6 Hz, 1H), 4.23 (d, J =5.6 Hz, 2H), 2.65 (s, 6H), 2.35 (s, 3H) ppm

LCMS (ESI) m/z: [M+H]+=459,0

Example 19. Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-[[4-(p-tolyl)thiazol-2-yl]amino]ethyl]benzamide (Compound 31)

Step 1: Preparation of tert-butyl N-[2-oxo-2-[[4-(p-tolyl)thiazol-2-yl]amino]ethyl]carbamate (intermediate i-19a)

To a solution of 2-( tert -butoxycarbonylamino)acetic acid (184.15 mg, 1.05 mmol) in DCM (2.0 mL) was added DIEA (407.57 mg, 3.15 mmol) and HATU (439.65 mg, 1.16 mmol). After 0.5 h, 4-(o-tolyl)thiazol-2-amine (200.0 mg, 1.05 mmol) was added to the mixture and stirred at 20°C for 1.5 h. The reaction mixture was poured into water (10.0 mL) and extracted with EtOAc (10.0 mL*3). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4filtered and concentrated. The crude product was triturated with MTBE (10.0 mL). The solid was collected by filtration and dried in vacuo to give intermediate i-19a as a white solid.

1H-NMR (400 MHz, CDCl3) δ 9,75 (br s, 1Η), 7,71 (d, J=8,0 Hz, 2H), 7,23 (d, J=8,0 Hz, 2H ), 7,11 (s, 1Η), 5,22-5,03 (m, 1Η), 4,20-3,94 (m, 2Η), 2,39 (s, 3Η), 1,50 ( s , 9 STUNDEN).

LCMS (ESI) m/z: [M+H]+=348,1

Step 2: Preparation of 2-amino-N-[4-(p-tolyl)thiazol-2-yl]acetamide (i-19b)

To a solution of intermediate i-19a (210 mg, 604.44 μmol) in DCM (2.1 mL) was added TFA (0.3 mL) at 20 °C. The mixture was stirred at 20°C for 16 hours. The reaction mixture was poured into saturated sodium hydroxide solution2CO3(10.0 mL) and extracted with EtOAc (20.0 mL*3). The combined organic extracts were washed with brine (20.0 mL) and dried over Na2AFTERWARD4filtered and concentrated. The crude product was triturated with MTBE (10.0 mL). The solid was collected by filtration and dried in vacuo to give intermediate i-19b as a white solid.

LCMS (ESI) m/z: [M+H]+=248,1

Step 3: Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-[[4-(p-tolyl)thiazol-2-yl]amino]ethyl]benzamide (Compound 31)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (139 mg, 606.32 μmol) in DCM (2.0 mL) was added DIEA (235.09 mg, 1.82 mmol) and HATU (253.59 mg, 666.95 μmol) and the mixture was stirred at 20 °C for 0.5 h. Then, intermediate D (150.00 mg, 606.51 μmol) was added at 20°C and the mixture was stirred at 20°C for 1.5 h. The reaction mixture was poured into water (10.0 mL) and extracted with EtOAc (10.0 mL*3). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4filtered and concentrated. The crude product was triturated with MTBE (10.0 mL). The solid was collected by filtration and dried in vacuo to give compound 31 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,45 (s, 1Η), 9,30-9,27 (m, 1Η), 8,26-8,22 (m, 2Η), 7, 94 (d, J=8,0 Hz, 1H), 7,82 -7,77 (m, 3H), 7,56 (s, 1H), 7,24 (d, J=8,0 Hz, 2H ), 4,23 (d, J=5,6 Hz, 2H), 2,65 (s, 6H), 2,32 (s, 3H) ppm

LCMS (ESI) m/z: [M+H]+=459,0

Example 20. Preparation of N-[2-[[4-(2,6-dimethylphenyl)thiazol-2-yl]amino]-2-oxoethyl]-3-(dimethylsulfamoyl)benzamide (Compound 29)

In a solution of N-[2-[(4-bromothiazol-2-yl)amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (50 mg, 111.78 μmol) and (2,6-dimethylphenyl) boronic acid (50.29 mg, 335.33 μmol) in dioxane (2 mL) and H20 (0.1 mL) of DIPEA (28.89 mg, 223.55 μmol) was added, followed by Pd(t-Bu).3PI)2(28.56 mg, 55.89 μmol). The reaction mixture was degassed three times and then stirred at 100°C for 12 hours. The reaction solution was diluted with EtOAc (100 mL) and then washed with NaHCO 33(100 mL) and brine (100 mL), the organic layer was dried over Na2AFTERWARD4and then concentrated in vacuo. The residue was purified by Prep-HPLC (Combiflash, CH).3CN/H2O: 5-95 %, SMALL3OH) to give compound 29 as a pink solid.

1HNMR (400 MHz, MeOD) δ=8.34 (s, 1H), 8.23 ​​(d, J=7.2 Hz, 1H), 8.01 (d, J=8.0 Hz, 1H), 7.80–7.76 (m, 1H), 7.20–7.16 (m, 1H), 7.11–7.09 (m, 2H), 6.91 (s, 1H), 4, 36 (s, 2H), 2.76 (s, 6H), 2.12 (s, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=473,3.

Example 21. Preparation of the compound N-[2-Oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]-5,6-dihydro-4H-cyclopenta[b]thiophene-2-carboxamide (Compound 79 ). )

For a solution of 5,6-dihydro-4H-cyclopenta[b]thiophene-2-carboxylic acid (30.0 mg, 178.35 μmol), HATU (67.81 mg, 178.35 μmol) and DIPEA (83, 82 mg, 648.513 μmol. μL) To DCM (0.5 mL) was added 2-amino-N-(4-phenylthiazol-2-yl)acetamide (56.31 mg, 162.13 μmol, TFA-salt). The mixture was stirred at 20°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was triturated with MeOH (10.0 mL), the solid collected by filtration and dried in vacuo to give compound 79 as a white solid

1H-NMR (400 ΜΗζ, DMSO-d6) δ=12.41 (s, 1H), 8.79-8.76 (m, 1H), 7.91 (d, J=7.2 Hz, 2H), 7.64-7.58 (m , 2H), 7.46-7.42 (m, 2H), 7.37 -7.30 (m, 1H), 4.15 (d, J=6.0 Hz, 2H), 2.90- 2.87 (m, 2H), 2.78-2.69 (m, 2H), 2.41-2.37 (m, 2H) ppm.

LCMS (ESI) m/z: [M+H]+=384,0.

Example 22. Preparation of N-[(1S)-4-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]pentyl]benzamide (Compound 46)

Step 1: Preparation of tert-butyl N-[(1S)-4-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]pentyl]carbamate (intermediate i-22a)

In a solution of (2S)-2-(tert-butoxycarbonylamino)-5-methylhexanoic acid (153.11 mg, 624.15 μmol) and EEDQ (210.47 mg, 851.12 μmol) in DCM (2.0 mL ) of 4-2-phenylol 100.0 mg, 567.41 µmol was added), the mixture was stirred at 20°C for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2, petroleum ether/ethyl acetate = 10:1 to 2:1) to give intermediate i-22a as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,33 (s, 1Η), 7,91 (d, J=7,2 Hz, 2Η), 7,63 (s, 1Η), 7,49 - 7,40 (m, 2H), 7,37-7,28 (m, 1H), 7,20 (d, J=7,6 Hz, 1H), 4,31-4,11 (m, 1H ), 2,50-2,06 (m, 5Η), 1,73-1,56 (m, 2Η), 1,55-1,45 (m, 1Η), 1,39 (s, 7Η), 1,27 (s, 2Η), 0,86-0,84 (m, 6Η) ppm.

LCMS (ESI) m/z: [M+H]+=404,2.

Step 2: Preparation of (2S)-2-amino-5-methyl-N-(4-phenylthiazol-2-yl)hexanamide trifluoroacetate (intermediate i-22b)

To a solution of intermediate i-22a (150.0 mg, 371.71 μmol) in DCM (1.0 mL) was added TFA (0.1 mL, 1.35 mmol) and the mixture was stirred at 20° for 12 h C stirred. The reaction mixture was concentrated under reduced pressure to give intermediate i-22b (TFA salt) as a white solid, which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=304,2.

Step 3: Preparation of N-[(1S)-4-methyl-1-[(4-phenylthiazol-2-yl)carbamoyl]pentyl]benzamide (Compound 46)

To a solution of benzoic acid (16.09 mg, 131.75 μmol, 20.11 μL), HATU (50.10 mg, 131.75 μmol) and DIPEA (61.92 mg, 479.11 μmol) in DCM are added (1.0 mg, intermediate (1.0 mL, i.2) 119.78 μmol, TFA salt was added), the mixture was stirred at 30 °C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 58%-88%, 9 min) and lyophilized to give Compound 46 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12.46 (s, 1H), 8.68 (d, J=7.2 Hz, 1H), 7.93-7.90 (m, 4H), 7.63 (s, 1H), 7.60-7.54 (m, 1H), 7.53-7.47 (m, 2H), 7.44-7.42 (m, 2H), 7, 37 -7.28 (m, 1H), 4.67-4.61 (m, 1H), 1.87-1.83 (m, 2H), 1.59-1.56 (m, 1H), 1.48-1.32 (m, 1H), 1.31-1.15 (m, 1H), 0.90-0.88 (m, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=408,2

HPLC solution: Cellucoat-MeOH(DEA)-5-40-3 mL-35T.Icm, 1.982 min.

Example 23. Preparation of 3-(dimethylsulfamoyl)-N-[(1S)-1-[(4-phenylthiazol-2-yl)carbamoyl]butyl]benzamide (Compound 17)

To a mixture of HATU (36.25 mg, 95.35 μmol) and 3-(dimethylsulfamoyl)benzoic acid (20.04 mg, 87.40 μmol) in DCM (2.0 mL) was added DIPEA (41.07 mg, 317.82 μmol in 1 μmol in 1 μmol) C under N2. The mixture was stirred at 20°C for 10 min. Then (2S)-2-amino-N-(4-phenylthiazol-2-yl)pentanamide (50.0 mg, 128.41 μmol, TFA salt) was added and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition, column: Boston pH-lex 150*25 10 μm, mobile phase: [water (0.1% TFA)-ACN], B%: 57%-76%, 8 min) and lyophilized to give compound 17 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12.52 (s, 1H), 9.05 (d, J=7.6 Hz, 1H), 8.29-8.27 (m, 2H), 7.93-7.90 (m, 3H), 7.81- 7.79 (m, 1H), 7.64 (s, 1H), 7.54-7.39 (m, 2H), 7, 38 -7.27 (m, 1H), 4.76-4.70 (m, 1H), 2.68-2.65 (m, 6H), 1.86-1.83 (m, 2H), 1.58-1.31 (m, 2H), 0.96-0.92 (m, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=487,1.

HPLC solution: Amycoat-MeOH(DEA)-40-7 min-3 mL, 2.022 min

Example 24. Preparation of compound N-[(1S)-1-methyl-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 44)

Step 1: Preparation of tert-butyl N-[(1S)-1-methyl-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]carbamate (intermediate i-24a)

To a solution of (2S)-2-(tert-butoxycarbonylamino)propanoic acid (2.0 g, 10.57 mmol) in DCM (20.0 mL) was added EEDQ (3.56 g, 14.41 mmol) and 4- phenylthiazol-2-amine (1.69 g, 9.61 mmol) were given in one portion at 20 °C under N2. The mixture was stirred at 20°C for 8 hours. The reaction mixture was diluted with EtOAc (50.0 mL) and washed with citric acid (10%) (50.0 mL*3). The organic phase was washed with brine (50.0 mL * 3) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2, petroleum ether/ethyl acetate = 10/1 to 3/1) to give intermediate i-24a as a white solid.

1H-NMR (400 ΜΗζ, DMSO-d6) δ=12,30 (s, 1Η), 7,90 (d, J=8,0 Hz, 2Η), 7,63 (s, 1Η), 7,45-7,41 (m, 2Η), 7,36–7,30 (m, 1H), 7,26 (d, J=8,0 Hz, 1H), 4,31–4,24 (m, 1H), 1,38–1,28 ( m, 12Η) ppm.

LCMS (ESI) m/z: [M+H]+=348,2.

Step 2: Preparation of (2S)-2-amino-N-(4-phenylthiazol-2-yl)propanamide trifluoroacetate (intermediate i-24b)

To a solution of intermediate i-24a (200.0 mg, 575.65 μmol) in DCM (10.0 mL) was added trifluoroacetic acid (1.0 mL) in one portion at 20 °C under N22. The mixture was stirred at 20°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give intermediate i-24b (TFA salt) as a yellow oil, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=248,2.

Step 3: Preparation of N-[(1S)-1-methyl-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 44)

To benzoic acid (37.18 mg, 304.42 μmol) in DCM (5.0 mL) was added DIPEA (143.07 mg, 1.11 mmol) and HATU (157.84 mg, 415.12 μmol) in one dose given at 20 °C under N.2and the mixture was stirred at 20°C for 10 min. Then, intermediate i-24b (100.0 mg, 276.75 μmol) was added and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Boston pH-lex 150*25 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 52%-72%, 8 min) purified and lyophilized, for to give compound 44 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12,38 (br s, 1Η), 8,75 (d, J=8,0 Hz, 1Η), 7,94-7,90 (m, 4Η) , 7,64 (s, 1Η), 7,60-7,54 (m, 1Η), 7,53-7,47 (m, 2Η), 7,47-7,41 (m, 2Η), 7 , 37-7,29 (m, 1Η), 4,74-4,67 (m, 1Η), 1,48 (d, J=8,0 Hz, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=352,3.

HPLC solution: Cellucoat-MeOH(DEA)-5-40-3 mL-35T.Icm, 2,262 min.

Example 25. Preparation of (S)-3-Methyl-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide (Compound 76 ) . )

To a solution of 3-methylbenzoic acid (17.77 mg, 130.54 μmol) in DCM (1 mL) was added HATU (74.45 mg, 195.81 μmol) and DIPEA (50.61 mg, 391.61 μmol) µmol), then (2S) Amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (40.13 mg, 130.54 µmol) was added. The mixture was stirred at 25°C for 2 hours. The reaction mixture was concentrated under reduced pressure. The residue was analyzed by preparative HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 μm; mobile phase: [water(0.1% TFA)-ACN]; B%: 55%-85%, 9 min) purified and lyophilized to give compound 76 as a white solid.

1Η NMR (400 MHz, METANOL-d4) δ 7,90 (d, J=7,2 Hz, 2Η), 7,73-7,70 (m, 2Η), 7,42-7,34 (m, 5Η), 7,30-7,27 (m, 1Η), 4,93-4,89 (m, 1Η), 2,71-2,60 (m, 2Η), 2,42 (s, 3Η) , 2,30-2,17 (m, 2Η), 2,14 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=426,0.

SFC-HPLC: OD-3_5CM_MEOH(DEA)_5_40_3ML_T3, 2.236 Min.

Example 26. Preparation of the compound (S)-4-Methyl-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide ( Compound 54)

To a solution of 4-methylbenzoic acid (17.77 mg, 130.54 μmol) in DCM (1 mL) was added HATU (74.45 mg, 195.81 μmol) and DIPEA (50.61 mg, 391.62 μmol) and then (2S)-2- 4-Methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (40.13 mg, 130.54 µmol) was added. The mixture was stirred at 25°C for 2 hours. The reaction mixture was concentrated under reduced pressure. The residue was analyzed by preparative HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 μm; mobile phase: [water(0.1% TFA)-ACN]; B%: 55%-85%, 9 min) purified Compound 54 is obtained as a white solid.

1HNMR (400 MHz, METANOL-d4) δ 7,90 (d, J=7,2 Hz, 2H), 7,81 (d, J=8,0 Hz, 2H), 7,42-7,36 ( m, 3Η), 7,33-7,28 (m, 3Η), 4,93-4,89 (m, 1Η), 2,75-2,62 (m, 2Η), 2,41 (s, 3Η), 2,30-2,21 (m, 2Η), 2,14 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=426,0.

HPLC-Quirale: OD-3_5CM_MEOH(DEA)_40_3ML_T35.M, 1.673 Min.

Example 27. Preparation of (S)-3-(Methylsulfonyl)-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide ( connection 3)

To a solution of 3-methylsulfonylbenzoic acid (19.00 mg, 94.91 μmol) in DCM (2 mL) was added HATU (54.13 mg, 142.36 μmol) and DIPEA (49.07 mg, 379.64 μmol) . µmol), then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (40 mg, 94.91 µmol) was added. The mixture was stirred at 10°C for 2 hours. The reaction mixture was concentrated under reduced pressure. The rest was preparation. HPLC (TFA condition; Column: Phenomenex Synergi C18 150*25*10 µm; Mobile phase: [Water(0.1%TFA)-ACN]; B%: 48%-78%, 9 min) yielding compound 3 as white solid.

1HNMR (400 MHz, METANOL-d4) δ 8,50 (s, 1Η), 8,24 (d, J=8,0 Hz, 1Η), 8,16 (d, J=8,0 Hz, 1Η) , 7,91-7,88 (m, 2Η) 7,79 -7,77 (m, 1Η), 7,42-7,36 (m, 3Η), 7,31-7,30 (m, 1Η ), 4,96-4,92 (m, 1Η), 3,18 (s, 3Η), 2,77-2,66 (m, 2Η), 2,36-2,20 (m, 2Η), 2,15 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=490,0.

Example 28. Preparation of 3-(tert-butyl)-N-(2-oxo-2-((4-phenylthiazol-2-yl)amino)ethyl)benzamide (Compound 65)

To a solution of 3-tert-butylbenzoic acid (25.66 mg, 143.96 μmol) in DCM (1 mL) was added HATU (82.11 mg, 215.94 μmol) and DIPEA (93.03 mg, 719.81 μmol, 125). The reaction mixture was stirred at 25°C for 0.5 h, then 2-amino-N-(4-phenylthiazol-2-yl)acetamide (50 mg, 143.96 μmol) was added. The reaction mixture was stirred at 25°C for 2 h. The reaction mixture was diluted with MeCN (2 mL) and a white precipitate formed. The precipitate was filtered and lyophilized to give compound 65 as a white solid.

LCMS (ESI) m/z: [M+H]+=394,3.

1H-NMR (400 MHz, DMSO-d6) δ=12,44 (s, 1Η), 8,96-8,94 (m, 1Η), 7,94-7,90 (m, 3Η), 7, 73 (d, J=8,0 Hz, 1Η), 7,64 (s, 1Η), 7,60 (br d, J=8,4 Hz, 1Η), 7,47-7,42 (m, 3Η), 7,34-7,31 (m, 1Η), 4,21 (d, J=5,6 Hz, 2Η), 1,33 (s, 9Η) ppm.

Example 29. Preparation of 3-(2-Hydroxypropan-2-yl)-N-(2-oxo-2-((4-phenylthiazol-2-yl)amino)ethyl)benzamide (Compound 61)

Step 1: Preparation of 3-(2-hydroxypropan-2-yl)benzoic acid (intermediate i-29a)

To a solution of 3-bromobenzoic acid (500 mg, 2.49 mmol) in THE (10 mL) was added n-BuLi (2 mL, 4.97 mmol, 2.5 M) at -65 °C under N22. The reaction mixture was stirred at -65°C for 10 min, then acetone (365 µL, 4.97 mmol) was added at -65°C. The reaction mixture was gradually warmed to 25°C and then stirred at room temperature for 50 min. The reaction mixture was poured into 20 mL of saturated aqueous ammonium chloride. The mixture was adjusted to pH=4 with 2M HCl. The mixture was then extracted with EtOAc (30 mL x 5). The combined organic extracts were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. MeCN was added to the residue to produce a precipitate. The precipitate was filtered and dried in vacuo to give intermediate i-29a as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=8.09 (m, 1H), 7.80-7.76 (m, 1H), 7.72-7.67 (m, 1H), 7, 43 (m, 1H), 5.15 (br s, 1H), 1.44 (s, 6H) ppm.

LCMS (ESI) m/z: [M-H2O+H]+=163,1.

Step 2: Preparation of 3-(2-Hydroxypropan-2-yl)-N-(2-oxo-2-((4-phenylthiazol-2-yl)amino)ethyl)benzamide (Compound 61)

To a solution of intermediate i-29a (25.94 mg, 143.96 μmol) in DCM (1 ml) was added HATU (82.11 mg, 215.94 μmol) and DIPEA (93.03 mg, 719.80 μmol ). µmol). The reaction mixture was stirred at 25°C for 0.5 h. 2-Amino-N-(4-phenylthiazol-2-yl)acetamide (50 mg, 143.96 µmol, TFA salt) was then added. The reaction mixture was stirred at 25°C for 2 h. The reaction mixture was diluted with MeCN (2 mL) and a white precipitate formed. The precipitate was filtered and lyophilized to give compound 61 as a white solid.

LCMS (ESI) m/z: [M+H]+=396,3.

1H-NMR (400 MHz, DMSO-d6) δ=8,93-8,90 (m, 1Η), 8,01 (s, 1Η), 7,90 (d, J=7,6 Hz, 2Η) , 7,73 (d, J=8,0 Hz, 1H), 7,65 (d, J=8,0 Hz, 1H), 7,63 (s, 1H), 7,45-7,40 ( m, 3Η), 7,35-7,30 (m, 1Η), 5,14 (s, 1Η), 4,19 (d, J=5,6 Hz, 2H), 1,46 (s, 6H ) ppm.

Example 30. Preparation of (S)-3-(tert-butyl)-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide (Union 73)

To a solution of 3-tert-butylbenzoic acid (21.14 mg, 118.64 μmol), DIPEA (82.66 μmol) in DCM (0.5 mL) was added HATy (67.66 mg, 177.96 μmol), giving 0°C. After the addition, the mixture was stirred at this temperature for 15 min and then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 μmol) was added ) 0 °C The resulting mixture was stirred at 20 °C for 2 h. The reaction mixture was diluted with water (1 ml) and extracted with DCM (2x1 ml). The combined organic extracts were washed with brine (2 ml) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give the residue. The residue was purified by prep-HPLC (Combiflash, CH).3CN/H20.5-95%, FA condition) to give compound 73 as a white solid.

LCMS (ESI) m/z: [M+H]+=468,3.

1H-NMR (400 MHz, DMSO-d6) δ=12,62-12,30 (br s, 1Η), 8,73 (d, J=7,2 Hz, 1Η), 7,91-7,89 (m, 3Η), 7,76 (d, J=8,0 Hz, 1Η), 7,63 (s, 1Η), 7,62-7,57 (m, 1Η), 7,43-7, 41 (m, 3Η), 7,35-7,30 (m, 1Η), 4,79-4,74 (m, 1Η), 2,66- 2,50 (m, 2Η), 2,14- 2,12 (m, 2Η), 2,10 (s, 3Η), 1,32 (s, 9Η) ppm.

Example 31. Preparation of (S)-3-(2-Hydroxypropan-2-yl)-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butane-2 -yl)benzamide (Compound 55)

In a solution of 3-(1-hydroxy-1-methylethyl)benzoic acid (21.38 mg, 118.64 μmol), DIPEA (82.66 μL) in DCM (0.5 mL), HATU (67.66 mg, 177.96 µmol) given at 0 °C. The mixture was stirred at this temperature for 15 min and then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 μmol) was added at 0 ° The resulting mixture was stirred at 20°C for 12 hours. The reaction mixture was diluted with water (1 ml) and extracted with DCM (2x1 ml). The combined organic extracts were washed with brine (2 ml) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give the residue. The residue was purified by reverse phase HPLC (FA condition) to give compound 55 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,48 (s, 1Η), 8,71 (br d, J=7,2 Hz, 1Η), 8,00 (s, 1Η), 7, 90 (d, J=7,6 Hz, 2H), 7,77 (d, J=8,0 Hz, 1H), 7,65-7,62 (m, 2H), 7,45-7,41 (m, 3Η), 7,33-7,32 (m, 1Η), 5,11 (s, 1Η), 4,78-4,77 (m, 1Η), 2,66-2,56 (m , 2Η), 2,16-2,12 (m, 2Η), 2,10 (s, 3Η), 1,45 (s, 6Η) ppm.

LCMS (ESI) m/z: [M+H]+=470,1.

Example 32. Preparation of 4-methyl-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]-3-methylsulfonylbenzamide (Compound 2)

Step 1: Preparation of 4-methyl-3-methylsulfonylbenzoic acid (intermediate i-32a)

For a mixture of methyl 4-methyl-3-methylsulfonylbenzoate (100 mg, 438.09 μmol) in THE (1 mL), MeOH (1 mL), and H20 (0.5 mL) of LiOH.H was added2(55.15 mg, 1.31 mmol) and the mixture was stirred at 30°C for 2 h. Water (20 ml) was added and the pH was adjusted to 4 with 2N HCl and then the mixture was extracted with EtOAc (20 ml*2). The combined organic phase was washed with brine and dried over anhydrous Na2AFTERWARD4filtered and concentrated in vacuo to give intermediate i-32a as a white solid which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=215,2.

Step 2: Preparation of 4-methyl-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]-3-methylsulfonylbenzamide (Compound 2)

To a mixture of intermediate i-32a (20.33 mg, 94.91 μmol) in DCM (1 ml) was added HATU (43.30 mg, 113.89 μmol) and DIEA (36.80 mg, 284.73 μmol ) and the mixture was stirred at 15°C. for 5 min Then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (40 mg, 94.91 μmol, TFA salt) was added and the mixture was allowed to stir for 16 h at 15 °C. The solvent was removed under reduced pressure and then the residue was purified by prep. Purified by HPLC (column: Phenomenex Synergi C18 150*25*10 µm, mobile phase: [water (0.1% TFA)-ACN]). B%: 42%-72%, 9 min) and lyophilized to give compound 2 as a white solid

1H-NMR (400 MHz, MeOD) δ=8.53 (d, J=1.6 Hz, 1H), 8.11-8.09 (m, 1H), 7.91-7.89 (m, 2H), 7.57 (d, J=8.0, 1H), 7.40–7.37 (m, 3H), 7.31–7.27 (m, 1H), 4.93–4, 91 (m, 1H), 3.19 (s, 3H), 2.77 (s, 3H), 2.72-2.61 (m, 2H), 2.33-2.18 (m, 2H) , 2.14 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=504,2.

Example 33. Preparation of methyl 3-[2-[[2-[[3-(dimethylsulfamoyl)benzoyl]amino]acetyl]amino]thiazol-4-yl]benzoate (Compound 9)

In a solution of N-[2-[(4-bromothiazol-2-yl)amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (30 mg, 67.07 μmol) and (3-methoxycarbonylphenyl)boric acid (36.21 mg, 201.21 µmol) in 1,4-dioxane (3 mL) and H20 (0.2 ml) of Pd(t-Bu) was added.3PI)2(17.14 mg, 33.53 μmol) e DIEA (43.34 mg, 335.35 μmol, 58.41 μL) sob N2. The mixture was stirred at 100°C for 2 h and then poured into H 22O (20 mL) and extracted with EtOAc (5 mL*3). The combined organic extracts were washed with H2O (5 mL*2) and brine (5 mL*1) were dried over Na2AFTERWARD4filtered and concentrated in vacuo. The residue was purified by Prep-HPLC (column: Phenomenex Gemini 150*25 mm*10 µm; mobile phase: [water (0.04% NH)]3H 2 O + 10 mM NH4HCO3)-ACN]; B%: 36-66%, 10 min) and lyophilized to give compound 9 as a white solid.

1H-NMR (400 ΜΗζ, DMSO-d6) δ 12,58 (s, 1H), 9,33-9,31 (m, 1H), 8,56 (s, 1H), 8,27-8,24 (m, 2H), 8,18 ( d, J=8,0 Hz, 1Η), 7,94-7,91 (m, 2Η), 7,83-7,81 (m, 2Η), 7,62-7,60 (m, 1Η) , 4,25 (d, J=5,6 Hz, 2H), 3,89 (s, 3H), 2,66 (s, 6H) ppm

LCMS (ESI) m/z: [M+H]+=503,3.

Example 34. Preparation of 3-[2-[[2-[[3-(dimethylsulfamoyl)benzoyl]amino]acetyl]amino]thiazol-4-yl]-N-methylbenzamide (Compound 4)

For a solution of N-[2-[(4-bromothiazol-2-yl)amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (30 mg, 67.07 μmol) and [3-(methylcarbamoyl) phenyl ] boronic acid (36.01 mg, 201.21 μmol) in 1,4-dioxane (3 mL) and H2O (0.15 ml) Pd(t-Bu) was added.3PI)2(17.14 mg, 33.53 μmol) e DIEA (43.34 mg, 335.35 μmol, 58.41 μL) sob N2. The mixture was stirred at 100°C for 2 h, then poured into H 2 H (20 mL) and extracted with EtOAc (5 mL*3). The combined organic layers were washed with H2O (5 mL*2) and brine (5 mL) were dried over Na2AFTERWARD4filtered and concentrated in vacuo. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 35%-65%, 9 min) and lyophilized compound 4 is obtained as a white solid.

1H-NMR (400 ΜΗζ, DMSO-d6) δ=12,55 (s, 1H), 9,33-9,30 (m, 1H), 8,52 (s, 1H), 8,40 (s, 1H), 8,27-8,24 (m, 2Η), 8,02 (d, J=7,6 Hz, 1Η), 7,94 (d, J=7,6 Hz, 1Η), 7,81-7,76 (m, 3Η) , 7,53-7,51 (m, 1Η), 4,25 (d, J=5,6 Hz, 2H), 2,81 (d, J=4,4 Hz, 3H), 2,66 ( s, 6Η), ppm.

LCMS (ESI) m/z: [M+H]+=502,3.

Example 35. Preparation of N-[2-[[4-(2-Bromophenyl)thiazol-2-yl]amino]-2-oxoethyl]-3-(dimethylsulfamoyl)benzamide (Compound 18)

Step 1: Preparation of tert-butyl N-[2-[[4-(2-bromophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]carbamate (intermediate i-35a)

To a solution of 2-( tert -butoxycarbonylamino)acetic acid (82.40 mg, 470.34 μmol) in DCM (10 mL) was added HATU (223.55 mg, 587.93 μmol) and DIEA (151.97 μmol) . mg, 1.18 mmol) and the mixture was stirred at 30°C for 30 min. Then 4-(2-bromophenyl)thiazol-2-amine (100 mg, 391.95 µmol) was added. The mixture was stirred at 30°C for another 12 h and then diluted with DCM (50.0 mL) and washed with H2O (10.0 mL*3) and brine (10.0 mL*2), then dried over Na2AFTERWARD4filtered and concentrated in vacuo to give intermediate i-35a as a yellow oil which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=412,3.

Step 2: Preparation of 2-amino-N-[4-(2-bromophenyl)thiazol-2-yl]acetamide trifluoroacetate (intermediate i-35b)

To a solution of intermediate i-35a (150 mg, 363.81 μmol) in DCM (5.0 mL) was added TFA (1.0 mL). The solution was stirred at 30°C for 2 h. The mixture was then diluted with DCM (20.0 mL) and concentrated in vacuo. This procedure was repeated three times. The residue was washed with MTBE (5.0 mL*2) and dried in vacuo to give intermediate i-35b (TFA salt) as a yellow oil, which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=312,3

Step 3: Preparation of N-[2-[[4-(2-Bromophenyl)thiazol-2-yl]amino]-2-oxoethyl]-3-(dimethylsulfamoyl)benzamide (Compound 18)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (96.82 mg, 422.33 μmol) in DMF (3.0 mL) was added HATU (401.46 mg, 1.06 mmol) and DIEA (227.43 mmol) which was given. mg, 1.76 mmol, 306.51 µL) and the mixture was stirred at 30°C for 30 min. Intermediate i-35b (150 mg, 351.94 μmol) was then added. The mixture was stirred at 30°C for 16 h and then poured into H2O (30.0 mL) and extracted with EtOAc (5.0 mL*3). The combined organic extracts were washed with H2O (5.0 mL * 2) and brine (5.0 mL), then dried over Na2AFTERWARD4filtered and concentrated in vacuo. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 42%-72%, 9 min) and lyophilized compound 18 is obtained as a pale yellow solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,51 (s, 1Η), 9,34-9,31 (m, 1Η), 8,27-8,23 (m, 2Η), 7, 94 (d, J=8,0 Hz, 1Η), 7,83 -7,81 (m, 1Η), 7,75-7,69 (m, 2Η), 7,54 (s, 1Η), 7 , 48–7,46 (m, 1H), 7,32–7,30 (m, 1H), 4,24 (d, J=5,6 Hz, 2H), 2,66 (s, 6H) ppm

LCMS (ESI) m/z: [M+H]+=525,0.

Example 36. Preparation of (N-[2-[[4-(3-Bromophenyl)thiazol-2-yl]amino]-2-oxoethyl]-3-(dimethylsulfamoyl)benzamide (Compound 7)

Step 1: Preparation of tert-butyl N-[2-[[4-(3-bromophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]carbamate (intermediate i-36a)

To a solution of 2-( tert -butoxycarbonylamino)acetic acid (82.40 mg, 470.34 μmol) in DCM (10 mL) was added HATU (223.55 mg, 587.93 μmol) and DIEA (151.97 μmol) . mg, 1.18 mmol, 204.81 µL) and the mixture was stirred at 30°C for 30 min. Then 4-(3-bromophenyl)thiazol-2-amine (100.00 mg, 391.95 µmol) was added. The mixture was stirred at 30°C for another 12 h and then diluted with DCM (50.0 mL). The solution was washed with H2O (10.0 mL*3) and brine (10 mL*2), then dried over Na2AFTERWARD4filtered and concentrated in vacuo to give intermediate i-36a as a yellow oil which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=412,3

Step 2: Preparation of 2-amino-N-[4-(3-bromophenyl)thiazol-2-yl]acetamide trifluoroacetate (intermediate i-36b)

To a solution of intermediate i-36a (150.00 mg, 363.81 μmol) in DCM (5 ml) was added TFA (1 ml) and the solution was stirred at 30 °C for 2 h. The mixture was then diluted with DCM (20.0 mL) and concentrated in vacuo. This procedure was repeated three times. The residue was washed with MTBE (5.0 mL*2) and dried in vacuo to give intermediate i-36b (TFA salt) as a yellow oil, which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=312,3

Step 3: Preparation of (N-[2-[[4-(3-Bromophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (Compound 7)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (96.82 mg, 422.33 μmol) in DMF (3.0 mL) was added HATU (401.46 mg, 1.06 mmol) and DIEA (227.43 mmol) which was given. mg, 1.76 mmol, 306.51 µL) and the mixture was stirred at 30°C for 30 min. Intermediate i-36b (150 mg, 351.94 μmol) was then added. The mixture was stirred at 30°C for 16 h and then poured into H2O (30.0 mL) and extracted with EtOAc (5.0 mL*3). The combined organic extracts were washed with H2O (5.0 mL * 2) and brine (5 mL), then dried over Na2AFTERWARD4filtered and concentrated in vacuo. The residue was purified by pre-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 42%-72%, 9 min) and lyophilized to give compound 7 as a pink solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,51 (s, 1Η), 9,33-9,30 (m, 1Η), 8,27-8,23 (m, 2Η), 8, 11 (s, 1Η), 7,94-7,91 (m, 2Η), 7,82-7,80 (m, 2Η), 7,43-7,42 (m, 1Η), 7,41- 7,40 (m, 1Η), 4,24 (d, J=5,6 Hz, 2H), 2,66 (s, 6H) ppm

LCMS (ESI) m/z: [M+H]+=523,2.

Example 37. Preparation of (S)-N-(4-(Methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)nicotinamide (Compound 70)

Step 1: Preparation of (S)-tert-butyl (4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)carbamate (intermediate i-37a)

To a solution of (2S)-2-(tert-butoxycarbonylamino)-4-methylsulfanylbutanoic acid (5.0 g, 20.05 mmol) in DCM (20.0 mL) was added EEDQ (6.20 g, 25.07 mmol) and then 4-phenylthiazole-amine (2.95 g, 16.71 mmol) was added to the mixture. The mixture was stirred at 25°C for 3 hours. 10% citric acid (800.0 mL) was added and the reaction mixture was extracted with EtOAc (200 mL). The combined organic extracts were washed with brine (100 mL) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2petroleum ether/ethyl acetate = 50/1 to 8:1) to give intermediate i-37a as a white solid.

LCMS (ESI) m/z: [M+H]+=408,0.

SFC-HPLC: Amycoat-MeOH(DEA)-5-40-3 mL-35, 2.284 Min.

Step 2: Preparation of (S)-2-amino-4-(methylthio)-N-(4-phenylthiazol-2-yl)butanamide trifluoroacetate (intermediate i-37b)

To a solution of intermediate i-37a (5.8 g, 14.23 mmol) in DCM (20.0 mL) was added TFA (4.0 mL). The mixture was stirred at 10°C for 2 hours. The reaction mixture was concentrated under reduced pressure. The crude intermediate i-37b (TFA salt) was lyophilized and obtained as a white solid.

This material was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=307,9.

SFC HPLC: Cellucoat-MeOH(DEA)-10-12 min-3, 8.970 min.

Step 3: Preparation of (S)-N-(4-(Methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)nicotinamide (Compound 70)

To a solution of nicotinic acid (14.61 mg, 118.64 μmol) in DCM (2 mL) was added HATU (67.66 mg, 177.95 μmol) and DIPEA (61.33 mg, 474.54 μmol, 82, 66 µL) and then i-3 (50 mg), 118.64 µmol) was added. The mixture was stirred at 10°C for 2 hours. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC (Phenomenex Gemini 150*25 mm*10 µm; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 45%-75%, 10 min) and lyophilized to give compound in gave 70 as a white solid.

1H-NMR (400 MHz, METANOL-d4) δ 9,06 (m, 1Η), 8,72-8,71 (m, 1Η), 8,33-8,31 (m, 1Η), 7,91 - 7,89 (m, 2Η), 7,58-7,55 (m, 1Η), 7,40-7,38 (m, 3Η), 7,37-7,30 (m, 1Η), 4 , 94-4,91 (m, 1Η), 2,75-2,65 (m, 2Η), 2,31-2,29 (m, 2Η), 2,14 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=413,1.

HPLC solution: Cellucoat-MeOH(DEA)-5-40-3 mL-35T.Icm, 2,269 min.

Example 38. Preparation of 4-methoxy-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 67)

To a solution of 4-methoxybenzoic acid (21.66 mg, 142.36 μmol) in DCM (2.0 mL) was added HATU (90.22 mg, 237.27 μmol) and DIEA (76.66 mg, 593.18 μmol) , 103). The mixture was stirred at 30°C for 30 minutes. Then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 µmol) was added. The mixture was further stirred at 30°C for 2 hours. The reaction solution was diluted with DCM (20.0 mL) and washed with (5.0 mL * 3) and brine (5 mL) and dried over Na2AFTERWARD4filtered and concentrated in vacuo. The residue was purified by pre-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 45%-75%, 9 min) and lyophilized to give compound 67 as a white solid.

1HNMR (400 MHz, METANOL-d4) δ 7,92-7,90 (m, 4H), 7,42-7,38 (m, 3H), 7,33-7,29 (m, 1H), 7 ,03 (d, J=9,2 Hz, 2Η), 4,94 -4,91 (m, 1Η), 3,88 (s, 3Η), 2,75-2,65 (m, 2Η), 2,36-2,15 (m, 2Η), 2,14 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=442,1.

Example 39. Preparation of 2-hydroxy-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 69)

In a mixture of (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 μmol) and 2-hydroxybenzoic acid (16.39 mg, 118.64 μmol ) in DCM (1 mL) DIPEA (61.33 mg, 474.54 μmol) was added. The mixture was stirred at 30°C for 15 min, then HATU (67.66 mg, 177.95 μmol) was added and stirred at 30°C for 2 h. The reaction mixture was evaporated to dryness to give the crude product. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150 x 25 x 10 µm, mobile phase: [water (0.1% TFA)-ACN], B%: 51%-71%, 12 min) purified and lyophilized compound 69 as a white solid.

1H-NMR (400 MHz, MeOD-d4) δ=7.92–7.89 (m, 3H), 7.43–7.37 (m, 4H), 7.31–7.28 (m, 1H), 6.96–6.92 ( m, 2H), 4.98-4.94 (m, 1H), 2.72-2.60 (m, 2H), 2.35-2.27 (m, 1H), 2.25-2, 17 (m, 1H), 2.13 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=428,1.

HPLC-Lösung: OD-3_5CM_MEOH(DEA)_40_3ML_T35.M, 0.822 min.

Example 40. Preparation of the compound (S)-3-Hydroxy-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide ( Compound 59)

In a solution of 3-hydroxybenzoic acid (16.39 mg, 118.64 μmol, 9.94 μL) and (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg , 118.64 μmol) in DCM (2.0 mL) was added DIPEA (61.33 mg, 474.56 μmol), then HATU (67.66 mg, 177.96 μmol) was added. The mixture was stirred at 10°C for 2 hours. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC (TFA condition; Column: Column: Phenomenex Synergi C18 150*25*10 µm; Mobile phase: [Water(0.1% TFA)-ACN]; B%: 45%-75%, 9 min ) and lyophilized to give compound 59 as a white solid.

1HNMR (400 MHz, METANOL-d4) δ=7,90–7,88 (m, 2H) 7,38–7,34 (m, 4H), 7,30–7,27 (m, 3H), 6 ,98-6,95 (m, 1Η), 4,90-4,87 (m, 1Η), 2,69-2,62 (m, 2Η), 2,26-2,20 (m, 2Η) , 2,13 (s, 3Η) ppm

LCMS (ESI) m/z: [M+H]+=428,1.

HPLC-Lösung: AS-3_5CM_MEOH(DEA)_40_3ML_5MIN_T35.M, 1.341 Min.

Example 41. Preparation of 4-hydroxy-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 53)

For a solution of 4-hydroxybenzoic acid (16.39 mg, 118.64 μmol) and (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 μmol) ) in DCM (2.0 mL added), DIEA (76.66 mg, 593.18 μmol, 103.32 μL), and HATU (90.22 mg, 237.27 μmol). The mixture was stirred at 30°C for 2.5 hours. The reaction was diluted with DCM (20.0 mL) and washed with water (5.0 mL * 3) and brine (5.0 mL) and dried over Na2AFTERWARD4filtered and concentrated in vacuo to give the crude product. The crude product was purified by pre-HPLC (column: Boston pH-lex 150*25 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 48%-68%, 8 min) and lyophilized to give compound 53 as a yellow solid.

1H-NMR (400 MHz, METANOL-d4) δ=7,91 (d, J=8,4 Hz, 2H), 7,82 (d, J=8,4 Hz, 2H), 7,42-7 , 38 (m, 3Η), 7,32-7,30 (m, 1Η), 6,87-6,85 (m, 2Η), 4,90-4,86 (m, 1Η), 2,72 - 2,64 (m, 2Η), 2,28-2,21 (m, 2Η), 2,15 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=428,3.

Example 42. Preparation of 2-amino-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 63)

In a mixture of 2-aminobenzoic acid (16.27 mg, 118.64 μmol) and (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 μmol) ) in DCM (1 mL) added DIPEA (61.33 mg, 474.54 μmol). The mixture was stirred at 30°C for 15 min, then HATU (67.66 mg, 177.95 μmol) was added and stirred at 30°C for 2 h. The reaction mixture was evaporated to dryness to give the crude product. The rest was preparation. HPLC (column: Boston pH-lex 150*25 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 55%-75%, 8 min) and lyophilized to give Compound 63 (TFA salt) as a white solid.

1H-NMR (400 MHz, MeOD-d4) δ=7.91–7.89 (m, 2H), 7.71–7.69 (m, 1H), 7.41–7.37 (m, 3H), 7.35–7.28 ( m, 2H), 6.93-6.86 (m, 2H), 4.91-4.87 (m, 1H), 2.75-2.61 (m, 2H), 2.33-2, 26 (m, 1H), 2.25-2.16 (m, 1H), 2.15 (s, 3H) ppm; LCMS (ESI) m/z: [M+H]+=427,3.

HPLC-Lösung: Cellucoat-MeOH(DEA)-40-3 mL-35T, 1 cm, 1.540 Min.

Example 43. Preparation of the compound (S)-3-amino-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide ( Compound 56)

Step 1: Preparation of (S)-tert-butyl (3-((4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)carbamoyl)phenyl) Carbamate (intermediate i-42a)

To a solution of 3-( tert -butoxycarbonylamino)benzoic acid (28.15 mg, 118.65 μmol) in DCM (2.0 mL) was added HATU (67.66 mg, 177.94 μmol) and DIPEA (61.33 mg, 474 μmol)- and then 4 μmol. 2-Amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 µmol) was added. The mixture was stirred at 10°C for 12 hours. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC (TFA condition; Column: Column: Phenomenex Synergi C18 150*25*10 µm; Mobile phase: [Water(0.1% TFA)-ACN]; B%: 55%-85%, 9 min ) to give intermediate i-42a as a white solid.

LCMS (ESI) m/z: [M+H]+=527,1.

Step 2: Preparation of (S)-3-amino-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide (Compound 56 ). ) trifluoroacetate

To a solution of intermediate i-42a (20 mg, 37.97 μmol) in DCM (2.0 mL) was added TFA (1.0 mL). The mixture was stirred at 10°C for 2 hours. The reaction mixture was concentrated under reduced pressure. The rest was preparation. HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 μm; mobile phase: [water(0.1% TFA)-ACN]; B%: 35%-65%, 9 min) purified and lyophilized, for to give compound 56 (TFA salt) as a white solid.

1Η NMR (400 MHz, METANOL-d4) δ=7,90–7,88 (m, 2Η), 7,84–7,83 (m, 1Η), 7,72–7,71 (m, 1Η) , 7,57–7,55 (m, 1Η), 7,40–7,36 (m, 4H), 7,31–7,279 (m, 1H), 4,93–4,91 (m, 1H) , 2,70-2,64 (m, 2Η), 2,30-2,20 (m, 2Η), 2,14 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=427,1.

Example 44. Preparation of 4-amino-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 52)

To a solution of 4-aminobenzoic acid (19.52 mg, 142.36 μmol) in DCM (2 mL) was added HATU (90.22 mg, 237.27 μmol) and DIEA (76.66 mg, 593.18 μmol, 103.32 µmol in 3 µL) and the mixture was 3 µL) °C for 30 min Then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg) was added , 118.64 μmol). The mixture was stirred at 30°C for another 2 h and then diluted with DCM (20.0 mL). The organic layer was washed with brine (5.0 mL * 3) and brine (5 mL) and dried over Na2AFTERWARD4filtered and concentrated in vacuo. The residue was purified by pre-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.225% FA)-ACN]; B%: 40%-70%, 9 min) and lyophilized atom gives compound 52 (TFA salt) as a white solid.

1H-NMR (400 MHz, METANOL-d4) δ 7,92-7,89 (m, 2Η), 7,76-7,74 (m, 2Η), 7,42-7,38 (m, 3Η) , 7,33-7,31 (m, 1Η), 6,80 (d, J=8,8 Hz, 2Η), 4,94-4,88 (m, 1Η), 2,71-2,64 (m, 2Η), 2,28-2,21 (m, 2Η), 2,15 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=427,1.

Example 45. Preparation of (S)-N-(3-amino-1-oxo-1-((4-phenylthiazol-2-yl)amino)propan-2-yl)-3-(N,N-dimethylsulfamoyl)benzamide (Compound 25)

Step 1: Preparation of (S)-3-((((9H-Fluoren-9-yl)methoxy)carbonyl)amino)-2-((tert-butoxycarbonyl)amino)propanoic acid (intermediate i-45a)

To a solution of (2S)-3-amino-2-(tert-butoxycarbonylamino)propanoic acid (1.0 g, 4.90 mmol) in THE (10.0 mL) and water (10.0 mL) was added FmocOSu (1 .82 g, 5.39 mmol). and NaHCO3(822.69 mg, 9.79 mmol). The mixture was stirred at 20°C for 3 hours. The reaction mixture was adjusted to pH 4 with citric acid solution. The solution was extracted with EtOAc (10.0 mL*3). The organic layer was washed with brine (20.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was triturated with MTBE (10.0 mL). The solution was filtered and the filter cake dried in vacuo to give intermediate i-45a as a white solid.

LCMS (ESI) m/z: [M+Na]+=449,3

Step 2: Preparation of (S)-(9H-fluoren-9-yl)methyl-tert-butyl-(3-oxo-3-((4-phenylthiazol-2-yl)amino)propan-1,2-diyl) dicarbamate (intermediate i-45b)

To a solution of intermediate i-45a (1.8 g, 4.22 mmol) in DCM (18.0 mL) was added EEDQ (1.57 g, 6.33 mmol) to give 4-phenylthiazol-2-amine ( 743 .86 mg, 4.22 mmol). . The reaction mixture was stirred at 20°C for 12 h and then poured into water (20.0 mL). The solution was extracted with EtOAc (20.0 mL*3). The combined organic layers were washed with brine (50.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiO).2, PE:EA=100:1-50:1-10:1-5:1-2:1) to give intermediate i-45b as a yellow solid.

LCMS (ESI) m/z: [M+H]+=585,4

HPLC-Lösung: Cellucoat-MeOH(DEA)-40-7MIN-3ML, 3.031 Min.

Step 3: Preparation of (S)-(9H-Fluoren-9-yl)methyl(2-amino-3-oxo-3-((4-phenylthiazol-2-yl)amino)propyl)carbamate (intermediate i-45c ). )

To a solution of intermediate i-45b (2.5 g, 4.28 mmol) in DCM (25.0 mL) was added TFA (4.0 mL). The reaction mixture was stirred at 20°C for 1.5 h. The reaction mixture was concentrated to give a residue. The residue was triturated with PE (10.0 mL). The mixture was then filtered and the solid dried in vacuo to give intermediate i-45c (TFA salt) as a white solid.

LCMS (ESI) m/z: [M+Na]+=507,3

HPLC-Lösung: OJ-3-MeOH(DEA)-60-7MIN-3ML-35T, 5.436 Min

Step 4: Preparation of (S)-(9H-Fluoren-9-yl)methyl (2-(3-(N,N-dimethylsulfamoyl)benzamido)-3-oxo-3-((4-phenylthiazol-2-yl) ) ) )amino)propyl)carbamate (intermediate i-45c)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (38.30 mg, 167.06 μmol) in DCM (1.0 mL) was added HATU (69.87 mg, 183.76 μmol), DIEA (64.77 μmol mg , 501.18 μmol mg, 501.18 μmol, 501.18 μmol, intermedia. gave -45b (100 mg, 167.06 μmol). The reaction mixture was stirred at 20°C for 12 h and then poured into water (2.0 mL). The solution was extracted with EtOAc (2.0 mL*3). The combined organic extracts were washed with brine (5.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiO).2. PE/EA=50/1-1/1) to give intermediate i-45c as a white solid.

LCMS (ESI) m/z: [M+H]+=696,0

HPLC-Lösung: OJ-3-MeOH(DEA)-60-7MIN-3ML-35T, 5.276 Min.

Step 5: Preparation of (S)-N-(3-amino-1-oxo-1-((4-phenylthiazol-2-yl)amino)propan-2-yl)-3-(N,N-dimethylsulfamoyl) Formate Benzamide salt (Compound 25)

To a solution of intermediate i-45c (100.0 mg, 140.84 μmol) in DCM (1.0 mL) was added piperidine (98.00 μL). The solution was stirred at 20°C for 2 h. The mixture was poured into water (2.0 mL) and extracted with EtOAc (2.0 mL*3). The combined organic extracts were washed with brine (3.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by Prep-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.225% FA)-ACN]; B%: 17%-37%, 7 min) and lyophilized compound The 25 (FA salt) is obtained as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=8,30–8,28 (m, 2H), 8,23–8,22 (m, 2H), 7,96–7,89 (m, 3H ), 7,81 (d, J=8,0 Hz, 1Η), 7,63 (s, 1Η), 7,45-7,43 (m, 2Η), 7,41-7,32 (m, 1Η), 4,74 (s, 1Η), 3,11 (s, 2Η), 2,66 (s, 6Η) ppm

LCMS (ESI) m/z: [M+H]+=474,3

HPLC-Lösung: OJ-3-MeOH (DEA)-40-7MIN-3ML-35T, 2.956 Min.

HPLC-Lösung: AD-3_5CM_MEOH(DEA)_40_3ML_8MIN_T35.M

Example 46. Preparation of (S)-N-(3-Acetamido-1-oxo-1-((4-phenylthiazol-2-yl)amino)propan-2-yl)-3-(N,N-dimethylsulfamoyl)benzamide (Union 21)

N-[(1S)-1-(aminomethyl)-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]-3-(dimethylsulfamoyl)benzamide (20.0 mg, 38, 49 μmol) in a.c2(0.2 ml) was stirred at 20°C for 2 h. The reaction mixture was then poured into water (2.0 mL) and extracted with EtOAc (2.0 mL*3). The combined organic extracts were washed with brine (5.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was lyophilized to give compound 21 (51.86% ee) as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,54 (s, 1Η), 9,12 (d, J=6,8 Hz, 1Η), 8,23-8,22 (m, 3Η) , 7,95 (d, J=7,6 Hz, 1Η), 7,89 (d, J=7,2 Hz, 2Η), 7,81 (s, 1Η), 7,65 (s, 1Η) , 7,44-7,42 (m, 2H), 7,40-7,31 (m, 1H), 4,72 (d, J=6,0 Hz, 1H), 3,62 (m, 2H ), 2,65 (s, 6H), 1,83 (s, 3H) ppm

LCMS (ESI) m/z: [M+H]+=516,4

HPLC solution: Amycoat-MeOH(DEA)-40-5MIN-3ML-35T, 1,514 min and 3,407 min

Example 47. Preparation of N-(2-((4-(2-cyanophenyl)thiazol-2-yl)amino)-2-oxoethyl)-3-(N,N-dimethylsulfamoyl)benzamide (Compound 27)

Step 1: Preparation of tert-Butyl (2-((4-(2-cyanophenyl)thiazol-2-yl)amino)-2-oxoethyl)carbamate (intermediate i-47a)

In a solution of tert-butyl N-[2-[(4-bromothiazol-2-yl)amino]-2-oxo-ethyl]carbamate (200.0 mg, 558.65 µmol) in toluene (3.0 mL). 2-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)benzonitrile (383.93 mg, 1.68 mmol), Ad2nBuP-Pd-G3 (406.85, mg, 558.65 μmol) and K3AFTER4(177.87 mg, 837.97 μmol) N sob2. The reaction mixture was stirred at 60°C for 12 hours. The reaction mixture was concentrated to give a residue. The residue was purified by column chromatography (SiO).2, PE/EA=50/1-1/1) to give intermediate i-47a as a brown solid.

LCMS (ESI) m/z: [M+H]+=359,2

Step 2: Preparation of 2-amino-N-(4-(2-cyanophenyl)thiazol-2-yl)acetamide (intermediate i-47b)

To a solution of intermediate i-47a (230 mg, 628.88 μmol) in DCM (2.5 mL) was added TFA (0.25 mL). The mixture was stirred at 30°C for 2 hours. The reaction mixture was concentrated to give intermediate i-47b (TFA salt) as a yellow solid, which was used directly in the next step.

LCMS (ESI) m/z: [M+Na]+=281,2

Step 3: Preparation of N-(2-((4-(2-cyanophenyl)thiazol-2-yl)amino)-2-oxoethyl)-3-(N,N-dimethylsulfamoyl)benzamide (Compound 27)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (81.41 mg, 355.13 μmol) in DCM (2.0 mL) was added HATU (162.04 mg, 426.15 μmol), DIEA (183.59 mg, 1 .42 mmol i-4) and 110.0 mg, 355.13 μmol). The reaction mixture was stirred at 30°C for 12 h. The reaction mixture was poured into water (5.0 mL). The solution was then extracted with EtOAc (5.0 mL*3). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by pre-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.225% FA)-ACN]; B%: 37%-57%, 7 min) and lyophilized compound 27 obtained as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.43 (br s, 1H), 9.32 (m, 1H), 8.26-8.25 (m, 2H), 7.97-7 , 92 (m, 3H), 7.80-7.79 (m, 3H), 7.56 (m, 1H), 4.26 (d, J=5.6 Hz, 2H), 2.65 ( s, 6H) ppm

LCMS (ESI) m/z: [M+H]+=470,2

Example 48. Preparation of N-[2-[[4-[3-(aminomethyl)phenyl]thiazol-2-yl]amino]-2-oxoethyl]-3-(dimethylsulfamoyl)benzamide (Compound 10)

In a solution of N-[2-[[4-(3-cyanophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (100.0 mg, 212.98 μmol) in To MeOH (4.0 mL) under N was added Raney Ni (99.99 mg, 1.17 mmol).2. The mixture was stirred under H2(50psi) at 15°C for 16 hours. The mixture was filtered and the filtrate was concentrated in vacuo to give a residue. The rest was preparation. HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 μm; mobile phase: [water (0.1% TFA)-ACN]; B%: 15%-45%, 9 min) purified and lyophilized, for to give compound 10 (TFA salt) as a white solid.

1H-NMR (400 ΜΗζ, DMSO-d6) δ=12.48 (s, 1H), 9.34–9.31 (m, 1H), 8.27–8.18 (m, 5H), 7.97–7.91 (m, 3H) , 7.83-7.79 (m, 1H), 7.62 (s, 1H), 7.53-7.49 (m, 1H), 7.42 (d, J=7.6, 1H) , 4.24 (d, J=5.6 Hz, 2H), 4.08 (m, 2H), 2.66 (m, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=474,2

Example 49. Preparation of N-[2-[[4-(3-cyanophenyl)thiazol-2-yl]amino]-2-oxoethyl]-3-(dimethylsulfamoyl)benzamide (Compound 8)

Step 1: Preparation of 3-(2-aminothiazol-4-yl)benzonitrile 2 (intermediate i-49a)

For a solution of 3-(2-bromoacetyl)benzonitrile (200.00 mg, 892.64 μmol) in H2Thiurea (74.74 mg, 981.91 µmol) was added to O (5.0 mL). The mixture was stirred at 25°C for 1.5 h. The reaction mixture was diluted with water (10.0 mL) and extracted with EtOAc (35.0 mL*3). The combined organic extracts were washed with brine (20.0 mL * 2) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give intermediate i-49a as a yellow solid.

LCMS (ESI) m/z: [M+H]+= 202,1.

Step 2: Preparation of tert-butyl N-[2-[[4-(3-cyanophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]carbamate (intermediate i-49b)

To a solution of 2-( tert -butoxycarbonylamino)acetic acid (129.27 mg, 737.90 μmol) in DCM (8.0 mL) was added HATU (280.57 mg, 737.90 μmol) and DIEA (260.09 mg , 2.05 μmol). and intermediate i-49a (135.0 mg, 670.82 μmol). The mixture was stirred at 25°C for 12 hours. The reaction mixture was diluted with water (10.0 mL) and extracted with EtOAc (20.0 mL*3). The combined organic extracts were washed with brine (20.0 mL * 2) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was analyzed by Prep-HPLC (TFA condition, column: Boston pH-lex 150*25 10 μm, mobile phase: [water (0.1% TFA)-ACN], B%: 40%-70%, 10th min ) clean up. and lyophilized to give intermediate i-49b as a white solid.

LCMS (ESI) m/z: [M+H]+=359,4

Step 3: Preparation of 2-amino-N-[4-(3-cyanophenyl)thiazol-2-yl]acetamide (intermediate i-49c)

To a solution of intermediate i-49b (93.0 mg, 259.48 μmol) in DCM (1.0 mL) was added TFA (0.1 mL). The mixture was stirred at 25°C for 1.5 h. The mixture was concentrated under reduced pressure to give intermediate i-49c (TFA salt) as a brown oil, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=259,1.

Step 4: Preparation of N-[2-[[4-(3-cyanophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (Compound 8)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (75.74 mg, 330.36 μmol) in DCM (2.0 mL) was added HATU (138.17 mg, 363.40 μmol), DIEA (170.79 mg, 1.32 μmol) and Intermedia (1.32 μL) (17 mmol) 123.0 mg, 330.36 μmol The mixture was stirred for 12 h at 25° C. The reaction mixture was diluted with water (10.0 mL) and washed with EtOAc (30.0 mL*3) The combined organic extracts were washed with brine (20.0 mL*2), dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 35%-65%, 9 min) purified and lyophilized, for to give compound 8 as a white solid.

1HNMR (400 MHz, DMSO-d6) δ 12.53 (s, 1H), 9.34-9.31 (m, 1H), 8.33 (s, 1H), 8.26-8.22 (m , 3H), 7.94 (d, J=8.0 Hz, 1H), 7.88 (s, 1H), 7.82-7.78 (m, 2H), 7.68-7.64 ( m, 1H), 4.24 (d, J=5.6 Hz, 2H), 2.65 (s, 6H).

LCMS (ESI) m/z: [M+H]+=470,0.

Example 50. Preparation of N-[2-[[4-[3-(Acetamidomethyl)phenyl]thiazol-2-yl]amino]-2-oxoethyl]-3-(dimethylsulfamoyl)benzamide (Compound 5)

In a solution of N-[2-[[4-(3-cyanophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (100 mg, 212.98 μmol) in MeOH ( 2.0 mL) was added to NiCl2.6H2O (126.56 mg, 532.44 μmol) in NaBH4(80.57 mg, 2.13 mmol), the mixture was stirred at 25°C for 2 h, then Ac2To the mixture were added O (21.74 mg, 212.98 µmol) and TEA (43.10 mg, 425.95 µmol). The mixture was stirred at 25°C for another 2 h and then concentrated in vacuo to give a residue. The residue was diluted with water (20.0 mL) and extracted with EtOAc (15.0 mL*2). The combined organic extracts were washed with brine (5.0 mL * 2) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 30%-53%, 7 min) purified and lyophilized, for to give compound 5 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12,52 (s, 1Η), 9,31-9,29 (m, 1Η), 8,37-8,36 (m, 1Η), 8,26 - 8,23 (m, 2Η), 7,94 (d, J =8,0 Hz, 1Η), 7,82-7,76 (m, 3Η), 7,61 (s, 1Η), 7, 39 -7,36 (m, 1H), 7,21 (d, J=7,6 Hz, 1H), 4,29 (d, J= 5,6 Hz, 2H), 4,23 (d, J = 5,6 Hz, 2Η), 2,65 (s, 6Η), 1,88 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=516,3.

Example 51. Preparation of N-[2-[[4-[4-(aminomethyl)phenyl]thiazol-2-yl]amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (Compound 22)

Step 1: Preparation of 4-(2-aminothiazol-4-yl)benzonitrile (intermediate i-51a)

In an aqueous mixture of 4-(2-bromoacetyl)benzonitrile (500 mg, 2.23 mmol, 79.78 μL) and NaF (0.1 g, 2.38 mmol) in H2Thiurea (186.86 mg, 2.45 mmol) was added to O (8.0 mL) and MeOH (8.0 mL). The mixture was stirred at 25°C for 30 min and then diluted with water (10.0 mL) and extracted with EtOAc (35.0 mL*3). The combined organic extracts were washed with sat.3(20.0 ml*2), dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give intermediate i-51a as a yellow solid.

1H RMN (400 MHz, DMSO-d6) δ=7,97-7,95 (m, 2H), 7,82-7,80 (m, 2H), 7,32 (s, 1H), 7,19 (s, 2Η) ppm.

Step 2: Preparation of tert-butyl N-[2-[[4-(4-cyanophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]carbamate (intermediate i-51b)

To a solution of 2-(tert-butoxycarbonylamino)acetic acid (287.26 mg, 1.64 mmol) in pyridine (6 mL) was added EDCI (1.43 g, 7.45 mmol) and intermediate i-51a (300 .0 mg, 1.49 mmol) The mixture was stirred at 25°C for 48 h. The reaction mixture was diluted with water (15.0 mL) and extracted with EtOAc (30.0 mL*3). The combined organic extracts were washed with brine (20.0 mL * 2) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was analyzed by Prep-HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]). B%: 38%-68%, 9 min purified) and lyophilized to give intermediate i-51b as a white solid.

LCMS (ESI) m/z: [M+H]+=359,2.

Step 3: Preparation of 2-amino-N-[4-(4-cyanophenyl)thiazol-2-yl]acetamide (intermediate i-51c)

To a solution of intermediate i-51b (250 mg, 697.52 μmol) in DCM (2.0 ml) was added TFA (0.2 ml) and stirred at 25 °C for 4 h. The mixture was concentrated under reduced pressure to give intermediate i-51c (TFA salt) as a brown solid, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=259,1.

Step 4: Preparation of N-[2-[[4-(4-cyanophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (intermediate i-51d)

To a solution of 3-(dimethylsulfamoyl)benzoic acid (244.08 mg, 1.06 mmol) in DCM (5.0 mL) was added HATU (404.82 mg, 1.06 mmol), DIEA (500.36 mg, 3.87 mmol, 674 mL) and intermediate 674. - was added. 51c (250.0 mg, 967.87 μmol. The mixture was stirred at 25 °C for 24 h. The reaction mixture was diluted with water (10.0 mL) and extracted with EtOAc (15.0 mL * 3). combined organic extracts were washed with brine (10.0 mL * 2), dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 45%-75%, 9 min) purified and lyophilized, for to give the intermediate i-51d as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12.27 (s, 1H), 9.33-9.30 (m, 1H), 8.26-8.23 (m, 2H), 8.08 (d, J=8.4 Hz, 2H), 7.95-7.89 (m, 4H), 7.82-7.78 (m, 1H), 4.24 (d, J=5.6 Hz, 2H), 2.65 (s, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=469,2.

Step 5: Preparation of N-[2-[[4-[4-(aminomethyl)phenyl]thiazol-2-yl]amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (Compound 22)

To a solution of intermediate i-51d (200.0 mg, 425.95 μmol) in MeOH (1.0 mL) was added Raney Ni (200.00 mg, 2.33 mmol) under N22. The mixture was shaken under H 2 (50 psi) at 15°C for 16 h. The mixture was filtered and the filtrate was concentrated in vacuo. The rest was preparation. HPLC (core column: Phenomenex Gemini 150*25 mm*10 µm; mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN]; B%: 28%–58%, 11.5 min) purified) and lyophilized to give compound 22 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 9.16 (s, 1H), 8.26-8.23 (m, 2H), 7.93 (d, J=7.6 Hz, 1H), 7.82-7.77 (m, 3H), 7.43 (s, 1H), 7.35 (d, J=7.6, 2H), 4.15 (m, 2H), 3.72 ( s, 2H), 2.65 (s, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=474,3.

Example 52. Preparation of N-[2-[[4-[4-(acetamidomethyl)phenyl]thiazol-2-yl]amino]-2-oxoethyl]-3-(dimethylsulfamoyl)benzamide (Compound 11)

In a solution of N-[2-[[4-(4-cyanophenyl)thiazol-2-yl]amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (100.00 mg, 212.98 μmol) in MeOH (2.0 mL) was added with NiCl2.6H2O (126.56 mg, 532.44 μmol) in NaBH4(80.57 mg, 2.13 mmol), the mixture was stirred at 25°C for 2 h, then Ac2O (21.74 mg, 212.98 μmol, 19.94 μL) and TEA (43.10 mg, 425.95 μmol, 59.29 μL) were added. The mixture was stirred at 25°C for another 2 h and then concentrated in vacuo to give a residue. The residue was diluted with water (20.0 mL) and extracted with EtOAc (15.0 mL*2). The combined organic extracts were washed with brine (5.0 ml*2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 30%-53%, 7 min) purified and lyophilized, for to give compound 11 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,49 (s, 1Η), 9,32-9,29 (m, 1Η), 8,38-8,35 (m, 1Η), 8, 27 -8,23 (m, 2Η), 7,95 (d, J=8,0 Hz, 1Η), 7,86-7,79 (m, 3Η), 7,60 (s, 1Η), 7 , 31 (d, J=8,4 Hz, 2H), 4,27 (d, J=5,6 Hz, 2H), 4,23 (d, J=5,6 Hz, 2H), 2,66 (s, 6Η), 1,88 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=516,3.

Example 53. Preparation of (S)-N-(6-Acetamido-1-oxo-1-((4-phenylthiazol-2-yl)amino)hexan-2-yl)benzamide (Compound 43)

Step 1: Preparation of (S)-6-((((9H-Fluoren-9-yl)methoxy)carbonyl)amino)-2-((tert-butoxycarbonyl)amino)hexanoic acid (intermediate i-53a)

To a solution of (2S)-6-amino-2-(tert-butoxycarbonylamino)hexanoic acid (1 g, 4.06 mmol) in THE (10.0 mL) was added a solution of NaHCO 33(682.14 mg, 8.12 mmol) in water (10.0 mL) and FmocOSu (1.51 g, 4.47 mmol). The reaction mixture was stirred at 20°C for 3 hours. The reaction mixture was poured into water (10.0 mL). The solution was adjusted to pH 4 with saturated citric acid solution. The mixture was extracted with EtOAc (15 mL*3). The combined organic extracts were washed with brine (20.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give intermediate i-53a as a white solid. The compound was used directly in the next step.

LCMS (ESI) m/z: [M+Na]+=491,4

Step 2: Preparation of (6-oxo-6-((4-phenylthiazol-2-yl)amino)hexane-1,5-diyl)dicarbamate (S)-(9H-Fluoren-9-yl)methyl tert-butyl Intermediate i-53b)

To a solution of intermediate i-53a (500 mg, 977.50 μmol) in DCM (5.0 mL) was added EEDQ (362.59 mg, 1.47 mmol) and 4-phenylthiazol-2-amine (172, 27n mg, 977.50 μmol). The reaction mixture was stirred at 20°C for 18 h. The reaction mixture was poured into water (10.0 mL). The solution was extracted with EtOAc (10.0 mL*3). The combined organic extracts were washed with brine (20.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiO).2, petroleum ether/ethyl acetate = 100:1 to 1:1) to give intermediate i-53b as a yellow oil.

LCMS (ESI) m/z: [M+H]+=627,5

Step 3: Preparation of (S)-(9H-Fluoren-9-yl)methyl (5-amino-6-oxo-6-((4-phenylthiazol-2-yl)amino)hexyl)carbamate (intermediate i-53c ). )

To a solution of intermediate i-53b (440 mg, 586.68 μmol) in DCM (5.0 mL) was added TFA (417.85 μL). The reaction mixture was stirred at 20°C for 2 hours. The reaction mixture was adjusted to pH 8 with saturated NaHCO 33Solution. The solution was extracted with EtOAc (5.0 mL*3). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was triturated with MTBE (10.0 mL). The solution was filtered. The filter cake was dried in vacuo to give intermediate i-53c as a white solid.

LCMS (ESI) m/z: [M+Na]+=549,4.

HPLC-Lösung: AS-3-MeOH (DEA)-5-40-3ML-35T, 2.386 Min.

Step 4: Preparation of (S)-(9H-Fluoren-9-yl)methyl(5-benzamido-6-oxo-6-((4-phenylthiazol-2-yl)amino)hexyl)carbamate (intermediate i-53d ). )

To a solution of benzoic acid (41.68 mg, 341.33 μmol, 52.10 μL) in DCM (2.0 mL) was added HATU (142.76 mg, 375.46 μmol), DIEA (176.46 mg, 1 .37 immol-) and intermediate 341.33 μmol). The reaction mixture was stirred at 20°C for 3 hours. The reaction mixture was poured into water (2.0 mL). The solution was extracted with EtOAc (2.0 mL*3). The combined organic extracts were washed with brine (5.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by silica gel flash chromatography (PE:EA=50:1-10:1-3:1-1:1) to give intermediate i-53d as a white solid.

LCMS (ESI) m/z: [M+H]+=631,4.

HPLC-Lösung: OJ-3-MeOH(DEA)-60-7MIN-3ML-35T, 3.881 Min.

Step 5: Preparation of (S)-N-(6-amino-1-oxo-1-((4-phenylthiazol-2-yl)amino)hexan-2-yl)benzamide (intermediate i-53e)

To a solution of intermediate i-53d (120 mg, 179.50 μmol) in DCM (1.2 mL) was added piperidine (120.00 μL). The solution was stirred at 20°C for 2 h. The mixture was poured into water (2.0 mL). The reaction mixture was diluted with saturated NaHCO 33solution (5 mL) and extracted with EtOAc (2.0 mL*3). The combined organic extracts were washed with brine (5.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by preparative HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.225% FA)-ACN]; B%: 17%-37%, 7 min) to give the intermediate i to give -53e (FA salt) as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 8,73 (d, J=6,8 Hz, 1Η), 8,42 (s, 1Η), 7,93-7,89 (m, 4Η), 7,62 (s, 1Η), 7,58-7,54 (m, 1Η), 7,50-7,47 (m, 2Η), 7,44-7,40 (m, 2Η), 7, 33 -7,30 (m, 1Η), 4,67-4,63 (m, 1Η), 2,73 (s, 2H), 1,85 (d, J=6,4 Hz, 2H), 1 53-1,40 (m, 4Η) ppm.

LCMS (ESI) m/z: [M+H]+=409,3.

HPLC-Quirale: OD-RH_0-60_1ML.M, 8.037 Min.

Step 6: Preparation of (S)-N-(6-Acetamido-1-oxo-1-((4-phenylthiazol-2-yl)amino)hexan-2-yl)benzamide (Compound 43)

A solution of intermediate i-53e (13 mg, 31.82 μmol) in Ac2(0.2 ml) was stirred at 30°C for 2 h. The reaction mixture was filtered to give a filter cake. The filter cake was washed with MTBE (2.0 mL) and dried in vacuo to give a solid. The solid was poured into water (2.0 mL) and the solution was lyophilized to give compound 43 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12.46 (br s, 1H), 8.67 (d, J=7.2 Hz, 1H), 7.94-7.90 (m, 4H) , 7.84 (s, 1H), 7.64 (s, 1H), 7.57-7.51 (m, 1H), 7.53-7.47 (m, 2H), 7.49-7 , 44 (m, 2H), 7.44-7.42 (m, 1H), 4.67-4.62 (m, 1H), 3.04 (m, 2H), 1.85 (m, 2H ), 1.78 (s, 3H), 1.44-1.35 (m, 4H) ppm.

LCMS (ESI) m/z: [M+H]+=451,3.

HPLC-Lösung: Cellucoat-MeOH(DEA)-40-3ML-35T, 0,767 Min.

Example 54. Preparation of 3-(Isopropylsulfonyl)-N-(2-oxo-2-((4-phenylthiazol-2-yl)amino)ethyl)benzamide (Compound 6)

Step 1: Preparation of methyl 3-(isopropylthio)benzoate (intermediate i-54a)

To a solution of methyl 3-sulfanylbenzoate (200.0 mg, 1.19 mmol) in THE (2.0 mL) was added 2-iodopropane (303.17 mg, 1.78 mmol). NaH (57.07 mg, 1.43 mmol, 60% purity) was then added in small portions over 5 min. The reaction mixture was stirred at 20°C for 1 h and then poured into 1N HCl (6 mL). The solution was extracted with EtOAc (5.0 mL*3). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiO).2, PE/EA=100/1-1/1) to give intermediate i-54a as a yellow oil.

LCMS (ESI) m/z: [M+H]+= 211,3.

Step 2: Preparation of methyl 3-(isopropylsulfonyl)benzoate (intermediate i-54b)

To a solution of intermediate i-54a (200.0 mg, 883.15 μmol) in DCM (2.0 mL) was added m-CPBA (320.04 mg, 1.58 mmol, 85% purity). The mixture was stirred at 20°C for 1 hour and then poured into saturated aqueous NaHCO 3 .3(30.0 mL). The solution was extracted with EtOAc (20.0 mL*3). The combined organic extracts were washed with brine (50.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give intermediate i-54b as a white solid, which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=243,3.

Step 3: Preparation of 3-(isopropylsulfonyl)benzoic acid (intermediate i-54c)

To a solution of intermediate i-54b (300.0 mg, 1.24 mmol) in dioxane (3.0 mL) and water (2.0 mL) was added LiOH (59.30 mg, 2.48 mmol). The reaction mixture was stirred at 20°C for 1 h and then adjusted to pH 6 with saturated citric acid solution. The solution was then extracted with EtOAc (10.0 mL*3). The combined organic extracts were washed with brine (20.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiO).2, PE:EA=50:1-20:1-10:1-3:1) to give intermediate i-54c as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 13,58 (s, 1Η), 8,31-8,27 (m, 2Η), 8,11-8,03 (m, 1Η), 7,82 (d, J=7,6 Hz, 1Η), 3,53-3,45 (m, 1Η), 1,16 (d, J=6,8 Hz, 6Η) ppm.

LCMS (ESI) m/z: [M+H]+=229,2.

Step 4: Preparation of 3-(Isopropylsulfonyl)-N-(2-oxo-2-((4-phenylthiazol-2-yl)amino)ethyl)benzamide (Compound 6)

To a solution of intermediate i-54c (30.0 mg, 129.42 μmol) in DCM (0.3 mL) was added HATU (59.05 mg, 155.30 μmol), DIEA (66.90 mg, 517, 66 μmol) and 2-amino-N-(4-phenylthiazol-2-yl)acetamide (30.19 mg, 86.93 μmol). The reaction mixture was stirred at 30°C for 12 h and then poured into water (2.0 mL). The solution was extracted with EtOAc (2.0 mL*3). The combined organic extracts were washed with brine (5.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. MeOH (0.5 mL) was added to the residue and stirred for 5 min, then filtered. The solid was lyophilized to give Compound 6 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 12,49 (s, 1Η), 9,31-9,28 (m, 1Η), 8,38 (s, 1Η), 8,27 (d, J = 8,0 Hz, 1H), 8,05 (d, J=7,6 Hz, 1H), 7,90 (d, J=7,2 Hz, 2H), 7,84-7,80 (m , 1Η), 7,65 (s, 1Η), 7,45-7,43 (m, 2Η), 7,41-7,32 (m, 1Η), 4,24 (d, J=5,6 Hz, 2Η), 3,54-3,47 (m, 1 Η), 1,18 (d, J=6,8 Hz, 6Η) ppm.

LCMS (ESI) m/z: [M+H]+=444,3.

Example 55. Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-[(6-phenyl-2-pyridyl)amino]ethyl]benzamide (Compound 41)

Step 1: Preparation of tert-butyl N-[2-oxo-2-[(6-phenyl-2-pyridyl)amino]ethyl]carbamate (intermediate i-55a)

In a solution of 2-( tert -butoxycarbonylamino)acetic acid (113.21 mg, 646.26 μmol), DIPEA (303.73 mg, 2.35 mmol) and HATU (245.73 mg, 646.26 μmol) in DCM (2.0 mL)pyridin. -2-amine (100.0 mg, 587.51 μmol) and the mixture was stirred at 20°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 5:1 to 2:1) to give intermediate i-55a as a pale yellow oil.

1H RMN (400 MHz, CDCl3) δ 8.48 (s, 1H), 8.14 (d, J=8 Hz, 1H), 7.96 (d, J=7.2 Hz, 2H), 7.82-7.78 (m , 1H), 7.52-7.45 (m, 4H), 5.22-5.21 (m, 1H), 4.03 (s, 2H), 1.52 (s, 9H) ppm.

LCMS (ESI) m/z: [M+H]+=328,0.

Step 2: Preparation of 2-amino-N-(6-phenyl-2-pyridyl)acetamide (intermediate i-55b)

To a solution of intermediate i-55a (130 mg, 381.21 μmol) in DCM (2.0 mL) was added TFA (192.00 g L) and the mixture was stirred at 20 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was triturated with MTBE (5 ml), the solid collected by filtration and dried in vacuo to give intermediate i-55b (TFA salt) as a white solid.

LCMS (ESI) m/z: [M+H]+=228,0.

Step 3: Preparation of 3-(dimethylsulfamoyl)-N-[2-oxo-2-[(6-phenyl-2-pyridyl)amino]ethyl]benzamide trifluoroacetate (Compound 41)

In a solution of 3-(dimethylsulfamoyl)benzoic acid (36.58 mg, 159.54 µmol), HATU (60.66 mg, 159.54 µmol) and DIPEA (74.98 mg, 580.16 µmol) in DCM HCl ( 1.0 mg Interbte i-05 ml) ). , 145.04 μmol) were added and the mixture was stirred at 25°C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 35%-65%, 9 min) and lyophilized to give Compound 41 (TFA salt) as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ 10.68 (s, 1H), 9.24-9.21 (m, 1H), 8.27-8.26 (m, 2H), 8.10 - 8.08 (m, 2H), 8.02 (d, J =8.0 Hz, 1H), 7.96-7.93 (m, 1H), 7.90-7.88 (m, 1H ), 7.82-7.78 (m, 1H), 7.69 (d, J=8.0 Hz, 1H), 7.51-7.46 (m, 3H), 4.23 (d, J =6.0 Hz, 2H), 2.68-2.66 (m, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=439,0.

Example 56. Preparation of N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]thiophene-2-carboxamide (Compound 58)

In a solution of thiophene-2-carboxylic acid (22.93 mg, 178.90 μmol), HATU (68.02 mg, 178.90 μmol), and DIPEA (84.08 mg, 650.55 μmol) in DCM (1st , 0 ml)-2-amino-(2S)-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50.00 mg, 162.64 μmol) was added and the mixture was stirred at 20° for 2 h C stirred.

The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150 x 25 x 10 µm, mobile phase: [water (0.1% TFA)-ACN], B%: 50%-80%, 9 min) purified and lyophilized compound 58 as a whiter solid.

1H RMN (400 MHz, CDCl3) δ=7.69 (d, J=7.2 Hz, 2H), 7.62-7.61 (m, 1H), 7.48 (d, J=4.8 Hz, 1H), 7, 40-7.36 (m, 2H), 7.34-7.30 (m, 1H), 7.26-7.24 (d, J=8.0 Hz, 1H), 7.07 (s, 1H), 7.06-7.03 (m, 1H), 5.00-4.95 (m, 1H), 2.68-2.63 (m, 2H), 2.29-2.20 ( m, 2H), 2.10 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=418,0

HPLC solution: Amycoat-MeOH(DEA)-40-7 min-3 mL-35T, 3.772 min

Example 57. Preparation of (S)-N-(4-(Methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzo[b]thiophene-2-amide carboxylic acid (Compound 71)

To a solution of benzothiophene-2-carboxylic acid (50.0 mg, 280.57 μmol, 47.17 μL) in DCM (1.0 mL) was added HATU (128.02 mg, 336.69 μmol), DIEA (108 .71 μmol) and 108 .71 μmol (284 μl). )-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (118.25 mg, 280.57 µmol). The reaction mixture was stirred at 20°C for 12 h. The reaction mixture was then poured into water (2.0 mL) and extracted with EtOAc (2.0 mL * 3). The combined organic extracts were washed with brine (5.0 mL) and dried over Na2AFTERWARD4filtered and concentrated in vacuo to give a residue. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 52%-82%, 9 min) and lyophilized to give Compound 71 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.58 (s, 1H), 9.08 (d, J=7.2 Hz, 1H), 8.31 (s, 1H), 8.03 - 7.92 (m, 2H), 7.91 (d, J=7.2 Hz, 2H), 7.66 (s, 1H), 7.48-7.43 (m, 4H), 7, 41 -7.33 (m, 1H), 4.79-4.74 (m, 1H), 2.68-2.66 (m, 1H), 2.59-2.54 (m, 1H), 2.16-2.11 (m, 5H) ppm.

LCMS (ESI) m/z: [M+H]+=468,3.

HPLC-Lösung: Cellucoat-MeOH+CAN(DEA)-40-7MIN-3ML-35T, 5.321 Min.

Example 58. Preparation of 5-(dimethylsulfamoyl)-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]thiophene-2-carboxamide (Compound 62)

Step 1: Preparation of 4-chlorosulfonylthiophene-2-carboxylic acid (intermediate i-58a) and 5-chlorosulfonylthiophene-2-carboxylic acid (intermediate i-58b)

Thiophene-2-carboxylic acid (300.0 mg, 2.34 mmol) was added to sulfuric acid (1.36 g, 11.71 mmol) and the mixture was stirred at 100°C for 1 h. The reaction mixture was carefully added dropwise to ice water (30.0 mL) and stirred for 5 min. The precipitate was filtered and dried in vacuo to give a mixture of i-58a and i-58b as a white solid which was used in the next step without further purification.

1H-NMR (400 MHz, DMSO-d6) δ 7.81 (d, J=1.6 Hz, 2H), 7.60 (d, J=1.6 Hz, 2H), 7.51 (d, J = 4 Hz, 1 H), 7.13 (d, J=4 Hz, 1 H) ppm.

Step 2: Preparation of 4-(dimethylsulfamoyl)thiophene-2-carboxylic acid (intermediate i-58c) and 5-(dimethylsulfamoyl)thiophene-2-carboxylic acid (intermediate i-58d)

To a mixture of i-58a and i-58b (260.0 mg, crude) in THE (2.0 mL) was added N-methylmethanamine (2.29 mL, 4.59 mmol, 2.0 M in THF) at 0° C, the mixture was stirred at 0°C for 2 h at 25°C. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150 x 25 x 10 µm, mobile phase: [water (0.1% TFA)-ACN], B%: 18%-40%, 12 min) intermediate i-58c was purified and lyophilized as a white solid and intermediate i-58d as a white solid.

Intermediate i-58c:

1H RMN (400 MHz, DMSO-d6) δ 8,51 (d, J=1,6 Hz, 1Η), 7,78 (d, J=1,6 Hz, 1Η), 2,73-2,66 (m, 7Η) ppm.

LCMS (ESI) m/z: [M+H]+=236,0.

Intermediate i-58d:

1H RMN (400 MHz, DMSO-d6) δ 7,82 (d, J=4,0 Hz, 1Η), 7,66 (d, J=4,0 Hz, 1Η), 2,70-2,67 (m, 6Η) ppm.

LCMS (ESI) m/z: [M+H]+=236,0.

Step 3: Preparation of 5-(dimethylsulfamoyl)-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]thiophene-2-carboxamide (Compound 62)

In a solution of intermediate i-58d (40.0 mg, 170.01 μmol), DIPEA (87.89 mg, 680.04 μmol), and HATU (71.11 mg, 187.01 μmol) in DCM (1.0 mL ) (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (78.82 mg, 187.01 μmol) was added and the mixture was stirred at 30 °C for 2 hours on tap. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 43%-73%, 10 min) and lyophilized to give Compound 62 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.61 (s, 1H), 9.15 (d, J=7.6 Hz, 1H), 8.07 (d, J=4.0 Hz , 1H), 7.92-7.90 (m, 2H), 7.71 (d, J=4.0 Hz, 1H), 7.66 (s, 1H), 7.46-7.42 ( m, 2H), 7.35-7.31 (m, 1H), 4.78-4.67 (m, 1H), 2.69 (s, 6H), 2.65-2.56 (m, 2H), 2.17-2.07 (m, 5H) ppm.

LCMS (ESI) m/z: [M+H]+=525,1.

Example 59. Preparation of N-(2-((4-(2,3-Dihydrobenzo[b][1,4]dioxin-6-yl)thiazol-2-yl)amino)-2-oxoethyl)-3-( N,N-Dimethylsulfamoyl)benzamide (Compound 15)

To a solution of N-[2-[(4-bromothiazol-2-yl)amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (30.0 mg, 67.07 μmol) and 2,3- dihydro-1,4-benzodioxin-6-ylboronic acid (36.21 mg, 201.20 μmol) in dioxane (2.0 mL) and H2O (0.2 mL), DIEA (43.34 mg, 335.33 μmol) and palladium; tri-tert-butylphosphane (17.14 mg, 33.53 μmol) were added under N2. The mixture was stirred at 100°C for 2 hours. The reaction mixture was filtered. The filtrate was concentrated to give a residue. The residue was purified by prep-HPLC (column: Boston pH-lex 150*25 10 μm; mobile phase: [water (0.1% TFA)-ACN]; B%: 39%-59%, 8 min) and lyophilized to give compound 15 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.42 (s, 1H), 9.30-9.28 (m, 1H), 8.26-8.22 (m, 2H), 7, 93 (d, J=7.6 Hz, 1H), 7.82 -7.80 (m, 1H), 7.48 (s, 1H), 7.39-7.35 (m, 2H), 6 , 89 (d, J=8.0 Hz, 1H), 4.27 (s, 4H), 4.22 (d, J=6.0 Hz, 2H), 2.65 (s, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=503,3.

Example 60. Preparation of (S)-3-(isopropylsulfonyl)-N-(4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)benzamide ( compound 14)

To a solution of 3-(isopropylsulfonyl)benzoic acid (30.0 mg, 129.42 μmol) in DCM (0.3 mL) was added HATU (59.05 mg, 155.30 μmol), DIEA (66.90 mg, 517 .66 μmol)-2- and given. Amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (54.54 mg, 129.42 µmol). The reaction mixture was stirred at 30°C for 12 h. The reaction mixture was then poured into water (2.0 mL) and extracted with EtOAc (2.0 mL * 3). The combined organic extracts were washed with brine (5.0 mL) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by pre-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 45%-75%, 9 min) and lyophilized to give compound 14 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,59 (s, 1Η), 9,12 (d, J=7,2 Hz, 1Η), 8,39 (s, 1Η), 8,30 (d, J=8,0 Hz, 1H), 8,05 (d, J=7,6 Hz, 1H), 7,91 (d, J=7,2 Hz, 2H), 7,83-7 , 79 (m, 1Η), 7,66 (s, 1Η), 7,45-7,41 (m, 2Η), 7,34 -7,30 (m, 1Η), 4,82-4,77 (m, 1Η), 3,51 (s, 1Η), 2,64-2,55 (m, 2Η), 2,16-2,09 (m, 2Η), 2,10 (s, 3Η), 1,18 -1,16 (m, 6Η) ppm.

LCMS (ESI) m/z: [M+H]+=518,3

HPLC-Lösung: Cellucoat-MeOH(DEA)-40-3ML-35T, 1.119 Min.

Example 61 Preparation of N-[(1S)-3-Methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]-3-(trifluoromethyl)benzamide (Compound 77)

To a solution of 3-(trifluoromethyl)benzoic acid (22.55 mg, 118.64 μmol) in DCM (3 mL) was added DIPEA (46.00 mg, 355.91 μmol) and HATU (49.62 mg, 130, 50 μmol). The mixture was stirred at 30°C for 10 min. (2S)-2-Amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 µmol) was added and the reaction mixture was stirred at 30°C for 2 h. The reaction mixture was evaporated to dryness to give a residue. The residue was purified by preparative HPLC (column: Phenomenex Synergi C18 150 x 25 x 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 53%-80%, 12 min) and lyophilized for to give Compound 77 (97.3% ee value) as a white solid.

1H-NMR (400 MHz, METANOL-d4) δ 8,27 (m, 1Η) 8,20 (d, J=8,0 Hz, 1Η), 7,93-7,89 (m, 3Η), 7 , 73 (m, 1Η), 7,42-7,39 (m, 3Η), 7,34-7,28 (m, 1Η), 4,97-4,95 (m, 1Η), 2,73 - 2,66 (m, 2Η), 2,30-2,24 (m, 2Η), 2,16 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H] + =480.1.

SFC-Quiral: OD-3_5CM_MEOH(DEA)_40_3ML_5MIN_T35.M., 0,601 λεπτά.

Example 62. Preparation of N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]-4-(trifluoromethyl)benzamide (Compound 68)

To a solution of 4-(trifluoromethyl)benzoic acid (22.55 mg, 118.64 μmol) in DCM (3 mL) was added DIPEA (46.00 mg, 355.91 μmol) and HATU (49.62 mg, 130, 50 μmol). The mixture was stirred at 30°C for 10 min. (2S)-2-Amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 µmol) was added and the reaction mixture was stirred at 30°C for 2 h. The reaction mixture was evaporated to dryness to give a residue. The residue was purified by preparative HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 65%-85%, 8 min) and lyophilized for to give compound 68 as a white solid.

1HNMR (400 MHz, METANOL-d4) δ 8,09 (d, J=8,0 Hz, 1H), 7,92 (d, J=7,2 Hz, 1H), 7,83 (d, J= 8,4 Hz, 2Η), 7,42-7,39 (m, 3Η), 7,33-7,31 (m, 1Η), 4,97-4,94 (m, 1Η), 2,73 -2,66 (m, 2Η), 2,33-2,24 (m, 2Η), 2,16 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H] + =480.1.

SFC-Quiral: Cellucoat-MeOH(DEA)-40-7 Min.-3 ml-35T, 0,937 Min.

Example 63. Paraskevi N4-Methyl-N1-[(1S)-3-Methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]terephthalamide (Verbindung 72)

Step 1: Preparation of methyl 4-[[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]carbamoyl]benzoate (intermediate i-63a)

To a solution of 4-methoxycarbonylbenzoic acid (85.49 mg, 474.54 μmol) in DCM (5 mL) was added DIPEA (183.99 mg, 1.42 mmol) and HATU (198.48 mg, 522.00 μmol) . The mixture was stirred at 30°C for 10 min. (2S)-2-Amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (200 mg, 474.54 μmol) was added and the reaction mixture was stirred at 30°C for 2 h. The reaction mixture was quenched by addition of water (10 mL) and extracted with ethyl acetate (10 mL x 2). The combined organic extracts were washed with water (10 ml) and dried over Na2AFTERWARD4, filtered and concentrated under reduced pressure to give intermediate i-63a as a colorless oil, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H]+=470,3.

Step 2: Preparation of 4-[[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]carbamoyl]benzoic acid (intermediate i-63b)

To a solution of intermediate i-63a (258.82 mg, 468.50 μmol) in MeOH (2.0 mL) and H2O (2.0 mL) was treated with NaOH (36.39 mg, 909.75 μmol). The mixture was stirred at 30°C for 18 hours. The reaction mixture was concentrated in vacuo. The pH of the residue was adjusted to pH 4 with 20% HCl solution. A white precipitate formed and the mixture was filtered. The filter cake was washed with water (4.0 mL) and concentrated to dryness to give intermediate i-63b as an off-white solid.

LCMS (ESI) m/z: [M+H]+=456,3.

Step 3: Preparation of N4-Methyl-N1-[(1S)-3-Methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]terephthalamide (Verbindung 72)

To a solution of intermediate i-63b (40 mg, 81.66 μmol) in DCM (3 ml) was added HATU (36.15 mg, 95.07 μmol) and DIPEA (20.48 mg, 158.46 μmol). The mixture was stirred at 30°C for 10 min. Methanamine (2M, 396.05 µL) was added and the reaction mixture was stirred at 30°C for 3 hours. The reaction mixture was evaporated to dryness to give a residue. The rest was preparation. HPLC (column: Boston pH-lex 150*25 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 50%-70%, 8 min) and lyophilized to give Compound 72 as a white solid.

1H-NMR (400 MHz, METANOL-d4) δ 8,00 (d, J=8,4 Hz, 2H), 7,94-7,91 (m, 4H), 7,42-7,39 (m , 3Η), 7,33-7,29 (m, 1Η), 4,97-4,94 (m, 1Η), 2,95 (s, 3Η), 2,73-2,66 (m, 2Η ), 2,33-2,24 (m, 2Η), 2,16 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H] + =469.3.

Example 64. Preparation of (S)-tert-butyl 3-((4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)carbamoyl)benzylcarbamate ( connection 75)

Step 1: Preparation of (S)-tert-butyl 3-((4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)carbamoyl)benzylcarbamate ( Intermediate i-64a)

To a solution of 3-[( tert -butoxycarbonylamino)methyl]benzoic acid (29.81 mg, 118.64 μmol) in DCM (2 mL) was then added HATU (67.66 mg, 177.96 μmol) and DIPEA ( 61n 34 mg, 47 µmol) Added (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 µmol). The mixture was stirred at 10°C for 12 hours. The reaction mixture was concentrated under reduced pressure.

The residue was purified by prep-TLC (SiO 2 , PE:EA=1:1) to give intermediate i-64a as a white solid.

LCMS (ESI) m/z: [M+H]+=541,1.

Step 2: Preparation of (S)-tert-butyl 3-((4-(methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)carbamoyl)benzylcarbamate ( Compound 75) trifluoroacetate

To a solution of intermediate i-64a (50 mg, 76.37 μmol) in DCM (2.0 mL) was added TFA (1.0 mL). The mixture was stirred at 12°C for 2 h. The reaction mixture was concentrated under reduced pressure to remove DCM. The rest was preparation. HPLC (TFA condition; column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water(0.1% TFA)-ACN]; B%: 25%-55%, 9 min) purified and lyophilized, for to give compound 75 (TFA salt) as a white solid.

1H NMR (400 MHz, METHANOL-d4) δ=7.99–7.97 (m, 2H), 7.91–7.89 (m, 2H), 7.66–7.64 (m, 1H) , 7.61-7.59 (m, 1H), 7.41- 7.36 (m, 3H), 7.31-7.29 (m, 1H), 4.95-4.91 (m, 1H), 4.19 (s, 2H), 2.72-2.65 (m, 2H), 2.30-2.20 (m, 2H), 2.15 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=441,0.

Example 65. Preparation of 4-(aminomethyl)-N-[3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 57)

Step 1: Preparation of tert-butyl N-[[4-[[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]carbamoyl]phenyl]methyl]carbamate (intermediate i - 65a)

To a mixture of 4-[( tert -butoxycarbonylamino)methyl]benzoic acid (89.43 mg, 355.91 μmol) in DCM (1 mL) was added HATU (202.99 mg, 533.86 μmol) and DIPEA (247, 97.2 mmol). The mixture was stirred at 30°C for 15 min, then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (150 mg, 355.91 μmol) was added and under stirring at 30°C for 16 hours. The reaction mixture was evaporated to dryness to give the crude product. The rest was preparation. HPLC (column: Boston pH-lex 150*25 10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 59%-79%, 8 min) and lyophilized to give Intermediate i -65a as a white solid.

1H-NMR (400 MHz, MeOD-d4) δ=7,91–7,86 (m, 4H), 7,41–7,37 (m, 5H), 7,31–7,28 (m, 1H), 4,92 (brd, J= 5,2 Hz, 1Η), 4,30 (s, 2H), 2,74 -2,60 (m, 2H), 2,33-2,17 (m, 2H), 2,14 (s, 3H ), 1,46 (s, 9Η) ppm.

LCMS (ESI) m/z: [M+H]+=541,3.

Step 2: Preparation of 4-(aminomethyl)-N-[3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide trifluoroacetate (Compound 57)

To a solution of intermediate i-65a (150.00 mg, 277.42 μmol) in DCM (1 mL) was added TFA (205.40 μL). The mixture was stirred at 30°C for 2 hours. The reaction mixture was evaporated to dryness to give the crude product. The rest was preparation. Compound 57 (TFA-salt) was purified and lyophilized as a white solid.

1H-NMR (400 MHz, MeOD-d4) δ=8.00 (d, J=8.4 Hz, 2H), 7.91-7.89 (m, 2H), 7.58 (d, J=8.4 Hz, 2H), 7, 41-7.37 (m, 3H), 7.32-7.28 (m, 1H), 4.94-4.91 (m, 1H), 4.20 (s, 2H), 2.71- 2.64 (m, 2H), 2.31-2.22 (m, 2H), 2.14 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=441,2.

Example 66. Preparation of 4-[(dimethylamino)methyl]-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 60)

In 4-[(dimethylamino)methyl]benzoic acid mixt. Hydrochloride (25.59 mg, 118.64 μmol) in DCM (1 mL) was added to HATU (67.66 mg, 177.96 μmol) and DIPEA (82.66 μL, 474.56 μmol). The mixture was stirred at 30°C for 15 min, then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 μmol) was added and under stirring at 30°C for 2 hours. The reaction mixture was evaporated to dryness. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 25%-55%, 9 min) and lyophilized to give Compound 60 as a non-white solid.

1H-NMR (400 MHz, MeOD-d4) δ=8.03 (d, J=8.0 Hz, 2H), 7.90 (d, J=8.0 Hz, 2H), 7.63 (d, J=8.4 Hz, 2H) , 7.40-7.36 (m, 3H), 7.31- 7.29 (m, 1H), 4.95-4.91 (m, 1H), 4.39 (s, 2H), 2 .88 (s, 6H), 2.71-2.64 (m, 2H), 2.29-2.19 (m, 2H), 2.14 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=469,1.

Example 67. Preparation of 4-(acetamidomethyl)-N-[3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 74)

In a solution of 4-(aminomethyl)-N-[3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (50 mg, 90.15 μmol, TFA salt) in DCM (1 mL) TEA (62.74 μL, 450.77 μmol) was added followed by Ac2O (21.11 µL, 225.39 µmol) at 0°C. The mixture was stirred at 30°C for 1 hour. The reaction mixture was evaporated to dryness. The residue was purified by preparative HPLC (column: and lyophilized to give compound 74 as an off-white solid.

1H-NMR (400 MHz, MeOD-d4) δ=7,90–7,87 (m, 4H), 7,41–7,36 (m, 5H), 7,31–7,27 (m, 1H), 4,92–4,90 ( m, 1Η), 4,43 (s, 2H), 2,73-2,60 (m, 2H), 2,27-2,21 (m, 2H), 2,13 (s, 3H), 2 ,01 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=483,3.

Example 68. Preparation of compound N-[(1S)-3-Methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]-4-methylsulfonylbenzamide (Compound 68)

To a solution of 4-methylsulfonylbenzoic acid (23.75 mg, 118.64 μmol) in DCM (3 mL) was added DIPEA (46.00 mg, 355.91 μmol) and HATU (49.62 mg, 130.50 μmol) . The mixture was stirred at 30°C for 10 min. (2S)-2-Amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50 mg, 118.64 µmol) was added and the reaction mixture was stirred at 30°C for 2 h. The reaction mixture was evaporated to dryness to give a residue. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 45%-69%, 12 min) and lyophilized to give compound 68 as a white solid.

1H-NMR (400 MHz, METANOL-d4) δ=8,19-8,05 (m, 4H), 7,92 (d, J=7,2 Hz, 2H), 7,42-7,39 ( m, 3Η), 7,33-7,31 (m, 1Η), 4,97-4,94 (m, 1Η), 3,19 (s, 3Η), 2,73-2,66 (m, 2Η), 2,33-2,24 (m, 2Η), 2,16 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H] + =490.1.

Example 69. Preparation of 4-Methoxy-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]-3-methylsulfonylbenzamide (Compound 20)

To a solution of 4-methoxy-3-methylsulfonylbenzoic acid (21.85 mg, 94.91 μmol) in DCM (1 mL) was added HATU (43.30 mg, 113.89 μmol) and DIEA (36.80 mg, 284 .73 µmol) The mixture was stirred at 15°C for 5 min. Then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (40 mg, 94.91 μmol) was added and the mixture was stirred at 15°C for 1 h. The solvent was removed under reduced pressure and the residue was purified by preparative HPLC (column: Phenomenex Synergi C18 150 x 25 x 10 µm, mobile phase: [water (0.1% TFA)-ACN], B%: 42% - 72) . %, 9 min) and lyophilized to give compound 20 as a white solid.

1H-NMR (400 MHz, MeOD) δ=8.49 (d, J=2.4, 1H), 8.26-8.23 (m, 1H), 7.90-7.88 (m, 2H ), 7.40–7.36 (m, 4H), 7.31–7.27 (m, 1H), 4.92–4.91 (m, 1H), 4.08 (s, 3H), 3.26 (s, 3H), 2.73-2.60 (m, 2H), 2.34-2.17 (m, 2H), 2.14 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=520,1.

Example 70. Preparation of 4-methyl-3-(methylsulfonyl)-N-(2-oxo-2-((4-phenylthiazol-2-yl)amino)ethyl)benzamide (Compound 19)

Step 1: Preparation of 4-methyl-3-(methylsulfonyl)benzoic acid (intermediate i-70a)

NaOH (13.14 mg, 328.57 μmol) was dissolved in H2O (1 mL) and then to a solution of methyl 4-methyl-3-methylsulfonylbenzoate (50 mg, 219.04 μmol) in THE (0.5 mL) and MeOH (0.2 mL). The reaction mixture was stirred at 30°C for 17 hours. The reaction mixture was diluted with water (3 mL) and then washed with EtOAc (5 mL). The aqueous layer was adjusted to pH=3 with 2M aqueous solution. HCl, then extracted with EtOAc (5 mL*3). The combined organic extracts were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated to give intermediate i-70a as a white solid.

LCMS (ESI) m/z: [M+H] + =215.0.

Step 2: Preparation of 4-methyl-3-(methylsulfonyl)-N-(2-oxo-2-((4-phenylthiazol-2-yl)amino)ethyl)benzamide (Compound 19)

To a solution of intermediate i-70a (25 mg, 116.69 μmol) in DCM (1 ml) were added HATU (66.56 mg, 175.04 μmol) and DIPEA (75.41 mg, 583.46 μmol) and then 2-amino-N-(4)-phenylthiazol-2-yl)acetamide (40.53 mg, 116.69 µmol). After stirring at 30°C for 2 h, a precipitate formed. The precipitate was collected by filtration and lyophilized to give compound 19 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.09-10.54 (br s, 1H), 9.20-9.19 (m, 1H), 8.44 (s, 1H), 8 , 13-7.10 (m, 1H), 7.91-7.89 (m, 2H), 7.63-7.60 (m, 2H), 7.45-7.40 (m, 2H) , 7.35-7.32 (m, 1H), 4.22 (d, J=5.6 Hz, 2H), 3.27 (s, 3H), 2.71 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=430,3.

Example 71. Preparation of 4-(dimethylsulfamoyl)-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]thiophene-2-carboxamide (Compound 64)

For a solution of 4-(dimethylsulfamoyl)thiophene-2-carboxylic acid (40.00 mg, 170.01 μmol), DIPEA (87.89 mg, 680.04 μmol, 118.45 μL), and HATU (71.11 mg , 187 μL) to D. was added (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (78.82 mg, 187.01 μmol) μmol and the mixture was stirred at 30 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The rest was preparation. HPLC (column: Phenomenex Synergi C18 150 x 25 x 10 µm, mobile phase: [water (0.1% TFA)-ACN], B%: 45%-75%, 10 min) purified and lyophilized compound 64 as a whiter solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.60 (s, 1H), 9.09 (d, J=7.2 Hz, 1H), 8.46 (d, J=1.6 Hz , 1H), 8.27 (d, J=1.6 Hz, 1H), 7.92-7.90 (m, 2H), 7.67 (s, 1H), 7.46-7.42 ( m, 2H), 7.35–7.33 (m, 1H), 4.77–4.73 (m, 1H), 2.73–2.67 (m, 6H), 2.66–2, 53 (m, 2H), 2.10-2.03 (m, 5H) ppm.

LCMS (ESI) m/z: [M+H]+=525,0

Example 72. Preparation of 3-cyclopropylsulfonyl-N-[2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 39)

Step 1: Preparation of 1-cyclopropylsulfanyl-3-methylbenzene (intermediate i-72a)

A mixture of 3-methylbenzenethiol (100.0 mg, 805.13 μmol), bromocyclopropane (292.21 mg, 2.42 mmol), and t -BuOK (99.38 mg, 885.65 μmol) in DMSO (0.5 ml) was stirred at 1102°C. hours. The reaction mixture was diluted with water (2.0 mL) and extracted with EtOAc (2.0 mL*3). The combined organic extracts were dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2, petroleum ether) to give intermediate i-72a as a colorless oil.

1H RMN (400 MHz, CDCl3) δ=7.18 (s, 1H), 7.10 (d, J=4.0 Hz, 3H), 6.88-6.86 (m, 1H), 2.29-2.25 (m , 3H), 2.14-2.09 (m, 1H), 1.01-0.96 (m, 2H), 0.63-0.60 (m, 2H) ppm.

Step 2: Preparation of 3-cyclopropylsulfonylbenzoic acid (intermediate i-72b)

For a solution of intermediate i-72a (60.0 mg, 365.26 μmol) and KMnO4(230.89 mg, 1.46 mmol) in water (1.0 mL) was added NaOH (7.30 mg, 182.63 μmol). The mixture was stirred at 100°C for 16 hours. The reaction mixture was poured into N/D2AFTERWARD3(20.0 mL), then 1N HCl (20.0 mL) was added to the mixture and extracted with EtOAc (20.0 mL*3). The combined organic extracts were washed with brine (20.0 mL) and dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give intermediate i-72b as a yellow oil.

1H-NMR (400 MHz, DMSO-d6) δ=8,35 (s, 1H), 8,26 (d, J=7,6 Hz, 1H), 8,13 (d, J=7,6 Hz , 1Η), 7,82-7,78 (m, 1Η), 2,99-2,93 (m, 1Η), 1,17-1,06 (m, 4Η) ppm.

LCMS (ESI) m/z: [M+H]+=227,0

Step 3: Preparation of 3-cyclopropylsulfonyl-N-[2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 39)

In a solution of intermediate i-72b (30.0 mg, 132.60 µmol), HATU (60.50 mg, 159.12 µmol), and DIPEA (68.55 mg, 530.39 µmol) in DCM (1, 0 mL) 2-amino-N-(4-phenylthiazol-2-yl)acetamide (50.66 mg, 145.86 µmol) was added. The mixture was then stirred at 30°C for 4 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was triturated with MeOH (4.0 mL), the solid collected by filtration and dried in vacuo to give a white solid. The solid was then suspended in water (3.0 mL) and lyophilized to give compound 39 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,46 (s, 1Η), 9,27 (m, 1Η), 8,42 (s, 1Η), 8,25 (d, J=8, 0 Hz, 1H), 8,10 (d, J =8,4 Hz, 1H), 7,92-7,90 (m, 2H), 7,81-7,79 (m, 1H), 7, 64 (s, 1H), 7,44-7,42 (m, 2H), 7,33-7,31 (m, 1H), 4,24 (d, J=5,6 Hz, 2H), 2 , 95-2,90 (m, 1Η), 1,19-1,07 (m, 4Η) ppm.

LCMS (ESI) m/z: [M+H]+=442,3.

Example 73. Preparation of 3-[2-[[2-[[3-(dimethylsulfamoyl)benzoyl]amino]acetyl]amino]thiazol-4-yl]-N,N-dimethylbenzamide (Compound 37)

To a solution of N-[2-[(4-bromothiazol-2-yl)amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (50.0 mg, 111.78 μmol) and [3-( dimethylcarbamoyl)phenyl]boronic acid (64.72 mg, 335.33 μmol) in dioxane (3.0 mL) and H20 (0.3 ml) of Pd(t-Bu) was added.3PI)2(28.56 mg, 55.89 μmol) e DIEA (72.23 mg, 558.88 μmol, 97.34 μL) sob N2. The mixture was stirred at 100°C for 2 hours. The solution was cooled to 30°C and a solid formed. The mixture was filtered to give the crude product as a solid. The crude product was dissolved in MeOH (1.0 mL) and DMSO (1.0 mL) and purified by pre-HPLC (column: Phenomenex Synergi C18 150*25*10 μm; mobile phase: [water(0.1% TFA )-ACN]) ; B%: 30-60%, 9 min) and lyophilized to give compound 37 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.47 (s, 1H), 9.30-9.29 (m, 1H), 8.27-8.23 (m, 2H), 7, 98 -7.93 (m, 3H), 7.81-7.75 (m, 2H), 7.50-7.52 (m, 1H), 7.34 (d, J=7.60 Hz, 1H), 4.25 (d, J=5.6 Hz, 2H), 3.01-2.94 (m, 6H), 2.66 (s, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=516,0.

Example 74. Preparation of 3-methylsulfonyl-N-[2-oxo-2-[[4-(3-phenylphenyl)thiazol-2-yl]amino]ethyl]benzamide (Compound 36)

Step 1: Preparation of tert-butyl N-[2-[(4-bromothiazol-2-yl)amino]-2-oxo-ethyl]carbamate (intermediate i-74a)

In a solution of 4-bromothiazol-2-amine (3.0 g, 16.76 mmol) and 2-( tert -butoxycarbonylamino)acetic acid (4.40 g, 25.13 mmol) in pyridine (30.0 mL) EDCI (16.06 g, 87 mmol). added). The mixture was stirred at 15°C for 16 hours. The reaction mixture was then concentrated under reduced pressure. The residue was diluted with 0.5N HCl (100.0 mL) and extracted with EtOAc (50 mL*3). The combined organic extracts were washed with NaHCO3(50.0 mL) and brine (50.0 mL) were dried over Na2AFTERWARD4filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO).2, petroleum ether/ethyl acetate = 7/1 to 3:1) to give intermediate i-74a as a white solid.

LCMS (ESI) m/z: [M+Na]+=358,1

Step 2: Preparation of 2-amino-N-(4-bromothiazol-2-yl)acetamide (intermediate i-74b)

A mixture of intermediate i-74a (300 mg, 892.31 μmol) in DCM (5.0 ml) and TFA (1.0 ml) was stirred at 30 °C for 2 h. The mixture was diluted with DCM (20.0 mL) and concentrated in vacuo.

This procedure was repeated three times. The residue was washed with MTBE (5 mL*2) to give intermediate i-74b (TFA salt) as a yellow oil, which was used directly in the next step.

LCMS (ESI) m/z: [M+Na]+=236,1

Step 3: Preparation of N-[2-[(4-bromothiazol-2-yl)amino]-2-oxo-ethyl]-3-methylsulfonylbenzamide (intermediate i-74c)

To a solution of 3-methylsulfonylbenzoic acid (171.55 mg, 856.87 μmol) in DMF (10.0 mL) was added HATU (977.42 mg, 2.57 mmol) and DIEA (553.72 mg, 4.28 mmol). The mixture was stirred at 30°C for 30 minutes. Intermediate i-74b (300.0 mg, 856.87 μmol) was then added and the mixture was stirred at 30 °C for a further 2 h. The mixture was poured into H2O (100.0 mL) and extracted with EtOAc (10.0 mL*3). The combined organic layers were washed with H2O (5.0 mL*3) and brine (5.0 mL*2), then dried over Na2AFTERWARD4filtered and concentrated under reduced pressure. The residue was purified by pre-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 32%-62%, 9 min) and lyophilized to give intermediate i-74c as a pale yellow solid.

LCMS (ESI) m/z: [81BrM+H]+=419,9.

Step 4: Preparation of 3-methylsulfonyl-N-[2-oxo-2-[[4-(3-phenylphenyl)thiazol-2-yl]amino]ethyl]benzamide (Compound 36)

For a solution of intermediate i-74c (60.0 mg, 143.44 µmol) and (3-phenylphenyl)boronic acid (85.22 mg, 430.33 µmol) in dioxane (3.0 mL) and H2O (0.15 ml) Pd(t-Bu) was added.3PI)2(36.65 mg, 71.72 μmol) and DIEA (92.69 mg, 717.21 μmol, 124.92 μL). The mixture was stirred at 100°C for 2 hours. The mixture was cooled to 30°C and a solid formed. The mixture was filtered to give the crude product as a yellow solid, which was dissolved in MeOH (1.0 mL) and DMSO (1.0 mL) and analyzed by pre-HPLC (column: Phenomenex Synergi C18 150*25*10 µm , mobile phase ): [water(0.1%TFA)-ACN]; B%: 45%-75%, 9 min) and lyophilized to give compound 36 as a white solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,49 (s, 1Η), 9,27-9,25 (m, 1Η), 8,46 (s, 1Η), 8,25-8, 21 (m, 2H), 8,13 (d, J= 7,80 Hz, 1H), 7,90 (d, J=7,80 Hz, 1H), 7,81-7,79 (m, 2H ), 7,73-7,71 (m, 2Η), 7,65-7,60 (m, 1Η), 7,54-7,49 (m, 3Η), 7,48-7,40 (m , 1Η), 4,25 (d, J=5,6 Hz, 2Η), 3,29 (s, 3H) ppm

LCMS (ESI) m/z: [M+H]+=492,0.

Example 75. Preparation of 4-(Methanesulfonamido)-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 38)

In a mixture of (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (50.0 mg, 118.64 μmol) and 4-(methanesulfonamido)benzoic acid (30, 64 mg , 142.36 ). μmol) in pyridine (1 mL) was added EDCI (68.23 mg, 355.91 μmol). The mixture was stirred at 30°C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Boston Green ODS 150*30 5m; mobile phase: [water (0.1% TFA)-ACN]; B%: 42%-72%, 9 min) and lyophilized, for to give compound 38 as a white solid.

1H-NMR (400 MHz, MeOD-d4) δ=7,91-7,88 (m, 4H), 7,40-7,29 (m, 6H), 4,92-4,88 (m, 1H), 3,03 (s, 3H) , 2,70-2,63 (m, 2Η), 2,30-2,20 (m, 2Η), 2,13 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=505,3.

Example 76. Preparation of 3-(dimethylsulfamoyl)-N-[2-[(4-isopropylthiazol-2-yl)amino]-2-oxoethyl]benzamide (Compound 35)

In a solution of 2-[[3-(dimethylsulfamoyl)benzoyl]amino]acetic acid (50.0 mg, 174.64 μmol), 4-isopropylthiazol-2-amine (24.84 mg, 174.64 μmol) in DCM ( 1.0 mL) DIEA (6. ). mg, 523.92 μmol, 91.26 μL), EDCI (50.22 mg, 261.96 μmol) and HOBt (35.40 mg, 261.96 μL) (μmol) were added. The mixture was stirred at 15°C for 16 hours. The reaction mixture was concentrated in vacuo to give a residue. The rest was preparation. HPLC (FA conditions) to give compound 35 as a white solid.

1HNMR (400 MHz, DMSO-d6) δ=12.26 (br s, 1H), 9.26–9.24 (m, 1H), 8.26–8.22 (m, 2H), 7.94 (d, J=8.0 Hz, 1H), 7.81 -7.78 (m, 1H), 6.75 (s, 1H), 4.17 (d, J=5.6 Hz, 2H) , 2.96-2.90 (m, 1H), 2.65 (s, 6H), 1.22 (d, J=6.8 Hz, 6H) ppm.

LCMS (ESI) m/z: [M+H]+=411,1.

Example 77. Preparation of N-[2-[(4-cyclohexylthiazol-2-yl)amino]-2-oxoethyl]-3-(dimethylsulfamoyl)benzamide (Compound 34)

In a solution of 2-[[3-(dimethylsulfamoyl)benzoyl]amino]acetic acid (50 mg, 174.64 μmol), 4-cyclohexylthiazol-2-amine (31.83 mg, 174.64 μmol) in DCM (2nd 0 .0 ml) DIEA (67.71). mg, 523.92 μmol, 91.26 μL), EDCI (50.22 mg, 261.96 μmol) and HOBt (35.40 mg, 261.96 μmol) were added. The mixture was stirred at 15°C for 16 hours. The reaction mixture was concentrated in vacuo to give a residue. The rest was preparation. HPLC (FA conditions) to give compound 34 as a yellow solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.23 (br s, 1H), 9.26-9.24 (m, 1H), 8.25-8.21 (m, 2H), 7 , 94 (d, J=8.0 Hz, 1H), 7.81-7.77 (m, 1H), 6.72 (s, 1H), 4.17 (d, J=5.6 Hz, 2H), 2.64 (s, 6H), 2.59-2.56 (m, 1H), 1.94-1.92 (m, 2H), 1.76-1.73 (m, 3H) , 1.39-1.31 (m, 4H), 1.18-1.15 (m, 1H) ppm.

LCMS (ESI) m/z: [M+H]+=451,0.

Example 78. Preparation of 3-[2-[[2-[[3-(dimethylsulfamoyl)benzoyl]amino]acetyl]amino]thiazol-4-yl]benzoic acid (Compound 33)

Step 1: Preparation of methyl 3-[2-[[2-[[3-(dimethylsulfamoyl)benzoyl]amino]acetyl]amino]thiazol-4-yl]benzoate (intermediate i-78a)

In a solution of N-[2-[(4-bromothiazol-2-yl)amino]-2-oxo-ethyl]-3-(dimethylsulfamoyl)benzamide (30.0 mg, 67.07 μmol) and acid (3o-Methoxycarbonylphenyl )boronic acid (36.21 mg, 201.21 μmol) in dioxane (2.0 mL) and H20 (0.1 ml) of Pd(t-Bu) was added.3PI)2(17.14 mg, 33.53 μmol) e DIEA (43.34 mg, 335.35 μmol, 58.41 μL) sob N2. The mixture was stirred at 100°C for 2 hours. A solid formed on cooling to 30°C. The solid was filtered and triturated in EtOAc (3 mL) and then dried in vacuo to give intermediate i-78a as a pale yellow solid which was used directly in the next step.

LCMS (ESI) m/z: [M+H]+=503,2.

Step 2: Preparation of 3-[2-[[2-[[3-(dimethylsulfamoyl)benzoyl]amino]acetyl]amino]thiazol-4-yl]benzoic acid (Compound 34)

For a solution of intermediate i-78a (15.0 mg, 29.85 μmol) in MeOH (2.0 mL) and H2O (1.0 mL) was treated with NaOH (1.19 mg, 29.85 μmol). The mixture was stirred at 30°C for 12 h, then diluted with MeOH (10.0 mL) and treated with HCl (1 M) to pH 5-6. The mixture was concentrated in vacuo to give a residue. The residue was purified by pre-HPLC (column: Phenomenex Synergi C18 150*25*10 µm; mobile phase: [water (0.1% TFA)-ACN]; B%: 28%-58%, 9 min) and lyophilized to give compound 34 as an off-white solid.

1H-NMR (400 MHz, DMSO-d6) δ 13,02 (br s, 1Η), 12,53 (s, 1Η), 9,31-9,30 (m, 1Η), 8,54 (s, 1Η), 8,27-8,24 (m, 2Η), 8,13 (d, J=8,0 Hz, 1Η), 7,94 (d, J=7,6 Hz, 1Η), 7, 90 (d, J=7,60 Hz, 1Η), 7,83-7,81 (m, 2Η), 7,77-7,57 (m, 1Η), 4,25 (d, J=5, 6 Hz, 2Η), 2,66 (s, 6Η) ppm.

LCMS (ESI) m/z: [M+H]+=488,9.

Example 79. Preparation of 3-(Hydroxymethyl)-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 84)

To a solution of 3-(hydroxymethyl)benzoic acid (14.85 mg, 97.58 μmol) and (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (30.00 mg, 97.58 µmol). μmol) in DCM (0.5 ml) were added at 25 °C with DIPEA (37.83 mg, 292.75 μmol) and HATU (55.66 mg, 146.37 μmol) and at 25 °C for 16 h touching . The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (FA condition) to give compound 84 as a white solid.

LCMS (ESI) m/z: [M+H]+=442,1.

1HNMR (400 MHz, METANOL-d4): 5=7,90–7,88 (m, 3H), 7,80 (d, J=8,0 Hz, 1H), 7,56 (d, J=7 ,6 Hz, 1Η), 7,48-7,46 (m, 1Η), 7,40-7,36 (m, 3Η), 7,29 (m, 1Η), 4,94-4,90 ( m, 1Η), 4,68 (m, 2Η), 2,71-2,64 (m, 2Η), 2,28-2,22 (m, 2Η), 2,13 (s, 3Η) ppm.

Example 80. Preparation of 4-(Hydroxymethyl)-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 85)

In a solution of 4-(hydroxymethyl)benzoic acid (14.85 mg, 97.58 μmol) and (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (30.00 mg , 97.58 μmol). μmol) in DCM (0.5 ml) were added at 25 °C with DIPEA (37.83 mg, 292.75 μmol) and HATU (55.66 mg, 146.37 μmol) and at 25 °C for 16 h touching . The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (FA condition) to give compound 85 as a white solid.

1HNMR (DMSO-d6, 400 MHz): 5=12,37 (br s, 1Η), 8,68 (d, J=7,2 Hz, 1Η), 7,92-7,89 (m, 4Η) , 7,63 (s, 1Η), 7,45- 7,42 (m, 4Η), 7,35-7,33 (m, 1Η), 5,32 (br s, 1Η), 4,79- 4,73 (m, 1Η), 4,57 (m, 2Η), 2,59-2,50 (m, 3Η), 2,15-2,10 (m, 5Η) ppm.

LCMS (ESI) m/z: [M+H]+=441,9.

Example 81. Preparation of 4-amino-N-[(1S)-1-methyl-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 82)

Step 1: Preparation of tert-butyl N-[(1S)-1-methyl-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]carbamate (intermediate i-81a)

In a mixture of 4-phenylthiazol-2-amine (200.0 mg, 1.13 mmol) and (2S)-2-(tert-butoxycarbonylamino)propanoic acid (214.72 mg, 1.13 mmol) in DCM (2nd 0.0 mL) DIPEA (44. mg, 3.40 mmol) and HATU (647.24 mg, 1.70 mmol) were added in one portion at 25°C under N.2. The mixture was stirred at 25°C for 2 hours. The mixture was poured into water (10.0 ml) and stirred for 5 min. The aqueous phase was extracted with DCM (10.0 ml*2). The combined organic extracts were washed with brine (10.0 mL) and dried over Na2AFTERWARD4filtered and concentrated in vacuo to give intermediate i-81a as a pale yellow oil, which was used in the next step without purification.

LCMS (ESI) m/z: [M+H]+=348,1.

Step 2: Preparation of (S)-2-amino-N-(4-phenylthiazol-2-yl)propanamide hydrochloride (intermediate i-81b)

To a mixture of intermediate i-81a (500 mg, 1.44 mmol) in dioxane (5.0 mL) was added HCl/dioxane (4M, 1.80 mL) in one portion at 25°C, 30 min at 25° C was stirred and a solid precipitate. The mixture was filtered and the filter cake was washed with EtOAc (10 mL). The solid was then dried in vacuo to give intermediate i-81b (HCl salt) as a white solid.

1H RMN (400 MHz, D2Ο) δ=7,84–7,79 (m, 1H), 7,49–7,43 (m, 1H), 4,39–4,35 (m, 1H), 2,80 (s, 1H ), 1,67 (d, J=6,8 Hz, 3Η).

HPLC-Lösung: OD-3_5CM_MeOH (DEA)_5_40_3 ML_T35.

LCMS (ESI) m/z: [M+H]+=248,1.

Step 3: Preparation of 4-amino-N-[(1S)-1-methyl-2-oxo-2-[(4-phenylthiazol-2-yl)amino]ethyl]benzamide (Compound 82)

To a mixture of intermediate i-81b (50 mg, 176.19 μmol) and 4-aminobenzoic acid (24.16 mg, 176.19 μmol) in DCM (1.0 mL) was added DIPEA (91.09 mg, 704 , 78 μmol) and HATU9 (1.0 mL) 264.29 μmol) in one dose at 25°C. The mixture was stirred at 25°C for 12 hours. The mixture was concentrated in vacuo to give a residue. The residue was purified via a reverse phase column (FA conditions) to give compound 82 as a white solid.

1H-NMR (400 MHz, METANOL-d4) δ=7,88 (d, J=7,2 Hz, 2H), 7,69 (d, J=8,6 Hz, 2H), 7,40-7 , 37 (m, 3Η), 7,31-7,27 (m, 1Η), 6,68 (d, J=8,6 Hz, 2Η), 4,77-4,72 (m, 1Η), 1,56 (d, J=7,2 Hz, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=367,0.

HPLC-Lösung: Amycoat_MeOH+CAN(DEA)_40_3 mL-35 T, 1.474 Min.

Example 82. Preparation of 4-(Methylamino)-N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]benzamide (Compound 81)

To a solution of 4-(methylamino)benzoic acid (10.76 mg, 71.18 μmol) in DCM (0.5 mL) was added DIPEA (27.60 mg, 213.55 μmol) and HATU (40.60 mg, 106 .77 μmol) at 25 °C. The mixture was stirred at 25°C for 1 hour. To the reaction mixture was added (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (30 mg, 71.18 µmol). The reaction mixture was stirred at 25°C for 16 hours. The mixture was then concentrated under reduced pressure to give a residue. The residue was purified by reverse phase HPLC (FA condition) followed by prep-TLC (PE/EA=1/1) to give compound 81 as a white solid.

1H-NMR (400 MHz, METANOL-d4) δ=7,89 (d, J=7,6 Hz, 2H), 7,74 (d, J=8,4 Hz, 2H), 7,40 (m , 3Η), 7,30 (m, 1Η), 6,61 (d, J=8,8 Hz, 2Η), 4,89 (m, 1Η), 2,81 (s, 3Η), 2,69 (m, 2Η), 2,26 (m, 2Η), 2,12 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H] + =441.0.

HPLC-Lösung: Cellucoat-MeOH+ACN(DEA)-40-5 Min.-3 ml-35T, 1.895 Min.

Example 83. Preparation of N-[(1S)-3-methylsulfanyl-1-[(4-phenylthiazol-2-yl)carbamoyl]propyl]indoline-5-carboxamide (Compound 80)

To a solution of indoline-5-carboxylic acid (11.61 mg, 71.18 µmol) in DCM (0.5 mL) was added DIPEA (27.60 mg, 213.55 µmol) and HATU (40.60 mg, 106 , 77 µmol) at 25°C. 1 h at 25°C and then (2S)-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (30.0 mg, 71, 18 µmol) was added. The reaction mixture was stirred at 25°C for 16 hours. The reaction mixture was concentrated in vacuo to give a residue. The residue was purified by reverse phase HPLC (FA condition) followed by preparative TLC (PE/EA=1/1) to give compound 80 as a white solid.

1HNMR (400 MHz, MeOD) δ=7.88–7.86 (m, 2H), 7.63–7.59 (m, 2H), 7.39–7.36 (m, 3H), 7, 28-7.26 (m, 1H), 6.58-6.52 (m, 1H), 4.89-4.86 (m, 1H), 3.59-3.54 (m, 2H), 3.04–3.00 (m, 2H), 2.68–2.63 (m, 2H), 2.20–2.12 (m, 2H), 2.11 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=453,0.

HPLC-Lösung: Cellucoat-MeOH+ACN(DEA)-40-5 Min.-3 ml-35T, 1.444 Min.

Example 84. Preparation of (S)-N-(4-(Methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)-1H-indole-5-carboxamide ( connection 83)

To a solution of 1H-indole-5-carboxylic acid (19.12 mg, 118.64 μmol) in DCM (1.0 mL) was added DIPEA (46.00 mg, 355.91 μmol), HATU (67.66 mg , 177.95 μmol) followed by (2Smol) )-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (36.47 mg, 118.64 μmol). The reaction mixture was stirred at 30°C for 2 hours. The reaction mixture was then poured into water (5.0 mL) and extracted with EtOAc (5.0 mL x 2). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The rest was preparation. HPLC (FA condition) to give compound 83 as a brown solid.

1H-NMR (400 MHz, DMSO-d6) δ=12.43 (br s, 1H), 11.34 (s, 1H), 8.56 (d, J=7.2 Hz, 1H), 8, 24 (s, 1H), 7.91-7.89 (m, 2H), 7.70-7.68 (m, 1H), 7.63 (s, 1H), 7.44-7.41 ( m, 4H), 7.32-7.31 (m, 1H), 6.56 (s, 1H), 4.79-4.74 (m, 1H), 2.68-2.65 (m, 1H), 2.59-2.55 (m, 1H), 2.14-2.12 (m, 2H), 2.10 (s, 3H) ppm.

LCMS (ESI) m/z: [M+H]+=451,3.

SFC-Quiral: OD-3_5 CM_MeOH (DEA)_40_3 ML_T35.M, 2.392 Min.

Example 85. Preparation of (S)-N-(4-(Methylthio)-1-oxo-1-((4-phenylthiazol-2-yl)amino)butan-2-yl)-1H-benzo[d]imidazole - 5-Carboxamide (Compound 66)

To a solution of 1H-benzimidazole-5-carboxylic acid (19.24 mg, 118.64 μmol) in DCM (1.0 mL) was added DIPEA (46.00 mg, 355.91 μmol), HATU (67.66 mg , 177.95 μmol) and then (20 mL). )-2-amino-4-methylsulfanyl-N-(4-phenylthiazol-2-yl)butanamide (36.47 mg, 118.64 μmol) was added. The reaction mixture was stirred at 30°C for 2 hours. The reaction mixture was poured into water (5.0 mL) and extracted with EtOAc (5.0 mL x 2). The combined organic extracts were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by preparative HPLC (FA condition) to give compound 66 as a yellow solid.

1H-NMR (400 MHz, DMSO-d6) δ=12,50 (br s, 1Η), 8,73 (d, J=7,2 Hz, 1Η), 8,34 (s, 1Η), 8, 25 (s, 1H), 7,91-7,81 (m, 2H), 7,80-7,79 (m, 1H), 7,62-7,61 (m, 2H), 7,44- 7,40 (m, 2Η), 7,32-7,31 (m, 1Η), 4,80-4,75 (m, 1Η), 2,68-2,65 (m, 1Η), 2, 59 -2,56 (m, 1Η), 2,17-2,14 (m, 2Η), 2,10 (s, 3Η) ppm.

LCMS (ESI) m/z: [M+H]+=452,3.

SFC solution: Cellucoat-MeOH (DEA)-40-3 mL-35 T, 0.942 min

Example 86. Synthesis of compound 87

Compound 87 was synthesized using the method described in the following scheme:

Preparation of (S)-4-amino-N-(4-(methylthio)-1-oxo-1-((4-(3-(pyridin-4-yl)phenyl)thiazol-2-yl)amino)butane - 2-yl)benzamide (Compound 87)

Step 1: Preparation of 2-bromo-1-(3-bromophenyl)ethenone (intermediate B)

In a solution of 1-(3-bromophenyl)ethanone (200 g, 1.00 mol, 132.45 mL) in CHCl3Added (250 mL) Br2(240.86 g, 1.51 mol, 77.70 mL) N sob2dropwise at 20°C. The mixture was stirred at 80°C for 1 hour. The reaction mixture was concentrated to give Intermediate B (279.27 g, crude) as a yellow oil, which was used directly in the next step.

Step 2: Preparation of 4-(3-bromophenyl)thiazol-2-amine (intermediate C)

To a solution of thiourea (229.23 g, 3.01 mol) in EtOH (1.5 L) was added intermediate B (279 g, 1.00 mol). The mixture was stirred at 85°C for 2 hours. The mixture was then concentrated in vacuo to give a residue. The residue was poured into NaHCO3(2 L) to adjust the pH to 8, then the solution was extracted with EA (2 L*3). The combined organic layers were washed with brine (4 L) and dried over Na2AFTERWARD4and filtered. The filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiO).2. PE:EA=5:1) gave Intermediate C (130 g, 494.40 mmol, 49.25% yield) as a yellow solid. LCMS (ESI) m/z: [M+H]+=257,0.1H-NMR (400 MHz, DMSO-d6) δ=7.98 (m, 1H), 7.79 (d, J=8.0 Hz, 1H), 7.44-7.42 (m, 1H) , 7.33-7.29 (m, 1H), 7.14 (s, 1H), 7.09 (s, 2H) ppm.

Step 3: Preparation of 4-[3-(4-pyridyl)phenyl]thiazol-2-amine (intermediate E)

4-(3-Bromophenyl)thiazol-2-amine (20 g, 78.40 mmol), 4-pyridylborohydride (28.9 g, 239.18 mmol), Di-tert-butyl(cyclopentyl)phosphane; Dichloropalladium; Eisen (2.56 g, 3.92 mmol) and K3AFTER4(66.56 g, 313.56 mmol) was taken up in dioxane (240 mL) and H2O (24 mL). The mixture was flushed with N 22stirred three times and then stirred at 80°C for 7 hours. Water (800 mL) was added and the mixture was extracted with EtOAc (800 mL*3). The combined organic layers were washed with brine and dried over Na2AFTERWARD4filtered and concentrated in vacuo. The residue was poured into DCM (30 ml) and MTBE (100 ml) and then stirred for 5 min. The precipitate was collected by filtration and washed with MTBE (10 mL) to give Intermediate E (16.2 g, 61.17 mmol, 78.03% yield) as a yellow solid.

LCMS (ESI) m/z: [M+H]+=254,0.

Step 4: Preparation of tert-butyl N-[(1S)-3-methylsulfanyl-1-[[4-[3-(4-pyridyl)phenyl]thiazol-2-yl]carbamoyl]propyl]carbamate (intermediate G)

To a mixture of intermediate E (12.6 g, 49.74 mmol) and (2S)-2-(tert-butoxycarbonylamino)-4-methylsulfanylbutanoic acid (18.6 g, 74.61 mmol) in DCM (900 mL) EEDQ (24.4 g, 9 mmol). ) was added and the mixture was stirred at 25°C for 2 h. The solvent was removed in vacuo. The residue was triturated with DCM (100 mL) followed by MeOH (200 mL) to give Intermediate G (11.7 g, 23.73 mmol, 47.71% yield, ee%=99.44%) as a white solid to be delivered. LCMS (ESI) m/z: [M+H]+=485,1.1H NMR (400MHz, DMSO) δ = 12.39 (s, 1H), 8.68-8.66 (m, 2H), 8.30 (s, 1H), 8.02-7.99 (m, 1H), 7.83 (s, 1H), 7.76-7.74 (m, 3H), 7.61-7.57 (m, 1H), 7.28 (d, J=7, 6 Hz , 1H), 4.31-4.30 (m, 1H), 2.65-2.44 (m, 2H), 2.06 ( s, 3H) 2.01-1.85 (m, 2H) , 1.38 (s, 9H) ppm. Note: The key point of this reaction is concentration. (1 mmol E with 17–18 mL DCM was the best situation)

Step 5: Preparation of (2S)-2-amino-4-methylsulfanyl-N-[4-[3-(4-pyridyl)phenyl]thiazol-2-yl]butanamide (intermediate H)

To a mixture of intermediate G (11.5 g, 23.73 mmol) in MeOH (50 mL) was added HCl/dioxane (4M, 100 mL) and the mixture was stirred at 25 °C for 1 h. The mixture was poured into MTBE (1000 mL) and the precipitate was collected by filtration to give Intermediate H (9.99 g, 23.73 mmol, 100.00% yield, HCl salt) as a yellow solid. LCMS (ESI) m/z: [M+H]+=385,0

Step 6: Preparation of 4-amino-N-[(1S)-3-methylsulfanyl-1-[[4-[3-(4-pyridyl)phenyl]thiazol-2-yl]carbamoyl]propyl]benzamide (intermediate E )

To a mixture of intermediate H (4 g, 9.50 mmol) and 4-aminobenzoic acid (1.30 g, 9.50 mmol) in DMF (40 mL) was added DIEA (4.91 g, 38.01 mmol, 6 0.62 mL), EDCI (2.73 g, 14.25 mmol) and HOBt (1.93 g, 14.25 mmol). The solution was stirred at 25°C for 14 h and then poured into water (200 ml) and the precipitate collected by filtration. The solid was triturated in MeOH (200 mL) (gelatinous solid formed). The precipitate was collected by filtration and the solid was further purified by column chromatography (SiO).2DCM:MeOH=80:1-20:1) to give compound 87 (2.13 g, 4.19 mmol, 44.11% yield, ee%=99.28%) as a white solid. LCMS (ESI) m/z: [M+H]+=504,0.1H-NMR (400 MHz, DMSO) δ=12.40 (s, 1H), 8.68-8.66 (m, 2H), 8.31-8.30 (m, 1H), 8.22 ( d, J=7.2 Hz, 1H), 8.02-7.99 (m, 1H), 7.82 (s, 1H), 7.76-7.74 (m, 3H), 7.67 - 7.63 (m, 2H), 7.61-7.57 (m, 1H), 6.58-6.54 (m, 2H), 5.67 (s, 2H), 4.72-4 , 67 (m, 1H), 2.65-2.54 (m, 2H), 2.12-2.06 (pol, 5H) ppm.

Example 87. Experimental data for compounds of the invention

LCMS m/z: # Compound name M + H HNMR 88 3-(methylsulfonyl)-N-(2-oxo-2-((4-(1- 483,1 1H-NMR (400 MHz, DMSO-d6) δ = 12,57 Oxo-1,2-dihydroisoquinoline- (br s, 1H), 11,25 (br s, 1H), 7-yl)thiazole-2- 9,29 (br s, 1Η), 8,78 (br s, 1Η), 8,46 (br s, il)amino)ethyl)benzamide 1H), 8.32-8.13 (m, 3H), 7,79-7,71 (m, 3Η), 7,19 (br s, 1Η), 6,56 (br s, 1H), 4,25 (br s, 2H), 3,29 (br s, 3Η) ppm. 89 3-(methylsulfonyl)-N-(2-oxo-2-((4-(3- 493,0 1H-NMR (400 MHz, DMSO-d6) δ = 12,50 (pyridin-4-yl)phenyl) (br s, 1H), 9,26 (br s, 1H), Thiazol-2-yl)amino)ethyl)benzamide 8,68 (br s, 2Η), 8,46 (br s, 1Η), 8,31 (br s, 1Η), 8,24 (br d, J = 8,4 Hz, 1 Η), 8,13 (br d, J = 6,8 Hz, 1H), 8,02 (br d, J = 7,6 Hz, 1 H), 7.85-7.76 (m, 5H), 7,62-7,58 (m, 1Η), 4,26 (br d, J = 4,8 Hz, 2Η), 3,29 (br s, 3Η) ppm. 90 3-(methylsulfonyl)-N-(2-oxo-2-((4-(3- 494,1 1H-NMR (400 MHz, DMSO-d6) δ = 12,59 (pyrimidin-4-yl)phenyl) (br s, 1Η), 9,29 (br s, 2Η), 8,91-8,82 (m, Thiazol-2-yl)amino)ethyl)benzamide 2Η), 8,45 (br s, 1Η), 8,25-8,09 (m, 5Η), 7,82 (br s, 2Η), 7,62 (br s, 1Η), 4,25 (br s, 2Η), 3,28 (br s, 3Η) ppm 91 3-(methylsulfonyl)-N-(2-oxo-2-((4-(3- 500,1 1H-NMR (400 MHz, DMSO-d6) δ = 12,46 (Tetrahydro-2H-pyran-4- (br s, 1Η), 9,27-9,24 (m, 1Η), 8,45 (s, 1Η), yl)phenyl)thiazole-2- 8.23 (d, J=8.0Hz, 1H), 8.12 (d, J=8.0Hz, il)amino)ethyl)benzamide 1Η), 7,82-7,78 (m, 2Η), 7,73 (d, J=8,0Hz, 1H), 7.65 (s, 1H), 7.38-7.34 (m, 1H), 7.22 (d, J=7.60Hz, 1H), 4.24 (d, J=6.0Hz, 2H), 3.98-3.95 (m, 2H), 3.48-3.42 (m, 2H), 3.28 (s, 3Η), 2,85-2,77 (m, 1Η), 1,74-1,65 (m, 4Η) ppm 92 N-(2-((4-(2,3- 474,3 1H-NMR (400 MHz, DMSO-d6) δ = 12,40 Dihydrobenzo[b][1,4]dioxin-6-yl)thiazol- (br s, 1Η), 9,25-9,23 (m, 1Η), 2-yl)amino)- 8,45-8,44 (m, 1Η), 8,23 (d, J = 8,0 Hz, 1Η), 2-Oxoetil)-3- 8.15-8.10 (m, 1H), 7.82-7.80 (m, 1H), (Methylsulfonyl)benzamide 7,47 (s, 1Η), 7,40-7,34 (m, 2Η), 6,89 (d, J = 8,2 Hz, 1H), 4,27 (s, 4H), 4,23 (d, J=5,8Hz, 2H), 3,28 (s, 3Η) ppm 93 N-(2-((4-(3-methyl-4-oxo-3,4- 498,0 1H-NMR (400 MHz, DMSO-d6) δ = 12,57 Dihydroquinazolin-6-yl)thiazol-2-yl) (s, 1Η), 9,27-9,25 (m, 1Η), amino)-2-oxoethyl)-3- 8,73 (d, J = 2,0 Hz, 1Η), 8,46-8,45 (m, 1Η), (Methylsulfonyl)benzamide 8,37 (s, 1Η), 8,34-7,32 (m, 1Η), 8.24 (d, J=7.6Hz, 1H), 8.13 (d, J=8.4Hz, 1H), 7.85 (s, 1H), 7.82-7.79 (m, 1H), 7.72 (d, J=8.4Hz, 1H), 4.26 (d, J=6.0Hz, 2H), 3,51 (s, 3Η), 3,28 (s, 3Η) ppm 94 N-(2-((4-(3- 523,3 1H-NMR (400 MHz, DMSO) δ = 12.31 (br s, (Methylsulfonamidomethyl)phenyl)thiazole- 1Η), 9,25 (br s, 1Η), 8,45 (s, 1Η), 8,24 (d, 2-il) J = 7,2 Hz, 1H), 8,12 (d, J = 7,2 Hz, 1H), 7,92 amino)-2-oxoethyl)-3- (s, 1H), 7,81 (d, J = 7,6 Hz, 2H), 7,62 (s, (Methylsulfonyl)benzamide 1H), 7.58 (br s, 1H), 7.41-7.40 (m, 1H), 7.30 (d, J=7.2Hz, 1H), 4.23 (d, J=5.2Hz, 2Η), 4,20 (d, J = 4,8 Hz, 2Η), 3,28 (s, 3Η), 2,89 (s, 3Η) ppm 95 N-(2-oxo-2-((4-(3-(pyridin-4- 456,2 1H-NMR (400 MHz, DMSO) δ = 12.39 (s, (yl)phenyl)thiazol-2-yl) 1Η), 8,88 (d, J=6,4Hz, 2Η), Amino)ethyl)indoline-5-carboxamide 8,48-8,47 (m, 1Η), 8,42 (s, 1Η), 8,18 (br d, J = 6,0Hz, 2H), 8.11 (d, J=8.0Hz, 1Η), 7.94-7.84 (m, 2Η), 7.68-7.67 (m, 1H), 7,63–7,53 (m, 2Η), 6,53 (d, J=8,0Hz, 1Η), 4.15-4.13 (m, 2H), 3,02-2,96 (m, 1Η), 2,98 (m, 2Η), 2,55 (br s, 2Η) ppm 96 N-(2-oxo-2-((4-(3-(pyridin-4- 455,2 1H-NMR (400 MHz, DMSO) δ = 13.37 (s, yl)phenyl)thiazol-2-yl)amino) 1H), 12.54 (s, 1H), 9.00 (s, 1H), ethyl)-1H-indazole-5-carboxamide 8,75-8,74 (m, 2Η), 8,47 (s, 1Η), 8,37 (s, 1H), 8.31 (s, 1H), 8.09-8.07 (m, 1H), 7.91-7.90 (m, 1H), 7.85 (s, 1H), 7,83-7,82 (m, 3Η), 7,69-7,66 (m, 2Η), 4,30- 4,28 (m, 2Η) ppm 97 (S)-N-(4-(methylthio)-1-oxo-1-((4-(3- 489,2 1H-NMR (400 MHz, DMSO) δ = 12.55 (s, (pyridin-4-yl)phenyl) 1H), 8.77 (d, J = 7.2 Hz, 1H), Thiazol-2-yl)amino)butan-2- 8,71-8,65 (m, 2Η), 8,32 (s, 1Η), 8,02 (d, J = il) Benzamide 8,0 Hz, 1H), 7,97–7,92 (m, 2H), 7,85 (s, 1H), 7,80–7,75 (m, 3Η), 7,63-7,55 (m, 2Η), 7.54-7.47 (m, 1H), 7.54-7.47 (m, 1H), 4.80- 4,75 (m, 1H), 2.60-2.57 (m, 2H), 2.18-2.12 (m, 2H), 2.11 (s, 3Η) ppm 98 3-(methylsulfonyl)-N-(2-oxo-2-((4-(3- 498,9 1H-NMR (400 MHz, DMSO) δ = 12.57 (s, (thiazol-2-yl)phenyl) 1 H), 9.28-9.25 (m, 1 H), Thiazol-2-yl)amino)ethyl)benzamide 8,55-8,54 (m, 1Η), 8,45 (d, J = 1,6 Hz, 1Η), 8,24 (d, J = 8,0 Hz, 1H), 8.12 (d, J=8.4Hz, 1H), 8.01 (d, J=8.0Hz, 1H), 7,96 (d, J = 3,2 Hz, 1Η), 7,92-7,90 (m, 1Η), 7.82-7.79 (m, 3H), 7,59-7,55 (m, 1Η), 4,25 (d, J=6,0Hz, 2Η), 3,28 (s, 3Η) ppm 99 N-(2-oxo-2-((4-(3-(pyridin-4- 470,3 1H-NMR (400 MHz, DMSO) δ = 12.36 (br s, yl)phenyl)thiazol-2-yl)amino)ethyl)- 1H), 8.73-8.64 (m, 2H), 8.38 (s, 1H), 8.32- 1,2,3,4-Tetrahidroquinolina-6- 8,31 (m, 1H), 8,01 (d, J = 7,8 Hz, 1H), 7,82 carboxylic acid amide (s, 1Η), 7,79-7,73 (m, 3Η), 7,60 (m, 1Η), 7,49–7,40 (m, 2H), 6,44 (d, J=9,0Hz, 1H), 6,29 (s, 1H), 4,12 (d, J = 5,8 Hz, 2Η), 3,23 (br s, 2Η), 2,71–2,70 (m, 2H), 1,89–1,75 (m, 2H) ppm 100 4-(aminomethyl)-N-(2-oxo-2-((4-(3- 444 1H-NMR (400 MHz, DETIUM OXIDE) (pyridin-4-yl)phenyl) δ = 8,61 (d, J = 6,8Hz, 2H), Thiazol-2-yl)amino)ethyl)benzamide 8.08 (d, J=6.8Hz, 2H), 7.85 (d, J=8.4Hz, 2H), 7.81 (s, 1H), 7,71–7,69 (m, 1H), 7,59–7,57 (m, 1H), 7,56 (d, J = 8.4Hz, 2H), 7,43-7,41 (m, 1Η), 7,27 (s, 1Η), 4,30 (s, 2Η), 4,24 (s, 2Η) ppm 101 N-(2-oxo-2-((4-(3-(pyridin-4- 455 1H-NMR (400 MHz, DMSO) δ = 12.24 (br s, yl)phenyl)thiazol-2-yl)amino) 1H), 9.03-8.95 (m, 2H), Ethyl)imidazo[1,5-a]pyridine-6- 8,71-8,65 (m, 2Η), 8,55 (s, 1Η), 8,31 (s, carboxylic acid amide 1H), 8.02 (d, J = 7.8 Hz, 1H), 7,84 (s, 1Η), 7,79-7,73 (m, 3H), 7,66-7,56 (m, 2H), 7,43 (s, 1H), 7,22-7,21 (m, 1Η), 4,23 (d, J=5,8Hz, 2H) ppm 102 N-(2-oxo-2-((4-(3-(pyridin-4- 471.1 1H-NMR (400 MHz, DMSO) δ = 8.63 (d, yl)phenyl)thiazol-2-yl)amino)ethyl)- J=5.8Hz, 2H), 8.28–8.25 (m, 2H), 5,6,7,8-Tetrahydro-1,8-naphthyridine-3- 7,98 (d, J = 7,8 Hz, 1Η), 7,75-7,72 (m, 4Η), carboxylic acid amide 7.60-7.58 (m, 2H), 4.11 (s, 2H), 3.30-3.27 (m, 2H), 2.68-2.67 (m, 2Η), 1,77-1,74 (m, 2Η) ppm 103 4-(3-Hydroxyazetidin-1-yl)-N-(2-oxo-2- 486,1 1H-NMR (400 MHz, DMSO) δ = 12.36 (br s, ((4-(3-(pyridin-4-yl)\ 1Η), 8,68 (d, J=4,8Hz, 2Η), phenyl)thiazole-2- 8,53 (s largo, 1Η), 8,31 (s, 1H), 8,02 (d, J = 7,2 il)amino)ethyl)benzamide Hz, 1Η), 7,80-7,74 (m, 6Η), 7,64-7,56 (m, 1Η), 6,45 (d, J=8,0Hz, 2Η), 5,69 (d, J = 6,4 Hz, 1Η), 4,60 (d, J = 6,0 Hz, 1Η), 4,18-4,10 (m, 4Η), 3,62-3,59 (m, 2Η) ppm 104 4-Methylsulfonyl-N-[2-oxo-2-[[4-[3-(4- 494,1 1H-NMR (400 MHz, DMSO) δ = 12.51 (s, Pyridyl)phenyl]thiazole-2- 1Η), 9,32-9,29 (m, 1Η), 9,06 (d, J = 4,8 Hz, yl]amino]ethyl]pyridine-2-carboxamide 1H), 8.80 (d, J = 5.6 Hz, 2H), 8.44 (s, 1H), 8,37 (s, 1Η), 8,17-8,16 (m, 1Η), 8,07-8,01 (m, 3Η), 7,86-7,83 (m, 2Η), 7,66-7,62 (m, 1Η), 4,31 (d, J = 6,0 Hz, 2Η), 3,41 (s, 3Η) ppm 105 3-Hydroxy-N-[2-oxo-2-[[4-[3-(4- 486,3 1H-NMR (400 MHz, DMSO) δ = 8.87 (d, Pyridyl)phenyl]thiazol-2-yl]amino]ethyl]- J=4,8Hz, 2H), 8,46-8,35 (m, 2H), 8,21- 1,2,3,4-Tetrahidroquinolina-6- 8,06 (m, 3Η), 7,92-7,83 (m, 2Η), 7,67-6,64 carboxylic acid amide (m, 1H), 7,51-7,43 (m, 2H), 6,45 (d, J = 9,0 Hz, 1H), 4.13 (d, J = 5.7 Hz, 2H), 4.01-3.92 (m, 1Η), 3,33-3,25 (m, 1Η), 2,99-2,97 (m, 1Η), 2,87 (d, J = 13,0 Hz, 1 Η), 2,69-2,65 (m, 1Η), 2,61 (d, J=7,6Hz, 2Η), 2,44-2,43 (m, 1Η) ppm 106 4-Amino-N-(2-oxo-2-((4-phenylthiazole- 353.1 1H-NMR (400 MHz, DMSO-d6) δ = 2-yl)amino)ethyl)benzamide 12,32 (br s, 1Η), 8,42-8,39 (m, 1Η), 7,90 (d, J = 7,6 Hz, 2Η), 7,62-7,60 (m, 3Η), 7.44-7.41 (m, 2H), 7.34-7.30 (m, 1H), 6.57 (d, J=7.6Hz, 2H), 5.65 (s, 2H), 4,12 (d, J = 5,2 Hz, 2Η) ppm. 107 (S)-4-Amino-N-(1-((4-(4- 452.1 1H-NMR (400 MHz, DMSO-d6) δ = 12,44 Cyanophenyl)thiazol-2-yl)amino)- (s, 1H), 8,23 (d, J = 7,2 Hz, 1H), 4-(Methylthio)-1-oxobutane-2- 8,08 (d, J = 8,4 Hz, 2Η), 7,92-7,88 (m, 3Η), il) Benzamide 7,65 (d, J=8,0Hz, 2Η), 6,58 (d, J=8.4Hz, 2H), 4.72-4.67 (m, 1H), 2.64- 2,60 (m, 2Η), 2,08 (s, 5Η) ppm 108 4-Amino-N-(2-((4-(4- 383,1 1HNMR (400 MHz, DMSO-d6) δ = 12,28 (br Methoxyphenyl)thiazol-2-yl)amino)- s, 1 H), 8.44-8.43 (m, 1 H), 2-oxoethyl)benzamide 7,84-7,82 (m, J = 8,8 Hz, 2Η), 7,62-7,60 (m, 2H), 7.44 (s, 1H), 7.00–6.98 (m, 2H), 6.57–6.55 (m, 2H), 5.66 (s, 2H), 4,11 (d, J = 5,6 Hz, 2H), 3,79 (s, 3H) ppm 109 4-Amino-N-(2-((4-(2,3- 411.3 1H-NMR (400 MHz, DMSO-d6) δ = 12,27 Dihydrobenzo[b][1,4]dioxin-6-yl) (br s, 1 Η), 8,40-8,38 (m, 1 Η), Thiazol-2-yl)amino)-2- 7.61 (d, J=8.8Hz, 2H), 7.45 (s, 1H), 7.41- Oxoethyl)benzamide 7,34 (m, 2Η), 6,89 (d, J=8,0Hz, 1Η), 6,56 (d, J=8,8Hz, 2H), 5,65 (s, 2H), 4,26 (s, 4Η), 4,10 (d, J = 6,0 Hz, 2Η) ppm 110 Methyl-3-(2-(2-(4-) 416.1 1H-NMR (400 MHz, MeOD) δ = 7,67 (d, Aminobenzamido)acetamido)thiazole-4- J = 8,6 Hz, 2Η), 6,92 (s, 1Η), il)- 6.69 (d, J=8.6Hz, 2H), 6.66 (s, 1H), 4.30 5,6-Dihydropyridine-1(2H)-carboxylate (s, 2H), 4,22 (s, 2H), 3,73 (s, 3H), 3,60–3,58 (m, 2H), 2,32–2,31 (m, 2H) ppm 111 4-Amino-N-(2-((4-(2,3- 395,3 1H-NMR (400 MHz, DMSO-d6) δ = 12,24 (s, Dihydrobenzofuran-5-yl)thiazol-2-yl) 1H), 8.40 (s, 1H), 7.75 (s, 1H), Amino)-2-oxoethyl)benzamide 7,69-7,56 (m, 3Η), 7,38 (s, 1Η), 6,80 (d, J=8.4Hz, 1H), 6.56 (d, J=8.8Hz, 2H), 5,65 (s, 2Η), 4,56 (m, 2Η), 4,10 (d, J = 5,6 Hz, 2H), 3.26-3.16 (m, 2H) ppm 112 4-Amino-N-(2-((4-(2,3- 395,2 1H-NMR (400 MHz, DMSO-d6) δ = 12,47- Dihydrobenzofuran-6-yl)thiazol-2-yl) 11,92 (m, 1Η), 8,30 (br s, 1Η), Amino)-2-oxoethyl)benzamide 7,60 (d, J = 8,4 Hz, 2Η), 7,46 (br s, 1Η), 7,38 (s, 1H), 7,27 (s, 1H), 7.24 (d, J=7.6Hz, 1H), 6.56 (d, J=8.4Hz, 2H), 5.64 (s, 2H), 4.55 (m, 2H), 4,06 (d, J = 5,6 Hz, 2H), 3,18 (m, 2H) ppm 113 4-amino-N-(2-((4-(3-(5-methyl-1,3,4- 435,1 1H-NMR (400 MHz, DMSO-d6) δ = 12,47 Oxadiazol-2-yl)phenyl) (s, 1Η), 8,54 (s, 1Η), 8,46-8,43 (m, 1Η), Thiazol-2-yl)amino)-2- 8,13 (br d, J = 8,0 Hz, 1H), 7,93 (d, J = 8,0 Oxoethyl)benzamide Hz, 1 H), 7.84-7.83 (m, 1 H), 7,67-7,60 (m, 3Η), 6,56 (d, J = 8,8 Hz, 2Η), 5.68(s, 2H), 4,12 (d, J = 5,6 Hz, 2H), 2,60 (s, 3H) ppm 114 4-Amino-N-(2-((4-(2- 369,3 1H-NMR (400 MHz, DMSO-d6) δ = 12,40 Hydroxyphenyl)thiazol-2-yl)amino)- (s, 1H), 10,90 (s, 1H), 8,47 (s, 1H), 2-oxoethyl)benzamide 7,92-7,91 (m, 1Η), 7,72 (s, 1Η), 7,62 (d, J = 8,6 Hz, 2Η), 7,22-7,13 (m, 1Η), 6,96–6,85 (m, 2H), 6,57 (d, J=8,6Hz, 2H), 5,68 (br s, 2H), 4,13 (d, J = 5,8 Hz, 2Η) ppm 115 4-Amino-N-(2-((4-(4- 369,1 1HNMR (400 MHz, DMSO-d6) δ = 12,22 Hydroxyphenyl)thiazol-2-yl) (br s, 1Η), 9,81-9,42 (m, 1Η), 8,41 (br s, Amino)-2-oxoethyl)benzamide 1H), 7.78-7.51 (m, 4H), 7.34 (s, 1H), 6,81 (br d, J = 8,8 Hz, 2H), 6,57 (br d, J = 8,4 Hz, 2Η), 5,66 (br s, 2Η), 4,11 (br d, J = 5,2 Hz, 2Η) ppm 116 (S)-4-Amino-N-(1-((4-(5-Cyano-[1,1'-- 528,3 1H-NMR (400 MHz, CDCl3) δ = 11,10 (br s, Diphenyl]-3-yl)thiazol-2-yl)amino)- 1H), 8.29 (s, 1H), 8.14 (s, 1H), 4-(Methylthio)-1-oxobutane-2- 7,83-7,80 (m, 3Η), 7,66-7,64 (m, 2Η), 7,53- il) Benzamide 7.50 (m, 2H), 7.47-7.43 (m, 1H), 7,31 (s, 1H), 7,12 (d, J = 8,0 Hz, 1H), 6,72 (d, J=8.0Hz, 2H), 5.31-5.26 (m, 1H), 2.81-2.69 (m, 2H), 2.44- 2,39 (m, 1H), 2,35–2,28 (m, 1H), 2,18 (s, 3H) ppm 117 4-Amino-N-(2-oxo-2-((4-(3-(pyridin-4- 430,0 1H-NMR (400 MHz, MeOD) δ = 8,89 (d, (yl)phenyl)thiazol-2-yl) J=6,8Hz, 2H), 8,51(s, 1H), 8,43(d, J=6,8 amino) ethyl) benzamide Hz, 2H), 8,21 (d, J = 7,6 Hz, 1H), 7,92 (br d, J=8.0Hz, 1H), 7.75 (d, J=8.8Hz, 2H), 7,71-7,67 (m, 1Η), 7,64 (s, 1Η), 6,81 (d, J = 8,8 Hz, 2Η), 4,30 (s, 2Η) ppm 118 (S)-4-Amino-N-(1-((4-(4'-- 532,4 1H-NMR (400 MHz, MeOD) δ = 8,19 (s, (Aminomethyl)-[1,1'-diphenyl]-3- 1H), 7.91 (d, J = 7.6 Hz, 1H), il)thiazol-2-yl) 7,79-7,75 (m, 4H), 7,59-7,54 (m, 3H), 7,51- Amino)-4-(methylthio)-1-oxobutane-2- 7,48 (m, 2Η), 6,84 (br d, J = 8,4 Hz, 2Η), il) Benzamide 4,89 (br s, 1Η), 4,17 (s, 2Η), 2,73-2,59 (m, 2Η), 2,32-2,17 (m, 2Η), 2,13 (br s, 3Η) ppm 119 (S)-4-Amino-N-(1-((4-(6-cyano-[1,1'-) 528,2 1H-NMR (400 MHz, DMSO-d6) δ = 12,43 Diphenyl]-3-yl)thiazol-2-yl) (s, 1H), 8,19 (s, 1H), 8,14 (s, 1H), Amino)-4-(methylthio)-1-oxobutane-2- 8.09 (d, J=8.4Hz, 1H), 8.04 (d, J=8.0Hz, il) Benzamide 2Η), 7,64 (d, J=7,2Hz, 4Η), 7,58-7,51 (m, 3Η), 6,55 (d, J = 8,8 Hz, 2Η), 5,66 (s, 2H), 4,66 (s, 1H), 2,64-2,61 (m, 2H), 2,07 (m, 5H) ppm 120 (S)-N-(1-((4-(3- 498,1 1H-NMR (400 MHz, DMSO-d6) δ = 12,31 (acetamidomethyl)phenyl)thiazole-2- (s, 1Η), 8,38-8,34 (m, 1Η), il)amino)-4- 8,21 (br d, J = 7,2 Hz, 1Η), 7,79 (s, 1Η), (metiltio)-1-oxobutan-2-il)-4- 7.76 (d, J=7.6Hz, 1H), 7.64 (d, J=8.8Hz, Aminobenzamide 2H), 7.59 (s, 1H), 7.39-7.35 (m, 1H), 7.20 (d, J=7.2Hz, 1H), 6.55 (d, J=8.4Hz, 2H), 5,67 (s, 2Η), 4,67 (br d, J = 7,2 Hz, 1Η), 4.28 (d, J=6.0Hz, 2Η), 2.64-2.61 (m, 2Η), 2,08-2,05 (m, 5H), 1,88 (s, 3H) ppm 121 (S)-N-(1-((4-(4- 498,1 1H-NMR (400 MHz, DMSO) δ = 12.36 (s, (acetamidomethyl)phenyl)thiazole-2- 1Η), 8,35-8,32 (m, 1Η), 8,21 (d, J = 8,0 Hz, the)amino)- 1Η), 7,83 (d, J=8,4Hz, 2Η), 7,66-7,63 (m, 4-(metiltio)-1-oxobutan-2-il)-4- 2Η), 7,57 (s, 1Η), 7,29 (d, J = 8,4 Hz, 2Η), Aminobenzamide 6.57 (d, J=8.4Hz, 2Η), 4.70-4.65 (m, 1Η), 4.26 (d, J=5.6Hz, 2Η), 2.64-2.50 (m, 2Η), 2,08–2,04 (m, 5H), 1,87 (s, 3H) ppm 122 (S)-Methyl-3-(2-(2-(4-) 418.1 1H-NMR (400 MHz, MeOD) δ = 7,67–7,65 Aminobenzamide)acetamide) (m, 2H), 6.76 (s, 1H), 6.69-6.67 (m, 2H), Tiazol-4-il)piperidina-1-carboxilato 4,25 (br d, J = 14,0 Hz, 1Η), 4,21 (s, 2Η), 4,03 (br d, J = 10,4 Hz, 1Η), 3,68 (s, 3Η), 2,98–2,92 (m, 2H), 2,78–2,76 (m, 1H), 2,10–2,07 (m, 1H), 1,77–1,74 (m, 2H), 1,71-1,56 (m, 1 Η) ppm 123 N-(2-((4-(4'-(acetamidomethyl)-[1,1'- 500,1 1H-NMR (400 MHz, DMSO) δ = 12.36 (s, Diphenyl]-3-yl)thiazol-2-yl) 1H), 8.42-8.35 (m, 2H), 8.18 (s, 1H), 7.89 Amino)-2-oxoethyl)-4-aminobenzamide (d, J = 7,6 Hz, 1Η), 7,75 (s, 1Η), 7,67 (d, J = 8,0 Hz, 2H), 7,62-7,59 (m, 3H), 7,53- 7,49 (m, 1Η), 7,38 (d, J=8,0Hz, 2Η), 6,57 (d, J=8.8Hz, 2H), 5.66 (s, 2H), 4.31 (d, J=6.0Hz, 2H), 4.12 (d, J=6.0Hz, 2H), 1.90 (s, 3Η) ppm 124 N-(2-((4-(3'-(acetamidomethyl)-[1,1'- 500,3 1H-NMR (400 MHz, MeOD) δ = 8,20-8,19 Diphenyl]-3-yl)thiazol-2-yl) (m, 1Η), 7,90 (d, J = 7,6 Hz, 1Η), Amino)-2-oxoethyl)-4-aminobenzamide 7,86-7,77 (m, 2Η), 7,66-7,56 (m, 3Η), 7,53- 7,43 (m, 3H), 7,32 (d, J = 8,0 Hz, 1H), 6,97 (d, J=8.4Hz, 2H), 4.46 (s, 2H), 4.31 (s, 2Η), 2,03 (s, 3Η) ppm 125 (S)-4-amino-N-(1-oxo-1-((4-(3-(pyridine- 444.1 H-NMR (400 MHz, DMSO-d6) δ = 12,33 (br 4-yl)phenyl) s, 1H), 8.68-8.66 (m, 2H), 8.30 (s, 1H), Thiazol-2-yl)amino)propan-2- 8,20 (br d, J = 6,4 Hz, 1H), 8,01 (br d, J = 8,0 il) Benzamide Hz, 1Η), 7,82 (s, 1Η), 7,76-7,74 (m, 3Η), 7.65 (d, J=8.8Hz, 2Η), 7.61-7.57 (m, 1Η), 6.56 (d, J=8.4Hz, 2H), 5.65 (s, 2H), 4.67- 4,61 (m, 1H), 1,43 (d, J = 7,2 Hz, 3H) ppm 126 4-(methylamino)-N-(2-oxo-2-((4-(3- 444,3 1H-NMR (400 MHz, DMSO-d6) δ = 12,24- (pyridin-4-yl)phenyl) 12.23 (m, 1Η), 8.67 (d, J=6.4Hz, 2Η), Thiazol-2-yl)amino)ethyl)benzamide 8,47-8,44 (m, 1Η), 8,30 (s, 1Η), 8,01 (br d, J = 7,6 Hz, 1Η), 7,82 (s, 1Η), 7,76 (br d, J=6.0Hz, 3H), 7.69 (d, J=8.8Hz, 2H), 7.61-7.57 (m, 1H), 6,55 (d, J=8,8Hz, 2Η), 6,25-6,22 (m, 1Η), 4,13 (br d, J = 6,0 Hz, 2H), 2,72 (d, J = 4,8 Hz, 3Η) ppm 127 4-Amino-N-(2-((4-(3-(6-Methoxypyridine- 460,0 1H-NMR (400 MHz, DMSO) δ = 12.34 (br s, 3-yl)phenyl) 1H), 8.54 (d, J = 2.4 Hz, 1H), Thiazol-2-yl)amino)-2- 8,41 (br s, 1Η), 8,16 (s, 1Η), 8,07-8,05 (m, Oxoethyl)benzamide 1H), 7.90 (d, J = 8.0 Hz, 1H), 7,77 (s, 1H), 7,61 (d, J = 8,4 Hz, 3H), 7,54- 7,50 (m, 1H), 6.95 (d, J=8.4Hz, 1H), 6.56 (d, J=8.4Hz, 2H), 5.65 (s, 2H), 4,12 (br d, J = 5,6 Hz, 2H), 3,91 (s, 3H) ppm 128 (S)-N-(1-((4-(3-(1,3,4-oxadiazole-2- 495,3 1H-NMR (400 MHz, DMSO-d6) δ = 12,46 yl)phenyl)thiazol-2-yl)amino)- (s, 1H), 9,37 (s, 1H), 8,61 (s, 1H), 4-(metiltio)-1-oxobutan-2-il)-4- 8.21 (d, J=6.8Hz, 1H), 8.15 (d, J=8.4Hz, Aminobenzamide 1H), 7.98 (d, J = 7.6 Hz, 1H), 7,84 (s, 1Η), 7,69-7,64 (m, 3H), 6,56 (d, J = 8.8 Hz, 2H), 5.66 (s, 2H), 4,71–4,67 (m, 1H), 2,64–2,54 (m, 2H), 2,12-2,09 (m, 5Η) ppm 129 4-amino-N-(2-((4-(3'-((N- 514,3 1H-NMR (400 MHz, DMSO) δ = 12.37(br s, Methylacetamido)methyl)-[1,1'- 1 Η), 8,43 (br s, 1 Η), 8,17 (br s, 1 Η), 7,91 Biphenyl]- (br d, J = 7,2 Hz, 1H), 7,76 (s, 1H), 7,65- 3-yl)thiazol-2-yl)amino)-2- 7,32 (m, 7Η), 7,28-7,19 (m, 1Η), 6,57 (d, Oxoethyl)benzamide J = 8,4 Hz, 2Η), 5,67 (s,2H), 4,69-4,54 (m, 2H), 4.13 (m, 2H), 3.02-2.82 (m, 3H), 2.12- 2,03 (m, 3Η) ppm 130 3'-(2-(2-(4- 486,1 1H-NMR (400 MHz, DMSO) δ = 12.37 (s, Aminobenzamido)acetamido)thiazole-4- 1H), 8.50-8.49 (m, 1H), il)- 8,44–8,41 (m, 1H), 8,25 (s, 1H), 8,00–7,92 N-Methyl-[1,1'-diphenyl]-4- (m, 3Η), 7,85-7,79 (m, 3Η), carboxylic acid amide 7.69 (d, J=8.2Hz, 1H), 7.62 (d, J=8.6 Hz, 2Η), 7,59-7,53 (m, 1Η), 6.57 (d, J=8.6Hz, 2H), 5.67 (s, 2H), 4.13 (d, J = 5,6 Hz, 1H), 4,16-4,11 (m, 1Η), 2,82 (d, J = 4,6 Hz, 3Η) ppm 131 3'-(2-(2-(4- 500,3 1H-NMR (400 MHz, DMSO) δ = 12.38 (s, Aminobenzamido)acetamido)thiazole-4- 1H), 8.44-8.41 (m, 1H), 8.24 (s, 1H), 7.94 il)- (d, J=7,8Hz, 1H), 7,83–7,78 (m, 2H), 7,72 N,N-dimethyl-[1,1'-diphenyl]-3- (s, 1H), 7,67 (d, J = 7,8 Hz, 1H), 7,62 (d, J = carboxylic acid amide 8,6Hz, 2H), 7,59-7,53 (m, 2H), 7,42 (d, J= 7,6 Hz, 1Η), 6,57 (d, J = 8,6 Hz, 2Η), 5,67 (br s, 2Η), 4,13 (d, J = 5,6 Hz, 2Η), 3,08-2,91 (m, 6Η) ppm 132 4-(Dimethylamino)-N-(2-oxo-2-((4-(3- 458,1 1H-NMR (400 MHz, DMSO-d6) δ = 12,40 (pyridin-4-yl)phenyl) (s, 1Η), 8.82 (d, J=6.4Hz, 2H), Thiazol-2-yl)amino)ethyl)benzamide 8,56-8,53 (m, 1Η), 8,38 (s, 1Η), 8,07 (br d, J = 6.4 Hz, 3H), 7,85 (s, 2H), 7,76 (d, J=8,8Hz, 2H), 7,66- 7,63 (m, 1H), 6,73 (d, J=8,8Hz, 2H), 4,15 (d, J = 5,6 Hz, 2H), 2,98 (s, 6H) ppm 133 4-Amino-N-(2-((4-(3-(2-Aminopyridine-4- 445 1H-NMR (400 MHz, DMSO) δ = 12.38 (br s, phenyl) 1H), 8.44-8.43 (m, 1H), Thiazol-2-yl)amino)-2- 8.23-8.13 (m, 2H), 8.04-7.92 (m, 2H), 7.76 Oxoethyl)benzamide (s, 1Η), 7,68-7,51 (m, 4Η), 6,82 (m, 1Η), 6,75 (s, 1H), 6,57 (d, J = 8,8 Hz, 2H), 6.00 (s, 2H), 5,67 (br s, 1H), 4,13 (d, J = 5,6 Hz, 2H) ppm 134 4-amino-N-(2-((4-(4'-(methylsulfonyl)- 507,2 1H-NMR (400 MHz, DMSO) δ = 8.43 (s, [1,1'-Diphenyl]-3-yl) 1H), 8.27 (s, 1H), 8.08-7.96 (m, 5H), 7.81 Thiazol-2-yl)amino)-2- (s, 1H), 7,72 (br d, J = 7,6 Hz, 1H), 7,67- Oxoethyl)benzamide 7,54 (m, 3H), 6,57 (d, J = 8,6 Hz, 2H), 5,71- 5,63 (m, 1Η), 5,67 (s, 1Η), 4,13 (br d, J = 5,6 Hz, 2Η), 3,28 (s, 3Η) ppm 135 4-Amino-N-(2-((4-(3'-(aminomethyl)- 458,3 1H-NMR (400 MHz, DMSO) δ = 12.35 (s, [1,1'-Diphenyl]-3-yl) 1H), 8.47-8.46 (m, 1H), 8.21 (s, 4H), 7.94 Thiazol-2-yl)amino)-2- (d, J = 7,6 Hz, 1H), 7,86 (s,1H), 7,79-7,73 Oxoethyl)benzamide (m, 2H), 7.42-7.69 (m, 7H), 6.59 (d, J=8.8 Hz, 2H), 4.18-4.11 (m, 4H) ppm 136 3'-(2-(2-(4- 473.1 1H-NMR (400 MHz, DMSO) δ = 13.12 (br Aminobenzamido)acetamido)thiazole-4- s, 1 H), 12,40 (s, 1 H), il)- 8,43 (br s, 1 H), 8,25 (br s, 2 H) 8,04-7,92 [1,1'-Diphenyl]-3-carboxylic acid (m, 3 H), 7,80 (s, 1 H), 7,72-7,53 (m, 5 H), 6,57 (br d, J = 8,2 Hz, 2 Η), 5,67 (br s, 2 Η), 4,20-4,08 (m, 1 Η), 4,13 (br d, J = 5,2 Hz, 1 Η) ppm 137 3'-(2-(2-(4- 500,1 1H-NMR (400 MHz, DMSO) δ = 12.37 (s, Aminobenzamido)acetamido)thiazole-4- 1H), 8.44-8.41 (m, 1H), 8.24 (s, 1H), 7.96- il)- 7,92 (m, 1Η), 7,79 (d, J = 6,6 Hz, 3H), 7,67 N,N-Dimethyl-[1,1'-diphenyl]-4- (d, J = 8,2 Hz, 1H), carboxylic acid amide 7.62 (d, J=8.6Hz, 2H), 7.58 (s, 1H), 7.59- 7,52 (m, 2Η), 6,57 (d, J=8,6Hz, 2H), 5,67 (s, 2H), 4,13 (d, J = 5,8 Hz, 2H), 2,99 (br s, 6Η) ppm 138 4-amino-N-(2-((4-(4'-((N- 514.1 1H-NMR (400 MHz, DMSO) δ = 12.38 (s, Methylacetamido)methyl)-[1,1'- 1H), 8.47-8.44 (m, 1H), Biphenyl]- 8.19-8.17 (m, 1H), 7.90-7.88 (m, 1H), 7.77 3-yl)thiazol-2-yl)amino)-2- (d, J = 2,4 Hz, 1H), Oxoethyl)benzamide 7,74-7,66 (m, 2Η), 7,63-7,59 (m, 3Η), 7,54- 7,49 (m, 1H), 7,35-7,32 (m, 2Η), 6,60 (d, J=8,4Hz, 2Η), 4.61-4.54 (m, 2H), 4,13 (d, J = 5,6 Hz, 2Η), 2,94-2,83 (m, 3Η), 2,08-2,07 (m, 3Η) ppm 139 4-amino-N-(2-((4-(4'-hydroxy-[1,1'- 445,2 1H-NMR (400 MHz, DMSO) δ = 12.35 (s, Diphenyl]-3-yl)thiazole- 1H), 9.57 (s, 1H), 2-yl)amino)-2-oxoethyl)benzamide 8,43-8,41 (m, 1Η), 8,10 (s, 1Η), 7,80 (d, J = 7,6 Hz, 1H), 7,72 (s, 1H), 7,61 (d, J=8,8Hz, 2H), 7,54- 7.51 (m, 3H), 7,47–7,43 (m, 1H), 6,88–6,86 (m, 2H), 6,55 (d, J = 8.4Hz, 2H), 5,6 (s, 2H), 4,11 (d, J = 5,6 Hz, 2H) ppm 140 4-Amino-N-(2-((4-(3-(1-methyl-2-oxo- 460,2 1H-NMR (400 MHz, DMSO) δ = 12.48- 1,2-Dihydropyridin-4-yl) 12,23 (m, 1Η), 8,43 (br s, 1Η), 8,24 (s, phenyl)thiazol-2-yl)amino)-2- 1Η), 8,00 (d, J = 8,2 Hz, 1Η), 7,86-7,79 (m, Oxoethyl)benzamide 2Η), 7,67 (d, J=7,6Hz, 1Η), 7,62 (d, J=8,6 Hz, 2Η), 7,58-7,52 (m, 1 Η), 6,75 (d, J=2,2). Hz, 1H), 6,63 (dd, J = 2,2, 7,2 Hz, 1H), 6,57 (d, J = 8,8 Hz, 2Η), 5,68 (s, 2Η), 4,12 (d, J = 5,8 Hz, 2H), 3,47 (s, 3H) ppm 141 4-amino-N-(2-((4-(3'-hydroxy-[1,1'- 445,2 1H-NMR (400 MHz, DMSO) δ = 12.37 (br s, Diphenyl]-3-yl)thiazol-2-yl) 1H), 9.56 (s, 1H), Amino)-2-oxoethyl)benzamide 8,45-8,43 (m, 1Η), 8,15 (s, 1Η), 7,89 (d, J = 7,6 Hz, 1H), 7.75 (s, 1H), 7.62 (d, J=8.4Hz, 2H), 7.56- 7.48 (m, 2H), 7.31-7.27 (m, 1H), 7,12 (d, J = 8,4 Hz, 1Η), 7,08-7,06 (m, 1Η), 6,80 (dd, J = 2,0, 7,6 Hz, 1H), 6.57 (d, J=8.8Hz, 2H), 5.67 (s, 2H), 4.12 (d, J = 6,0 Hz, 2Η) ppm 142 4-amino-N-(2-((4-(2'-fluoro-[1,1'- 446,9 1H-NMR (400 MHz, DMSO) δ = 12.26 (br s, Diphenyl]-3-yl)thiazol-2-yl) 1H), 8.43-8.42 (m, 1H), Amino)-2-oxoethyl)benzamide 8,10 (d, J = 1,2 Hz, 1Η), 7,94-7,93 (m, 1Η), 7.72 (s, 1H), 7.62-7.51 (m, 6H), 7.34-7.33 (m, 2H), 6,57 (d, J=8,8Hz, 2H), 5,65 (s, 2Η), 4,12 (d, J = 6,0 Hz, 2Η) ppm 143 4-Amino-N-(2-((4-(2-methyl-1-oxo-1,2- 434.1 1H-NMR (400 MHz, DMSO-d6) δ = 12,43 dihydroisoquinolin-7-yl) (br s, 1Η), 8,81 (s, 1Η), 8,40 (br s, 1Η), Thiazol-2-yl)amino)-2- 8,21-8,19 (m, 1Η), 7,74 (br s, 1Η), 7,70 (d, Oxoethyl)benzamide J=8.0Hz, 1H), 7.61 (d, J=8.4Hz, 2H), 7.48 (d, J = 7,2 Hz, 1H), 6,63 (d, J = 7,2 Hz, 1H), 6.57 (d, J=8.4Hz, 2H), 5.65 (s, 2H), 4.12 (br d, J = 5,6 Hz, 2H), 3,52 (s, 3H) ppm 144 3-[3-[2-12-1(4- 486,4 1H-NMR (400 MHz, DMSO) δ = 12.42 (s, Aminobenzoyl)amino]acetyl]amino]thiazole- 1 Η), 8,58 (br d, J = 4,6 Hz, 1 Η), 8,48- 4-yl]phenyl]-N-methylbenzamide 8,47 (m, 1 H), 8,26 (s, 1 H), 8,17 (s, 1 H), 7.94 (d, J = 7.8 Hz, 1 H), 7.90-7.84 (m, 2 H), 7,78 (s, 1 H), 7,70 (br d, J = 8,2 Hz, 1 H), 7,66 (d, J = 8,6 Hz, 2 H), 7,59- 7,57 (m, 2 H), 6,64 (d, J = 8,7 Hz, 2 H), 4,14 (d, J = 5,6 Hz, 2 H), 2,84 (d, J = 4,6 Hz, 3 H) ppm 145 4-Amino-N-(2-((4-(4'-- 486,1 1H-NMR (400 MHz, MeOD) δ = 8,18 (s, ((Dimethylamino)methyl)-[1,1'- 1H), 7.87 (d, J = 8.0 Hz, 1H), Diphenyl]-3-yl) 7,69-7,65 (m, 4Η), 7,57 (d, J = 8,0 Hz, 1Η), Thiazol-2-yl)amino)-2- 7,49-7,40 (m, 4H), Oxoethyl)benzamide 6,70-6,68 (m, 2Η), 4,27 (s, 2Η), 3,54 (s, 2Η), 2,28 (s, 6Η) ppm 146 4-Amino-N-(2-((4-(3-(2-Methoxypyridine- 460,2 1H-NMR (400 MHz, DMSO) δ = 12.37 (s, 4-yl)phenyl)thiazol-2-yl) 1Η), 8,44 (br s, 1Η), Amino)-2-oxoethyl)benzamide 8,28–8,26 (m, 2H), 8,00 (d, J=7,6Hz, 1H), 7,82 (s, 1H), 7,73 (d, J = 8,0 Hz, 1H), 7,63 (br d, J = 6,8 Hz, 2Η), 7,59-7,55 (m, 1Η), 7,37-7,35 (m, 1Η), 7,16 (s, 1Η), 6,59 (br d, J=4,8Hz, 2H), 4,12 (br d, J = 5,2 Hz, 2H), 3,91 (s, 3H) ppm 147 4-Amino-N-(2-oxo-2-((4-(1-oxo-1,2- 420.1 1H-NMR (400 MHz, DMSO) δ = 12.43 (br s, dihydroisoquinolin-7-yl) 1Η), 11,24 (br d, J = 5,2 Hz, 1 Η), Thiazol-2-yl)amino)ethyl)benzamide 8,78 (s, 1Η), 8,43-8,42 (m, 1Η), 8,22-8,20 (m, 1Η), 7,76 (s, 1Η), 7.71 (d, J=8.6Hz, 1H), 7.63 (d, J=8.6Hz, 2H), 7.20-7.17 (m, 1H), 6,58-6,56 (m, 3Η), 5,66 (s, 2Η), 4,13 (d, J = 5,6 Hz, 2Η) ppm 148 4-Amino-N-(2-((4-(3-methyl-4-oxo-3,4- 435,3 1H-NMR (400 MHz, DMSO-d6) δ = 12,31 dihydroquinazolin-6-yl) (br s, 1H), 8,71 (s, 1H), Thiazol-2-yl)amino)-2- 8,38–8,37 (m, 2H), 8,33–8,31 (m, 1H), Oxoethyl)benzamide 7,79 (s, 1Η), 7,71 (d, J = 8,6 Hz, 1Η), 7.61 (d, J=8.6Hz, 2H), 6.56 (d, J=8.6Hz, 2H), 5.65 (s, 2H), 4,11 (d, J = 5,6 Hz, 2H), 3,51 (s, 3H) ppm 149 4-Amino-N-(2-((4-(4'-cyano-[1,1'- 454 1H-NMR (400 MHz, DMSO-d6) δ = 12,37 Diphenyl]-3-yl)thiazol-2-yl) (br s, 1 Η), 8,42-8,26 (m, 1 Η), Amino)-2-oxoethyl)benzamide 7,99 (s, 1Η), 7,97-7,93 (m, 5Η), 7,81 (s, 1H), 7.62 (d, J = 8.8 Hz, 1H), 7,60-7,58 (m, 3Η), 6,57 (d, J = 8,8 Hz, 2Η), 5.66(s, 2H), 4,12 (d, J = 5,6 Hz, 2Η). ppm 150 4-Amino-N-(2-((4-(3-methyl-4-oxo-3,4- 435,1 1H-NMR (400 MHz, DMSO) δ = 12.53 (br s, dihydrophthalazin-6-yl) 1H), 8.82 (s, 1H), Thiazol-2-yl)amino)-2- 8,46-8,42 (m, 3Η), 8,01-7,99 (m, 2Η), Oxoethyl)benzamide 7.62 (d, J=8.6Hz, 2H), 6.57 (d, J=8.6Hz, 2H), 5.68 (s, 2H), 4.13 (d, J = 5.6Hz, 2H), 3,74 (s, 3Η) ppm 151 4-Amino-N-(2-oxo-2-((4-(3-(1-(2,2,2- 500,8 1H-NMR (400 MHz, DMSO-d6) δ = 12,31 Trifluoretil)-1H-pirazol-3-il) (br s, 1Η), 8,57-8,32 (m, 2Η), phenyl)thiazole-2- 7,93 (d, J=2,3Hz, 1H), 7,85 (d, J=8,0Hz, il)amino)ethyl)benzamide 1H), 7.76 (d, J = 7.6 Hz, 1H), 7.69 (s, 1H), 7.61 (d, J=8.4Hz, 2H), 7.49- 7,48 (m, 1H), 6.89 (d, J=2.4Hz, 1H), 6.56 (d, J=8.4Hz, 2H), 5.66 (s, 2H), 5,21–5,20 (m, 2H), 4,12 (d, J=5,6Hz, 2Η).ppm 152 4-amino-N-(2-((4-(3'-fluoro-[1,1'- 447,1 1H-NMR (400 MHz, DMSO-d6) δ = 12,34 Diphenyl]-3-yl)thiazol-2-yl) (br s, 1Η), 8,42-8,41 (m, 1Η), Amino)-2-oxoethyl)benzamide 8,22 (d, J = 1,6 Hz, 1Η), 7,93-7,92 (m, 1Η), 7,80 (s, 1H), 7.63-7.62 (m, 1H), 7.60-7.54 (m, 6H), 7.24-7.23 (m, 1H), 6.57 (d, J=8.8Hz, 2H), 5.65 (s, 2H), 4.12 (d, J = 6,0 Hz, 2Η) ppm 153 4-amino-N-(2-((4-(4'-(hydroxymethyl)- 458,8 1H-NMR (400 MHz, DMSO-d6) δ = 8,49- [1,1'-Diphenyl]-3-yl) 8,38 (m, 2Η), 8,20 (s, 1Η), Thiazol-2-yl)amino)-2- 7,89 (d, J = 7,8 Hz, 1H), 7,76 (s, 1H), 7,69 Oxoethyl)benzamide (d, J = 8.2Hz, 2H), 7,62 (d, J = 8,4 Hz, 3Η), 7,56-7,49 (m, 1Η), 7.45 (d, J=8.0Hz, 2H), 6.57 (d, J=8.6Hz, 2H), 5.67 (s, 2H), 5.25 (br s, 1H), 4,57 (s, 2H), 4,13 (d, J = 5,6 Hz, 2H) ppm 154 4-amino-N-(2-((4-(3'-(hydroxymethyl)- 459,1 1H-NMR (400 MHz, DMSO-d6) δ = 12,37 [1,1'-Diphenyl]-3-yl) (br s, 1H), 8,42 (s, 1H), Thiazol-2-yl)amino)-2- 8,20 (s, 1Η), 7,94-7,85 (m, 1Η), 7,75 (s, Oxoethyl)benzamide 1H), 7.69-7.56 (m, 5H), 7,55-7,49 (m, 1Η), 7,45 (m, 1Η), 7,34 (br d, J = 7,6 Hz, 1H), 6.56 (d, J=8.4Hz, 2H), 5.66 (s, 2H), 5.28- 5,27 (m, 1H), 4,59 (d, J=6,0Hz, 2H), 4,12 (br d, J=5,6 Hz, 2Η) ppm 155 (R)-4-Amino-N-(4-(methylthio)-1-oxo-1- 504,3 1H-NMR (400 MHz, DMSO-d6) δ = 12,38 ((4-(3-(Pyridin-4-yl))) (s, 1H), 8,67 (d, J=6,0Hz, 2H), phenyl)thiazol-2-yl)amino)butan-2- 8,30 (s, 1H), 8,20 (d, J = 6,8 Hz, 1H), 8,01 il) Benzamide (d, J = 7,6 Hz, 1H), 7,82 (s, 1H), 7,76-7,75 (m, 3Η), 7,65 (d, J = 8,4 Hz, 2Η), 7.61-7.57 (m, 1H), 6,56 (d, J=8,8Hz, 2Η), 5,65-5,64 (m, 2Η), 4.72-4.67 (m, 1H), 2,64–2,60 (m, 2H), 2,12–2,06 (m, 5H) ppm 156 4-(3-(2-(2-(4- 446,2 1H-NMR (400 MHz, DMSO) δ = 12.36 (s, Aminobenzamido)acetamido)thiazole-4- 1Η), 8,42 (s, 1Η), 8,31 (d, J = 7,6 Hz, 2Η), il) 8,27 (s, 1Η), 7,97 (s, 1Η), 7,83-7,81 (m, Phenyl)pyridine-1-oxide 3Η), 7,73 (d, J = 8,8 Hz, 1 Η), 7,61 (d, J = 8,4 Hz, 2H), 7.58-7.56 (m, 1H), 6.57 (d, J=8.4 Hz, 2H), 4,12 (d, J = 5,6 Hz, 2Η) ppm 157 4-Amino-N-(2-((4-(3'-cyano-[1,1'- 454,2 1H-NMR (400 MHz, DMSO) δ = 12.36 (br s, Diphenyl]-3-yl)thiazol-2-yl) 1Η), 8,42 (br s, 1Η), Amino)-2-oxoethyl)benzamide 8,25 (br d, J = 10,0 Hz, 2Η), 8,15-7,94 (m, 2H), 7.89-7.55 (m, 7H), 6.57 (d, J=3.8Hz, 2H), 5.66 (s, 2H), 4.12 (s, 2Η) ppm 158 4-Amino-N-(2-((4-(4'-(aminomethyl)- 462,3 1H-NMR (400 MHz, DMSO) δ = 12.31- 2',3',4',5'-Tetrahydro-[1,1'-diphenyl]- 12.24 (m, 1H), 8.45-8.43 (m, 1H), 3-yl)thiazol-2-yl)amino)-2- 7,95 (s, 1Η), 7,78-7,77 (m, 3Η), 7,67 (s, Oxoethyl)benzamide 1Η), 7,62 (d, J=7,2Hz, 2Η), 7.39–7.38 (m, 2H), 6.58 (d, J=8.8Hz, 2H), 6,21 (br s, 1H), 4.12 (d, J=5.6Hz, 2Η), 2.89-2.81 (m, 2Η), 2,38 (br d, J = 17,2 Hz, 3H), 1,99–1,93 (m, 3H), 1,46–1,40 (m, 1H) ppm 159 4-Amino-N-(2-oxo-2-((4-(3-(2- 514,3 1H-NMR (400 MHz, DMSO) δ = 12.36 (s, (piperazin-1-yl)pyridin-4-yl) 1Η), 8,81 (br s, 2Η), phenyl)thiazole-2- 8,47–8,44 (m, 1H), 8,26–8,24 (m, 2H), 7,98 il)amino)ethyl)benzamide (d, J = 8,0 Hz, 1H), 7,79 (s, 1H), 7,72 (d, J = 8,0 Hz, 1H), 7,63- 7.58 (m, 3H), 7,24 (s, 1H), 7,12 (d, J = 6,4 Hz, 1H), 6,60 (d, J = 8.4Hz, 2H), 4,13 (d, J=5,6Hz, 2H), 3,85-3,82 (m, 4H), 3,23-3,017 (m, 4Η) ppm 160 4-Amino-N-(2-((4-(3-(2-((2- 488,2 1H-NMR (400 MHz, MeOD) δ = 8,30 (s, (Aminoethyl)amino)pyridin-4-yl)phenyl) 1H), 8.14-8.04 (m, 2H), Thiazol-2-yl)amino)-2- 7,77-7,71 (m, 3Η), 7,65-7,57 (m, 2Η), 7,35- Oxoethyl)benzamide 7.25 (m, 2H), 6.82-6.74 (m, 2H), 4.29 (s, 2H), 3.81-3.80 (m, 2Η), 3,30-3,28 (m, 2Η) ppm 161 4-Amino-N-(2-((4-(3- 378 1H-NMR (400 MHz, DMSO-d6) δ = 12,38 Cyanophenyl)thiazol-2-yl) (br s, 1Η), 8,43-8,42 (m, 1Η), Amino)-2-oxoethyl)benzamide 8,32 (s, 1Η), 8,23-8,21 (m, 1Η), 7,85 (s, 1H), 7.78 (d, J = 7.6 Hz, 1H), 7.69-7.64 (m, 1H), 7.64-7.57 (m, 2H), 6.56 (d, J = 8.8Hz, 2H), 5,65 (s, 2H), 4,12 (d, J = 5,6 Hz, 2H) ppm 162 4-Amino-N-(2-oxo-2-((4-(3-(1- 503.1 1H-NMR (400 MHz, DMSO-d6) δ = 12,37 (Tetrahydro-2H-pyran-4-yl)-1H- (br s, 1Η), 8,43-8,42 (m, 1Η), Pyrazol-3-yl)phenyl)thiazole-2- 8,37 (s, 1H), 7,87 (d, J = 2,0 Hz, 1H), 7,81 il)amino)ethyl)benzamide (br d, J = 7,6 Hz, 1H), 7,73 (br d, J = 7,6 Hz, 1Η), 7,67 (s, 1Η), 7,62 (br d, J = 8,4 Hz, 2H), 7,45-7,44 (m, 1Η), 6,75 (d, J = 2,0 Hz, 1Η), 6.57 (d, J=8.4Hz, 2H), 5,66 (s, 2H), 4,52-4,40 (m, 1H), 4,12 (br d, J = 5.6Hz, 2H), 4.00–3.97 (m, 2H), 3.54–3.45 (m, 2H), 2,06-1,96 (m, 4Η) ppm 163 4-Amino-N-(2-((4-(3-(2-(2- 489 1H-NMR (400 MHz, MeOD) δ = 8,28-8,21 Aminoethoxy)pyridin-4-yl) (m, 2H), 8,02 (d, J=7,8Hz, 1H), phenyl)thiazol-2-yl)amino)-2- 7,77-7,70 (m, 2Η), 7,67 (d, J=7,8Hz, 1Η), Oxoethyl)benzamide 7,60-7,51 (m, 2Η), 7,39 (dd, J = 5,6 Hz, 1,6 Hz, 1H), 7,22 (d, J = 1,2 Hz, 1H), 6.81-6.71 (m, 2H), 4.66-4.58 (m, 2H), 4.29 (s, 2Η), 3,45-3,39 (m, 2Η) ppm 164 4-Amino-N-(2-((4-(3-(2- 459,2 1H-NMR (400 MHz, DMSO) δ = 8.61 (d, (Aminomethyl)pyridine-4- J = 4,8 Hz, 1Η), 8,44-8,41 (m, 1Η), yl)phenyl)thiazole- 8,32-8,30 (m, 2Η), 8,01 (d, J=8,0Hz, 1Η), 2-yl)amino)-2-oxoethyl)benzamide 7,84 (s, 1Η), 7,79 (s, 1Η), 7.77-7.74 (m, 1H), 7.66-7.58 (m, 4H), 6.58- 6,54 (m, 2Η), 5,66 (s, 2Η), 4,12 (d, J = 5,6 Hz, 2H), 4,01 (s, 2H) ppm 165 4-Amino-N-(2-((4-(3-(2- 460,2 1H-NMR (400 MHz, MeOD) δ = 8,74 (d, (Hydroxymethyl)pyridin-4-yl) J = 6,2 Hz, 1Η), 8,49 (s, 1Η), phenyl)thiazol-2-yl)amino)-2- 8,34 (s, 1Η), 8,28 (br d, J = 6,2 Hz, 1Η), Oxoethyl)benzamide 8,19 (d, J = 7,8 Hz, 1Η), 7,91 (d, J=8,6Hz, 1Η), 7,78-7,72 (m, 2Η), 7.69-7.67 (m, 1H), 7,63 (s, 1Η), 6,85-6,77 (m, 2Η), 5,08 (s, 2Η), 4,30 (s, 2Η) ppm 166 4-Amino-N-(2-((4-(3-(1-(2-aminoethyl)- 489,2 1H-NMR (400 MHz, MeOD) δ = 8,25 (s, 2-Oxo-1,2-di-hydropyridin-4-yl) 1H), 8.04 (d, J = 8.0 Hz, 1H), phenyl)thiazol-2-yl)amino)-2- 7,81-7,69 (m, 3Η), 7,66 (br d, J = 7,2 Hz, Oxoethyl)benzamide 1H), 7.60-7.53 (m, 2H), 6,92 (d, J = 2,0 Hz, 1H), 6,86 (dd, J = 7,2 Hz, 2,0 Hz, 1H), 6.80 (d, J=8.8Hz, 2Η), 4.38-4.34 (m, 2Η), 4,29 (s, 2Η), 3,40 (m, 2Η) ppm 167 4-Amino-N-(2-((4-(2-(2- 491 1H-NMR (400 MHz, DMSO) δ = 12.42 (br s, (Dimethylamino)ethyl)-1-oxo-1,2- 1H), 8.80 (d, J = 2.0 Hz, 1H), Dihydroisoquinoline- 8.43-8.40 (m, 1H), 8.21-8.19 (m, 1H), 8.15 7-yl)thiazol-2-yl)amino)-2- (s, 1H), 7,76 (s, 1H), Oxoethyl)benzamide 7.69 (d, J=8.4Hz, 1H), 7.62 (d, J=8.8Hz, 2Η), 7,45 (d, J = 7,2Hz, 1Η), 6.62 (d, J=7.2Hz, 1H), 6.57 (d, J=8.8Hz, 2Η), 5,65 (s, 2Η), 4,13 (d, J = 6,0 Hz, 2Η), 4.08-4.05 (m, 2H), 2.58-2.55 (m, 2H), 2.20 (s, 6Η) ppm 168 4-Amino-N-(2-((4-(3-(2- 487 1H-NMR (400 MHz, DMSO) δ = 12.37 (s, ((Dimethylamino)methyl)pyridin-4-yl) 1H), 10.01 (s, 1H), phenyl)thiazol-2-yl)amino)-2- 8,78 (d, J = 5,2 Hz, 1Η), 8,46-8,43 (m, 1Η), Oxoethyl)benzamide 8,32 (s, 1H), 8,04 (d, J = 8,0 Hz, 1H), 7,94 (s, 1H), 7,88- 7,86 (m, 1H), 7,80-7,75 (m, 2Η), 7,65-7,61 (m, 3Η), 6,58 (d, J = 8.4Hz, 2H), 4.53 (s, 2H), 4.13 (d, J=5.6Hz, 2H), 2.87 (s, 6Η) ppm 169 4-Amino-N-(2-((4-(3-(4- 464,2 1H-NMR (400 MHz, MeOD) δ = 7,78 (br d, (Aminomethyl)cyclohexyl)phenyl) J = 13,2 Hz, 1H), 7,71-7,67 (m, 3H), 7,36- Thiazol-2-yl)amino)-2- 7,35 (m, 2Η), 7,32-7,16 (m, 1Η), 6,70 (d, Oxoethyl)benzamide J=8.6Hz, 2H), 4.25(s, 2H), 3.08(d, J=7.6 Hz, 1 Η), 2,86 (d, J = 7,2 Hz, 1 Η), 2,72-2,55 (m, 1H), 1.80-1.89 (m, 2H), 1.95-1.75 (m, 5H), 1.61- 1,58 (m, 1Η), 1,29-1,23 (m, 1Η) ppm 170 4-Amino-N-(2-((4-(3-(1-(2- 517,3 1H-NMR (400 MHz, MeOD) δ = 8,40 (br s, (Dimethylamino)ethyl)-6-oxo-1,6- 1H), 8.10-8.05 (m, 2H), Dihydropyridin-3-yl) 7.99 (d, J=8.0Hz, 1H), 7.90 (d, J=8.0 phenyl)thiazol-2-yl)amino)-2- Hz, 1Η), 7,70 (d, J = 8,0 Hz, 2Η), Oxoethyl)benzamide 7.51-7.47 (m, 3H), 8.73-8.70 (m, 3H), 4.39- 4,36 (m, 2Η), 4,28 (s, 2Η), 3,22–3,19 (m, 2H), 2,72 (s, 6H) ppm 171 4-Amino-N-(2-((4-(2-(2-aminoethyl)-1- 463 1H-NMR (400 MHz, DMSO) δ = 8.82 (d, Oxo-1,2-dihydroisoquinolin-7-yl) J = 1,6 Hz, 1Η), 8,46-8,43 (m, 1Η), Thiazol-2-yl)amino)-2- 8,31 (br s, 1Η), 8,23-8,21 (m, 1Η), 7,77 (s, Oxoethyl)benzamide 1H), 7.71 (d, J = 8.4 Hz, 1H), 7.62 (d, J=8.8Hz, 2H), 7.46 (d, J=7.2Hz, 1H), 6.65 (d, J = 7.2 Hz, 1H), 6,57 (d, J = 8,4 Hz, 2Η), 5,66 (br s, 2Η), 4.13 (d, J=5.6Hz, 2H), 4,06 (br s, 2H), 2,99 (br s, 2H) ppm 172 (S)-4-amino-N-(3-hydroxy-1-oxo-1-((4- 460,2 1H-NMR (400 MHz, DMSO-d6) δ = 12,35 (s, (3-(Pyridin-4-yl) 1Η), 8,92 (d, J=6,4Hz, 2Η), phenyl)thiazol-2-yl)amino)propan-2- 8.44 (s, 1H), 8.28 (d, J=6.4Hz, 2H), 8.12 il) Benzamide (d, J = 8,0 Hz, 1H), 8,00 (d, J = 7,2 Hz, 1H), 7,92 (br d, J = 8,8 Hz, 1Η), 7,87 (s, 1Η), 7,69-7,64 (m, 3Η), 6,61 (d, J = 8,8 Hz, 2Η), 4.72-4.68 (m, 1H), 3,85-3,83 (m, 2Η) ppm 173 4-Amino-N-(2-oxo-2-((4-(1-(pyridin-3- 437,4 1H-NMR (400 MHz, DMSO) δ = 12.17 (s, il)piperidin-3-yl) 1 H), 8.39-8.37 (m, 1 H), Thiazol-2-yl)amino)ethyl)benzamide 8,32 (d, J = 2,6 Hz, 1Η), 7,96-7,95 (m, 1Η), 7.60 (d, J=8.6Hz, 2H), 7,37–7,32 (m, 1H), 7,22–7,20 (m, 1H), 6,91 (s, 1H), 6,56 (d, J=8,6Hz, 2H), 5,65 (s, 2H), 4,07 (d, J=5,8Hz, 2H), 3,93 (d, J = 11,0 Hz, 1H), 3,78 (d, J = 11,4 Hz, 1Η), 2,97-2,75 (m, 3Η), 2,07 (d, J = 8,0 Hz, 1H), 1,84–1,78 (m, 1H), 1,68–1,66 (m, 2H) ppm 174 4-Amino-N-(2-((4-(3-((1r, 4r)-4- 464,3 1H-NMR (400 MHz, MeOD) δ = 7,78–7,67 (Aminomethyl)cyclohexyl)phenyl) (m, 4Η), 7,37-7,27 (m, 2Η), Thiazol-2-yl)amino)-2- 7.17 (d, J=7.6Hz, 1Η), 6.91-6.76 (m, 2Η), Oxoethyl)benzamide 4,26 (s, 2H), 2,85 (d, J=7,0Hz, 2H), 2,62- 2.53 (m, 1H), 2.03-1.92 (m, 4H), 1.74-1.72 (m, 1H), 1,65–1,52 (m, 2H), 1,31–1,17 (m, 2H) ppm 175 4-Amino-N-(2-((4-(3-((1s, 4s)-4- 464,3 1H-NMR (400 MHz, MeOD) δ = 7,85–7,75 (Aminomethyl)cyclohexyl)phenyl)thiazole- (m, 3H), 7,71 (br d, J = 8,0 Hz, 1H), 7,41- 2-yl)amino)-2-oxoethyl)benzamide 7,30 (m, 2H), 7,24 (d, J=7,8Hz, 1H), 6,97 (br s, 2Η), 4,29 (d, J = 2,6 Hz, 2H), 3,09 (d, J = 7,6 Hz, 2H), 2,73 (br s, 1H), 2,07 (br s, 1Η), 1,80 (br s, 8Η) ppm 176 2-(4-(3-(2-(2-(4- 477,2 1H-NMR (400 MHz, DMSO-d6) δ = 13,13 (s, Aminobenzamido)acetamido)thiazole-4- 1H), 12.35 (s, 1H), 8.43-8.40 (m, 1H), 8.18 il) (s, 1H), 8,09 (s, 1H), 7,94 (s, 1H), 7,74 (br phenyl)-1H-pyrazol-1-yl)acetic acid d, J = 8,0 Hz, 1H), 7,71 (s, 1H), 7,61 (d, J=8,8Hz, 2H), 7,52 (br d, J = 8,0 Hz, 1H), 7.44-7.40 (m, 1H), 6.59-6.55 (m, 2H), 5.67 (br s, 2H), 4,99 (s, 2H), 4,12 (d, J = 6,0 Hz, 2Η) ppm 177 Methyl-2-(4-(3-(2-(2-(4-))) 502,3 1H-NMR (400 MHz, DMSO) δ = 12.37 (s, Aminobenzamide)acetamide) 1H), 8.58 (d, J = 5.2 Hz, 1H), Thiazol-4-yl)phenyl)pyridin-2-yl)acetate 8,43-8,40 (m, 1Η), 8,28 (s, 1Η), 8,01 (br d, J = 8,0 Hz, 1H), 7,80 (s, 1Η), 7,74-7,72 (m, 2Η), 7,66-7,57 (m, 4Η), 6.56 (d, J=8.8Hz, 2H), 5,65 (s, 2H), 4,12 (br d, J = 5,6 Hz, 2H), 3,94 (s, 2Η), 3,64 (s, 3Η) ppm 178 (S)-4-amino-N-(4-methoxy-1-oxo-1-((4- 488,2 1H-NMR (400 MHz, MeOD) δ = 8,88 (d, (3-(Pyridin-4-yl) J = 7,2 Hz, 2Η), 8,50 (s, 1Η), phenyl)thiazol-2-yl)amino)butan-2- 8.43 (d, J=7.2Hz, 2H), 8.20 (d, J=8.0Hz, il) Benzamide 1H), 7.92 (d, J = 8.8 Hz, 1H), 7,72 (d, J=8,4Hz, 2Η), 7,70-7,66 (m, 1Η), 7,64 (s, 1Η), 6,81-6,78 (m, 2Η), 4.85-4.82 (m, 1H), 3.61-3.58 (m, 2H), 3.37 (s, 3Η), 2,32-2,23 (m, 1Η), 2,20-2,12 (m, 1Η) ppm 179 4-Amino-N-(2-((4-(3- 438,3 1H-NMR (400 MHz, DMSO) δ = 12.29 (s, Morpholinophenyl)thiazol-2-yl) 1 H), 8.41-8.38 (m, 1 H), Amino)-2-oxoethyl)benzamide 7,63-7,57 (m, 3H), 7,46 (s, 1H), 7,38-7,32 (m, 1H), 7.31-7.24 (m, 1H), 6.92 (d, J=8.0Hz, 1H), 6.56 (d, J=8.8Hz, 2H), 5.65 (s, 2H), 4,10 (d, J=5,6Hz, 2H), 3,78-3,74 (m, 4H), 3,18-3,12 (m, 4H) ppm 180 Methyl-2-(4-(3-(2-(2-(4-))) 491,2 1H-NMR (400 MHz, DMSO) δ = 12.25 (br s, Aminobenzamido)acetamido)thiazole-4- 1H), 8.43-8.40 (m, 1H), il) 8,20 (s, 1Η), 8,09 (s, 1Η), 7,97 (s, 1Η), Phenyl)-1H-pyrazol-1-yl)acetate 7,74 (d, J = 7,6 Hz, 1Η), 7,70 (s, 1H), 7,61 (d, J=8,8Hz, 2H), 7,53 (d, J = 8,0 Hz, 1H), 7,44-7,40 (m, 1Η), 6,56 (d, J=8,8Hz, 2Η), 5.66(s, 2H), 5.13(s, 2H), 4,12 (d, J = 5,6 Hz, 2H), 3,71 (s, 3H) ppm 181 Methyl-4-(3-(2-(2-(4-))) 488,2 1H-NMR (400 MHz, DMSO) δ = 12.36 (s, Aminobenzamido)acetamido)thiazole-4- 1H), 8.81 (d, J = 5.2 Hz, 1H), il) 8,40-8,35 (m, 3Η), 8,06-8,02 (m, 2Η), 7,83- Phenylpicoline 7.80 (m, 2H), 7,64-7,60 (m, 3Η), 6,56 (d, J = 8,4 Hz, 2Η), 5.65(s, 2H), 4,11 (br d, J = 6,0 Hz, 2H), 3,93 (s, 3H) ppm 182 Methyl-2-(4-(3-(2-(2-(4-))) 530.1 1H-NMR (400 MHz, DMSO) δ = 12.36 (s, Aminobenzamido)acetamido)thiazole-4- 1H), 8.58 (d, J = 5.2 Hz, 1H), il) 8,44-8,41 (m, 1Η), 8,25 (s, 1Η), 8,01 (d, phenyl)pyridin-2-yl)-2- J = 8,0 Hz, 1H), 7,82 (s, 1H), Methylpropanoate 7,75 (d, J = 7,6 Hz, 1H), 7,68 (s, 1H), 7,62- 7,57 (m, 4H), 6.56 (d, J=8.4Hz, 2H), 5.66 (s, 2H), 4.12 (d, J=6.0Hz, 2H), 3,60 (s, 3Η), 1,60 (s, 6Η) ppm 183 4-amino-N-(2-((4-(3-(2-(N- 501,4 1H-NMR (400 MHz, DMSO) δ = 12.38 (s, (Methylacetamido)pyridin-4-yl)phenyl) 1H), 8.57 (d, J = 5.2 Hz, 1H), Thiazol-2-yl)amino)-2- 8,47-8,46 (m, 1Η), 8,31 (s, 1Η), 8,03 (d, Oxoethyl)benzamide J = 7,8 Hz, 1H), 7,89 (s, 1H), 7,83 (s, 1H), 7,79 (d, J = 7,6 Hz, 1H), 7,69- 8,68 (m, 1Η), 7,67-7,56 (m, 3Η), 6,62 (d, J=8,4Hz, 2Η), 4,18-4,10 (m, 2Η), 3,34 (s, 3Η), 2,08 (s, 3Η) ppm 184 5-Amino-N-(2-oxo-2-((4-(3-(pyridin-4- 431.1 1H NMR (400MHz, DMSO) δ = 8.69-8.67 phenyl) (m, 2H), 8.65-8.62 (m, 1H), Thiazol-2-yl)amino)ethyl)picolinamide 8,30 (s, 1H), 8,02 (d, J = 8,0 Hz, 1H), 7,97 (d, J = 2,4 Hz, 1H), 7,83 (s, 1H), 7,77-7,61 (m, 3Η), 7,72 (d, J = 8,4 Hz, 1Η), 7.62-7.58 (m, 1H), 7,01-6,98 (m, 1Η), 5,99 (s, 2Η), 4,21 (d, J = 6,0 Hz, 2Η) ppm 185 4-Amino-N-(2-((4-(3-(1,1- 486,4 1H-NMR (400 MHz, DMSO) δ = 12.31 (br s, Dioxythiomorpholino)phenyl) 1H), 8.40 (s, 1H), Thiazol-2-yl)amino)-2- 7,67–7,58 (m, 3H), 7,51 (s, 1H), 7,41–7,36 Oxoethyl)benzamide (m, 1H), 7.33-7.27 (m, 1H), 7,02-6,95 (m, 1Η), 6,56 (d, J=8,8Hz, 2Η), 5.66(s, 2H), 4,10 (d, J = 5,6 Hz, 2Η), 3,81 (br s, 4Η), 3,16 (br s, 4Η) ppm 186 4-Amino-N-[2-[[4-[3-[2-[2-[2- 558,3 1H-NMR (400 MHz, DMSO) δ = 12.60 (s, (Dimethylamino)ethylamino]-2-oxo- 1Η), 8,56 (d, J = 5,2 Hz, 1Η), 8,43-8,40 (m, Ethyl]-4-pyridyl]phenyl]thiazole-2- 1H), 8.27 (s, 1H), 8.18 (s, 1H), 8.07-8.06 yl]amino]-2-oxo-ethyl]benzamide (m, 1Η), 8,00 (br d, J = 7,6 Hz, 1Η), 7,79 (s, 1H), 7.72-7.69 (m, 2H), 7.62-7.57 (m, 4H), 6.56 (d, J=8.4Hz, 2H), 5.65 (s, 2H), 4.12 (br d, J = 6,0 Hz, 2H), 3,69 (s, 2H), 3,20- 3.15 (m, 2Η), 2.33-2.32 (m, 2Η), 2.14 (s, 6Η) ppm 187 1-[3-[2-[[2-[(4- 480,1 1H-NMR (400 MHz, MeOD) δ = 7,71–7,66 Aminobenzoyl)amino]acetyl]amino]thiazole- (m, 2H), 7.56 (s, 1H), 7.38-7.33 (m, 2H), 4-yl]phenyl]piperidine-4-carbons 7.26-7.24 (m, 1H), 6.95-6.94 (m, 1H), 6.72- acid 6,67 (m, 2Η), 4,26 (s, 2Η), 3,70 (d, J = 12,8 Hz, 2H), 2.89-2.77 (m, 2H), 2.46-2.45 (m, 1H), 2.09-1.99 (m, 2H), 1.92-1.78 (m, 2H) ppm 188 4-Amino-N-[2-[[4-[4-methyl-3-(4- 444.1 1H-NMR (400 MHz, DMSO) δ = 12.30 (br s, Pyridyl)phenyl]thiazol-2-yl]amino]-2- 1Η), 8,66 (d, J=5,6Hz, 2Η), 8,42-8,40 (m, Oxoethyl]benzamide 1Η), 7,87 (br d, J = 8,0 Hz, 1Η), 7,80 (s, 1Η), 7,66 (s, 1Η), 7,61 (br d, J = 8,8 Hz, 2Η), 7,45 (d, J=6,0Hz, 2Η), 7,41 (d, J=8,0 Hz, 1H), 6,56 (d, J=8,8Hz, 2H), 5,66 (s, 2Η), 4,10 (br d, J = 5,6 Hz, 2Η), 2,28 (s, 3Η) ppm 189 4-Amino-N-[2-[[4-[3-[2-[2- 516,4 1H-NMR (400 MHz, DMSO) δ = 12.38 (br (Dimethylamino)ethylamino]-4- s, 1Η), 8,42-8,44 (m, 1Η), 8,32 (s, 1Η), Pyridyl]phenyl]thiazol-2-yl]amino]-2- 8,19 (s, 1H), 8,06 (d, J = 5,8 Hz, 1H), 7,96 Oxoethyl]benzamide (br d, J = 7,6 Hz, 1H), 7,76 (s, 1H), 7,65- 7,50 (m, 4Η), 6,83-6,79 (m, 2Η), 6,57 (d, J = 8,6 Hz, 2H), 6,42 (br s, 1H), 5,67 (br s, 2Η), 4,13 (br d, J = 5,6 Hz, 2Η), 3,41 (br s, 2Η), 2,47 (br s, 2H), 2,22 (s, 6H) ppm 190 4-Amino-N-[2-[[4-[3-[2-(4- 528,2 1H-NMR (400 MHz, MeOD) δ = 8,27 (s, Methylpiperazine-1-yl)-4- 1Η), 8,22 (d, J = 5,6 Hz, 1Η), 8,03 (d, J = 8,0 Pyridyl]phenyl]thiazol-2-yl]amino]-2- Hz, 1Η), 7,79-7,77 (m, 2Η), 7,72-7,70 (m, Oxoethyl]benzamide 1Η), 7,58-7,54 (m, 2Η), 7,41 (br d, J = 6,4 Hz, 1Η), 7,30-7,27 (m, 1Η), 6,91-6,88 (m, 2Η), 4,28-3,99 (m, 6Η), 3,47 (br s, 4Η), 2,99 (s, 3Η) ppm 191 4-Amino-N-[2-[[4-[3-[2-[2- 530,0 1H-NMR (400 MHz, DMSO) δ = 12.35 (br s, (Dimethylamino)ethyl-methyl-amino]-4- 1H), 8.43-8.41 (m, 1H), 8.23-8.14 (m, 2H), Pyridyl]phenyl]thiazol-2-yl]amino]-2- 7,96 (d, J = 7,6 Hz, 1H), 7,78 (s, 1H), 7,67 Oxoethyl]benzamide (d, J=8.0Hz, 1H), 7.61 (d, J=8.8Hz, 2H), 7,58–7,51 (m, 1H), 6,94–6,71 (m, 2H), 6,56 (d, J = 8,4 Hz, 2H), 5,66 (br s, 2H), 4,12 (d, J = 6,0 Hz, 2Η), 3,72-3,69 (m, 2Η), 3,08 (s, 3H), 2,47–2,44 (m, 2H), 2,22 (s, 6H) ppm 192 4-Amino-N-[2-[[4-(1H-indazole-5- 392,9 1H-NMR (400 MHz, DMSO-d6) δ = 13,09 (s, yl)thiazol-2-yl]amino]-2-oxo- 1H), 12.29 (s, 1H), 8.42-8.39 (m, 1H), Ethyl]benzamide 8,27 (s, 1Η), 8,11 (s, 1Η), 7,91-7,87 (m, 1Η), 7,62-7,53 (m, 4Η), 6,56 (d, J = 8,4 Hz, 2Η), 5,65 (s, 2Η), 4,12 (d, J=5,6Hz, 2Η) ppm 193 5-Amino-N-[2-oxo-2-[[4-[3-(4- 432,2 1H-NMR (400 MHz, DMSO) δ = 12.44 (s, Pyridyl)phenyl]thiazole-2- 1H), 8.86-8.76 (m, 3H), 8.39 (s, 1H), 8.19 yl]amino]ethyl]pyrimidine-2- (s, 2H), 8,08 (d, J = 6,0 Hz, 3H), 7,89-7,83 carboxylic acid amide (m, 2H), 7,66-7,64 (m, 1H), 4,21 (d, J = 6,0 Hz, 2Η) ppm 194 4-amino-N-[2-[[4-[3-(3-methyl-4- 444,2 1H-NMR (400 MHz, DMSO) δ = 12.33 (br s, Pyridyl)phenyl]thiazol-2-yl]amino]-2- 1Η), 8,54 (s, 1Η), 8,49 (d, J = 4,8 Hz, 1Η), Oxoethyl]benzamide 8,43-8,40 (m, 1Η), 7,98 (d, J = 8,4 Hz, 1Η), 7,94 (d, J = 1,6 Hz, 1H), 7,75 (s, 1H), 7,62 (d, J=8,8Hz, 2H), 7,57–7,55 (m, 1H), 7,38 (d, J = 8,0 Hz, 1H), 7,30 (d, J = 4,8 Hz, 1H), 6.57 (d, J=8.8Hz, 2H), 5.66 (s, 2H), 4.12 (d, J = 6,0 Hz, 2H), 2,29 (s, 3H) ppm 195 4-amino-N-[2-[[4-[3-(2-methyl-4- 444,0 1H-NMR (400 MHz, DMSO) δ = 12.36 (m, Pyridyl)phenyl]thiazol-2-yl]amino]-2- 1Η), 8,53 (d, J = 5,2 Hz, 1Η), 8,43-8,40 (m, Oxoethyl]benzamide 1Η), 8,28 (s, 1Η), 7,99 (d, J = 7,6 Hz, 1Η), 7,81 (s, 1Η), 7,73 (br d, J = 7,6 Hz, 1Η), 7,62 (d, J = 2,4 Hz, 2Η), 7,60-7,53 (m, 3Η), 6.56 (d, J=8.8Hz, 2H), 5.66 (s, 2H), 4.12 (d, J = 5,6 Hz, 2H), 2,55 (s, 3H) ppm 196 4-Amino-N-[2-[[4-[3-[2- 473,0 1H RMN (400 MHz, METANOL-d4) δ = (Methylaminomethyl)-4- 8,72 (d, J = 5,2 Hz, 1H), 8,31 (s, 1H), 8,04 Pyridyl]phenyl]thiazol-2-yl]amino]-2- (d, J = 7,6 Hz, 1 Η), 7,81-7,77 (m, 4Η), Oxoethyl]benzamide 7.73 (d, J=8.0Hz, 1Η), 7.61-7.56 (m, 2Η), 6,97-6,92 (m, 2Η), 4,45 (s, 2Η), 4,30 (s, 2Η), 2,85 (s, 3Η) ppm 197 4-Amino-N-[2-oxo-2-[[4-[3-(4- 460,2 1H-NMR (400 MHz, DMSO) δ = 12.33 (s, Pyridyloxymethyl)phenyl]thiazole-2- 1H), 8.41-8.39 (m, 3H), 8.13 (s, 1H), 8.00 il]amino]ethyl]benzamide (s, 1Η), 7,88-7,84 (m, 1Η), 7,64-7,59 (m, 3H), 7.48-7.40 (m, 2H), 7.06-7.05 (m, 2H), 6,56 (d, J = 8,4 Hz, 2Η), 5,65 (br s, 2Η), 5,24 (s, 1H), 5,25 (s, 1H), 4,11 (d, J = 5,6 Hz, 2Η) ppm 198 4-Amino-N-[2-[[4-[3-(1-hydroxy-1- 411.2 1H-NMR (400 MHz, DMSO) δ = 12.35 (s, Methyl-ethyl)phenyl]thiazol-2-yl]amino]- 1H), 8.46-8.43 (m, 1H), 8.04 (s, 1H), 7.71 in 2-oxoethyl]benzamide (br d, J = 7,6 Hz, 1H), 7,63 (d, J = 8,4 Hz, 2H), 7.57 (s, 1H), 7.42-7.40 (m, 1H), 7.36- 7,32 (m, 1H), 6,62 (d, J=8,8Hz, 2H), 4,12 (br d, J = 5,6 Hz, 2H), 1,45 (s, 6H) ppm 199 5-amino-N-[2-[[4-[3-(2-methoxy-4- 461,2 1H-NMR (400 MHz, DMSO) δ = 12.43 (br s, Pyridyl)phenyl]thiazol-2-yl]amino]-2- 1H), 8.64-8.62 (m, 1H), 8.27-8.23 (m, 2H), Oxo-ethyl]pyridine-2-carboxamide 8,04–7,92 (m, 2H), 7,83 (s, 1H), 7,75–7,67 (m, 2Η), 7,62-7,52 (m, 1Η), 7,36-7,35 (m, 1H), 7.17 (s, 1H), 7.0-6.97 (m, 1H), 5.99 (s, 2H), 4,20 (d, J=6,0Hz, 2H), 3,92 (s, 3Η) ppm 200 2-[[4-[3-[2-[[2-[(4- 517,3 1H NMR (400MHz, DMSO) δ = 8.44-8.40 Aminobenzoyl)amino]acetyl]amino]thiazole- (m, 1Η), 8,29 (s, 1Η), 8,20 (s, 1Η), 8,13 (d, 4-yl]phenyl]-2-pyridyl]-methyl- J = 5,2 Hz, 1H), 7,97 (d, J = 7,6 Hz, 1H), 7,78 aminoacetic acid (s, 1Η), 7,68 (d, J = 7,6 Hz, 1Η), 7,61 (d, J=8,8Hz, 2H), 7,58-7,52 (m, 1H), 6,90 (d, J = 5,2 Hz, 1H), 6,85 (s, 1H), 6,56 (d, J = 8,8 Hz, 2H), 5,65 (br s, 2H), 4,25 (s, 2H), 4,12 (br d, J = 6,0 Hz, 2H), 3,12 (s, 3H) ppm 201 4-Amino-N-[2-oxo-2-[[4-[6-(4-pyridyl)-2- 431.1 1H-NMR (400 MHz, DMSO) δ = 12.44 (s, Piridil]tiazol-2- 1Η), 8,87 (d, J=4,8Hz, 2Η), 8,49-8,42 (m, il]amino]ethyl]benzamide 3H), 8.21-8.17 (m, 1H), 8.15-8.09 (m, 2H), 8,08–8,04 (m, 1H), 7,63 (d, J=8,8Hz, 2H), 6.59 (d, J=8.8Hz, 2H), 4.14 (d, J=5.6Hz, 2Η) ppm 19F-NMR (376 MHz, DMSO-d6) ä = -74,27 (s, 1F) 202 4-Amino-N-[2-[[4-[3-[2-[methyl-[2- 530,2 1H-NMR (400 MHz, DMSO) δ = 12.36 (br s, (Methylamino)-2-oxo-ethyl]amino]-4- 1H), 8.44-8.41 (m, 1H), 8.21 (s, 1H), 8.15 Pyridyl]phenyl]thiazol-2-yl]amino]-2- (d, J = 5,2 Hz, 1H), 7,98 (d, J = 8,0 Hz, 1H), Oxoethyl]benzamide 7,81-7,74 (m, 2Η), 7,68 (d, J=7,6Hz, 1Η), 7.62 (d, J=8.8Hz, 2Η), 7.58-7.54 (m, 1Η), 6,97-6,91 (m, 1Η), 6,87 (s, 1Η), 6,57 (d, J = 8,8 Hz, 2Η), 5,66 (s, 2Η), 4,20 (s, 2Η), 4,13 (d, J = 6,0 Hz, 2H), 3,13 (s, 3H), 2,60 (d, J = 4,4 Hz, 3Η) ppm 203 4-Amino-N-[2-[[4-[3-[2-(1,1- 563,2 1H-NMR (400 MHz, DMSO) δ = 12.38 (s, Dioxotiazinan-2-il)-4- 1Η), 8,53 (d, J = 5,2 Hz, 1 Η), 8,42-8,28 (m, Pyridyl]phenyl]thiazol-2-yl]amino]-2- 1Η), 8,27 (s, 1Η), 8,02 (d, J = 7,6 Hz, 1Η), Oxoethyl]benzamide 7,81 (s, 1Η), 7,72 (br d, J = 8,0 Hz, 1Η), 7,64-7,60 (m, 5Η), 6,57 (d, J = 8,4 Hz, 2Η), 5,65 (s, 2H), 4,13 (d, J=5,6Hz, 2H), 4,01- 3,98 (m, 2Η), 3,33 (br s, 2Η), 2,20-2,17 (m, 2Η), 1,98-1,84 (m, 2Η) ppm 204 4-Amino-N-[2-[[4-[3-(4-methyl-3-oxo- 465,2 1H-NMR (400 MHz, DMSO) δ = 12.33 (br s, Piperazin-1-yl)phenyl]thiazole-2- 1H), 8.41 (m, 1H), 7.68-7.59 (m, 3H), 7.47 yl]amino]-2-oxo-ethyl]benzamide (s, 1Η), 7,40-7,33 (m, 1Η), 7,32-7,26 (m, 1Η), 6,93 (m, 1Η), 6,58 (d, J = 8,6 Hz, 2Η), 5,67 (s, 2H), 4,11 (br d, J = 5,6 Hz, 2H), 3.82 (s, 2H), 3.56-3.52 (m, 2H), 3.47-3.44 (m, 2Η), 2,92 (s, 3Η) ppm 205 4-Amino-N-[2-[[4-[3-[2-[2- 501,2 1H-NMR (400 MHz, DMSO) δ = 12.40 (br s, (Methylamino)-2-oxo-ethyl]-4- 1Η), 8,57 (d, J = 5,4 Hz, 1 Η), 8,41 (br s, Pyridyl]phenyl]thiazol-2-yl]amino]-2- 1Η), 8,28 (s, 1Η), 8,01 (br d, J = 7,8 Hz, Oxoethyl]benzamide 2H), 7.79 (s, 1H), 7.75-7.68 (m, 2H), 7.67- 7,56 (m, 4Η), 6,57 (br d, J = 8,6 Hz, 2Η), 5,66 (s, 2H), 4,13 (br d, J = 5,6 Hz, 2H), 3,69 (s, 2H), 2,62 (d, J = 4,6 Hz, 3H) ppm 206 4-Amino-N-[2-[[4-[3-[2-[[2- 544,2 1H-NMR (400 MHz, DMSO) δ = 12.36 (s, (Dimethylamino)-2-oxo-ethyl]-methyl- 1H), 8.44-8.41 (m, 1H), 8.21 (s, 1H), 8.12 Amino]-4-pyridyl]phenyl]thiazole-2- (d, J = 5,2 Hz, 1H), 7,98 (d, J = 7,6 Hz, 1H), yl]amino]-2-oxo-ethyl]benzamide 7,79 (s, 1H), 7,68 (d, J = 8,4 Hz, 1H), 7,62 (d, J=8.4Hz, 2H), 7.59-7.53 (m, 1H), 6.92- 6,83 (m, 2Η), 6,57 (d, J=8,4Hz, 2H), 5,66 (s, 2H), 4,52 (s, 2H), 4,13 (d, J=5,6Hz, 2Η), 3,12-3,01 (m, 6Η), 2,82 (s, 3Η) ppm 207 4-amino-N-[2-[[4-[3-(2-isopropyl-4- 472,2 1H-NMR (400 MHz, DMSO) δ = 12.37 (s, Pyridyl)phenyl]thiazol-2-yl]amino]-2- 1Η), 8,57 (d, J = 4,8 Hz, 1 Η), 8,45-8,42 (m, Oxoethyl]benzamide 1Η), 8,27 (s, 1Η), 7,99 (d, J = 7,6 Hz, 1Η), 7,82 (s, 1H), 7,74 (d, J = 8,4 Hz, 1H), 7,62- 7.54 (m, 5Η), 6.56 (d, J=8.8Hz, 2H), 5.67 (s, 2H), 4.12 (d, J=5.6Hz, 2H), 3.15–3.08 (m, 1H), 1,29 (d, J = 6,8 Hz, 6H) ppm 208 4-Amino-N-[2-oxo-2-[[4-[5-(4-pyridyl)-3- 431,2 1H-NMR (400 MHz, DMSO) δ = 12.68- Piridil]tiazol-2- 12,28 (m, 1Η), 9,20 (d, J = 2,0 Hz, 1Η), il]amino]ethyl]benzamide 8,96 (d, J=2,2Hz, 1Η), 8,74-8,70 (m, 2Η), 8.62-8.61 (m, 1H), 8.44-8.42 (m, 1H), 7.99 (s, 1H), 7,87-7,84 (m, 2H), 7,62 (d, J = 8,6 Hz, 2H), 6,57 (d, J=8,6Hz, 2H), 5,66 (s, 2Η), 4,13 (d, J = 5,6 Hz, 2Η) ppm 209 Methyl-2-[3-[3-[2-[[2-[(4- 519,2 1H-NMR (400 MHz, DMSO) δ = 12.41 (s, Aminobenzoyl)amino]acetyl]amino]thiazole- 1Η), 8,44 (s, 1Η), 8,34 (s, 1Η), 8,00 (s, 4-yl]phenyl]pyrazol-1-yl]-2-methyl- 1Η), 7,83 (d, J = 6,4 Hz, 1 Η), 7,77-7,68 (m, Propanoato 2H), 7.63 (d, J = 7.2 Hz, 2H), 7.46 (s, 1H), 6,81 (s, 1H), 6,58 (d, J=7,2Hz, 2H), 5,68 (s, 2H), 4,13 (s, 2H), 3,65 (s, 3H), 1,84 (s, 6Η) ppm 210 2-[3-[3-[2-[[2-[(4- 477,1 1H-NMR (400 MHz, DMSO) δ = 12.45 (s, Aminobenzoyl)amino]acetyl]amino]thiazole- 1H), 8.64-8.30 (m, 2H), 7.87-7.79 (m, 2H), 4-yl]phenyl]pyrazol-1-yl]isigour 7.76–7.68 (m, 2H), 7.63 (d, J=8.8Hz, 2H), 7,46 (m, 1Η), 6,78 (d, J = 2,2 Hz, 1Η), 6,58 (d, J = 8,6 Hz, 2H), 5,01 (s, 2H), 4,13 (br d, J = 5,6 Hz, 2Η) ppm 211 4-Amino-N-[2-[[4-[3-[2-(1-amino-1- 487,4 1H-NMR (400 MHz, DMSO) δ = 8.61 (d, Methyl-ethyl)-4-pyridyl]phenyl]thiazole- J = 5,2 Hz, 1Η), 8,45-8,37 (m, 1Η), 8,28 (s, 2-yl]amino]-2-oxo-ethyl]benzamide 1H), 8.04-7.94 (m, 2H), 7.83-7.72 (m, 2H), 7,64-7,53 (m, 5Η), 6,56 (d, J = 8,8 Hz, 2Η), 5.66 (s, 2H), 4.12 (d, J=5.6Hz, 2H), 1.49 (s, 6Η) ppm 212 2-3-[3-12-1[2-1(4- 505,2 1H-NMR (400 MHz, DMSO) δ = 12.42 (s, Aminobenzoyl)amino]acetyl]amino]thiazole- 1H), 8.42 (s, 1H), 8.36 (s, 1H), 7.96 (d, 4-yl]phenyl]pyrazol-1-yl]-2-methyl- J = 2,4 Hz, 1H), 7,82 (br d, J = 8,0 Hz, 1H), propanoic acid 7,74 (br d, J = 7,6 Hz, 1Η), 7,69 (s, 1Η), 7.63 (d, J=8.6Hz, 2H), 7.46 (m, 1H), 6.79 (d, J=2.4Hz, 1H), 6.58 (d, J=8.6Hz, 2H), 4,14 (d, J = 5,6 Hz, 2H), 1,81 (s, 6H) ppm 213 4-Amino-N-[2-[[4-[3-[1-[1,1-dimethyl-2- 518,3 1H-NMR (400 MHz, DMSO) δ = 12.43 (br s, (Methylamino)-2-oxo-ethyl]pyrazole-3- 1H), 8.43 (m, 1H), 8.37 (s, 1H), 7.92 (d, yl]phenyl]thiazol-2-yl]amino]-2-oxo- J = 2,4 Hz, 1H), 7,83 (d, J = 8,0 Hz, 1H), 7,76 Ethyl]benzamide (d, J = 7,8 Hz, 1Η), 7,69 (s, 1Η), 7,62 (d, J = 8,8 Hz, 2Η), 7,46 (m, 1Η), 7,34 (br d, J = 4,4 Hz, 1H), 6,81 (d, J = 2,4 Hz, 1H), 6,57 (d, J=8.8Hz, 2H), 5.68 (s, 2H), 4.13 (d, J=5.6Hz, 2H), 2.59 (d, J=4.4Hz, 3H), 1.76 (s, 6Η) ppm 214 (S)-4-Amino-N-(1-((4-(3-(2- 518.1 1H-NMR (400 MHz, DMSO) δ = 12.44 (s, Methylpyridin-4-yl)phenyl)thiazol-2- 1H), 8.92-8.79 (m, 1H), 8.46 (s, 1H), 8.36- yl)amino)-4-(methylthio)-1-oxobutan-2- 8,09 (m, 4Η), 7,94 (br d, J = 7,4 Hz, 1Η), il) Benzamide 7,89 (s, 1H), 7,74-7,59 (m, 3H), 6,59 (br dd, J = 8,6, 2,6 Hz, 2Η), 4,75-4,65 (m, 1Η), 2,76 (d, J = 2,4 Hz, 3Η), 2,66-2,52 (m, 3Η), 2,15-2,04 (m, 5Η) ppm 215 N-[1-[4-[3-[2-[[2-[(4- 597,5 1H-NMR (400 MHz, DMSO) δ = 12.46- Aminobenzoyl)amino]acetyl]amino]thiazole- 12,29 (m, 1Η), 9,59 (s, 1H), 8,65 (d, J = 4-yl]phenyl]-2-pyridyl]-1-methyl- 5,2 Hz, 1Η), 8,44-8,43 (m, 1Η), 8,27 (s, Ethyl]-5-methyl-1,3,4-oxadiazole-2- 1H), 8.01 (d, J = 7.6 Hz, 1 H), 7.89 (s, 1 H), carboxylic acid amide 7,81 (s, 1H), 7,75 (d, J = 7,6 Hz, 1H), 7,68- 7,67 (m, 1Η), 7,58 (s, 3H), 6,57 (d, J = 8,8 Hz, 2H), 5,67 (s, 2H), 4,12 (d, J=5,6Hz, 2Η), 2,58 (s, 3H), 1,81 (s, 6H) ppm 216 4-Amino-N-[2-[[4-[3-[1-[2- 490,2 1H-NMR (400 MHz, DMSO) δ = 12.45 (br s, (Methylamino)-2-oxo-ethyl]pyrazole-3- 1Η), 8,52-8,34 (m, 2Η), 8,04 (br d, J = 4,8 yl]phenyl]thiazol-2-yl]amino]-2-oxo- Hz, 1Η), 7,82 (br d, J = 7,8 Hz, 1Η), 7,79 (d, Ethyl]benzamide J = 2,4 Hz, 1H), 7,72 (br d, J = 7,8 Hz, 1H), 7,69 (s, 1H), 7,62 (d, J=8,6Hz, 2H), 7,46 (m, 1Η), 6,77 (d, J = 2,4 Hz, 1Η), 6,57 (d, J = 8,6 Hz, 2Η), 5,68 (s, 2Η), 4,84 (s, 2Η), 4,13 (br d, J=5,6Hz, 2H), 2,65 (d, J=4,6 Hz, 3Η) ppm 217 4-Amino-N-[2-oxo-2-[[4-[2-(4-pyridyl)-4- 431,2 1HNMR (400 MHz, DMSO) δ = 12,48 (m, Piridil]tiazol-2- 1Η), 8,78 (d, J = 4,8 Hz, 1 Η), 8,74-8,72 (m, il]amino]ethyl]benzamide 2H), 8.52 (s, 1H), 8.45-8.42 (m, 1H), 8.18 (s, 1H), 8,10-8,09 (m, 2H), 7,61 (d, J = 8,8 Hz, 2H), 7.69-7.53 (m, 1H), 6.56 (d, J=8.8 Hz, 2H), 5,66 (s, 2H), 4,13 (d, J=5,6Hz, 2Η) ppm 218 4-Amino-N-[2-oxo-2-[[4-[3-[2-(4- 458,2 1HNMR (400 MHz, MeOD) δ = 8,68 (d, Pyridyl)ethyl]phenyl]thiazole-2- J=6,8Hz, 2H), 7,91 (d, J=6,8Hz, 2H), il]amino]ethyl]benzamide 7,77-7,73 (m, 4H), 7,36 (s, 1H), 7,31-7,29 (m, 1H), 7,15 (d, J = 8,0 Hz, 1H), 6,86-6,84 (m, 2Η), 4,27 (s, 2Η), 3,36-3,34 (m, 2Η), 3,16-3,12 (m, 2Η) ppm 219 1-[4-[3-[2-[[2-[(4- 514,3 1H-NMR (400 MHz, DMSO) δ = 13.40- Aminobenzoyl)amino]acetyl]amino]thiazole- 12.88 (m, 1H), 12.53-12.29 (m, 1H), 8.60- 4-yl]phenyl]-2- 8.52 (m, 1H), 8.48-8.38 (m, 1H), 8.31-8.25 Pyridyl]cyclopropane carbons (m, 1Η), 8,05-7,97 (m, 1Η), 7,89-7,80 (m, 2H), 7.78-7.71 (m, 1H), 7.66-7.55 (m, 4H), 6.63-6.53 (m, 2H), 5.72-5.61 (m, 2H), 4.17- 4,09 (m, 2H), 1,61-1,44 (m, 4H) ppm 220 4-Amino-N-[2-[[4-[3-(1-methylimidazole-4- 433.1 1H NMR (400MHz, DMSO) δ = 8.43-8.40 yl)phenyl]thiazol-2-yl]amino]-2-oxo- (m, 1Η), 8,35 (s, 1H), 8,19 (s, 1H), 7,72- Ethyl]benzamide 7,60 (m, 7Η), 7,40-7,38 (m, 1Η), 6,56 (d, J=8.8Hz, 2H), 5.66(s, 2H), 4.12(d, J=6.0 Hz, 2Η), 3,70 (s, 3Η) ppm 221 4-Amino-N-[2-[[4-[3-[2-(3-Amino-1- 528,3 1HNMR (400 MHz, DMSO) δ = 8,45–8,42 Piperidyl)-4-pyridyl]phenyl]thiazole-2- (m, 1H), 8.35-8.32 (m, 1H), 8.20 (s, 1H), yl]amino]-2-oxo-ethyl]benzamide 8.18 (d, J=5.2Hz, 1H), 7.97 (d, J=7.6 Hz, 1H), 7,79 (s, 1H), 7,69 (br d, J = 8,0 Hz, 1H), 7.61 (d, J = 8.8 Hz, 2H), 7.57-7.53 (m, 1Η), 7,08 (s, 1Η), 6,95 (d, J = 5,2 Hz, 1Η), 6,56 (d, J = 8,4 Hz, 2Η), 5,67 (s, 2Η), 4,36-4,33 (m, 1Η), 4,15-4,11 (m, 3Η), 3,03- 2.78 (m, 4H), 2.00-1.88 (m, 1H), 1.77-1.67 (m, 2Η), 1,57-1,34 (m, 2Η) ppm 222 4-Amino-N-[2-[[4-[3-(1,2- 447,2 1H-NMR (400 MHz, DMSO) δ = 12.41 (s, Dimethylimidazol-4-yl)phenyl]thiazol-2- 1H), 8.44-8.41 (m, 1H), 8.31-8.30 (m, 1H), yl]amino]-2-oxo-ethyl]benzamide 7.68-7.66 (m, 1H), 7.62-7.59 (m, 4H), 7.51 (s, 1Η), 7,37-7,33 (m, 1Η), 6,57-6,55 (m, 2Η), 5,67 (s, 2Η), 4,12 (d, J = 6,0 Hz, 2Η), 3,58 (s, 3Η), 2,32 (s, 3Η) ppm 223 4-Amino-N-[2-[[4-[3-[2-[1- 527,3 1H-NMR (400 MHz, DMSO) δ = 12.49- (Metilcarbamoil) Ciclopropil]-4- 12,20 (m, 1Η), 8,61 (d, J = 5,4 Hz, 1Η), Pyridyl]phenyl]thiazol-2-yl]amino]-2- 8,46-8,38 (m, 1Η), 8,27 (s, 1Η), 8,01 (d, J = Oxoethyl]benzamide 7,8 Hz, 1H), 7,82 (s, 1H), 7,74 (d, J = 7,8 Hz, 1Η), 7,67-7,56 (m, 6Η), 6,57 (d, J = 8,6 Hz, 2H), 5.67 (s, 2H), 4.13 (d, J=5.8Hz, 2Η), 2,62 (d, J = 4,6 Hz, 3Η), 1,43-1,37 (m, 2Η), 1,30-1,24 (m, 2Η) ppm 224 [2-[4-[3-[2-[[2-[(4- 488,4 1H RMN (400 MHz, METANOL-d4) δ = Aminobenzoyl)amino]acetyl]amino]thiazole- 8,46 (d, J = 5,2 Hz, 1H), 8,30 (s, 1H), 7,98 4-yl]phenyl]-2- (d, J = 8,0 Hz, 1Η), 7,77 (s, 1Η), 7,72 (d, Pyridyl]acetyl]oxysodium J=8.8Hz, 2H), 7.67 (d, J=7.6Hz, 1H), 7.60 (d, J = 5,2 Hz, 1Η), 7,53-7,49 (m, 1Η), 7,36 (s, 1H), 6,70 (d, J = 8,4 Hz, 2H), 4,20 (s, 2H), 3,80 (s, 2H) 225 4-Amino-N-[2-[[4-[3-[2-[3- 556,2 1H-NMR (400 MHz, MeOD) δ = 8,29-8,27 (Dimethylamino)-1-piperidyl]-4- (m, 1H), 8,17 (d, J = 6,0 Hz, 1H), 8,07-8,05 Pyridyl]phenyl]thiazol-2-yl]amino]-2- (m, 1Η), 7,78-7,73 (m, 3Η), 7,59-7,56 (m, Oxoethyl]benzamide 2H), 7.46 (s, 1H), 7.31-7.30 (m, 1H), 6.87- 6,83 (m, 2Η), 4,50-4,47 (m, 1Η), 4,27 (s, 2H), 4.09-4.05 (m, 1H), 3.68-3.63 (m, 1H), 3.54-3.47 (m, 1H), 3.39-3.34 (m, 1H), 3.00 (s, 6Η), 2,30-2,26 (m, 1Η), 2,08-1,77 (m, 3Η) ppm 226 4-Amino-N-[2-[[4-[3-[2-[1,1-dimethyl-2- 529,3 1H-NMR (400 MHz, DMSO) δ = 12.48- (Methylamino)-2-oxo-ethyl]-4- 12,23 (m, 1Η), 8,61 (d, J = 5,4 Hz, 1Η), Pyridyl]phenyl]thiazol-2-yl]amino]-2- 8,47-8,37 (m, 1Η), 8,24 (s, 1Η), 8,01 (d, J = Oxoethyl]benzamide 7,8 Hz, 1H), 7,82-7,68 (m, 2H), 7,66-7,55 (m, 5H), 7,40 (br d, J = 4,4 Hz, 1H), 6,57 (d, J = 8,8 Hz, 2H), 5,67 (s, 2H), 4,13 (br d, J=5.4Hz, 2H), 2.58 (d, J=4.4Hz, 3H), 1,56 (s, 6Η) ppm 227 4-Amino-N-[2-oxo-2-[[4-(3-pyrimidin-4- 431,2 1H-NMR (400 MHz, DMSO) δ = 9.30 (s, ylphenyl)thiazole-2- 1H), 8.90 (d, J = 5.2 Hz, 1 H), 8.82 (s, 1 H), il]amino]ethyl]benzamide 8,45-8,43 (m, 1Η), 8,20-8,04 (m, 3Η), 7,80 (s, 1H), 7,62 (d, J = 8,8 Hz, 3H), 6,57 (d, J = 8.4Hz, 2H), 5.67(s, 2H), 4.13(d, J=6.0 Hz, 2Η) ppm 228 4-Amino-N-[2-[[4-(1-methylindazole-6- 407,0 1H-NMR (400 MHz, DMSO-d6) δ = 12,39 (s, yl)thiazol-2-yl]amino]-2-oxo- 1H), 8.45-8.43 (m, 1H), 8.11 (s, 1H), 8.04 Ethyl]benzamide (d, J = 0,8 Hz, 1Η), 7,82-7,76 (m, 1Η), 7.75–7.67 (m, 2H), 7.62 (d, J=8.8Hz, 2Η), 6,57 (d, J = 8,4 Hz, 2Η), 5,68 (s, 2Η), 4,13 (d, J = 5,6 Hz, 2H), 4,07 (s, 3H) ppm 229 4-Amino-N-[2-[[4-[3-[1-(2-hydroxy-2- 491,4 1H-NMR (400 MHz, DMSO-d6) δ = 12,37 Methyl-propyl)pyrazole-3- (br s, 1Η), 8,44-8,43 (m, 1Η), 8,36 (s, 1Η), yl]phenyl]thiazol-2-yl]amino]-2-oxo- 7,81 (d, J = 7,6 Hz, 1Η), 7,75-7,72 (m, Ethyl]benzamide 2H), 7.67 (s, 1H), 7.63 (d, J = 8.6 Hz, 2H), 7,45-7,44 (m, 1Η), 6,75 (d, J = 2,0 Hz, 1Η), 6.57 (d, J=8.6Hz, 2H), 5.66 (s, 2H), 4,92-4,56 (m, 1Η), 4,13 (d, J = 5,6 Hz, 2H), 4.09 (s, 2H), 1.12 (s, 6H). 230 4-Amino-N-[2-oxo-2-[[4-[3- 467,0 1 H-NMR (400 MHz, DMSO) δ (ppm) = (Tetrahydropyran-4- 12,33 (br s, 1Η), 8,43-8,40 (m, 1Η), 7,88 yloxymethyl)phenyl]thiazole-2- (s, 1H), 7,81 (d, J = 7,6 Hz, 1H), 7,62-7,59 il]amino]ethyl]benzamide (m, 3H), 7,42-7,38 (m, 1H), 7,30 (d, J = 7,6 Hz, 1Η), 6,57 (d, J=8,4Hz, 2Η), 5,66 (s, 2Η), 4,58 (s, 2Η), 4,12 (d, J = 5,6 Hz, 2Η), 3.85-3.80 (m, 2H), 3.63-3.56 (m, 1H), 3.40- 3,35 (m, 2Η), 1,99-1,85 (m, 2Η), 1,55- 1.41 (m, 2H) 231 4-Amino-N-[2-[[4-[3-(3-Methylisoxazole- 434,2 1H-NMR (400 MHz, DMSO-d6) δ = 12,36 5-yl)phenyl]thiazol-2-yl]amino]-2-oxo- (s, 1Η), 8,43 (t, J = 6,0 Hz, 1Η), 8,37 (s, Ethyl]benzamide 1 H), 8.01 (d, J = 8.0 Hz, 1 H), 7.80-7.77 (m, 2Η), 7,64-7,59 (m, 3Η), 6,92 (s, 1Η), 6.57 (d, J=8.4Hz, 2H), 5.66 (s, 2H), 4.12 (d, J = 6,0 Hz, 2H), 2,31 (s, 3H) 232 4-Amino-N-[2-[[4-[3-[1-(2-methoxy-2- 505,1 1 H-NMR (400 MHz, DMSO) δ (ppm) = Methyl-propyl)pyrazole-3- 12.48-12.30 (m, 1H), 8.43-8.40 (m, 1H), yl]phenyl]thiazol-2-yl]amino]-2-oxo- 8,34 (s, 1Η), 7,83-7,78 (m, 1Η), 7,73 (d, J = Ethyl]benzamide 8,0 Hz, 1Η), 7,69 (d, J = 2,4 Hz, 1Η), 7,67 (s, 1H), 7,61 (d, J=8,7Hz, 2H), 7,44 (t, J = 7,8 Hz, 1H), 6,74 (d, J = 2,3 Hz, 1H), 6.56 (d, J=8.4Hz, 2H), 5.66 (s, 2H), 4.18 (s, 2H), 4,12 (d, J=6,0Hz, 2H), 3,20 (s, 3H), 1,12 (s, 6H) 233 (S)-4-Amino-N-(4-(dimethylamino)-1- 501,3 1H NMR (400MHz, DMSO) δ = 8.73-8.64 Oxo-1-((4-(3-(Pyridin-4- (m, 2Η), 8,52 (d, J = 7,0 Hz, 1Η), 8,31 (s, yl)phenyl)thiazol-2-yl)amino)butan-2- 1Η), 8,02 (d, J = 7,6 Hz, 1Η), 7,82 (s, 1Η), il) Benzamide 7.79-7.72 (m, 3H), 7.65-7.56 (m, 3H), 6.57 (d, J = 8,8 Hz, 2H), 5,68 (s, 2H), 4,64 (br d, J = 5,6 Hz, 1Η), 2,42-2,36 (m, 2Η), 2,20 (s, 6Η), 2,03-1,89 (m, 2Η) ppm 234 4-Amino-N-[2-[[4-[3-[(3R)-3-hydroxy-1-) 452,0 11H-NMR (400 MHz, DMSO-d6) δ = 12,35 Piperidyl]phenyl]thiazol-2-yl]amino]-2- (s, 1H) 8.48 (m, 1H), 7.58-7.67 (m, 4H), Oxoethyl]benzamide 7,40 (br s, 1Η), 7,31 (m, 1Η), 7,02 (m, 1Η), 6.64 (d, J=8.4Hz, 2H), 4.12 (m, 2H), 3.58 (m, 3Η), 2,83 (br s, 1Η), 2,68-2,74 (m, 1Η), 1,80-1,96 (m, 2Η), 1,61 (m, 1Η), 1,36 (m, 1Η) ppm 235 4-Amino-N-[2-oxo-2-[[4-[3-(1- 481.1 1H-NMR (400 MHz, DMSO-d6) δ = 12,32 Tetrahydropyran-4- (br s, 1Η), 8,43 (m, 1Η), 7,90 (s, 1Η), yloxyethyl)phenyl]thiazole-2- 7,79 (d, J = 7,6 Hz, 1Η), 7,66-7,57 (m, 3Η), il]amino]ethyl]benzamide 7,40 (m, 1H), 7,30 (d, J = 7,6 Hz, 1H), 6,57 (d, J=8,8Hz, 2H), 5,68 (s, 2H), 4,70 (m, 1H), 4.12 (m, 2H), 3.86-3.68 (m, 2H), 3,41–3,39 (m, 1H), 3,30–3,24 (m, 2H), 1.92 (m, 1H), 1.71-1.61 (m, 1H), 1.50- 1,31 (m, 5Η) ppm 236 4-Amino-N-[2-[[4-[3-[(3S)-3-hydroxy-1- 452.1 1H-NMR (400 MHz, DMSO-d6) δ = 12,33- Piperidyl]phenyl]thiazol-2-yl]amino]-2- 12.32 (m, 1H), 8.42-8.39 (m, 1H), 7.62- Oxoethyl]benzamide 7,58 (m, 3Η), 7,47 (s, 1Η), 7,28-7,21 (m, 2Η), 6,88-6,86 (m, 1Η), 6,56 (d, J=6,8Hz, 2H), 5.66 (s, 2H), 4.85 (d, J = 2.0 Hz, 1H), 4.11 (d, J=5.6Hz, 2Η), 3.62-3.51 (m, 2Η), 2.68-2.66 (m, 1H), 2.71-2.66 (m, 2H), 1.93-1.89 (m, 1H), 1.79-1.74 (m, 1H), 1.59-1.50 (m, 1H), 1.33-1.24 (m, 1H) ppm 237 4-Amino-N-[(1S)-1-[[4-[3-[3- 532,3 1H RMN (400 MHz, METANOL-d4) δ = (Aminomethyl)phenyl]phenyl]thiazole-2- 8,21 (s, 1H), 8,09 (d, J=8,8Hz, 2H), 7,92 il]carbamoil]-3-methylsulfanil- (d, J = 7,6 Hz, 1H), 7,82 (s, 1H), 7,77 (d, J = Propyl]benzamide 8,0 Hz, 1H), 7,64 (d, J = 8,0 Hz, 1H), 7.58-7.46 (m, 6H), 4.95-4.92 (m, 1H), 4,22 (s, 2Η), 2,76-2,65 (m, 2Η), 2,36- 2,18 (m, 2Η), 2,14 (s, 3Η) ppm 238 4-Amino-N-[2-[[4-[3-[(3S)-3-methoxy-1- 466,0 1H-NMR (400 MHz, DMSO-d6) δ = 12,29 Piperidyl]phenyl]thiazol-2-yl]amino]-2- (s, 1Η), 8,41-8,38 (s, 1Η), 7,60 (d, J = 8,4, Oxoethyl]benzamide 3H), 7.46 (s, 1H), 7.30-7.22 (m, 2H), 6.92- 6,89 (m, 1Η), 6,56 (d, J = 8,4, 2Η), 5,65 (s, 2Η), 4,10 (d, J=5,6, 2Η), 3,69-3,65 (m, 1H), 3.47-3.43 (m, 1H), 3.37-3.35 (m, 1H), 3,29 (s, 3H), 2,84-2,77 (1H), 2,73-2,68 (m, 1H), 2,02-1,98 (m, 1H), 1,82-1,77 (m, 1 H), 1.57-1.52 (s.1 H), 1.38-1.28 (m, 1Η) ppm. 239 4-Amino-N-[2-oxo-2-[[4-(3- 437,2 1H-NMR (400 MHz, DMSO-d6) δ = 12,36 Tetrahydropyran-2-ylphenyl)thiazol-2- (br s, 1Η), 8,46 (br s, 1Η), 7,91 (br s, 1Η), il]amino]ethyl]benzamide 7.76 (d, J=7.2Hz, 1H), 7.65 (s, 2H), 7.59 (s, 1Η), 7,36 (m, 1Η), 7,24 (d, J = 6,8 Hz, 1Η), 6,71-6,51 (m, 2Η), 4,34 (d, J = 10,8 Hz, 1Η), 4,13 (s, 2Η), 4,08-3,98 (m, 2Η), 1,96-1,75 (m, 2Η), 1,71-1,32 (m, 4Η) 1H RMN (400MHz, DMSO + D2 O) δ = 7,88 (br s, 1Η), 7,74 (d, J = 7,6 Hz, 1Η), 7,65 (d, J = 8.0 Hz, 2H), 7.55 (s, 1H), -7.41 7,30 (m, 1H), 7,23 (d, J = 7,6 Hz, 1H), 6,70 (d, J=8.4Hz, 2H), 4.32 (d, J=10.8Hz, 1Η), 4,12 (s, 2Η), 4,01 (d, J=11,2Hz, 1H), 3.59-3.42 (m, 1H), 1.90-1.75 (m, 2H), 1.70-1.35 (m, 4H) 19F-NMR (376 MHz, DMSO + D2O) δ = −74,24 (s) 240 4-Amino-N-[2-oxo-2-[(4-quinazolin-7- 405,3 1H-NMR (400 MHz, DMSO-d6) δ = 9,58 (s, Illthiazol-2-yl)amino]ethyl]benzamide 1Η), 9,29 (s, 1Η), 8,50 (s, 1Η), 8,45 (t, J = 6,0 Hz, 1Η), 8,33-8,31 (m, 1Η), 8,21 (d, J = 8,4 Hz, 1H), 8,10 (s, 1H), 7,62 (d, J = 8.4Hz, 2H), 6.56 (d, J=8.4Hz, 2H), 5,67 (s, 2H), 4,14 (d, J=6,0Hz, 2H) 241 4-Amino-N-[2-[[4-[3-[1-(2-amino-2- 490,1 1H-NMR (400 MHz, DMSO-d6) δ = 12,34 Methyl-propyl)pyrazole-3- (s, 1Η), 8,44-8,43 (m, 1Η), 8,33 (s, 1Η), yl]phenyl]thiazol-2-yl]amino]-2-oxo- 8,00 (br s, 3H), 7,88-7,79 (m, 3H), 7,69- Ethyl]benzamide 7,64 (m, 1H), 7,61 (d, J=8,6Hz, 2H), 7,50- 7,48 (m, 1Η), 6,85 (d, J = 2,2 Hz, 1Η), 6,56 (d, J = 8,7 Hz, 2H), 5,66 (br s, 2H), 4,33 (s, 2Η), 4,12 (br d, J = 5,7 Hz, 2Η), 1,26 (s, 6Η)

Example 88. Assay for BRM and BRG-1 ATPase catalytic activity

The ATPase catalytic activity of BRM or BRG-1 was measured by an in vitro biochemical assay using ADP-Glo™ (Promega, V9102). The ADP-Glo™ kinase assay is performed in two steps once the reaction is complete. The first step is to consume unused ATP in the reaction. The second step is the conversion of the reaction product, ADP, to ATP, which is used by luciferase to generate luminescence and recognized by a luminescence reader such as Envision.

The assay reaction mixture (10 µL) contains 30 nM BRM or BRG-1, 20 nM salmon sperm DNA (from Invitrogen, UltraPure™ Salmon Sperm DNA Solution, Cat. No. 15632011) and 400 µM ATP in the ATPase assay buffer consisting of 20 mM Tris, pH 8, 20 mM MgCl2, 50 mM NaCl, 0.1% Tween-20, and 1 mM fresh DTT (Pierce™ DTT (Dithiothreitol), Cat # 20290). The reaction is prepared by adding 2.5 µl of ATPase solution to 2.5 µl of ATP/DNA solution in a small volume Proxiplate 384 Plus white plate (Perkin Elmer, Cat# 6008280) and incubating for 1 hour at room temperature. The reaction is then incubated for 40 min at room temperature after the addition of 5 µl of the ADP-Glo™ reagent provided in the kit. Then 10 µL of the kinase detection reagent provided in the kit is added to convert ADP to ATP and the reaction is incubated at room temperature for 60 min. Finally, the luminescence reading is obtained with a plate-meter photometer such as the Envision.

BRM and BRG-1 were synthesized from five insect cell lines with greater than 90% purity. IP50Data for compounds 1-241 from the ATPase catalytic activity assay described herein are presented in Tables 5 and 6 below.

TABLE 5 is made up BRM-IP50 BRM max % IP BRG150 BRG1 Max % NO. (μM)* inhibition (μM)* inhibition 1 ++ >95 % ++ >70 % 2 +++ >95 % ++ >95 % 3 ++ >95 % ++ >90 % 4 ++ >90 % + >70 % 5 + >90 % + >70 % 6 ++ >90 % + >60 % 7 ++ >90 % ++ >60 % 8 ++ >90 % + >70 % 9 +++ >90 % +++ >60 % 10 + >90 % + >90 % 11 ++ >80 % + >70 % 12 ++ >80 % + >60 % 13 ++ >80 % ++ >30 % 14 ++ >80 % + >60 % 15 ++ >80 % ++ >60 % 16 ++ >80 % + >60 % 17 ++ >80 % + >60 % 18 + >80 % + >60 % 19 +++ >70 % + >30 % 20 + >70 % + >60 % 21 + >70 % + >60 % 22 + >70 % + >60 % 23 ++ >70 % ++ >50 % 24 ++ >70 % NT >99 % 25 + >70 % + >50 % 26 + >60 % + <30 % 27 + >60 % + >30 % 28 ++ >50 % ++ >30 % 29 + >50 % + >60 % 30 NT >70 % NT >50 % 31 ++ >30 % ++ <30 % 32 + >30 % + <30 % 33 + >80 % + >80 % 34 + >90 % + >60 % 35 + >95 % + >90 % 36 +++ >95 % ++ >90 % 37 + >50 % + >30 % 38 + >80 % + >80 % 39 + >30 % + <30 % 40 NT >50 % NT <30 % 41 ++ >90 % ++ >80 % 42 ++ >90 % ++ >80 % 43 + >80 % + >70 % 44 + >70 % + >70 % 45 + NT + NT 46 ++ >60 % ++ >70 % 47 + >70 % + >70 % 48 + >50 % + >30 % 49 ++ >50 % ++ >30 % 50 + >30 % + >30 % 51 ++ >90 % + >80 % 52 +++ >95 % +++ >95 % 53 ++ >95 % ++ >90 % 54 ++ >90 % ++ >90 % 55 ++ >90 % ++ >80 % 56 + >90 % + >80 % 57 + >90 % + >90 % 58 ++ >90 % ++ >90 % 59 + >90 % + >90 % 60 + >90 % + >80 % 61 ++ >80 % ++ >50 % 62 ++ >80 % ++ >70 % 63 + >80 % + >80 % 64 ++ >70 % ++ >60 % 65 ++ >70 % ++ >30 % 66 + >70 % + >50 % 67 + >70 % + >70 % 68 + >60 % + >60 % 69 + >60 % + >60 % 70 + >60 % + >50 % 71 ++ >60 % + >50 % 72 + >60 % + >50 % 73 +++ >50 % ++ >30 % 74 + >50 % + >30 % 75 + >50 % + >60 % 76 + >30 % + >30 % 77 + >30 % + >30 % 78 + NT + NT 79 +++ >30 % ++ >30 % 80 +++ >95 % +++ >95 % 81 +++ >95 % +++ >95 % 82 ++ >95 % ++ >95 % 83 ++ >90 % + >80 % 84 + >80 % + >80 % 85 ++ >95 % ++ >90 % 86 + >70 % + >70 % "+" indicates inhibitory effect >10 µM. “++” indicates an inhibitory effect of 1–10 μM. "+++" indicates inhibitory effect <1 µM. "NT" means not tested

TABLE 6 is made up BRM-IP50 BRM max % IP BRG150 BRG1 Max % NO. (μM)* inhibition (μM)* inhibition 87 ++++ >95 % ++++ >95 % 88 +++ >95 % ++ >95 % 89 ++++ >80 % +++ >80 % 90 ++++ >70 % ++++ >50 % 91 +++ >95 % ++ >90 % 92 +++ >90 % +++ >80 % 93 +++ >90 % +++ >90 % 94 +++ >95 % ++ >90 % 95 ++++ >95 % ++++ >95 % 96 +++ >70 % ++ >70 % 97 +++ >70 % +++ >70 % 98 +++ >90 % +++ >70 % 99 ++++ >90 % ++++ >95 % 100 +++ >80 % ++ >80 % 101 +++ >95 % +++ >95 % 102 +++ >80 % +++ >80 % 103 +++ >70 % ++ >80 % 104 ++++ >70 % ++++ >60 % 105 +++ >80 % +++ >50 % 106 +++ >70 % +++ >60 % 107 +++ >95 % ++ >95 % 108 +++ >80 % +++ >90 % 109 +++ >90 % +++ >90 % 110 +++ >80 % ++ >80 % 111 +++ >90 % +++ >90 % 112 +++ >90 % +++ >80 % 113 +++ >95 % +++ >95 % 114 +++ >80 % ++ >80 % 115 +++ >90 % +++ >90 % 116 +++ >95 % +++ >95 % 117 ++++ >95 % ++++ >95 % 118 +++ >90 % ++ >90 % 119 +++ >95 % ++ >95 % 120 +++ >90 % +++ >90 % 121 +++ >95 % +++ >95 % 122 +++ >95 % ++ >90 % 123 +++ >80 % +++ >80 % 124 +++ >95 % +++ >95 % 125 ++++ >95 % ++++ >95 % 126 ++++ >80 % ++++ >80 % 127 +++ >90 % +++ >80 % 128 ++++ >95 % ++++ >95 % 129 +++ >95 % +++ >95 % 130 +++ >70 % ++ >70 % 131 ++++ >95 % ++++ >95 % 132 ++++ >80 % ++++ >70 % 133 ++++ >95 % ++++ >95 % 134 +++ >50 % +++ >50 % 135 ++++ >95 % ++++ >95 % 136 +++ >95 % +++ >95 % 137 +++ >90 % +++ >95 % 138 +++ >90 % +++ >90 % 139 ++++ >95 % ++++ >95 % 140 +++ >95 % +++ >90 % 141 ++++ >95 % +++ >95 % 142 +++ >90 % +++ >90 % 143 ++++ >95 % ++++ >95 % 144 +++ >95 % +++ >95 % 145 +++ >90 % +++ >90 % 146 ++++ >95 % ++++ >95 % 147 ++++ >95 % ++++ >95 % 148 ++++ >95 % ++++ >95 % 149 ++++ >80 % ++++ >80 % 150 ++++ >95 % ++++ >90 % 151 +++ >80 % +++ >80 % 152 +++ >90 % +++ >80 % 153 ++++ >90 % ++++ >80 % 154 ++++ >95 % ++++ >95 % 155 +++ >80 % +++ >80 % 156 +++ >95 % +++ >90 % 157 ++++ >95 % ++++ >90 % 158 +++ >90 % +++ >95 % 159 +++ >90 % +++ >95 % 160 +++ >95 % +++ >95 % 161 +++ >70 % +++ >50 % 162 ++++ >95 % ++++ >95 % 163 ++++ >95 % +++ >95 % 164 ++++ >95 % ++++ >95 % 165 ++++ >95 % ++++ >95 % 166 +++ >95 % +++ >95 % 167 +++ >95 % +++ >95 % 168 ++++ >95 % ++++ >95 % 169 +++ >95 % +++ >95 % 170 +++ >95 % +++ >95 % 171 +++ >95 % +++ >95 % 172 +++ >95 % +++ >90 % 173 +++ >90 % +++ >90 % 174 +++ >80 % +++ >80 % 175 +++ >90 % +++ >80 % 176 +++ >90 % +++ >80 % 177 ++++ >95 % ++++ >95 % 178 ++++ >95 % ++++ >95 % 179 +++ >90 % +++ >95 % 180 +++ >90 % +++ >95 % 181 ++++ >95 % ++++ >95 % 182 ++++ >95 % +++ >90 % 183 ++++ >95 % ++++ >95 % 184 ++++ >80 % ++++ >70 % 185 +++ >80 % +++ >70 % 186 +++ >95 % +++ >95 % 187 +++ >70 % +++ >60 % 188 ++++ >95 % ++++ >80 % 189 +++ >90 % +++ >90 % 190 +++ >80 % +++ >80 % 191 ++++ >95 % +++ >95 % 192 +++ >90 % +++ >90 % 193 +++ >80 % +++ >90 % 194 +++ >90 % +++ >80 % 195 ++++ >95 % ++++ >95 % 196 +++ >95 % +++ >90 % 197 ++++ >90 % ++++ >80 % 198 +++ >80 % +++ >60 % 199 +++ >95 % +++ >95 % 200 ++++ >95 % ++++ >95 % 201 ++++ >95 % ++++ >90 % 202 ++++ >95 % ++++ >95 % 203 +++ >80 % +++ >80 % 204 +++ >80 % +++ >70 % 205 ++++ >95 % ++++ >95 % 206 +++ >95 % +++ >95 % 207 ++++ >95 % ++++ >95 % 208 +++ >90 % +++ >80 % 209 ++++ >95 % ++++ >95 % 210 +++ >95 % +++ >80 % 211 +++ >95 % +++ >90 % 212 +++ >95 % +++ >95 % 213 ++++ >95 % ++++ >95 % 214 ++++ >95 % ++++ >95 % 215 +++ >90 % +++ >90 % 216 ++++ >90 % +++ >95 % 217 ++++ >90 % ++++ >90 % 218 ++++ >95 % +++ >90 % 219 ++++ >95 % ++++ >95 % 220 +++ >95 % ++ >80 % 221 +++ >95 % +++ >80 % 222 +++ >95 % +++ >80 % 223 ++++ >95 % ++++ >90 % 224 ++++ >95 % ++++ >90 % 225 +++ >80 % +++ >90 % 226 ++++ >95 % ++++ >95 % 227 ++++ >95 % ++++ >95 % 228 +++ >90 % +++ >90 % 229 +++ >95 % +++ >95 % 230 +++ >95 % +++ >90 % 231 +++ >90 % +++ >90 % 232 +++ >80 % +++ >80 % 233 +++ >90 % +++ >80 % 234 +++ >90 % +++ >90 % 235 +++ >90 % +++ >70 % 236 +++ >90 % +++ >90 % 237 +++ >90 % +++ >90 % 238 +++ >90 % +++ >95 % 239 +++ >90 % +++ >80 % 240 +++ >80 % +++ >80 % 241 +++ >90 % +++ >90 % "+" indicates inhibitory effect >10 µM. “++” indicates an inhibitory effect of 1–10 μM. "+++" indicates an inhibitory effect of 0.1-1 µM. "++++" indicates an inhibitory activity <0.1 µM

Example 89. Effects of BRG1/BRM ATPase Inhibition on the Growth of Choroidal Melanoma and Hematological Cancer Cell Lines

Methods: Uveal melanoma cell lines (92-1, MP41, MP38, MP46), prostate cancer cell lines (LNCAP), lung cancer cell lines (NCIH1299), and immortalized embryonic kidney lines (HEK293T) were seeded by growth in 96-well plates (see Table 7). The BRG1/BRM ATPase inhibitor, compound 87, was dissolved in DMSO and added to the cells at the time of plating in a concentration gradient from 0 to 10 µM. Cells were incubated at 37°C for 3 days. After three days of treatment, the medium was removed from the cells and 30 μl of TrypLE (Gibco) was added to the cells for 10 min. Cells were detached from the plates and resuspended by adding 170 microliters of growth medium. Cells from two DMSO-treated control wells were counted and the original number of cells plated at the start of the experiment were replated on fresh compound plates for an additional four days at 37 degrees Celsius. On day 7, cells were harvested as described above. On day 3 and day 7, relative cell growth was measured by adding Glo cell titer (Promega) and luminescence was measured in an Envision plate reader (Perkin Elmer). The concentration of compound at which the growth of each cell line was inhibited by 50% (Eq50), calculated with Graphpad Prism and shown below. For multiple myeloma cell lines (OPM2, MM1 S, LP1), ALL cell lines (TALL1, JURKAT, RS411), DLBCL cell lines (SUDHL6, SUDHL4, DB, WSUDLCL2, PFEIFFER), AML cell lines (OCIAML5), MDS, (SKM1), ovarian cancer cell lines (OV7, TYKNU), esophageal cancer cell lines (KYSE150), rhabdomyosarcoma cell lines (RD, G402, G401, HS729, A204), liver cancer (HLF, HLE, PLCRPF5) and lung cancer cell lines (SW1573, NCIH2444) performed the above methods with the following modifications: cells were seeded in 96-well plates, and the next day the BRG1/BRM ATPase inhibitor, compound 87, was dissolved in DMSO and added to the cells in gradient concentration from 0 to 10 micromolar. At the time of cell division on days 3 and 7, cells were split into new 96-well plates and new compound was added four hours after replating.

Table 7 lists the cell lines tested and the growth media used.

TABLE 7 cell lines and growth media cell line Those means of development 92-1 Sigma RPMI1640 + 20 % FBS A204 ATCC McCoys 5A + 10 % FBS Database ATCC RPMI1640 + 10 % FBS G401 ATCC McCoys 5A + 10 % FBS G402 ATCC McCoys 5A + 10 % FBS HEK293T ATCC DMEM + 10 % FBS PLEASE JCRB DMEM + 10 % FBS HLF JCRB DMEM + 10 % FBS HS729 ATCC DMEM + 10 % FBS JURKAT ATCC RPMI1640 + 10 % FBS KISS150 DSMZ RPMI1640/Ham's F12 + 10 % FBS LNCAP ATCC RPMI1640 + 10 % FBS LP1 DSMZ IMDM + 20 % FBS MM1S ATCC RPMI1640 + 10 % FBS MP38 ATCC RPMI1640 + 20 % FBS MP41 ATCC RPMI1640 + 20 % FBS MP46 ATCC RPMI1640 + 20 % FBS NCIH1299 ATCC RPMI1640 + 10 % FBS NCIH2444 ATCC RPMI1640 + 20 % FBS OCIAML5 DSMZ alfa-MEM + 20 % FBS + 10 ng/ml GM-CSF OPM2 DSMZ RPMI1640 + 10 % FBS OV7 etc DMEM/Ham's F12 (1:1) + 2 mM Glutamine + 10 % FBS + 0,5 μg/ml Hydrocortison + 10 µg/ml Insulin PFEIFFER ATCC RPMI1640 + 10 % FBS PLCPRF5 ATCC EMEM + 10% FBS RD ATCC DMEM + 10 % FBS RS411 ATCC RPMI1640 + 10 % FBS SKM1 JCRB RPMI1640 + 10 % FBS SUDHL4 DSMZ RPMI1640 + 10 % FBS SUDHL6 ATCC RPMI1640 + 20 % FBS SW1573 ATCC DMEM + 10 % FBS NUMBER 1 JCRB RPMI1640 + 10 % FBS TYKNU JCRB EMEM + 20% FBS WSUDLCL2 DSMZ RPMI1640 + 10 % FBS

Results: As shown in FIGFIGO.1, choroidal melanoma and hematological cancer cell lines were more sensitive to BRG1/BRM inhibition than the other cell lines tested. Inhibition of choroidal melanoma and hematologic cancer cell lines persisted until day 7.

Example 90. Comparison of BRG1/BRM inhibitors with clinical PKC and MEK inhibitors in uveal melanoma cell lines

Methods: Uveal melanoma cell lines, 92-1 or MP41, were plated in 96-well plates in the presence of growth medium (see Table 7). BAF-ATPase inhibitor (Compound 87), PKC inhibitor (LXS196, MedChemExpress), or MEK inhibitor (Selumetinib, Selleck Chemicals) were dissolved in DMSO and added to cells at the time of plating in a concentration gradient from 0 to 10 micromolar. Cells were incubated at 37°C for 3 days. After three days of treatment, cell growth was measured by Cell-Titer Glow (Promega) and luminescence was read on an Envision plate reader (Perkin Elmer).

Results: As shown in FIGFIGO.2Compound 87 showed comparable growth inhibition of choroidal melanoma cells as clinical inhibitors of PKC and MEK. Furthermore, compound 87 was found to result in a faster onset of inhibition than clinical PKC and MEK inhibitors.

Example 91. Composition of Union B

The BRG1/BRM B inhibitory compound has the structure:

Compound B was synthesized as shown in Scheme 2 below.

Step 1: Preparation of (S)-1-(Methylsulfonyl)-N-(4-(methylthio)-1-oxo-1-((4-(3-(pyridin-4-yl)phenyl)thiazol-2-yl) )amino)butan-2-yl)-1H-pyrrole-3-carboxamide (Compound B)

In a mixture of (2S)-2-amino-4-methylsulfanyl-N-[4-[3-(4-pyridyl)phenyl]thiazol-2-yl]butanamide (2 g, 4.75 mmol, HCl salt) and 1 -methylsulfonylpyrrole-3-carboxylic acid (898.81 mg, 4.75 mmol) in DMF (20 mL) was added to EDCI (1.37 g, 7.13 mmol), HOBt (962.92 mg, 7.13 mmol ) and DIEA (2,400 mmol). 3.31 ml) and the mixture was stirred at 25°C for 3 hours. The mixture was poured into H2(100 mL) and the precipitate was collected by filtration. The solid was triturated in MeOH (20 mL) and the precipitate was collected by filtration. The solid was dissolved in DMSO (10 mL) and then the mixture was poured into MeOH (50 mL) and the precipitate formed was collected by filtration and lyophilized to give compound B (2.05 g, 3.66 mmol, 77.01 % yield) as a white solid. LCMS (ESI) m/z [M+H]+=555,9.1H-NMR (400 MHz, DMSO) δ 12,49 (s, 1Η), 8,68-8,66 (m, 2Η), 8,46 (d, J=7,2 Hz, 1Η), 8, 31 -8,30 (m, 1Η), 8,02-8,00 (m, 1Η), 7,94-7,96 (m, 1Η), 7,83 (s, 1Η), 7,73- 7,74 (m, 3Η), 7,61–7,57 (m, 1Η), 7,31–7,29 (m, 1Η), 6,79–6,77 (m, 1Η), 4, 74 -4,69 (m, 1Η), 3,57 (s, 3Η), 2,67-2,53 (m, 2Η), 2,13-2,01 (m, 5Η). SFC: AS-3-MeOH (DEA)-40-3 mL-35T.Icm, t=0,932 min, ee %=100 %.

Example 92. Effects of BRG1/BRM ATPase Inhibition on the Growth of Choroidal Melanoma, Hematologic Cancer, Prostate Cancer, Breast Cancer, and Ewing Sarcoma Cell Lines

Methods: All cell lines described above in Example 89 were also tested with Compound B as described above. In addition, the following cell lines were also tested as follows. Briefly, for Ewing sarcoma cell lines (CADOES1, RDES, SKES1), retinoblastoma cell lines (WERIRB1), ALL cell lines (REH), AML cell lines (KASUMI1), prostate cancer cell lines (PC3, DU145, 22RV1 Cells) Lines (SH4, SKMEL28, WM115, COLO829, SKMEL3, A375), Breast Cancer Cell Lines (MDAMB415, CAMA1, MCF7, BT474, HCC1419, DU4475, BT549), B-ALLBB125 Cell Lines, (CSUMLP Cell Lines) MEG01 ), Burkitt lymphoma cell lines (RAMOS2G64C10, DAUDI), mantle cell lymphoma cell lines (JEKO1, REC1), bladder cancer cell lines (HT1197) and lung cancer cell lines (SBC5), the above methods were performed with the following modifications: Cells were seeded in 96-well plates and the next day the BRG1/BRM ATPase inhibitor, compound B, was dissolved in DM10 and cells were added in a concentration gradient from 0 to 10 µM. At the time of cell division on days 3 and 7, cells were split into new 96-well plates and new compound was added four hours after replating.

Table 8 lists the cell lines tested and the growth media used.

cell line Those means of development 22RV1 ATCC RPMI1640 + 10 % FBS A375 ATCC DMEM + 10 % FBS BT474 ATCC Hybricare-Medium + 1,5 g/L Natrium Bicarbonate + 10% FBS BT549 ATCC RPMI1640 + 0.023 IU/ml Insulin + 10% FBS CADOES1 DSMZ RPMI1640 + 10 % FBS CAMA1 ATCC EMEM + 10% FBS COLO829 ATCC RPMI1640 + 10 % FBS DAVID ATCC RPMI1640 + 10 % FBS DU145 ATCC EMEM + 10% FBS DU4475 ATCC RPMI1640 + 10 % FBS HCC1419 ATCC RPMI1640 + 10 % FBS HT1197 ATCC EMEM + 10% FBS JEKO1 ATCC RPMI1640 + 20 % FBS K562 ATCC IMDM + 10 % FBS KASOUMI 1 ATCC RPMI1640 + 10 % FBS MCF7 ATCC EMEM + 0,01 mg/ml Rinderinsulin + 10% FBS MDAMB415 ATCC Leibovitz's L-15 + 2 mM L-Glutamina + 10 µg/ml Insulin + 10 µg/ml Glutathione + 15 % FBS MEG01 ATCC RPMI1640 + 10 % FBS PC3 ATCC F-12K + 10 % FBS RAMOS2G64C10 ATCC RPMI1640 + 10 % FBS RDES ATCC RPMI1640 + 15 % FBS REC1 ATCC RPMI1640 + 10 % FBS REH ATCC RPMI1640 + 10 % FBS SBC5 JCRB EMEM + 10% FBS SH4 ATCC DMEM + 10 % FBS SKES1 ATCC McCoys 5A + 15 % FBS SKMEL28 ATCC EMEM + 10% FBS SKMEL3 ATCC McCoys 5A + 15 % FBS SUPB15 ATCC IMDM + 4 mM L-Glutamine + 1.5 g/L Natriumbicarbonat + 0,05 mM 2-Mercaptoethanol + 20 % FBS WERIRB1 ATCC RPMI1640 + 10 % FBS WM115 ATCC EMEM + 10% FBS

Results: As shown in FIGFIGO.3, choroidal melanoma, hematologic cancer, prostate cancer, breast cancer, and Ewing sarcoma cell lines were more sensitive to BRG1/BRM inhibition than the other cell lines tested. Inhibition in choroidal melanoma, hematologic cancer, prostate cancer, breast cancer, and Ewing sarcoma cell lines persisted until day 7.

Other ways

While the invention has been described with respect to specific embodiments thereof, it should be understood that the invention may be further modified and this application is intended to cover any variations, uses or adaptations of the invention which generally follow the principles of the invention and including such Deviations from the present disclosure which are within the known or ordinary practice in the art to which the invention relates and are applicable to the essential features set forth herein and falling within the scope of the claims.

See the claims for further details.

References

Top Articles
Latest Posts
Article information

Author: Greg O'Connell

Last Updated: 06/03/2023

Views: 5821

Rating: 4.1 / 5 (62 voted)

Reviews: 85% of readers found this page helpful

Author information

Name: Greg O'Connell

Birthday: 1992-01-10

Address: Suite 517 2436 Jefferey Pass, Shanitaside, UT 27519

Phone: +2614651609714

Job: Education Developer

Hobby: Cooking, Gambling, Pottery, Shooting, Baseball, Singing, Snowboarding

Introduction: My name is Greg O'Connell, I am a delightful, colorful, talented, kind, lively, modern, tender person who loves writing and wants to share my knowledge and understanding with you.