WO2024011307A1 - Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1 and treatment of cancer - Google Patents

Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1 and treatment of cancer Download PDF

Info

Publication number
WO2024011307A1
WO2024011307A1 PCT/CA2023/050681 CA2023050681W WO2024011307A1 WO 2024011307 A1 WO2024011307 A1 WO 2024011307A1 CA 2023050681 W CA2023050681 W CA 2023050681W WO 2024011307 A1 WO2024011307 A1 WO 2024011307A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
compound
group
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/CA2023/050681
Other languages
French (fr)
Inventor
Marc Vidal
Maroua KHALIFA
Eric Campeau
Delphine Labit
Martin Maguire
Original Assignee
Thryv Therapeutics Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Thryv Therapeutics Inc. filed Critical Thryv Therapeutics Inc.
Publication of WO2024011307A1 publication Critical patent/WO2024011307A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast

Definitions

  • the technical field relates to pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives and pharmaceutical compositions that inhibit SGK-1 , and more particularly relates to pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives and pharmaceutical compositions for the treatment of heart conditions treatable by SGK-1 inhibition such as Long QT syndrome, and for the treatment of cancer.
  • Serine/threonine-protein kinase (also known as serum/glucocorticoid-regulated kinase 1) is a protein kinase that plays a role in a cell's response to stress. SGK-1 activates certain potassium, sodium, and chloride channels. For instance, SGK-1 is known to regulate the myo-inositol transporter during osmotic stress.
  • SGK-1 inhibitor for the treatment of heart conditions such as LOTS, and/or for the treatment of cancer.
  • Formula VII or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer or tumor or in the manufacture of a medicament for the treatment of cancer or tumor, wherein:
  • Y 1 is H or F
  • Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN;
  • W 1 is selected from the group consisting of H and halogen;
  • R 35 is H or methyl.
  • a pharmaceutical composition comprising one or more compounds or pharmaceutically acceptable salts thereof as defined herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, for use in the treatment of cancer or tumor or in the manufacture of a medicament for the treatment of cancer or tumor.
  • described herein is a combination comprising one or more compounds or pharmaceutically acceptable salts thereof as defined herein and at least one inhibitor of AKT, PI3K, and/or mTOR.
  • a combination comprising one or more compounds or pharmaceutically acceptable salts thereof as defined herein and at least one inhibitor of AKT, PI3K, mTOR, PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1, SOS2, SHP1 , and/or MAPK.
  • a combination comprising one or more compounds or pharmaceutically acceptable salts thereof as defined herein and ipatasertib, alpelisib, everolimus, lapatinib, AT13148, gedatolisib, BAY-1082439, GSK112212, dabrafenib, trametinib, erlotinib, and/or GSK2334470.
  • described herein is a combination comprising at least two compounds or pharmaceutically acceptable salts thereof as defined herein.
  • described herein is a use of the pharmaceutical composition defined herein or the combination defined herein for the treatment of cancer or tumor or for the manufacture of a medicament for the treatment of cancer or tumor.
  • described herein is a method for the treatment of cancer, comprising administering to a subject in need thereof one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein.
  • a method for the treatment of cancer or tumor in a subject comprising: i. determining that cells from said cancer or tumor (e.g., cancer or tumor sample or biopsy): have an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; have an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; have a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; have an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and//
  • cancer or tumor e.g., cancer or tumor sample or biopsy
  • the cancer or tumor has been predetermined as: having an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; having an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; having a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRA
  • described herein is a use of one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein, for the treatment of one or more side effects or toxicities caused by an anticancer therapy in a subject undergoing said anticancer therapy.
  • described herein is a method for treating one or more side effects or toxicities caused by an anticancer therapy, comprising administering to a subject undergoing said anticancer therapy one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein.
  • a method for monitoring the progression of an anticancer therapy in a subject that was previously administered with one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein wherein the method comprises determining in a sample from said subject: a loss of phosphorylation of NDRG1 ; an increase in phosphorylation of MAPK14 (i.e.
  • p38 an increase in phosphorylation of MAPK14 (i.e., p38); an increase in phosphorylation of heat shock protein family B1 (i.e., HSPB1 or HSP27); an increase in phosphorylation of tumor suppressor p53 (TP53); an increase in phosphorylation of ribosomal protein S6 kinase A1 (RSK1); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERKI); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERK1); and/or an increase in phosphorylation of AKT1
  • Figs. 1A and 1B is a graph (Fig. 1A) and a Dose-response matrix (Fig. 1B) showing synergistic combination of Compound 79 with AKT inhibitor Ipatasertib on the growth of JIMT-1 breast cancer cells.
  • Figs. 2A and 2B is a graph (Fig. 2A) and a Dose-response matrix (Fig. 2B) showing synergistic combination of Compound 79 with AKT inhibitor Capivasertib.
  • Figs. 3A and 3B is a graph (Fig. 3A) and a Dose-response matrix (Fig. 3B) showing synergistic combination of Compound 79 with AKT inhibitor MK-2206.
  • Fig. 4A is a Western blot showing inhibition of NDRG1 phosphorylation in PBMCs by Compound 79.
  • Fig. 4B a Western blot showing inhibition of NDRG1 phosphorylation in BT-459 breast cancer cells by Compound 79.
  • Fig. 5 is a series of Western blots showing inhibition of NDRG1 phosphorylation in ex vivo treated blood of healthy volunteers by Compound 84.
  • Fig. 6 is a series of Western blots showing NDRG1 is a direct target of SGK1 phosphorylation.
  • Figs. 7A-7B shows the effect of the combination of Compound 156 with the PIK3CA inhibitor Alpelisib (Fig. 7A) or the AKT inhibitor Ipatasertib (Fig. 7B) on the growth of JIMT- 1 breast cancer cells.
  • Figs. 8A-8B shows the effect of the combination of Compound 175 with the PIK3CA inhibitor Alpelisib (Fig. 8A) or the AKT inhibitor Ipatasertib (Fig. 8B) on the growth of JIMT- 1 breast cancer cells.
  • Figs. 9A-9C shows the effect of the combination of Compounds 79 (Fig. 9A), 175 (Fig. 9B), or 156 (Fig. 9C) with the PIK3CA inhibitor Alpelisib on the growth of BT-549 breast cancer cells.
  • Figs. 10A-10D shows the effect of the combination of Compound 79 with the mTOR inhibitor Everolimus (Fig. 10A), the HER2/EGFR inhibitor Lapatinib (Fig. 10B), the multi- AGC kinase inhibitor AT13148 (Fig. 10C), or the mT0R/PI3K inhibitor Gedatolisib (Fig. 10D) on the growth of JIMT-1 breast cancer cells.
  • Figs. 11A-11C shows the effect of the combination of Compound 79 with the mTOR inhibitor Everolimus (Fig. 11A), the PIK3a/b/g inhibitor BAY-1082439 (Fig. 11 B), or the MEK inhibitor GSK112212 (Fig. 11C) on the growth of BT-549 breast cancer cells.
  • Figs. 12A-12C shows the effect of the combination of Compounds 79 (Fig. 12A), 175 (Fig. 12B), or 156 (Fig. 12C) with the PIK3CA inhibitor Alpelisib on the growth of HT-29 colorectal cancer cells.
  • Figs. 13A-13C shows the effect of the combination of Compounds 79 (Fig. 13A), 175 (Fig. 13B), or 156 (Fig. 13C) with the BRAF inhibitor Dabrafenib on the growth of HT-29 colorectal cancer cells.
  • Figs. 14A-14D shows the effect of the combination of Compound 79 with the PI3K/mT0R inhibitor Gedatolisib (Fig. 14A), the MEK inhibitor Trametinib (Fig. 14B), the EGFR inhibitor Erlotinib (Fig. 14C), or the PDK1 inhibitor GSK2334470 (Fig. 14D) on the growth of HT-29 colorectal cancer cells.
  • Figs. 15A-15B shows the effect of the combination of Compound 79 with the BRAF inhibitor Dabrafenib (Fig. 15A) or the MEK inhibitor Trametinib (Fig. 15B) on the growth of 8305c anaplastic thyroid cancer cells.
  • Figs. 16A-16D shows the effect of the triple combination of Compound 79 (5 ⁇ M [Fig. 16A]; 2.5 ⁇ M [Fig. 16B]; 1.25 ⁇ M [Fig. 16C]; and 0.63 ⁇ M [Fig. 16D]) with the BRAF inhibitor Dabrafenib and the MEK inhibitor Trametinib on the growth of 8305c anaplastic thyroid cancer cells.
  • Figs. 17A-17F shows the results from Figs. 16A-16D at different concentrations of Compound 79 (5 ⁇ M [Fig. 17A], 2.5 ⁇ M [Fig. 17B], 1.25 ⁇ M [Fig. 17C], and 0.625 ⁇ M [Fig. 17D]) in combination with Dabrafenib (0.08 nM) and Trametinib (0.08 nM).
  • Figs. 17E-F show the results from Figs. 16A-16D at 5 ⁇ M of Compound 79 in combination with Dabrafenib (5 nM) and Trametinib (5 nM [Fig. 17E]) and 1.25 nM [Fig.17F],
  • Figs. 18A-18B shows the effect of the combination of Compound 79 with the BRAF inhibitor Dabrafenib (Fig. 18A) or the MEK inhibitor Trametinib (Fig. 18B) on the growth of A375 melanoma cancer cells.
  • Figs. 19A-19B shows the effect of the combination of Compound 79 with the MEK inhibitor Trametinib (Fig. 19A) or the EGFR inhibitor Erlotinib (Fig. 19B) on the growth of SW403 colorectal cancer cells.
  • Figs. 20A-20B shows the effect of the combination of Compound 79 with the BRAF inhibitor Dabrafenib (Fig. 20A) or the MEK inhibitor Trametinib (Fig. 20B) on the growth of RKO colorectal cancer cells that are resistant to Dabrafenib.
  • Figs. 21A-21S shows the proliferation results obtained from the 45 PIK3CA mutant cell lines screened after treatment with compound 79 alone, the PIK3CA inhibitor Alpelisib alone, or in combination. Graphs represent viability (%) with respect to concentration ( ⁇ M).
  • Fig. 22 shows the synergistic effect of the combination of compound 79 and the PIK3CA inhibitor Alpelisib alone on PIK3CA mutant cancer cell lines.
  • Cell lines were grouped according to the level of viability decreased when treated with the combination at different compound 79 concentrations.
  • Group 1 represents cell lines whereby there was >10% decrease in viability when combined with ⁇ 1 ⁇ M Compound 79;
  • Group 2 represents cell lines whereby there was a 5-10% decrease in viability when combined with ⁇ 1 ⁇ M Compound 79;
  • Group 3 represents cell lines whereby there was ⁇ 10% decrease in viability when with any concentration of compound 79; and
  • Group 1 represents cell lines whereby there was >10% decrease in viability when combined with >1 ⁇ M Compound 79;
  • Fig. 23 shows the enrichment of RAS/MAPK activating mutations in cell lines associated with synergy between Compound 79 and Alpelisib in inhibiting proliferation. Cell lines were grouped as shown in Fig. 22.
  • Figs. 24A-24D shows the luminal and basal breast cancer tumor signatures associated with poor synergy of the combination of compound 79 and Alpelisib.
  • Figs. 25A-25E shows the upregulation of ribosomal signatures that are enriched in cell lines that showed synergy when Compound 79 and Alpelisib were combined compared to cell lines that did not show synergy.
  • Figs. 26A-26B show the results of the Western Blot of the phosphorylation of ribosomal protein S6 upon incubation with Alpelisib and Compound 79 in HT-29 colorectal cancer cells at 4 hours and 24 hours (Fig. 26A) and at different concentrations of Compound 79 (Fig. 26B).
  • Figs. 27A-27B show the Kegg-Ribosome (Fig. 27A) and Reactome Translation initiation (Fig. 27B) activity scores in the grouped cell lines as described in Fig. 22.
  • Fig. 28A-28D shows the gene sets from MsigDB (The Molecular Signatures Database) (Fig. 28A) that are significantly upregulated in Group 1 vs. Group 3 cell lines, according to the grouping described in Fig. 22.
  • Figs. 28-C-28D correspond to the genes specified in Tables X-Z.
  • Figs. 29A-29D show the Anaplastic thyroid cancer (ATC) tumor xenograft model using 8305c cells treated with Compound 79 (45 mg/kg QD), BRAF inhibitor dabrafenib (30 mg/kg QD), MEK inhibitor trametinib (0.6 mg/kg QD) or the triple combination showing tumor volume (Fig. 29A), total body weight (Fig. 29B), tumor response (waterfall pit) (Fig, 29C), and tumor volumes in individual mice after treatment with the triple combination.
  • ATC Anaplastic thyroid cancer
  • Fig. 30A shows the tumor volumes of the Anaplastic thyroid cancer (ATC) tumor xenograft model using 8305c cells, whereby mice were treated with BRAF inhibitor dabrafenib (30 mg/kg QD) and MEK inhibitor trametinib (0.6 mg/kg QD) at Day 0 and Compound 79 (45 mg/kg QD) at Day 13.
  • Fig. 30B shows the tumor volumes of the same model as Fig. 30A whereby mice were treated with Compound 79 (45 mg/kg QD) at Day 0 and BRAF inhibitor dabrafenib (30 mg/kg QD) and MEK inhibitor trametinib (0.6 mg/kg QD) at Day 13.
  • Fig. 30C shows the tumor waterfall of the different groups of the model of Figs. 30A and 30B.
  • Figs. 31A-31 B shows the phosphoproteomic array of peripheral blood mononuclear cells (PBMCs) incubated with 1 uM of Compound 79 for 4 hours.
  • PBMCs peripheral blood mononuclear cells
  • Figs. 32A and 32B show the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof.
  • Fig. 32C and 32D show the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof.
  • Figs. 32A and 32B show the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof.
  • 32E and 32F shows the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (5, 15, or 45 mg/kg mg/kg), or combinations thereof.
  • Figs. 33A-33B shows the results of mice blood C-peptide levels (Fig. 33A) and insulin levels (Fig. 33B) after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof.
  • Fig. 33C shows the results of mice blood C-peptide after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK- 2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof.
  • Figs. 33D-33E shows the results of mice blood C-peptide levels (Fig.
  • Fig. 33D shows insulin levels (Fig. 33E) after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (5, 15, or 45 mg/kg), or combinations thereof.
  • Fig. 33F shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof.
  • Fig. 33D shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof.
  • FIG. 33G shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof.
  • Fig. 33H shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (5, 15, or 45 mg/kg), or combinations thereof.
  • the present description relates to compounds of Formula I, or pharmaceutically acceptable salts thereof.
  • the compounds of Formula I and their pharmaceutically acceptable salts are pharmacologically active compounds that modulate protein kinase activity, specifically the activity of serum and glucocorticoid regulated kinase isoform 1 (SGK-1).
  • the compounds of Formula I or their pharmaceutically acceptable salts can be suitable for the treatment of conditions in which SGK-1 activity is inappropriate. Non-limiting examples of such conditions can include Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, stent failure, cancer and epilepsy.
  • the compounds of Formula I and their pharmaceutically acceptable salts are described in more detail herein.
  • an alkyl group can have 1 to 20 carbon atoms (i.e, C 1 -C 20 alkyl), 1 to 8 carbon atoms (i.e., C 1 -C 8 alkyl), or 1 to 4 carbon atoms (i.e., C 1 -C 4 alkyl).
  • alkyl groups include, but are not limited to, methyl (Me, — CH 3 ), ethyl (Et, — CH 2 CH 3 ), 1- propyl (n-Pr, n-propyl, — CH 2 CH 2 CH 3 ), 2-propyl (i-Pr, i-propyl, — CH(CH 3 ) 2 ), 1-butyl (n-Bu, n-butyl, — CH 2 CH 2 CH 2 CH 3 ), 2-methyl-1 -propyl (i-Bu, i-butyl, — CH 2 CH(CH 3 ) 2 ), 2-butyl
  • Alkoxy means a group having the formula — O-alkyl, in which an alkyl group, as defined above, is attached to the parent molecule via an oxygen atom.
  • the alkyl portion of an alkoxy group can have 1 to 20 carbon atoms (i.e., C 1 -C 20 alkoxy), 1 to 12 carbon atoms (i.e., C 1 -C 12 alkoxy), or 1 to 4 carbon atoms (i.e., C 1 -C 4 alkoxy).
  • alkoxy groups include, but are not limited to, methoxy ( — O — CH 3 or — OMe), ethoxy ( — OCH 2 CH 3 or — OEt), t-butoxy ( — O — C(CH 3 ) 3 or -OtBu), and the like.
  • Haloalkyl is an alkyl group, as defined above, in which one or more hydrogen atoms of the alkyl group is replaced with a halogen atom.
  • the alkyl portion of a haloalkyl group can have 1 to 20 carbon atoms (i.e., C 1 -C 20 haloalkyl), 1 to 12 carbon atoms (i.e., C 1 -C 12 haloalkyl), or 1 to 4 carbon atoms (i.e., C 1 -C 4 haloalkyl).
  • suitable haloalkyl groups include, but are not limited to, —CF 3 , — CHF 2 , — CFH 2 , — CH 2 CF 3 , and the like.
  • Cycloalkyl means a mono or bicyclic carbocyclic ring functional group including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, bicyclo[2.2.1]heptanyl, bicyclo[3.2.1]octanyl, and bicyclo[5.2.0]nonanyl.
  • the cycloalkyl can have 3 to 12 carbon atoms (i.e., C 3 -C 12 cycloalkyl), 3 to 7 carbon atoms (i.e., C 3 -C 7 cycloalkyl) or 3 to 6 carbon atoms (i.e., C 3 -C 6 cycloalkyl).
  • the term “( C 3 -C 7 )cycloalkyl” refers to a cycloalkyl group containing from 3 to 8 carbons.
  • (C 3 -C 7 )cycloalkyl encompasses a monocyclic cycloalkyl group containing from 3 to 7 carbons and a bicyclic cycloalkyl group containing from 6 to 7 carbons.
  • Alkenyl is a hydrocarbon containing primary, secondary or tertiary carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp2 double bond.
  • an alkenyl group can have 2 to 20 carbon atoms (i.e., C 2 -C 20 alkenyl), 2 to 12 carbon atoms (i.e., C 2 -C 12 alkenyl), or 2 to 6 carbon atoms (i.e., C 2 -C 6 alkenyl).
  • Alkynyl is a hydrocarbon containing primary, secondary or tertiary carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp triple bond.
  • an alkynyl group can have 2 to 20 carbon atoms (i.e., C 2 -C 20 alkynyl), 2 to 12 carbon atoms (i.e., C 2 -C 12 alkynyl), or 2 to 6 carbon atoms (i.e., C 2 -C 6 alkynyl).
  • Alkylene refers to a saturated, branched or straight hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane.
  • an alkylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms.
  • Typical alkylene radicals include, but are not limited to, methylene ( — CH 2 — ), 1 ,1-ethylene (— CH(CH 3 )— ), 1 ,2-ethylene (— CH 2 CH 2 — ), 1 ,1-propylene (— CH(CH 2 CH 3 )— ), 1 ,2- propylene ( — CH 2 CH(CH 3 ) — ), 1 ,3-propylene ( — CH 2 CH 2 CH 2 — ), 1 ,4-butylene (— CH 2 CH 2 CH 2 CH 2 — ), and the like.
  • Alkenylene refers to an unsaturated, branched or straight hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene.
  • alkenylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms.
  • Alkynylene refers to an unsaturated, branched or straight hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne.
  • an alkynylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms.
  • Aryl means a monovalent aromatic hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system.
  • an aryl group can have 6 to 20 carbon atoms, 6 to 14 carbon atoms, or 6 to 12 carbon atoms.
  • Typical aryl groups include, but are not limited to, radicals derived from benzene (e.g., phenyl), substituted benzene, naphthalene, anthracene, biphenyl, and the like.
  • “Arylene” refers to an aryl as defined above having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent aryl.
  • Typical arylene radicals include, but are not limited to, phenylene, such as 1 ,4- phenylene.
  • Arylalkyl refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl radical.
  • Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, naphthobenzyl, 2-naphthophenylethan-1-yl and the like.
  • the arylalkyl group can comprise 6 to 20 carbon atoms, e.g., the alkyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
  • Arylalkenyl refers to an acyclic alkenyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also an sp2 carbon atom, is replaced with an aryl radical.
  • the aryl portion of the arylalkenyl can include, for example, any of the aryl groups disclosed herein, and the alkenyl portion of the arylalkenyl can include, for example, any of the alkenyl groups disclosed herein.
  • the arylalkenyl group can comprise 6 to 20 carbon atoms, e.g., the alkenyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
  • Arylalkynyl refers to an acyclic alkynyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also an sp carbon atom, is replaced with an aryl radical.
  • the aryl portion of the arylalkynyl can include, for example, any of the aryl groups disclosed herein, and the alkynyl portion of the arylalkynyl can include, for example, any of the alkynyl groups disclosed herein.
  • the arylalkynyl group can comprise 6 to 20 carbon atoms, e.g., the alkynyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
  • halogen refers to F, Cl, Br, or I.
  • haloalkyl refers to an alkyl group, as defined herein, that is substituted with at least one halogen.
  • branched or straight chained “haloalkyl” groups as used herein include, but are not limited to, methyl, ethyl, propyl, isopropyl, n- butyl, and t-butyl substituted independently with one or more halogens, for example, fluoro, chloro, bromo, and iodo.
  • haloalkyl should be interpreted to include such substituents as perfluoroalkyl groups such as — CF 3 .
  • substituted in reference to alkyl, aryl, arylalkyl, carbocyclyl, heterocyclyl, and other groups used herein, for example, “substituted alkyl”, “substituted cycloalkyl”, “substituted aryl”, “substituted arylalkyl”, “substituted heterocyclyl”, and “substituted carbocyclyl” means a group, alkyl, alkylene, aryl, arylalkyl, heterocyclyl, carbocyclyl respectively, in which one or more hydrogen atoms are each independently replaced with a non-hydrogen substituent.
  • substituted or are shown diagrammatically to be substituted (or optionally substituted, e.g., when the number of substituents ranges from zero to a positive integer), then the terms “alkyl”, “aryl”, “heterocyclyl”, etc. are understood to be interchangeable with “alkylene”, “arylene”, “heterocyclylene”, etc.
  • Heteroalkyl refers to an alkyl group where one or more carbon atoms have been replaced with a heteroatom, such as, O, N, or S.
  • a heteroatom e.g., O, N, or S
  • the resulting heteroalkyl groups are, respectively, an alkoxy group (e.g., — OCH 3 , etc.), an amine (e.g., — NHCH 3 , — N(CH 3 ) 2 , and the like), or a thioalkyl group (e.g., — SCH 3 ).
  • heteroalkyl groups are, respectively, an alkyl ether (e.g., — CH 2 CH 2 — O — CH 3 , etc.), an alkyl amine (e.g., — CH 2 NHCH 3 , — CH 2 N(CH 3 ) 2 , and the like), or a thioalkyl ether (e.g., — CH 2 — S— CH 3 ).
  • an alkyl ether e.g., — CH 2 CH 2 — O — CH 3 , etc.
  • alkyl amine e.g., — CH 2 NHCH 3 , — CH 2 N(CH 3 ) 2
  • thioalkyl ether e.g., — CH 2 — S— CH 3
  • the resulting heteroalkyl groups are, respectively, a hydroxyalkyl group (e.g., — CH 2 CH 2 — OH), an aminoalkyl group (e.g., — CH 2 NH 2 ), or an alkyl thiol group (e.g., — CH 2 CH 2 — SH).
  • a heteroalkyl group can have, for example, 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms.
  • a C 1 -C 6 heteroalkyl group means a heteroalkyl group having 1 to 6 carbon atoms.
  • Heterocycle or “heterocyclyl” as used herein includes by way of example and not limitation those heterocycles described in Paquette, Leo A.; Principles of Modern Heterocyclic Chemistry (W. A. Benjamin, New York, 1968), particularly Chapters 1 , 3, 4, 6, 7, and 9; The Chemistry of Heterocyclic Compounds, A Series of Monographs” (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566.
  • heterocycle includes a “carbocycle” as defined herein, wherein one or more (e.g.
  • heterocycle or “heterocyclyl” includes saturated rings, partially unsaturated rings, and aromatic rings (i.e., heteroaromatic rings). Heterocycles includes aromatic and non- aromatic mono-, bi-, and poly-cyclic rings, whether fused, bridged, or spiro.
  • heterocycle encompasses, but is not limited to “heteroaryl.”
  • heterocycles include by way of example and not limitation pyridyl, dihydroypyridyl, tetrahydropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, azetidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofur
  • carbon bonded heterocycles are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1 , 3, 4, 5, 6, 7, or 8 of an isoquinoline.
  • carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3- pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5- pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4- thiazolyl, or 5-thiazolyl.
  • nitrogen bonded heterocycles are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3- pyrazoline, piperidine, piperazine, indole, indoline, 1 H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or p-carboline.
  • nitrogen bonded heterocycles include 1-aziridyl, 1-azetedyl, 1-pyrrolyl, 1-imidazolyl, 1-pyrazolyl, and 1-piperidinyl.
  • Heterocyclylene refers to a heterocyclyl, as defined herein, derived by replacing a hydrogen atom from a carbon atom or heteroatom of a heterocyclyl, with an open valence.
  • heteroarylene refers to an aromatic heterocyclylene.
  • Heterocyclylalkyl refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heterocyclyl radical (i.e. , a heterocyclyl-alkylene- moiety).
  • Typical heterocyclyl alkyl groups include, but are not limited to heterocyclyl-CH 2 — , 2-(heterocyclyl)ethan-1-yl, and the like, wherein the “heterocyclyl” portion includes any of the heterocyclyl groups described above, including those described in Principles of Modern Heterocyclic Chemistry.
  • heterocyclyl group can be attached to the alkyl portion of the heterocyclyl alkyl by means of a carbon-carbon bond or a carbon- heteroatom bond, with the proviso that the resulting group is chemically stable.
  • the heterocyclyl alkyl group comprises 2 to 20 carbon atoms, e.g., the alkyl portion of the arylalkyl group comprises 1 to 6 carbon atoms and the heterocyclyl moiety comprises 1 to 14 carbon atoms.
  • heterocyclylalkyls include by way of example and not limitation 5-membered sulfur, oxygen, and/or nitrogen containing heterocycles such as thiazolylmethyl, 2-thiazolylethan-1-yl, imidazolylmethyl, oxazolylmethyl, thiadiazolylmethyl, and the like, 6-membered sulfur, oxygen, and/or nitrogen containing heterocycles such as piperidinylmethyl, piperazinylmethyl, morpholinylmethyl, pyridinylmethyl, pyridizylmethyl, pyrimidylmethyl, pyrazinylmethyl, and the like.
  • heterocycles such as thiazolylmethyl, 2-thiazolylethan-1-yl, imidazolylmethyl, oxazolylmethyl, thiadiazolylmethyl, and the like
  • 6-membered sulfur, oxygen, and/or nitrogen containing heterocycles such as piperidinylmethyl, piperazinylmethyl, morpholinylmethyl, pyridin
  • Heterocyclylalkenyl refers to an acyclic alkenyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also a sp2 carbon atom, is replaced with a heterocyclyl radical (i.e., a heterocyclyl-alkenylene- moiety).
  • the heterocyclyl portion of the heterocyclyl alkenyl group includes any of the heterocyclyl groups described herein, including those described in Principles of Modern Heterocyclic Chemistry, and the alkenyl portion of the heterocyclyl alkenyl group includes any of the alkenyl groups disclosed herein.
  • heterocyclyl group can be attached to the alkenyl portion of the heterocyclyl alkenyl by means of a carbon-carbon bond or a carbon-heteroatom bond, with the proviso that the resulting group is chemically stable.
  • the heterocyclyl alkenyl group comprises 2 to 20 carbon atoms, e.g., the alkenyl portion of the heterocyclyl alkenyl group comprises 1 to 6 carbon atoms and the heterocyclyl moiety comprises 1 to 14 carbon atoms.
  • “Heteroaryl” refers to a monovalent aromatic heterocyclyl having at least one heteroatom in the ring.
  • Non-limiting examples of suitable heteroatoms which can be included in the aromatic ring include oxygen, sulfur, and nitrogen.
  • suitable heteroaryl rings include all of those listed in the definition of “heterocyclyl”, including pyridinyl, pyrrolyl, oxazolyl, indolyl, isoindolyl, purinyl, furanyl, thienyl, benzofuranyl, benzothiophenyl, carbazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, quinolyl, isoquinolyl, pyridazyl, pyrimidyl, pyrazyl, and the like.
  • Carbocycle or “carbocyclyl” refers to a saturated, partially unsaturated or aromatic ring having 3 to 7 carbon atoms as a monocycle, 7 to 12 carbon atoms as a bicycle, and up to about 20 carbon atoms as a polycycle.
  • Monocyclic carbocycles have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms.
  • Bicyclic carbocycles have 7 to 12 ring atoms, e.g., arranged as a bicyclo (4,5), (5,5), (5,6) or (6,6) system, or 9 or 10 ring atoms arranged as a bicyclo (5,6) or (6,6) system.
  • Carbocycles includes aromatic and non-aromatic mono-, bi-, and poly-cyclic rings, whether fused, bridged, or spiro.
  • monocyclic carbocycles include the cycloalkyls group such as cyclopropyl, cyclobutyl, cyclopentyl, 1 -cyclopent- 1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1- cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl or aryl groups such as phenyl, and the like.
  • “carbocycle,” as used herein, encompasses but is not limited to “aryl”, “phenyl” and “biphenyl.”
  • Carbocyclylene refers to a carbocyclyl or carbocycle as defined above having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent carbocyclyl. Typical carbocyclylene radicals include, but are not limited to, phenylene. Thus, “carbocyclylene,” as used herein, encompasses but is not limited to “arylene.”
  • Carbocyclylalkyl refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a carbocyclyl radical as defined above.
  • Typical carbocyclylalkyl groups include, but are not limited to the arylalkyl groups such as benzyl, 2-phenylethan-1-yl, naphthylmethyl, 2- naphthylethan-1-yl, naphthobenzyl, 2-naphthophenylethan-1-yl or the cycloalkylalkyl groups such as cyclopropylmethyl, cyclobutylethyl, cyclohexyl methyl and the like.
  • the arylalkyl group can comprise 6 to 20 carbon atoms, e.g., the alkyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
  • the cycloalkylalkyl group can comprise 4 to 20 carbon atoms, e.g., the alkyl moiety is 1 to 6 carbon atoms and the cycloalkyl group is 3 to 14 carbon atoms.
  • Arylheteroalkyl refers to a heteroalkyl as defined herein, in which a hydrogen atom, which may be attached either to a carbon atom or a heteroatom, has been replaced with an aryl group as defined herein.
  • the aryl groups may be bonded to a carbon atom of the heteroalkyl group, or to a heteroatom of the heteroalkyl group, provided that the resulting aryl heteroalkyl group provides a chemically stable moiety.
  • an arylheteroalkyl group can have the general formulae -alkylene-O-aryl, -alkylene-O-alkylene-aryl, - alkylene-NH-aryl, -alkylene-NH-alkylene-aryl, -alkylene-S-aryl, -alkylene-S-alkylene-aryl, and the like.
  • any of the alkylene moieties in the general formulae above can be further substituted with any of the substituents defined or exemplified herein.
  • Heteroarylalkyl refers to an alkyl group, as defined herein, in which a hydrogen atom has been replaced with a heteroaryl group as defined herein.
  • Non-limiting examples of heteroaryl alkyl include — CH 2 -pyridinyl, — CH 2 -pyrrolyl, — CH 2 -oxazolyl, — CH 2 -indolyl, — CH 2 -isoindolyl, — CH 2 -purinyl, — CH 2 -furanyl, — CH 2 -thienyl, — CH 2 -benzofuranyl, — CH 2 - benzothiophenyl, — CH 2 -carbazolyl, — CH 2 -imidazolyl, — CH 2 -thiazolyl, — CH 2 -isoxazolyl, — CH 2 -pyrazolyl, — CH 2 -isothiazolyl, — CH 2
  • optionally substituted in reference to a particular moiety of the compound of the Formulae of the invention, for example an optionally substituted aryl group, refers to a moiety having 0, 1 , or more substituents.
  • prodrug refers to any compound that when administered to a biological system generates the drug substance, i.e., active ingredient, as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), photolysis, and/or metabolic chemical reaction(s).
  • a prodrug is thus a covalently modified analog or latent form of a therapeutically active compound.
  • substituents and other moieties of the compounds of the present description should be selected in order to provide a compound which is sufficiently stable to provide a pharmaceutically useful compound which can be formulated into an acceptably stable pharmaceutical composition.
  • crystalline polymorphism means the ability of a crystalline compound to exist in different crystal structures. Polymorphism generally can occur as a response to changes in temperature, pressure, or both. Polymorphism can also result from variations in the crystallization process.
  • Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility, and melting point.
  • the crystalline polymorphism may result from differences in crystal packing (packing polymorphism) or differences in packing between different conformers of the same molecule (conformational polymorphism).
  • crystalline pseudopolymorphism means the ability of a hydrate or solvate of a compound to exist in different crystal structures.
  • the pseudopolymorphs of some of the compounds of the present description may exist due to differences in crystal packing (packing pseudopolymorphism) or due to differences in packing between different conformers of the same molecule (conformational pseudopolymorphism). It is understood that all polymorphs and pseudopolymorphs of the compounds described herein and their pharmaceutically acceptable salts are included within the scope of the present description.
  • an amorphous solid is a solid in which there is no long-range order of the positions of the atoms in the solid. This definition applies as well when the crystal size is two nanometers or less. Additives, including solvents, may be used to create amorphous forms the compounds of the present description.
  • Certain of the compounds described herein contain one or more chiral centers or may otherwise be capable of existing as multiple stereoisomers.
  • the scope of the present description includes mixtures of stereoisomers as well as purified enantiomers or enantiomerically/diastereomerically enriched mixtures. Also included within the scope of the present description are the individual isomers of the compounds described herein, as well as any wholly or partially equilibrated mixtures thereof.
  • the compounds of the present description and their pharmaceutically acceptable salts also includes the individual isomers of the compounds represented by the formulas above as mixtures with isomers thereof in which one or more chiral centers are inverted.
  • the compounds of the present description may exist in solvated, for example hydrated, as well as unsolvated forms.
  • the salts of the compounds of the present description are pharmaceutically acceptable salts. Salts encompassed within the term “pharmaceutically acceptable salts” refer to non-toxic salts of the compounds of the present description.
  • Suitable pharmaceutically acceptable salts include inorganic acid addition salts such as chloride, bromide, sulfate, phosphate, and nitrate; organic acid addition salts such as acetate, galactarate, propionate, succinate, lactate, glycolate, malate, tartrate, citrate, maleate, fumarate, methanesulfonate, p-toluenesulfonate, and ascorbate; salts with acidic amino acid such as aspartate and glutamate; alkali metal salts such as sodium salt and potassium salt; alkaline earth metal salts such as magnesium salt and calcium salt; ammonium salt; organic basic salts such as trimethylamine salt, triethylamine salt, pyridine salt, picoline salt, dicyclohexylamine salt, and N,N'-dibenzylethylenediamine salt; and salts with basic amino acid such as lysine salt and arginine salt.
  • the salts may be in some cases hydrate
  • Inoperable species or compounds means compound structures that violates relevant scientific principles (such as, for example, a carbon atom connecting to more than four covalent bonds) or compounds too unstable to permit isolation and formulation into pharmaceutically acceptable dosage forms.
  • the compounds of the present description can be formulated with conventional carriers and excipients, which will be selected in accordance with ordinary practice. Tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form, and when intended for delivery by other than oral administration generally will be isotonic. All formulations will optionally contain excipients such as those set forth in the Handbook of Pharmaceutical Excipients (1986), herein incorporated by reference in its entirety. Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH of the formulations ranges from about 3 to about 11 but is ordinarily about 7 to 10.
  • compositions of the invention both for veterinary and for human use, comprise at least one active ingredient, together with one or more acceptable carriers and optionally other therapeutic ingredients.
  • the carrier(s) must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and physiologically innocuous to the recipient thereof.
  • the formulations include those suitable for the foregoing administration routes.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa.), herein incorporated by reference in its entirety. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be administered as a bolus, electuary or paste.
  • a tablet is made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingredient.
  • compositions according to the present description include one or more compounds together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents.
  • Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration.
  • tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
  • Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc.
  • inert diluents such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate
  • granulating and disintegrating agents such as maize starch, or alginic acid
  • binding agents such as cellulose, microcrystalline cellulose, starch,
  • Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • Such excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate).
  • a suspending agent such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyr
  • the aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents, such as those set forth herein, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules of the invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium tartrate
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate
  • the pharmaceutical compositions may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these.
  • Suitable emulsifying agents include naturally- occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate.
  • the emulsion may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • sweetening agents such as glycerol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • compositions of the invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • a sterile injectable preparation such as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned herein.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1 ,3-butane-diol or prepared as a lyophilized powder.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may likewise be used in the preparation of injectables.
  • a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weightweight).
  • the pharmaceutical composition can be prepared to provide easily measurable amounts for administration.
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 pg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations are presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use.
  • sterile liquid carrier for example water for injection
  • Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
  • Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
  • formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
  • compositions comprising one or more compounds of the present description formulated for sustained or controlled release.
  • the effective dose of an active ingredient depends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses) or against an active disease or condition, the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies.
  • the effective dose can be expected to be from about 0.0001 to about 10 mg/kg body weight per day, typically from about 0.001 to about 1 mg/kg body weight per day, more typically from about 0.01 to about 1 mg/kg body weight per day, even more typically from about 0.05 to about 0.5 mg/kg body weight per day.
  • the daily candidate dose for an adult human of approximately 70 kg body weight will range from about 0.05 mg to about 100 mg, or between about 0.1 mg and about 25 mg, or between about 0.4 mg and about 4 mg, and may take the form of single or multiple doses.
  • SGK-1 and associated conditions will range from about 0.05 mg to about 100 mg, or between about 0.1 mg and about 25 mg, or between about 0.4 mg and about 4 mg, and may take the form of single or multiple doses.
  • the present description relates to compounds or pharmaceutically acceptable salts thereof, for the treatment various conditions treatable by inhibiting SGK-1.
  • the condition can be Long QT syndrome (LQTS), such as genetic LQTS or acquired LQTS, or other cardiovascular diseases (e.g., dilated cardiomyopathy - genetic or acquired) that are treatable by inhibiting SGK-1.
  • LQTS Long QT syndrome
  • cardiovascular diseases e.g., dilated cardiomyopathy - genetic or acquired
  • SGK- 1 inhibition in vivo has a protective effect and can alleviate symptoms associated with LQTS; can reduce and alleviate symptoms associated with heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, genetic or acquired dilated cardiomyopathy, hypertrophic cardiomyopathy, and stent failure.
  • Long QT syndrome can be genetic (e.g. caused by a mutation in the KCNQ1 gene, the KCNH 2 gene, or the SCN5a gene).
  • Long QT syndrome is not associated with a genetic mutation and is acquired as a result of exposure to an external stimulus.
  • acquired Long QT syndrome can be a side effect of drugs such as erythromycin or haloperidol.
  • Acquired Long QT syndrome is also associated with other heart conditions such as myocardial ischemia.
  • the present description also relates to compounds or pharmaceutically acceptable salts thereof, forthe treatment of other conditions related to SGK-1 mediated mechanisms, such as cancer, Parkinson’s disease and Lafora disease.
  • the present description provides compounds or pharmaceutically acceptable salts thereof for treating cancer, tumor, or another proliferative disorder.
  • the terms “inhibition of cancer”, “inhibition of cancer cell proliferation”, and “inhibition of cancer invasion and metastasis” refer to the inhibition, or decrease in the rate, of the growth, division, maturation, viability, or ability to invade and colonize other organs and tissues of cancer cells, and/or causing the death of cancer cells, individually or in aggregate with other cancer cells, by cytotoxicity, nutrient depletion, induction of differentiation or apoptosis, or recognition by the immune system in order to elicit an immune response to the cancer cells.
  • tissues containing cancerous cells whose proliferation can be inhibited by a compound, salt or composition thereof described herein and against which the methods described herein are useful include but are not limited to breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
  • the cancer treated by a provided compound, salt or composition thereof is a cancer or tumor that is a melanoma, liposarcoma, lung cancer (e.g., non-small cell lung cancer), breast cancer (e.g., ER+, ER-, HER2+, HER2-, PR-, PR+, triple negative, luminal, basal), Luminal androgen receptor (LAR)) prostate cancer, leukemia, kidney cancer, esophageal cancer, thyroid cancer (e.g., Anaplastic Thyroid Cancer (ATC), Poorly Differentiated Thyroid Cancer (PDTC)), ovarian cancer, endometrial cancer, Head and Neck Squamous Cell Carcinoma, brain cancer, lymphoma, rectal cancer, colon cancer, uterine cancer, cervical cancer, Hepatocellular carcinoma, Papillary thyroid carcinoma (PTC), or colorectal cancer
  • lung cancer e.g., non-small cell lung cancer
  • breast cancer e.g., ER+, ER-, HER
  • the compounds of the present description can be used to treat cancer by inhibiting signaling of the AKT/PI3K/mTOR pathway in patients whose tumors have activation of this pathway through mutations in PIK3CA, AKT1 , and/or PTEN for example.
  • the compounds of the present description can be used in combination with compounds that inhibit AKT/PI3K/mTOR signaling to treat cancer in patients whose tumors have activation of this pathway through mutations in PIK3CA, AKT1 , and/or PTEN for example.
  • Non-limiting examples of AKT/PI3K/mTOR inhibitors include NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI-587), PQR 3 09 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL- 101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK- 1117, ML
  • the inhibitor of AKT, PI3K, and/or mTOR may be selected from the group consisting of vistusertib (AZD2014), NU7441 (KU-57788), KU-0063794, TGX-221 , RLY-2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223, ABTL0812, DCBCI0901, AZD-8055, sapanisertib, JR-AB2-011 , omipalisib, and torkinibm PI-103.
  • the compounds of the present description can be used in combination with at least one inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK.
  • the inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK may be selected from the group consisting of dabrafenib, trametinib (GSK1120212), encorafenib (LGX818), binimetinib, cobimetinib, naporafenib (LXH 2 54), sorafenib (BAY 43-9006), vemurafenib (PLX4032), PLX-4720, regorafenib (BAY 73- 4506), GDC_0879, RAF265 (CHIR-265), AZ 628, lifirafenib (BGB-283), NVP-BHG712, SB590885, ZM 336372, GW5074, TAK-632, agerafenib (RXDX-105), G
  • Sotorasib (AMG510), lonafamib (SCH66336), ASP2453, BI-3406, BI-2852, BAY-293, KRpep-2d, ARS-853, adagrasib (MRTX849), adagrasib (MRTX849), ARS1620, divarasib (GDC-6036), GDC-1971 , IACS- 13909, JAB-3068, RMC-4550, RMC-4630, SHP099, TNO155, GDC-1971 , HBI-2376, BBP-398 (IACS-15509), PF-07284892, selpercatinib (LOXO-292), pralsetinib (BLU-667), LOXO-260, TAS-0953/HM06, TPX-0046, EP0031 , APS03118, TP-0903, BGB324 (R428), bosutinib (SKI-606),
  • the compounds of the present description can be combined with an inhibitor of the RAS/RAF/MAPK pathway in cells that have a mutation activating RAS, RAF, MEK, or ERK signaling.
  • RAF/RAS/MEK/ERK/SOS1/SOS2/SHP2 inhibitors include dabrafenib, trametinib (GSK1120212), encorafenib (LGX818), binimetinib, cobimetinib, naporafenib (LXH 2 54), sorafenib (BAY 43-9006), vemurafenib (PLX4032), PLX-4720, regorafenib (BAY 73- 4506), GDC_0879, RAF265 (CHIR-265), AZ 628, lifirafenib (BGB-283), NVP-BHG712, SB590885, ZM 336372, GW5074,
  • Sotorasib (AMG510), lonafamib (SCH66336), ASP2453, BI-3406, BI-2852, BAY-293, KRpep-2d, ARS-853, adagrasib (MRTX849), adagrasib (MRTX849), ARS1620, divarasib (GDC-6036), GDC-1971 , IACS- 13909, JAB-3068, RMC-4550, RMC-4630, SHP099, TNO155, GDC-1971 , HBI-2376, BBP-398 (IACS-15509), and PF-07284892.
  • the compounds of the present description can be combined with an inhibitor of the RTK, PI3K, AKT, RAS, RAF or MAPK pathway in cancer cells that, for example, have up-regulation of gene expression associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation.
  • the compounds of the present description can be combined with an inhibitor of the PI3K or AKT pathway in cancer cells that, for example, have upregulation of gene signatures associated with basal and/or mesenchymal breast cancer. In some embodiments, the compounds of the present description can be combined with an inhibitor of the PI3K or AKT pathway in cancer cells that, for example, have a downregulation of gene signatures associated with luminal breast cancer.
  • Non-limiting examples of AKT/PI3K/mT0R inhibitors include PDK1 inhibitors, and other mT0RC1/2 inhibitors such as vistusertib (AZD2014), NU7441 (KU-57788), KU-0063794, TGX-221 , RLY-2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223, ABTL0812, DCBCI0901 , AZD-8055, sapanisertib, JR-AB2-011 , omipalisib, and torkinibm PI-103.
  • PDK1 inhibitors include PDK1 inhibitors, and other mT0RC1/2 inhibitors such as vistusertib (AZD2014), NU7441 (KU-57788), KU-0063794, TGX-221 , RLY-2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223,
  • the compounds of the present description can be used to treat cancer by inhibiting signaling of the RAF/MEK/ERK pathway in patients whose tumors have activation of this pathway through, for example, mutations in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIG.
  • the compounds of the present description can be used to treat cancer by inhibiting signaling of the RAF/MEK/ERK and/or PI3K, AKT, mTOR pathway in patients whose tumors have concomitant activation of these pathways through, for example, concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN.
  • the compounds of the present description can be used to treat cancer in patients that had prior progression on prior a treatment (i.e., cancers that are resistant to one or more anticancer therapies) (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof).) (e.g., combination of inhibitors).
  • a treatment i.e., cancers that are resistant to one or more anticancer therapies
  • BRAF inhibitors e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof.
  • EGFR inhibitors e.g., EGFR inhibitors, or combinations thereof.
  • combination of inhibitors e.g., a combination of BRAF inhibitors, MEK inhibitor, and/or EGFR.
  • the compounds of the present description can be combined with an inhibitor of the RTK, PI3K, AKT, RAS, RAF or MAPK pathway in cancers or tumors that have an upregulation of a mitogen activated protein kinase (MAPK) gene expression signature, such as CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and/or SPRY4.
  • a mitogen activated protein kinase (MAPK) gene expression signature such as CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and/or SPRY4.
  • MAPK mitogen activated protein kinase
  • the compounds of the present description can be combined with an inhibitor of one or more inhibitors of the pathways described in Table V. In some embodiments, the compounds of the present description can be combined with an inhibitor of one or more inhibitors described herein in the cancers or tumors have an upregulation in one or more genes described in Tables W-Z.
  • the compounds of the present description can be combined with an inhibitor of a receptor tyrosine kinase (RTK).
  • RTK inhibitors include epidermal growth factor receptor (EGFR) inhibitors erlotinib, osimertinib, neratinib, gefitinib, cetuximab, panitumumab, dacomitinib, lapatinib, necitumumab, mobercitinib, vandetanib, sapitinib, afatinib, canertinib, CP-724714, CUDC-101 , mubritinib (TAK 165), OSI-420, pelitinib, irbinitinib (ARRY-380, ONT-380), varlitinib, TAK-285, BIBX 1382, PD168393, raciletinib, poziotinib, CL-387785
  • EGFR epiderma
  • the compounds of the present description can be used to treat inflammatory and fibrotic diseases that can include fatty liver diseases, endometriosis, types 1 or 2 diabetes mellitus, inflammatory bowel disease, asthma, rheumatoid arthritis, obesity, systemic sclerosis, sclerodermatous graft vs. host disease, nephrogenic systemic fibrosis, as well as organ-specific fibrosis, including radiation-induced fibrosis, and auto- immune diseases.
  • inflammatory and fibrotic diseases can include fatty liver diseases, endometriosis, types 1 or 2 diabetes mellitus, inflammatory bowel disease, asthma, rheumatoid arthritis, obesity, systemic sclerosis, sclerodermatous graft vs. host disease, nephrogenic systemic fibrosis, as well as organ-specific fibrosis, including radiation-induced fibrosis, and auto- immune diseases.
  • Serine/threonine-protein kinase (also known as serum/glucocorticoid-regulated kinase 1) is a protein kinase that plays a role in a cell's response to stress. In vivo, SGK- 1 activates certain potassium, sodium, and chloride channels. For instance, the protein is known to regulate the myo-inositol transporter during osmotic stress.
  • inhibitor of SGK-1 refers to any compound that can block, arrest, interfere with, or reduce the biological activity of SGK-1.
  • the compounds of the present description can be used for increasing fetal hemoglobin (HbF) in erythrocytes. In some embodiments, the compounds of the present description can be used for the treatment of a p-hemoglobinopathy. In some embodiments, the compounds of the present description can be used for the treatment of sickle cell disease.
  • HbF fetal hemoglobin
  • the compounds of the present description can be used for the treatment of prostate cancer. In other embodiments, the compounds of the present description can be used for the treatment of epilepsy. in some embodiments, there is described a method for the treatment of cancer or tumor in a subject, comprising: i.
  • cells from said cancer or tumor e.g., cancer or tumor sample or biopsy: have an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; have an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; have a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; have an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIO; have an activation of a RAF, MEK, ERK, PI3K, AKT, and/
  • cancer or tumor e.g., cancer or tumor sample or biopsy
  • the cancer or tumor has been predetermined as: having an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; having an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; having a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; having an activation of a RAF
  • the pharmaceutical compositions defined herein, or the combinations defined herein for the treatment of one or more side effects or toxicities caused by an anticancer therapy in a subject undergoing said anticancer therapy.
  • the one or more side effects or toxicities caused by the anticancer therapy may be selected from the group consisting of: hyperglycemia and hyperinsulinemia, increased ALT/AST, pyrexia, rash, diarrhea, nausea, fatigue, thrombocytopenia, anemia, dermatitis acneiform, QT elongation, and dyspnea.
  • a method for treating one or more side effects or toxicities caused by an anticancer therapy comprising administering to a subject undergoing said anticancer therapy one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical compositions defined herein, or the combinations defined herein.
  • the one or more side effects or toxicities caused by the anticancer therapy may be selected from the group consisting of: hyperglycemia and hyperinsulinemia, increased ALT/AST, pyrexia, rash, diarrhea, nausea, fatigue, thrombocytopenia, anemia, dermatitis acneiform, QT elongation, and dyspnea.
  • a method for monitoring the progression of an anticancer therapy in a subject that was previously administered with one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical compositions defined herein, or the combinations defined herein wherein the method comprises determining in a sample from said subject: a loss of phosphorylation of NDRG1 ; an increase in phosphorylation of MAPK14 (i.e.
  • p38 an increase in phosphorylation of MAPK14 (i.e., p38); an increase in phosphorylation of heat shock protein family B1 (i.e., HSPB1 or HSP27); an increase in phosphorylation of tumor suppressor p53 (TP53); an increase in phosphorylation of ribosomal protein S6 kinase A1 (RSK1); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERKI); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERK1); and/or an increase in phosphorylation of AKT1
  • said loss of phosphorylation of NDRG1 is at threonine 346 of the NDRG1 ; said increase in phosphorylation of MAPK14 (i.e., p38) is at threonine 180 of the MAPK14; said increase in phosphorylation of MAPK14 (i.e., p38) is at tyrosine 182 of the MAPK14; said increase in phosphorylation of heat shock protein family B1 (i.e., HSPB1 or HSP27) at serine 82 of the HSPB1 ; said increase in phosphorylation of tumor suppressor p53 (TP53) is at serine 15 of the TP53; said increase in phosphorylation of ribosomal protein S6 kinase A1 (RSK1) is at serine 380 of the RSK1 ; said increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2) is at ty
  • said loss or increase in phosphorylation is in comparison to a sample from a healthy subject or a subject that is not undergoing said anticancer therapy.
  • said monitoring the progression of said anticancer therapy includes determining if the anticancer therapy is effective or if the cancer is becoming resistant to the anticancer therapy.
  • the compounds of the present description and their pharmaceutically acceptable salts thereof are pharmacologically active compounds that modulate protein kinase activity, specifically the activity of serum and glucocorticoid regulated kinase isoform 1 (SGK-1).
  • the compounds of the present description or their pharmaceutically acceptable salts can be suitable for the treatment of conditions in which SGK-1 activity is inappropriate.
  • Non- limiting examples of such conditions can include Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, stent failure, prostate cancer and epilepsy.
  • Other non- limiting examples of such conditions include ⁇ -hemoglobinopathies, such as sickle cell disease.
  • Z is selected from the group consisting of a direct bond, -O-, -S-, - CH(R 9 )- and -N(Rw)-, wherein R 9 and R 10 are independently of one another selected from the group consisting of H and (C 1 -C 4 )-alkyl.
  • Z is selected from the group consisting of a direct bond, -O-, -S-, -CH 2 - and -NH-.
  • Z is a direct bond.
  • Z is selected from the group consisting of -O- and - NH-.
  • R 3 is selected from the group consisting of H, (C 1 -C 8 )-alkyl, R 30 and ( C 1 -C 6 )-alkyl- R 30 , wherein (C 1 -C 8 )-alkyl is unsubstituted or substituted by one or more identical or different substituents R 31 .
  • R 30 is a 3-membered to 12-membered, monocyclic or bicyclic, saturated, partially unsaturated or aromatic, cyclic group which comprises 0, 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, which is unsubstituted or substituted by one or more identical or different substituents R 32 .
  • R 31 is selected from the group consisting of halogen, -OH, -CF 3 , -O-(C 1 -C 4 )-alkyl, -N(R 33 )-R 34 and -CN.
  • R 33 and R 34 are independently of one another selected from the group consisting of H, (C 1 -C 4 )-alkyl and (C 3 -C 7 )-cycloalkyl, wherein (C 1 -C 4 )-alkyl and (C3- C 7 )-cycloalkyl are unsubstituted or substituted by one or more identical or different substituents R 50 , wherein R 50 is selected from the group consisting of halogen, -OH, -O- (C 1 -C 4 )-alkyl, -CF 3 and -CN.
  • R 37 , R 38 , R 39 , R 40 , R 41 , R 42 and R 43 are independently of one another selected from the group consisting of H and (C 1 -C 4 )-alkyl.
  • R 3 is selected from the group consisting of H, -CH 2 OH,
  • Z is a direct bond and R 3 is selected from the group consisting of
  • Z is selected from the group consisting of - O- and -NH- and R 3 is selected from the group consisting of:
  • R 1 is selected from the group consisting of H, -N(R 11 )R 12 , -N(R 13 )- C(O)-R 14 , -NR 13 -S(O) 2 -R 15 , -NR 13 -C(O)-NH-R 16 , -(C 1 -C 4 )-alkyl, — (C 1 -C 4 )-alkyl-OR 17 and —
  • (C 1 -C 4 )-alkyl-N(Ris) R 19 wherein R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 and R 19 are independently of one another selected from the group consisting of H and (C 1 -C 4 )-alkyl.
  • R 1 is selected from the group consisting of -(C 1 -C 4 )-alkyl, and - (C 1 -C 4 )-alkyl-N( R 18 )R 19 . In some embodiments, R 1 is selected from the group consisting of -CH 3, -CH 2 N(CH 3 ) 2 and -CH 2 -CH 2 -N(CH 3 ) 2 .
  • Y is selected from the group consisting of carbocyclylene and heterocyclylene, which is unsubstituted or substituted by one or more identical or different substituents R 5 , wherein R 5 is selected from the group consisting of halogen, (C 1 -C 4 )-alkyl, -O-( C 1 -C 4 )-alkyl and -CN.
  • R 5 is selected from the group consisting of halogen, (C 1 -C 4 )-alkyl, -O-( C 1 -C 4 )-alkyl and -CN.
  • Y is selected from the group consisting of arylene and heteroarylene, which is unsubstituted or substituted by one or more identical or different substituents R 5 .
  • Y is selected from the group consisting of:
  • A is selected from the group of a direct bond or -CH 2 -.
  • -Y- is directly linked to the nitrogen of the sulfonamide group.
  • R 2 is selected from the group consisting of (C 1 -C 4 )-alkyl, (C 3 -C 7 )- cycloalkyl, (C 2 -C 4 )-alkenyl, (C 2 -C 4 )-alkynyl, phenyl and a 5-membered or 6-membered monocyclic, saturated, partially unsaturated or aromatic, heterocyclic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and is bonded via a ring carbon atom or a ring nitrogen atom, wherein R 2 is unsubstituted or substituted by one or more identical or different substituents R 20 , wherein R 20 is selected from the group consisting of halogen, -CF 3 , (C 1 -C 4 )-alkyl, -OR 21 , -N(R 22 )R 23 , (C 1 -C 4 )-alkyl-
  • R 2 is selected from the group consisting of (C 1 -C 4 )-alkyl, (C 3 - C 7 )-cycloalkyl, (C 2 -C 4 )-alkenyl, (C 2 -C 4 )-alkynyl, phenyl and a 5-membered or 6-membered monocyclic, saturated, partially unsaturated or aromatic, heterocyclic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and is bonded via a ring carbon atom or a ring nitrogen atom, wherein R 2 is unsubstituted or substituted by one or more identical or different substituents R 20 , wherein R 20 is selected from the group consisting of
  • Y is 1 ,4-phenylene and R1 is H, -N(R 11 )R 12 , -N(R 13 )-C(O)-R 14 , -NR 13 -S(O) 2 -Ri5, -NR 13 -C(O)-NH-Ri6, -(C 1 -C 4 )-alkyl or
  • R 2 is selected from the group consisting of (C 1 -C 4 )-alkyl, (C 3 -C 7 )- cycloalkyl, (C 2 -C 4 )-alkenyl, (C 2 -C 4 )-alkynyl and a 5-membered or 6-membered monocyclic, saturated or partially unsaturated, heterocyclic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and is bonded via a ring carbon atom or a ring nitrogen atom, wherein R 2 is unsubstituted or substituted by one or more identical or different substituents R 20 , wherein R 20 is selected from the group consisting of halogen, -CF 3 , (C 1 -C 4 )-alkyl, -OR 21 , -N(R 22 )R 23 , (C 1 -
  • R 2 is selected from the group consisting of:
  • Y is 1 ,4-phenylene and R 2 is selected from the group consisting of: -CH 3 ,
  • the compound of Formula I is selected from the group consisting of:
  • the compound of Formula I is selected from the group consisting of
  • Z is selected from the group consisting of O, CH 2 , S and NH;
  • R 1 is selected from the group consisting of H, and -(C 1 -C 4 )-alkyl;
  • R 3 is selected from the group consisting of -(CH 2 ) P -N(R 33 )R 34 ;
  • p is 1 , 2, 3 or 4;
  • R 2 is selected from the group consisting of a 5-membered or 6-membered monocyclic, aromatic or heteroaromatic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, wherein R 2 is unsubstituted or substituted by one or more identical or different substituents R 20 ;
  • R 20 is selected from the group consisting of halogen, -CF 3 , (C 1 -C 4 )-alkyl, -OR 21 , -N(R 22 )R 23 , (C 1 -C 4 )-alkyl-OR 24 , (C 1 -C 4 )-alkyl-N(R
  • Z is NH. In other embodiments, Z is O. In some embodiments, Ri is methyl. In some embodiments, p is 2, 3 or 4.
  • R 2 is selected from the group consisting of:
  • R 33 is methyl. In some embodiments, R 34 is methyl.
  • R 3 is selected from the group consisting of:
  • the compound of Formula II is selected from the group consisting of:
  • Z is selected from the group consisting of a direct bond, O, S, CH(Rg) and N(R 10 );
  • R 1 is selected from the group consisting of H, -N(R 11 )R 12 , -N(R 13 )-C(O)-R 14 , -NR 13 -S(O) 2 "R 15 , -NR 13 -C(O)-NH-R 16 , -(C 1 -C 4 )-alkyl, — (C 1 -C 4 )-alkyl-OR 17 and — (C 1 -C 4 )-alkyl-N(R 18 )R 19 ;
  • R 3 is selected from the group consisting of H, (C 1 -C 8 )-alkyl, R 30 and (C 1 -C 4 )-alkyl- R 30 , wherein (C 1 -C 8 )-alkyl is unsubstituted or substituted by one or more identical or different substituents R 31 ;
  • R 30 is a 3-membered to 12-membered, monocyclic or bicyclic, saturated, partially unsaturated or aromatic, cyclic group which comprises 0, 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, which is unsubstituted or substituted by one or more identical or different substituents R 32 ;
  • R 31 is selected from the group consisting of halogen, -OH, -CF 3 , -O-(C 1 -C 4 )-alkyl, -N(R 3 3)-R 34 and -CN;
  • R 32 is selected from the group consisting of halogen
  • R 3 8, R 39 , R40, R41 , R42 and R43 are independently of one another selected from the group consisting of H and (C 1 -C 4 )-alkyl;
  • R 33 and R 34 are independently of one another selected from the group consisting of H, (C 1 -C 4 )-alkyl and (C 3 -C 7 )-cycloalkyl wherein (C 1 -C 4 )-alkyl and (C 3 -C 7 )-cycloalkyl are unsubstituted or substituted by one or more identical or different substituents R 50 ; and R 50 is selected from the group consisting of halogen, -OH, -O-(C 1 -C 4 )-alkyl, -CF 3 and -CN.
  • R 1 is selected from the group consisting of -(C 1 -C 4 )-alkyl, and - (C 1 -C 4 )-alkyl-N(R 18 )R 19 .
  • R 1 is selected from the group consisting of -CH 3 , -CH 2 N(CH 3 ) 2 and -CH 2 -CH 2 -N(CH 3 ) 2 .
  • Z is selected from the group consisting of -O- and -NH-.
  • R 3 is selected from the group consisting of:
  • Z is a direct bond.
  • R 3 is selected from the group consisting of H, -CH 2 OH and -CH 3 .
  • R 2 is selected from the group consisting of: In some embodiments, the compound of Formula II is selected from the group consisting of:
  • Z is selected from the group consisting of O, CH 2 , S and NH;
  • R 1 is selected from the group consisting of H, and -(C 1 -C 4 )-alkyl
  • R 3 is selected from the group consisting of -(CH 2 ) P -N(R 33 )R 34 , wherein zero, one or two hydrogen atoms of the group -(CH 2 ) P - are independently replaced with F; p is 1 , 2, 3 or 4; R 2 is selected from the group consisting of a 5-membered or 6-membered monocyclic, aromatic or heteroaromatic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, wherein R 2 is unsubstituted or substituted by one or more identical or different substituents R 20 ; R 20 is selected from the group consisting of halogen, -CF 3 , (C 1 -C 4 )-alkyl, -OR 21 , -N(R 22 )R 23 , (C 1 -C 4 )-alkyl-OR 24 , (C 1 -C 4 )-alkyl-N(R 25 )R 26 and
  • R 50 is selected from the group consisting of halogen, -OR27, -O-(C 1 -C 4 )-alkyl, CF 3 , and -CN;
  • W 1 , W 2 , W 3 , W 4 are independently of one another selected from the group consisting of H, halogen, -OR 21 , -CF 3 , (C 1 -C 4 )-alkyl, and -CN.
  • Z is NH.
  • Z is O.
  • R1 is methyl.
  • p is 2, 3 or 4.
  • Z 1 and Z 2 are independently from one another selected from the group consisting of Cl, F, -OMe and - CN, and Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN.
  • R 2 is selected from the group consisting of:
  • R 2 is selected from the group consisting of:
  • R 2 is
  • R 3 is selected from the group consisting of: wherein R 51 is (C 1 -C 4 )-alkyl.
  • W 1 , W 2 , W 3 and W 4 are each H;
  • W 1 is F or Cl, W 2 , W 3 and W 4 are each H; or
  • W 1 and W 2 are each F, W 3 and W 4 are each H.
  • W 1 is F or Cl
  • W 2 is H
  • W 3 is H and W 4 is H.
  • W 1 is F
  • W 2 is H
  • W 3 is H and W 4 is H.
  • W 1 is Cl
  • W 2 is H
  • W 3 is H and W 4 is H.
  • Z is selected from the group consisting of O and NH;
  • R 1 is selected from the group consisting of H and (C 1 -C 4 )-alkyl
  • R 3 is selected from the group consisting of -(CH 2 ) P -N(R 33 )R 34 ; p is 2, 3 or 4; Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • R 33 and R 34 are independently of one another a (C 1 -C 4 )-alkyl
  • W 1 , W 2 , W 3 , W 4 are independently of one another selected from the group consisting of H, halogen, -OR 21 , -CF 3 , (C 1 -C 4 )-alkyl, and -CN; and R 21 , is selected from the group consisting of H and (C 1 -C 4 )-alkyl.
  • R 1 is methyl
  • R 3 is selected from the group consisting of:
  • R 3 is
  • W 1 , W 2 , W 3 and W 4 are each H;
  • W 1 is F, W 2 , W 3 and W 4 are each H; or
  • W 1 and W 2 are each F, W 3 and W 4 are each H.
  • W 1 is F
  • W 2 , W 3 and W 4 are each H.
  • Z is NH
  • Z is selected from the group consisting of O, and NH;
  • R 1 is selected from the group consisting of H, and -(C 1 -C 4 )-alkyl;
  • R 3 is a nitrogen-bearing heterocycle selected from the group consisting of , wherein zero, one or two hydrogens on the -CH 2 - groups of the nitrogen-bearing heterocycle is replaced with halogen, -OH, -CN, -CF 3 or (C 1 -C 4 )-alkyl;
  • R 35 is H or (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ; and R 50 is selected from the group consisting of halogen, -OH, -O-(C 1 -C 4 )- alkyl, CF 3 , and -CN; Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • W 1 is F
  • W 2 is H or F; W 3 is H; and
  • W 4 is H.
  • Z is O.
  • R 2 is selected from the group consisting of:
  • R 1 is methyl
  • R 3 is
  • R 35 is methyl or isopropyl.
  • W 2 is H.
  • Z is selected from the group consisting of O, and NH;
  • R 1 is selected from the group consisting of H, and -(C 1 -C 4 )-alkyl;
  • R 3 is a nitrogen-bearing heterocycle selected from the group consisting of , wherein zero, one or two hydrogens on any of the -CH 2 - groups of the nitrogen-bearing heterocycle is replaced with halogen, -OH, -CN, -CF 3 or (C 1 -C 4 )-alkyl;
  • R 35 is H or (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ; and R 50 is selected from the group consisting of halogen, -OH, -O-(C 1 -C 4 )- alkyl, CF 3 , and -CN; Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • W 1 is F, Cl or OMe
  • W 2 is H or F; W 3 is H; and
  • W 4 is H.
  • Z is O.
  • R 1 is methyl
  • R 3 is
  • R 35 is methyl or isopropyl.
  • W 2 is H.
  • Z is selected from the group consisting of O, and NH;
  • R 1 is selected from the group consisting of H, and -(C 1 -C 4 )-alkyl;
  • R 3 is a nitrogen-bearing heterocycle selected from the group consisting of , wherein zero, one or two hydrogens on any of the -CH 2 - groups of the nitrogen-bearing heterocycle is replaced with halogen, -OH, -CN, -CF 3 or (C 1 -C 4 )-alkyl;
  • R 35 is H or (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ; and R 50 is selected from the group consisting of halogen, -OH, -O-(C 1 -C 4 )- alkyl, CF 3 , and -CN;
  • R 2 is Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN;
  • Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • W 1 , W 2 , W 3 , W 4 are independently of one another selected from the group consisting of H, halogen, -OR 21 , -CF 3 , (C 1 -C 4 )-alkyl, and -CN, R 21 is selected from the group consisting of H and (C 1 -C 4 )-alkyl.
  • Z is O.
  • R 1 is methyl
  • R 3 is
  • R 35 is methyl or isopropyl.
  • W 1 is H, F, Cl or OMe; W 2 is H or F; W 3 is H; and
  • W 4 is H.
  • W 1 is F
  • W 2 is H
  • W 3 is H and W 4 is H.
  • W 1 is Cl
  • W 2 is H
  • W 3 is H and W 4 is H.
  • a compound selected from the group consisting of: or a pharmaceutically acceptable sale thereof is provided.
  • Z is selected from the group consisting of O and NH;
  • R 3 is selected from the group consisting of: -(CH 2 ) P -N(R 33 )R 34 , p is 2, 3 or 4;
  • R 33 and R 34 are independently from one another a (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ;
  • R 35 is H or a (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ;
  • R 50 is selected from the group consisting of halogen, -OR 27 , -O-(C 1 -C 4 )-alkyl, -CF 3 , and -CN;
  • R 2 is Z 1 and Z 2 are independently of one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN;
  • Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN; and
  • W 1 is halogen
  • R 3 is . In some embodiments, R 35 is methyl or isopropyl.
  • R 3 is . In some embodiments, R 35 is H.
  • Z-R 3 is selected from the group consisting of
  • Z-R 3 is selected from the group consisting of and
  • Z-R 3 is selected from the group consisting of and In some embodiments, Z-R 3 is
  • W 1 is F. In other embodiments, W 1 is Cl.
  • R 2 is Z 1 and Z 2 can be independently from one another selected from the group consisting of Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe.
  • R 2 is In some embodiments, R 2 is
  • R 2 is .
  • Z 3 can be selected from the group consisting of H, Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe, or from the group consisting of H, -CH 3 , -CF 3 , -OCH(CH 3 ) 2 and -OMe, or from the group consisting of -CH 3 , -OCH(CH 3 ) 2 and - OMe.
  • the compound of Formula IV is a compound of Formula I a: or a pharmaceutically acceptable salt thereof, wherein: R 2 is selected from the group consisting of:
  • W 1 is selected from the group consisting of Cl and F;
  • R 27 is selected from the group consisting of: H,
  • the compound of Formula IV is a compound of Formula IVb: or a pharmaceutically acceptable salt thereof, wherein:
  • R 2 is selected from the group consisting o
  • W 1 is selected from the group consisting of Cl and F.
  • the compound of Formula IV is a compound of Formula IVc:
  • R 2 is selected from the group consisting of:
  • W 1 is selected from the group consisting of Cl and F;
  • R 35 is selected from the group consisting of methyl and isopropyl.
  • the compound of Formula IV is a compound of Formula IVd: or a pharmaceutically acceptable salt thereof, wherein: R 2 is selected from the group consisting of: W 1 is selected from the group consisting of Cl and F.
  • the compound of Formula IV is selected from the group consisting of Compounds 28, 38, 78, 79, 84, 85, 99, 100, 101, 102, 103,104, 105, 107, 106, 108, 109, 110, 111, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124,
  • Z-R 3 is selected from the group consisting of: and R 2 is selected from the group consisting of
  • W 1 is selected from the group consisting of Cl and F. In some embodiments, W 1 is Cl. In other embodiments, W 1 is F. In some embodiments, R 2 is . in some embodiments, R 27 is H. In some embodiments, Z-R 3 is
  • Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN;
  • Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • W 1 is selected from the group consisting of H and halogen;
  • R 33 is -CH 3 or -(CH 2 )-(CH 2 )-OR 2 7;
  • W 1 is F. In some embodiments, W 1 is Cl..
  • R 2 is 2 .
  • Z 1 and Z 2 can be independently from one another selected from the group consisting of Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe.
  • R 2 is In some embodiments, R 2 is
  • R 2 is .Z 3 can be selected from the group consisting of H, Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe, or from the group consisting of H, -CH 3 , - CF 3 , -OCH(CH 3 ) 2 and -OMe, or from the group consisting of -CH 3 , -OCH(CH 3 ) 2 and - OMe.
  • R 33 is -CH 3 . In other embodiments, R 33 is-(CH 2 )-(CH 2 )-OR 27 . In some embodiments, R 27 is selected from the group consisting of H and
  • the compound of Formula V is a compound of Formula Va: or a pharmaceutically acceptable salt thereof, wherein: R 2 is selected from the group consisting of:
  • W 1 is selected from the group consisting of H, Cl and F;
  • R 27 is selected from the group consisting of: H,
  • the compound of Formula V is a compound of Formula Vb: or a pharmaceutically acceptable salt thereof, wherein: R 2 is selected from the group consisting of:
  • W 1 is selected from the group consisting of H, Cl and F.
  • the compound of Formula V is selected from the group consisting of Compounds 9, 20, 22, 24, 27, 30, 38, 45, 72, 73, 78, 84, 85, 99, 100, 103, 104, 105, 106, 107, 108, 109, 110, 111 , 112, 113, 114, 115, 116, 117, 118, 119, 120, 121 , 122, 123, 124, 125, 150, 151 , 152 and 153, as numbered in Table A hereinbelow, or a pharmaceutically acceptable salt thereof.
  • Formula Vb or a pharmaceutically acceptable salt thereof wherein: Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN; and
  • W 1 is selected from the group consisting of H and halogen. In some embodiments, W 1 is F. In other embodiments, W 1 is Cl.
  • R 2 is z 2 .
  • Z 1 and Z 2 can be independently from one another selected from the group consisting of Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe.
  • R 2 is
  • R 2 is .
  • Z 3 can be selected from the group consisting of H, Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe, or from the group consisting of H, -CH 3 , - CF 3 , -OCH(CH 3 ) 2 and -OMe, or from the group consisting of -CH 3 , -OCH(CH 3 ) 2 and - OMe.
  • the compound of Formula Vb is selected from the group consisting of Compounds 9, 38, 45, 84, 85 and 111 , as numbered in Table A hereinbelow, or a pharmaceutically acceptable salt thereof.
  • the compound of Formula Vb is selected from the group consisting of Compounds 24, 27, 30, 73, 100, 103, 104, 105, 106, 107, 108, 109, 110, 113 and 112, as numbered in Table A hereinbelow, or a pharmaceutically acceptable salt thereof.
  • Y 1 is H or F; q is 0 or 1; Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • W 1 is selected from the group consisting of H and halogen
  • R 35 is H or a (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ;
  • R 50 is selected from the group consisting of halogen, -OR 27 , -O-(C 1 -C 4 )-alkyl, -CF 3 , and -CN;
  • Y 1 is H.
  • the compound of Formula VI is selected from the group consisting of:
  • Formula Vic Formula Vid or a pharmaceutically acceptable salt thereof.
  • the compound of Formula VI is a compound of Formula Via: or a pharmaceutically acceptable salt thereof.
  • q 1.
  • W 1 is F. In other embodiments, W 1 is Cl.
  • R 2 is .
  • Z 1 and Z 2 can be independently from one another selected from the group consisting of Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe.
  • R 2 is In some embodiments, R 2 is .
  • Z 3 can be selected from the group consisting of H, Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe, or from the group consisting of H, -CH 3 , - CF 3 , -OCH(CH 3 ) 2 and -OMe, or from the group consisting of -CH 3 , -OCH(CH 3 ) 2 and - OMe.
  • R 35 is H or a (C 1 -C 4 )-alkyl a (C 1 -C 4 )-alkyl which is unsubstituted.
  • R 35 is H.
  • the compound of Formula VI is a compound of Formula Via: or a pharmaceutically acceptable salt thereof, wherein: R 2 is selected from the group consisting of:
  • W 1 is selected from the group consisting of H, Cl and F.
  • the compound of Formula VI is selected from the group consisting of Compounds 29, 41 , 42, 44, 46, 47, 49, 50, 53, 54, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 66, 67, 69, 70, 71, 92, 155 and 156, as numbered in Table A hereinbelow, or a pharmaceutically acceptable salt thereof.
  • the compound of Formula VI is preferably not a compound of Formula Via in racemic form: or a pharmaceutically acceptable salt thereof, wherein:
  • Y 2 is H or F; and R 52 is selected from the group consisting of:
  • Formula Vb or a pharmaceutically acceptable salt thereof wherein: Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN; and
  • W 1 is selected from the group consisting of H and halogen.
  • Formula V or a pharmaceutically acceptable salt thereof wherein: Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • W 1 is selected from the group consisting of H and halogen
  • R 33 is -CH 3 or -(CH 2 )-(CH 2 )-OR 2 7;
  • Z 1 and Z 2 are independently from one another selected from the group consisting of Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe.
  • W 1 is selected from the group consisting of H, Cl and F; and o
  • R 27 is selected from the group consisting of: H, The compound of embodiment 14, which is: or a pharmaceutically acceptable salt thereof, wherein:
  • W 1 is selected from the group consisting of H, Cl and F.
  • Y 1 is H or F; q is 0 or 1; Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • W 1 is selected from the group consisting of H and halogen
  • R 35 is H or a (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ;
  • R 50 is selected from the group consisting of halogen, -OR 27 , -O-(C 1 -C 4 )-alkyl, -CF 3 , and -CN;
  • Z 1 and Z 2 are independently from one another selected from the group consisting of Cl, F, -CH 3 , -CN, -OCH(CH 3 ) 2 and -OMe.
  • W 1 is selected from the group consisting of H, Cl and F.
  • Z is selected from the group consisting of O and NH;
  • R 3 is selected from the group consisting of: -(CH 2 ) P -N(R 33 )R 34 , p is 2, 3 or 4;
  • R 33 and R 34 are independently from one another a (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ;
  • R 35 is H or a (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ;
  • R 50 is selected from the group consisting of halogen, -OR 27 , -O-(C 1 -C 4 )-alkyl, -CF 3 , and -CN;
  • Z 1 and Z 2 are independently of one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN;
  • Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN; and
  • W 1 is halogen
  • W 1 is selected from the group consisting of Cl and F;
  • W 1 is selected from the group consisting of Cl and F.
  • R 35 is selected from the group consisting of methyl and isopropyl.
  • W 1 is selected from the group consisting of Cl and F.
  • Z-R 3 is selected from the group consisting of:
  • R 27 is selected from the group consisting of: H,
  • W 1 is selected from the group consisting of Cl and F. .
  • Z is selected from the group consisting of O, CH 2 , S and NH;
  • R 1 is selected from the group consisting of H, and -(C 1 -C 4 )-alkyl;
  • R 3 is selected from the group consisting of -(CH 2 ) P -N(R 33 )R 34 ; p is 1 , 2, 3 or 4; R 2 is selected from the group consisting of a 5-membered or 6-membered monocyclic, aromatic or heteroaromatic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, wherein R 2 is unsubstituted or substituted by one or more identical or different substituents R 20 ; R 20 is selected from the group consisting of halogen, -CF 3 , (C 1 -C 4 )-alkyl, -OR 21 , -N(R 22 )R 23 , (C 1 -C 4 )-alkyl-OR 24 , (C 1 -C 4 )-alkyl-N(R 25 )R 26 and -CN; R 21 , R 22 , R 23 , R 24 , R 25 and R 26 , are independently of one
  • Z is selected from the group consisting of a direct bond, O, S, CH(R 9 ) and N(R 10 );
  • R 1 is selected from the group consisting of H, -N(R 11 )R 12 , -N(R 13 )-C(O)-R 14 , -NR 13 -S(O) 2 "R 15 , -NR 13 -C(O)-NH-R 16 , -(C 1 -C 4 )-alkyl, — (C 1 -C 4 )-alkyl-OR 17 and — (C 1 -C 4 )-alkyl-N(R 18 )R 19 ;
  • R 3 is selected from the group consisting of H, (C 1 -C 8 )-alkyl, R 30 and (C 1 -C 4 )-alkyl- R 30 , wherein (C 1 -C 8 )-alkyl is unsubstituted or substituted by one or more identical or different substituents R 31 ;
  • R 30 is a 3-membered to 12-membered, monocyclic or bicyclic, saturated, partially unsaturated or aromatic, cyclic group which comprises 0, 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, which is unsubstituted or substituted by one or more identical or different substituents R 32 ;
  • R 31 is selected from the group consisting of halogen, -OH, -CF 3 , -O-(C 1 -C 4 )-alkyl, -N(R 33 )-R 34 and -CN;
  • R 32 is selected from the group consisting of halogen
  • R 2 is a 6-membered monocyclic, heteroaromatic group which comprises 1 or 2 nitrogen atoms, wherein R 2 is unsubstituted or substituted by one or more identical or different substituents R 20 ;
  • R 20 is selected from the group consisting of halogen, -CF 3 , (C 1 -C 4 )-alkyl, -OR 21 , -N(R 22 )R 23 , (C 1 -C 4 )-alkyl-OR 24 ,
  • R 38 , R 39 , R 40 , R 41 , R 42 and R 43 are independently of one another selected from the group consisting of H and (C 1 -C 4 )-alkyl;
  • R 1 is selected from the group consisting of -(C 1 -C 4 )-alkyl, and -(C 1 -C 4 )- alkyl-N(R 18 )R 19 .
  • Z is selected from the group consisting of a direct bond, O, S, CH(R 9 ) and N(R 10 );
  • R 1 is selected from the group consisting of H, -N(R 11 )R 12 , -N(R 13 )-C(O)-R 14 , -NR 13 -S(O) 2 "R 15 , -NR 13 -C(O)-NH-R 16 , -(C 1 -C 4 )-alkyl, — (C 1 -C 4 )-alkyl-OR 17 and — (C 1 -C 4 )-alkyl-N(R 18 )R 19 ;
  • R 3 is selected from the group consisting of H, (C 1 -C 8 )-alkyl, R 30 and (C 1 -C 4 )-alkyl- R 30 , wherein (C 1 -C 8 )-alkyl is unsubstituted or substituted by one or more identical or different substituents R 31 ;
  • R 30 is a 3-membered to 12-membered, monocyclic or bicyclic, saturated, partially unsaturated or aromatic, cyclic group which comprises 0, 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, which is unsubstituted or substituted by one or more identical or different substituents R 32 ;
  • R 31 is selected from the group consisting of halogen, -OH, -CF 3 , -O-(C 1 -C 4 )-alkyl, -N(R 3 3)-R 34 and -CN;
  • R 32 is selected from the group consisting of halogen
  • A is a direct bond or -CH 2 -;
  • Y is selected from the group consisting of carbocyclylene and heterocyclylene, which is unsubstituted or substituted by one or more identical or different substituents R 5 ;
  • R 5 is selected from the group consisting of halogen, (C 1 -C 4 )-alkyl, -O-( C 1 -C 4 )- alkyl and -CN; when Y is not 1 ,4-phenylene, or when Y is 1 ,4-phenylene and R 1 is -(C 1 -C 4 )- alkyl-N(R 18 )R 19 :
  • R 2 is selected from the group consisting of from the group consisting of (C 1 -C 4 )-alkyl, (C 3 -C 7 )-cycloalkyl, (C 2 -C 4 )-alkenyl, (C 2 -C 4 )-alkynyl, phenyl and a 5- membered or 6-membered monocyclic,
  • R 3 8, R 39 , R40, R41 , R42 and R43 are independently of one another selected from the group consisting of H and (C 1 -C 4 )-alkyl;
  • Y is selected from the group consisting of arylene and heteroarylene, which is unsubstituted or substituted by one or more identical or different substituents R 5 .
  • Z is selected from the group consisting of O, CH 2 , S and NH;
  • R 1 is selected from the group consisting of H, and -(C 1 -C 4 )-alkyl;
  • R 3 is selected from the group consisting of -(CH 2 ) P -N(R 33 )R 34 ; p is 1 , 2, 3 or 4; R 2 is a 6-membered monocyclic, heteroaromatic group which comprises 1 or 2 nitrogen atoms, wherein R 2 is unsubstituted or substituted by one or more identical or different substituents R 20 ; R 20 is selected from the group consisting of halogen, -CF 3 , (C 1 -C 4 )-alkyl, -OR 21 , -N(R 22 )R 23 , (C 1 -C 4 )-alkyl-OR 24 , (C 1 -C 4 )-alkyl-N(R 25 )R 26 and -CN; R 21 , R 22 , R 23 , R 24 , R 25 and R 26 , are independently of one another selected from the group consisting of H and (C 1 -C 4 )-alkyl;
  • Z is selected from the group consisting of O, CH 2 , S and NH;
  • R 1 is selected from the group consisting of H, and -(C 1 -C 4 )-alkyl;
  • R 3 is selected from the group consisting of -(CH 2 ) P -N(R 33 )R 34 , wherein zero, one or two hydrogen atoms of the group -(CH 2 ) P - are independently replaced with F; p is 1 , 2, 3 or 4; R 2 is selected from the group consisting of a 5-membered or 6-membered monocyclic, aromatic or heteroaromatic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, wherein R 2 is unsubstituted or substituted by one or more identical or different substituents R 20 ; R 20 is selected from the group consisting of halogen, -CF 3 , (C 1 -C 4 )-alkyl, -OR 21 , -N(R 22 )R 23 , (C 1 -C 4 )-alkyl-OR 24 , (C 1 -C 4 )-alkyl-N(R 25 )R 26 and
  • R 50 is selected from the group consisting of halogen, -OR 27 , -O-(C 1 -C 4 )-alkyl, CF 3 , and -CN;
  • W 1 , W 2 , W 3 , W 4 are independently of one another selected from the group consisting of H, halogen, -OR 21 , -SH, -CF 3 , (C 1 -C 4 )-alkyl, and -CN.
  • Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; and Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN.
  • W 1 is F or Cl, W 2 , W 3 and W 4 are each H; or
  • W 1 and W 2 are each F, W 3 and W 4 are each H.
  • Z is selected from the group consisting of O and NH;
  • R 1 is selected from the group consisting of H and (C 1 -C 4 )-alkyl;
  • R 3 is -(CH 2 ) P -N(R 33 )R 34 ; p is 2, 3 or 4; Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN;
  • Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • R 33 and R 34 are independently of one another a (C 1 -C 4 )-alkyl
  • Wi, W 2 , W 3 , W 4 are independently of one another selected from the group consisting of H, halogen, -OR 2 I, -CF 3 , (C 1 -C 4 )-alkyl, and -CN,
  • R 2 I is selected from the group consisting of H and (C 1 -C 4 )-alkyl.
  • W 1 , W 2 , W 3 and W 4 are each H;
  • W 1 is F or Cl, W 2 , W 3 and W 4 are each H; or
  • W 1 and W 2 are each F, W 3 and W 4 are each H.
  • a compound of Formula III or a pharmaceutically acceptable salt thereof, wherein:
  • Z is selected from the group consisting of O, and NH;
  • R 1 is selected from the group consisting of H, and -(C 1 -C 4 )-alkyl;
  • R 3 is a nitrogen-bearing heterocycle selected from the group consisting of or two hydrogens on any of the -CH 2 - groups of the nitrogen-bearing heterocycle is replaced with halogen, -OH, -CN, -CF 3 or (C 1 -C 4 )-alkyl;
  • R 35 is H or (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ; and R 50 is selected from the group consisting of halogen, -OH, -O-(C 1 -C 4 )- alkyl, CF 3 , and -CN; Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • W 1 is F, OMe or Cl
  • W 2 is H or F; W 3 is H; and
  • W 4 is H.
  • R 3 is a nitrogen-bearing heterocycle selected from the group consisting of and
  • R 35 is H or (C 1 -C 4 )-alkyl which is unsubstituted or substituted by one or more identical or different substituents R 50 ; and R 50 is selected from the group consisting of halogen, -OH, -O-(C 1 -C 4 )- alkyl, CF 3 , and -CN; Z1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN; Z 3 is selected from the group consisting of H, halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 and -CN;
  • W 1 , W 2 , W 3 , W 4 are independently of one another selected from the group consisting of H, halogen, -OR 2 I, -CF 3 , (C 1 -C 4 )-alkyl, and -CN,
  • R 2 I is selected from the group consisting of H and (C 1 -C 4 )-alkyl.
  • W 1 is H, F, OMe or CI
  • W 2 is H or F; W 3 is H; and
  • W 4 is H.
  • a pharmaceutical composition comprising the compound as defined in any one of embodiments 1 to 174 or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient.
  • cancer affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
  • embodiment 177 wherein the cancer is a melanoma, liposarcoma, lung cancer, breast cancer, prostate cancer, leukemia, kidney cancer, esophageal cancer, brain cancer, lymphoma, colon cancer or colorectal cancer.
  • embodiment 177 wherein the cancer is prostate cancer, colorectal cancer or breast cancer.
  • embodiment 181 wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI- 587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclidectact
  • embodiment 181 wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
  • a cardiovascular disease selected from the group consisting of Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, and stent failure.
  • cancer affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
  • embodiment 191 wherein the cancer is a melanoma, liposarcoma, lung cancer, breast cancer, prostate cancer, leukemia, kidney cancer, esophageal cancer, brain cancer, lymphoma, colon cancer or colorectal cancer.
  • embodiment 191 wherein the cancer is prostate cancer, colorectal cancer or breast cancer.
  • embodiment 195 wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI- 587), PQR 3 09 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclidectact
  • embodiment 195 wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
  • a cardiovascular disease selected from the group consisting of Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, and stent failure.
  • a method for the treatment of cancer comprising administering to a subject a therapeutically effective amount of the compound as defined in any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof.
  • cancer affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
  • cancer is a melanoma, liposarcoma, lung cancer, breast cancer, prostate cancer, leukemia, kidney cancer, esophageal cancer, brain cancer, lymphoma, colon cancer or colorectal cancer.
  • the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF- 05212384 (Gedatolisib, PKI-587), PQR 3 09 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101, Idelalisib), GSK2636771 , INCB050465 (Parsaclisi
  • a method for the treatment of epilepsy comprising administering to a subject a therapeutically effective amount of a compound as defined in any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof.
  • a method for the treatment of a cardiovascular disease comprising administering to a subject a therapeutically effective amount of a compound as defined in any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, the cardiovascular disease being selected from the group consisting of Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, and stent failure. 213.
  • a method for the treatment of Long QT syndrome the method comprising administering to a subject a therapeutically effective amount of a compound as defined in any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof.
  • Y 1 is H or F;
  • Z 1 and Z 2 are independently from one another selected from the group consisting of halogen, (C 1 -C 4 )alkyl, -OH, -O-(C 1 -C 4 )alkyl, -CF 3 , and -CN;
  • W 1 is selected from the group consisting of H and halogen
  • R 35 is H or methyl.
  • Z 2 is F
  • W 1 is H, Cl or F.
  • the compound of embodiment 216, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer which is: or a pharmaceutically acceptable salt thereof.
  • the compound of embodiment 216 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer which is a compound of Formula VI lb:
  • Z 2 is F; R 35 is H or methyl; and
  • W 1 is H, Cl or F.
  • the compound of embodiment 216, for use in the treatment of cancer which is: or a pharmaceutically acceptable salt thereof.
  • the compound of embodiment 243, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer wherein the at least one inhibitor of AKT/PI3K/mT0R is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI- 587), PQR 3 09 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib),
  • the compound of any one of embodiments 216 to 245, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer wherein the cancer affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
  • tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pit
  • the compound of any one of embodiments 216 to 245, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer wherein the cancer is a melanoma, liposarcoma, lung cancer, breast cancer, prostate cancer, leukemia, kidney cancer, esophageal cancer, brain cancer, lymphoma, colon cancer or colorectal cancer.
  • a pharmaceutical composition for use in the treatment of cancer, comprising the compound of any one of embodiments 216 to 242, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • composition of embodiment 249 further comprising at least one inhibitor of AKT/PI3K/mTOR.
  • composition of embodiment 250, for the treatment of cancer wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI-587), PQR 3 09 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL- 101 (GS-1101 , Idelalisib), GSK2636771 , INCB0504
  • composition of embodiment 250 for use in the treatment of cancer, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
  • a method for the treatmenf of cancer comprising administering to a subject in need thereof the compound of any one of embodiments 216 to 242, or a pharmaceutically acceptable salt thereof.
  • the prepared compounds were in general characterized by spectroscopic data and chromatographic data, in particular mass spectra (MS) and/or nuclear magnetic resonance (NMR) spectra. 'H-NMR spectra were generally recorded at 600 MHz. In the NMR characterization, the chemical shift ⁇ (in ppm), the number of hydrogen atoms (H), the coupling constant J (in Hz) and the multiplicity (s: singlet, d: doublet, dd: double doublet, t: triplet, dt: double triplet, m: multiplet; br: broad) of the peaks are given.
  • the mass number (m/z) of the peak of the molecular ion (M) or of a related ion such as the ion [M+1], i.e. the protonated molecular ion [M+H)] or the ion [M- 1], which was formed depending on the ionization method used, is given.
  • the ionization method was electrospray ionization (ES+ or ES-).
  • the resulting mixture was concentrated under reduced pressure.
  • the crude product was purified by Prep-HPLC with the following conditions: Column: XBridge Prep C18 OBD Column, 19*150mm 5um; Mobile Phase A:Water(10MMOL/L NH 4 HCO 3 +0.1 %NH 3 .H 2 O), Mobile Phase B:ACN; Flow rate:25 mL/min; Gradient:18 B to 38 B in 7 min; Director: 220 nm.
  • the resulting solution was stirred for 40 hours at 100 °C in an oil bath.
  • the reaction mixture was cooled with a water bath.
  • the resulting solution and E08786-007 were diluted with 20 mL of water.
  • the resulting mixture was washed with 2 x20 ml of DCM.
  • the resulting mixture was concentrated.
  • the residue was applied onto a C18 gel with H 2 O (0.5% NH4HCO3)/ACN (90:10 to 10:90) in 45 minutes.
  • the collected fractions were combined and concentrated.
  • 6-chloro-3-methyl-N-[2-(morpholin-4- yl)ethyl]-1 H-pyrazolo[3,4-d]pyrimidin-4-amine (200.00 mg, 0.674 mmol, 1.00 equiv.), dioxane (16.00 mL), H 2 O (4.00 mL), 5-chloro-2-fluoro-N-[4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenyl]benzenesulfonamide (305.19 mg, 0.741 mmol, 1.10 equiv.), Cs 2 CO 3 (658.75 mg, 2.022 mmol, 3 equiv.), Pd(dppf)CI 2 (49.31 mg, 0.067 mmol, 0.1 equiv.).
  • the resulting solution was stirred for 3 hr at 100 °C.
  • the resulting solution was extracted with 3x50 mL of ethyl acetate dried in an oven under reduced pressure and concentrated.
  • the crude product was purified by Flash-Prep-HPLC with the following conditions: Column, silica gel C18 (210 g); mobile phaseA: Water-10 mM NH 4 HCO 3 , mobile phaseB:Acetonitrile; Flow rate:50 mL/min; Gradient:55 B to 60 B; 254 nm;. The solution was concentrated. The solid was washed with CH 3 CN (3 mLx2). The solid was collected by filtration.
  • the reaction mixture was cooled to ambient temperature and filtered.
  • the filtrate was purified by prep-HPLC with the following condition: Column: YMC-Actus Triart C18, 30 mm X 150 mm, 5um; Mobile Phase A: Water(0.05%TFA ), Mobile Phase B: ACN; Flow rate:60 mL/min; Gradient:20% B to 50% B in 7 min; 254 nm; RT:5.12.
  • reaction mixture was purged with nitrogen 3 times and stirred at 100 °C for 12 hours under nitrogen.
  • the reaction was cooled to the room temperature and neutralized with saturated aqueous 1 N HCI to pH 7 ⁇ 8 and washed by DCM (3*100 mL).
  • the combined organic phase was dried over Na 2 SO 4 and concentrated under reduced pressure to provide the crude. Then the crude purified by HPLC chromatography to provide the desired product (350.0 mg, 90% purity, off-white solid).
  • the reaction mixture was purged with nitrogen 3 times and stirred at 100 °C for 18 h under nitrogen.
  • the reaction was cooled to the room temperature and neutralized with saturated aqueous 1 N HCI to pH 7 ⁇ 8 and washed by DCM (3*5 mL).
  • the combined organic phase was dried over Na 2 SO 4 and concentrated under reduced pressure to provide the crude.
  • the crude purified by reversed-phase chromatography(10 mmol/L NH 4 HCO 3 /ACN) to provide the desired product (75 mg, 50% purity, off-white solid).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Biomedical Technology (AREA)
  • Hospice & Palliative Care (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Compounds of Formula I: Formula I and pharmaceutically acceptable salts thereof are provided for as inhibitors of SGK-1 for example for the treatment of conditions such as Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, stent failure, cancer, Parkinson's disease and Lafora disease.

Description

PYRAZOLO[3,4-D]PYRIMIDIN-6-YL-SULFONAMIDE DERIVATIVES FOR THE INHIBITION OF SGK-1 AND TREATMENT OF CANCER
TECHNICAL FIELD
The technical field relates to pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives and pharmaceutical compositions that inhibit SGK-1 , and more particularly relates to pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives and pharmaceutical compositions for the treatment of heart conditions treatable by SGK-1 inhibition such as Long QT syndrome, and for the treatment of cancer.
BACKGROUND
Serine/threonine-protein kinase (SGK-1) (also known as serum/glucocorticoid-regulated kinase 1) is a protein kinase that plays a role in a cell's response to stress. SGK-1 activates certain potassium, sodium, and chloride channels. For instance, SGK-1 is known to regulate the myo-inositol transporter during osmotic stress. Several challenges remain in the development of an SGK-1 inhibitor for the treatment of heart conditions such as LOTS, and/or for the treatment of cancer.
SUMMARY
In a first aspect, described herein is a compound of Formula VII:
Figure imgf000003_0001
Formula VII or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer or tumor or in the manufacture of a medicament for the treatment of cancer or tumor, wherein:
Y1 is H or F;
Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; W1 is selected from the group consisting of H and halogen; and
R35 is H or methyl.
In a further aspect, described herein is a pharmaceutical composition comprising one or more compounds or pharmaceutically acceptable salts thereof as defined herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, for use in the treatment of cancer or tumor or in the manufacture of a medicament for the treatment of cancer or tumor.
In a further aspect, described herein is a combination comprising one or more compounds or pharmaceutically acceptable salts thereof as defined herein and at least one inhibitor of AKT, PI3K, and/or mTOR.
In a further aspect, described herein is a combination comprising one or more compounds or pharmaceutically acceptable salts thereof as defined herein and at least one inhibitor of AKT, PI3K, mTOR, PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1, SOS2, SHP1 , and/or MAPK.
In a further aspect, described herein is a combination comprising one or more compounds or pharmaceutically acceptable salts thereof as defined herein and ipatasertib, alpelisib, everolimus, lapatinib, AT13148, gedatolisib, BAY-1082439, GSK112212, dabrafenib, trametinib, erlotinib, and/or GSK2334470.
In a further aspect, described herein is a combination comprising at least two compounds or pharmaceutically acceptable salts thereof as defined herein.
In a further aspect, described herein is a use of the pharmaceutical composition defined herein or the combination defined herein for the treatment of cancer or tumor or for the manufacture of a medicament for the treatment of cancer or tumor.
In a further aspect, described herein is a method for the treatment of cancer, comprising administering to a subject in need thereof one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein.
In a further aspect, described herein is a method for the treatment of cancer or tumor in a subject, comprising: i. determining that cells from said cancer or tumor (e.g., cancer or tumor sample or biopsy): have an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; have an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; have a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; have an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIO; have an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by one or more concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN; have an upregulation of a mitogen activated protein kinase (MAPK) gene expression (e.g., CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and SPRY4); and/or are resistant to one or more anticancer therapies (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof); and ii. administering to the subject one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein.
In a further aspect, described herein is a use of one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein, for the treatment of cancer or tumor or for the manufacture of a medicament for the treatment of cancer or tumor, wherein the cancer or tumor (e.g., cancer or tumor sample or biopsy) has been predetermined as: having an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; having an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; having a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIO; having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by one or more concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN; having an upregulation of a mitogen activated protein kinase (MAPK) gene expression (e.g., CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and SPRY4); and/or being resistant to one or more anticancer therapies (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof).
In a further aspect, described herein is a use of one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein, for the treatment of one or more side effects or toxicities caused by an anticancer therapy in a subject undergoing said anticancer therapy.
In a further aspect, described herein is a method for treating one or more side effects or toxicities caused by an anticancer therapy, comprising administering to a subject undergoing said anticancer therapy one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein.
In a further aspect, described herein is a method for monitoring the progression of an anticancer therapy in a subject that was previously administered with one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical composition defined herein, or the combination defined herein, wherein the method comprises determining in a sample from said subject: a loss of phosphorylation of NDRG1 ; an increase in phosphorylation of MAPK14 (i.e. , p38); an increase in phosphorylation of MAPK14 (i.e., p38); an increase in phosphorylation of heat shock protein family B1 (i.e., HSPB1 or HSP27); an increase in phosphorylation of tumor suppressor p53 (TP53); an increase in phosphorylation of ribosomal protein S6 kinase A1 (RSK1); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERKI); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERK1); and/or an increase in phosphorylation of AKT1 substrate 1 (PRAS40).
BRIEF DESCRIPTION OF THE FIGURES
Figs. 1A and 1B is a graph (Fig. 1A) and a Dose-response matrix (Fig. 1B) showing synergistic combination of Compound 79 with AKT inhibitor Ipatasertib on the growth of JIMT-1 breast cancer cells.
Figs. 2A and 2B is a graph (Fig. 2A) and a Dose-response matrix (Fig. 2B) showing synergistic combination of Compound 79 with AKT inhibitor Capivasertib.
Figs. 3A and 3B is a graph (Fig. 3A) and a Dose-response matrix (Fig. 3B) showing synergistic combination of Compound 79 with AKT inhibitor MK-2206.
Fig. 4A is a Western blot showing inhibition of NDRG1 phosphorylation in PBMCs by Compound 79. Fig. 4B a Western blot showing inhibition of NDRG1 phosphorylation in BT-459 breast cancer cells by Compound 79.
Fig. 5 is a series of Western blots showing inhibition of NDRG1 phosphorylation in ex vivo treated blood of healthy volunteers by Compound 84.
Fig. 6 is a series of Western blots showing NDRG1 is a direct target of SGK1 phosphorylation. Figs. 7A-7B shows the effect of the combination of Compound 156 with the PIK3CA inhibitor Alpelisib (Fig. 7A) or the AKT inhibitor Ipatasertib (Fig. 7B) on the growth of JIMT- 1 breast cancer cells.
Figs. 8A-8B shows the effect of the combination of Compound 175 with the PIK3CA inhibitor Alpelisib (Fig. 8A) or the AKT inhibitor Ipatasertib (Fig. 8B) on the growth of JIMT- 1 breast cancer cells.
Figs. 9A-9C shows the effect of the combination of Compounds 79 (Fig. 9A), 175 (Fig. 9B), or 156 (Fig. 9C) with the PIK3CA inhibitor Alpelisib on the growth of BT-549 breast cancer cells.
Figs. 10A-10D shows the effect of the combination of Compound 79 with the mTOR inhibitor Everolimus (Fig. 10A), the HER2/EGFR inhibitor Lapatinib (Fig. 10B), the multi- AGC kinase inhibitor AT13148 (Fig. 10C), or the mT0R/PI3K inhibitor Gedatolisib (Fig. 10D) on the growth of JIMT-1 breast cancer cells.
Figs. 11A-11C shows the effect of the combination of Compound 79 with the mTOR inhibitor Everolimus (Fig. 11A), the PIK3a/b/g inhibitor BAY-1082439 (Fig. 11 B), or the MEK inhibitor GSK112212 (Fig. 11C) on the growth of BT-549 breast cancer cells.
Figs. 12A-12C shows the effect of the combination of Compounds 79 (Fig. 12A), 175 (Fig. 12B), or 156 (Fig. 12C) with the PIK3CA inhibitor Alpelisib on the growth of HT-29 colorectal cancer cells.
Figs. 13A-13C shows the effect of the combination of Compounds 79 (Fig. 13A), 175 (Fig. 13B), or 156 (Fig. 13C) with the BRAF inhibitor Dabrafenib on the growth of HT-29 colorectal cancer cells.
Figs. 14A-14D shows the effect of the combination of Compound 79 with the PI3K/mT0R inhibitor Gedatolisib (Fig. 14A), the MEK inhibitor Trametinib (Fig. 14B), the EGFR inhibitor Erlotinib (Fig. 14C), or the PDK1 inhibitor GSK2334470 (Fig. 14D) on the growth of HT-29 colorectal cancer cells. Figs. 15A-15B shows the effect of the combination of Compound 79 with the BRAF inhibitor Dabrafenib (Fig. 15A) or the MEK inhibitor Trametinib (Fig. 15B) on the growth of 8305c anaplastic thyroid cancer cells.
Figs. 16A-16D shows the effect of the triple combination of Compound 79 (5 μM [Fig. 16A]; 2.5 μM [Fig. 16B]; 1.25 μM [Fig. 16C]; and 0.63 μM [Fig. 16D]) with the BRAF inhibitor Dabrafenib and the MEK inhibitor Trametinib on the growth of 8305c anaplastic thyroid cancer cells.
Figs. 17A-17F shows the results from Figs. 16A-16D at different concentrations of Compound 79 (5 μM [Fig. 17A], 2.5 μM [Fig. 17B], 1.25 μM [Fig. 17C], and 0.625 μM [Fig. 17D]) in combination with Dabrafenib (0.08 nM) and Trametinib (0.08 nM). Figs. 17E-F show the results from Figs. 16A-16D at 5 μM of Compound 79 in combination with Dabrafenib (5 nM) and Trametinib (5 nM [Fig. 17E]) and 1.25 nM [Fig.17F],
Figs. 18A-18B shows the effect of the combination of Compound 79 with the BRAF inhibitor Dabrafenib (Fig. 18A) or the MEK inhibitor Trametinib (Fig. 18B) on the growth of A375 melanoma cancer cells.
Figs. 19A-19B shows the effect of the combination of Compound 79 with the MEK inhibitor Trametinib (Fig. 19A) or the EGFR inhibitor Erlotinib (Fig. 19B) on the growth of SW403 colorectal cancer cells.
Figs. 20A-20B shows the effect of the combination of Compound 79 with the BRAF inhibitor Dabrafenib (Fig. 20A) or the MEK inhibitor Trametinib (Fig. 20B) on the growth of RKO colorectal cancer cells that are resistant to Dabrafenib.
Figs. 21A-21S’ shows the proliferation results obtained from the 45 PIK3CA mutant cell lines screened after treatment with compound 79 alone, the PIK3CA inhibitor Alpelisib alone, or in combination. Graphs represent viability (%) with respect to concentration (μM).
Fig. 22 shows the synergistic effect of the combination of compound 79 and the PIK3CA inhibitor Alpelisib alone on PIK3CA mutant cancer cell lines. Cell lines were grouped according to the level of viability decreased when treated with the combination at different compound 79 concentrations. Group 1 represents cell lines whereby there was >10% decrease in viability when combined with ≤ 1 μM Compound 79; Group 2 represents cell lines whereby there was a 5-10% decrease in viability when combined with < 1 μM Compound 79; Group 3 represents cell lines whereby there was <10% decrease in viability when with any concentration of compound 79; and Group 1 represents cell lines whereby there was >10% decrease in viability when combined with >1 μM Compound 79;
Fig. 23 shows the enrichment of RAS/MAPK activating mutations in cell lines associated with synergy between Compound 79 and Alpelisib in inhibiting proliferation. Cell lines were grouped as shown in Fig. 22.
Figs. 24A-24D shows the luminal and basal breast cancer tumor signatures associated with poor synergy of the combination of compound 79 and Alpelisib.
Figs. 25A-25E shows the upregulation of ribosomal signatures that are enriched in cell lines that showed synergy when Compound 79 and Alpelisib were combined compared to cell lines that did not show synergy.
Figs. 26A-26B show the results of the Western Blot of the phosphorylation of ribosomal protein S6 upon incubation with Alpelisib and Compound 79 in HT-29 colorectal cancer cells at 4 hours and 24 hours (Fig. 26A) and at different concentrations of Compound 79 (Fig. 26B).
Figs. 27A-27B show the Kegg-Ribosome (Fig. 27A) and Reactome Translation initiation (Fig. 27B) activity scores in the grouped cell lines as described in Fig. 22.
Fig. 28A-28D shows the gene sets from MsigDB (The Molecular Signatures Database) (Fig. 28A) that are significantly upregulated in Group 1 vs. Group 3 cell lines, according to the grouping described in Fig. 22. Figs. 28-C-28D correspond to the genes specified in Tables X-Z.
Figs. 29A-29D show the Anaplastic thyroid cancer (ATC) tumor xenograft model using 8305c cells treated with Compound 79 (45 mg/kg QD), BRAF inhibitor dabrafenib (30 mg/kg QD), MEK inhibitor trametinib (0.6 mg/kg QD) or the triple combination showing tumor volume (Fig. 29A), total body weight (Fig. 29B), tumor response (waterfall pit) (Fig, 29C), and tumor volumes in individual mice after treatment with the triple combination.
Fig. 30A shows the tumor volumes of the Anaplastic thyroid cancer (ATC) tumor xenograft model using 8305c cells, whereby mice were treated with BRAF inhibitor dabrafenib (30 mg/kg QD) and MEK inhibitor trametinib (0.6 mg/kg QD) at Day 0 and Compound 79 (45 mg/kg QD) at Day 13. Fig. 30B shows the tumor volumes of the same model as Fig. 30A whereby mice were treated with Compound 79 (45 mg/kg QD) at Day 0 and BRAF inhibitor dabrafenib (30 mg/kg QD) and MEK inhibitor trametinib (0.6 mg/kg QD) at Day 13. Fig. 30C shows the tumor waterfall of the different groups of the model of Figs. 30A and 30B.
Figs. 31A-31 B shows the phosphoproteomic array of peripheral blood mononuclear cells (PBMCs) incubated with 1 uM of Compound 79 for 4 hours.
Figs. 32A and 32B show the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof. Fig. 32C and 32D show the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof. Figs. 32E and 32F shows the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (5, 15, or 45 mg/kg mg/kg), or combinations thereof.
Figs. 33A-33B shows the results of mice blood C-peptide levels (Fig. 33A) and insulin levels (Fig. 33B) after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof. Fig. 33C shows the results of mice blood C-peptide after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK- 2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof. Figs. 33D-33E shows the results of mice blood C-peptide levels (Fig. 33D) and insulin levels (Fig. 33E) after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (5, 15, or 45 mg/kg), or combinations thereof. Fig. 33F shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof. Fig. 33G shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof. Fig. 33H shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (5, 15, or 45 mg/kg), or combinations thereof.
DETAILED DESCRIPTION
The present description relates to compounds of Formula I, or pharmaceutically acceptable salts thereof.
Figure imgf000012_0001
Formula I
The compounds of Formula I and their pharmaceutically acceptable salts are pharmacologically active compounds that modulate protein kinase activity, specifically the activity of serum and glucocorticoid regulated kinase isoform 1 (SGK-1). The compounds of Formula I or their pharmaceutically acceptable salts can be suitable for the treatment of conditions in which SGK-1 activity is inappropriate. Non-limiting examples of such conditions can include Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, stent failure, cancer and epilepsy. The compounds of Formula I and their pharmaceutically acceptable salts are described in more detail herein.
Definitions
Unless stated otherwise, the following terms and phrases as used herein are intended to have the following meanings. The fact that a particular term or phrase is not specifically defined should not be correlated to indefiniteness or lacking clarity, but rather terms herein are used within their ordinary meaning. When trade names are used herein, it is intended to independently include the tradename product and the active pharmaceutical ingredient(s) of the tradename product. “Alkyl” is hydrocarbon containing primary, secondary or tertiary carbon atoms. For example, an alkyl group can have 1 to 20 carbon atoms (i.e, C1-C20 alkyl), 1 to 8 carbon atoms (i.e., C1-C8 alkyl), or 1 to 4 carbon atoms (i.e., C1-C4 alkyl). Examples of suitable alkyl groups include, but are not limited to, methyl (Me, — CH3), ethyl (Et, — CH2CH3), 1- propyl (n-Pr, n-propyl, — CH2CH2CH3), 2-propyl (i-Pr, i-propyl, — CH(CH3)2), 1-butyl (n-Bu, n-butyl, — CH2CH2CH2CH3), 2-methyl-1 -propyl (i-Bu, i-butyl, — CH2CH(CH3)2), 2-butyl
(s-Bu, s-butyl, — CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, — C(CH3)3), 1-pentyl
(n-pentyl, — CH2CH2CH2CH2CH3), 2-pentyl (— CH(CH3)CH2CH2CH3), 3-pentyl
(— CH(CH2CH3)2), 2-methyl-2-butyl (— C(CH3)2CH2CH3), 3-methyl-2-butyl
(— CH(CH3)CH(CH3)2), 3-methyl-1 -butyl (— CH2CH2CH(CH3)2), 2-methyl-1 -butyl
(— CH2CH(CH3)CH2CH3), 1-hexyl (— CH2CH2CH2CH2CH2CH3), 2-hexyl
(— CH(CH3)CH2CH2CH2CH3), 3-hexyl (— CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (— C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (— CH(CH3)CH(CH3)CH2CH3), 4-methyl-2- pentyl (— CH(CH3)CH2CH(CH3)2), 3-methyl-3-pentyl (— C(CH3)(CH2CH3)2), 2-methyl-3- pentyl (— CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (— C(CH3)2CH(CH3)2), 3,3- dimethyl-2-butyl (— CH(CH3)C(CH3)3, and octyl (— (CH2)7CH3).
Alkoxy” means a group having the formula — O-alkyl, in which an alkyl group, as defined above, is attached to the parent molecule via an oxygen atom. The alkyl portion of an alkoxy group can have 1 to 20 carbon atoms (i.e., C1-C20 alkoxy), 1 to 12 carbon atoms (i.e., C1-C12alkoxy), or 1 to 4 carbon atoms (i.e., C1-C4alkoxy). Examples of suitable alkoxy groups include, but are not limited to, methoxy ( — O — CH3 or — OMe), ethoxy ( — OCH2CH3 or — OEt), t-butoxy ( — O — C(CH3)3 or -OtBu), and the like.
“Haloalkyl” is an alkyl group, as defined above, in which one or more hydrogen atoms of the alkyl group is replaced with a halogen atom. The alkyl portion of a haloalkyl group can have 1 to 20 carbon atoms (i.e., C1-C20 haloalkyl), 1 to 12 carbon atoms (i.e., C1-C12 haloalkyl), or 1 to 4 carbon atoms (i.e., C1-C4 haloalkyl). Examples of suitable haloalkyl groups include, but are not limited to, —CF3, — CHF2, — CFH2, — CH2CF3, and the like.
“Cycloalkyl” means a mono or bicyclic carbocyclic ring functional group including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, bicyclo[2.2.1]heptanyl, bicyclo[3.2.1]octanyl, and bicyclo[5.2.0]nonanyl. The cycloalkyl can have 3 to 12 carbon atoms (i.e., C3-C12 cycloalkyl), 3 to 7 carbon atoms (i.e., C3-C7 cycloalkyl) or 3 to 6 carbon atoms (i.e., C3-C6 cycloalkyl). Unless otherwise indicated, the term “( C3-C7)cycloalkyl” refers to a cycloalkyl group containing from 3 to 8 carbons. Thus, the term “(C3-C7)cycloalkyl” encompasses a monocyclic cycloalkyl group containing from 3 to 7 carbons and a bicyclic cycloalkyl group containing from 6 to 7 carbons.
“Alkenyl” is a hydrocarbon containing primary, secondary or tertiary carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp2 double bond. For example, an alkenyl group can have 2 to 20 carbon atoms (i.e., C2-C20 alkenyl), 2 to 12 carbon atoms (i.e., C2-C12 alkenyl), or 2 to 6 carbon atoms (i.e., C2-C6 alkenyl). Examples of suitable alkenyl groups include, but are not limited to, ethylene, vinyl ( — CH=CH2), allyl ( — CH2CH=CH2), cyclopentenyl (— C5H7), and 5-hexenyl (— CH2CH2CH2CH2CHMDH2).
“Alkynyl” is a hydrocarbon containing primary, secondary or tertiary carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp triple bond. For example, an alkynyl group can have 2 to 20 carbon atoms (i.e., C2-C20 alkynyl), 2 to 12 carbon atoms (i.e., C2-C12 alkynyl), or 2 to 6 carbon atoms (i.e., C2-C6 alkynyl). Examples of suitable alkynyl groups include, but are not limited to, acetylenic ( — C=CH), propargyl ( — CH2C=CH), and the like.
“Alkylene” refers to a saturated, branched or straight hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. For example, an alkylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkylene radicals include, but are not limited to, methylene ( — CH2 — ), 1 ,1-ethylene (— CH(CH3)— ), 1 ,2-ethylene (— CH2CH2— ), 1 ,1-propylene (— CH(CH2CH3)— ), 1 ,2- propylene ( — CH2CH(CH3) — ), 1 ,3-propylene ( — CH2CH2CH2 — ), 1 ,4-butylene (— CH2CH2CH2CH2— ), and the like.
“Alkenylene” refers to an unsaturated, branched or straight hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene. For example, and alkenylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkenylene radicals include, but are not limited to, 1 ,2-ethylene ( — CH=CH — ). “Alkynylene” refers to an unsaturated, branched or straight hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne. For example, an alkynylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkynylene radicals include, but are not limited to, acetylene ( — C=C — ), propargyl ( — CH2C=C— ), and 4-pentynyl (— CH2CH2CH2C=C— ).
“Aryl” means a monovalent aromatic hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. For example, an aryl group can have 6 to 20 carbon atoms, 6 to 14 carbon atoms, or 6 to 12 carbon atoms. Typical aryl groups include, but are not limited to, radicals derived from benzene (e.g., phenyl), substituted benzene, naphthalene, anthracene, biphenyl, and the like.
“Arylene” refers to an aryl as defined above having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent aryl. Typical arylene radicals include, but are not limited to, phenylene, such as 1 ,4- phenylene.
“Arylalkyl” refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl radical. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, naphthobenzyl, 2-naphthophenylethan-1-yl and the like. The arylalkyl group can comprise 6 to 20 carbon atoms, e.g., the alkyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
“Arylalkenyl” refers to an acyclic alkenyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also an sp2 carbon atom, is replaced with an aryl radical. The aryl portion of the arylalkenyl can include, for example, any of the aryl groups disclosed herein, and the alkenyl portion of the arylalkenyl can include, for example, any of the alkenyl groups disclosed herein. The arylalkenyl group can comprise 6 to 20 carbon atoms, e.g., the alkenyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
“Arylalkynyl” refers to an acyclic alkynyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also an sp carbon atom, is replaced with an aryl radical. The aryl portion of the arylalkynyl can include, for example, any of the aryl groups disclosed herein, and the alkynyl portion of the arylalkynyl can include, for example, any of the alkynyl groups disclosed herein. The arylalkynyl group can comprise 6 to 20 carbon atoms, e.g., the alkynyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
As used herein, the “halogen” refers to F, Cl, Br, or I.
As used herein the term “haloalkyl” refers to an alkyl group, as defined herein, that is substituted with at least one halogen. Examples of branched or straight chained “haloalkyl” groups as used herein include, but are not limited to, methyl, ethyl, propyl, isopropyl, n- butyl, and t-butyl substituted independently with one or more halogens, for example, fluoro, chloro, bromo, and iodo. The term “haloalkyl” should be interpreted to include such substituents as perfluoroalkyl groups such as — CF3.
The term “substituted” in reference to alkyl, aryl, arylalkyl, carbocyclyl, heterocyclyl, and other groups used herein, for example, “substituted alkyl”, “substituted cycloalkyl”, “substituted aryl”, “substituted arylalkyl”, “substituted heterocyclyl”, and “substituted carbocyclyl” means a group, alkyl, alkylene, aryl, arylalkyl, heterocyclyl, carbocyclyl respectively, in which one or more hydrogen atoms are each independently replaced with a non-hydrogen substituent. Typical substituents include, but are not limited to, — X, — R, — O— , =0, —OR, —SR, — S— , — NR2, — N(+)R3, =NR, — CX3, — CRX2, — CR2X, — CN, — OCN, —SON, — N=C=O, — NCS, —NO, — NO2, =N2, — N3, — NRC(=O)R, — NRC(=O)OR, — NRC(=O)NRR, — C(=O)NRR, — C(=O)OR, — OC(=O)NRR, — OC(=O)OR, — C(=O)R, — S(=O)2OR, — S(=O)2R, — OS(=O)2OR, — S(=O)2NR, — S(=O)R, — NRS(=O)2R, — NRS(=O)2NRR, — NRS(=O)2OR, — OP(=O)(OR)2, — P(=O)(OR)2, — P(O)(OR)(O)R, — C(=O)R, — C(=S)R, — C(=O)OR, — C(=S)OR, — C(=O)SR, — C(=S)SR, — C(=O)NRR, — C(=S)NRR, — C(=NR)NRR,
— NRC(=NR)NRR, where each X is independently a halogen: F, Cl, Br, or I; and each R is independently H, alkyl, cycloalkyl, aryl, arylalkyl, a heterocycle, or a protecting group or prodrug moiety. Divalent groups may also be similarly substituted.
Those skilled in the art will recognize that when moieties such as “alkyl”, “aryl”, “heterocyclyl”, etc. are substituted with one or more substituents, they could alternatively be referred to as “alkylene”, “arylene”, “heterocyclylene”, etc. moieties (i.e. , indicating that at least one of the hydrogen atoms of the parent “alkyl”, “aryl”, “heterocyclyl” moieties has been replaced with the indicated substituent(s)). When moieties such as “alkyl”, “aryl”, “heterocyclyl”, etc. are referred to herein as “substituted” or are shown diagrammatically to be substituted (or optionally substituted, e.g., when the number of substituents ranges from zero to a positive integer), then the terms “alkyl”, “aryl”, “heterocyclyl”, etc. are understood to be interchangeable with “alkylene”, “arylene”, “heterocyclylene”, etc.
“Heteroalkyl” refers to an alkyl group where one or more carbon atoms have been replaced with a heteroatom, such as, O, N, or S. For example, if the carbon atom of the alkyl group which is attached to the parent molecule is replaced with a heteroatom (e.g., O, N, or S) the resulting heteroalkyl groups are, respectively, an alkoxy group (e.g., — OCH3, etc.), an amine (e.g., — NHCH3, — N(CH3)2, and the like), or a thioalkyl group (e.g., — SCH3). If a non-terminal carbon atom of the alkyl group which is not attached to the parent molecule is replaced with a heteroatom (e.g., O, N, or S) and the resulting heteroalkyl groups are, respectively, an alkyl ether (e.g., — CH2CH2 — O — CH3, etc.), an alkyl amine (e.g., — CH2NHCH3, — CH2N(CH3)2, and the like), or a thioalkyl ether (e.g., — CH2— S— CH3). If a terminal carbon atom of the alkyl group is replaced with a heteroatom (e.g., O, N, or S), the resulting heteroalkyl groups are, respectively, a hydroxyalkyl group (e.g., — CH2CH2 — OH), an aminoalkyl group (e.g., — CH2NH2), or an alkyl thiol group (e.g., — CH2CH2 — SH). A heteroalkyl group can have, for example, 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. A C1-C6 heteroalkyl group means a heteroalkyl group having 1 to 6 carbon atoms.
“Heterocycle” or “heterocyclyl” as used herein includes by way of example and not limitation those heterocycles described in Paquette, Leo A.; Principles of Modern Heterocyclic Chemistry (W. A. Benjamin, New York, 1968), particularly Chapters 1 , 3, 4, 6, 7, and 9; The Chemistry of Heterocyclic Compounds, A Series of Monographs” (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566. In one specific embodiment of the invention “heterocycle” includes a “carbocycle” as defined herein, wherein one or more (e.g. 1 , 2, 3, or 4) carbon atoms have been replaced with a heteroatom (e.g. O, N, P or S). The terms “heterocycle” or “heterocyclyl” includes saturated rings, partially unsaturated rings, and aromatic rings (i.e., heteroaromatic rings). Heterocycles includes aromatic and non- aromatic mono-, bi-, and poly-cyclic rings, whether fused, bridged, or spiro. As used herein, the term “heterocycle” encompasses, but is not limited to “heteroaryl.” Examples of heterocycles include by way of example and not limitation pyridyl, dihydroypyridyl, tetrahydropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, azetidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofuranyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, octahydroisoquinolinyl, azocinyl, triazinyl, 6H-1 ,2,5-thiadiazinyl, 2H.6H-1 ,5,2-dithiazinyl, thienyl, thianthrenyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathinyl, 2H- pyrrolyl, isothiazolyl, isoxazolyl, pyrazinyl, pyridazinyl, indolizinyl, isoindolyl, 3H-indolyl, 1 H-indazoly, purinyl, 4H-quinolizinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, quinazolinyl, cinnolinyl, pteridinyl, 4aH-carbazolyl, carbazolyl, p-carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, furazanyl, phenoxazinyl, isochromanyl, chromanyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperazinyl, indolinyl, isoindolinyl, quinuclidinyl, morpholinyl, oxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl and isatinoyl.
By way of example and not limitation, carbon bonded heterocycles are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1 , 3, 4, 5, 6, 7, or 8 of an isoquinoline. Still more typically, carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3- pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5- pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4- thiazolyl, or 5-thiazolyl.
By way of example and not limitation, nitrogen bonded heterocycles are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3- pyrazoline, piperidine, piperazine, indole, indoline, 1 H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or p-carboline. Still more typically, nitrogen bonded heterocycles include 1-aziridyl, 1-azetedyl, 1-pyrrolyl, 1-imidazolyl, 1-pyrazolyl, and 1-piperidinyl. “Heterocyclylene” refers to a heterocyclyl, as defined herein, derived by replacing a hydrogen atom from a carbon atom or heteroatom of a heterocyclyl, with an open valence. Similarly, “heteroarylene” refers to an aromatic heterocyclylene.
“Heterocyclylalkyl” refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heterocyclyl radical (i.e. , a heterocyclyl-alkylene- moiety). Typical heterocyclyl alkyl groups include, but are not limited to heterocyclyl-CH2 — , 2-(heterocyclyl)ethan-1-yl, and the like, wherein the “heterocyclyl” portion includes any of the heterocyclyl groups described above, including those described in Principles of Modern Heterocyclic Chemistry. One skilled in the art will also understand that the heterocyclyl group can be attached to the alkyl portion of the heterocyclyl alkyl by means of a carbon-carbon bond or a carbon- heteroatom bond, with the proviso that the resulting group is chemically stable. The heterocyclyl alkyl group comprises 2 to 20 carbon atoms, e.g., the alkyl portion of the arylalkyl group comprises 1 to 6 carbon atoms and the heterocyclyl moiety comprises 1 to 14 carbon atoms. Examples of heterocyclylalkyls include by way of example and not limitation 5-membered sulfur, oxygen, and/or nitrogen containing heterocycles such as thiazolylmethyl, 2-thiazolylethan-1-yl, imidazolylmethyl, oxazolylmethyl, thiadiazolylmethyl, and the like, 6-membered sulfur, oxygen, and/or nitrogen containing heterocycles such as piperidinylmethyl, piperazinylmethyl, morpholinylmethyl, pyridinylmethyl, pyridizylmethyl, pyrimidylmethyl, pyrazinylmethyl, and the like.
“Heterocyclylalkenyl” refers to an acyclic alkenyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also a sp2 carbon atom, is replaced with a heterocyclyl radical (i.e., a heterocyclyl-alkenylene- moiety). The heterocyclyl portion of the heterocyclyl alkenyl group includes any of the heterocyclyl groups described herein, including those described in Principles of Modern Heterocyclic Chemistry, and the alkenyl portion of the heterocyclyl alkenyl group includes any of the alkenyl groups disclosed herein. One skilled in the art will also understand that the heterocyclyl group can be attached to the alkenyl portion of the heterocyclyl alkenyl by means of a carbon-carbon bond or a carbon-heteroatom bond, with the proviso that the resulting group is chemically stable. The heterocyclyl alkenyl group comprises 2 to 20 carbon atoms, e.g., the alkenyl portion of the heterocyclyl alkenyl group comprises 1 to 6 carbon atoms and the heterocyclyl moiety comprises 1 to 14 carbon atoms. “Heteroaryl” refers to a monovalent aromatic heterocyclyl having at least one heteroatom in the ring. Non-limiting examples of suitable heteroatoms which can be included in the aromatic ring include oxygen, sulfur, and nitrogen. Non-limiting examples of heteroaryl rings include all of those listed in the definition of “heterocyclyl”, including pyridinyl, pyrrolyl, oxazolyl, indolyl, isoindolyl, purinyl, furanyl, thienyl, benzofuranyl, benzothiophenyl, carbazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, quinolyl, isoquinolyl, pyridazyl, pyrimidyl, pyrazyl, and the like.
“Carbocycle” or “carbocyclyl” refers to a saturated, partially unsaturated or aromatic ring having 3 to 7 carbon atoms as a monocycle, 7 to 12 carbon atoms as a bicycle, and up to about 20 carbon atoms as a polycycle. Monocyclic carbocycles have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms. Bicyclic carbocycles have 7 to 12 ring atoms, e.g., arranged as a bicyclo (4,5), (5,5), (5,6) or (6,6) system, or 9 or 10 ring atoms arranged as a bicyclo (5,6) or (6,6) system. Carbocycles includes aromatic and non-aromatic mono-, bi-, and poly-cyclic rings, whether fused, bridged, or spiro. Non-limiting examples of monocyclic carbocycles include the cycloalkyls group such as cyclopropyl, cyclobutyl, cyclopentyl, 1 -cyclopent- 1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1- cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl or aryl groups such as phenyl, and the like. Thus, “carbocycle,” as used herein, encompasses but is not limited to “aryl”, “phenyl” and “biphenyl.”
“Carbocyclylene” refers to a carbocyclyl or carbocycle as defined above having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent carbocyclyl. Typical carbocyclylene radicals include, but are not limited to, phenylene. Thus, “carbocyclylene,” as used herein, encompasses but is not limited to “arylene.”
“Carbocyclylalkyl” refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a carbocyclyl radical as defined above. Typical carbocyclylalkyl groups include, but are not limited to the arylalkyl groups such as benzyl, 2-phenylethan-1-yl, naphthylmethyl, 2- naphthylethan-1-yl, naphthobenzyl, 2-naphthophenylethan-1-yl or the cycloalkylalkyl groups such as cyclopropylmethyl, cyclobutylethyl, cyclohexyl methyl and the like. The arylalkyl group can comprise 6 to 20 carbon atoms, e.g., the alkyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms. The cycloalkylalkyl group can comprise 4 to 20 carbon atoms, e.g., the alkyl moiety is 1 to 6 carbon atoms and the cycloalkyl group is 3 to 14 carbon atoms.
“Arylheteroalkyl” refers to a heteroalkyl as defined herein, in which a hydrogen atom, which may be attached either to a carbon atom or a heteroatom, has been replaced with an aryl group as defined herein. The aryl groups may be bonded to a carbon atom of the heteroalkyl group, or to a heteroatom of the heteroalkyl group, provided that the resulting aryl heteroalkyl group provides a chemically stable moiety. For example, an arylheteroalkyl group can have the general formulae -alkylene-O-aryl, -alkylene-O-alkylene-aryl, - alkylene-NH-aryl, -alkylene-NH-alkylene-aryl, -alkylene-S-aryl, -alkylene-S-alkylene-aryl, and the like. In addition, any of the alkylene moieties in the general formulae above can be further substituted with any of the substituents defined or exemplified herein.
“Heteroarylalkyl” refers to an alkyl group, as defined herein, in which a hydrogen atom has been replaced with a heteroaryl group as defined herein. Non-limiting examples of heteroaryl alkyl include — CH2-pyridinyl, — CH2-pyrrolyl, — CH2-oxazolyl, — CH2-indolyl, — CH2-isoindolyl, — CH2-purinyl, — CH2-furanyl, — CH2-thienyl, — CH2-benzofuranyl, — CH2- benzothiophenyl, — CH2-carbazolyl, — CH2-imidazolyl, — CH2-thiazolyl, — CH2-isoxazolyl, — CH2-pyrazolyl, — CH2-isothiazolyl, — CH2-quinolyl, — CH2-isoquinolyl, — CH2-pyridazyl, — CH2-pyrimidyl, — CH2-pyrazyl, — CH(CH3)-pyridinyl, — CH(CH3)-pyrrolyl, — CH(CH3)- oxazolyl, — CH(CH3)-indolyl, — CH(CH3)-isoindolyl, — CH(CH3)-purinyl, — CH(CH3)- furanyl, — CH(CH3)-thienyl, — CH(CH3)-benzofuranyl, — CH(CH3)-benzothiophenyl, — CH(CH3)-carbazolyl, — CH(CH3)-imidazolyl, — CH(CH3)-thiazolyl, — CH(CH3)-isoxazolyl, — CH(CH3)-pyrazolyl, — CH(CH3)-isothiazolyl, — CH(CH3)-quinolyl, — CH(CH3)- isoquinolyl, — CH(CH3)-pyridazyl, — CH(CH3)-pyrimidyl, — CH(CH3)-pyrazyl, and the like.
The term “optionally substituted” in reference to a particular moiety of the compound of the Formulae of the invention, for example an optionally substituted aryl group, refers to a moiety having 0, 1 , or more substituents.
The term “prodrug” as used herein refers to any compound that when administered to a biological system generates the drug substance, i.e., active ingredient, as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), photolysis, and/or metabolic chemical reaction(s). A prodrug is thus a covalently modified analog or latent form of a therapeutically active compound. One skilled in the art will recognize that substituents and other moieties of the compounds of the present description should be selected in order to provide a compound which is sufficiently stable to provide a pharmaceutically useful compound which can be formulated into an acceptably stable pharmaceutical composition.
Some compounds of the present description and their pharmaceutically acceptable salts may exist as different polymorphs or pseudopolymorphs. As used herein, crystalline polymorphism means the ability of a crystalline compound to exist in different crystal structures. Polymorphism generally can occur as a response to changes in temperature, pressure, or both. Polymorphism can also result from variations in the crystallization process.
Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility, and melting point. The crystalline polymorphism may result from differences in crystal packing (packing polymorphism) or differences in packing between different conformers of the same molecule (conformational polymorphism). As used herein, crystalline pseudopolymorphism means the ability of a hydrate or solvate of a compound to exist in different crystal structures. The pseudopolymorphs of some of the compounds of the present description may exist due to differences in crystal packing (packing pseudopolymorphism) or due to differences in packing between different conformers of the same molecule (conformational pseudopolymorphism). It is understood that all polymorphs and pseudopolymorphs of the compounds described herein and their pharmaceutically acceptable salts are included within the scope of the present description.
The compounds of the present description and their pharmaceutically acceptable salts may exist as an amorphous solid. As used herein, an amorphous solid is a solid in which there is no long-range order of the positions of the atoms in the solid. This definition applies as well when the crystal size is two nanometers or less. Additives, including solvents, may be used to create amorphous forms the compounds of the present description.
Certain of the compounds described herein contain one or more chiral centers or may otherwise be capable of existing as multiple stereoisomers. The scope of the present description includes mixtures of stereoisomers as well as purified enantiomers or enantiomerically/diastereomerically enriched mixtures. Also included within the scope of the present description are the individual isomers of the compounds described herein, as well as any wholly or partially equilibrated mixtures thereof. The compounds of the present description and their pharmaceutically acceptable salts also includes the individual isomers of the compounds represented by the formulas above as mixtures with isomers thereof in which one or more chiral centers are inverted.
The compounds of the present description may exist in solvated, for example hydrated, as well as unsolvated forms. Typically, but not absolutely, the salts of the compounds of the present description are pharmaceutically acceptable salts. Salts encompassed within the term “pharmaceutically acceptable salts” refer to non-toxic salts of the compounds of the present description.
Examples of suitable pharmaceutically acceptable salts include inorganic acid addition salts such as chloride, bromide, sulfate, phosphate, and nitrate; organic acid addition salts such as acetate, galactarate, propionate, succinate, lactate, glycolate, malate, tartrate, citrate, maleate, fumarate, methanesulfonate, p-toluenesulfonate, and ascorbate; salts with acidic amino acid such as aspartate and glutamate; alkali metal salts such as sodium salt and potassium salt; alkaline earth metal salts such as magnesium salt and calcium salt; ammonium salt; organic basic salts such as trimethylamine salt, triethylamine salt, pyridine salt, picoline salt, dicyclohexylamine salt, and N,N'-dibenzylethylenediamine salt; and salts with basic amino acid such as lysine salt and arginine salt. The salts may be in some cases hydrates or ethanol solvates.
The definitions and substituents for various genus and subgenus of the present compounds are described and illustrated herein. It should be understood by one skilled in the art that any combination of the definitions and substituents described above should not result in an inoperable species or compound. “Inoperable species or compounds” means compound structures that violates relevant scientific principles (such as, for example, a carbon atom connecting to more than four covalent bonds) or compounds too unstable to permit isolation and formulation into pharmaceutically acceptable dosage forms.
Pharmaceutical compositions
The compounds of the present description can be formulated with conventional carriers and excipients, which will be selected in accordance with ordinary practice. Tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form, and when intended for delivery by other than oral administration generally will be isotonic. All formulations will optionally contain excipients such as those set forth in the Handbook of Pharmaceutical Excipients (1986), herein incorporated by reference in its entirety. Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH of the formulations ranges from about 3 to about 11 but is ordinarily about 7 to 10.
While it is possible for the active ingredients to be administered alone it may be preferable to present them as pharmaceutical formulations. The formulations of the invention, both for veterinary and for human use, comprise at least one active ingredient, together with one or more acceptable carriers and optionally other therapeutic ingredients.
The carrier(s) must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and physiologically innocuous to the recipient thereof.
The formulations include those suitable for the foregoing administration routes. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa.), herein incorporated by reference in its entirety. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be administered as a bolus, electuary or paste.
A tablet is made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent.
Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingredient.
Pharmaceutical formulations according to the present description include one or more compounds together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents. Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil. Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth herein, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
Dispersible powders and granules of the invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.
The pharmaceutical compositions may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally- occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
The pharmaceutical compositions of the invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned herein. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1 ,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables.
The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weightweight). The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion may contain from about 3 to 500 pg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
The formulations are presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
The compounds of the present description can also be formulated to provide controlled release of the active ingredient to allow less frequent dosing or to improve the pharmacokinetic or toxicity profile of the active ingredient. Accordingly, there is also provided compositions comprising one or more compounds of the present description formulated for sustained or controlled release.
The effective dose of an active ingredient depends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses) or against an active disease or condition, the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. The effective dose can be expected to be from about 0.0001 to about 10 mg/kg body weight per day, typically from about 0.001 to about 1 mg/kg body weight per day, more typically from about 0.01 to about 1 mg/kg body weight per day, even more typically from about 0.05 to about 0.5 mg/kg body weight per day. For example, the daily candidate dose for an adult human of approximately 70 kg body weight will range from about 0.05 mg to about 100 mg, or between about 0.1 mg and about 25 mg, or between about 0.4 mg and about 4 mg, and may take the form of single or multiple doses. SGK-1 and associated conditions
The present description relates to compounds or pharmaceutically acceptable salts thereof, for the treatment various conditions treatable by inhibiting SGK-1. For example, the condition can be Long QT syndrome (LQTS), such as genetic LQTS or acquired LQTS, or other cardiovascular diseases (e.g., dilated cardiomyopathy - genetic or acquired) that are treatable by inhibiting SGK-1. Without being bound by theory, it is believed that SGK- 1 inhibition in vivo has a protective effect and can alleviate symptoms associated with LQTS; can reduce and alleviate symptoms associated with heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, genetic or acquired dilated cardiomyopathy, hypertrophic cardiomyopathy, and stent failure.
Long QT syndrome (LQTS) can be genetic (e.g. caused by a mutation in the KCNQ1 gene, the KCNH2 gene, or the SCN5a gene). Alternatively, Long QT syndrome is not associated with a genetic mutation and is acquired as a result of exposure to an external stimulus. For instance, acquired Long QT syndrome can be a side effect of drugs such as erythromycin or haloperidol. Acquired Long QT syndrome is also associated with other heart conditions such as myocardial ischemia.
The present description also relates to compounds or pharmaceutically acceptable salts thereof, forthe treatment of other conditions related to SGK-1 mediated mechanisms, such as cancer, Parkinson’s disease and Lafora disease.
In some embodiments, the present description provides compounds or pharmaceutically acceptable salts thereof for treating cancer, tumor, or another proliferative disorder. As used herein, the terms “inhibition of cancer”, “inhibition of cancer cell proliferation”, and “inhibition of cancer invasion and metastasis” refer to the inhibition, or decrease in the rate, of the growth, division, maturation, viability, or ability to invade and colonize other organs and tissues of cancer cells, and/or causing the death of cancer cells, individually or in aggregate with other cancer cells, by cytotoxicity, nutrient depletion, induction of differentiation or apoptosis, or recognition by the immune system in order to elicit an immune response to the cancer cells.
Examples of tissues containing cancerous cells whose proliferation can be inhibited by a compound, salt or composition thereof described herein and against which the methods described herein are useful include but are not limited to breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
In some embodiments, the cancer treated by a provided compound, salt or composition thereof is a cancer or tumor that is a melanoma, liposarcoma, lung cancer (e.g., non-small cell lung cancer), breast cancer (e.g., ER+, ER-, HER2+, HER2-, PR-, PR+, triple negative, luminal, basal), Luminal androgen receptor (LAR)) prostate cancer, leukemia, kidney cancer, esophageal cancer, thyroid cancer (e.g., Anaplastic Thyroid Cancer (ATC), Poorly Differentiated Thyroid Cancer (PDTC)), ovarian cancer, endometrial cancer, Head and Neck Squamous Cell Carcinoma, brain cancer, lymphoma, rectal cancer, colon cancer, uterine cancer, cervical cancer, Hepatocellular carcinoma, Papillary thyroid carcinoma (PTC), or colorectal cancer
In some embodiments, the compounds of the present description can be used to treat cancer by inhibiting signaling of the AKT/PI3K/mTOR pathway in patients whose tumors have activation of this pathway through mutations in PIK3CA, AKT1 , and/or PTEN for example.
In some embodiments, the compounds of the present description can be used in combination with compounds that inhibit AKT/PI3K/mTOR signaling to treat cancer in patients whose tumors have activation of this pathway through mutations in PIK3CA, AKT1 , and/or PTEN for example. Non-limiting examples of AKT/PI3K/mTOR inhibitors include NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI-587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL- 101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK- 1117, MLN-1117, TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC- 907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-100-115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
The inhibitor of AKT, PI3K, and/or mTOR may be selected from the group consisting of vistusertib (AZD2014), NU7441 (KU-57788), KU-0063794, TGX-221 , RLY-2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223, ABTL0812, DCBCI0901, AZD-8055, sapanisertib, JR-AB2-011 , omipalisib, and torkinibm PI-103.
In some embodiments, the compounds of the present description can be used in combination with at least one inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK.
The inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK may be selected from the group consisting of dabrafenib, trametinib (GSK1120212), encorafenib (LGX818), binimetinib, cobimetinib, naporafenib (LXH254), sorafenib (BAY 43-9006), vemurafenib (PLX4032), PLX-4720, regorafenib (BAY 73- 4506), GDC_0879, RAF265 (CHIR-265), AZ 628, lifirafenib (BGB-283), NVP-BHG712, SB590885, ZM 336372, GW5074, TAK-632, agerafenib (RXDX-105), GNE-9815, TBAP- 001 , L-779450, belvarafenib (HM95573), AZ304, PLX8394, RAF709, CCT196969, BAW2881 (NVP-BAW2881), LY3009120, avutometinib (RO5126766, CH5126766), doramapimod (BIRB 796), MCP110, PLX7904, tovorafenib (MLN2480), selumetinib (AZD6244), mirdametinib (PD0325901), U0126, PD98059, PD184352 (CI-1040), BIX 02189, pimasertib (AS-70326), pelitinib (EKB-569), BIX 02188, TAK-733, AZD8330, binimetinib (MEK162), SL-327, refametinib (RDEA119), zapnometinib (PD0184264), GDC-0623, BI-847325, cobimetinib (GDC_0973), PD318088, SCH772984, ASN007, MRTX-1257, AZD0634 (ATG-017), MK-8353 (SCH900353), temuterkib (LY3214996), VX- 11e, ulixertinib (BVD-523), ravoxertinib (GDC-0994), exarafenib, ERAS-601 , BDTX-4933, ADT007, HM99462, DCC-3084, PHI-501 , MRTX1133. Sotorasib (AMG510), lonafamib (SCH66336), ASP2453, BI-3406, BI-2852, BAY-293, KRpep-2d, ARS-853, adagrasib (MRTX849), adagrasib (MRTX849), ARS1620, divarasib (GDC-6036), GDC-1971 , IACS- 13909, JAB-3068, RMC-4550, RMC-4630, SHP099, TNO155, GDC-1971 , HBI-2376, BBP-398 (IACS-15509), PF-07284892, selpercatinib (LOXO-292), pralsetinib (BLU-667), LOXO-260, TAS-0953/HM06, TPX-0046, EP0031 , APS03118, TP-0903, BGB324 (R428), bosutinib (SKI-606), and gilteritinib (ASP2215).
In some embodiments, the compounds of the present description can be combined with an inhibitor of the RAS/RAF/MAPK pathway in cells that have a mutation activating RAS, RAF, MEK, or ERK signaling. Non-limiting examples of RAF/RAS/MEK/ERK/SOS1/SOS2/SHP2 inhibitors include dabrafenib, trametinib (GSK1120212), encorafenib (LGX818), binimetinib, cobimetinib, naporafenib (LXH254), sorafenib (BAY 43-9006), vemurafenib (PLX4032), PLX-4720, regorafenib (BAY 73- 4506), GDC_0879, RAF265 (CHIR-265), AZ 628, lifirafenib (BGB-283), NVP-BHG712, SB590885, ZM 336372, GW5074, TAK-632, agerafenib (RXDX-105), GNE-9815, TBAP- 001 , L-779450, belvarafenib (HM95573), AZ304, PLX8394, RAF709, CCT196969, BAW2881 (NVP-BAW2881), LY3009120, avutometinib (RO5126766, CH5126766), doramapimod (BIRB 796), MCP110, PLX7904, tovorafenib (MLN2480), selumetinib (AZD6244), mirdametinib (PD0325901), U0126, PD98059, PD184352 (CI-1040), BIX 02189, pimasertib (AS-70326), pelitinib (EKB-569), BIX 02188, TAK-733, AZD8330, binimetinib (MEK162), SL-327, refametinib (RDEA119), zapnometinib (PD0184264), GDC-0623, BI-847325, cobimetinib (GDC_0973), PD318088, SCH772984, ASN007, MRTX-1257, AZD0634 (ATG-017), MK-8353 (SCH900353), temuterkib (LY3214996), VX- 11e, ulixertinib (BVD-523), ravoxertinib (GDC-0994), exarafenib, ERAS-601 , BDTX-4933, ADT007, HM99462, DCC-3084, PHI-501 , MRTX1133. Sotorasib (AMG510), lonafamib (SCH66336), ASP2453, BI-3406, BI-2852, BAY-293, KRpep-2d, ARS-853, adagrasib (MRTX849), adagrasib (MRTX849), ARS1620, divarasib (GDC-6036), GDC-1971 , IACS- 13909, JAB-3068, RMC-4550, RMC-4630, SHP099, TNO155, GDC-1971 , HBI-2376, BBP-398 (IACS-15509), and PF-07284892.
In some embodiments, the compounds of the present description can be combined with an inhibitor of the RTK, PI3K, AKT, RAS, RAF or MAPK pathway in cancer cells that, for example, have up-regulation of gene expression associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation.
In some embodiments, the compounds of the present description can be combined with an inhibitor of the PI3K or AKT pathway in cancer cells that, for example, have upregulation of gene signatures associated with basal and/or mesenchymal breast cancer. In some embodiments, the compounds of the present description can be combined with an inhibitor of the PI3K or AKT pathway in cancer cells that, for example, have a downregulation of gene signatures associated with luminal breast cancer.
Non-limiting examples of AKT/PI3K/mT0R inhibitors include PDK1 inhibitors, and other mT0RC1/2 inhibitors such as vistusertib (AZD2014), NU7441 (KU-57788), KU-0063794, TGX-221 , RLY-2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223, ABTL0812, DCBCI0901 , AZD-8055, sapanisertib, JR-AB2-011 , omipalisib, and torkinibm PI-103.
In some embodiments, the compounds of the present description can be used to treat cancer by inhibiting signaling of the RAF/MEK/ERK pathway in patients whose tumors have activation of this pathway through, for example, mutations in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIG.
In some embodiments, the compounds of the present description can be used to treat cancer by inhibiting signaling of the RAF/MEK/ERK and/or PI3K, AKT, mTOR pathway in patients whose tumors have concomitant activation of these pathways through, for example, concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN.
In some embodiments, the compounds of the present description can be used to treat cancer in patients that had prior progression on prior a treatment (i.e., cancers that are resistant to one or more anticancer therapies) (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof).) (e.g., combination of inhibitors). For example, a combination of BRAF inhibitors, MEK inhibitor, and/or EGFR.
In some embodiments, the compounds of the present description can be combined with an inhibitor of the RTK, PI3K, AKT, RAS, RAF or MAPK pathway in cancers or tumors that have an upregulation of a mitogen activated protein kinase (MAPK) gene expression signature, such as CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and/or SPRY4.
In some embodiments, the compounds of the present description can be combined with an inhibitor of one or more inhibitors of the pathways described in Table V. In some embodiments, the compounds of the present description can be combined with an inhibitor of one or more inhibitors described herein in the cancers or tumors have an upregulation in one or more genes described in Tables W-Z.
In some embodiments, the compounds of the present description can be combined with an inhibitor of a receptor tyrosine kinase (RTK). Non-limiting examples of RTK inhibitors include epidermal growth factor receptor (EGFR) inhibitors erlotinib, osimertinib, neratinib, gefitinib, cetuximab, panitumumab, dacomitinib, lapatinib, necitumumab, mobercitinib, vandetanib, sapitinib, afatinib, canertinib, CP-724714, CUDC-101 , mubritinib (TAK 165), OSI-420, pelitinib, irbinitinib (ARRY-380, ONT-380), varlitinib, TAK-285, BIBX 1382, PD168393, raciletinib, poziotinib, CL-387785, icotinib, CTX-2006, WHI-P154, AZ5104, AZD3759, Erb-B2 receptor tyrosine kinase 2 (HER2) such as neratinib, trastuzumab and its derivatives such as trastuzumab deruxtecan, lapatinib, dacomitinib, tucatinib, pertuzumab, margetuximab, afatinib, AZD8931 , AST1306, AEE788, canertinib, CP724.714, CUDC101 , TAK285, dacomitinib, pelitinib, AC480, canertinib, sapitinib, mubritinib (TAK 165), BDTX-189, epertinib, TAS0728, poziotinib, multikinase inhibitors cabozantinib, imatinib, Lenvatinib, regorafenib, sorafenib, sunitinib, vandetinib, MET inhibitors crizotinib, cabozantinib, foretinib, PHA-665752, SU11274, SGX-523, BMS- 777607, tivantinib (ARQ 197), JNJ-38877605, PF-04217903, MGCD-265, capmatinib (INCB28060), BMS-754807, BMS-794833, AMG-208, MK-2461 , golvatinib (E7050), AMG-458, NVP-BVU972, XL092, UNC2025, elzovantinib (TPX-0022), AMG-1 , JNJ- 38877618 (OMO-1), altiratinib, SAR125844, glumetinib (SCC244), savolitinib (AZD6094), RXDX-106 (CEP-40783), S49076, merestinib (LY2801653), AMG 337, tepotinib, neurotrophic receptor tyrosine kinase 1 (NTRK1) inhibitors larotrectinib, entrectinib, danusertib, BMS-754807, GW441756, UNC2025, taletrectinib, altiratinib, selitrectinib (LOXO-195), CH7057288, BMS-935177, PF-06273340, sitravatinib (MGCD516), GNF- 5837, vascular endothelial growth factor receptor inhibitors bevacizumab, ramucirumab, SU5408, linifanib (ABT-869), axitinib (AG 013736), nintedanib (BIBF 1120), cediranib (AZD2171), motesanib (AMG-706), pazopanib (GW786034), vandetanib, sunitinib (SU11248), sorafenib (BAY 43-9006), brivanib (BMS-540215), vatalanib (PTK787), foretinib (GSK1363089), cabozantinib (BMS-907351), brivanib (BMS-582664), Lenvatinib (E7080), CYC116, regorafenib (BAY 73-4506), ENMD-2076, tivozanib, ponatinib (AP24534), apatinib (YN968D1), telatinib, PP121 , pazopanib, dovitinib, SAR131675, semaxanib (SU5416), golvatinib (E7050), fibroblast growth factor receptor inhibitors ponatinib (AP24534), infigratinib (BGJ398), nintedanib (BIBF 1120), PD173074, AZD4547, dovitinib, pemigatinib, futibatinib, infigratinib, and RLY-4008.
In some embodiments, the compounds of the present description can be used to treat inflammatory and fibrotic diseases that can include fatty liver diseases, endometriosis, types 1 or 2 diabetes mellitus, inflammatory bowel disease, asthma, rheumatoid arthritis, obesity, systemic sclerosis, sclerodermatous graft vs. host disease, nephrogenic systemic fibrosis, as well as organ-specific fibrosis, including radiation-induced fibrosis, and auto- immune diseases.
Serine/threonine-protein kinase (SGK-1) (also known as serum/glucocorticoid-regulated kinase 1) is a protein kinase that plays a role in a cell's response to stress. In vivo, SGK- 1 activates certain potassium, sodium, and chloride channels. For instance, the protein is known to regulate the myo-inositol transporter during osmotic stress. The term “inhibitor of SGK-1”, as used herein, refers to any compound that can block, arrest, interfere with, or reduce the biological activity of SGK-1.
In some embodiments, the compounds of the present description can be used for increasing fetal hemoglobin (HbF) in erythrocytes. In some embodiments, the compounds of the present description can be used for the treatment of a p-hemoglobinopathy. In some embodiments, the compounds of the present description can be used for the treatment of sickle cell disease.
In some embodiments, the compounds of the present description can be used for the treatment of prostate cancer. In other embodiments, the compounds of the present description can be used for the treatment of epilepsy. in some embodiments, there is described a method for the treatment of cancer or tumor in a subject, comprising: i. determining that cells from said cancer or tumor (e.g., cancer or tumor sample or biopsy): have an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; have an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; have a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; have an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIO; have an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by one or more concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN; have an upregulation of a mitogen activated protein kinase (MAPK) gene expression (e.g., CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and SPRY4); and/or are resistant to one or more anticancer therapies (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof); and ii. administering to the subject one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical compositions defined herein, or the combinations defined herein. in some embodiments, there is described a use of one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical compositions defined herein, or the combinations defined herein, for the treatment of cancer or tumor or for the manufacture of a medicament for the treatment of cancer or tumor, wherein the cancer or tumor (e.g., cancer or tumor sample or biopsy) has been predetermined as: having an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; having an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; having a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIO; having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by one or more concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN; having an upregulation of a mitogen activated protein kinase (MAPK) gene expression (e.g., CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and SPRY4); and/or being resistant to one or more anticancer therapies (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof).
In some embodiments, there is described a use of one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical compositions defined herein, or the combinations defined herein, for the treatment of one or more side effects or toxicities caused by an anticancer therapy in a subject undergoing said anticancer therapy. The one or more side effects or toxicities caused by the anticancer therapy may be selected from the group consisting of: hyperglycemia and hyperinsulinemia, increased ALT/AST, pyrexia, rash, diarrhea, nausea, fatigue, thrombocytopenia, anemia, dermatitis acneiform, QT elongation, and dyspnea.
In some embodiments, there is described a method for treating one or more side effects or toxicities caused by an anticancer therapy, comprising administering to a subject undergoing said anticancer therapy one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical compositions defined herein, or the combinations defined herein. The one or more side effects or toxicities caused by the anticancer therapy may be selected from the group consisting of: hyperglycemia and hyperinsulinemia, increased ALT/AST, pyrexia, rash, diarrhea, nausea, fatigue, thrombocytopenia, anemia, dermatitis acneiform, QT elongation, and dyspnea. In some embodiments, there is described a method for monitoring the progression of an anticancer therapy in a subject that was previously administered with one or more compounds or pharmaceutically acceptable salts thereof as defined herein, the pharmaceutical compositions defined herein, or the combinations defined herein, wherein the method comprises determining in a sample from said subject: a loss of phosphorylation of NDRG1 ; an increase in phosphorylation of MAPK14 (i.e. , p38); an increase in phosphorylation of MAPK14 (i.e., p38); an increase in phosphorylation of heat shock protein family B1 (i.e., HSPB1 or HSP27); an increase in phosphorylation of tumor suppressor p53 (TP53); an increase in phosphorylation of ribosomal protein S6 kinase A1 (RSK1); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERKI); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERK1); and/or an increase in phosphorylation of AKT1 substrate 1 (PRAS40).
In some embodiments, said loss of phosphorylation of NDRG1 is at threonine 346 of the NDRG1 ; said increase in phosphorylation of MAPK14 (i.e., p38) is at threonine 180 of the MAPK14; said increase in phosphorylation of MAPK14 (i.e., p38) is at tyrosine 182 of the MAPK14; said increase in phosphorylation of heat shock protein family B1 (i.e., HSPB1 or HSP27) at serine 82 of the HSPB1 ; said increase in phosphorylation of tumor suppressor p53 (TP53) is at serine 15 of the TP53; said increase in phosphorylation of ribosomal protein S6 kinase A1 (RSK1) is at serine 380 of the RSK1 ; said increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2) is at tyrosine 185 of the MAPK1 ; said increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2) is at tyrosine 187 of the MAPK1 ; said increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERK1) is at threonine 202 of the MAPK3; said increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERK1) is at tyrosine 204 of the MAPK3; and/or said increase in phosphorylation of AKT1 substrate 1 (PRAS40) is at threonine 246 of the AKT1 substrate 1.
In some embodiments, said loss or increase in phosphorylation is in comparison to a sample from a healthy subject or a subject that is not undergoing said anticancer therapy. In some embodiments, said monitoring the progression of said anticancer therapy includes determining if the anticancer therapy is effective or if the cancer is becoming resistant to the anticancer therapy.
Inhibitors of SGK-1
The compounds of the present description and their pharmaceutically acceptable salts thereof are pharmacologically active compounds that modulate protein kinase activity, specifically the activity of serum and glucocorticoid regulated kinase isoform 1 (SGK-1). The compounds of the present description or their pharmaceutically acceptable salts can be suitable for the treatment of conditions in which SGK-1 activity is inappropriate. Non- limiting examples of such conditions can include Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, stent failure, prostate cancer and epilepsy. Other non- limiting examples of such conditions include β-hemoglobinopathies, such as sickle cell disease.
In one aspect, compounds of Formula I, or pharmaceutically acceptable salts thereof are provided.
Figure imgf000039_0001
Formula I
In some embodiments, Z is selected from the group consisting of a direct bond, -O-, -S-, - CH(R9)- and -N(Rw)-, wherein R9 and R10 are independently of one another selected from the group consisting of H and (C1-C4)-alkyl. In some embodiments, Z is selected from the group consisting of a direct bond, -O-, -S-, -CH2- and -NH-. In some embodiments, Z is a direct bond. In some embodiments, Z is selected from the group consisting of -O- and - NH-.
In some embodiments, R3 is selected from the group consisting of H, (C1-C8)-alkyl, R30 and ( C1-C6)-alkyl- R30, wherein (C1-C8)-alkyl is unsubstituted or substituted by one or more identical or different substituents R31. R30 is a 3-membered to 12-membered, monocyclic or bicyclic, saturated, partially unsaturated or aromatic, cyclic group which comprises 0, 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, which is unsubstituted or substituted by one or more identical or different substituents R32. R31 is selected from the group consisting of halogen, -OH, -CF3, -O-(C1-C4)-alkyl, -N(R33)-R34 and -CN. R32 is selected from the group consisting of halogen, (C1-C4)-alkyl, (C3-C7)-cycloalkyl, -(C1-C4)-alkyl-(C3-C7)-cycloalkyl, -(C1-C4)-alkyl-O-R37, - (C1-C4)-alkyl-N(R38)-R39, -(C1-C4)-alkyl-CN, -C(O)-(C1-C4)-alkyl, -CN, -OH, =0, -O-(C1-C4)- alkyl, -N(R4O)-R4I, -C(O)-O-(C1-C4)-alkyl and -C(O)-N(R42)-R43.
In some embodiments, R33 and R34 are independently of one another selected from the group consisting of H, (C1-C4)-alkyl and (C3-C7)-cycloalkyl, wherein (C1-C4)-alkyl and (C3- C7)-cycloalkyl are unsubstituted or substituted by one or more identical or different substituents R50, wherein R50 is selected from the group consisting of halogen, -OH, -O- (C1-C4)-alkyl, -CF3 and -CN. In some embodiments, R37, R38, R39, R40, R41, R42 and R43 are independently of one another selected from the group consisting of H and (C1-C4)-alkyl.
In some embodiments, R3 is selected from the group consisting of H, -CH2OH,
-CH3,
Figure imgf000040_0001
Figure imgf000041_0001
In some embodiments, Z is a direct bond and R3 is selected from the group consisting of
H, -CH2OH and -CH3. In other embodiments, Z is selected from the group consisting of - O- and -NH- and R3 is selected from the group consisting of:
Figure imgf000041_0002
In some embodiments, R1 is selected from the group consisting of H, -N(R11)R12, -N(R13)- C(O)-R14, -NR13-S(O)2-R15, -NR13-C(O)-NH-R16, -(C1-C4)-alkyl, — (C1-C4)-alkyl-OR17 and —
(C1-C4)-alkyl-N(Ris) R19, wherein R11, R12, R13, R14, R15, R16, R17, R18 and R19 are independently of one another selected from the group consisting of H and (C1-C4)-alkyl.
In some embodiments, R1 is selected from the group consisting of -(C1-C4)-alkyl, and - (C1-C4)-alkyl-N( R18)R19. In some embodiments, R1 is selected from the group consisting of -CH3, -CH2N(CH3)2 and -CH2-CH2-N(CH3)2.
In some embodiments, Y is selected from the group consisting of carbocyclylene and heterocyclylene, which is unsubstituted or substituted by one or more identical or different substituents R5, wherein R5 is selected from the group consisting of halogen, (C1-C4)-alkyl, -O-( C1-C4)-alkyl and -CN. In some embodiments, Y is selected from the group consisting of arylene and heteroarylene, which is unsubstituted or substituted by one or more identical or different substituents R5. In some embodiments, Y is selected from the group consisting of:
Figure imgf000042_0001
It is understood that when two symmetrical Y groups are listed, such
Figure imgf000042_0002
and
Figure imgf000042_0003
In some embodiments, A is selected from the group of a direct bond or -CH2-. When A is a direct bond, -Y- is directly linked to the nitrogen of the sulfonamide group.
In some embodiments R2 is selected from the group consisting of (C1-C4)-alkyl, (C3-C7)- cycloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl, phenyl and a 5-membered or 6-membered monocyclic, saturated, partially unsaturated or aromatic, heterocyclic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and is bonded via a ring carbon atom or a ring nitrogen atom, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20, wherein R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21, -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN, and wherein R21, R22, R23, R24, R25 and R26 are independently of one another selected from the group consisting of H and (C1-C4)-alkyl.
In some embodiments, when Y is not 1 ,4-phenylene, or when Y is 1 ,4-phenylene and R1 is -(C1-C4)-alkyl-N(Ris)R19: R2 is selected from the group consisting of (C1-C4)-alkyl, (C3- C7)-cycloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl, phenyl and a 5-membered or 6-membered monocyclic, saturated, partially unsaturated or aromatic, heterocyclic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and is bonded via a ring carbon atom or a ring nitrogen atom, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20, wherein R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21, -N( R22) R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN, and wherein R21, R22, R23, R24, R25 and R26 are independently of one another selected from the group consisting of H and (C1-C4)-alkyl.
In other embodiments, when Y is 1 ,4-phenylene and R1 is H, -N(R11)R12, -N(R13)-C(O)-R14, -NR13-S(O)2-Ri5, -NR13-C(O)-NH-Ri6, -(C1-C4)-alkyl or
-(C1-C4)-alkyl-OR17, R2 is selected from the group consisting of (C1-C4)-alkyl, (C3-C7)- cycloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl and a 5-membered or 6-membered monocyclic, saturated or partially unsaturated, heterocyclic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and is bonded via a ring carbon atom or a ring nitrogen atom, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20, wherein R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21, -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN, and wherein R21, R22, R23, R24, R25 and R26 are independently of one another selected from the group consisting of H and (C1-C4)-alkyl.
In some embodiments, R2 is selected from the group consisting of:
Figure imgf000044_0001
In some embodiments, Y is 1 ,4-phenylene and R2 is selected from the group consisting of: -CH3,
Figure imgf000044_0002
In some embodiments, the compound of Formula I is selected from the group consisting of:
Figure imgf000044_0003
Figure imgf000045_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound of Formula I is selected from the group consisting
5 of:
Figure imgf000045_0002
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000047_0002
or a pharmaceutically acceptable salt thereof.
In another aspect, compounds of Formula II, or pharmaceutically acceptable salts thereof, are provided:
Figure imgf000047_0003
Formula II wherein:
Z is selected from the group consisting of O, CH2, S and NH;
R1 is selected from the group consisting of H, and -(C1-C4)-alkyl; R3 is selected from the group consisting of -(CH2)P-N(R33)R34; p is 1 , 2, 3 or 4; R2 is selected from the group consisting of a 5-membered or 6-membered monocyclic, aromatic or heteroaromatic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20; R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21, -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN; R21, R22, R23, R24, R25 and R26 , are independently of one another selected from the group consisting of H and (C1-C4)-alkyl;
R33 and R34 are independently of one another selected from the group consisting of -CH=O, (C1-C4)-alkyl and (C3-C7)-cycloalkyl, wherein (C1-C4)-alkyl and (C3-C7)- cycloalkyl are each unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)-alkyl, CF3, and -CN.
In some embodiments, Z is NH. In other embodiments, Z is O. In some embodiments, Ri is methyl. In some embodiments, p is 2, 3 or 4.
In some embodiments, R2 is selected from the group consisting of:
Figure imgf000048_0001
In some embodiments, R33 is methyl. In some embodiments, R34 is methyl.
In some embodiments, R3 is selected from the group consisting of:
Figure imgf000048_0002
In some embodiments, the compound of Formula II is selected from the group consisting of:
Figure imgf000049_0001
Figure imgf000050_0001
or a pharmaceutically acceptable salt thereof.
5 In yet another aspect, compounds of Formula II, or pharmaceutically acceptable salts thereof are provided:
Figure imgf000050_0002
Formula II wherein:
Z is selected from the group consisting of a direct bond, O, S, CH(Rg) and N(R10); R1 is selected from the group consisting of H, -N(R11)R12, -N(R13)-C(O)-R14, -NR13-S(O)2"R15, -NR13-C(O)-NH-R16, -(C1-C4)-alkyl, — (C1-C4)-alkyl-OR17 and — (C1-C4)-alkyl-N(R18)R19;
R3 is selected from the group consisting of H, (C1-C8)-alkyl, R30 and (C1-C4)-alkyl- R30, wherein (C1-C8)-alkyl is unsubstituted or substituted by one or more identical or different substituents R31; R30 is a 3-membered to 12-membered, monocyclic or bicyclic, saturated, partially unsaturated or aromatic, cyclic group which comprises 0, 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, which is unsubstituted or substituted by one or more identical or different substituents R32; R31 is selected from the group consisting of halogen, -OH, -CF3, -O-(C1-C4)-alkyl, -N(R33)-R34 and -CN; R32 is selected from the group consisting of halogen, (C1-C4)-alkyl, (C3-C7)- cycloalkyl, -(C1-C4)-alkyl-(C3-C7)-cycloalkyl, -(C1-C4)-alkyl-O-R37, -(C1-C4)-alkyl-N(R38)- R39, -(C1-C4)-alkyl-CN, -C(O)-(C1-C4)-alkyl, -CN, -OH, =0, -O-(C1-C4)-alkyl, -N(R4O)-R41, - C(O)-O-(C1-C4)-alkyl and -C(O)-N(R42)-R43; R2 is a 6-membered monocyclic, heteroaromatic group which comprises 1 or 2 nitrogen atoms, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20; R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21 , -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN;
R9, R10, R1 1, R12, R13, R14, R15, R16, R17, R18, R19, R21 , R22, R23, R24, R25, R26, R37,
R38, R39, R40, R41 , R42 and R43 are independently of one another selected from the group consisting of H and (C1-C4)-alkyl;
R33 and R34 are independently of one another selected from the group consisting of H, (C1-C4)-alkyl and (C3-C7)-cycloalkyl wherein (C1-C4)-alkyl and (C3-C7)-cycloalkyl are unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)-alkyl, -CF3 and -CN. In some embodiments, R1 is selected from the group consisting of -(C1-C4)-alkyl, and - (C1-C4)-alkyl-N(R18)R19. For example, in some embodiments, R1 is selected from the group consisting of -CH3, -CH2N(CH3)2 and -CH2-CH2-N(CH3)2.
In some embodiments, Z is selected from the group consisting of -O- and -NH-. In some embodiments, R3 is selected from the group consisting of:
Figure imgf000052_0001
In some embodiments, Z is a direct bond. In some embodiments, R3 is selected from the group consisting of H, -CH2OH and -CH3.
In some embodiments, R2 is selected from the group consisting of:
Figure imgf000052_0002
In some embodiments, the compound of Formula II is selected from the group consisting of:
Figure imgf000053_0001
5 or a pharmaceutically acceptable salt thereof. In yet another aspect, a compound of Formula III is provided:
Figure imgf000054_0001
or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O, CH2, S and NH;
R1 is selected from the group consisting of H, and -(C1-C4)-alkyl;
R3 is selected from the group consisting of -(CH2)P-N(R33)R34, wherein zero, one or two hydrogen atoms of the group -(CH2)P- are independently replaced with F; p is 1 , 2, 3 or 4; R2 is selected from the group consisting of a 5-membered or 6-membered monocyclic, aromatic or heteroaromatic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20; R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21, -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN; R21, R22, R23, R24, R25 and R26 , are independently of one another selected from the group consisting of H and (C1-C4)-alkyl;
R33 and R34 are independently of one another selected from the group consisting of -CH=O, (C1-C4)-alkyl and (C3-C7)-cycloalkyl, wherein (C1-C4)-alkyl and (C3-C7)- cycloalkyl are each unsubstituted or substituted by one or more identical or different substituents R50; R50 is selected from the group consisting of halogen, -OR27, -O-(C1-C4)-alkyl, CF3, and -CN;
R27 is selected from the group consisting of H, -C(=O)-(C1-C4)-alkyl, a natural amino acid bound by the a-carboxyl group, or P(=O)(OH)2; and
W1, W2, W3, W4 are independently of one another selected from the group consisting of H, halogen, -OR21, -CF3, (C1-C4)-alkyl, and -CN. In some embodiments, Z is NH. In other embodiments, Z is O. In some embodiments, R1 is methyl. In some embodiments, p is 2, 3 or 4.
Figure imgf000055_0001
wherein Z1 and Z2 are independently from one another selected from the group consisting of Cl, F, -OMe and - CN, and Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN.
In some embodiments, R2 is selected from the group consisting of:
Figure imgf000055_0002
Figure imgf000055_0003
In some embodiments, R2 is selected from the group consisting of:
Figure imgf000055_0004
Figure imgf000055_0005
In some embodiments, R2 is
Figure imgf000055_0006
Figure imgf000055_0007
In some embodiments, R33 = methyl. In some embodiments, R34 = methyl.
In some embodiments, R3 is selected from the group consisting of:
Figure imgf000056_0001
wherein R51 is (C1-C4)-alkyl.
In some embodiments:
W1, W2, W3 and W4 are each H;
W1 is F or Cl, W2, W3 and W4 are each H; or
W1 and W2 are each F, W3 and W4 are each H.
In some embodiments, W1 is F or Cl, W2 is H, W3 is H and W4 is H. In some embodiments, W1 is F, W2 is H, W3 is H and W4 is H. In some embodiments, W1 is Cl, W2 is H, W3 is H and W4 is H.
In yet another aspect, a compound of Formula III is provided:
Figure imgf000056_0002
Formula III or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O and NH;
R1 is selected from the group consisting of H and (C1-C4)-alkyl;
R3 is selected from the group consisting of -(CH2)P-N(R33)R34; p is 2, 3 or 4;
Figure imgf000056_0003
Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
R33 and R34 are independently of one another a (C1-C4)-alkyl;
W1, W2, W3, W4 are independently of one another selected from the group consisting of H, halogen, -OR21, -CF3, (C1-C4)-alkyl, and -CN; and R21, is selected from the group consisting of H and (C1-C4)-alkyl.
In some embodiments, R1 is methyl.
In some embodiments, R3 is selected from the group consisting of:
Figure imgf000057_0002
Figure imgf000057_0001
In some embodiments, R3 is
Figure imgf000057_0003
Figure imgf000057_0004
Figure imgf000057_0005
Figure imgf000057_0006
In some embodiments, W1, W2, W3 and W4 are each H;
W1 is F, W2, W3 and W4 are each H; or
W1 and W2 are each F, W3 and W4 are each H.
In some embodiments, W1 is F, W2, W3 and W4 are each H.
In some embodiments, Z is NH.
In some embodiments, there is provided a compound selected from the group consisting
Figure imgf000058_0001
In yet another aspect, a compound of Formula III is provided:
Figure imgf000058_0002
or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O, and NH; R1 is selected from the group consisting of H, and -(C1-C4)-alkyl; R3 is a nitrogen-bearing heterocycle selected from the group consisting of
Figure imgf000059_0001
Figure imgf000059_0002
, wherein zero, one or two hydrogens on the -CH2- groups of the nitrogen-bearing heterocycle is replaced with halogen, -OH, -CN, -CF3 or (C1-C4)-alkyl;
R35 is H or (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)- alkyl, CF3, and -CN;
Figure imgf000059_0003
Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
W1 is F;
W2 is H or F; W3 is H; and
W4 is H.
In some embodiments, Z is O.
In some embodiments, R2 is selected from the group consisting of:
In some embodiments, R1 is methyl.
N-R35
In some embodiments, R3 is
In some embodiments, R35 is methyl or isopropyl.
In some embodiments, W2 is H.
In yet another aspect, a compound of Formula III is provided: or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O, and NH; R1 is selected from the group consisting of H, and -(C1-C4)-alkyl;
R3 is a nitrogen-bearing heterocycle selected from the group consisting of
Figure imgf000061_0001
, wherein zero, one or two hydrogens on any of the -CH2- groups of the nitrogen-bearing heterocycle is replaced with halogen, -OH, -CN, -CF3 or (C1-C4)-alkyl;
R35 is H or (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)- alkyl, CF3, and -CN;
Figure imgf000061_0002
Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
W1 is F, Cl or OMe;
W2 is H or F; W3 is H; and
W4 is H.
In some embodiments, Z is O.
In some embodiments, R1 is methyl.
In some embodiments, R3 is
Figure imgf000061_0003
In some embodiments, R35 is methyl or isopropyl.
In some embodiments, W2 is H.
Figure imgf000062_0001
In some embodiments, there is provided a compound selected from the group consisting of:
Figure imgf000062_0002
a pharmaceutically acceptable sale thereof.
In yet another aspect, a compound of Formula III is provided:
Figure imgf000062_0003
or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O, and NH; R1 is selected from the group consisting of H, and -(C1-C4)-alkyl; R3 is a nitrogen-bearing heterocycle selected from the group consisting of
Figure imgf000063_0001
Figure imgf000063_0002
, wherein zero, one or two hydrogens on any of the -CH2- groups of the nitrogen-bearing heterocycle is replaced with halogen, -OH, -CN, -CF3 or (C1-C4)-alkyl;
R35 is H or (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)- alkyl, CF3, and -CN; R2 is
Figure imgf000063_0003
Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
W1, W2, W3, W4 are independently of one another selected from the group consisting of H, halogen, -OR21, -CF3, (C1-C4)-alkyl, and -CN, R21 is selected from the group consisting of H and (C1-C4)-alkyl.
In some embodiments, Z is O.
In some embodiments, R1 is methyl.
In some embodiments, R3 is
Figure imgf000063_0004
In some embodiments, R35 is methyl or isopropyl.
In some embodiments,
W1 is H, F, Cl or OMe; W2 is H or F; W3 is H; and
W4 is H. In some embodiments, W1 is F, W2 is H, W3 is H and W4 is H. In some embodiments, W1 is Cl, W2 is H, W3 is H and W4 is H.
Figure imgf000064_0001
In some embodiments, there is provided a compound selected from the group consisting of:
Figure imgf000064_0002
or a pharmaceutically acceptable sale thereof.
In yet another aspect, a compound of Formula IV is provided:
Figure imgf000065_0001
Formula IV or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O and NH;
R3 is selected from the group consisting of: -(CH2)P-N(R33)R34,
Figure imgf000065_0003
Figure imgf000065_0002
p is 2, 3 or 4;
R33 and R34 are independently from one another a (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50;
R35 is H or a (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50; R50 is selected from the group consisting of halogen, -OR27, -O-(C1-C4)-alkyl, -CF3, and -CN;
R27 is selected from the group consisting of H, -C(=O)-(C1-C4)alkyl, a natural amino acid bound by the a-carboxyl group, and P(=O)(OH)2; R2 is
Figure imgf000065_0004
Z1 and Z2 are independently of one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN;
Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN; and
W1 is halogen.
In some embodiments, Z is O. In other embodiments, Z is NH. In some embodiments, R27 is selected from the group consisting of H or -C(=O)-(C1-C4)alkyl.
In some embodiments, R3 is -(CH2)P-N(R33)R34. In some embodiments, p = 2. In some embodiments, R33 is methyl.
In some embodiments, R34 is methyl. In other embodiments, R34 is is -(CH2)2-OH or -(CH2)2-O-C(=O)-(C1-C4)alkyl.
In some embodiments, R3 is . In some embodiments, R35 is methyl or
Figure imgf000066_0001
isopropyl.
In some embodiments, R3 is
Figure imgf000066_0002
. In some embodiments, R35 is H.
In some embodiments, Z-R3 is selected from the group consisting of
Figure imgf000066_0003
Figure imgf000066_0004
In some embodiments, Z-R3 is selected from the group consisting of
Figure imgf000066_0005
and
Figure imgf000066_0006
In some embodiments, Z-R3 is selected from the group consisting of
Figure imgf000066_0008
and
Figure imgf000066_0007
In some embodiments, Z-R3 is
Figure imgf000067_0001
In some embodiments, W1 is F. In other embodiments, W1 is Cl.
In some embodiments, R2 is
Figure imgf000067_0002
Z1 and Z2 can be independently from one another selected from the group consisting of Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe.
In some embodiments, R2 is In some embodiments, R2 is
Figure imgf000067_0003
Figure imgf000067_0004
In some embodiments, R2 is
Figure imgf000067_0005
. Z3 can be selected from the group consisting of H, Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe, or from the group consisting of H, -CH3, -CF3, -OCH(CH3)2 and -OMe, or from the group consisting of -CH3, -OCH(CH3)2 and - OMe. In some embodiments, the compound of Formula IV is a compound of Formula I a:
Figure imgf000067_0006
or a pharmaceutically acceptable salt thereof, wherein: R2 is selected from the group consisting of:
Figure imgf000068_0001
Figure imgf000068_0002
W1 is selected from the group consisting of Cl and F; and
R27 is selected from the group consisting of: H,
Figure imgf000068_0003
Figure imgf000068_0004
In some embodiments, the compound of Formula IV is a compound of Formula IVb:
Figure imgf000068_0005
or a pharmaceutically acceptable salt thereof, wherein:
R2 is selected from the group consisting o
Figure imgf000068_0006
Figure imgf000068_0007
W1 is selected from the group consisting of Cl and F. In some embodiments, the compound of Formula IV is a compound of Formula IVc:
or a pharmaceutically acceptable salt thereof, wherein: R2 is selected from the group consisting of:
W1 is selected from the group consisting of Cl and F; and
R35 is selected from the group consisting of methyl and isopropyl.
In some embodiments, the compound of Formula IV is a compound of Formula IVd: or a pharmaceutically acceptable salt thereof, wherein: R2 is selected from the group consisting of: W1 is selected from the group consisting of Cl and F.
In some embodiments, the compound of Formula IV is selected from the group consisting of Compounds 28, 38, 78, 79, 84, 85, 99, 100, 101, 102, 103,104, 105, 107, 106, 108, 109, 110, 111, 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, 155, 156, 163, 164, 165, 166, 167, 168, 169,
170, 171, 172, 173 and 174, as numbered in Table A hereinbelow, or a pharmaceutically acceptable salt thereof.
In another aspect, a compound of Formula IV is provided:
Formula IV or a pharmaceutically acceptable salt thereof, wherein:
Z-R3 is selected from the group consisting of:
Figure imgf000070_0001
and R2 is selected from the group consisting of
Figure imgf000071_0001
Figure imgf000071_0002
W1 is selected from the group consisting of Cl and F. In some embodiments, W1 is Cl. In other embodiments, W1 is F. In some embodiments, R2 is
Figure imgf000071_0003
. in some embodiments, R27 is H. In some embodiments, Z-R3 is
Figure imgf000071_0004
In another aspect, a compound of Formula V is provided:
Figure imgf000071_0005
Formula V or a pharmaceutically acceptable salt thereof, wherein:
Figure imgf000071_0006
Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN; W1 is selected from the group consisting of H and halogen;
R33 is -CH3 or -(CH2)-(CH2)-OR27; and
R27 is selected from the group consisting of H, -C(=O)-(C1-C4)alkyl, a natural amino acid bound by the a-carboxyl group, and -P(=O)(OH)2.
In some embodiments, W1 is F. In some embodiments, W1 is Cl..
In some embodiments, R2 is
Figure imgf000072_0001
2 . Z1 and Z2 can be independently from one another selected from the group consisting of Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe.
In some embodiments, R2 is
Figure imgf000072_0002
Figure imgf000072_0003
In some embodiments, R2 is
Figure imgf000072_0004
In some embodiments, R2 is
Figure imgf000072_0005
.Z3 can be selected from the group consisting of H, Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe, or from the group consisting of H, -CH3, - CF3, -OCH(CH3)2 and -OMe, or from the group consisting of -CH3, -OCH(CH3)2 and - OMe.
In some embodiments, R33 is -CH3. In other embodiments, R33 is-(CH2)-(CH2)-OR27. In some embodiments, R27 is selected from the group consisting of H and
-C(=O)-(C1-C4)alkyl. In some embodiments, the compound of Formula V is a compound of Formula Va: or a pharmaceutically acceptable salt thereof, wherein: R2 is selected from the group consisting of:
W1 is selected from the group consisting of H, Cl and F; and
R27 is selected from the group consisting of: H,
In some embodiments, the compound of Formula V is a compound of Formula Vb: or a pharmaceutically acceptable salt thereof, wherein: R2 is selected from the group consisting of:
W1 is selected from the group consisting of H, Cl and F.
In some embodiments, the compound of Formula V is selected from the group consisting of Compounds 9, 20, 22, 24, 27, 30, 38, 45, 72, 73, 78, 84, 85, 99, 100, 103, 104, 105, 106, 107, 108, 109, 110, 111 , 112, 113, 114, 115, 116, 117, 118, 119, 120, 121 , 122, 123, 124, 125, 150, 151 , 152 and 153, as numbered in Table A hereinbelow, or a pharmaceutically acceptable salt thereof.
In one aspect, a compound of Formula Vb is provided:
Formula Vb or a pharmaceutically acceptable salt thereof, wherein: Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN; and
W1 is selected from the group consisting of H and halogen. In some embodiments, W1 is F. In other embodiments, W1 is Cl.
In some embodiments, R2 is z2 . Z1 and Z2 can be independently from one another selected from the group consisting of Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe.
In some embodiments, R2 is
In some embodiments, R2 is . Z3 can be selected from the group consisting of H, Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe, or from the group consisting of H, -CH3, - CF3, -OCH(CH3)2 and -OMe, or from the group consisting of -CH3, -OCH(CH3)2 and - OMe.
In some embodiments, the compound of Formula Vb is selected from the group consisting of Compounds 9, 38, 45, 84, 85 and 111 , as numbered in Table A hereinbelow, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound of Formula Vb is selected from the group consisting of Compounds 24, 27, 30, 73, 100, 103, 104, 105, 106, 107, 108, 109, 110, 113 and 112, as numbered in Table A hereinbelow, or a pharmaceutically acceptable salt thereof.
In another aspect, there is provided a compound of Formula VI:
Formula VI or a pharmaceutically acceptable salt thereof, wherein:
Y1 is H or F; q is 0 or 1; Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
W1 is selected from the group consisting of H and halogen;
R35 is H or a (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50; R50 is selected from the group consisting of halogen, -OR27, -O-(C1-C4)-alkyl, -CF3, and -CN;
R27 is selected from the group consisting of H, -C(=O)-(C1-C4)alkyl, a natural amino acid bound by the a-carboxyl group, and P(=O)(OH)2.
In some embodiments, Y1 is H.
In some embodiments, the compound of Formula VI is selected from the group consisting of:
Formula Via Formula Vlb
Figure imgf000077_0001
Formula Vic Formula Vid or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of Formula VI is a compound of Formula Via:
Figure imgf000077_0002
or a pharmaceutically acceptable salt thereof.
In some embodiments, q = 1. In some embodiments, W1 is F. In other embodiments, W1 is Cl.
In some embodiments, R2 is
Figure imgf000077_0003
. Z1 and Z2 can be independently from one another selected from the group consisting of Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe.
In some embodiments, R2 is
Figure imgf000077_0004
In some embodiments, R2 is . Z3 can be selected from the group consisting of H, Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe, or from the group consisting of H, -CH3, - CF3, -OCH(CH3)2 and -OMe, or from the group consisting of -CH3, -OCH(CH3)2 and - OMe.
In some embodiments, R27 is selected from the group consisting of H and -C(=O)-(Ci- C4)alkyl.
In some embodiments, R35 is H or a (C1-C4)-alkyl a (C1-C4)-alkyl which is unsubstituted.
In some embodiments, R35 is H.
In some embodiments, the compound of Formula VI is a compound of Formula Via: or a pharmaceutically acceptable salt thereof, wherein: R2 is selected from the group consisting of:
W1 is selected from the group consisting of H, Cl and F.
In some embodiments, the compound of Formula VI is selected from the group consisting of Compounds 29, 41 , 42, 44, 46, 47, 49, 50, 53, 54, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 66, 67, 69, 70, 71, 92, 155 and 156, as numbered in Table A hereinbelow, or a pharmaceutically acceptable salt thereof.
For all the embodiments pertaining to Formula VI described herein, the compound of Formula VI is preferably not a compound of Formula Via in racemic form: or a pharmaceutically acceptable salt thereof, wherein:
Y2 is H or F; and R52 is selected from the group consisting of:
The present description also discloses the following embodiments:
1 . A compound of Formula Vb:
Formula Vb or a pharmaceutically acceptable salt thereof, wherein: Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN; and
W1 is selected from the group consisting of H and halogen.
2. The compound of embodiment 1 , wherein W1 is F.
3. The compound of embodiment 1 , wherein W1 is Cl.
4. The compound of any one of embodiments 1 to 3, wherein R2 is
5. The compound of embodiment 1 to 4, wherein Z1 and Z2 are independently from one another selected from the group consisting of Cl, F, -CH3, -CN, -OCH(CH3)2 and -
OMe. 6. The compound of any one of embodiments 1 to 5, wherein R2 is
Figure imgf000081_0002
or
The compound of any one of embodiments 1 to 6, wherein R2 is
8. The compound of any one of embodiments 1 to 3, wherein R2 is
9. The compound of embodiment 8, wherein Z3 is selected from the group consisting of H, Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe.
10. The compound of embodiment 8 or 9, wherein Z3 is selected from the group consisting of H, -CH3, -CF3, -OCH(CH3)2 and -OMe.
11. The compound of any one of embodiments 8 to 10, wherein Z3 is selected from the group consisting of -CH3, -OCH(CH3)2 and -OMe.
12. The compound of embodiment 1, which is:
Figure imgf000081_0001
5 13. The compound of embodiment 1, which is:
or a pharmaceutically acceptable salt thereof. 14. A compound of Formula V:
Formula V or a pharmaceutically acceptable salt thereof, wherein: Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
W1 is selected from the group consisting of H and halogen;
R33 is -CH3 or -(CH2)-(CH2)-OR27; and
R27 is selected from the group consisting of H, -C(=O)-(C1-C4)alkyl, a natural amino acid bound by the a-carboxyl group, and -P(=O)(OH)2.
15. The compound of embodiment 14, wherein W1 is F.
16. The compound of embodiment 14, wherein W1 is Cl.
The compound of any one of embodiments 14 to 16, wherein R2 is
18. The compound of any one of embodiments 14 to 17, wherein Z1 and Z2 are independently from one another selected from the group consisting of Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe.
19. The compound of any one of embodiments 14 to 18, wherein R2 is
20. The compound of any one of embodiments 14 to 19, wherein R2 is F
21. The compound of any one of embodiments 14 to 16, wherein R2 is
22. The compound of embodiment 21 , wherein Z3 is selected from the group consisting of H, Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe. 23. The compound of embodiment 21 or 22, wherein Z3 is selected from the group consisting of H, -CH3, -CF3, -OCH(CH3)2 and -OMe.
24. The compound of any one of embodiments 21 to 23, wherein Z3 is selected from the group consisting of -CH3, -OCH(CH3)2 and -OMe.
25. The compound of any one of embodiments 14 to 24, wherein R33 is -CH3.
26. The compound of any one of embodiments 14 to 24, wherein R33 is-(CH2)- (CH2)-OR27.
27. The compound of embodiment 26, wherein R27 is selected from the group consisting of H and -C(=O)-(C1-C4)alkyl.
28. The compound of embodiment 14, which is: or a pharmaceutically acceptable salt thereof, wherein:
W1 is selected from the group consisting of H, Cl and F; and o
R27 is selected from the group consisting of: H, The compound of embodiment 14, which is: or a pharmaceutically acceptable salt thereof, wherein:
W1 is selected from the group consisting of H, Cl and F.
30. The compound of embodiment 14, which is:
31. A compound of Formula VI:
Formula VI or a pharmaceutically acceptable salt thereof, wherein:
Y1 is H or F; q is 0 or 1; Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
W1 is selected from the group consisting of H and halogen;
R35 is H or a (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50; R50 is selected from the group consisting of halogen, -OR27, -O-(C1-C4)-alkyl, -CF3, and -CN;
R27 is selected from the group consisting of H, -C(=O)-(C1-C4)alkyl, a natural amino acid bound by the a-carboxyl group, and P(=O)(OH)2.
32. The compound of embodiment 31, wherein Y1 is H.
33. The compound of embodiment 31 or 32, which is selected from the group consisting of: or a pharmaceutically acceptable salt thereof.
34. The compound of any one of embodiments 31 to 33, which is:
or a pharmaceutically acceptable salt thereof.
35. The compound of any one of embodiments 31 to 34, wherein q is 1.
36. The compound of any one of embodiments 31 to 35, wherein W1 is F.
37. The compound of any one of embodiments 31 to 35, wherein W1 is Cl.
38. The compound of any one of embodiments 31 to 37, wherein R2 is
Figure imgf000095_0001
39. The compound of any one of embodiments 31 to 38, wherein Z1 and Z2 are independently from one another selected from the group consisting of Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe.
40. The compound of any one of embodiments 31 to 39, wherein R2 is
41. The compound of any one of embodiments 31 to 40, wherein R2 is
42. The compound of any one of embodiments 31 to 37, wherein R2 is 43. The compound of embodiment 42, wherein Z3 is selected from the group consisting of H, Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe.
44. The compound of embodiment 42 or 43, wherein Z3 is selected from the group consisting of H, -CH3, -CF3, -OCH(CH3)2 and -OMe.
45. The compound of any one of embodiments 42 to 44, wherein Z3 is selected from the group consisting of -CH3, -OCH(CH3)2 and -OMe.
46. The compound of any one of embodiments 31 to 45, wherein R27 is selected from the group consisting of H and -C(=O)-(C1-C4)alkyl.
47. The compound of any one of embodiments 31 to 45, wherein R35 is H, a (C1-C4)- alkyl or a (C1-C4)-alkyl which is unsubstituted.
48. The compound of any one of embodiments 31 to 45, wherein R35 is H.
49. The compound of embodiment 31 , which is: or a pharmaceutically acceptable salt thereof, wherein:
W1 is selected from the group consisting of H, Cl and F.
50. The compound of embodiment 31 , which is:
Formula IV or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O and NH; R3 is selected from the group consisting of: -(CH2)P-N(R33)R34,
Figure imgf000101_0002
Figure imgf000101_0001
p is 2, 3 or 4;
R33 and R34 are independently from one another a (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50;
R35 is H or a (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50; R50 is selected from the group consisting of halogen, -OR27, -O-(C1-C4)-alkyl, -CF3, and -CN;
R27 is selected from the group consisting of H, -C(=O)-(C1-C4)alkyl, a natural amino acid bound by the a-carboxyl group, and P(=O)(OH)2; Z1 and Z2 are independently of one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN; and
W1 is halogen.
52. The compound of embodiment 51 , wherein Z is O.
53. The compound of embodiment 51 , wherein Z is NH.
54. The compound of any one of embodiments 51 to 53, wherein R27 is selected from the group consisting of H or -C(=O)-(C1-C4)alkyl.
55. The compound of any one of embodiments 51 to 54, wherein R3 is -(CH2)P-
N(R33)R34-
56. The compound of embodiment 55, wherein p = 2. 57. The compound of embodiment 55 or 56, wherein R33 is methyl.
58. The compound of any one of embodiments 55 to 57, wherein R34 is methyl.
59. The compound of any one of embodiments 55 to 57, wherein R34 is -(CH2)2-OH or
-(CH2)2-O-C(=O)-(C1-C4)alkyl.
60. The compound of any one of embodiments 51 to 54, wherein R3 is
61. The compound of embodiment 60, wherein R35 is methyl or isopropyl.
62. The compound of any one of embodiments 51 to 54, wherein R3 is
R35
63. The compound of embodiment 62, wherein R35 is H.
64. The compound of embodiment 51, wherein Z-R3 is selected from the group
N consisting of
65. The compound of embodiment 64, wherein Z-R3 is selected from the group
N N consisting of and
66. The compound of embodiment 64 or 65, wherein R27 is selected from the group consisting of H and -C(=O)-(C1-C4)alkyl. 67. The compound of embodiment 66, wherein R27 is H.
68. The compound of embodiment 64, wherein Z-R3 is selected from the group
69. The compound of embodiment 64, wherein Z-R3 is
70. The compound of any one of embodiments 51 to 69, wherein W1 is F.
71. The compound of any one of embodiments 51 to 69, wherein W1 is Cl
The compound of any one of embodiments 51 to 71, wherein R2 is
73. The compound of embodiment 72, wherein Z1 and Z2 are independently from one another selected from the group consisting of Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe.
74. The compound of embodiment 72 or 73, wherein R2 is F
II
The compound of any one of embodiments 72 to 74, wherein R2 is
76. The compound of any one of embodiments 51 to 71, wherein R2 is
77. The compound of embodiment 76, wherein Z3 is selected from the group consisting of H, Cl, F, -CH3, -CN, -OCH(CH3)2 and -OMe. 78. The compound of embodiment 76 or 77, wherein Z3 is selected from the group consisting of H, -CH3, -CF3, -OCH(CH3)2 and -OMe. 79. The compound of any one of embodiments 76 to 77, wherein Z3 is selected from the group consisting of -CH3, -OCH(CH3)2 and -OMe.
80. The compound of embodiment 51 , which is:
Figure imgf000104_0001
or a pharmaceutically acceptable salt thereof, wherein: R2 is selected from the group consisting of
Figure imgf000104_0002
Figure imgf000104_0003
W1 is selected from the group consisting of Cl and F; and
Figure imgf000104_0004
The compound of embodiment 51 , which is: or a pharmaceutically acceptable salt thereof, wherein:
W1 is selected from the group consisting of Cl and F.
The compound of embodiment 51, which is: or a pharmaceutically acceptable salt thereof, wherein: W1 is selected from the group consisting of Cl and F; and
R35 is selected from the group consisting of methyl and isopropyl.
The compound of embodiment 51 , which is:
R. or a pharmaceutically acceptable salt thereof, wherein:
W1 is selected from the group consisting of Cl and F.
The compound of embodiment 51 , which is:
Figure imgf000106_0001
Figure imgf000107_0001
 
Figure imgf000113_0001
or a pharmaceutically acceptable salt thereof. 85. A compound of Formula IV:
Figure imgf000114_0001
Formula IV or a pharmaceutically acceptable salt thereof, wherein: Z-R3 is selected from the group consisting of:
Figure imgf000114_0002
, and R27 is selected from the group consisting of: H,
W1 is selected from the group consisting of Cl and F. . The compound of embodiment 85, wherein W1 is Cl. . The compound of embodiment 85, wherein W1 is F. . The compound of any one of embodiments 85 to 87, wherein R2 is. The compound of any one of embodiments 85 to 88, wherein R27 is H.. The compound of any one of embodiments 85 to 89, wherein Z-R3 is
H . A compound of Formula II:
Formula II or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O, CH2, S and NH; R1 is selected from the group consisting of H, and -(C1-C4)-alkyl;
R3 is selected from the group consisting of -(CH2)P-N(R33)R34; p is 1 , 2, 3 or 4; R2 is selected from the group consisting of a 5-membered or 6-membered monocyclic, aromatic or heteroaromatic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20; R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21 , -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN; R21 , R22, R23, R24, R25 and R26 , are independently of one another selected from the group consisting of H and (C1-C4)-alkyl;
R33 and R34 are independently of one another selected from the group consisting of -CH=O, (C1-C4)-alkyl and (C3-C7)-cycloalkyl, wherein (C1-C4)-alkyl and (C3-C7)- cycloalkyl are each unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)-alkyl, CF3, and -CN.
92. The compound of embodiment 91 , or a pharmaceutically acceptable salt thereof, wherein Z is NH.
93. The compound of embodiment 91 , or a pharmaceutically acceptable salt thereof, wherein Z is O.
94. The compound of any one of embodiments 91 to 93, or a pharmaceutically acceptable salt thereof, wherein R1 is methyl.
95. The compound of any one of embodiments 91 to 94, or a pharmaceutically acceptable salt thereof, wherein p is 2, 3 or 4. 96. The compound of any one of embodiments 91 to 95, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group consisting of:
97. The compound of any one of embodiments 91 to 95, or a pharmaceutically
98. The compound of any one of embodiments 91 to 97, or a pharmaceutically acceptable salt thereof, wherein R33 = methyl.
99. The compound of any one of embodiments 91 to 98, or a pharmaceutically acceptable salt thereof, wherein R34 = methyl.
100. The compound of any one of embodiments 91 to 97, wherein R3 is selected from the group consisting of: or a pharmaceutically acceptable salt thereof.
102. A compound of Formula II:
Formula II or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of a direct bond, O, S, CH(R9) and N(R10); R1 is selected from the group consisting of H, -N(R11)R12, -N(R13)-C(O)-R14, -NR13-S(O)2"R15, -NR13-C(O)-NH-R16, -(C1-C4)-alkyl, — (C1-C4)-alkyl-OR17 and — (C1-C4)-alkyl-N(R18)R19;
R3 is selected from the group consisting of H, (C1-C8)-alkyl, R30 and (C1-C4)-alkyl- R30, wherein (C1-C8)-alkyl is unsubstituted or substituted by one or more identical or different substituents R31; R30 is a 3-membered to 12-membered, monocyclic or bicyclic, saturated, partially unsaturated or aromatic, cyclic group which comprises 0, 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, which is unsubstituted or substituted by one or more identical or different substituents R32; R31 is selected from the group consisting of halogen, -OH, -CF3, -O-(C1-C4)-alkyl, -N(R33)-R34 and -CN; R32 is selected from the group consisting of halogen, (C1-C4)-alkyl, (C3-C7)- cycloalkyl, -(C1-C4)-alkyl-(C3-C7)-cycloalkyl, -(C1-C4)-alkyl-O-R37, -(C1-C4)-alkyl-N(R38)-
R39, -(C1-C4)-alkyl-CN, -C(O)-(C1-C4)-alkyl, -CN, -OH, =0, -O-(C1-C4)-alkyl, -N(R4O)-R4I, - C(O)-O-(C1-C4)-alkyl and -C(O)-N(R42)-R43; R2 is a 6-membered monocyclic, heteroaromatic group which comprises 1 or 2 nitrogen atoms, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20; R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21 , -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN;
R9, R10, R11, R12, R13, R14, R15, R16, R17, R18, R19, R21 , R22, R23, R24, R25, R26, R37,
R38, R39, R40, R41, R42 and R43 are independently of one another selected from the group consisting of H and (C1-C4)-alkyl;
R33 and R34 are independently of one another selected from the group consisting of H, -CH=O, (C1-C4)-alkyl and (C3-C7)-cycloalkyl wherein (C1-C4)-alkyl and (C3-C7)- cycloalkyl are unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)-alkyl, -CF3 and -CN. 103. The compound of embodiment 102, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from the group consisting of -(C1-C4)-alkyl, and -(C1-C4)- alkyl-N(R18)R19.
104. The compound of embodiment 103, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from the group consisting of -CH3, -CH2N(CH3)2 and -CH2-CH2-N(CH3)2.
105. The compound of embodiment 104, or a pharmaceutically acceptable salt thereof, wherein R1 is -CH3.
106. The compound of any one of embodiments 102 to 105, or a pharmaceutically acceptable salt thereof, wherein Z is selected from the group consisting of -O- and -NH-.
107. The compound of any one of embodiments 102 to 106, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from the group consisting of:
108. The compound of any one of embodiments 102 to 105, or a pharmaceutically acceptable salt thereof, wherein Z a direct bond.
109. The compound of embodiment 108, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from the group consisting of H, -CH2OH and -CH3. 110. The compound of any one of embodiments 102 to 109, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group consisting of:
111. A compound selected from the group consisting of:
or a pharmaceutically acceptable salt thereof. 112. A compound of Formula I:
Formula I or a pharmaceutically acceptable salt thereof, wherein: Z is selected from the group consisting of a direct bond, O, S, CH(R9) and N(R10); R1 is selected from the group consisting of H, -N(R11)R12, -N(R13)-C(O)-R14, -NR13-S(O)2"R15, -NR13-C(O)-NH-R16, -(C1-C4)-alkyl, — (C1-C4)-alkyl-OR17 and — (C1-C4)-alkyl-N(R18)R19;
R3 is selected from the group consisting of H, (C1-C8)-alkyl, R30 and (C1-C4)-alkyl- R30, wherein (C1-C8)-alkyl is unsubstituted or substituted by one or more identical or different substituents R31; R30 is a 3-membered to 12-membered, monocyclic or bicyclic, saturated, partially unsaturated or aromatic, cyclic group which comprises 0, 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, which is unsubstituted or substituted by one or more identical or different substituents R32; R31 is selected from the group consisting of halogen, -OH, -CF3, -O-(C1-C4)-alkyl, -N(R33)-R34 and -CN; R32 is selected from the group consisting of halogen, (C1-C4)-alkyl, (C3-C7)- cycloalkyl, -(C1-C4)-alkyl-(C3-C7)-cycloalkyl, -(C1-C4)-alkyl-O-R37, -(C1-C4)-alkyl-N(R3s)-
R39, -(C1-C4)-alkyl-CN, -C(O)-(C1-C4)-alkyl, -CN, -OH, =0, -O-(C1-C4)-alkyl, -N(R4O)-R4I, - C(O)-O-(C1-C4)-alkyl and -C(O)-N(R42)-R43;
A is a direct bond or -CH2-;
Y is selected from the group consisting of carbocyclylene and heterocyclylene, which is unsubstituted or substituted by one or more identical or different substituents R5; R5 is selected from the group consisting of halogen, (C1-C4)-alkyl, -O-( C1-C4)- alkyl and -CN; when Y is not 1 ,4-phenylene, or when Y is 1 ,4-phenylene and R1 is -(C1-C4)- alkyl-N(R18)R19: R2 is selected from the group consisting of from the group consisting of (C1-C4)-alkyl, (C3-C7)-cycloalkyl, (C2-C4)-alkenyl, (C2-C4)-alkynyl, phenyl and a 5- membered or 6-membered monocyclic, saturated, partially unsaturated or aromatic, heterocyclic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and is bonded via a ring carbon atom or a ring nitrogen atom, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20; when Y is 1 ,4-phenylene and R1 is H, -N(R11)R12, -N(R13)-C(O)-R14, -NR13-S(O)2-R15, -NR13-C(O)-NH-R16, -(C1-C4)-alkyl or -(C1-C4)-alkyl-ORi7: R2 is selected from the group consisting of (C1-C4)-alkyl, (C3-C7)-cycloalkyl, (C2-C4)-alkenyl, (C2-C4)- alkynyl and a 5-membered or 6-membered monocyclic, saturated or partially unsaturated, heterocyclic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and is bonded via a ring carbon atom or a ring nitrogen atom, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20; R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21 , -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN;
R9, R10, R11, R12, R13, R14, R15, R16, R17, R18, R19, R21 , R22, R23, R24, R25, R26, R37,
R38, R39, R40, R41 , R42 and R43 are independently of one another selected from the group consisting of H and (C1-C4)-alkyl;
R33 and R34 are independently of one another selected from the group consisting of H, -CH=O, (C1-C4)-alkyl and (C3-C7)-cycloalkyl, wherein (C1-C4)-alkyl and (C3-C7)- cycloalkyl are unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)-alkyl, -CF3 and -CN.
113. The compound of embodiment 112, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from the group consisting of -(C1-C4)-alkyl, and
— (C1-C4)-alkyl-N(R18)R19.
114. The compound of embodiment 113, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from the group consisting of -CH3, -CH2N(CH3)2 and - CH2-CH2-N(CH3)2.
115. The compound of embodiment 114, or a pharmaceutically acceptable salt thereof, wherein R1 is -CH3.
116. The compound of any one of embodiments 112 to 115, or a pharmaceutically acceptable salt thereof, wherein Z is selected from the group consisting of -O- and -NH-.
117. The compound of any one of embodiments 112 to 116, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from the group consisting of:
118. The compound of any one of embodiments 112 to 115, or a pharmaceutically acceptable salt thereof, wherein Z a direct bond.
119. The compound of embodiment 118, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from the group consisting of H, -CH2OH and -CH3.
120. The compound of any one of embodiments 112 to 119, or a pharmaceutically acceptable salt thereof, wherein A is a direct bond.
121. The compound of any one of embodiments 112 to 120, or a pharmaceutically acceptable salt thereof, wherein:
Y is selected from the group consisting of arylene and heteroarylene, which is unsubstituted or substituted by one or more identical or different substituents R5.
122. The compound of any one of embodiments 112 to 120, or a pharmaceutically acceptable salt thereof, wherein Y is selected from the group consisting of: 123. The compound of embodiment 122, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group consisting of:
124. The compound of any one of embodiments 112 to 120, or a pharmaceutically acceptable salt thereof, wherein Y is
125. The compound of embodiment 124, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group consisting of:
-CH3, 126. A compound selected from the group consisting of:
Figure imgf000128_0001
or a pharmaceutically acceptable salt thereof. A compound selected from the group consisting of:
or a pharmaceutically acceptable salt thereof.
128. A compound of Formula II:
Formula II or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O, CH2, S and NH; R1 is selected from the group consisting of H, and -(C1-C4)-alkyl;
R3 is selected from the group consisting of -(CH2)P-N(R33)R34; p is 1 , 2, 3 or 4; R2 is a 6-membered monocyclic, heteroaromatic group which comprises 1 or 2 nitrogen atoms, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20; R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21, -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN; R21, R22, R23, R24, R25 and R26 , are independently of one another selected from the group consisting of H and (C1-C4)-alkyl;
R33 and R34 are independently of one another selected from the group consisting of -CH=O, (C1-C4)-alkyl and (C3-C7)-cycloalkyl, wherein (C1-C4)-alkyl and (C3-C7)- cycloalkyl are each unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)-alkyl, CF3, and -CN.
129. The compound of embodiment 128, or a pharmaceutically acceptable salt thereof, wherein Z is NH.
130. The compound of embodiment 128, or a pharmaceutically acceptable salt thereof, wherein Z is O.
131. The compound of any one of embodiments 128 to 130, or a pharmaceutically acceptable salt thereof, wherein R1 is methyl.
132. The compound of any one of embodiments 128 to 131 , or a pharmaceutically acceptable salt thereof, wherein p is 2, 3 or 4.
133. The compound of any one of embodiments 128 to 132, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group consisting of:
Figure imgf000131_0001
134. The compound of any one of embodiments 128 to 133, or a pharmaceutically acceptable salt thereof, wherein R33 = methyl.
135. The compound of any one of embodiments 128 to 134, or a pharmaceutically acceptable salt thereof, wherein R34 = methyl.
136. The compound of any one of embodiments 128 to 135, wherein R3 is selected from the group consisting of:
137. A compound of Formula III: or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O, CH2, S and NH; R1 is selected from the group consisting of H, and -(C1-C4)-alkyl;
R3 is selected from the group consisting of -(CH2)P-N(R33)R34, wherein zero, one or two hydrogen atoms of the group -(CH2)P- are independently replaced with F; p is 1 , 2, 3 or 4; R2 is selected from the group consisting of a 5-membered or 6-membered monocyclic, aromatic or heteroaromatic group which comprises 1 , 2 or 3 identical or different ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, wherein R2 is unsubstituted or substituted by one or more identical or different substituents R20; R20 is selected from the group consisting of halogen, -CF3, (C1-C4)-alkyl, -OR21, -N(R22)R23, (C1-C4)-alkyl-OR24, (C1-C4)-alkyl-N(R25)R26 and -CN; R21, R22, R23, R24, R25 and R26 , are independently of one another selected from the group consisting of H and (C1-C4)-alkyl;
R33 and R34 are independently of one another selected from the group consisting of -CH=O, (C1-C4)-alkyl and (C3-C7)-cycloalkyl, wherein (C1-C4)-alkyl and (C3-C7)-cycloalkyl are each unsubstituted or substituted by one or more identical or different substituents R50; R50 is selected from the group consisting of halogen, -OR27, -O-(C1-C4)-alkyl, CF3, and -CN;
R27 is selected from the group consisting of H, -C(=O)-(C1-C4)-alkyl, a natural amino acid bound by the a-carboxyl group, or P(=O)(OH)2; and
W1, W2, W3, W4 are independently of one another selected from the group consisting of H, halogen, -OR21, -SH, -CF3, (C1-C4)-alkyl, and -CN.
138. The compound of embodiment 137, or a pharmaceutically acceptable salt thereof, wherein Z is NH.
139. The compound of embodiment 137, or a pharmaceutically acceptable salt thereof, wherein Z is O.
140. The compound of any one of embodiments 137 to 139, or a pharmaceutically acceptable salt thereof, wherein R1 is methyl.
141 . The compound of any one of embodiments 137 to 140, or a pharmaceutically acceptable salt thereof, wherein p is 2, 3 or 4.
142. The compound of any one of embodiments 137 to 141 , or a pharmaceutically acceptable salt thereof, wherein: Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; and Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN.
143. The compound of any one of embodiments 137 to 141 , or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group consisting of:
144. The compound of any one of embodiments 137 to 143, or a pharmaceutically acceptable salt thereof, wherein R33 = methyl.
145. The compound of any one of embodiments 137 to 144, or a pharmaceutically acceptable salt thereof, wherein R34 = methyl.
146. The compound of any one of embodiments 137 to 143, wherein R3 is selected from the group consisting of: wherein R51 is (C1-C4)-alkyl.
147. The compound of any one of embodiments 137 to 146, wherein: W1, W2, W3 and W4 are each H;
W1 is F or Cl, W2, W3 and W4 are each H; or
W1 and W2 are each F, W3 and W4 are each H.
148. The compound of any one of embodiments 137 to 146, wherein W1 is F or Cl, W2 is H, W3 is H and W4 is H.
149. A compound of Formula III: or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O and NH; R1 is selected from the group consisting of H and (C1-C4)-alkyl;
R3 is -(CH2)P-N(R33)R34; p is 2, 3 or 4; Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN;
Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
R33 and R34 are independently of one another a (C1-C4)-alkyl; and
Wi, W2, W3, W4 are independently of one another selected from the group consisting of H, halogen, -OR2I, -CF3, (C1-C4)-alkyl, and -CN,
R2I, is selected from the group consisting of H and (C1-C4)-alkyl.
150. The compound of embodiment 149, wherein R1 is methyl. 151. The compound of embodiment 149 or 150, wherein R3 is selected from the group consisting of:
Figure imgf000136_0001
152. The compound of embodiment 151, wherein R3 is
Figure imgf000136_0002
153. The compound of any one of embodiments 149 to 152, wherein R2 is selected from the group consisting of
Figure imgf000136_0003
:
Figure imgf000136_0004
154. The compound of any one of embodiments 149 to 153, wherein:
W1, W2, W3 and W4 are each H;
W1 is F or Cl, W2, W3 and W4 are each H; or
W1 and W2 are each F, W3 and W4 are each H.
155. The compound of any one of embodiments 149 to 154, wherein W1 is F or Cl, W2, W3 and W4 are each H.
156. The compound of any one of embodiments 149 to 155, wherein Z is NH. 157. A compound selected from the group consisting of: pharmaceutically acceptable sale thereof.
158. A compound of Formula III: or a pharmaceutically acceptable salt thereof, wherein:
Z is selected from the group consisting of O, and NH; R1 is selected from the group consisting of H, and -(C1-C4)-alkyl;
R3 is a nitrogen-bearing heterocycle selected from the group consisting of or two hydrogens on any of the -CH2- groups of the nitrogen-bearing heterocycle is replaced with halogen, -OH, -CN, -CF3 or (C1-C4)-alkyl;
R35 is H or (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)- alkyl, CF3, and -CN; Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
W1 is F, OMe or Cl;
W2 is H or F; W3 is H; and
W4 is H.
159. The compound of embodiment 158, wherein Z is O.
160. The compound of embodiment 158 or 159, wherein R1 is methyl.
161. The compound of any one of embodiments 158 to 160, wherein R3 is
35
162. The compound of any one of embodiments 158 to 161, wherein R35 is methyl or isopropyl.
163. The compound of any one of embodiments 158 to 162, wherein W2 is H.
164. The compound of any one of embodiments 158 to 163, wherein R2 is selected
165. A compound selected from the group consisting of: 166. A compound of Formula III: or a pharmaceutically acceptable salt thereof, wherein: Z is selected from the group consisting of O, and NH; R1 is selected from the group consisting of H, and -(C1-C4)-alkyl;
R3 is a nitrogen-bearing heterocycle selected from the group consisting of and
, wherein zero, one or two hydrogens on any of the -CH2- groups of the nitrogen-bearing heterocycle is replaced with halogen, -OH, -CN, -CF3 or (C1-C4)-alkyl;
R35 is H or (C1-C4)-alkyl which is unsubstituted or substituted by one or more identical or different substituents R50; and R50 is selected from the group consisting of halogen, -OH, -O-(C1-C4)- alkyl, CF3, and -CN; Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN; Z3 is selected from the group consisting of H, halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3 and -CN;
W1, W2, W3, W4 are independently of one another selected from the group consisting of H, halogen, -OR2I, -CF3, (C1-C4)-alkyl, and -CN,
R2I is selected from the group consisting of H and (C1-C4)-alkyl.
167. The compound of embodiment 166, wherein Z is O.
168. The compound of embodiment 166 or 167, wherein R1 is methyl.
169. The compound of any one of embodiments 166 to 168, wherein R3 is
170. The compound of any one of embodiments 166 to 169, wherein R35 is methyl or isopropyl.
171. The compound of any one of embodiments 166 to 170, wherein
W1 is H, F, OMe or CI;
W2 is H or F; W3 is H; and
W4 is H.
172. The compound of any one of embodiments 166 to 170, wherein W1 is F, W2 is H, W3 is H and W4 is H. 173. The compound of any one of embodiments 166 to 172, wherein R2 is selected
174. A compound selected from the group consisting of: or a pharmaceutically acceptable sale thereof.
175. A pharmaceutical composition, comprising the compound as defined in any one of embodiments 1 to 174 or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient.
176. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, as an inhibitor of SGK-1 .
177. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the treatment of cancer.
178. The use of embodiment 177, wherein the cancer affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
179. The use of embodiment 177, wherein the cancer is a melanoma, liposarcoma, lung cancer, breast cancer, prostate cancer, leukemia, kidney cancer, esophageal cancer, brain cancer, lymphoma, colon cancer or colorectal cancer.
180. The use of embodiment 177, wherein the cancer is prostate cancer, colorectal cancer or breast cancer.
181. The use of any one of embodiments 177 to 180, wherein the compound or pharmaceutically acceptable salt thereof is used in combination with at least one inhibitor of AKT/PI3K/mTOR.
182. The use of embodiment 181 , wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI- 587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK-1117, MLN-1117, TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC-907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG- 100- 115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
183. The use of embodiment 181 , wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
184. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the treatment of Parkinson’s disease or Lafora disease. 185. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the treatment of epilepsy.
186. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the treatment of a cardiovascular disease selected from the group consisting of Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, and stent failure.
187. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the treatment of Long QT syndrome.
188. The use of embodiment 187, wherein the Long QT syndrome is genetic Long QT syndrome.
189. The use of embodiment 187, wherein The Long QT syndrome is acquired Long QT syndrome.
190. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament that inhibits SGK-1 in a subject.
191 . Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of cancer.
192. The use of embodiment 191 , wherein the cancer affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
193. The use of embodiment 191 , wherein the cancer is a melanoma, liposarcoma, lung cancer, breast cancer, prostate cancer, leukemia, kidney cancer, esophageal cancer, brain cancer, lymphoma, colon cancer or colorectal cancer. 194. The use of embodiment 191 , wherein the cancer is prostate cancer, colorectal cancer or breast cancer.
195. The use of any one of embodiments 191 to 194, wherein the compound or pharmaceutically acceptable salt thereof is used in combination with at least one inhibitor of AKT/PI3K/mTOR.
196. The use of embodiment 195, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI- 587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK-1117, MLN-1117, TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC-907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-100- 115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
197. The use of embodiment 195, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
198. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of Parkinson’s disease or Lafora disease.
199. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of epilepsy. 200. Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a cardiovascular disease selected from the group consisting of Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, and stent failure.
201 . Use of the compound of any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of Long QT syndrome.
202. The use of embodiment 201 , wherein the Long QT syndrome is genetic Long QT syndrome.
203. The use of embodiment 201 , wherein The Long QT syndrome is acquired Long QT syndrome.
204. A method for the treatment of cancer, the method comprising administering to a subject a therapeutically effective amount of the compound as defined in any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof.
205. The method of embodiment 204, wherein the cancer affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
206. The method of embodiment 204, wherein the cancer is a melanoma, liposarcoma, lung cancer, breast cancer, prostate cancer, leukemia, kidney cancer, esophageal cancer, brain cancer, lymphoma, colon cancer or colorectal cancer.
207. The method of embodiment 204, wherein the cancer is prostate cancer, colorectal cancer or breast cancer. 208. The method of any one of embodiments 204 to 207, wherein the compound or pharmaceutically acceptable salt thereof is used in combination with at least one inhibitor of AKT/PI3K/mTOR.
209. The method of embodiment 208, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF- 05212384 (Gedatolisib, PKI-587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101, Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK-1117.MLN- 1117.TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC-907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-100-115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
210. The method of embodiment 208, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
211. A method for the treatment of epilepsy, the method comprising administering to a subject a therapeutically effective amount of a compound as defined in any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof.
212. A method for the treatment of a cardiovascular disease, the method comprising administering to a subject a therapeutically effective amount of a compound as defined in any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof, the cardiovascular disease being selected from the group consisting of Long QT syndrome, heart failure, arrhythmia, ischemic injury, ischemic infarction, cardiac fibrosis, vascular proliferation, restenosis, dilated cardiomyopathy, and stent failure. 213. A method for the treatment of Long QT syndrome, the method comprising administering to a subject a therapeutically effective amount of a compound as defined in any one of embodiments 1 to 174, or a pharmaceutically acceptable salt thereof.
214. The method of embodiment 193, wherein the Long QT syndrome is genetic Long QT syndrome.
215. The method of embodiment 193, wherein The Long QT syndrome is acquired Long QT syndrome.
216. A compound of Formula VII:
Formula VII or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein:
Y1 is H or F; Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN;
W1 is selected from the group consisting of H and halogen; and
R35 is H or methyl.
217. The compound of embodiment 216 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein Y1 is H.
218. The compound of embodiment 216 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein Y1 is F.
219. The compound of any one of embodiments 216 to 218 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein Z1 is F and Z2 is F. 220. The compound of any one of embodiments 216 to 218 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein Z1 is Cl and Z2 is F.
221 . The compound of any one of embodiments 216 to 220 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein W1 is H.
222. The compound of any one of embodiments 216 to 220 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein W1 is Cl.
223. The compound of any one of embodiments 216 to 220 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein W1 is F.
224. The compound of any one of embodiments 216 to 223 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein R35 is H.
225. The compound of any one of embodiments 216 to 223 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein R35 is methyl.
226. The compound of embodiment 216 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, which is a compound of Formula Vila: wherein:
Figure imgf000148_0001
Z1 is F or Cl;
Z2 is F; and
W1 is H, Cl or F.
227. The compound of embodiment 226, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein Z1 is F and Z2 is F. 228. The compound of embodiment 226, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein Z1 is Cl and Z2 is F.
229. The compound of any one of embodiments 226 to 228, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein W1 is H.
230. The compound of any one of embodiments 226 to 228, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein W1 is Cl.
231. The compound of any one of embodiments 226 to 228, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein W1 is F.
232. The compound of embodiment 216, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, which is: or a pharmaceutically acceptable salt thereof.
233. The compound of embodiment 216 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, which is a compound of Formula VI lb:
Formula VI lb wherein: Z1 is F or Cl;
Z2 is F; R35 is H or methyl; and
W1 is H, Cl or F.
234. The compound of embodiment 233 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein R35 is H.
235. The compound of embodiment 233 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein R35 is methyl.
236. The compound of any one of embodiments 233 to 235, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein Z1 is F and Z2 is F.
237. The compound of any one of embodiments 233 to 235, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein Z1 is Cl and Z2 is F.
238. The compound of any one of embodiments 233 to 237, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein W1 is H.
239. The compound of any one of embodiments 233 to 237, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein W1 is Cl.
240. The compound of any one of embodiments 233 to 237, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein W1 is F.
241. The compound of embodiment 216, for use in the treatment of cancer, which is: or a pharmaceutically acceptable salt thereof.
Figure imgf000150_0001
242. The compound of embodiment 216, for use in the treatment of cancer, which is:
or a pharmaceutically acceptable salt thereof.
243. The compound of any one of embodiments 216 to 242, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, in combination with at least one inhibitor of AKT/PI3K/mT0R.
244. The compound of embodiment 243, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein the at least one inhibitor of AKT/PI3K/mT0R is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI- 587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK-1117, MLN-1117, TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC-907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-100- 115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
245. The compound of embodiment 243, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
246. The compound of any one of embodiments 216 to 245, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein the cancer affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
247. The compound of any one of embodiments 216 to 245, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein the cancer is a melanoma, liposarcoma, lung cancer, breast cancer, prostate cancer, leukemia, kidney cancer, esophageal cancer, brain cancer, lymphoma, colon cancer or colorectal cancer.
248. The compound of any one of embodiments 216 to 245, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein the cancer is prostate cancer, colorectal cancer or breast cancer.
249. A pharmaceutical composition, for use in the treatment of cancer, comprising the compound of any one of embodiments 216 to 242, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
250. The pharmaceutical composition of embodiment 249, further comprising at least one inhibitor of AKT/PI3K/mTOR.
251 . The pharmaceutical composition of embodiment 250, for the treatment of cancer, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI-587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL- 101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK- 1117, MLN-1117, TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC- 907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-100-115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
252. The pharmaceutical composition of embodiment 250, for use in the treatment of cancer, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
253. A method for the treatmenf of cancer, comprising administering to a subject in need thereof the compound of any one of embodiments 216 to 242, or a pharmaceutically acceptable salt thereof.
EXAMPLES
Some abbreviations and acronyms are used in the description of the experimental procedures and Examples below. Although most of these abbreviations and acronyms would be understood by a person skilled in the art.
EXAMPLE 1 : Preparation of Compounds
ACS grade solvents and reagents were used without further purification.
The prepared compounds were in general characterized by spectroscopic data and chromatographic data, in particular mass spectra (MS) and/or nuclear magnetic resonance (NMR) spectra. 'H-NMR spectra were generally recorded at 600 MHz. In the NMR characterization, the chemical shift δ (in ppm), the number of hydrogen atoms (H), the coupling constant J (in Hz) and the multiplicity (s: singlet, d: doublet, dd: double doublet, t: triplet, dt: double triplet, m: multiplet; br: broad) of the peaks are given. In the MS characterization, the mass number (m/z) of the peak of the molecular ion (M) or of a related ion such as the ion [M+1], i.e. the protonated molecular ion [M+H)] or the ion [M- 1], which was formed depending on the ionization method used, is given. Generally, the ionization method was electrospray ionization (ES+ or ES-).
Compound 1 N-(4-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)ethenesulfonamide
Figure imgf000154_0001
(i) 4,6-dichloro-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1 H-pyrazolo[3,4-d]pyrimidine
Commercially available 4,6-Dichloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidine (1.00 g, 5.29 mmol, 1.00 equiv.) was dissolved in THF (13.3 ml, 0.4 M) in a reaction vessel containing a magnetic stirring bar, followed by addition of 3,4-dihydro-2H-pyran (2.42 ml, 26.5 mmol, 5.00 equiv.) and pyridinium 4-toluenesulfonate (66.3 mg, 0.264 mmol, 0.05 equiv.) at RT.The colorless reaction mixture was heated to 60° C for 3h (the solution became slightly yellow) and allowed to cool down before evaporation of the volatiles. The residue was dissolved in ethyl acetate (20 ml) and washed with a saturated aqueous sodium hydrogenocarbonate solution (3x20ml), dried over sodium sulfate, filtered and evaporated to afford the desired product (1.28 g, 94% yield) as a slightly yellow solid.
(ii) 6-chloro-4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidine
1 -lsopropylpiperidin-4-ol (498 mg, 3.30 mmol, 4.74 equiv.) was dissolved in dry THF (5.00 ml) in a reaction vessel containing a magnetic stirring bar under an argon atmosphere, and the mixture cooled on an ice bath. Then sodium hydride (26.5 mg, 60% suspension in mineral oil) was added and the mixture stirred on an ice bath for approximately 30 min Addition of 4,6-dichloro-3-methyl-1-(tetrahydro-pyran-2-yl) 1 H-pyrazolo3,4-dpyrimidine (200 mg, 0.70 mmol, 1.00 equiv.) dissolved in THF (2.00 ml). The ice bath was removed and the mixture stirred at RT until complete conversion of the starting material as monitored by TLC (AcOEt/hexanes). the reaction mixture was quenched with water (10ml) and extracted with ethyl acetate (3x 20ml) and the combined organic phases dried over sodium sulfate, filtered and evaporated. The crude product was purified by flash chromatography on silica gel using a mixture of ethyl acetate and Hexanes as the eluent to afford the desired product after evaporation (241 mg, 88% yield) as a colorless oil. LCMS (ESI, m/z): 394.5 [M+H]+.
(iii) N-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)ethenesulfonamide
Ethenesulfonyl chloride (217 μL, 2.28 mmol, 1.00 equiv.) and 4-(4.4.5.5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)phenylamine (500 mg, 2.28 mmol, 1.00 equiv.) were added to a reaction vessel containing a magnetic stirring bar, followed by 9.58 ml dry DCM and 196 pl pyridine. The reaction mixture was stirred at RT. After 20h, the reaction mixture was cooled on an ice-bath and quenched with 1M aqueous Sodium hydroxide solution (formation of yellow solution). The organic phase was separated and the aqueous phase acidified with 2M aqueous hydrochloric acid (formation of a white precipitate) and extracted three times with ethyl acetate. The combined organic phases were washed with brine and dried over sodium sulfate and evaporated to afford the crude product. Purification by flash chromatography on silica gel using a mixture of ethyl acetate and hexanes as the eluent afforded the desired product as a white solid (234 mg, 33% yield). LCMS (ESI, m/z): 309.2 [M+H]+.
(iv) N-(4-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H-pyrazolo[3,4-c(]pyrimidin-6- yl)phenyl)ethenesulfonamide N-(4-(4,4,5,5-Tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)ethanesulfonamide (60.0 mg, 1.00 equiv.) was added to a reaction vessel containing a magnetic stirring bar together with MK0016 (76.4 mg, 1.00 equiv.). BDFP (11.3 mg) and cesium carbonate (196 mg, 2.2 equiv.), followed by 1.94 mL Dioxane and 324 ul water, and the mixture heated to 100 °C. under stirring. Reaction was monitored by LC-MS After 3 h the reaction mixture was cooled to RT and quenched with a saturated aqueous sodium hydrogencarbonate solution (10 ml) and extracted with ethyl acetate (3x10 ml). The combined aqueous phases were dried over sodium sulfate, filtered and evaporated to afford the crude product as a brown oil.
The crude product was dissolved in a mixture of 4M HCI in Diox (1 ml) and iPrOH (1 ml) and stirred for 2 h at RT before evaporation of the solvent. Reaction was monitored by LC- MS. The crude product was purified by C18 reversed phase column, elution with a water/MeCN gradient with 0.1 % TFA. The fractions containing the product were lyophilized to yield pure desired product (18 mg, 20% yield) as an off-white TFA salt. LCMS (ESI, m/z): 457.7 [M+H]+. 1H NMR (600 MHz, DMSO-d6) δ 13.49 (brs, 1 H), 10.40 (d, J=12.75 Hz, 1 H), 9.41 (br s, 1 H), 8.37 (dd, J=18.80, 8.99 Hz, 2 H), 7.29 (dd, J=8.57, 4.81 Hz, 2 H), 6.20 (dd, J= 17.61 , 5.14 Hz, 1 H), 6.13 - 6.00 (m, 2H), 5.93-5.57 (m, 1 H), 3.65 - 3.11 (m, 7H), 2.59 (s, 3H), 2.38 - 2.17 (m, 2H), 2.00 (t, J=13.57, 1 H), 1.32 (d, J = 6.88 Hz, 6H).
Compound 2 N-(4-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)pyrrolidine-1-sulfonamide
This compound was prepared according to the procedure described in example 1. The desired product (5.6 mg, 8% yield) was obtained as an off-white TFA salt. LCMS (ESI, m/z): 500.7 [M+H]+. 1H NMR (600 MHz, DMSO-d6) δ 13.47 (brs, 1 H), 10.21 (d, J=14.67 Hz, 1 H), 9.39 (br s, 1 H), 8.37 (dd, J=18.66, 9.03 Hz, 2 H), 7.33 (br t, J=8.76 Hz, 2 H) , 5.87-5.66 (m,1 H), 3.58 - 3.36 (m, 10H), 2.59 (s, 3H), 2.40-2.16 (m, 2H), 2.11-1.93 (m, 1 H), 1.76-1.69 (m, 4H) ,1.46 -1.41 (m, 1 H), 1.32 (d, J = 6.9 Hz, 6H).
Compound 3 4-cyano-N-(4-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)benzenesulfonamide
This compound was prepared according to the procedure described in example 1. The desired product (103 mg, 75% yield) was obtained as an off-white TFA salt. LCMS (ESI, m/z): 532.4 [M+H]+.1H NMR (600 MHz, DMSO-d6) δ 13.51 (br s, 1 H), 10.95-10.93 (m, 1 H), 9.43 (br d, 1 H), 8.41 - 8.30 (m, 2H), 8.08 - 7.96 (m, 4H), 7.27 - 7.25 (m, 2H), 5.83 - 5.62 (m, 1 H), 3.58 - 3.18 (m, 9H), 2.58 - 2.50 (m, 1 H), 2.58 (s, 3H), 2.33 - 2.18 (m, 2H), 2.01-
1.96( m, 1 H), 1.30 (d, J = 6.6 Hz, 6H).
Compound 4 4-(aminomethyl)-N-(4-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
A solution of the compound 3 (33.5 mg. 0.07 mmol, 1.00 equiv.) in methanol (630 μL, 0.1M), at 0 °C was treated with cobalt (II) chloride (16.4 mg. 0.13 mmol, 2.00 equiv.) and the resulting mixture left to stir for 5 minutes prior to the addition of sodium borohydride (23.8 mg, 0.63 mmol, 10.0 equiv.). The resulting suspension was allowed to warm gradually to room temperature. After 16 hours 10ml of 3N solution of hydrochloric acid was added and the mixture stirred for 10 minutes followed by of 1 ml of 880 Ammonia solution. The crude product was purified by C18 reversed phase column, elution with a water/MeCN gradient with 0.1% TFA. The fractions containing the product were lyophilized to yield pure desired product (15.7 mg, 47% yield) was obtained as an off-white TFA salt. LCMS (ESI, m/z): 536.7 [M+H]+. 1H NMR (400 MHz, CDCI3): δ 13.45 (br s, 1 H), 10.91-10.65 (m, 1 H), 9.43-9.17 (br d 1 H), 8.29 (d, J= 8.6 Hz, 2H), 8.19 (br, s, 2H), 7.87 (dd, J=8.21 , 3.91 Hz, 2H), 7.60 (d, J=8.60, 2H), 7.25 (dd, J=8.79, 4.10 Hz, 2H), 5.92 - 5.47 (m, 1 H), 3.70 - 3.05 (m, 10H), 2.55 (s, 3H), 2.38 - 2.10 (m, 2H), 2.02-1.89(m, 1 H), 1.28 (d, J = 6.6 Hz, 6H). Compound 5 N-(4-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)propane-2-sulfonamide
This compound was prepared according to the procedure described in example 1. The desired product (82.7 mg, 67% yield) was obtained as an off-white TFA salt. LCMS (ESI, m/z): 473.5 [M+H]+. 1H NMR (600 MHz, DMSO-d6) δ 13.45 (br s, 1 H), 10.14-10.12 (m, 1 H), 9.31 (br s, 1 H), 8.41 - 8.36 (m, 2H), 7.39 - 7.36 (m, 1 H), 5.86 - 5.55 (m, 1 H), 3.57 - 3.18 (m, 5H), 2.58 (s, 3H), 2.30 - 2.21 (m, 2H), 1.99 -1.97 (m, 1 H), 1.47-1.41 (m, 2H), 1.30 (d, J = 6.6 Hz, 6H). Compound 6 N-(4-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)piperidine-1-sulfonamide
This compound was prepared according to the procedure described in example 1. The desired product (11.9 mg, 14% yield) was obtained as an off-white TFA salt. LCMS (ESI, m/z): 514.6 [M+H]+. 1H NMR (600 MHz, CDCI3): δ 13.46 (br, s, 1 H), 10.32-10.17(m, 1 H),
9.24(br,d, 1 H), 8.35 (dd, J=12.5, 8.99 Hz, 2H), 7.42 - 7.15 ( m, 2H), 5.85 (m, 1 H), 3.39 - 2.89 (m, 10H), 2.58 (s, 3H), 2.38 - 2.12 (m, 3H), 2.01-1.89 (m, 1 H), 1.41-1.38 (m, 6H), 1.29 (d, J = 6.64 Hz, 6H).
Compound 7 N-(4-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)pyridine-2-sulfonamide
This compound was prepared according to the procedure described in example 1. The desired product (113 mg, 79% yield) was obtained as an off-white TFA salt. LCMS (ESI, m/z): 508.6 [M+H]+. 1H NMR (600 MHz, DMSO-d6) δ 13.49 (br s, 1 H), 10.91-10.89 (m, 1 H), 9.41 (br s, 1 H), 8.96 - 8.90 (m, 1 H), 8.80 - 8.78 (m, 1 H), 8.36 - 8.19 (m, 3H), 7.63 - 7.60 (m, 1 H), 7.27 - 7.25 (m, 2H), 5.83 - 5.61 (m, 1 H), 3.57 - 3.18 (m, 5H), 2.58 (s, 3H), 2.30 - 2.21 (m, 2H), 1.99 -1.97 (m, 1 H), 1.47-1.41 (m, 2H), 1.30 (d, J = 6.6 Hz, 6H).
Compound 8 5-chloro-N-(4-(4-(2-(dimethylamino)ethoxy)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)-2-fluorobenzenesulfonamide
(i) 5-chloro-2-fluoro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl]benzenesulfonamide
A 50-mL 3-necked round-bottom flask was charged with 5-chloro-2-fluorobenzenesulfonyl chloride (1.00 g, 4.36 mmol, 1.00 equiv.), dichloromethane (10 mL), 4-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)aniline (0.956 g, 4.36 mmol, 1.00 equiv.), pyridine (0.345 g, 4.36 mmol, 1.00 equiv.) under N2. The reaction mixture was stirred overnight at room temperature and diluted with dichloromethane (50 mL). The resulting mixture was washed with water (3 x 20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with ethyl acetate/hexane (1/3) to afford desired product 5-chloro- 2-fluoro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]benzenesulfonamide (1.03 g, 57% yield) as a light yellow solid. LCMS (ESI, m/z): 412 [M+H]+.
(ii) 2-[[6-chloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- yl]oxy]ethyl)dimethylamine A 20-mL vial was charged with 4,6-dichloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4- d]pyrimidine (75.0 mg, 0.261 mmol, 1.00 equiv.), dimethylaminoethanol (23.8 mg, 0.261 mmol, 1.00 equiv.), potassium carbonate (72.2 mg, 0.522 mmol, 2.00 equiv.) and acetonitrile (3 mL). The resulting solution was stirred overnight at room temperature. The solids were filtered off and the filtrate was concentrated under reduce pressure. The residue was purified by silica gel column chromatography, eluted withdichloromethane/methanol (95/5) to afford desired product (65.0 mg, 73% yield) as an off-white solid. LCMS (ESI, m/z): 340 [M+H]+.
(iii) 5-chloro-N-(4-(4-(2-(dimethylamino)ethoxy)-3-methyl-1H-pyrazolo[3,4-c(]pyrimidin-6- yl)phenyl)-2-fluorobenzenesulfonamide
A 8 mL vial was charged with 5-chloro-2-fluoro-N-/4-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)phenyl]benzenesulfonamide (87.3 mg, 0.212 mmol, 1.20 equiv.), (2-[[6- chloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4-yl]oxy]ethyl)dimethylamine (60.0 mg, 0.177 mmol, 1.00 equiv.), 1 ,4-dioxane (2 mL), water (0.2 mL), cesium carbonate (115 mg, 0.353 mmol, 2.00 equiv.), [1 ,T-bis(diphenylphosphino)ferrocene]dichloropalladium (14.4 mg, 0.0180 mmol, 0.10 equiv.) under N2. The resulting solution was stirred overnight at 100 °C. The solids were filtered off and the filtrate was concentrated under reduce pressure. The crude product was purified by reverse phase column chromatography with the following conditions: Column: Agela C18 Column, 120 g, Mobile Phase A: Water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 40 mL/min; Gradient: 0% B to 45% B in 45 min; Detector: 220 nm to afford desired product 5-chloro-N-(4-[4-[2- (dimethylamino)ethoxy]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-6-yl]phenyl)-2- fluorobenzenesulfonamide (50.0 mg, 40% yield) as a brown solid. LCMS (ESI, m/z): 589 [M+H]+.
A 25-mL 2-necked round-bottom flask was charged with 5-chloro-N-(4-[4-[2- (dimethylamino)ethoxy]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-6-yl]phenyl)-2- fluorobenzenesulfonamide (54.0 mg, 0.092 mmol, 1.00 equiv.), isopropyl alcohol (2.6 mL),
2 M hydrochloric acid (gas) in 1 ,4-dioxane (1.5 mL). The resulting solution was stirred for
3 h at room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: Xselect CSH OBD Column 30 x 50 mm, 5 urn, Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1 %NH3.H2 O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient:18% B to 38% B in 7 min; Detector: 220 nm to afford desired product 5-chloro-N-(4-(4-(2- (dimethylamino)ethoxy)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)-2- fluorobenzenesulfonamide (12.7 mg, 27% yield)as an off-white solid. LCMS (ESI, m/z): 505 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 13.42 (s, 1 H), 10.82 (s, 1 H), 8.35 - 8.21 (m, 2H), 7.86 - 7.84 (m, 1 H), 7.74 - 7.72 (m, 1 H), 7.47 (t, J = 8.0 Hz, 1 H), 7.26 - 7.17 (m, 2H), 4.75 (t, J = 4.0 Hz, 2H), 2.89 (t, J = 6.0 Hz, 2H), 2.50 (s, 3H), 2.36 (s, 6H).
Compound 9 5-chloro-N-(4-(4-((2-(dimethylamino)ethyl)amino)-3-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)-2-fluorobenzenesulfonamide
(i) 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- amine
A 8-mL vial was charged with 4,6-dichloro-3-methyl-1-(oxan-2-yl)-2H,3H-pyrazolo[3,4- d]pyrimidine (74.3 mg, 0.259 mmol, 1.00 equiv.), (2-aminoethyl)dimethylamine (22.8 mg, 0.259 mmol, 1.00 equiv.), dichloromethane (3.0 mL) and triethylamine (57.7 mg, 0.571 mmol, 2.20 equiv.). The resulting solution was stirred overnight at room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with dichloromethane/methanol (95/5) to afford desired product 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- amine (55.0 mg, 62% yield) as a colorless solid. LCMS (ESI, m/z): 339 [M+H]+.
(ii) 5-chloro-N-(4-(4-((2-(dimethylamino)ethyl)amino)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)-2-fluorobenzenesulfonamide
A 8-mL vial was charged with 5-chloro-2-fluoro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)phenyl]benzenesulfonamide (69.2 mg, 0.168 mmol, 1.00 equiv.), 6- chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- amine (prepared from 6-chloro-3-methyl-1 H-pyrazolo 3,4-dipyrimidine analogously to the procedure described in example 8), (57.0 mg, 0.168 mmol, 1.00 equiv.), 1 ,4-dioxane (2.9 mL), water (0.3 mL), cesium carbonate (109 mg, 0.336 mmol, 2.00 equiv.), [1 ,1'- bis(diphenylphosphino)ferrocene]dichloropalladium (13.7 mg, 0.0170 mmol, 0.10 equiv.). The resulting solution was stirred overnight at 100 °C. The solids were filtered off and the filtrate was concentrated under reduced pressure. The crude product was purified by reverse phase column chromatography with the following conditions: Column, Column: Agela C18 Column, 120 g, Mobile Phase A: Water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 40 mL/min; Gradient: 0% B to 55% B in 45 min; Detector: 220 nm to afford desired product 5-chloro-N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1-(oxan-2- yl)pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide (50.0 mg, 51% yield) as a brown solid. LCMS (ESI, m/z): 588 [M+H]+.
A 25-mL 2-necked round-bottom flask was charged with 5-chloro-N-[4-(4-[[2- (dimethylamino)ethyl]amino]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]- 2-fluorobenzenesulfonamide (55.0 mg, 0.094 mmol, 1.00 equiv.), isopropanol (3 mL), 2M hydrochloric acid (gas)in 1 ,4-dioxane (1 mL). The resulting solution was stirred for 3 h at room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: Xselect CSH OBD Column 30 x 150 mm, 5 urn. Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1 %NH3.H2 O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 18% B to 38% B in 7 min; Detector: 220 nm to afford desired product 5-chloro-N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide (13.6 mg, 29% yield) as an off-white solid. LCMS (ESI, m/z): 504 [M+H]+. 1H NMR (400 MHz, DMSO- d6): δ 12.95 (s, 1 H), 10.69 (s, 1 H), 8.27 - 8.20 (m, 2H), 7.83 (dd, J = 6.0, 2.7 Hz, 1 H), 7.73 (dt, J = 8.6, 3.4 Hz, 1 H), 7.46 (t, J = 9.2 Hz, 1 H), 7.16 (d, J = 8.5 Hz, 2H), 7.02 (d, J = 6.2 Hz, 1 H), 3.79 - 3.71 (m, 2H), 2.74 (s, 2H), 2.51 (s, 3H), 2.38 (s, 6H).
Compound 10 5-chloro-N-(4-(4-(4-(dimethylamino)butoxy)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)-2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 8. The desired product (18.4 mg, 39% yield) was obtained as an off-white solid. LCMS (ESI, m/z): 519 [M+H]+. 1H NMR (300 MHz, DMSO-d6) δ 13.39 (s, 1 H), 8.33 - 8.17 (m, 2H), 7.84 - 7.81 (m, 1 H), 7.71 - 7.69 (m, 1 H), 7.43 (t, J = 9.0 Hz, 1 H), 7.31 - 7.10 (m, 2H), 4.65 (t, J =
6.0 Hz, 2H), 2.67 (t, J = 6.0 Hz, 2H), 2.50 (s, 3H), 2.36 (s, 6H), 2.08 - 1.99 (m, 2H).
Compound 11 5-chloro-N-(4-(4-((4-(dimethylamino)butyl)amino)-3-methyl-1 H- pyrazolo[3,4-c(]pyrimidin-6-yl)phenyl)-2-fluorobenzenesulfonamide This compound was prepared according to the procedure described in example 9. The desired product (17.3 mg, 40% yield) was obtained as an off-white solid. LCMS (ESI, m/z): 518 [M+H]+. 1 H NMR (400 MHz, DMSO- d6): δ 12.90 (s, 1 H), 8.21 (d, J = 8.5 Hz, 2H), 7.82 (dd, J = 6.0, 2.7 Hz, 1 H), 7.69 (s, 1 H), 7.44 (t, J = 9.0 Hz, 2H), 7.13 (d, J = 8.5 Hz, 2H), 3.69 - 3.62 (m, 2H), 2.57 (s, 2H), 2.51 (s, 3H), 2.32 (s, 6H), 1.90 - 1.82 (m, 2H). Compound 12 5-chloro-2-fluoro-N-(5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H- pyrazolo[3,4-c(]pyrimidin-6-yl)pyrimidin-2-yl)benzenesulfonamide
(i) N-(5-bromopyrimidin-2-yl)-5-chloro-2-fluorobenzenesulfonamide
To a solution of 5-bromopyrimidin-2-amine (1.50 g, 8.62 mmol, 1.00 equiv.) in tetrahydrofuran (60 mL) was added dropwise lithium hexamethyldisilazide (9.00 mL, 9.00 mmol, 1.05 equiv., 1M solution in THF) at -40 °C under N2. The resulting mixture was stirred for 0.5 hour at -40 °C and more 1 hour at room temperature. Then a solution of 5- chloro-2-fluorobenzenesulfonyl chloride (2.40 g, 10.5 mmol, 1.20 equiv.) in tetrahydrofuran (5 mL) was added dropwise at -40 °C. The resulting mixture was stirred at -40 °C for 1 hour and additional 16 hours at room temperature. The reaction mixture was quenched with saturated ammonium chloride solution (100 mL) at 0 °C, extracted with ethyl acetate (3 x 100 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with 0-15% ethyl acetate in petroleum ether to afford the desired product (750 mg, 24% yield) as a yellow solid. LCMS (ESI, m/z): 366 [M+H]+.
(ii) 5-chloro-2-fluoro-N-[5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyrimidin-2- yl]benzenesulfonamide
A mixture of N-(5-bromopyrimidin-2-yl)-5-chloro-2-fluorobenzenesulfonamide (700 mg, 1.53 mol, 1.00 equiv.), bis(pinacolato)diboron (900 mg, 3.54 mmol, 2.30 equiv.), potassium acetate (380 mg, 3.87 mmol, 2.50 equiv.) and [1 ,1'- bis(diphenylphosphino)ferrocene]dichloropalladium (112 mg, 0.150 mmol, 0.10 equiv.) in 1 ,4-dioxane (30 mL) was stirred for 16 hours at 85 °C under N2 and diluted with water (100 mL). The mixture was extracted with ethyl acetate (3 x 100 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by reverse phase column chromatography with the following condition: Column: Agela C18 Column , Mobile Phase A: Water, Mobile Phase B: ACN; Flow rate:40 mL/min; Gradient: 0% B to 80% B in 7 min; Detector: 220 nm to afford desired product (140 mg, 18% yield) as a light yellow solid. LCMS (ESI, m/z): 332 [M+H-82]+
(iii) 5-chloro-2-fluoro-N-(5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1-(tetrahydro-2H- pyran-2-yl)-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)pyrimidin-2-yl)benzenesulfonamide
A mixture of 5-chloro-2-fluoro-N-[5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyrimidin- 2-yl]benzenesulfonamide (90.0 mg, 0.220 mmol, 1.20 equiv.), 4-[[6-chloro-3-methyl-1- (oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4-yl]oxy]-1-isopropylpiperidine (prepared from 6- chloro-3-methyl-1 H-pyrazolo 3,4-dipyrimidine analogously to the procedure described in example 1 (75.0 mg, 0.190 mmol, 1.00 equiv.), sodium bicarbonate (31 mg, 0.370 mmol, 2.00 equiv.) and [1 ,1'-bis(diphenylphosphino)ferrocene]dichloropalladium (16.00 mg, 0.022 mmol, 0.10 equiv.) in 1 ,4-dioxane (3.5 mL) I ethanol (1.5 mL) / water (2.0 mL) was stirred for 2 hours at 80 °C under N2 and concentrated under reduced pressure. The residue was purified by reverse phase column chromatography with the following condition: Column: Agela C18 Column, 120 g, Mobile Phase A: Water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 40 mL/min; Gradient: 30% B to 80% B in 7 min; Detector: 220 nm to afford desired product (30.0 mg, 25% yield) as a light yellow solid. LCMS (ESI, m/z): 645 [M+H]+.
(iv) 5-chloro-2-fluoro-N-(5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)pyrimidin-2-yl)benzenesulfonamide
To a solution of 5-chloro-2-fluoro-N-(5-[4-[(1-isopropylpiperidin-4-yl)methyl]-3-methyl-1- (oxan-2-yl)-2H,3H-pyrazolo[3,4-d]pyrimidin-6-yl]pyrimidin-2-yl)benzenesulfonamide (30.0 mg, 0.046 mmol, 1.00 equiv.) in methanol (2 mL) was added 4M HCI(gas) in 1 ,4-dioxane (0.2 mL) at 0 °C. The resulting mixture was stirred for 3 hours at room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XSelect CSH Prep C18 OBD Column, 19 x 250 mm, 5 urn; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 54% B to 74% B in 8 min; Detector: 254 nm to afford desired product (8.4 mg, 32% yield) as a white solid. LCMS (ESI, m/z): 561 [M+H]+. 1H NMR (300 MHz, DMSO-d6) δ 13.62 (brs, 1 H), 9.36 - 9.10 (m, 2H), 7.99 - 7.82 (m, 1 H), 7.82 - 7.79 (m, 1 H), 7.50 (t, J = 8.7 Hz, 1 H), 5.91 - 5.71 (m, 1 H), 3.57 - 3.06 (m, 9H), 2.58 - 2.50 (m, 1 H), 2.31 - 1.82 (m, 3H), 1.30 (d, J = 6.6 Hz, 6H).
Compound 13 5-chloro-2-fluoro-N-(4-(3-methyl-4-((pyridin-4-ylmethyl)amino)-1 /7- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
(i) 6-chloro-3-methyl-1-(oxan-2-yl)-N-(pyridin-4-ylmethyl)pyrazolo[3,4-d]pyrimidin-4- amine
A 20-mL vial was charged with 4,6-dichloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4- d]pyrimidine (150 mg, 0.522 mmol, 1.00 equiv.), dichloromethane (3 mL), 4- pyridinemethaneamine (62.1 mg, 0.575 mmol, 1.10 equiv.), trimethylamine (58.2 mg, 0.575 mmol, 1.10 equiv.). The resulting solution was stirred overnight at room temperature and quenched with water (10 mL). The mixture was extracted with dichloromethane (3 x 10 mL) and the organic layers were combined, washed with water (3 x 10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by reverse phase column chromatography with the following conditions: Column: Agela C18 Column, 120 g, Mobile Phase A: Water, Mobile Phase B: ACN; Flow rate: 40 mL/min; Gradient: 5% B to 95% B in 35 min; Detector: 220 nm to afford desired product (120 mg, 64% yield)) as an off-white solid. LCMS (ESI, m/z): 359 [M+H]+.
(ii) 5-chloro-2-fluoro-N-(4-(3-methyl-4-((pyridin-4-ylmethyl)amino)-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)benzenesulfonamide
The desired product (20.0 mg, 51% yield) was obtained as a beige solid following the general procedure described in example 1. LCMS (ESI, m/z): 524 [M+H]+. 1H NMR (300 MHz, DMSO-d6): δ 13.03 (s, 1 H), 11.00 (s, 1 H), 8.52 - 8.44 (m, 2H), 8.17 - 8.08 (m, 2H), 7.84 (dd, J = 6.0, 2.7 Hz, 2H), 7.76 (ddd, J = 8.8, 4.1 , 2.7 Hz, 1 H), 7.54 - 7.42 (m, 1 H), 7.46 - 7.38 (m, 2H), 7.21 - 7.10 (m, 2H), 4.83 (d, J = 5.7 Hz, 2H), 2.61 (s, 3H).
Compound 14 5-chloro-2-fluoro-N-((5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)furan-2-yl)methyl)benzenesulfonamide-2,2,2- trifluoroacetaldehyde
(i) 5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)furan-2-carbaldehyde
A mixture of 4-[[6-chloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4-yl]oxy]-1- isopropylpiperidine, (prepared from 6-chloro-3-methyl-1 H-pyrazolo 3,4-dipyrimidine analogously to the procedure described in example 1) (500 mg, 1.27 mmol, 1.00 equiv.) and 5-formylfuran-2-ylboronic acid (300 mg, 2.14 mmol, 1.69 equiv.), [1,1'- bis(diphenylphosphino)ferrocene]dichloropalladium (103 mg, 0.140 mmol, 0.11 equiv.) and cesium carbonate (799 mg, 2.45 mmol, 1.93 equiv.) in 1 ,4-dioxane (12 mL) and water (2 mL) was stirred at 100 °C for 4 hours under N2 and quenched with water (50 mL). The mixture was extracted with ethyl acetate (3 x 100 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with 0- 100% ethyl acetate in petroleum ether to afford 5-(4-((1-isopropylpiperidin-4-yl)oxy)-3- methyl-1-(tetrahydro-2H-pyran-2-yl)-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)furan-2- carbaldehyde (450 mg, 78% yield) as a light brown solid. LCMS (ESI, m/z): 454[M+H]+.
(ii) (E)-5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)furan-2-carbaldehyde oxime A mixture of 5-[4-[(1-isopropylpiperidin-4-yl)oxy]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4- d]pyrimidin-6-yl]furan-2-carbaldehyde (430 mg, 0.95 mmol, 1.00 equiv.), sodium acetate (172 mg, 2.10 mmol, 2.20 equiv.) and hydroxylamine hydrochloride (86.0 mg, 1.24 mmol, 1 .30 equiv.) in ethanol (20 mL) was stirred for 1 h at 50 °C and concentrated under reduced pressure. The residue was diluted with water (50 mL). The mixture was extracted with ethyl acetate (3 x 100 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to afford (E)-5-(4-((1- isopropylpiperidin-4-yl)oxy)-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)furan-2-carbaldehyde oxime (350 mg, crude) as brown oil, which was used in the next step without any further purification. LCMS (ESI, m/z): 469 [M+H]+.
(iii) (5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)furan-2-yl)methanamine
To a solution of (E)-N-[(5-[4-[(1-isopropylpiperidin-4-yl)oxy]-3-methyl-1-(oxan-2- yl)pyrazolo[3,4-d]pyrimidin-6-yl]furan-2-yl)methylidene]hydroxylamine (300 mg, 0.64 mmol, 1.00 equiv.) in acetic acid (10 mL) was added zinc powder (419 mg, 6.39 mmol, 10.0 equiv.) .The resulting mixture was stirred for 2 h at 50 °C. The solids were filtered off and the filter cake was washed with acetic acid (2 x 10 mL). The combined filtrate was concentrated under reduced pressure. The residue was purified by reverse phase column chromatography with the following condition: Column: Agela C18 Column, 120 g, Mobile Phase A: Water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 40 mL/min; Gradient: 50% B to 80% B in 10 min; Detector: 254 nm to afford (5-(4-((1-isopropylpiperidin-4-yl)oxy)-3- methyl-1-(tetrahydro-2H-pyran-2-yl)-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)furan-2- yl)methanamine (50.0 mg, 17% yield) as a light brown solid. LCMS (ESI, m/z): 455 [M+H]+.
5-chloro-2-fluoro-N-((5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1-(tetrahydro-2H- pyran-2-yl)-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)furan-2-yl)methyl)benzenesulfonamide
To a solution of 1-(5-[4-[(1-isopropylpiperidin-4-yl)oxy]-3-methyl-1-(oxan-2- yl)pyrazolo[3,4-d]pyrimidin-6-yl]furan-2-yl)methanamine (50.0 mg, 0.110 mmol, 1.00 equiv.) and trimethylamine (17.0 mg, 0.160 mmol, 1.50 equiv.) in dichloromethane (3 mL) was added 5-chloro-2-fluorobenzenesulfonyl chloride (27.0 mg, 0.110 mmol, 1.00 equiv.) at 0 °C under N2. The resulting mixture was stirred for 3 hours at room temperature and quenched with water (20 mL). The mixture was extracted with dichloromethane (3 x 50 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to afford 5-chloro-2-fluoro-N-((5-(4-((1- isopropylpiperidin-4-yl)oxy)-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)furan-2-yl)methyl)benzenesulfonamide (15.0 mg, crude) as brown oil which was used in the next step without any further purification. LCMS (ESI, m/z): 647 [M+H]+.
(iv) 5-chloro-2-fluoro-N-((5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)furan-2-yl)methyl)benzenesulfonamide-2,2,2-trifluoroacetaldehyde
A mixture of 5-chloro-2-fluoro-N-((5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1- (tetrahydro-2H-pyran-2-yl)-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)furan-2- yl)methyl)benzenesulfonamide (15.0 mg, 0.023 mmol, 1.00 equiv.) in methanol (2 mL) was added 4M HCI (gas) in 1 ,4-dioxane (0.2 mL) at 0 °C. The resulting mixture was stirred at room temperature for 3 hours and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 19 x 250 mm, 5 urn; Mobile Phase A: Water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 23% B to 42% B in 7 min; Detector: 254 nm to afford 5- chloro-2-fluoro-N-((5-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)furan-2-yl)methyl)benzenesulfonamide-2,2,2-trifluoroacetaldehyde (6.5 mg, 42% yield) as a white solid. LCMS (ESI, m/z): 563 [M+H-CF3 COOH]+. 1H NMR (400 MHz, DMSO-d6) δ 13.48 (brs, 1 H), 8.86 - 8.83 (m, 1 H), 7.60 - 7.53 (m, 2H), 7.33 - 6.99 (m, 3H), 6.41 (d, J = 3.9 Hz, 1 H), 5.74 - 5.51 (m, 1 H), 4.30 (d, J = 5.2 Hz, 2H), 3.58 - 3.20 (m, 7H), 2.70 - 2.58 (m, 2H), 2.31 - 1.96 (m, 3 H), 1.40 - 1.20 (m, 6H).
Compound 15 N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl) phenyl]-3-fluoropyridine-4-sulfonamid (i) 4-(benzylsulfanyl)-3-fluoropyridine
Into a 4-chloro-3-fluoropyridine (2.00 g, 15.2 mmol, 1.00 equiv.) solution of acetonitrile (7.5 mL) were added potassium carbonate (4.20 g, 30.4 mmol, 2.00 equiv.), then, added benzyl mercaptan (1.89 g, 0.0150 mmol, 1.00 equiv.) solution of acetonitrile (15 mL) at 0 °C. Then finally the mixture was stirred at room temperature, concentrated under reduced pressure. The reaction was quenched with water at room temperature. The resulting mixture was extracted with dichloromethane (2x50 mL). The combined organic layers were washed with brine (3x50 mL), dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. T The residue was purified by silica gel column chromatography, eluted with ethyl acetate/hexane (1/3) to afford desired product 4-(benzylsulfanyl)-3-fluoropyridine (2.5 g, 72% yield) as a Brown yellow oil. LCMS (ESI, m/z): 220 [M+H]+.
(ii) 3-fluoropyridine-4-sulfonyl chloride
To a stirred solution of hydrochloric acid (12 mL) in dichloromethane (8 mL) was added NaCIO (10 mL,), 4-(benzylsulfanyl)-3-fluoropyridine (800 mg, 3.65 mmol, 1.00 equiv.) at - 5 °C under air atmosphere. The resulting mixture was stirred for 1h at -5 °C under air atmosphere. The reaction was added with dichloromethane at room temperature. The combined organic layers were washed with brine (3x30 mL), dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The crude product 3-fluoropyridine-4-sulfonyl chloride was used in the next step directly without further purification. LCMS (ESI, m/z): 196 [M+H]+.
(iii) 4,6-dichloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidine A 50-mL 3-necked round-bottom flask was charged with 4,6-dichloro-3-methyl-1 H- pyrazolo[3,4-d]pyrimidine (750 mg, 3.69 mmol, 1.00 equiv.), tetrahydrofuran (20 mL), dihydropyran (2.25 mL, 26.7mmol, 7.10 equiv.), pyridinium p-toluenesulfonate (50.0 mg, 0.199 mmol, 0.05 equiv.) under N2. The resulting solution was stirred for 3 h at 60 °C and concentrated under reduced pressure. The mixture was diluted with dichloromethane (30 mL), washed with saturated sodium bicarbonate (3x20 mL) dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with ethyl acetate/hexane (3/7) to afford desired product 4, 6-dichloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidine (750 mg, 71% yield) as an off-white solid. LCMS (ESI, m/z): 287 [M+H]+.
(iv) 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin -
4-amine
A 8-mL vial was charged with 4,6-dichloro-3-methyl-1-(oxan-2-yl)-2H,3H-pyrazolo[3,4- d]pyrimidine (74.3 mg, 0.259 mmol, 1.00 equiv.), (2-aminoethyl)dimethylamine (22.8 mg, 0.259 mmol, 1.00 equiv.), dichloromethane (3.0 mL) and triethylamine (57.7 mg, 0.571 mmol, 2.20 equiv.). The resulting solution was stirred overnight at room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with dichloromethane/methanol (95/5) to afford desired product 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- amine (55.0 mg, 62% yield) as a colorless solid. LCMS (ESI, m/z): 339 [M+H]+.
(v) 6-(4-aminophenyl)-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d] pyrimidin-4-amine
To a stirred solution of 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2- yl)pyrazolo[3,4-d]pyrimidin-4-amine (100 mg, 0.295 mmol, 1.00 equiv.) and 4-(4, 4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (77.6 mg, 0.354 mmol, 1.20 equiv.) in dioxane (2 mL) was added [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium (32.4 mg, 0.0440 mmol, 0.150 equiv.), cesium carbonate (192 mg, 0.590 mmol, 2.00 equiv.), water (0.3 mL) portions at room temperature under N2 atmosphere. The resulting mixture was stirred for 3 h at 100 °C under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: Column, Column: Agela C18 Column, 120 g, Mobile Phase A: Water (0.05% trifluoroacetic acid), Mobile Phase B: acetonitrile; Flow rate: 40 mL/min; Gradient: 0% B to 55% B in 45 min; Detector: 220 nm to afford 6-(4-aminophenyl)-N-[2- (dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4-amine(89 mg, 76% yield) as a Brown yellow solid. LCMS (ESI, m/z): 396 [M+H]+.
(vi) N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d] pyrimidin-6-yl)phenyl]-3-fluoropyridine-4-sulfonamide
To a stirred solution of 6-(4-aminophenyl)-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2- yl) pyrazolo[3,4-d]pyrimidin-4-amine (50.0 mg, 0.126 mmol, 1.00 equiv.) and 3- fluoropyridine-4-sulfonyl chloride (123 mg, 0.632 mmol, 5.00 equiv.) in dichloromethane (8 mL) was added pyridine (50.0 mg, 0.632 mmol, 5.00 equiv.) at room temperature under air atmosphere. The resulting mixture was stirred for overnight at room temperature under air atmosphere. The reaction was quenched with Water (30 mL) at room temperature. The resulting mixture was extracted with dichloromethane (3x20mL). The combined organic layers were washed with brine (3x30 mL), dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: Column, Column: Agela C18 Column, 120 g, Mobile Phase A: Water (0.05% trifluoroacetic acid), Mobile Phase B: acetonitrile; Flow rate: 40 mL/min; Gradient: 0% B to 55% B in 45 min; Detector: 220 nm to afford N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1-(oxan-2-yl) pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]-3-fluoropyridine-4-sulfonamide(60.0 mg, 42% yield) as a brown yellow solid. LCMS (ESI, m/z): 555 [M+H]+.
(vii) N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl) phenyl]-3-fluoropyridine-4-sulfonamid
To a stirred solution of N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1-(oxan-2-yl) pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-3-fluoropyridine-4-sulfonamide(35.0 mg, 0.0630 mmol, 1.00 equiv.) in isopropanol (1 mL) was added trifluoroacetic acid (1 mL) at room temperature under air atmosphere. The resulting mixture was stirred for 3h at room temperature under air atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: Column: YMC-Actus Triart C18 30*250, 5um;Mobile Phase A:Water(10mmoL/L NH4HCO3+0.1 %NH3.H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 7% B to 32% B in 7 min; 254 nm; Rt: 6.47 min ; detector, UV 254 nm. This resulted in N-[4-(4-[[2-(dimethylamino)ethyl] amino]-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]-3-fluoropyridine-4-sulfonamide(5.9 mg, 19% yield) as an off-white solid. LCMS (ESI, m/z): 471 [M+H]+. 1H NMR (400 MHz, DMSO-d6): δ 12.90 (s, 1 H), 10.51 (s, 1 H), 8.69 (s, 1 H), 8.54 (d, J = 8.0 Hz, 1 H), 8.16 - 8.14 (m, 2H), 7.75 (t, J = 8.0 Hz, 1 H), 7.06 - 6.98 (m, 3H), 3.80 - 3.75 (m, 2H), 2.89 - 2.83 (m, 2H), 2.52 (s, 3H), 2.44 (s, 6H).
Compound 16 6-amino-N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1 H- pyrazolo[3,4-d] pyrimidin-6-yl)phenyl]pyridine-3-sulfonamide
(i) 6-chloro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]pyridine-3 - sulfonamide
To a stirred solution of 6-chloropyridine-3-sulfonyl chloride(200 mg, 0.943 mmol, 1.00 equiv.) and 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)aniline(206 mg, 0.943 mmol, 1.00 equiv.) in dichloromethane (10 mL) was added Pyridine(85.8 mg, 1.085 mmol, 1.15 equiv.) dropwise at 0 °C under air atmosphere. The resulting mixture was stirred for overnight at room temperature under air atmosphere. The reaction was quenched with Water at room temperature. The resulting mixture was extracted with dichloromethane (3 x 20 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with ethyl acetate/hexane (1/1) to afford desired product 6-chloro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)phenyl]pyridine-3-sulfonamide (270 mg, 72% yield) as a yellow solid. LCMS (ESI, m/z): 361 [M+H]+.
(ii) 6-[[(4-methoxyphenyl)methyl]amino]-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan -2- yl)phenyl]pyridine-3-sulfonamide
To a stirred solution of 6-chloro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- ylphenyl]pyridine -3-sulfonamide(100 mg, 0.253 mmol, 1.00 equiv.) in 1-methyl-2- pyrrolidinone (7.5 mL) was added benzenemethanamine, 4- meth oxy- (52.1 mg, 0.380 mmol, 1.50 equiv.) dropwise at room temperature under air atmosphere. The resulting mixture was stirred for overnight at 60 °C under air atmosphere. The reaction was quenched with Water at room temperature. The resulting mixture was extracted with ethyl acetate (3 x20 mL). The combined organic layers were washed with brine (3 x 20 mL), dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with ethyl acetate/hexane (1/1) to afford desired product This resulted in 6-[[(4- methoxyphenyl)methyl]amino]-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl) phenyl]pyridine-3-sulfonamide (90.0 mg, 71% yield) as a Brown yellow solid. LCMS (ESI, m/z): 496 [M+H]+.
(iii) 4,6-dichloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidine
A 50-mL 3-necked round-bottom flask was charged with 4,6-dichloro-3-methyl-1 H- pyrazolo[3,4-d]pyrimidine (750 mg, 3.69 mmol, 1.00 equiv.), tetrahydrofuran (20 mL), dihydropyran (2.25 mL, 26.7mmol, 7.10 equiv.), pyridinium p-toluenesulfonate (50.0 mg, 0.199 mmol, 0.05 equiv.) under N2. The resulting solution was stirred for 3 h at 60 °C and concentrated under reduced pressure. The mixture was diluted with dichloromethane (30 mL), washed with saturated sodium bicarbonate (3 x 20 mL) dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with ethyl acetate/hexane (3/7) to afford desired product 4, 6-dichloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidine (750 mg, 71 % yield) as an off-white solid. LCMS (ESI, m/z): 287 [M+H]+.
(iv) 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d] pyrimidin- 4-amine
A 8-mL vial was charged with 4,6-dichloro-3-methyl-1-(oxan-2-yl)-2H,3H-pyrazolo[3,4- d]pyrimidine (74.3 mg, 0.259 mmol, 1.00 equiv.), (2-aminoethyl)dimethylamine (22.8 mg, 0.259 mmol, 1.00 equiv.), dichloromethane (3.0 mL) and triethylamine (57.7 mg, 0.571 mmol, 2.20 equiv.). The resulting solution was stirred overnight at room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with dichloromethane/methanol (95/5) to afford desired product 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- amine (55.0 mg, 62% yield) as a colorless solid. LCMS (ESI, m/z): 339 [M+H]+.
(v) N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]-6-[[(4-methoxyphenyl)methyl]amino]pyridine-3-sulfonamide
To a stirred solution of 6-[[(4-methoxyphenyl)methyl]amino]-N-[4-(4,4,5,5-tetramethyl - 1 ,3,2-dioxaborolan-2-yl)phenyl]pyridine-3-sulfonamide(90.0 mg, 0.182 mmol, 1.00 equiv.) and 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin- 4-amine(61.5 mg, 0.182 mmol, 1.00 equiv.) in dioxane (2 mL) was added [1,1'- bis(diphenylphosphino)ferrocene] dichloropalladium (19.9 mg, 0.0270 mmol, 0.150 equiv.), cesium carbonate (118 mg, 0.363 mmol, 2.00 equiv.), water(0.3 mL) at room temperature under N2 atmosphere. The resulting mixture was stirred for 3 h at 100 °C under N2 atmosphere. The crude product was purified by reverse phase column chromatography with the following conditions: Column, Column: Agela C18 Column, 120 g, Mobile Phase A: Water (0.05% trifluoroacetic acid), Mobile Phase B: acetonitrile; Flow rate: 40 mL/min; Gradient: 0% B to 55% B in 45 min; Detector: 220 nm to afford N-[4-(4- [[2-(dimethylamino)ethyl]amino] -3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-6- yl)phenyl]-6-[[(4-methoxyphenyl)methyl]amino]pyridine-3-sulfonamide(62.0 mg, 50% yield) as a Brown yellow solid. LCMS (ESI, m/z): 672 [M+H]+.
(vi) 6-amino-N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1 H-pyrazolo[3,4-d] pyrimidin-6-yl)phenyl]pyridine-3-sulfonamide
To a stirred solution of N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1- (oxan-2- yl)pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-6-[[(4-methoxyphenyl)methyl]amino]pyridine-3- sulfonamide(30.0 mg, 0.0450 mmol, 1.00 equiv.) was added trifluoroacetic acid (2 mL) at 0 °C under air atmosphere. The resulting mixture was stirred for overnight at 25 °C under air atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: Column: XSelect CSH Prep C18 OBD Column, 5um, 19*150mm; Mobile Phase A:Water(10mmoL/L NH4HCO3+0.1 %NH3.H2O ), Mobile Phase B:ACN; Flow rate:25 mL/min; Gradient:10 B to 30 B in 7 min; 220 nm; RT1:5.93; RT2; Injection Volumn: ml; Number Of Runs:;detector, UV 254 nm to afford 6-amino-N-[4-(4-[[2- (dimethylamino)ethyl]amino] -3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]pyridine- 3-sulfonamide(1.9 mg, 9% yield) as an off-white solid. LCMS (ESI, m/z): 468 [M+H]+. 1H NMR (400 MHz, DMSO-d6): δ 12.93 (s, 1 H), 11 .03 (s, 1 H), 8.26 - 8.22 (m, 2H), 7.63 - 7.60 (m, 1 H), 7.18 - 7.15 (m, 2H), 7.01 - 6.98 (m, 1 H), 6.87 (s, 3H), 6.43 - 6.40 (m, 1 H), 3.70 -
3.68 (m, 2H), 2.50 - 2.48 (m, 2H), 2.47 (s, 3H), 2.25 (s, 6H).
Compound 17 5-chloro-2-fluoro-N-(4-[4-[(1-isopropylazetidin-3-yl)oxy]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl]phenyl)benzenesulfonamide This compound was prepared according to the procedure described in example 1. The desired product as an off-white solid (35.0 mg, 50%). LCMS (ESI, m/z): 531 [M+H]+.1H NMR (300 MHz, DMSO-d6) δ 13.45 (s, 1 H), 8.24 - 8.21 (d, J = 9.0 Hz 2H), 7.84 - 7.81 (m, 1 H), 7.74 - 7.69 (m, 1 H), 7.48-7.42 (t, J = 9.0 Hz, 1 H), 5.46-5.38 (m, 1 H), 3.96-3.91 (t, J = 9.0 Hz, 1 H), 3.37-3.32 (m, 3H), 2.49 (s, 3H), 0.99-0.97 (d, J = 6.0 Hz 6H). Compound 18 N-[2-([6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl]amino)ethyl]-N-methylformamid
(i) tert-butyl N-(2-[[6-chloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- yl]amino]ethyl)-N-methylcarbamate
To a stirred solution of 4,6-dichloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidine(100 mg, 0.348 mmol, 1.00 equiv.) and tert-butyl N-(2-aminoethyl)-N-methylcarbamate (91.0 mg, 0.522 mmol, 1.50 equiv.) in dichloromethane (4 mL), trimethylamine (70.9 mg, 0.697 mmol, 2.00 equiv.) was added dropwise at room temperature under N2 atmosphere. The resulting mixture was stirred for overnight at room temperature under air atmosphere. The resulting solution was diluted with 20 mL of dichloromethane. The resulting mixture was washed with 2 x10 ml of water. The resulting mixture was concentrated under vacuum. This resulted in 130 mg (83%) of tert-butyl N-(2-[[6-chloro-3-methyl-1-(oxan-2- yl)pyrazolo[3,4-d]pyrimidin-4-yl]amino]ethyl)-N-methylcarbamate as a light yellow solid. LCMS29 (ESI, m/z): 425 [M+H]+.
(ii) tert-butyl N-[2-([6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1-(oxan- 2-yl)pyrazolo[3,4-d]pyrimidin-4-yl]amino)ethyl]-N-methylcarbamate
To a stirred solution of 5-chloro-2-fluoro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl]benzenesulfonamide (151 mg, 0.367 mmol, 1.20 equiv.) and tert-butyl N-(2-[[6- chloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4-yl]amino]ethyl)-N- methylcarbamate (130 mg, 0.306 mmol, 1.00 equiv.) in dioxane ( 2mL) was added cesium carbonate (199 mg, 0.612 mmol, 2.00 equiv.), and water(0.30 mL) then added [1 ,1'- bis(diphenylphosphino)ferrocene]dichloropalladium(ll) (38.5 mg, 0.0470 mmol, 0.180 equiv.) at room temperature under N2 atmosphere. The resulting mixture was stirred for 3 h at 100 °C under N2 atmosphere. The reaction mixture was cooled. The solids were filtered out. The crude product was purified by Flash-Prep-HPLC with the following conditions (lntelFlash-1): Column, C18 silica gel; mobile phase, ACN/water=5/95 increasing to ACN/water=56/44 within 30; Detector, 220nm. This resulted in 140 mg (64 %) of tert-butyl N-[2-([6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1- (oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4-yl]amino)ethyl]-N-methylcarbamate as a light brown solid. LCMS (ESI, m/z): 674 [M+H]+.
(iii) 5-chloro-2-fluoro-N-(4-(3-methyl-4-((2-(methylamino)ethyl)amino)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)benzenesulfonamide To a stirred solution of tert-butyl N-[2-([6-[4-(5-chloro-2- fluorobenzenesulfonamido)phenyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- yl]amino)ethyl]-N-methylcarbamate (140 mg) in dichloromethane (2 mL) was added trifluoroacetic acid (1 mL) at room temperature under N2 atmosphere. The resulting mixture was stirred for 3h at room temperature under air atmosphere. The resulting mixture was concentrated under reduced pressure. The residue 70.0 mg 5-chloro-2-fluoro- N-(4-(3-methyl-4-((2-(methylamino)ethyl)amino)-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl)benzenesulfonamide was used directly. LCMS (ESI, m/z): 490 [M+H]+.
(iv) N-[2-([6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-4-yl]amino)ethyl]-N-methylformamide
To a stirred solution of 5-chloro-2-fluoro-N-[4-(3-methyl-4-[[2-(methylamino)ethyl]amino]- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzene sulfonamide (70.00 mg, 0.163 mmol, 1.00 equiv.) in NMP (2 mL) was added 1 ,1 ,3-trioxo-1 lambda6,2-benzothiazole-2- carbaldehyde (34.5 mg, 0.163 mmol, 1.00 equiv.) at room temperature under air atmosphere. The resulting mixture was stirred for 1h at room temperature under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: XBridge Prep C18 OBD Column, 19*150mm 5um; Mobile Phase A:Water(10MMOL/L NH4HCO3 +0.1 %NH3.H2O), Mobile Phase B:ACN; Flow rate:25 mL/min; Gradient:18 B to 38 B in 7 min; Director: 220 nm. This resulted in 23.2 mg (29%) of N-[2-([6-[4-(5-chloro-2- fluorobenzenesulfonamido)phenyl]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl]amino)ethyl]-N-methylformamide as a solid. LCMS (ESI, m/z): 518 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.99 (s, 1 H), 11.02(s, 1 H), 11.13 (s, 1 H), 8.32 - 8.25 (m, 2H), 8.01- 7.84 (m, 3H), 7.77 - 7.76 (m, 1 H), 7.52-7.48 (m, 2H), 7.32 - 7.20 (s, 1 H), 3.79-3.73(m, 2H), 3.57-3.53(m, 2H),2.98-2.80(m, 3H), 2.67 (s, 3H).
Compound 19 N-(4-(4-(2-(dimethylamino)ethylamino)-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)-4-methoxypyridine-3-sulfonamide (i) 4-chloro-N-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)pyridine-3- sulfonamide
Into a 50-mL round-bottom flask, was placed 4-chloropyridine-3-sulfonyl chloride (900.00 mg, 4.244 mmol, 1.00 equiv.), 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)aniline (929.92 mg, 4.244 mmol, 1.00 equiv.), DCM (15.00 mL), pyridine (671.48 mg, 8.488 mmol, 2.00 equiv.). The resulting solution was stirred for overnight at room temperature. The resulting solution was diluted with 50 mL of DCM. The resulting mixture was washed with 2 x30 ml of brine and 1 x30 mL of water. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (0:100 to 20: 80). The collected fractions were combined and concentrated. This resulted in 700 mg (37.61%) of 4-chloro-N-[4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)phenyl]pyridine-3-sulfonamide as a off-white solid. LCMS (ESI, m/z): 395 [M+H]+.
(ii) 4-(4-methoxypyridine-3-sulfonamido)phenylboronic acid
Into a 25-mL round-bottom flask, was placed 4-chloro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)phenyl]pyridine-3-sulfonamide (650.00 mg, 1.647 mmol, 1.00 equiv.), MeOH (3.00 mL, 0.094 mmol, 0.06 equiv.), CH3 ONa (1 .00 mL). The resulting solution was stirred for overnight at 70 °C in an oil bath. The resulting mixture was concentrated. The resulting solution was diluted with 10 mL of H2O. The resulting mixture was washed with 2 x10 ml of DCM. The pH value of the solution was adjusted to 2-3 with HCI (2 mol/L). The resulting mixture was washed with 2x10 mL of EA. The water layer was concentrated. This resulted in 350 mg (crude) of 4-(4-methoxypyridine-3-sulfonamido)phenylboronic acid as light yellow oil. LCMS (ESI, m/z): 309 [M+H]+. (iii) N-(4-(4-(2-(dimethylamino)ethylamino)-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)-4-methoxypyridine-3-sulfonamide
Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of N2, was placed 4-(4-methoxypyridine-3-sulfonamido)phenylboronic acid (NaN mg, 0.649 mmol, 2.75 equiv., 60%), 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl- 1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4-amine (80.00 mg, 0.236 mmol, 1.00 equiv.), dioxane (4.00 mL), Cs2CO3 (230.78 mg, 0.708 mmol, 3.00 equiv.), water (1.00 mL), Pd(dppf)CI2 (17.28 mg, 0.024 mmol, 0.10 equiv.). The resulting solution was stirred for 40 hours at 100 °C in an oil bath. The reaction mixture was cooled with a water bath. The resulting solution and E08786-007 were diluted with 20 mL of water. The resulting mixture was washed with 2 x20 ml of DCM. The resulting mixture was concentrated. The residue was applied onto a C18 gel with H2 O (0.5% NH4HCO3)/ACN (90:10 to 10:90) in 45 minutes. The collected fractions were combined and concentrated. This resulted in 50 mg (33.63%) of N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]-4-methoxypyridine-3-sulfonamide as light yellow oil. LCMS (ESI, m/z): 567 [M+H]+
(iv) N-(4-(4-(2-(dimethylamino)ethylamino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl)-4-methoxypyridine-3-sulfonamide
Into a 50-mL round-bottom flask, was placed N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3- methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-methoxypyridine-3- sulfonamide (50.00 mg, 0.088 mmol, 1.00 equiv.), IPA (5.00 mL), HCI(gas) in 1 ,4-dioxane (5.00 mL, 0.073 mmol, 0.83 equiv.). The resulting solution was stirred for 3 hours at room temperature. The resulting mixture was concentrated. The crude product was purified by prep HPLC. Column: Xselect CSH OBD Column 30*150mm*5um, Mobile Phase A: Water(10MMOL/L NH4HCO3+0.1%NH3.H2O) , Mobile Phase B:ACN; flow rate: 60ml/min; Gradient:13% B to 38% B in 7 min; 254/220nm; Rt:5.13 min (detected by Icms and collected). The combined fractions were lyophilized to afford the desired product 14.1 mg as a white solid. LCMS (ESI, m/z): 483 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 8.81 (s, 1 H), 8.55 (d, J = 6.0 Hz, 1 H), 8.27 - 8.30 (m, 2H), 7.21 - 7.24 (m, 3H), 4.63 (s, 1 H), 4.06 (s, 3H), 3.88 (t, J = 6.7 Hz, 2H), 2.75 (t, J = 6.7 Hz, 2H), 2.62 (s, 3H), 2.40 (s, 7H). Compound 20 N-[4-[4-([2-[(2-hydroxyethyl)(methyl)amino]ethyl]amino)-5-methyl-7-
(oxan-2-yl)pyrrolo[2,3-d]pyrirnidin-2-yl]phenyl]pyridine-2-sulfonamide
This compound was prepared according to the procedure described in example 9. The desired product as an off-white solid (13.9 mg). LCMS (ESI, m/z): 482 [M+H]+. 1H NMR (300 MHz, CD3OD) δ 8.66-8.64 (m, 1 H), 8.27-8.22 (m, 2H), 8.01 - 7.96 (m, 2H), 7.57 - 7.52(m, 1 H), 7.25-7.21 (m, 2H), 3.85-3.81 (t, J= 6.6Hz 2H), 3.69-3.65 (t, J= 6.0Hz 2H), 2.83-2.78 (t, J= 6.0Hz 2H), 2.68-2.64 (t, J= 6.0Hz 2H), 2.58 (s, 3H), 2.43 (s, 3H).
Compound 21 N-[4-[4-([2-[(2-hydroxyethyl)(methyl)amino]ethyl]amino)-3-methyl-1 H- pyrazolo[3,4-d] pyrimidin-6-yl]phenyl]pyridine-3-sulfonamide
This compound was prepared according to the procedure described in example 9. The desired product as an off-white solid (41.2 mg, 60% yield). LCMS (ESI, m/z): 483 [M+H]+. 1H NMR (400 MHz, DMSO-d6): δ 12.95 (s, 1 H), 10.64 (s, 1 H), 8.93 (s, 1 H), 8.77 - 8.75 (m, 1 H), 8.25 - 8.23 (m, 2H), 8.17 - 8.14 (m, 1 H), 7.61 - 7.58 (m, 1 H), 7.19 - 7.17 (m, 2H), 7.00
- 6.97 (m, 1 H), 4.45 (s, 1 H), 3.72 - 3.67 (m, 2H), 3.50 (t, J = 8.0 Hz, 2H), 2.71 - 2.69 (m, 2H), 2.55 - 2.52 (m, 2H), 2.50 (s, 3H), 2.33 (s, 3H). Compound 225-chloro-2-fluoro-N-[4-[4-([2-[(2- hydroxyethyl)(methyl)amino]ethyl]amino)-3-methyl-1H-pyrazolo[3,4-d]pyrimidin-6- yl]phenyl]benzenesulfonamide This compound was prepared according to the procedure described in example 9. The desired product as an off-white solid (28.8 mg, 29%). LCMS (ESI, m/z): 534 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.94 (s, 1H), 13.00 (s, 1H), 10.82 (s, 1H), 8.24-8.22 (d, J =8.0 Hz, 2H), 7.84-7.82 (m, 1H), 7.75-7.72 (m, 1H), 7.48-7.44 (t, J = 8.0 Hz, 1H), 7.17 - 7.15 (d, J =8.0 Hz, 2H), 7.01-6.98(d, J =8.0 Hz, 1H), 4.50 (s, 1H), 3.74 - 3.71 (t, J =4.0 Hz, 2H), 3.53-3.50 (t, J =4.0 Hz, 2H), 2.77-2.74 (t, J =6.0 Hz, 2H), 2.67 (s, 3H),2.33 (s, 3H).
Compound 23 N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl) phenyl]pyridine-3-sulfonamide This compound was prepared according to the procedure described in example 9. The desired product as an off-white solid (26.9 mg, 31% yield). LCMS (ESI, m/z): 453 [M+H]+. 1H NMR (400 MHz, DMSO-d6): δ 12.95 (s, 1H), 11.15 (s, 1H), 8.93 (s, 1H), 8.77-8.75 (m, 1 H), 8.25 - 8.23 (d, J = 8.0 Hz, 2H), 8.17 - 8.14 (m, 1 H), 7.61 - 7.58 (m, 1 H), 7.19 - 7.17 (m, 2H), 7.01 (t, J = 4.0 Hz, 1 H), 3.74 - 3.69 (m, 2H), 2.62 (t, J = 8.0 Hz, 2H), 2.52 (s, 3H), 2.29 (s, 6H).
Compound 24 N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]pyridine-2-sulfonamide
This compound was prepared according to the procedure described in example 9. The desired product as an off-white solid (14.9 mg, 17.67%). LCMS29 (ESI, m/z): 453 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 12.93 (s, 1 H), 10.77 (s, 1 H), 8.72 - 8.70 (m, 1 H), 8.22 - 8.01 (m, 2H), 8.07 - 8.01 (m, 3H), 7.65 - 7.62 (m, 1 H), 7.23-7.21 (d, J = 8.0 Hz,
2H), 7.00 - 6.98 (t, J = 4.0 Hz, 1 H),3.72-3.67(m, 2H), 2.58-2.55 (t, J = 8.0 Hz,3H), 2.52 (s, 2H), 2.25 (s, 6H).
Compound 25 5-chloro-2-fluoro-N-(4-(4-((1-isopropylazetidin-3-yl)amino)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
(i) 3-methyl-6-(4-nitrophenyl)-1 ,3a,7,7a-tetrahydro-4H-pyrazolo[3,4-d]pyrimidin-4-one
To a stirred mixture of 3-amino-5-methyl-2H-pyrazole-4-carboxamide (1 g, 7.143 mmol, 1.0 equiv.) in ACN (20 mL) was added 4-nitrobenzaldehyde (2.15 g, 14.29 mmol, 2.0 equiv.) and I2 (3.63 g, 14.290 mmol, 2.0 equiv.) at room temperature. The resulting mixture was stirred for 4 hours at 90 °C. The resulting mixture was cooled at room temperature and filtered, the filter cake was washed with ACN (2 x 100 mL) and dried under IR lamp to afford 3-methyl-6-(4-nitrophenyl)-1 H-pyrazolo[3,4-d]pyrimidin-4-ol (900 mg, crude) as a pink solid. LCMS (ESI, m/z): 272 [M+H]+
(ii) 4-chloro-3-methyl-6-(4-nitrophenyl)-3a,4,7,7a-tetrahydro-1 H-pyrazolo[3,4-d]pyrimidine
To a stirred mixture of 3-methyl-6-(4-nitrophenyl)-1 H-pyrazolo[3,4-d]pyrimidin-4-ol (2 g, 7.35 mmol, 1.0 equiv.) in SOCI2 (17.50 g, 147 mmol, 20.0 equiv.) was DMF (0.536 g, 7.35 mmol, 1 .0 equiv.) at room temperature. The resulting mixture was stirred for 4 hours at 80 °C. The resulting mixture was cooled and quenched with water (100 mL) at room temperature, and filtered, the filter cake was washed with Et20 (2 x 100 mL) and dried under IR lamp to afford 4-chloro-3-methyl-6-(4-nitrophenyl)-1 H-pyrazolo[3,4-d]pyrimidine (1.6 g, 88% purity, 75.05% yield) as a yellow solid. LCMS (ESI, m/z): 290 [M+H]+.
(iii) N-(1-isopropylazetidin-3-yl)-3-methyl-6-(4-nitrophenyl)-1 H-pyrazolo[3,4-d]pyrimidin-4- amine
To a stirred mixture of 4-chloro-3-methyl-6-(4-nitrophenyl)-1 H-pyrazolo[3,4-d]pyrimidine (300 mg, 1.034 mmol, 1.0 equiv.) in DMF (5 mL) was DIEA (668.5 mg, 5.172 mmol, 5.0 equiv.) and 1-isopropylazetidin-3-amine (235 mg, 2.068 mmol, 2.0 equiv.) at room temperature. The resulting mixture was stirred for 2 hours at 50 °C. The mixture was then diluted with water and extracted with EtOAc. The organic layer was dried over Na2SO4, concentrated, and purified by column chromatography to give the compound of 1- isopropyl-N-[3-methyl-6-(4-nitrophenyl)-1 H-pyrazolo[3,4-d]pyrimidin-4-yl]azetidin-3- amine 160 mg (93% purity, white solid) in 46% yield. LCMS (ESI, m/z): 368 [M+H]+.
(iv) 6-(4-aminophenyl)-N-(1-isopropylazetidin-3-yl)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-
4-amine
1-isopropyl-N-[3-methyl-6-(4-nitrophenyl)-1 H-pyrazolo[3,4-d]pyrimidin-4-yl]azetidin-3- amine (160 mg, 0.435 mmol, 1.0 equiv.) and 20% Pd/C (230 mg, 0.217 mmol, 0.5 equiv.) were dissolved under H2 (5 bar) atmosphere in DMF (5 mL). The mixture was stirred for 18 hours at 25 °C. The resulting mixture was filtered, the filtrate was removed under reduced pressure to afford the white solid crude of N-[6-(4-aminophenyl)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl]-1-isopropylazetidin-3-amine (130 mg, 86.4% purity, 88.4% yield, white solid). LCMS (ESI, m/z): 338 [M+H]+
(v) 5-chloro-2-fluoro-N-(4-(4-((1-isopropylazetidin-3-yl)amino)-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)benzenesulfonamide
A solution of N-[6-(4-aminophenyl)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl]-1- isopropylazetidin-3-amine (130 mg, 0.385 mmol, 1.0 equiv.) and Pyridine (30.8 mg, 0.385 mmol, 1.0 equiv.) in anhydrous DMF (1 mL) was stirred with chloro(5-chloro-2- fluorophenyl)methylidene-lambda6-sulfanone (87.3 mg, 0.385 mmol, 1.0 equiv.) for 24 hours at room temperature. The organic layer was worked up with aqueous HCI (5%, 10 mL) and DCM (10 mL), dried over anhydrous Na2SO4 , evaporated in vacuo and purified by HPLC( Column: XBridge Shield RP18 OBD Column, 19*250mm,10um; Mobile Phase A:Water(10MMOL/L NH4HCO3+0.1 %NH3.H2O), Mobile Phase B:ACN; Flow rate:25 mL/min; Gradient: 18 B to 48 B in 10 min, 220 nm; RT1 :9.60 ) provide the 5-chloro-2-fluoro- N-(4-[4-[(1-isopropylazetidin-3-yl)amino]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl]phenyl)benzenesulfonamide (17.8 mg, 95.7 % purity, 8.73% yield, yellow solid). LCMS (ESI, m/z): 530.2 [M+H]+. 1H NMR (300 MHz, DMSO-d6) δ 12.99 (s, 1 H), 8.20 (d, J = 8.5 Hz, 2H), 7.82 (dd, J = 6.0, 2.7 Hz, 1 H), 7.70 (dt, J = 8.8, 3.4 Hz, 1 H), 7.44 (dd, J = 10.9, 7.5 Hz, 1 H), 7.17 (dd, J = 15.9, 6.9 Hz, 3H), 4.77 (q, J = 6.8 Hz, 1 H), 3.86 (s, 2H), 2.98 (s, 2H), 2.58 (s, 3H), 1.18 (d, J = 6.8 Hz, 1 H), 0.96 (d, J = 6.2 Hz, 6H).
Compound 26 5-chloro-2-fluoro-N-(4-(3-methyl-4-((2-morpholinoethyl)amino)-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
(i) 6-chloro-3-methyl-N-(2-morpholinoethyl)-1 H-pyrazolo[3,4-d]pyrimidin-4-amine Into a 50-mL round-bottom flask, was placed 4,6-dichloro-3-methyl-1H-pyrazolo[3,4- d]pyrimidine (150.00 mg, 0.739 mmol, 1.00 equiv.), DCM (15.00 mL), DIEA (286.46 mg, 2.216 mmol, 3 equiv.), N-aminoethylmorpholine (115.42 mg, 0.887 mmol, 1.20 equiv.). The resulting solution was stirred for 16 hr at room temperature. The resulting solution was extracted with 2x30 mL of ethyl acetate dried over anhydrous sodium sulfate and concentrated. This resulted in 200 mg (82.10%) of 6-chloro-3-methyl-N-[2-(morpholin-4- yl)ethyl]-1 H-pyrazolo[3,4-d]pyrimidin-4-amine as a yellow solid. LCMS (ES. m/z): 297 [M+H]+.
(ii) 5-chloro-2-fluoro-N-(4-(3-methyl-4-((2-morpholinoethyl)amino)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)benzenesulfonamide
Into a 50-mL round-bottom flask, was placed 6-chloro-3-methyl-N-[2-(morpholin-4- yl)ethyl]-1 H-pyrazolo[3,4-d]pyrimidin-4-amine (200.00 mg, 0.674 mmol, 1.00 equiv.), dioxane (16.00 mL), H2O (4.00 mL), 5-chloro-2-fluoro-N-[4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenyl]benzenesulfonamide (305.19 mg, 0.741 mmol, 1.10 equiv.), Cs2CO3 (658.75 mg, 2.022 mmol, 3 equiv.), Pd(dppf)CI2 (49.31 mg, 0.067 mmol, 0.1 equiv.). The resulting solution was stirred for 3 hr at 100 °C. The resulting solution was extracted with 3x50 mL of ethyl acetate dried in an oven under reduced pressure and concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, silica gel C18 (210 g); mobile phaseA: Water-10 mM NH4HCO3, mobile phaseB:Acetonitrile; Flow rate:50 mL/min; Gradient:55 B to 60 B; 254 nm;. The solution was concentrated. The solid was washed with CH3CN (3 mLx2). The solid was collected by filtration. This resulted in 65.3 mg (17.05%) of 5-chloro-2-fluoro-N-[4-(3- methyl-4-[[2-(morpholin-4-yl)ethyl]amino]-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl]benzenesulfonamide as a light brown solid. 1H-NMR (CD3OD, 300 MHz) δ (ppm): 8.29 (d, J = 9.0 Hz, 2H), 7.86-7.89 (m, 1 H), 7.60-7.64 (m, 1 H), 7.21-7.32(m, 3H), 3.85-3.91 (m, 2H), 3.70-3.73 (m, 4H), 2.71-2.80 (m, 2H), 2.67 (s, 7H). LCMS(ES. m/z): 546 [M+H]+.
Compound 27 N-(4-(4-((2-(dimethylamino)ethyl)amino)-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)-4-methoxypyridine-2-sulfonamide
This compound was prepared according to the procedure described in example 9. The desired product as an off-white solid (21.9 mg, 11.6% yield). LCMS (ESI, m/z): 483.3 [M+H]+. 1H NMR (300 MHz, DMSO-d6) δ 12.95 (s, 1 H), 8.52 (d, J = 5.6 Hz, 1 H), 8.27 - 8.18 (m, 2H), 7.51 (d, J = 2.5 Hz, 1 H), 7.29 - 7.17 (m, 3H), 7.02 (t, J = 5.7 Hz, 1 H), 3.90
(s, 3H), 3.72 (q, J = 6.5 Hz, 2H), 2.62 (t, J = 6.8 Hz, 2H), 2.52 (s, 3H). 2.29 (s, 6H).
Compound 28 5-chloro-2-fluoro-N-(4-(4-((1-isopropylpiperidin-4-yl)oxy)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
This compound was prepared according to the procedure described in example 9. The desired product as a white solid (37.6 mg, 32.16%). 1H-NMR (DMSO-d6, 300 MHz) δ (ppm): 13.6 (s, 1 H), 7.89-7.96 (m, 2H), 7.69-7.23 (m, 1 H), 7.53-7.57 (m, 1 H), 7.25-7.34 (m, 2H), 5.70 (s, 1 H), 3.61 (s, 1 H), 3.22 (s, 4H), 2.48 (s, 3H), 1 .97-2.27 (m, 4H), 1 .25 (d, J = 6.6 Hz, 6H). LCMS (ES. m/z): 577 [M+H]+.
Compound 29 5-chloro-2-fluoro-N-(4-(4-(((3S,4R)-3-fluoropiperidin-4-yl)oxy)-3-methyl- 1H-pyrazolo[3,4-c(]pyrimidin-6-yl)phenyl)benzenesulfonamide
(i) tert-butyl-(3S,4R)-4-((6-chloro-3-methyl-1H-pyrazolo[3,4-c(]pyrimidin-4-yl)oxy)-3- fluoropiperidine-1 -carboxylate
Into a 50-mL round-bottom flask, was placed 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4- d]pyrimidine (100.00 mg, 0.493 mmol, 1.00 equiv.), tert-butyl (3S,4R)-3-fluoro-4- hydroxypiperidine-1 -carboxylate (118.79 mg, 0.542 mmol, 1.10 equiv.), THF (10.00 mL). The resulting solution was stirred for 20 min at room temperature. Then added NaH (59.10 mg, 2.463 mmol, 5 equiv.) at 0 °C. The resulting solution was stirred for 2 hr at room temperature. The reaction was then quenched by the addition of water. The resulting solution was extracted with 2x50 mL of ethyl acetate dried over anhydrous sodium sulfate and concentrated. The resulting mixture was washed with EtOAc. The solids were collected by filtration. This resulted in 110 mg (52.10%) of tert-butyl (3S,4R)-4-([6- chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl]oxy)-3-fluoropiperidine-1 -carboxylate as a light yellow solid. LCMS(ES. m/z): 386 [M+H]+.
(ii) tert-butyl (3S,4R)-4-((6-(4-((5-chloro-2-fluorophenyl)sulfonamido)phenyl)-3-methyl- 1H-pyrazolo[3,4-c(]pyrimidin-4-yl)oxy)-3-fluoropiperidine-1-carboxylate
Into a 40-mL sealed tube purged and maintained with an inert atmosphere of N2, was placed tert-butyl (3S,4R)-4-([6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl]oxy)-3- fluoropiperidine-1 -carboxylate (100.00 mg, 0.259 mmol, 1.00 equiv.), dioxane (8.00 mL), H2O (2.00 mL), 5-chloro-2-fluoro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl]benzenesulfonamide (106.70 mg, 0.259 mmol, 1.00 equiv.), Cs2CO3 (126.67 mg, 0.389 mmol, 1.5 equiv.), Pd(dppf)CI2 (37.93 mg, 0.052 mmol, 0.2 equiv.). The resulting solution was stirred for 16 hr at 100 °C. The resulting mixture was concentrated. The residue was purified by preparative TLC (DCM:MeOH = 20:1). This resulted in 150 mg (72.90%) of tert-butyl (3S,4R)-4-([6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]- 3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl]oxy)-3-fluoropiperidine-1-carboxylate as a yellow solid. LCMS(ES. m/z): 635 [M+H]+.
(iii) 5-chloro-2-fluoro-N-(4-(4-(((3S,4R)-3-fluoropiperidin-4-yl)oxy)-3-methyl-1H- pyrazolo[3,4-c]pyrimidin-6-yl)phenyl)benzenesulfonamide
Into a 50-mL round-bottom flask, was placed tert-butyl (3S,4R)-4-([6-[4-(5-chloro-2- fluorobenzenesulfonamido)phenyl]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl]oxy)-3- fluoropiperidine-1 -carboxylate (130.00 mg, 0.205 mmol, 1.00 equiv.), DCM (5.00 mL), HCI (4M in 1 ,4-dioxane) (5.00 mL, 87.587 mmol, 427.88 equiv.). The resulting solution was stirred for 2 hr at room temperature. The resulting mixture was concentrated. The solid by Prep-HPLC (Column: YMC-Actus Triart C18, 30 mm x150 mm, 5um; Mobile Phase A:Water(10MMOL/L NH4HCO3+0.1 %NH3.H2O), Mobile Phase B:ACN; Flow rate:60 mL/min; Gradient:25 B to 45 B in 7 min, 254 nm; RT1 :6.73; RT2:; Injection Volumn: ml; Number Of Runs:;). This resulted in 20.7 mg (18.18%) of 5-chloro-2-fluoro-N-[4-(4- [[(3S,4R)-3-fluoropiperidin-4-yl]oxy]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl]benzenesulfonamide as a white solid. 1H-NMR (CD3OD, 300 MHz) δ (ppm): 8.32 (d, J = 9 Hz, 2H), 7.86-7.89 (m, 1 H), 7.57-7.62 (m, 1 H), 7.22-7.30 (m, 3H), 5.68-5.89 (m, 1 H), 5.08 (s, 1 H), 3.06-3.21 (m, 2H), 2.79-3.02 (m, 2H), 2.59 (s, 3H), 2.04-3.01 (m, 2H).LCMS(ES. m/z): 535 [M+H]+.
Compound 30 N-(4-(4-((2-(dimethylamino)ethyl)amino)-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)-4-(trifluoromethyl)pyridine-2-sulfonamide
This compound was prepared according to the procedure described in example 9. The desired product as a white solid (41.6 mg). 1H NMR (300 MHz, DMSO-d6) δ 12.91 (s, 1 H), 8.98 (d, J = 5.0 Hz, 1 H), 8.24 - 8.15 (m, 3H), 8.10 - 7.99 (m, 1 H), 7.19 (d, J = 8.7 Hz, 2H), 6.99 (t, J = 5.6 Hz, 1 H), 3.72 (d, J = 6.3 Hz, 2H), 2.66 (t, J = 6.7 Hz, 2H), 2.50 (s, 3H), 2.31 (s, 6H). LCMS (ES, m/z): 521 [M+H]+.
Compound 31 N-(4-(4-((2-(dimethylamino)ethyl)amino)-3-methyl-1 H-pyrazolo[3,4- cflpyrimidin-6-yl)phenyl)-5-(trifluoromethyl)pyridine-2-sulfonamide
This compound was prepared according to the procedure described in example 9. The desired product as a white solid (17.8 mg). 1H NMR (300 MHz, DMSO-d6) δ 12.94 (s, 1 H), 9.14 (s, 1 H), 8.48 (dd, J = 8.3, 2.3 Hz, 1 H), 8.20 (dd, J = 8.3, 5.1 Hz, 3H), 7.20 (d, J = 8.5 Hz, 2H), 7.02 (s, 1 H), 3.72 (d, J = 6.2 Hz, 2H), 2.70 (s, 2H), 2.50 (s, 3H), 2.34 (s, 6H). LCMS (ES, m/z): 521 [M+H]+.
Compound 32 N-(4-(4-((2-(diethylamino)ethyl)amino)-3-methyl-1H-pyrazolo[3,4- cflpyrimidin-6-yl)phenyl)-2,5-difluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 28. The desired product as an off-white solid ( 39.1 mg, 16.4%). 1H-NMR (DMSO-d6, 300 MHz) δ (ppm): 12.95 (s, 1 H), 10.63 (s, 1 H), 8.22 (d, J = 8.7 Hz, 2H), 7.69-7.63 (m, 1 H), 7.57 (d, J = 3.6 Hz, 2H), 7.15 (d, J = 8.7 Hz, 2H), 7.03-6.99 (m, 1 H), 3.71 (d, J = 5.7 Hz, 2H), 2.82- 2.78 (m, 2H), 2.72-2.61 (m, 4H), 2.53 (s, 3H), 1.08 (s, 6H). LCMS (ES. m/z): 516 [M+H]+.
Compound 33 2,5-difluoro-N-(4-(3-methyl-4-((2-(pyrrolidin-1-yl)ethyl)amino)-1H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
This compound was prepared according to the procedure described in example 28. The desired product as a white solid to provide (60.0 mg, 99.4% purity). LCMS (ESI, m/z): 514.4 [M+H]+. 1H NMR (300 MHz, DMSO-d6) δ 12.94 (s, 1 H), 8.29 - 8.13 (m, 2H), 7.65 (ddd, J = 8.0, 5.4, 3.1 Hz, 1 H), 7.56 - 7.38 (m, 2H), 7.21 - 7.00 (m, 3H), 3.77 (q, J = 6.4 Hz, 2H), 2.89 (t, J = 6.7 Hz, 2H), 2.75 (d, J = 6.3 Hz, 4H), 2.53 (s, 3H), 1.74 (h, J = 3.0 Hz, 4H).
Compound 34 2,5-difluoro-N-(4-(3-methyl-4-((1-methylazetidin-3-yl)oxy)-1H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide This compound was prepared according to the procedure described in example 8. The desired product as a white solid to provide the desired product as a TFA salt (9.1 mg, 99.1 % purity).1H NMR (300 MHz, DMSO-d6) δ 13.62 (s, 1 H), 11.18 (s, 1 H), 8.28 (d, J = 8.5 Hz, 2H), 7.79 - 7.69 (m, 1 H), 7.56 (dtd, J = 18.3, 9.0, 8.6, 4.2 Hz, 2H), 7.32 - 7.23 (m, 2H), 5.71 (s, 1 H), 4.71 (s, 2H), 4.41 (s, 2H), 2.97 (s, 3H), 2.58 (s, 3H), 1.24 (s, 1 H). ). LCMS (ESI, m/z): 487.1 [M+H]+.
Compound 35 5-chloro-2-fluoro-N-(4-(4-((2-((2-hydroxyethyl)(methyl)amino) ethyl)(methyl)amino)-3-methyl-1H-pyrazolo[3,4-c(]pyrimidin-6-yl)phenyl)benzene sulfonamide
OH
(i) tert-butyl (2-((2-hydroxyethyl)(methyl)amino)ethyl)(methyl)carbamate
A mixture of 2-(methylamino)ethan-1-ol (1.10 g, 14.67 mmol), tert-butyl (2- (methylamino)ethyl)carbamate (2.54 g, 14.67 mmol), Sodium triacetoxyborohydride (6.22 g, 29.34 mmol) and AcOH (0.1 mL) in CHCI3 (40 mL) was stirred for 16 hours at room temperature. The reaction mixture was washed with saturated aqueous NaHCO3 (20 mL) then brine (20 mL). The organic layer was dried anhydrous Na2SO4 and filtered The filtrate was concentrated in vacuo. This resulted in tert-butyl (2-((2- hydroxyethyl)(methyl)amino)ethyl)(methyl)carbamate (720 mg, Y=23%) as light yellow oil, which was used in the next step directly without any further purification. LCMS (ES, m/z): 233 [M+H]+.
(ii) 2-(methyl(2-(methylamino)ethyl)amino)ethan-1-ol
To a solution of tert-butyl (2-((2-hydroxyethyl)(methyl)amino)ethyl)(methyl) carbamate (720 mg, 3.09 mmol) in DCM (12 mL) was added TFA (4 mL). The resulting mixture was stirred for 16 hours at room temperature. The reaction mixture was concentrated in vacuo. This resulted in 2-(methyl(2-(methylamino)ethyl)amino)ethan-1-ol (530 mg crude, TFA salt form) as light brown oil, which was used in the next step directly without any further purification. LCMS (ES, m/z): 133 [M+H]+.
(iii) 2-((2-((6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl)(methyl)amino)ethyl)(methyl)amino)ethan-1-ol
A mixture of 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidine (200 mg, 0.99 mmol), 2- (methyl(2-(methylamino)ethyl)amino)ethan-1-ol (530 mg crude, TFA salt form) and DIPEA (1.67 mL, 9.99 mmol) in DCM (10 mL) was stirred for 16 hours at room temperature. The reaction mixture was concentrated in vacuo. The residue was purified by reverse phase chromatography with the following condition: (Column: Agela C18 Column, 120 g; Mobile Phase A:Water(10mmol/L NH4HCO3), Mobile Phase B:ACN; Flow rate:40 mL/min; Gradient:O B to 100% B in 20 min; 254 nm. This resulted in 2-((2-((6-chloro-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl)(methyl)amino)ethyl)(methyl)amino)ethan-1-ol (280 mg, Y=95%) as a light yellow solid. LCMS (ES, m/z): 299 [M+H]+.
(iv) 5-chloro-2-fluoro-N-(4-(4-((2-((2-hydroxyethyl)(methyl)amino)ethyl)(methyl)amino)-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide trifluoroacetate
A mixture of 2-((2-((6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl)(methyl)amino)ethyl)(methyl)amino)ethan-1-ol (100 mg, 0.33 mmol), 5-chloro-2-fluoro- N-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)benzenesulfonamide (204 mg, 0.50 mmol), cesium carbonate (196 mg, 0.60 mmol) and Pd(dppf)CI2 (22 mg, 0.03 mmol) in 1 ,4-dioxane (4 mL) and water (1 mL) was degassed and refilled with N2 for three times, then stirred at 100 °C for 3 hours under N2. The reaction mixture was cooled to ambient temperature and filtered. The filtrate was purified by prep-HPLC with the following condition: Column: YMC-Actus Triart C18, 30 mm X 150 mm, 5um; Mobile Phase A: Water(0.05%TFA ), Mobile Phase B: ACN; Flow rate:60 mL/min; Gradient:20% B to 50% B in 7 min; 254 nm; RT:5.12. This resulted in 5-chloro-2-fluoro-N-(4-(4-((2-((2- hydroxyethyl)(methyl)amino)ethyl)(methyl)amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin- 6-yl)phenyl)benzenesulfonamide trifluoroacetate (11.9 mg) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 13.27 (brs, 1 H), 11.08 (s, 1 H), 9.36 (s, 1 H), 8.32 - 8.24 (m, 2H), 7.86 (dd, J = 6.1 , 2.7 Hz, 1 H), 7.80 (ddd, J = 8.8, 4.2, 2.7 Hz, 1 H), 7.52 (t, J = 9.3 Hz, 1 H), 7.29 - 7.20 (m, 2H), 4.27 - 4.16 (m, 2H), 3.80 - 3.50 (m, 4H), 3.50 - 3.30 (m, 4H), 3.20 (dd, J = 13.1 , 5.7 Hz, 1 H), 2.89 (d, J = 4.3 Hz, 3H), 2.61 (s, 3H). LCMS (ES, m/z): 548 [M+H]+- 114. Compound 36 2,5-difluoro-N-(3-methyl-4-(3-methyl-4-((2-(pyrrolidin-1-yl)ethyl)amino)- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
This compound was prepared according to the procedure described in example 9. The desired product was obtained as a white a TFA salt (40.6 mg, 99.6% purity). 1H NMR (300 MHz, DMSO-d6) δ 10.96 (s, 1 H), 9.56 (s, 1 H), 7.77 - 7.67 (m, 2H), 7.58 (dtt, J = 18.3, 9.1 , 4.8 Hz, 2H), 7.41 (s, 1 H), 7.11 - 7.00 (m, 2H), 3.84 (d, J = 5.8 Hz, 2H), 3.61 (s, 2H), 3.41 (t, J = 5.7 Hz, 2H), 3.01 (s, 2H), 2.59 (s, 3H), 2.44 (s, 3H), 1.90 (s, 2H), 1.66 (s, 2H). LCMS (ESI, m/z): 528 [M+H]+. Compound 37 5-chloro-2-fluoro-N-(4-(3-methyl-4-((2-(pyrrolidin-1-yl)ethyl)amino)-1H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
This compound was prepared according to the procedure described in example 9. The desired product was obtained as a white TFA salt solid (14.3 mg, 99.6% purity). 1H NMR (300 MHz, DMSO-d6) δ 11.09 (s, 1 H), 9.58 (s, 1 H), 8.34 - 8.24 (m, 2H), 7.90 - 7.75 (m,
2H), 7.53 (t, J = 9.3 Hz, 1 H), 7.39 - 7.19 (m, 3H), 3.95 (d, J = 6.0 Hz, 2H), 3.73 - 3.59 (m, 2H), 3.54 - 3.40 (m, 2H), 3.11 (d, J = 9.6 Hz, 2H), 2.57 (s, 3H), 2.00 (s, 2H), 1.90 - 1.73 (m, 2H). LCMS (ESI, m/z): 530 [M+H]+.
Compound 38 5-chloro-N-(4-(4-((2-(dimethylamino)ethyl)amino)-3-methyl-1 H- pyrazolo[3,4-c(]pyrimidin-6-yl)-2-fluorophenyl)-2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 9. The desired product was obtained as a white TFA salt solid (28.6 mg, 19%). LCMS (ES. m/z): 522 [M+H]+. 1H-NMR (CD3OD, 300 MHz) δ (ppm): 8.19-8.10 (m, 1 H), 8.09-8.06 (m, 1 H), 7.81-7.78 (m, 1 H), 7.68-7.53 (m, 2H), 7.35-7.29 (m, 1 H), 4.16-4.12 (m, 2H), 3.57-3.53 (m, 2H), 2.98 (s, 6H), 2.65 (s, 3H).
Compound 39 N-(4-(4-((2-(dimethylamino)ethyl)amino)-3-ethyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)-2,5-difluorobenzenesulfonamide
(i) 6-chloro-N-[2-(dimethylamino)ethyl]-1 H-pyrazolo[3,4-d]pyrimidin-4-amine A solution of 4,6-dichloro-1 H-pyrazolo[3,4-d]pyrimidine (2.0 g, 10.5 mmol, 1.0 equiv.) and (2-aminoethyl)dimethylamine (2.72 g, 21.1 mmol, 1.0 equiv.) in anhydrous THF (20 mL) was added DIEA (1.12 g, 10.5 mmol, 1.0 equiv.) in 0 °C, and stirred for 4 h at room temperature. The organic layer was worked up with H2O (20 mL), extracted with DCM (30 mL), dried over anhydrous Na2SO4, and evaporated in vacuo to provide the crude of the desired product (1.3 g, 93% purity, light yellow solid).
(ii) N-[4-(4-[[2-(dimethylamino)ethyl]amino]-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2,5- difluorobenzenesulfonamide
To the solution of 6-chloro-N-[2-(dimethylamino)ethyl]-1 H-pyrazolo[3,4-d]pyrimidin-4- amine (1.3 g, 5.39 mmol, 1.0 equiv.), Pd(dppf)CI2 (788.6 mg, 1.08 mmol, 0.2 equiv.), and Cs2CO3 (2.64 mg, 8.09 mmol, 1.5 equiv.) in 1.4-dioxane (24 mL) and H2O (6 ml) was added 2,5-difluoro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl]benzenesulfonamide (2.34 g, 5.93 mmol, 1.1 equiv.) under an atmosphere of nitrogen at room temperature. The reaction mixture was purged with nitrogen 3 times and stirred at 100 °C for 12 hours under nitrogen. The reaction was cooled to the room temperature and neutralized with saturated aqueous 1 N HCI to pH 7~8 and washed by DCM (3*100 mL). The combined organic phase was dried over Na2SO4 and concentrated under reduced pressure to provide the crude. Then the crude purified by HPLC chromatography to provide the desired product (350.0 mg, 90% purity, off-white solid).
(iii) N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-iodo-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl]-2,5-difluorobenzenesulfonamide
To a stirred solution of the N-[4-(4-[[2-(dimethylamino)ethyl]amino]-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]-2,5-difluorobenzenesulfonamide(300.0 mg, 0.634 mmol, 1.0 equiv.) in HOAc (10 mL) was added NIS(143 mg, 0.634 mol, 1.0 eq), The resulting mixture was stirred for overnight at rt . The resulting mixture was concentrated under reduced pressure and diluted with water (100 mL). The residue was acidified to pH=7 with NaOH aq. (1 M). The mixture was extracted with DCM. The organic layer was dried over Na2SO4, concentrated and purified by column chromatography to give the desired product (Peak A: 90 mg, 70 % purity, off-white solid; Peak B: 120 mg, 55 % purity, off-white solid).
(iv) N-[4-(4-[[2-(dimethylamino)ethyl]amino]-5-ethenyl-7H-pyrrolo[2,3-d]pyrimidin-2- yl)phenyl]-2,5-difluorobenzenesulfonamide To the solution of N-[4-(4-[[2-(dimethylamino)ethyl]amino]-5-iodo-7H-pyrrolo[2,3- d]pyrimidin-2-yl)phenyl]-2,5-difluorobenzenesulfonamide (120 mg, 0.2 mmol, 1.0 equiv.), Pd(dppf)CI2 (43.9 mg, 0.06 mmol, 0.3 equiv.), and K2CO3(55.2 mg, 0.4 mmol, 2 eq) in dioxane (4 mL) and H2O (1 ml) was added 2-ethenyl-4,4,5,5-tetramethyl-1 ,3,2- dioxaborolane (93 mg, 0.6 mmol, 3 equiv.) under an atmosphere of nitrogen at room temperature. The reaction mixture was purged with nitrogen 3 times and stirred at 100 °C for 18 h under nitrogen. The reaction was cooled to the room temperature and neutralized with saturated aqueous 1 N HCI to pH 7~8 and washed by DCM (3*5 mL). The combined organic phase was dried over Na2SO4 and concentrated under reduced pressure to provide the crude. Then the crude purified by reversed-phase chromatography(10 mmol/L NH4HCO3/ACN) to provide the desired product (75 mg, 50% purity, off-white solid).
(v) N-[4-(4-[[2-(dimethylamino)ethyl]amino]-5-ethyl-7H-pyrrolo[2,3-d]pyrimidin-2- yl)phenyl]-2,5-difluorobenzenesulfonamide
To the solution of N-[4-(4-[[2-(dimethylamino)ethyl]amino]-5-ethenyl-7H-pyrrolo[2,3- d]pyrimidin-2-yl)phenyl]-2,5-difluorobenzenesulfonamide (75 mg, 0.15 mmol, 1.0 equiv.) in CH3OH (5 mL) and DMF (5 mL) was add Pd/C (32 mg, 0.03 mmol, 0.2 equiv.) under an atmosphere of H2 at room temperature. The reaction mixture was purged with nitrogen 3 times and stirred at 25 °C for 20 h. The combined organic phase was filtrated and concentrated under reduced pressure to provide the crude. Then the crude purified by reversed-phase chromatography (Column: Xselect CSH OBD Column 30*150mm 5um, n; Mobile Phase A: Water(0.05%TFA ), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 9% B to 39% B in 9 min, 39% B; Wave Length: 254 nm; RT1(min): 7.58; Number Of Runs: 0) to provide the desired product as a TFA salt (24 mg, 96.5 % purity, white solid). LCMS (ESI, m/z): 502 [M-2TFA+H]*. 1H NMR (300 MHz, DMSO-d6) δ 11.10 (s, 1 H), 9.49 (s, 1 H), 8.33-8.24 (m, 2H), 7.76-7.49 (m, 3H), 7.29-7.18 (m, 3H), 3.97 (d, J = 5.7 Hz, 2H), 3.41 (d, J = 5.7 Hz, 2H), 2.98 (q, J = 7.5 Hz, 2H), 2.88 (d, J = 4.7 Hz, 6H), 1.28 (t, J = 7.5 Hz, 3H).
Compound 40 N-(2-((6-(4-((5-chloro-2-fluorophenyl)sulfonamido)-2-methylphenyl)-3- methyl-1H-pyrazolo[3,4-c(]pyrimidin-4-yl)amino)ethyl)-/\/-methylformamide
(i) tert-butyl (2-(N-methylformamido)ethyl)carbamate
A suspension of tert-butyl N-[2-(methylamino)ethyl]carbamate (2.00 g, 11.478 mmol) in ethyl formate (6 mL, 72.87 mmol) was stirred for 3 hours at 60 °C. The resulting mixture was cooled, concentrated in vacuo. The residue was applied onto a silica gel column with 0 - 3% MeOH in DCM. This resulted in tert-butyl N-[2-(N- methylformamido)ethyl]carbamate (2.12 g, Y = 82%) as yellow oil. LCMS (ESI, m/z): 203 [M+H]+.
(ii) N-(2-aminoethyl)-N-methylformamide
To a solution of tert-butyl (2-(N-methylformamido)ethyl)carbamate (2.12g, 10.50 mmol) in DCM (30 mL) was added TFA (10 mL). The resulting mixture was stirred for 16 hours at room temperature. The reaction mixture was concentrated in vacuo. This resulted in N-(2- aminoethyl)-N-methylformamide (2.00 g crude, TFA salt form) as light brown oil, which was used in the next step directly without any further purification. LCMS (ES, m/z): 103 [M+H]+
(iii) N-(2-((6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl)amino)ethyl)-N- methylformamide
A mixture of 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidine (1.97.g, 9.54 mmol), N- (2-aminoethyl)-N-methylformamide (2.00 mg crude, TFA salt form) and DI PEA (15.7 mL, 95.4 mmol) in DCM (30 mL) was stirred for 16 hours at room temperature. The reaction mixture was concentrated in vacuo. The residue was purified by reverse phase chromatography with the following condition: (Column: Agela C18 Column, 330 g; Mobile Phase A:Water(10mmol/L NH4HCO3), Mobile Phase B:ACN; Flow rate:40 mL/min; Gradient:0% B to 100% B in 20 min; 254 nm. This resulted in N-(2-((6-chloro-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-4-yl)amino)ethyl)-N-methylformamide (2.05 g, Y = 73%) as a light yellow solid. LCMS (ES, m/z): 269 [M+ H]+.
(iv) 5-chloro-2-fluoro-N-(3-methyl-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl) benzenesulfonamide
A mixture of 5-chloro-2-fluorobenzenesulfonyl chloride (97 mg, 0.429 mmol), pyridine (101 mg, 1.28 mmol) and 3-methyl-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)aniline (100 mg, 0.429 mmol) in DCM (5 mL) was stirred for 16 hours at room temperature. The resulting mixture was concentrated in vacuo. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1/3). This resulted in 5-chloro-2-fluoro-N-(3- methyl-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)benzenesulfonamide (148 mg, Y= 81%) as a white solid. LCMS (ESI, m/z): 424 [M-1]_.
(v) N-(2-((6-(4-((5-chloro-2-fluorophenyl)sulfonamido)-2-methylphenyl)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl)amino)ethyl)-N-methylformamide
A mixture of 5-chloro-2-fluoro-N-[3-methyl-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl]benzenesulfonamide (80 mg, 0.194 mmol)N-[2-([6-chloro-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl]amino)ethyl]-N-methylformamide (50 mg, 0.194 mmol), Pd(dppf)CI2.CH2Cl2 (15.83 mg, 0.019 mmol), and Cs2CO3 (126.63 mg, 0.389 mmol) in dioxane (2 mL) and water (0.5 mL) was stirred for 3 hours at 100 °C under nitrogen. The reaction mixture was cooled, concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: Column: Xselect CSH OBD Column 30*150mm 5um, n; Mobile Phase A:Water(0.05%TFA ), Mobile Phase B:ACN; Flow rate:60 mL/min; Gradient:5% B to 50% B in 13 min; 254 nm; RT:7.55. This resulted in N- [2-([6-[4-(5-chloro-2-fluorobenzenesulfonamido)-2-methylphenyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl]amino)ethyl]-N-methylformamide as a TFA salt (14.9 mg) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 11.04 (brs, 1 H), 7.92 - 7.80 (m, 3H), 7.75 - 7.73 (m, 1 H), 7.55 - 7.49 (m, 1 H), 7.11 - 7.04 (m, 2H), 3.76 - 3.65 (m, 3H), 3.64 - 3.59 (m, 2H), 2.94 - 2.71 (m, 3H), 2.67 - 2.58 (m, 2H), 2.51 - 2.42 (m, 3H). LCMS (ES, m/z): 532 [M+H]+-114.
Compound 41 5-chloro-2-fluoro-N-(4-(4-(((3S,4R)-3-fluoro-1-isopropylpiperidin-4- yl)oxy)-3-methyl-1H-pyrazolo[3,4-c(]pyrimidin-6-yl)phenyl)benzenesulfonamide
(i) 5-chloro-2-fluoro-N-[4-(4-[[(3S,4R)-3-fluoro-1-isopropylpiperidin-4-yl]oxy]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
To a stirred mixture of (3S,4R)-4-([6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl]oxy)-3-fluoro-1 -isopropylpiperidine (50 mg, 0.153 mmol, prepared following the procedure in example 29) and (3S,4R)-4-([6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin- 4-yl]oxy)-3-fluoro-1-isopropylpiperidine (50 mg, 0.153 mmol) in 1 ,4-dioxane (3.0 mL) and H2O (1.0 mL) were added Pd(dppf)CI2.CH2Cl2 (13 mg, 0.015 mmol) and Cs2CO3 (100 mg, 0.306 mmol) .The flask was evacuated and flushed with nitrogen for three times. The resulting mixture stirred at 100 °C for 3 hours under nitrogen. The reaction mixture was cooled to ambient temperature and filtered. The filtrate was purified by prep-HPLC with the following condition: Column: YMC-Actus Triart C18, 30 mm X 150 mm, 5um; Mobile Phase A: Water(0.05%TFA ), Mobile Phase B: ACN; Flow rate:60 mL/min; Gradient:20% B to 50% B in 7 min; 254 nm; RT:5.12. This resulted in afford 5-chloro-2-fluoro-N-[4-(4- [[(3S,4R)-3-fluoro-1-isopropylpiperidin-4-yl]oxy]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl]benzenesulfonamide trifluoroacetic acid (11.3 mg, Y = 11 %, mixture of two enantiomers) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 13.57 (s, 1 H), 11.18 (s, 1 H), 9.50 - 9.30 (m, 1 H), 8.40 - 8.35 (m, 2H), 7.88- 7.86 (m ,2H), 7.55 - 7.51 (m, 1 H), 7.29 -7.27 (m, 2H),5.85 - 5.45 (m, 2H),3.82 - 3.47 (m, 4H), 2.68 -2.66 (m, 2H), 2.52 - 2.50 (m, 3H),2.33 - 2.32 (m, 1 H), 1.32 (d, J = 6.24 Hz, 6H). LCMS (ES, m/z): 577 [M+H]+-114.
Compound 42 5-chloro-2-fluoro-N-(4-(4-(((3R,4S)-4-fluoropyrrolidin-3-yl)oxy)-3-methyl- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
This compound was prepared according to the procedure described in example 29. The desired product was obtained as a white TFA salt (40.2 mg, 99.5% purity). LCMS (ESI, m/z): 521 [M-TFA+H]+. 1H N MR (300 MHz, DMSO-d6) δ 13.63 (s, 1 H), 11.21 (s, 1 H), 9.57 (d, J = 112.0 Hz, 2H), 8.41-8.32 (m, 2H), 7.92-7.77 (m, 2H), 7.53 (t, J = 9.3 Hz, 1 H), 7.33-
7.24 (m, 2H), 5.97 (dtd, J = 20.4, 7.9, 3.7 Hz, 1 H), 5.82-5.56 (m, 1 H), 3.98 (s, 2H), 3.76 (s, 2H), 3.68 (s, 1 H), 3.43 (t, J = 10.1 Hz, 1 H), 2.53 (s, 3H).
Compound 43 5-chloro-2-fluoro-N-(4-(4-(((3S,4R)-3-fluoropiperidin-4-yl)oxy)-3-methyl- 1H-pyrazolo[3,4-c(]pyrimidin-6-yl)-3-methylphenyl)benzenesulfonamide
This compound was prepared according to the procedure described in example 29. The desired product was obtained as a white TFA salt (30.8 mg, 23%).1H-NMR (CD3OD, 300 MHz) δ (ppm): 7.87-7.81 (m, 2H), 7.65-7.60 (m, 1 H), 7.33-7.27 (m, 1 H), 7.14-7.07 (m, 2H), 5.84-5.71 (m, 1 H), 5.43-5.27 (m, 1 H), 3.81-3.73 (m, 1 H), 3.62-3.46 (m, 2H), 3.31-3.39 (m, 1 H), 2.59 (s, 6H), 2.39 (m, 2H). LCMS (ES. m/z): 549 [M+H]+.
Compound 44 (R)-5-chloro-N-(4-(4-((3,3-difluoropiperidin-4-yl)oxy)-3-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)-2-fluorobenzenesulfonamide This compound was prepared according to the procedure described in example 29. The desired product was obtained as a white TFA salt (4.8 mg, 5 %). 1H-NMR (CD3OD, 400 MHz) δ (ppm): 8.33 (d, J = 8.7 Hz, 2H), 7.89-7.87 (m, 1 H), 7.61-7.57 (m, 1 H), 7.29-7.22 (m, 3H), 5.99-5.96 (m, 1 H), 3.28-2.85 (m, 4H), 2.59 (s, 3H), 2.39-2.01 (m, 2H). LCMS (ES. m/z): 553 [M+H]+.
Compound 45 N-(4-(4-((2-(dimethylamino)ethyl)amino)-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)-2,5-difluorobenzenesulfonamide This compound was prepared according to the procedure described in example 9. The desired product was obtained as a white TFA salt (21.5 mg, 22%).1H NMR (400 MHz, DMSO-d6): δ 13.15 (s, 1 H), 11.1 (s, 1 H), 9.41-9.25 (m, 1 H), 8.26 (d, J=8.60 Hz, 2H), 7.66- 7.72 (m, 1 H), 7.62-7.55 (m, 1 H), 7.54-7.47 (m, 1 H), 7.22 (d, J = 8.60 Hz, 2H), 3.42 - 3.32 (m, 2H), 3.42-3.32 (m, 2H), 2.54 (s, 3H), 1.10 (s, 6H). LCMS (ES. m/z): 488.5 [M+H]+. Compound 46 5-chloro-2-fluoro-N-(4-(4-(((3R,4S)-3-fluoropiperidin-4-yl)oxy)-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide A solution of 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidine (200 mg, 0.984 mmol) and tert-butyl (3R,4S)-3-fluoro-4-hydroxypiperidine-1 -carboxylate (218 mg, 0.984 mmol) in anhydrous DMF (2 mL) was added NaH (60 mg, 1.476 mmol) in 0 °C, and stirred for 2 hours at room temperature. The organic layer was worked up with H2O (10 mL), extracted with DCM (15 mL), dried over anhydrous Na2SO4, and evaporated in vacuo to provide the crude of tert-butyl (3R,4S)-4-((6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl)oxy)-3- fluoropiperidine-1 -carboxylate (160 mg, 85% purity, white solid). LCMS (ESI, m/z): 386 [M+H]+.
(i) tert-butyl (3R,4S)-4-((6-(4-((5-chloro-2-fluorophenyl)sulfonamido)phenyl)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl)oxy)-3-fluoropiperidine-1-carboxylate
To the solution of tert-butyl (3R,4S)-4-((6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl)oxy)-3-fluoropiperidine-1-carboxylate (160 mg, 0.43 mmol, 1.0 equiv.), Pd(dppf)CI2 (63.4 mg, 0.09 mmol, 0.2 equiv.), and Cs2CO3 (212 mg, 0.65 mmol, 1.5 equiv.) in 1.4- dioxane (12 mL) and H2O (3 mL) was added 5-chloro-2-fluoro-N-(4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)phenyl)benzenesulfonamide (196 mg, 0.48 mmol, 1.1 equiv.) under an atmosphere of nitrogen at room temperature. The reaction mixture was purged with nitrogen 3 times and stirred at 100 °C for 16 hours under nitrogen. The reaction was cooled to the room temperature and neutralized with aqueous 1 N HCI to pH 7~8 and washed by DCM (3*20 mL). The combined organic phase was dried over Na2SO4 and concentrated under reduced pressure to provide the crude. Then the crude purified by reversed-phase chromatography (C18 column; mobile phase, MeCN in water, 10% to 36% gradient in 18 min; detector, UV 254 nm) to provide the desired product (130 mg, 48% purity, light brown solid). LCMS (ESI, m/z): 635 [M+H]+ (ii) 5-chloro-2-fluoro-N-(4-(4-(((3R,4S)-3-fluoropiperidin-4-yl)oxy)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide trifluoroacetic acid
A solution of tert-butyl (3R,4S)-4-((6-(4-((5-chloro-2-fluorophenyl)sulfonamido)phenyl)-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl)oxy)-3-fluoropiperidine-1-carboxylate (130 mg, 0.204 mmol) in ACN (2 mL) was added CF3COOH (2 mL) in 25 °C, and stirred for 10 hours at room temperature. The mixture was concentrated under reduced pressure to provide the crude. Then the crude purified by reversed-phase chromatography (Column: XBridge Shield RP18 OBD Column, 19*150 mm, 5pm; Mobile Phase A: Water(0.05%TFA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 20% B to 50% B in 9 min, 50% B; Wave Length: 254 nm; RT1(min): 7.38; Number Of Runs: 0) to provide 5-chloro-2-fluoro-N-(4-(4-(((3R,4S)-3-fluoropiperidin-4-yl)oxy)-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl)benzenesulfonamide trifluoroacetic acid (55.8 mg, 99.5% purity, white solid). LCMS (ESI, m/z): 535 [M-TFA+H]+. 1H NMR (300 MHz, DMSO-d6) δ 13.56 (s, 1 H), 9.11 (s, 1 H), 8.44 - 8.31 (m, 2H), 7.93 - 7.72 (m, 2H), 7.53 (t, J = 9.3 Hz, 1 H), 7.32 - 7.23 (m, 2H), 6.07 - 5.71 (m, 1 H), 5.38 (d, J = 48.2 Hz, 1 H), 3.71 - 3.47 (m, 2H), 3.35 - 3.23 (m, 4H), 2.58(s, 3H), 2.26 (td, J = 15.9, 15.0, 4.8 Hz, 2H).
Compound 47 5-chloro-2-fluoro-N-(4-(4-(((3S,4S)-3-fluoropiperidin-4-yl)oxy)-3-methyl-
1 H-pyrazolo[3,4-d]pyrimidin-6-yl) phenyl)benzenesulfonamide
This compound was prepared according to the procedure described in example 46. The desired product was obtained as a light pink TFA salt (66.5 mg, 95.8% purity). LCMS (ESI, m/z): 535 [M-TFA+H]+. 1H NMR (300 MHz, DMSO-d6) δ 13.56 (s, 1 H), 8.39 - 8.28 (m, 2H), 7.89 (dd, J = 6.0, 2.7 Hz, 1 H), 7.80 (ddd, J = 8.8, 4.2, 2.7 Hz, 1 H), 7.53 (t, J = 9.3 Hz, 1 H), 7.35 - 7.23 (m, 2H), 5.95 - 5.82 (m, 1 H), 5.30 - 5.02 (m, 1 H), 3.69 - 3.49 (m, 2H), 3.27 (d, J = 11.7 Hz, 4H), 2.55 (s, 3H), 2.39 (s, 1 H), 2.17 - 2.06 (m, 1 H). Compound 48 5-chloro-2-fluoro-N-(4-{3-methyl-4-[(3S)-pyrrolidin-3-yloxy] -1 H- pyrazolo[3,4-d]pyrimidin-6-yl}phenyl)benzenesulfonamide
This compound was prepared according to the procedure described in example 47. The desired product was obtained as a white TFA salt (19.5 mg, Y= 21%). LCMS (ES. m/z): 503 [M+H] +.1H-NMR (CD3OD, 300 MHz) δ (ppm):8.37 (d, J = 8.7 Hz, 2H), 7.88-7.86 (m, 1 H), 7.64-7.59 (m, 1 H), 7.32-7.25 (m, 3H), 6.11 (d, J = 3.0 Hz,1 H), 3.83-3.71 (m, 2H), 3.59- 3.55 (m, 2H), 2.59-2.49 (m, 5H).
Compound 49 5-chloro-N-(4-{4-[(4,4-difluoropyrrolidin-3-yl)oxy]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl}phenyl) -2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 47. The desired product was obtained as an off-white TFA salt (12.2 mg, 98.4% purity). LCMS (ESI, m/z): 539 [M-TFA+H]+. 1H N MR (300 MHz, DMSO-d6) δ 13.67 (s, 1 H), 11.17 (s, 1 H), 10.08 (s, 1 H), 8.47 - 8.31 (m, 2H), 7.98 - 7.73 (m, 2H), 7.53 (t, J = 9.3 Hz, 1 H), 7.29 (dd, J = 9.8, 3.0 Hz, 2H), 6.34 - 6.07 (m, 1 H), 4.15 - 3.87 (m, 3H), 3.78 (dt, J = 13.5, 2.8 Hz, 1 H), 2.56 (s, 3H).
Compound 50 5-chloro-2-fluoro-N-[4-(4-{[(3S,4R)-3-fluoro-1-(2-hydroxyethyl)piperidin-4- yl]oxy}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide (i) 2-[(3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-3- fluoropiperidin-1-yl]ethanol
A solution of (3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-3- fluoropiperidine (150 mg, 0.499 mmol, 1.0 equiv.) and 2-bromoethanol (311 mg, 2.495 mmol, 5.0 equiv.) in anhydrous DMF (2 mL) was added DIEA (257 mg, 1.996 mmol, 4.0 equiv.), and stirred for 2 hours at room temperature. The organic layer was worked up with H2O (20 mL), extracted with DCM (20 mL), dried over anhydrous Na2SO4, and evaporated in vacuo to provide the crude. The residue was purified by reverse flash chromatography with the following conditions: column, silica gel; mobile phase, MeCN in water, 10% to 35% gradient in 18 min; detector, UV 254 nm and the purified product (120 mg, 93% purity, white solid) was obtained. LCMS (ESI, m/z): 330 [M+H]+.
(ii) 5-chloro-2-fluoro-N-[4-(4-{[(3S,4R)-3-fluoro-1-(2-hydroxyethyl)piperidin-4-yl]oxy}-
3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
To the solution of 2-[(3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)- 3-fluoropiperidin-1-yl]ethanol (120 mg, 0.338 mmol, 1.0 equiv.), Pd(dppf)CI2 (49.5 mg, 0.068 mmol, 0.2 eq), and Cs2CO3 (165 mg, 0.51 mmol, 1.5 eq) in 1.4-dioxane (8 mL) and H2O (2 mL) was added 5-chloro-2-fluoro-N-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl)benzenesulfonamide (170 mg, 0.37 mmol, 1.1 eq) under an atmosphere of nitrogen at room temperature. The reaction mixture was purged with nitrogen 3 times and stirred at 100 °C for 16 hours under nitrogen. The reaction was cooled to the room temperature and neutralized with saturated aqueous 1 N HCI to pH 7~8 and washed by DCM (3*20 mL). The combined organic phase was dried over Na2SO4 and concentrated under reduced pressure to provide the crude. Then the crude purified by reversed-phase chromatography (Column: Xselect CSH OBD Column 30*150mm 5um, n; Mobile Phase A: Water(0.05%TFA ), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 18% B to 28% B in 13 min, 28% B; Wave Length: 254 nm; RT1 (min): 11.52; Number Of Runs: 0) to provide 5-chloro-2-fluoro-N-[4-(4-{[(3S,4R)-3-fluoro-1-(2-hydroxyethyl)piperidin-4-yl]oxy}- 3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide; trifluoroacetic acid (34.6 mg, 99.5% purity, yellow solid). LCMS (ESI, m/z): 579 [M-TFA+H]+. 1H NMR (300 MHz, Methanol-d4) δ 8.46 - 8.34 (m, 2H), 7.91 (dd, J = 6.1 , 2.7 Hz, 1 H), 7.65 (ddd, J = 8.9, 4.2, 2.7 Hz, 1 H), 7.41 - 7.26 (m, 3H), 5.97 (dt, J = 27.9, 7.9 Hz, 1 H), 5.50 (d, J = 48.0 Hz, 1 H), 4.22 - 3.40 (m, 8H), 2.65 (s, 3H), 2.54 (d, J = 7.7 Hz, 2H).
Compound 51 5-chloro-2-fluoro-N-[4-(4-{[1-(2-hydroxyethyl)piperidin-4-yl]oxy}-3-methyl-
1 H-pyrazolo[3,4-d]pyrimidin -6-yl)phenyl]benzenesulfonamide
(i) tert-butoxy[4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)piperidin-1- yl]methanol
A solution of 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidine (300 mg, 1.404 mmol, 1.0 eq) and tert-butyl 4-hydroxypiperidine-1-carboxylate (339 mg, 1.685 mmol, 1.2 equiv.) in anhydrous DMF (2 mL) was added NaH (84.22 mg, 2.106 mmol, 3.0 equiv.) in 0 °C, and stirred for 2 hours at room temperature. The organic layer was worked up with H2O (10 mL), extracted with DCM (15 mL), dried over anhydrous Na2SO4, and evaporated in vacuo to provide the crude of tert-butoxy[4-({6-chloro-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-4-yl}oxy)piperidin-1-yl]methanol (230 mg, 75% purity, orange oil). LCMS (ESI, m/z): 368 [M+H]+.
(ii) 4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)piperidine A solution of tert-butyl 4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl}oxy)piperidine-1-carboxylate (230 mg, 0.470 mmol, 1.0 eq) in ACN (3 mL) was added CF3COOH(3 mL), and stirred for 16 hours at room temperature. The mixture was concentrated under reduced pressure to provide the crude. Then the crude purified by reversed-phase chromatography with the following conditions: C18 column; mobile phase, MeCN in water, 10% to 35% gradient in 15 min; detector, UV 254 nm to provide the desired product (150 mg, 90 % purity, white solid). LCMS (ESI, m/z): 268 [M+H]+.
(iii) 2-[4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)piperidin-1-yl]ethanol
A solution of 4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)piperidine (150 mg, 0.504 mmol, 1.0 equiv.) and 2-bromoethanol (315 mg, 2.520 mmol, 5.0 equiv.) in anhydrous DMF (2 mL) was added DIEA (260 mg, 2.016 mmol, 4.0 equiv.), and stirred for 16 hours at room temperature. The organic layer was worked up with H2 O (20 mL), extracted with DCM (20 mL), dried over anhydrous Na2SO4, and evaporated in vacuo to provide the crude. The residue was purified by reverse flash chromatography with the following conditions: column, silica gel; mobile phase, MeCN in water, 10% to 37% gradient in 15 min; detector, UV 254 nm and the purified product (130 mg, 94 % purity, white solid) was obtained. LCMS (ESI, m/z): 312 [M+H]+.
(iv) 5-chloro-2-fluoro-N-[4-(4-{[1-(2-hydroxyethyl)piperidin-4-yl]oxy}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin -6-yl)phenyl]benzenesulfonamide
To the solution of 2-[4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)piperidin- 1-yl]ethanol (130 mg, 0.392 mmol), Pd(dppf)CI2 (57.3mg, 0.078 mmol), and Cs2CO3 (127.7 mg, 0.392 mmol) in 1.4-dioxane (8 mL) and H2O (2 mL) was added the boronic acid (233 mg, 0.51 mmol, 1.3 eq) under an atmosphere of nitrogen at room temperature. The reaction mixture was purged with nitrogen 3 times and stirred at 100 °C for 16 hours under nitrogen. The reaction was cooled to the room temperature and neutralized with aqueous 1 N HCI to pH 7~8 and washed by DCM (3*20 mL). The combined organic phase was dried over Na2SO4 and concentrated under reduced pressure to provide the crude. Then the crude purified by reversed-phase chromatography (Column: YMC-Actus Triart C18 ExRS, 30*150 mm, 5pm; Mobile Phase A: Water (0.05%TFA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 25% B in 16 min, 25% B; Wave Length: 254 nm; RT1 (min): 12.55;) to provide the desired product (39.9 mg, 99.9% purity, white solid). LCMS (ESI, m/z): 561 [M+H]+. 1 H NMR (300 MHz, Methanol-d4) δ 8.43 - 8.32 (m, 2H), 7.89 (dd, J = 6.1 , 2.6 Hz, 1 H), 7.68 - 7.58 (m, 1 H), 7.30 (dd, J = 14.3, 8.8 Hz, 3H), 5.98 - 5.57 (m, 1 H), 3.95 (dd, J = 6.3, 4.0 Hz, 2H), 3.74 (dd, J = 43.1 , 12.8 Hz, 2H), 3.38 (t, J = 5.3 Hz, 4H), 2.70 - 2.12 (m, 7H).
Compound 525-chloro-2-fluoro-N-[4-(4-{[1-(2-hydroxyethyl)pyrrolidin-3-yl]oxy}-3-methyl-
1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
This compound was prepared according to the procedure described in example 51. The desired product was obtained as a white free base (9.1 mg, 99.3% purity, white solid). LCMS (ESI, m/z): 547 [M+H]+. 1H NMR (300 MHz, DMSO-d6) δ 13.42 (s, 1 H), 8.34 - 8.21 (m, 2H), 7.85 (dd, J = 6.0, 2.7 Hz, 1 H), 7.77 - 7.68 (m, 1 H), 7.46 (t, J = 9.3 Hz, 1 H), 7.25 - 7.15 (m, 2H), 5.73 (td, J = 6.8, 3.3 Hz, 1 H), 4.58 (s, 1 H), 3.52 (d, J = 7.0 Hz, 2H), 3.16 (dd, J = 11.3, 6.3 Hz, 1 H), 2.98 - 2.85 (m, 2H), 2.64 (t, J = 6.1 Hz, 3H), 2.47 - 2.35 (m, 4H), 2.09 - 1.92 (m, 1 H).
Compound 53 5-chloro-2-fluoro-N-[4-(4-{[(3R,4R)-3-fluoropiperidin-4-yl]oxy}-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
This compound was prepared according to the procedure described in example 47. The desired product was obtained as a white TFA Salt (16.8 mg, 54%). LCMS (ES. m/z): 535 [M+H]+. 1H-NMR (DMSO-d6, 300 MHz) δ (ppm): 13.55 (s, 1 H), 9.28 (s, 1 H), 8.33 (d, J = 8.7 Hz, 2H), 7.89-7.78(m, 2H), 7.55-7.49 (m, 1 H), 7.28 (d, J = 8.7 Hz, 2H), 5.87 (s, 1 H), 5.26-5.09 (m, 1 H), 3.70-3.27 (m, 4H), 3.62 (s, 3H), 2.27 (s, 2H), 2.14-2.08 (m, 1 H).
Compound 54 5-chloro-N-[4-(4-{[(3S,4R)-1-ethyl-3-fluoropiperidin-4-yl]oxy}-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
(i) tert-butyl (3S,4R)-4-([6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl]oxy)-3-fluoropiperidine-1 -carboxylate
Into a 50-mL round-bottom flask, was placed 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4- d]pyrimidine (300.00 mg, 1.478 mmol, 1.00 equiv.), THF (15.00 mL), tert-butyl (3S,4R)-3- fluoro-4-hydroxypiperidine-1 -carboxylate (323.98 mg, 1.478 mmol, 1.00 equiv.). The resulting solution was stirred for 30 min at room temperature. Then added NaH (177.30 mg, 4.433 mmol, 3 equiv., 60%) at 0 °C. The resulting solution was stirred for 2 hr at room temperature. The resulting mixture was concentrated. The solid was washed with ethyl ethere. The solids were collected by filtration. This resulted in 280mg (44.20%) of tert- butyl (3S,4R)-4-([6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl]oxy)-3- fluoropiperidine-1 -carboxylate as a yellow solid. LCMS(ES. m/z): 386 [M+H]+.
(ii) (3S,4R)-4-([6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl]oxy)-3- fluoropiperidine hydrochloride Into a 50-mL round-bottom flask, was placed tert-butyl (3S,4R)-4-([6-chloro-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl]oxy)-3-fluoropiperidine-1 -carboxylate (280.00 mg, 0.726 mmol, 1.00 equiv.), DCM (6.00 mL), HCI (4M in 1 ,4-dioxane) (6.00 mL, 105.105 mmol, 115.86 equiv.). The resulting solution was stirred for 2 hr at room temperature. The resulting mixture was concentrated. This resulted in 200 mg (76.99%) of the desired compound as a white solid. LCMS (ES. m/z): 286 [M+H]+.
(iii) (3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-1-ethyl-3- fluoropiperidine
A solution of (3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-3- fluoropiperidine hydrochloride (180 mg, 0.559 mmol, 1.00 equiv.), MeOH (6 mL, 148.193 mmol, 265.24 equiv.), Acetaldehyde (49.23 mg, 1.118 mmol, 2 equiv.), AcOH (33.55 mg, 0.559 mmol, 1.00 equiv.), H2O (0.5 mL, 27.754 mmol, 49.68 equiv.) was stirred for 30 min at room temperature. NaBH3CN (70.22 mg, 1.118 mmol, 2 equiv.) was added and the mixture was stirred for 3h at 60 °C. Then added NaBH3CN (70.22 mg, 1.118 mmol, 2 equiv.) was stirred for 16h at 60 °C. The mixture was allowed to cool down to room temperature. The reaction was quenched with Water. The resulting mixture was extracted with EtOAc (3 x 30 mL), dried over anhydrous Na2SO4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2 CI2 I MeOH 15:1) to afford the desired product (100 mg, 51.34%) as a yellow oil. LCMS (ES. m/z): 314 [M+H]+.
(iv) 5-chloro-N-[4-(4-{[(3S,4R)-1-ethyl-3-fluoropiperidin-4-yl]oxy}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
A solution of (3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-1-ethyl- 3-fluoropiperidine (100 mg, 0.319 mmol, 1.00 equiv.), 5-chloro-2-fluoro-N-[4-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]benzenesulfonamide (144.33 mg, 0.351 mmol, 1.1 equiv.), Pd(dppf)CI2 (46.64 mg, 0.064 mmol, 0.2 equiv.), Cs2CO3 (155.77 mg, 0.479 mmol, 1.5 equiv.)and Cs2CO3 (155.77 mg, 0.479 mmol, 1.5 equiv.) in dioxane (4 mL, 47.216 mmol, 148.15 equiv.), H2O (1 mL, 55.508 mmol, 174.16 equiv.) was stirred for 16h at 100 °C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under vacuum. The sloid was purified by flash with the following conditions (Column, C18 (40 g); mobile phase A: Water- 10 mM NH4HCO3, mobile phase B: Acetonitrile; Flow rate:40 mL/min; Gradient 60 B to 65 B; 254 nm). Then The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH OBD Column 30x150mm 5um, n; Mobile Phase A: Water (0.05%TFA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 35% B in 9 min, 35% B; Wave Length: 254 nm; RT1 (min): 7.48;) to afford the desired product (19.3 mg, 8.91%) as a white solid. LCMS (ES. m/z): 563 [M+H]+. 1H-NMR (CD3OD, 300 MHz) δ (ppm): 7.58 (d, J = 8.7 Hz, 2H), 7.09-7.06 (m, 1 H), 6.85-6.82 (m, 1 H), 6.23-6.46 (m, 3H), 4.92-5.13 (m, 1 H), 4.76-4.60 (m, 1 H), 3.21 (d, J = 9.3 Hz, 1 H), 2.26-3.02 (m, 5H), 1.79-1.64 (m, 5H), 0.66-0.61 (m, 3H).
Compound 55 5-chloro-2-fluoro-N-{4-[3-methyl-4-(piperidin-4-yloxy)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl]phenyl}benzenesulfonamide
This compound was prepared according to the procedure described in example 47. The desired product was obtained as a white TFA Salt (3.4 mg, 8.29%) . LCMS(ES. m/z): 517 [M+H]+. 1H-NMR (300 MHz, DMSO-d6) δ (ppm): 13.49 (s, 1 H), 11.16 (s, 1 H), 8.67-8.41 (m, 2H), 8.33 (d, J = 9.0 Hz, 2H), 7.82-7.78 (m, 2H), 7.56-7.49 (m, 1 H), 7.27 (d, J = 8.7 Hz, 2H), 5.74-5.71 (m, 1 H), 3.58-3.48 (m, 4H),2.59 (s, 3H), 2.13-2.25 (m, 2H), 1.96-2.05 (m, 2H).
Compound 56 5-chloro-N-[4-(4-{[(3R,4S)-1-ethyl-4-fluoropyrrolidin-3-yl]oxy}-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 47. The desired product was obtained as an off white TFA Salt (20.9 mg, 9.41 %). LCMS (ES. m/z): 549 [M+H]+. 1H-NMR (CD3OD, 300 MHz) δ (ppm): 8.40-8.37 (m, 2H), 7.88-7.85 (m, 1 H), 7.64-7.59 (m, 1 H), 7.32-7.26 (m, 3H), 6.16-6.10 (m, 1 H), 5.85-5.68 (m, 1 H), 3.65-4.23 (m, 4H), 3.48-3.41 (m, 2H), 2.61 (s, 3H), 1.44-1.39 (m, 3H).
Compound 57 5-chloro-2-fluoro-N-[4-(4-{[(3R,4S)-4-fluoro-1-isopropylpyrrolidin-3- yl]oxy}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
(i) (3R,4S)-3-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-4-fluoro-1- isopropylpyrrolidine
To a solution of (3R,4S)-3-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-4- fluoropyrrolidine hydrochloride (200 mg, 0.649 mmol, 1.00 equiv.) in MeOH (10.00 mL, 246.97 mmol, 380.54 equiv.) was added acetone (376.97 mg, 6.490 mmol, 10 equiv. )The mixture was stirred for 30 min. NaBH3CN (81.58 mg, 1.298 mmol, 2 equiv.) was added and the mixture was stirred for 16h at rt. The resulting mixture was concentrated under reduced pressure. The crude product was purified by flash with the following conditions (Column, C18 (80 g); mobile phase A: Water-1 OmM NH4HCO3, mobile phase B: Acetonitrile; Flow rate:50 mL/min; Gradient 48 B to 60 B; 254 nm) to afford the desired product (120 mg, 53.03%) as a yellow oil. LCMS (ES. m/z): 314 [M+H]+.
(ii) 5-chloro-2-fluoro-N-[4-(4-{[(3R,4S)-4-fluoro-1-isopropylpyrrolidin-3-yl]oxy}-3- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
A solution of (3R,4S)-3-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-4- fluoro-1-isopropylpyrrolidine (120 mg, 0.382 mmol, 1.00 equiv.), 5-chloro-2-fluoro-N-[4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]benzenesulfonamide (173.20 mg, 0.420 mmol, 1.1 equiv.), Pd(dppf)CI2 (55.97 mg, 0.076 mmol, 0.2 equiv.) and Cs2CO3 (186.92 mg, 0.573 mmol, 1.5 equiv.) in dioxane (2.40 mL, 28.295 mmol, 74.07 equiv.), H2O (0.60 mL, 33.265 mmol, 87.08 equiv.) was stirred for 16h at 100 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The solid was purified by flash with the following conditions (Column, C18 (80 g); mobile phase A:Water-10 mM NH4HCO3, mobile phase B: Acetonitrile; Flow rate:50 mL/min; Gradient 65 B to 72 B; 254 nm). The crude product was purified by Prep-HPLC with the following conditions (Column: Kinetex EVO C18 Column, 30x150, 5um; Mobile Phase A: Water (0.05%TFA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 40% B in 9 min, 40% B; Wave Length: 254 nm; RT1(min): 6.78; Number Of Runs: 0) to afford (26.1 mg, 10.00%) as an off-white solid. LCMS (ES. m/z): 563 [M+H]+. 1H-NMR (CD3OD, 300 MHz) δ (ppm): 8.37 (d, J = 8.7 Hz, 2H), 7.88-7.85 (m, 1H), 7.64-7.58 (m, 1 H), 7.31-7.25 (m, 3H), 6.13-6.08 (m, 1H), 5.84-5.66 (m, 1 H), 4.07-3.59 (m, 5H), 2.59 (s, 3H), 1.47-1.44 (m, 6H).
Compound 585-chloro-2-fluoro-N-[4-(4-{[(3R,4S)-4-fluoro-1-methylpyrrolidin-3-yl]oxy}-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
This compound was prepared according to the procedure described in example 57. The desired product was obtained as an off white TFA Salt (14.3 mg, Y=6.95%). LCMS (ES. m/z): 535 [M+H]+.1H-NMR (CD3OD, 300 MHz) δ (ppm): 8.38 (d, J = 8.7 Hz, 2H), 7.88-7.85 (m, 1 H), 7.65-7.59 (m, 1 H), 7.32-7.26 (m, 3H), 6.18-6.08 (m, 1 H), 5.86-5.68 (m, 1 H), 4.13- 3.52 (m, 4H), 3.07 (s, 3H), 2.61 (s, 3H).
Compound 59 5-chloro-N-[4-(4-{[(4R)-1-ethyl-3,3-difluoropiperidin-4-yl]oxy}-3-methyl- 1 H-pyrazolo [3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 57. The desired product was obtained as a white TFA Salt (29.9 mg, Y=10.92%) LCMS (ES. m/z):
581 [M+H]+. 1H-NMR (CD3OD, 300 MHz) δ (ppm): 8.38 (d, J = 8.7 Hz, 2H), 7.89-7.86 (m, 1 H), 7.64-7.59 (m, 1 H), 7.31-7.25 (m, 3H), 6.22-6.15 (m, 1 H), 3.94 (s, 2H), 3.54-3.36 (m, 4H), 2.62-2.51 (m, 5H), 1.46-1.41 (m, 3H).
Compound 60 5-chloro-N-[4-(4-{[(4R)-3,3-difluoro-1-methylpiperidin-4-yl]oxy}-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 57. The desired product was obtained as a white TFA Salt (16.3mg, 12.55%). LCMS (ES. m/z): 567 [M+H]+.1H-NMR (CD3OD, 300 MHz) δ (ppm): 8.39-8.36 (m, 2H), 7.89-7.86 (m, 1 H), 7.64-7.60 (m, 1 H), 7.31-7.25 (m, 3H), 6.19-6.12 (m, 1 H), 3.97-3.89 (m, 2H), 3.53 (s, 2H), 3.06 (s, 3H), 2.62-2.52 (m, 5H).
Compound 61 5-chloro-2-fluoro-N-[4-(4-{[(3R,4S)-4-fluoro-1-(2-hydroxyethyl)pyrrolidin- 3-yl]oxy}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
This compound was prepared according to the procedure described in example 50. The desired product was obtained as a white TFA Salt (12.7 mg, 7.22%). LCMS (ES. m/z):
565 [M+H]+. 1H-NMR (CD3OD, 400 MHz) δ (ppm): 8.38 (d, J = 8.8 Hz, 2H), 7.88-7.86 (m, 1 H), 7.64-7.60 (m, 1 H), 7.31-7.26 (m, 3H), 6.13-6.08 (m, 1 H), 5.82-5.69 (m, 1 H), 4.15-4.07 (m, 2H), 3.93-3.78 (m, 4H), 3.55-3.50 (m, 2H), 2.61 (s, 3H).
Compound 62 N-[4-(4-{[(3S,4R)-3-fluoro-1-methylpiperidin-4-yl]oxy}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-methoxypyridine-2-sulfonamide
This compound was prepared according to the procedure described in example 57. The desired product was obtained as a white solid and a TFA salt (52.2 mg, 26.58%). 1H- NMR (DMSO-d6, 300 MHz) δ (ppm): 10.92 (s, 1 H), 9.94 (s, 1 H), 8.52 (d, J = 5.7 Hz, 1 H), 8.30 (d, J = 9.0 Hz, 2H), 7.53 (d, J = 2.4 Hz, 1 H), 7.32-7.21 (m, 3H), 5.67-5.94 (m, 1 H), 5.53-5.37 (m, 1 H), 3.91 (s, 3H), 3.76-3.42 (m, 4H), 2.87 (s, 3H), 2.58 (s, 3H), 2.48-2.19
(m, 2H). LCMS (ES. m/z): 528 [M+H]+.
Compound 63 N-[4-(4-{[(3S,4R)-3-fluoro-1-methylpiperidin-4-yl]oxy}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-(trifluoromethyl)pyridine-2-sulfonamide This compound was prepared according to the procedure described in example 57. The desired product was obtained as a white solid and a TFA salt (34.1 mg, 16.5%). 1 H- NMR (DMSO-d6, 300 MHz) δ (ppm): 13.56 (s, 1 H), 11.15 (s, 1 H), 9.95 (s, 1 H), 9.03 (d, J = 4.8 Hz, 2H), 8.34-8.12 (m, 4H), 7.33 (d, J = 8.7 Hz, 1 H), 5.62-5.97 (m, 1 H), 5.53-5.29 (m, 1 H), 3.92-3.43 (m, 4H), 2.95 (s, 3H), 2.59 (s, 3H), 2.47-2.23 (m, 2H). LCMS (ES. m/z): 566 [M+H]+.
Compound 645-chloro-N-[4-(4-{[(3S,4R)-1-(2,2-difluoroethyl)-3-fluoropiperidin-4-yl]oxy}-
3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
(i) (3S,4R)-3-fluoropiperidin-4-ol hydrochloride
To a solution of tert-butyl (3S,4R)-3-fluoro-4-hydroxypiperidine-1 -carboxylate (1.00 g, 4.561 mmol, 1.00 equiv.) in DCM (15 mL, 235.951 mmol, 51.73 equiv.). HCI (4M in 1 ,4- dioxane) (15 mL, 416.667 mmol, 91.36 equiv.) was added and stirred for 2h at rt. The resulting mixture was concentrated under reduced pressure. This resulted in (3S,4R)-3- fluoropiperidin-4-ol hydrochloride (700 mg, 88.77%) as a yellow solid. LCMS (ES. m/z): 120 [M+H-HCI]+.
(ii) (3S,4R)-1-(2,2-difluoroethyl)-3-fluoropiperidin-4-ol
A solution of (3S,4R)-3-fluoropiperidin-4-ol hydrochloride (680.94 mg, 4.376 mmol, 1.4 equiv.), 1 ,1-difluoro-2-iodoethane (600 mg, 3.126 mmol, 1.00 equiv.) and NaHCO3 (787.8 mg, 9.378 mmol, 3.00 equiv.) in EtOH (20 mL, 344.271 mmol, 110.1 equiv.) was stirred for 36h at 80 °C. The solution was collected by filtration. The resulting mixture was concentrated under reduced pressure. The crude product was re-crystallized from DCM/MeOH (98%/ 2%) to afford (3S,4F?)-1-(2,2-difluoroethyl)-3-fluoropiperidin-4-ol (30 mg, 4.72%) as a yellow oil. LCMS (ES. m/z): 184 [M+H]+.
(iii) (3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-1-(2,2- difluoroethyl)-3-fluoropiperidine
To a solution of (3S,4R)-1-(2,2-difluoroethyl)-3-fluoropiperidin-4-ol (30.0 mg, 0.164 mmol, 1.00 equiv.) in THF (4.00 mL, 51.7 mmol, 315.6 equiv.) was added NaH (26.20 mg, 0.66 mmol, 4.00 equiv.) at 0 °C. The mixture was stirred for 30 min. 4,6-dichloro-3-methyl-1 H- pyrazolo[3,4-d]pyrimidine (36.58 mg, 0.180 mmol, 1.10 equiv.) was added and the mixture was allowed to warm to RT and stirred for 16h at rt. The reaction was quenched with Water. The resulting mixture was extracted with EtOAc (3 x 30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The solid was purified by flash Prep-HPLC with the following conditions (Column, C18 spherical 20- 35 um 100A (40 g); mobile phase A: Water-10 mM NH4HCO3, mobile phase B: Acetonitrile; Flow rate:35 mL/min; Gradient: 56 B to 67 B; 254 nm) to afford (3S,4R)-4-({6-chloro-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-1-(2,2-difluoroethyl)-3-fluoropiperidine (20 mg, 31.42%) as a white solid. LCMS(ES. m/z): 350 [M+H]+. (iv) 5-chloro-N-[4-(4-{[(3S,4R)-1-(2,2-difluoroethyl)-3-fluoropiperidin-4-yl]oxy}-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
A mixture of (3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-1- (2,2-difluoroethyl)-3-fluoropiperidine (30 mg, 0.086 mmol, 1.00 equiv.) , 5-chloro-2-fluoro- N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]benzenesulfonamide (38.84 mg, 0.095 mmol, 1.10 equiv.) , Pd(dppf)CI2 (12.55 mg, 0.017 mmol, 0.2 equiv.) and Cs2CO3 (41.92 mg, 0.129 mmol, 1.5 equiv.) in 1 ,4-dioxane (3.00 mL, 34.05 mmol, 397.0 equiv.) , H2O (0.75 mL, 41.63 mmol, 485.34 equiv.) was stirred for 16h at 100 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The solid was purified by flash Prep-HPLC with the following conditions (Column, C18 spherical 20- 35 cum 100A (40 g); mobile phase A: Water-10 mM NH4HCO3, mobile phase B: Acetonitrile; Flow rate:35 mL/min; Gradient: 60 B to 68 B; 254 nm). The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30x150 mm, 5pm; Mobile Phase A: Water(0.05%TFA ), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 34% B to 55% B in 7 min, 55% B; Wave Length: 254 nm; RT1(min): 6.58;) to afford 5-chloro-N-[4-(4-{[(3S,4R)-1-(2,2-difluoroethyl)-3-fluoropiperidin-4-yl]oxy}-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide; trifluoroacetic acid (8.6 mg, 13.71%) as an off-white solid.1H-NMR (DMSO-d6, 300 MHz) δ (ppm): 11.14 (s, 1 H), 8.31 (d, J = 8.4 Hz, 2H), 7.89-7.77 (m, 2H), 7.46-7.58 (m, 1 H), 7.27 (d, J = 8.7 Hz, 2H), 6.03-6.49 (m, 1 H), 5.83-5.75 (m, 1 H), 5.14-4.92(m, 1 H), 3.25-2.73 (m, 6H), 2.59 (s, 3H), 2.12-2.05 (m, 2H). LCMS (ES. m/z): 599 [M+H]+.
Compound 65 5-chloro-2-fluoro-N-(4-(4-(((3S,4R)-3-fluoro-1-(methyl-d3)piperidin-4- yl)oxy)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
(i) 6-chloro-4-(((3S,4R)-3-fluoro-1 -(methyl-c/3)piperidin-4-yl)oxy)-3-methyl-1 /7-pyrazolo[3,4- cf]pyrimidine
A solution of and (3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl}oxy)-3-fluoropiperidine hydrochloride (300 mg, 0.931 mmol, 1.00 equiv.), CD3I (134.98 mg, 0.931 mmol, 1 equiv.), DIEA (361 mg, 2.79 mmol, 3.00 equiv.) in DMF (8.00 mL, 103.4 mmol, 111.0 equiv.) was stirred for 16h at room temperature. The resulting mixture was extracted with EtOAc (3 x 40 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The solid was purified by flash Prep-HPLC with the following conditions (Column, C18 spherical 20-35 um 100A (120 g); mobile phaseA: Water-10 mM NH4HCO3, mobile phaseB:Acetonitrile; Flow rate:50 mL/min; Gradient: 49 B to 68 B; 254 nm) to afford 6-chloro-4-(((3S,4R)-3-fluoro-1-(methyl- d3)piperidin-4-yl)oxy)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidine (120 mg, 38.31%) as a yellow solid. LCMS(ES. m/z): 303 [M+H]+.
(ii) 5-chloro-2-fluoro-N-(4-(4-(((3S,4R)-3-fluoro-1-(methyl-d3)piperidin-4-yl)oxy)-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)benzenesulfonamide
A mixture of (3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-3- fluoro-1-(2H3)methylpiperidine (120 mg, 0.396 mmol, 1.00 equiv.) , 5-chloro-2-fluoro-N- [4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]benzenesulfonamide (179.5 mg, 0.436 mmol, 1.10 equiv.) , Pd(dppf)CI2 (58.00 mg, 0.079 mmol, 0.2 equiv.) and Cs2CO3 (193.7 mg, 0.59 mmol, 1.50 equiv.) in 1 ,4-dioxane (4 mL ) and water (1 ml) was stirred for 16h at 100 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The solid was purified by flash Prep-HPLC with the following conditions (Column, C18 spherical 20-35 um 100A (80 g); mobile phase A: Water-10 mM NH4HCO3, mobile phase B: Acetonitrile; Flow rate:40 mL/min; Gradient: 40 B to 51 B; 254 nm). The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30x150 mm, 5pm; Mobile Phase A: Water(0.05%TFA ), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 23% B to 43% B in 7 min, 43% B; Wave Length:
254 nm; RT1(min): 4.93) to afford 5-chloro-2-fluoro-N-[4-(4-{[(3S,4R)-3-fluoro-1- (2H3)methylpiperidin-4-yl]oxy}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl]benzenesulfonamide; trifluoroacetic acid (21.4 mg, 7.79%) as an off-white solid.1H-NMR (DMSO-d6, 300 MHz) δ (ppm): 11.18 (s, 1 H), 10.02 (s, 1 H), 8.35 (d, J = 8.7 Hz, 2H), 7.95-7.78 (m, 2H), 7.56-7.50 (m, 1 H), 7.28 (d, J = 8.7 Hz, 2H), 5.82-5.70 (m, 1 H), 5.53-5.28 (m, 1 H), 3.92-3.43 (m, 4H), 2.59 (s, 3H), 2.45-2.19 (m, 2H).LCMS(ES. m/z): 552 [M+H]+.
Compound 66 N-[4-(4-{[(3S,4R)-3-fluoro-1-methylpiperidin-4-yl]oxy}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]pyridine-2-sulfonamide
This compound was prepared according to the procedure described in example 57. The desired product was obtained as a white solid and a free base (4.6 mg, 9.10%). 1H NMR (300 MHz, DMSO-d6) δ 13.54 (s, 1 H), 10.95 (s, 1 H), 9.96 (s, 1 H), 8.72 (dt, J = 4.7, 1 .4 Hz, 1 H), 8.33 - 8.22 (m, 2H), 8.20 - 7.96 (m, 2H), 7.67 (ddd, J = 7.2, 4.7, 1.6 Hz, 1 H), 7.46 - 7.28 (m, 3H), 7.04 (d, J = 51.1 Hz, 1 H), 5.81 (s, 1 H), 5.40 (d, J = 48.0 Hz, 1 H), 3.51 (s, 3H), 2.84 (s, 3H), 2.52 (s, 3H), 2.36 - 2.15 (m, 2H). LCMS (ESI, m/z): 498[M+H]+, Compound 67 N-[4-(4-{[(3S,4R)-3-fluoro-1-methylpiperidin-4-yl]oxy}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]pyridine-2-sulfonamide
Figure imgf000223_0001
This compound was prepared according to the procedure described in example 57. The desired product was obtained as a white solid and a TFA salt (4.6 mg, 9.10%). 1H NMR (300 MHz, DMSO-d6) δ 13.54 (s, 1 H), 10.95 (s, 1 H), 9.96 (s, 1 H), 8.72 (dt, J = 4.7, 1 .4 Hz, 1 H), 8.33 - 8.22 (m, 2H), 8.20 - 7.96 (m, 2H), 7.67 (ddd, J = 7.2, 4.7, 1.6 Hz, 1 H), 7.46 - 7.28 (m, 3H), 7.04 (d, J = 51.1 Hz, 1 H), 5.81 (s, 1 H), 5.40 (d, J = 48.0 Hz, 1 H), 3.51 (s, 3H), 2.84 (s, 3H), 2.52 (s, 3H), 2.36 - 2.15 (m, 2H). LCMS (ESI, m/z): 498[M+H]+. Compound 67 N-[4-(4-{[(3R,4S)-4-fluoro-1-methylpyrrolidin-3-yl]oxy}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-(trifluoromethyl)pyridine-2-sulfonamide
Figure imgf000223_0002
This compound was prepared according to the procedure described in example 57. The desired product was obtained as a white solid and a TFA salt (47.7 mg, 20.44%). 1H-NMR (CD3OD, 300 MHz) δ (ppm): 8.92 (d, J = 4.8 Hz, 1 H), 8.37 (d, J = 8.7 Hz, 2H), 8.23 (s, 1 H), 7.90 (d, J = 4.8 Hz, 1 H), 7.34 (d, J = 8.7 Hz, 2H), 6.17-6.09 (m, 1 H), 5.85-5.68 (m, 1 H), 4.11-3.97 (m, 4H), 3.07 (s, 3H), 2.60 (s, 3H). LCMS (ES. m/z): 552 [M+H]+. Compound 68 5-chloro-2-fluoro-N-[4-(4-{[(3S,4R)-3-fluoro-1-methylpiperidin-4- yl]oxy}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)-3-methylphenyl]benzenesulfonamide
This compound was prepared according to the procedure described in example 57. The desired product was obtained as a white solid and a TFA salt (24.4 mg, 10.78%). 1H- NMR (CD3OD, 300 MHz) δ (ppm): 7.87-7.81 (m, 2H), 7.66-7.61 (m, 1 H), 7.33-7.27 (m, 1 H), 7.13-7.07 (m, 2H), 5.85-5.64 (m, 1 H), 5.48-5.22 (m, 1 H), 3.67 (s, 1 H), 3.69-3.48 (m, 2H), 3.42-3.35 (m, 1 H), 2.96 (s, 3H), 2.60 (s, 6H), 2.53-2.38 (m, 2H). LCMS (ES. m/z): 563 [M+H]+. Compound 69 5-chloro-2-fluoro-N-[2-fluoro-4-(4-{[(3S,4R)-3-fluoro-1-methylpiperidin-4- yl]oxy}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
This compound was prepared according to the procedure described in example 57.
The desired product was obtained as an off-white solid and a TFA salt (23.5 mg, 17.17%). 1H-NMR (CD3OD, 300 MHz) δ (ppm) : 8.26(d, J=8.4Hz, 1h), 8.16-8.12(dd,
J=1.8&11.7Hz, 1 H), 7.81-7.78 (dd, J=2.7&6Hz, 1 H), 7.67-7.62 (m, 1 H), 7.59-7.54 (t,
J=8.1 Hz, 1 H), 7.35-7.29 (t, J=9.3Hz, 1 H), 5.95-5.78(m, 1 H), 5.43(d, J=47.7Hz, 1 H), 3.77 (s, 1 H), 3.79-3.59 (m, 2H), 3.49-3.46 (m, 1 H), 3.00 (s, 3H), 2.65 (s, 3H), 2.53-2.42 (m, 2H).
LCMS (ES. m/z): 567 [M+H]+.
Compound 70 N-[4-(4-{[(3R)-4,4-difluoro-1-methylpyrrolidin-3-yl]oxy}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-(trifluoromethyl)pyridine-2-sulfonamide
This compound was prepared according to the procedure described in example 57.
The desired product was obtained as a white solid and a TFA salt (6.3 mg, 5.49%). 1H NMR (CD3OD ,400 MHz) δ 8.91 (d, J = 4.8 Hz, 1 H), 8.37 (d, J = 8.4 Hz, 2H), 8.23 (s, 1 H), 7.90 - 7.89 (m, 1 H), 7.33 - 7.31 (m, 2H), 6.19 (d, J = 10.4 Hz, 1 H), 4.12(m, 1 H), 3.87 - 3.84 (m, 3H), 2.94 (s, 3H), 2.63 (s, 3H). LCMS (ESI, m/z): 570[M+H]+.
Compound 71 5-chloro-N-[4-(4-{[(4R)-3,3-difluoro-1-(2-hydroxyethyl)piperidin-4-yl]oxy}-
3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 51. The desired product was obtained as a white solid and a TFA salt (18.7 mg, 21.5%) as a white solid.1H-NMR (DMSO-d6, 300 MHz) δ (ppm): 8.34 (d, J = 8.4 Hz, 2H), 7.92-7.74 (m, 2H), 7.51-7.40 (m, 1 H), 7.23 (d, J = 8.7 Hz, 2H), 5.98-5.95 (m, 1 H), 4.85-4.81 (m, 1 H), 4.40-4.27 (m, 2H), 3.84-3.71 (m, 2H), 3.24-2.77 (m, 4H), 2.68 (s, 3H), 2.23-2.19 (m, 1 H),
1.95-1.79 (m, 1 H). LCMS (ES. m/z): 597 [M+H]+.
Compound 72 N-{4-[4-({2-[(2-hydroxyethyl)(methyl)amino]ethyl}amino)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}-4-(trifluoromethyl)pyridine-2-sulfonamide This compound was prepared according to the procedure described in example 51. The desired product was obtained as an off-white solid and a TFA salt (34.5 mg, 17.58%).1H-NMR (DMSO-d6, 300 MHz) δ (ppm): 12.95 (s, 1 H), 9.01 (d, J = 4.8 Hz, 1 H), 8.23 (d, J = 8.7 Hz, 3H), 8.07 (d, J = 4.8 Hz, 1 H), 7.23 (d, J = 8.7 Hz, 2H), 7.03-6.99 (m, 1 H), 4.50 (s, 1 H), 3.74-3.71 (m, 2H), 3.57-3.49 (m, 2H), 2.76-2.72 (m, 2H), 2.68-2.59 (m, 2H), 2.48 (s, 3H), 2.39 (s, 3H).LCMS (ES. m/z): 551 [M+H]+.
Compound 73 N-[4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]-4-isopropoxypyridine-2-sulfonamide
(i) 4-isopropoxy-2-methylpyridine
To a stirred mixture of2-chloropyridin-4-ol (4.5 g, 34.738 mmol, 1.00 equiv.) and K2CO3 (9.60 g, 69.48 mmol, 2.00 equiv.) in DMF (90 mL, 1163 mmol, 33.5 equiv.) was added 2- iodopropane (8.86 g, 52.11 mmol, 1.50 equiv.) dropwise and stirred at 60 °C for 16 hours. The resulting mixture was extracted with EA (100 ml). The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, silica gel; mobile phase, EA in PE, 0% to 50% gradient in 30 min; detector, UV 254 nm. This resulted in 4-isopropoxy-2- methylpyridine (5 g, 95.19%) as a yellow oil. LCMS (ESI, m/z): 172[M+H]+.
(ii) 2-(benzylsulfanyl)-4-isopropoxypyridine
To a stirred mixture of 4-isopropoxy-2-methylpyridine (5.00 g, 33.1 mmol, 1.00 equiv.) and Cs2CO3 (18.98 g, 58.27 mmol, 2.00 equiv.) in DMF (100 mL) were added KF (1.69 g, 29.14 mmol, 1.00 equiv.) and benzyl mercaptan (7.24 g, 58.27 mmol, 2.00 equiv.) in portions, and stirred at 60 °C for 16 hours. The resulting mixture was extracted with EA (500 ml). The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 50% gradient in 30 min; detector, UV 254 nm. This resulted in 2-(benzylsulfanyl)-4-isopropoxypyridine as a yellow oil. LCMS (ESI, m/z): 260[M+H]+
(iii) bis(4-isopropoxypyridine-2-sulfonyl chloride
To a stirred mixture of 2-(benzylsulfanyl)-4-isopropoxypyridine (4 g) in DCM (10 mL) was added HCI (20 mL) and NaCICh (20 mL) in portions at room temperature. After stirring for 2 hours, the resulting mixture was concentrated under reduced pressure. Desired product could be detected by LCMS. The crude product was used in the next step directly without further purification. LCMS (ESI, m/z): 236[M+H]+.
(iv) 4-isopropoxy-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]pyridine-2- sulfonamide
To a stirred mixture of bis(4-isopropoxypyridine-2-sulfonyl chloride) (3.3 g, 7.001 mmol, 1.00 equiv.) in DCM (10 mL) were added 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)aniline (1.53 g, 7.00 mmol, 1.00 equiv.) and pyridine (0.55 g, 7.00 mmol, 1.00 equiv.) in portions at room temperature, and stirred for 4 hours. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 90% gradient in 30 min; detector, UV 254 nm. This resulted in 4- isopropoxy-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]pyridine-2- sulfonamide (4.2 g, 86.05%) as an off-white solid. LCMS (ESI, m/z): 419[M+H]+.
(v) N-[4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl]-4-isopropoxypyridine-2-sulfonamide
To a solution of 4-isopropoxy-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl]pyridine-2-sulfonamide (180.6 mg, 0.43 mmol, 1.10 equiv.) and 6-chloro-N-[2- (dimethylamino)ethyl]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-amine (100 mg, 0.393 mmol, 1.00 equiv.) in 1 ,4-dioxane (4 mL, 1.179 mmol) and H2 O (1 mL) were added Cs2CO3 (191.87 mg, 0.590 mmol, 1.5 equiv.) and Pd(dppf)CI2 (57.45 mg, 0.079 mmol, 0.2 equiv.) . After stirring for 16 hours at 100 °C under a nitrogen atmosphere, the resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 90% gradient in 30 min; 40ml/min, detector, UV 254 nm. The residue was purified by Prep-TLC(Column: YMC-Actus Triart C18, 30*150 mm, 5pm; Mobile Phase A: Water(10 mmol/L NH4HCO3+0.1%NH3.H2 O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 52% B in 7 min, 52% B; Wave Length: 254 nm; RT1 (min): 6.17;) to afford N-[4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]-4-isopropoxypyridine-2-sulfonamide (23.7 mg, 11.78%) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 12.94 (s, 1 H), 10.74 (s, 1 H), 8.47 (d, J = 5.7 Hz, 1 H), 8.23 - 8.20 (m, 2H), 7.43 (d, J = 2.4 Hz, 1 H), 7.23 (d, J = 8.7 Hz, 2H), 7.16 (dd, J = 5.7, 2.4 Hz, 1 H), 7.01 (t, J = 5.6 Hz, 1 H), 4.84 (p, J = 6.2 Hz, 1 H), 3.70 (q, J = 6.4 Hz, 2H), 2.56 (t, J = 6.9 Hz, 2H), 2.51-2.50(m,3H), 2.24 (s, 6H), 1.27 (d, J = 6 Hz, 6H). LCMS (ESI, m/z): 511.15[M+H]+.
Compound 74 N-[4-(4-{[3-(dirnethylamino)-2,2-difluoropropyl]amino}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-(trifluoromethyl)pyridine-2-sulfonamide
(i) N1-{6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}-2,2-difluoropropane-1 ,3- diamine
To a stirred mixture of 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidine (500 mg, 2.46 mmol, 1.00 equiv.) and DIEA (1910 mg, 14.78 mmol, 6.00 equiv.) in DCM (50 mL) was added 2,2-difluoropropane-1 ,3-diamine dihydrochloride (450.7 mg, 2.46 mmol, 1.00 equiv.) dropwise at room temperature, and stirred for 48 hours. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 50ml/min, 10% to 90% gradient in 30 min; detector, UV 254 nm. This resulted in N1-{6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}-2,2-difluoropropane-1 ,3-diamine (400 mg, 58.70%) as a brown yellow solid. LCMS (ESI, m/z): 277[M+H]+.
(ii) 6-chloro-N-[3-(dimethylamino)-2,2-difluoropropyl]-3-methyl-1 H-pyrazolo[3,4- djpyrimidin-4-amine
To a stirred mixture of N1-{6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}-2,2- difluoropropane-1 ,3-diamine; bis(trifluoroacetic acid) (288 mg, 0.571 mmol, 1.00 equiv.) and DIEA (368.7 mg, 2.85 mmol, 5.00 equiv.) in DMF (30 mL) was added CH3 I (161.98 mg, 1.14 mmol, 2.00 equiv.) dropwise at room temperature, and stirred for 16 hours. The aqueous layer was extracted with EA (300 ml). The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 50m l/m in, 10% to 90% gradient in 30 min; detector, UV 254 nm. This resulted in 6-chloro-N-[3- (dimethylamino)-2,2-difluoropropyl]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-amine (60 mg, 34.51%) as a off-white solid. LCMS (ESI, m/z): 305 [M+H]+.
(iii) N-[4-(4-{[3-(dimethylamino)-2,2-difluoropropyl]amino}-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]-4-(trifluoromethyl)pyridine-2-sulfonamide
To a solution of 6-chloro-N-[3-(dimethylamino)-2,2-difluoropropyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-amine (30 mg, 0.098 mmol, 1.00 equiv.) and N-[4-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]-4-(trifluoromethyl)pyridine-2-sulfonamide (46.37 mg, 0.108 mmol, 1.10 equiv.) in 1 ,4-dioxane (1.5 mL) and H2O (0.37 mL) were added Cs2CO3 (48.11 mg, 0.147 mmol, 1.50 equiv.) and Pd(dppf)CI2 (14.41 mg, 0.020 mmol, 0.20 equiv.) . After stirring for 16 hours at 100 °C under a nitrogen atmosphere, the resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 40ml/min, 10% to 90% gradient in 10 min; detector, UV 254 nm. The residue was purified by Prep-TLC(Column: XBridge Shield RP18 OBD Column, 30*150 mm, 5pm; Mobile Phase A: Water(10 mmol/L NH4HCO3+0.1%NH3.H2 O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 22% B to 42% B in 7 min, 42% B; Wave Length: 254 nm; RT1 (min): 5.43;) to afford N-[4-(4-{[3-(dimethylamino)-2,2-difluoropropyl]amino}- 3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-(trifluoromethyl)pyridine-2- sulfonamide (5.1 mg, 8.94%) as a white solid.1H NMR (300 MHz, DMSO-d6) δ 12.95 (s, 1 H), 8.92 (s, 1 H), 8.25-8.13 (m, 3H), 7.92 (s, 1 H), 7.22-7.08 (m, 3H), 4.29-4.17 (m, 2H), 2.82 (t, J = 13.5 Hz, 2H), 2.53 (s, 3H), 2.29 (s, 6H). LCMS (ESI, m/z): 571.10 [M+H]+.
Compound 75 5-chloro-N-[4-(4-{[3-(dimethylamino)-2,2-difluoropropyl]amino}-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 51. The desired product was obtained (4.5 mg, 8.10%) as an off-white solid. 1H NMR (300 MHz, Methanol-d4) δ 8.29 (d, J = 8.7 Hz, 2H), 7.89 (dd, J = 6.0, 2.7 Hz, 1 H), 7.65-7.60 (m, 1 H), 7.53-7.26 (m, 3H), 4.36-4.32 (m, 2H), 3.96-3.86 (m, 2H), 2.89 (s, 6H), 2.66 (s, 3H), 1.37 (s, 1 H). LCMS (ESI, m/z): 554.10[M+H-TFA]+.
Compound 76 5-chloro-2-fluoro-N-{4-[4-({2-[(2-hydroxy-2-methylpropyl)(methyl)amino] ethyl}amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}benzenesulfonamide
(i) tert-butyl N-{2-[(2-hydroxy-2-methylpropyl)(methyl)amino]ethyl}carbamate
To a stirred mixture of tert-butyl N-[2-(methylamino)ethyl]carbamate (1.50 g, 8.61 mmol, 1.00 equiv.) and K2CO3 (3.57 g, 25.83 mmol, 3.00 equiv.) in ACN (20 mL) was 2,2- dimethyloxirane (0.93 g, 12.91 mmol, 1.50 equiv.) added dropwise at room temperature, and stirred at reflux for 16 hours. The resulting mixture was filtered, the filter cake was washed with ACN (100 ml). The filtrate was concentrated under reduced pressure. Desired product could be detected by LCMS. LCMS (ESI, m/z): 247 [M+H]+.
(ii) 1-[(2-aminoethyl)(methyl)amino]-2-methylpropan-2-ol
To a stirred solution of tert-butyl N-{2-[(2-hydroxy-2- methylpropyl)(methyl)amino]ethyl}carbamate (1.40 g, 1.00 equiv.) in dioxane (10 mL) was added HCI (gas)in 1 ,4-dioxane (10 mL) dropwise at room temperature, and stirred for 6 hours. The resulting mixture was concentrated under reduced pressure. Desired product could be detected by LCMS. LCMS (ESI, m/z): 147 [M+H]+.
(iii) 1-{[2-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl]
(methyl)amino}-2-methylpropan-2-ol
To a stirred solution of 1-[(2-aminoethyl)(methyl)amino]-2-methylpropan-2-ol (150 mg, 1.03 mmol, 1.00 equiv.) and DIEA (397.7 mg, 3.08 mmol, 3.00 equiv.) in THF (2 mL) was added 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidine (208.3 mg, 1.03 mmol, 1.00 equiv.) in portions at room temperature, and stirred for 3 hours. The resulting mixture was concentrated under reduced pressure. Desired product could be detected by LCMS. LCMS (ESI, m/z): 313 [M+H]+
(iv) 5-chloro-2-fluoro-N-{4-[4-({2-[(2-hydroxy-2-methylpropyl)(methyl)amino] ethyl}amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl]phenyl}benzenesulfonamide
To a solution of 1-{[2-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl}amino)ethyl](methyl)amino}-2-methylpropan-2-ol (100 mg, 0.320 mmol, 1.00 equiv.) and 5-chloro-2-fluoro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl]benzenesulfonamide (197.4 mg, 0.48 mmol, 1.50 equiv.) in 1 ,4-dioxane (4 mL) and H2O (1.00 mL) were added Pd(dppf)CI2 (46.78 mg, 0.064 mmol, 0.20 equiv.) and Cs2CO3 (156.2 mg, 0.480 mmol, 1.50 equiv.) . After stirring for 16 hours at 100°C under a nitrogen atmosphere, the resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 90% gradient in 30 min; detector, UV 254 nm, 40ml/min. The residue was purified by Prep-TLC(Column: XSelect CSH Prep C18 OBD Column, 19*150 mm, 5pm; Mobile Phase A: Water(10 mmol/L NH4HCO3+0.1 %NH3.H2 O), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 23% B to 53% B in 7 min, 53% B; Wave Length: 254 nm; RT1 (min): 6.8;) to afford 5-chloro-2- fluoro-N-{4-[4-({2-[(2-hydroxy-2-methylpropyl)(methyl)amino]ethyl}amino)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}benzenesulfonamide (3.9 mg, 2.09%) as an off-white solid. 1H NMR (400 MHz, Methanol-d4) δ 8.28 - 8.25 (m, 2H), 7.86-7.84 (m, 1 H), 7.61- 7.57 (m, 1 H), 7.29 - 7.24 (m, 1 H), 7.24 - 7.16 (m, 2H), 3.82 (t, J = 6.4 Hz, 2H), 2.85-2.81 (m, 2H), 2.65 (s, 3H), 2.48 (s, 5H), 1.17 (s, 6H). LCMS (ESI, m/z): 562.15 [M+H]+.
Compound 77 2-fluoro-N-{4-[4-({2-[(2-hydroxyethyl)(methyl)amino]ethyl}amino)-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}-5-methoxybenzenesulfonamide
This compound was prepared according to the procedure described in example 51. The desired product was obtained (20.6 mg, 8.96%) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 10.93 (s,1 H), 9.34 (s,1 H), 8.27 (d, J=8.7Hz, 2H), 7.39 - 7.20 (m, 6H), 4.00(s,2H), 3.78-3.69 (m,5H), 3.53-3.17 (m, 4H), 2.90 (d, J = 4.8 Hz, 3H), 2.55 (s, 3H). LCMS (ESI, m/z): 530.20 [M+H]+.
Compound 78 5-chloro-2-fluoro-N-{2-fluoro-4-[4-({2-[(2- hydroxyethyl)(methyl)amino]ethyl}amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl]phenyl}benzenesulfonamide This compound was prepared according to the procedure described in example 51. The desired product was obtained (19.5 mg, 14.80%) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 12.96 (s, 1 H), 7.95 - 7.90 (m, 2H), 7.71-7.69 (m, 1 H), 7.57 (d, J = 8 Hz, 1 H), 7.35-7.21 (m, 2H), 7.09 (t, J = 5.2 Hz, 1 H), 3.86 (d, J = 5.6 Hz, 2H), 3.62 (s, 2H), 3.11 (s,
2H), 2.95 (s, 2H), 2.65 (s, 3H), 2.53 (s, 3H). LCMS (ESI, m/z): 552.05 [M+H]+.
Compound 79 5-chloro-2-fluoro-N-(2-fluoro-4-{3-methyl-4-[(1-methylpiperidin-4-yl)oxy]- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl}phenyl)benzenesulfonamide This compound was prepared according to the procedure described in example 57. The desired product was obtained (10.8 mg, 8.37%) as an off-white solid. 1H NMR (300 MHz, DMSO-d6) δ 13.37 (s, 1 H), 7.95 - 7.88 (m, 2H), 7.73-7.70 (m, 1 H), 7.58-7.53 (m, 1 H), 7.35-7.25 (m, 2H), 5.63(s,1 H), 3.12 (s, 4H), 2.69 (s, 3H), 2.53 (s, 3H), 2.20 (s, 2H), 2.07 (s, 3H). LCMS (ESI, m/z): 549.10 [M+H]+. Compound 80 N-(4-{4-[(1-isopropylpiperidin-4-yl)oxy]-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl}phenyl)-4-methoxypyridine-2-sulfonamide
Figure imgf000235_0001
This compound was prepared according to the procedure described in example 57. The desired product was obtained (12.7 mg, 7.30%) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 13.38 (s, 1H), 8.50 (d, J = 5.6 Hz, 1 H), 8.23 - 8.21 (m, 2H), 7.51 (d, J = 2.4 Hz, 1 H), 7.28 - 7.24 (m, 2H), 7.19 (dd, J = 5.6, 2.5 Hz, 1H), 5.50 (tt, J = 7.3, 3.6 Hz, 1H), 3.89 (s, 3H), 2.78-2.67 (m, 3H), 2.08-2.03 (m, 2H), 1.83 - 1.80 (m, 2H), 1.02 (d, J = 6.8 Hz, 6H). LCMS (ESI, m/z): 538.25[M+H]+.
Compound 81
Figure imgf000235_0002
(i) 2-((2-(6-chloro-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3,4- d]pyrimidin-4-ylamino)ethyl)(methyl)amino)ethanol
Into a 50-mL round-bottom flask, was placed 4,6-dichloro-3-methyl-1-(oxan-2- yl)pyrazolo[3,4-d]pyrimidine (450.00 mg, 1.57 mmol, 1.00 equiv.), 2-[(2- aminoethyl)(methyl)amino]ethanol (277.8 mg, 2.35 mmol, 1.50 equiv.), DCM (10.0 mL), TEA (317.2 mg, 3.13 mmol, 2.00 equiv.). The resulting solution was stirred for 1 overnight at room temperature. The resulting mixture was concentrated. The residue was applied onto a silica gel column with dichloromethane/methanol (100:0 to 85:15). The collected fractions were combined and concentrated. This resulted in 500 mg (77.84%) of 2-[(2-[[6- chloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- yl]amino]ethyl)(methyl)amino]ethanol as colorless oil. LCMS (ESI, m/z): 369 [M+H]+.
(ii) 4-(N-(5-chloro-2-fluorophenyl)sulfamoyl)phenylboronic acid
Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 2-[(2-[[6-chloro-3-methyl-1-(oxan-2-yl)pyrazolo[3,4- d]pyrimidin-4-yl]amino]ethyl)(methyl)amino]ethanol (300.00 mg, 0.813 mmol, 1.00 equiv.), 4-(5-chloro-2-fluorobenzenesulfonamido)phenylboronic acid (219.76 mg, 0.667 mmol, 0.82 equiv.), dioxane (8.00 mL), H2 O (2.00 mL, 0.111 mmol, 0.14 equiv.), Cs2CO3 (529.98 mg, 1.627 mmol, 2.0 equiv.), Pd(dppf)CI2 (119.02 mg, 0.163 mmol, 0.2 equiv.). The resulting solution was stirred for overnight at 90 °C in an oil bath. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated. The resulting solution was diluted with 50 mL of DCM. The resulting mixture was washed with 2 x30 ml of brine and 1 x30 mL of water. The mixture was dried over anhydrous sodium sulfate. The residue was applied onto a silica gel column with dichloromethane/methanol (100:0 to 10:90). The collected fractions were combined and concentrated. This resulted in 90 mg (16.11%) of 5-chloro-2-fluoro-N-[4-[4-([2-[(2- hydroxyethyl)(methyl)amino]ethyl]amino)-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin- 6-yl]phenyl]benzenesulfonamide as colorless oil. LCMS (ESI, m/z): 618 [M+H]+.
(iii) (2S)-2-((2-(6-(4-(5-chloro-2-fluorophenylsulfonamido)phenyl)-3-methyl-1- (tetrahydro-2H-pyran-2-yl)-1 H-pyrazolo[3,4-d]pyrimidin-4- ylamino)ethyl)(methyl)amino)ethyl 2-(tert-butoxycarbonylamino)-3- methylbutanoate
Into a 50-mL round-bottom flask, was placed 5-chloro-2-fluoro-N-[4-[4-([2-[(2- hydroxyethyl)(methyl)amino]ethyl]amino)-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin- 6-yl]phenyl]benzenesulfonamide (80.00 mg, 0.129 mmol, 1.00 equiv.), (2S)-2-[(tert- butoxycarbonyl)amino]-3-methylbutanoic acid (30.93 mg, 0.142 mmol, 1.10 equiv.), DCM (8.00 mL), DCC (53.4 mg, 0.258 mmol, 2.00 equiv.), DMAP (15.81 mg, 0.129 mmol, 1.00 equiv.). The resulting solution was stirred for 4 hr at room temperature. The resulting mixture was concentrated. The crude product (150 mg) was purified by Flash-Prep-HPLC with the following conditions (CombiFlash-1): Column, C18 silica gel; mobile phase, water (NH4HCO3 0.05%)/ACN=80:20 increasing to water (NH4HCO3 0.05%)/ACN=30:70 within 45 minutes ; Detector, 220nm. This resulted in 15 mg (12.76%) of 2-[[2-([6-[4-(5- chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4- d]pyrimidin-4-yl]amino)ethyl](methyl)amino]ethyl (2S)-2-[(tert-butoxycarbonyl)amino]-3- methylbutanoate as an off-white solid. LCMS (ESI, m/z): 817 [M+H]+.
(iv) (S)-2-((2-(6-(4-(5-chloro-2-fluorophenylsulfonamido)phenyl)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-ylamino)ethyl)(methyl)amino)ethyl 2-amino-3- methylbutanoate
Into a 25-mL round-bottom flask, was placed 2-[[2-([6-[4-(5-chloro-2- fluorobenzenesulfonamido)phenyl]-3-methyl-1-(oxan-2-yl)pyrazolo[3,4-d]pyrimidin-4- yl]amino)ethyl](methyl)amino]ethyl (2S)-2-[(tert-butoxycarbonyl)amino]-3- methylbutanoate (15.00 mg, 0.018 mmol, 1.00 equiv.), HCI(gas)in 1 ,4-dioxane (2.00 mL).IPA (2.0 ml). The resulting solution was stirred for 4 hr at room temperature. The resulting mixture was concentrated. The crude product (30 mg ) was purified with HPLC. Column: XBridge Prep C 18 OBD column, 19*250mm*5um, Mobile Phase A: Water(0.05% TFA) , Phase B: Mobile ACN; flow rate: 25ml/min; Gradient: 30% B to 50% B in 7 min; 254/220nm; Rt:4.75 min (detected by Icms and collected). 13.6 mg product was obtained as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.62 (s, 1 H), 8.57 - 8.49 (m, 4H), 7.86 (d, J = 8.5 Hz, 2H), 7.49 (s, 1 H), 7.40 - 7.15 (m, 4H), 4.72 - 4.44 (m, 2H), 4.07 (s, 2H), 3.90 (s, 2H), 3.82 - 3.30 (m, 3H), 2.98 (s, 3H), 2.59 (d, J = 1.5 Hz, 3H), 0.89 (d, J = 6.0 Hz, 6H). LCMS (ESI, m/z): 633 [M+H-CF3COOH]+.
Compound 82 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethyl 2,2-dimethylpropanoate
To a stirred mixture of 5-chloro-2-fluoro-N-{4-[4-({2-[(2-hydroxyethyl)(methyl)amino]ethyl }amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}benzenesulfonamide (100 mg, 0.187 mmol, 1 equiv.) in pivalic acid (3 mL) was added trimethylacetic anhydride (69.76 mg, 0.374 mmol, 2.0 equiv.) dropwise at 40°C, and stirred at 120°C for 6 hours. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, can in water, 10% to 50% gradient in 30 min; detector, UV 254 nm, 40ml/min. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 30*150 mm, 5pm; Mobile Phase A: Water(0.05%TFA ), Mobile Phase canACN; Flow rate: 60 mL/min; Gradient: 27% B to 47% B in 7 min, 47% B; Wave Length: 254 nm; RT1(min): 4; Number Of Runs: 0) to afford 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethyl 2,2-dimethyl propanoate (21.2 mg, 17.93%) as a white solid.1 H NMR (400 MHz, Methanol-cL) 5 8.24 - 8.21 (m, 2H), 7.91-7.89 (m, 1 H), 7.65-7.61 (m, 1 H), 7.33 - 7.27 (m, 3H), 4.33 (t, J = 4.9 Hz, 2H), 4.22 (d, J = 6.1 Hz, 2H), 3.64 (s, 4H), 3.06 (d, J = 1.6 Hz, 3H), 2.70 - 2.65 (m, 3H), 1.10 (s, 9H). LCMS (ESI, m/z): 618.15[M+H]+.
Compound 83 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethyl 3-methylbutanoate
To a stirred solution 5-chloro-2-fluoro-N-{4-[4-({2-[(2-hydroxyethyl)(methyl) amino]ethyl}amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl]phenyl}benzenesulfonamide (100 mg, 0.187 mmol, 1.00 equiv.)in DMF (3 mL) and Et3N (37.90 mg, 0.374 mmol, 2.00 equiv.). pivaloyl chloride (24.84 mg, 0.206 mmol, 1.10 equiv.) dropwise at 0 °C. The reaction was stirred for 2 hours at 0 °C. The reaction was quenched by the addition of water. The resulted mixture was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column 30*150mm, 5um; Mobile Phase A: Water(0.05% TFA), Mobile B: ACN; Flow rate: 60ml/min, Gradient: 25% B to 45% in7 min, Wave Length 254nm; RT 4.57 min) to afford 2-{[2-({6-[4-(5-chloro-2- fluorobenzenesulfonamido)phenyl]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl}amino)ethyl](methyl)amino}ethyl 3-methylbutanoate TFA salt (21.3mg) pure: 98.2% as a white solid. 1 H NMR (300 MHz, DMSO-d6) δ 11.09 (s, 1 H), 9.71 (s, 1 H), 8.42 - 8.16 (m, 2H), 8.05 - 7.68 (m, 2H), 7.52 (t, J = 9.3 Hz, 1 H), 7.39 - 7.18 (m, 3H), 4.31 (t, J = 5.1 Hz, 2H), 4.00 (s, 2H), 3.65-3.51 (m, 2H), 2.95 (s, 3H), 2.57 (s, 3H), 2.06 (d, J = 7.1 Hz, 2H), 1.87 (dt, J = 13.5, 6.7 Hz, 1 H), 0.78 (d, J = 6.6 Hz, 5H). LCMS (ESI, m/z): 618 [M+H- CF3COOH]+.
Compound 84 N-[4-(4-[[2-(dimethylamino)ethyl]amino]-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)-2-fluorophenyl]-2,5-difluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 38. The desired product as a white solid (23.6 mg, 7.84%). LCMS (ES. m/z): 522 [M-TFA+H]+.1H- NMR (CD3OD, 300 MHz) δ (ppm): 8.19-8.16 (m, 1 H), 8.10-8.06 (dd, J = 1.8 &12 Hz, 1 H), 7.81-7.78 (m, 1 H), 7.68-7.63 (m, 2H), 7.57 (t, J = 8.2Hz, 1 H), 7.35-7.29 (t, J = 9.3Hz, 1 H), 4.16-4.13 (t, J = 5.7 Hz, 2H), 3.56-3.53 (t, J = 5.7 Hz, 2H), 2.98 (s, 6H), 2.65 (s, 3H).
Compound 85 N-(2-chloro-4-(4-((2-(dimethylamino)ethyl)amino)-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)-2,5-difluorobenzenesulfonamide This compound was prepared according to the procedure described in example 38. The desired product as a white solid (4.56 mg, 46%). LCMS: (ES, m/z): [M+H]+ = 522, 1H NMR (300 MHz, DMSO-d6) δ 12.97 (s, 1 H), 9.75 (s, 1 H), 8.23 (d, J = 2.1 Hz, 1 H), 8.94 - 7.08 (m, 1 H), 7.44 - 7.53 (m, 1 H), 7.32 (q, J = 8.7, 6.8 Hz, 3H), 7.18 (t, J = 5.8 Hz, 1 H), 5.76
(s, 2H), 3.94 (d, J = 6.2 Hz, 2H), 3.17 (s, 1 H), 2.81 (d, J = 5.8 Hz, 6H), 2.55 (d, J = 7.2 Hz, 3H).
Compound 86 5-chloro-2-fluoro-N-[4-(4-{2-[(2-hydroxyethyl)(methyl)amino]ethoxy}-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide
OH
This compound was prepared according to the procedure described in example 9. The desired product as an off-white solid (516.3 mg, 30% yield). LCMS (ESI, m/z): 535 [M+H]+. 1H NMR (300 MHz, DMSO-d6) δ 13.43 (s, 1 H), 8.33 - 8.23 (m, 2H), 7.86 (dd, J = 6.1 , 2.7 Hz, 1 H), 7.75 (ddd, J = 8.8, 4.1 , 2.7 Hz, 1 H), 7.48 (t, J = 9.3 Hz, 1 H), 7.27 - 7.16 (m, 2H), 4.73 (t, J = 5.6 Hz, 2H), 4.46 (s, 1 H), 3.51 (t, J = 6.2 Hz, 2H), 3.39 (s, 3H), 2.97 (t, J = 5.5
Hz, 2H), 2.63 (t, J = 6.2 Hz, 2H), 2.49 (s, 3H),2.39 (s, 3H). Compound 87 2-[[2-([6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl]amino)ethyl](methyl)amino]ethyl acetate
This compound was prepared according to the procedure described in example 83. The desired product as a white solid (50.3 mg, 95.1% purity). LCMS (ESI, m/z): 576+H]+. 1H NMR (300 MHz, DMSO-d6) δ 12.96 (s, 1 H), 10.98 (s, 1 H), 8.26 (d, J = 8.4 Hz, 2H), 7.89 - 7.68 (m, 2H), 7.49 (t, J = 9.3 Hz, 1 H), 7.20 (d, J = 8.4 Hz, 2H), 6.98 (t, J = 5.6 Hz, 1 H), 4.09 (t, J = 5.9 Hz, 2H), 3.69 (q, J = 6.4 Hz, 2H), 2.69 (dt, J = 8.4, 4.6 Hz, 4H), 2.55 (s, 3H), 2.33 (s, 3H), 1.94 (s, 3H). Compound 88 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethoxyphosphonic acid
Into tetra hydrofuran (3 mL) was added 5-chloro-2-fluoro-N-{4-[4-({2-[(2-hydroxyethyl) (methyl)amino]ethyl}amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl]phenyl}benzenesulfonamide (200 mg, 0.375 mmol, 1.00 equiv.) at room temperature. To the above mixture was added (dichlorophosphoryl)oxyphosphonoyl dichloride (0.16 mL, 0.636 mmol, 1.70 equiv.) dropwise at -40°C. The resulting mixture was stirred for additional 1 hour at -40°C. The residue was basified to pH 8 with NaHCO3. The residue was acidified with AcOH. The residue was purified by pre-HPLC: column: XBridge Shield RP18 OBD Column, 19*250 mm, 10pm; Mobile phase A: Water(10 mmol/L NH4HCO3+0.1 %NH3.H2O), Mobile phase B: ACN; Flow rate: 25 mL/min; Gradient: 12% B to 28% B in 5 min, 28% B; Wave Length: 220 nm; RT1 (min): 4.58 to afford 2-{[2-({6-[4- (5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl}amino)ethyl](methyl)amino}ethoxyphosphonic acid (98.3 mg, 41.12%) as a white solid. LCMS (ESI, m/z): 614.1 [M+H]+.1H NMR (300 MHz, DMSO-d6) δ 13.05 (s, 1 H), 11.07 (s, 1 H), 8.30 - 8.27 (m, 2H), 7.87-7.77 (m, 2H), 7.55-7.48 (m, 1 H), 7.29-7.22 (m, 3H), 4.15- 4.12 (m, 2H), 4.00-3.98 (m, 2H), 3.47 (s,4H),2.93 (s, 3H), 2.56 (s, 3H).
Compound 89 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethyl 2-methylpropanoate trifluoroacetic acid
O
To a stirred solution of 5-chloro-2-fluoro-N-{4-[4-({2-[(2- hydroxyethyl)(methyl)amino]ethyl}amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl]phenyl}benzenesulfonamide (1.4 g, 2.622 mmol, 1 equiv.) in isobutyric acid (15 mL, 170.249 mmol, 64.94 equiv.) was added isobutyric anhydride (0.62 g, 3.933 mmol, 1.5 equiv.) in portions, and stirred at 120°C for 16 hours. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (0.05% TFA), 10% to 50% gradient in 60 min; detector, UV 254 nm, 50ml/min. This resulted in 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethyl 2- methylpropanoate trifluoroacetic acid (1.0438 g, 54.39%) as a white solid. LCMS (ESI, m/z): 604.15 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 11.11 (s, 1 H), 9.89 (s, 1 H), 8.30 - 8.28 (m, 2H), 7.87-7.85 (m, 1 H), 7.82 - 7.78 (m, 1 H), 7.54 -7.50 (m, 1 H), 7.37 (s, 1 H), 7.25 - 7.23 (m, 2H), 4.31 (t, J = 5.1 Hz, 2H), 4.01 (s, 2H), 3.58 -3.44 (m,4H), 2.96 (s, 3H), 2.57 (s, 3H), 2.48-2.39 (m, 1 H), 1.00 (d, J = 6.8 Hz, 6H).
Compound 90 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethyl propanoate; bis(trifluoroacetic acid) bis(5-chloro-2-fluoro-N-{4-[4-({2-[(2-hydroxyethyl)(methyl)amino]ethyl}amino)-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}benzenesulfonamide) (200 mg, 0.187 mmol, 1.00 equiv.) in propionic acid (3 mL) , was added propionic anhydride (97.56 mg, 2.00 equiv.) . The mixture was stirred for 4 hours at 100°C. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under vacuum. The resulting mixture was diluted with DMF (3ml). The residue was purified by reverse flash chromatography with the following conditions: column, C18 gel; mobile phase, MeCN water (TFA 0.05%), 10% to 90% gradient in 40 min; detector, UV 254 nm&220nm. This result to give desired product 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]- 3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethyl propanoate; bis(trifluoroacetic acid) (114.2 mg, 85.43%) as a white solid. LCMS (ESI, m/z): 590[M+H]+.1H NMR (400 MHz, DMSO-d6) δ 11.12 (s, 1 H), 9.83 (s, 1 H), 8.30 - 8.28 (m, 2H), 7.87 (dd, J = 6.1 , 2.7 Hz, 1 H), 7.80 (m, 1 H), 7.52 (t, J = 9.3 Hz, 1 H), 7.40 (s, 1 H), 7.28 - 7.22 (m, 2H), 4.31 (t, J = 5.1 Hz, 2H), 4.01 (m, 2H), 3.58-3.44(m, 4H), 2.95 (s, 3H),
2.57 (s, 3H), 2.22 (q, J = 7.5 Hz, 2H), 0.93 (t, J = 7.5 Hz, 3H).
Compound 91 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethyl butanoate trifluoroacetic acid
To a stirred solution of 5-chloro-2-fluoro-N-{4-[4-({2-[(2- hydroxyethyl)(methyl)amino]ethyl}amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl]phenyl}benzenesulfonamide (200 mg, 0.375 mmol, 1.00 equiv.) in butanoic acid (3 mL) was added butyric anhydride (118.50 mg, 0.750 mmol, 2.00 equiv.) dropwise at room temperature, and stirred for 6 hours at 120°C.The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water(0.05%TFA), 10% to 50% gradient in 30 min; detector, UV 254 nm. This resulted in 2-{[2-({6-[4-(5-chloro-2-fluorobenzenesulfonamido)phenyl]-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-4-yl}amino)ethyl](methyl)amino}ethyl butanoate trifluoroacetic acid (176.6 mg, 63.04%) as a white solid. LCMS (ESI, m/z): 604.20 [M+H]+.1H NMR (300 MHz, DMSO- d6) δ 11.09 (s, 1 H), 9.75 (s,1 H), 8.30 - 8.27 (m, 2H), 7.87-7.79 (m, 2H), 7.52 (t, J = 9.3 Hz, 1 H), 7.36 - 7.22 (m, 3H), 4.32 (t, J = 5.1 Hz, 2H), 3.99 (s, 2H), 2.93 - 2.85 (m, 3H), 2.73 (s, 1 H), 2.57 (s, 3H), 2.15 (t, J = 7.4 Hz, 2H), 1.48-1.35 (m, 2H), 0.78 (t, J = 7.4 Hz, 3H).
Compound 92 5-chloro-2-fluoro-N-[4-(4-{[(3S,4R)-3-fluoro-1-(2-hydroxy-2- methylpropyl)piperidin-4-yl]oxy}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl]benzenesulfonamide (i) l-[(3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-3- fluoropiperidin-1-yl]-2-methylpropan-2-ol
A solution of (3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-3- fluoropiperidine hydrochloride (200 mg, 0.621 mmol, 1.00 equiv.) ,Cs2CO3 (257.39 mg, 1.863 mmol, 3 equiv.) in DMF (10 mL) was stirred for 16h at 60°C. The solution was collected by filtration. The solid was purified by flash Prep-HPLC with the following conditions (Column, C18 spherical 20-35 urn 100A (120 g); mobile phase A:Water-10 mM NH4HCO3, mobile phase B:Acetonitrile; Flow rate:50 mL/min; Gradient 41 B to 48 B; 254 nm). This resulted in 1-[(3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl}oxy)-3-fluoropiperidin-1-yl]-2-methylpropan-2-ol (30 mg, 10.80%) as a yellow oil. LCMS(ES. m/z): 358 [M+H]+.
(ii) 5-chloro-2-fluoro-N-[4-(4-{[(3S,4R)-3-fluoro-1-(2-hydroxy-2- methylpropyl)piperidin-4-yl]oxy}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenyl]benzenesulfonamide
A solution of 1-[(3S,4R)-4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-3- fluoropiperidin-1-yl]-2-methylpropan-2-ol (30 mg, 0.084 mmol, 1 equiv.) , 5-chloro-2- fluoro-N-[4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]benzenesulfonamide (34.52 mg, 0.084 mmol, 1.00 equiv.) , Pd(dppf)CI2 (12.27 mg, 0.017 mmol, 0.20 equiv.) , Cs2CO3 (40.98 mg, 0.126 mmol, 1.50 equiv.) in 1 ,4-dioxane (3 mL), H2O (0.75 mL) was stirred for 16h at 100°C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The solid was purified by flash Prep-HPLC with the following conditions (Column, C18 spherical 20-35 um 100A (800 g); mobile phase A: Water-10 mM NH4HCO3, mobile phase B: Acetonitrile; Flow rate:40 mL/min; Gradient 50 B to 58 B; 254 nm). The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30x150 mm, 5pm; Mobile Phase A: Water(10 mmol/L NH4HCC>3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 40% B to 50% B in 9 min, 50% B; Wave Length: 254 nm; RT1(min): 5.85). This resulted in 5- chloro-2-fluoro-N-[4-(4-{[(3S,4R)-3-fluoro-1-(2-hydroxy-2-methylpropyl)piperidin-4- yl]oxy}-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]benzenesulfonamide (4.5 mg, 8.69%) as a white solid. LCMS (ES. m/z): 607 [M+H]+ . 1H-NMR (DMSO-d6, 300 MHz) δ (ppm): 8.28-8.16 (m, 2H), 7.81-7.88 (m, 1 H), 7.68-7.65 (m, 1 H), 7.43-7.38 (m, 1 H), 7.18- 7.06 (m, 2H), 5.82-5.74 (m, 1 H), 5.09-4.86 (m, 1 H), 4.81 (s, 1 H), 4.18 (s, 2H), 3.13-2.91(m, 2H), 2.75 (s, 3H), 2.39 (s, 2H), 2.08-1.91 (m, 2H), 1.16 (s, 6H).
Compound 93 5-chloro-2-fluoro-N-{4-[4-({2-[(2-hydroxyethyl)(isopropyl)amino] ethyl}amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}benzenesulfonamide
(i) N-{2-[(2-hydroxyethyl)(isopropyl)amino]ethyl}carbamate
To a stirred mixture of tert-butyl N-(2-bromoethyl)carbamate (433 mg, 1.93 mmol, 1.00 equiv.) and 2-(isopropylamino)ethanol (199.3 mg, 1.93 mmol, 1.00 equiv.) in ACN (15 mL) was added K2CO3 (534.1 mg, 3.86 mmol, 2.00 equiv.) in portions, and stirred at 60°C for 16 hours. The residue was purified by flash chromatography with the following conditions: column, silica gel; mobile phase, DCM in MeOH 0% to 10% gradient in 30 min; detector, UV 254 nm. This resulted in tert-butyl N-{2-[(2-hydroxyethyl)(isopropyl) amino]ethyl}carbamate (100 mg, 21.01 %) as an off-white solid. LCMS (ESI, m/z): 247 [M+H]+.
(ii) 2-[(2-aminoethyl)(isopropyl)amino]ethanol hydrochloride To a stirred solution of tert-butyl N-{2-[(2-hydroxyethyl)(isopropyl)amino]ethyl}carbamate (100 mg, 0.406 mmol, 1.00 equiv.) in DCM (3 mL) was added HCI (gas)in 1 ,4-dioxane (3 mL, 98.74 mmol, 243.2 equiv.) dropwise at room temperature, and stirred for 2 hours. The resulting mixture was concentrated under reduced pressure. This resulted in 2-((2- aminoethyl)(isopropyl)amino)ethan-1-ol dihydrochloride (80 mg, 100%) as an off-white solid. LCMS (ESI, m/z): 147 [M-2HCI+H]+.
(iii) 5-chloro-2-fluoro-N-{4-[4-({2-[(2-hydroxyethyl)(isopropyl)amino]ethyl}amino)-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}benzenesulfonamide
This compound was prepared according to the procedure described in example 9. The desired product as a white solid (50.3 mg, 95.1% purity (3.2 mg, 11.78%) as a white solid. LCMS (ESI, m/z): 562.10[M+H]+.1H NMR (300 MHz, DMSO-d6) δ 12.94 (s, 1 H), 8.24 (d, J = 8.3 Hz, 2H), 7.83-7.67 (m, 2H), 7.47 (t, J = 9.2 Hz, 1 H), 7.16 (d, J = 8.4 Hz, 2H), 7.02 (m, 1 H), 4.45 (s, 1 H), 3.63 (q, J = 6.7, 6.2 Hz, 2H), 3.45 (m, 2H), 3.07 - 2.96 (m, 1 H), 2.70 (t, J = 6.9 Hz, 2H), 2.55 (m, 4H), 0.96 (d, J = 6.5 Hz, 6H)
Compound 94 5-chloro-N-{4-[4-({2-[ethyl(2-hydroxyethyl)amino]ethyl}amino)-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}-2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 93. The desired product as a white solid (22.1 mg, 19.64%). LCMS (ES. m/z): 548 [M+H]+. 1H- NMR (DMSO-d6, 300 MHz) δ (ppm): 12.93 (s, 1 H), 11.11 (s, 1 H), 8.23 (d, J = 8.7 Hz, 2H), 7.84-7.70 (m, 2H), 7.49-7.43 (m, 1 H), 7.17-6.99 (m, 3H), 4.48 (s, 1 H), 3.69-3.61 (m, 2H), 3.51-3.47 (m, 2H), 2.98-2.61 (m, 6H), 2.53 (s, 3H), 1.02-0.97 (m, 3H). Compound 95 5-chloro-2-fluoro-N-{4-[4-({3-[(2- hydroxyethyl)(methyl)amino]propyl}amino)-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6- yl]phenyl}benzenesulfonamide This compound was prepared according to the procedure described in example 9. The desired product as a white solid (3.7 mg, 3.88%). LCMS (ES. m/z): 548 [M+H]+. 1H-NMR (DMSO-d6, 300 MHz) δ (ppm): 12.86 (s, 1 H), 8.17 (d, J = 8.4 Hz, 2H), 7.81-7.74 (m, 1 H), 7.67-7.64 (m, 1 H), 7.49-7.29 (m, 2H), 7.07 (d, J = 8.4 Hz, 2H), 4.52 (s, 1 H), 3.72-3.56 (m, 4H), 2.68-2.59 (m, 4H), 2.48 (s, 3H), 2.25 (s, 3H), 1.92-1.79 (m, 2H). Compound 96 N-[4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)-2-methoxyphenyl]-2,5-difluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 9. The desired product as an off white solid (120.7 mg, 19.76%). LCMS (ESI, m/z): 518.2 [M+H]+.1H NMR (300 MHz, DMSO-d6) δ 13.01 (s, 1 H), 7.94 - 7.82 (m, 2H), 7.73-7.51 (m, 3H), 7.29 (d, J = 8.2 Hz, 1 H), 7.09 (t, J = 5.7 Hz, 1 H), 3.77-3.71 (m, 2H), 3.62 (s, 3H), 2.65 (t, J = 6.8 Hz, 2H), 2.53-2.50 (s, 3H), 2.30 - 2.14 (m, 6H).
Compound 995-chloro-N-{2-chloro-4-[4-({2-[(2-hydroxyethyl)(methyl)amino]ethyl}amino)
-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}-2-fluorobenzenesulfonamide
(i) 5-chloro-N-[2-chloro-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]-2- fluorobenzenesulfonamide
Into a 50 mL round-bottom flask were added 2-chloro-4-(4, 4,5, 5-tetramethyl-1 , 3,2- dioxaborolan-2-yl)aniline (1 g, 3.944 mmol, 1 equiv) , Pyridine (467.99 mg, 5.916 mmol, 1.5 equiv) in DCM (10 mL) at room temperature. The resulting mixture was stirred for 6 h at room temperature . The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE I EA (10:1) to afford 5-chloro-N-[2-chloro-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl]-2-fluorobenzenesulfonamide (1 g, 56.83%) as an off-white solid. LCMS: (ES, m/z): [M-HJ+ = 446
(ii) 2-[[2-([6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- yl]amino)ethyl](methyl)amino]ethanol
Into a 50-mL round-bottom flask, was placed 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4- d]pyrimidine (240.00 mg, 1.182 mmol, 1.00 equiv), DCM (10.00 mL), DIEA (458.33 mg, 3.546 mmol, 3 equiv), 2-[(2-aminoethyl)(methyl)amino]ethanol (209.55 mg, 1.773 mmol, 1 .50 equiv). The resulting solution was stirred for 16 hr at room temperature. The resulting mixture was concentrated. The solid was washed with EA and MeOH(10/1). The solids were collected by filtration. This resulted in 220 mg (60.78%) of 2-[[2-([6-chloro-3-methyl- 1 H-pyrazolo[3,4-d]pyrimidin-4-yl]amino)ethyl](methyl)amino]ethanol as a white solid. LCMS: (ES, m/z): [M+H]+ = 285
(iii) 5-chloro-N-{2-chloro-4-[4-({2-[(2-hydroxyethyl)(methyl)amino]ethyl}amino)-3- methyl-1 H-pyrazolo[3,4-d]pyrimidin-6-yl]phenyl}-2-fluorobenzenesulfonamide
A solution of 5-chloro-N-[2-chloro-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]- 2-fluorobenzenesulfonamide (300 mg, 0.672 mmol, 1 equiv) , Cs2CO3 (328.65 mg, 1.008 mmol, 1.5 equiv) , Pd(dppf)CI2 (98.41 mg, 0.134 mmol, 0.2 equiv) in H2 O (30.00 mL, 1664.127 mmol, 2476.38 equiv) and dioxane (9.00 mL, 106.163 mmol, 157.98 equiv) was treated for 10 min at room temperature under nitrogen atmosphere .The resulting mixture was stirred for 16h at 100 °C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The residue was purified by silica gel column chromatography, eluted with CH2 CI2 I MeOH (10:1) to afford the desired product (138.8 mg, 35.37%) as a off-white solid. LCMS: (ES, m/z): [M+H]+ = 568 1 H NMR (400 MHz, DMSO-d6) δ 12.95 (s, 1 H), 9.59 (s, 1 H), 8.23 (s, 1 H), 8.02 (d, J = 8.6 Hz, 1 H), 7.70 (d, J = 4.8 Hz, 1 H), 7.54 (d, J = 8.7 Hz, 1 H), 7.35 - 7.27 (m, 2H), 7.13 (d, J = 5.7 Hz, 1 H), 5.13 (s, 1 H), 3.93 (q, J = 6.1 Hz, 2H), 3.68 (s, 6H), 3.15 (d, J = 11.7 Hz, 2H), 2.80 (s, 3H), 2.54 (s, 3H).
Compound 100 N-[2-chloro-4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-isopropoxypyridine-2-sulfonamide
(i) 4-isopropoxypyridine-2-sulfonyl chloride Into a 100 mL round-bottom flask were added 2-(benzylsulfanyl)-4-isopropoxypyridine (1 g, 3.855 mmol, 1 equiv.), HCI (5 mL, 164.564 mmol) and NaCIO2 (15 mL, 8%) in DCM (20 mL, 314.612 mmol) at room temperature. The resulting mixture was stirred for 4 h at rt. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2x15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 4-isopropoxypyridine- 2-sulfonyl chloride (1 g, 93.54%) as a yellow oil. LCMS: (ES, m/z): [M-H]+ = 236
(ii) N-[2-chloro-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]-4- isopropoxypyridine-2-sulfonamide
Into a 50 mL round-bottom flask were added 4-({6-chloro-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-4-yl}oxy)-1-methylpiperidine (1 g, 3.549 mmol, 1 equiv) , 5-chloro-N-[2- chloro-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]-2- fluorobenzenesulfonamide (1.58 g, 3.542 mmol, 1.00 equiv.) and Pyridine (0.42 g, 5.324 mmol, 1.5 equiv). in DCM (15 mL, 235.959 mmol, 66.48 equiv.) at room temperature. The resulting mixture was stirred for 4 h at rt. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE I EA (10:1) to afford N-[2-chloro-4-(4, 4,5, 5-tetramethyl-1 , 3, 2-dioxaborolan-2- yl)phenyl]-4-isopropoxypyridine-2-sulfonamide (600 mg, 26.55%) as a yellow solid. CMS: (ES, m/z): [M+H]+ = 453
(iii) 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- amine
Into a 100-mL round-bottom flask, was placed 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4- d]pyrimidine (1.00 g, 4.925 mmol, 1.00 equiv.), DCM (30.00 mL), (2- aminoethyl)dimethylamine (521.03 mg, 5.910 mmol, 1.2 equiv.), DIEA (1.90 g, 14.701 mmol, 2.98 equiv.). The resulting solution was stirred for 3 hr at room temperature. The resulting mixture was concentrated. The solids were wished with Ethyl ether. The solids were collected by filtration. This resulted in 1.2 g (86.08%) of 6-chloro-N-[2- (dimethylamino)ethyl]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-amine as a white solid. LCMS: (ES, m/z): [M+H]+ = 255 (iv) N-[2-chloro-4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]-4-isopropoxypyridine-2-sulfonamid
A solution of 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4- amine (500 mg, 1.963 mmol, 1 equiv.) , N-[2-chloro-4-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)phenyl]-4-isopropoxypyridine-2-sulfonamide (888.74 mg, 1.963 mmol, 1 equiv.) , Pd(dppf)CI2 (287.26 mg, 0.393 mmol, 0.2 equiv.) , Cs2CO3 (959.34 mg, 2.945 mmol, 1.5 equiv.) in dioxane (10 mL) and H2 O (3 mL) was treated for 10 min at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16h at 100 °C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2 CI2 I MeOH (10:1) to afford the desired product (129.9 mg, 11.44%) as a brown solid. LCMS: (ES, m/z): [M+H]+ = 5451 H NMR (400 MHz, DMSO-d6) δ 13.04 (s, 1 H), 11.18 (s, 1 H), 8.13-8.43 (d, J = 5.6 Hz, 3H), ,7.25-7.47 (d, J = 8.6 Hz, 3H), 7.11 (dd, J = 5.7, 2.5 Hz, 1 H), 4.79 (p, J = 6.0 Hz, 1 H), 3.91 (q, J = 6.1 Hz, 2H), 3.18 (t, J = 6.4 Hz, 2H), 2.70 (s, 4H), 2.57 (s, 2H), 1.92 (s, 4H), 1.26 (dd, J = 14.0, 6.5 Hz, 6H), 0.90 (t, J = 7.0 Hz, 1 H).
Compound 101 5-chloro-N-(2-chloro-4-{3-methyl-4-[(1-methylpiperidin-4-yl)oxy]-1 H- pyrazolo[3,4-d]pyrimidin-6-yl}phenyl)-2-fluorobenzenesulfonamid
(i) 4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-1-methylpiperidine
Into a 50 mL round-bottom flask were added 4,6-dichloro-3-methyl-1 H-pyrazolo[3,4- d]pyrimidine (500 mg, 2.463 mmol, 1 equiv.) , 4-piperidinol, 1-methyl- (0.28 g, 2.463 mmol, 1 equiv.) in THF (10 mL) and added NaH (0.09 g, 3.695 mmol, 1.5 equiv.) at 0 °C. The resulting mixture was stirred for 4 h at room temperature. The reaction was quenched by the addition of Water (10 mL) at room temperature. The aqueous layer was extracted with EtOAc (3x50 mL). The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE I EA (5:1) to afford 4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-1 -methylpiperidine (300 mg, 43.24%) as a white solid. LCMS: (ES, m/z): [M-HJ+ = 282
(ii) 5-chloro-N-(2-chloro-4-{3-methyl-4-[(1-methylpiperidin-4-yl)oxy]-1 H-pyrazolo[3,4- d]pyrimidin-6-yl}phenyl)-2-fluorobenzenesulfonamid
A solution of 4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-1- methylpiperidine (400 mg, 1.420 mmol, 1 equiv) , 5-chloro-N-[2-chloro-4-(4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]-2-fluorobenzenesulfonamide (633.38 mg, 1.420 mmol, 1 equiv.) , Pd(dppf)CI2 (207.77 mg, 0.284 mmol, 0.2 equiv.) , Cs2CO3 (693.87 mg, 2.130 mmol, 1.5 equiv.) in H2 O (3 mL) and dioxane (12ml) was treated for 10 min at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 100 °C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2 CI2 I MeOH (10:1) to afford the desired product (113.4 mg, 14.05%) as a white solid. LCMS: (ES, m/z): [M+H]+ = 565, 1 H NMR (300 MHz, DMSO-d6) δ 13.33 (s, 1 H), 9.56 (s, 1 H), 8.21 (d, J = 2.1 Hz, 1 H), 8.01 (dd, J = 8.7, 2.2 Hz, 1 H), 7.71 (dd, J = 6.0, 2.8 Hz, 1 H), 7.52 (dt, J = 8.7, 3.4 Hz, 1 H), 7.23 - 7.35 (m, 2H), 5.64 (s, 1 H), 3.39 (s, 4H), 3.17 (d, J = 4.0 Hz, 3H), 2.79 (s, 3H), 2.53 (s, 3H), 2.26 (s, 2H), 2.12 (s, 2H).
Compound 102 N-(2-chloro-4-{3-methyl-4-[(1-methylpiperidin-4-yl)oxy]-1 H-pyrazolo[3,4- d]pyrimidin-6-yl}phenyl)-2,5-difluorobenzenesulfonamide
To a stirred mixture of 4-({6-chloro-3-methyl-1 H-pyrazolo[3,4-d]pyrimidin-4-yl}oxy)-1- methylpiperidine (0.5 g, 1.775 mmol, 1 equiv) and N-[2-chloro-4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)phenyl]-2,5-difluorobenzenesulfonamide (0.84 g, 1.953 mmol, 1.1 equiv) in 1 ,4-dioxane (8 mL) and H2 O (2 mL),was added Cs2CO3 (0.87 g, 2.662 mmol, 1.5 equiv) and Pd(dppf)cl2 (0.26 g, 0.355 mmol, 0.2 equiv) in portions at 100°C under N2 atmosphere for 16h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, silica gel; mobile phase, MeOH in DCM, 0% to 15% gradient in 30 min; detector, UV 254 nm.This resulted in N-(2-chloro-4-{3-methyl-4-[(1-methylpiperidin-4-yl)oxy]-1 H- pyrazolo[3,4-d]pyrimidin-6-yl}phenyl)-2,5-difluorobenzenesulfonamide (0.2104 g, 21.23%) as an off-white solid. LCMS (ESI, m/z): 549[M+H]+. 1 H NMR (300 MHz, DMSO- d6) δ 13.33 (s, 1 H), 9.60 (s, 1 H), 8.21 (d, J = 2.2 Hz, 1 H), 8.00 (dd, J = 8.7, 2.2 Hz, 1 H), 7.49 (ddd, J = 8.1 , 5.4, 2.9 Hz, 1 H), 7.29 (dq, J = 8.1 , 2.9, 1.7 Hz, 3H), 5.63 (s, 1 H), 3.21 (s, 4H), 2.76 (s, 3H), 2.53 (s, 3H), 2.16 (d, J = 48.7 Hz, 4H).
Compound 103 N-[2-chloro-4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]pyridine-2-sulfonamide
(i) Pyridine-2-sulfonyl chloride
To a stirred mixture of 2-pyridinethiol (0.5 g, 4.498 mmol, 1 equiv) in DCM (5 mL, 78.653 mmol, 17.49 equiv.) was added HCI (10 mL, 329.128 mmol, 73.17 equiv.) and NaOCI (10 mL, 147.770 mmol, 32.85 equiv.) in portions at 0°C.The aqueous layer was extracted with H2O(10 ml x 3) and DCM(10ml x 3 ).The resulting mixture was concentrated under reduced pressure. This resulted in pyridine-2-sulfonyl chloride (0.4 g, 30.04%) as a yellow oil. LCMS (ESI, m/z): 178 [M+H]+. (ii) N-[2-chloro-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl]pyridine-2- sulfonamide
To a stirred mixture of pyridine-2-sulfonyl chloride (0.4 g, 2.252 mmol, 1 equiv.) and 2- chloro-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)aniline (0.57 g, 2.252 mmol, 1 equiv.) in DCM (4 mL, 62.922 mmol, 27.94 equiv.) was added Pyridine (0.53 g, 6.756 mmol, 3 equiv.) in portions at RT .The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, silica gel; mobile phase, PE in EA, 0% to 50% gradient in 30 min; detector, UV 254 nm.This resulted in N-[2-chloro-4-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)phenyl]pyridine-2-sulfonamide (0.2 g, 15.34%) as an off-white solid. LCMS (ESI, m/z): 395 [M+H]+.
(iii) N-[2-chloro-4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-6-yl)phenyl]pyridine-2-sulfonamide
To a stirred mixture of 6-chloro-N-[2-(dimethylamino)ethyl]-3-methyl-1 H-pyrazolo[3,4- d]pyrimidin-4-amine (0.15 g, 0.589 mmol, 1 equiv.) and N-[2-chloro-4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)phenyl]pyridine-2-sulfonamide (0.26 g, 0.648 mmol, 1.1 equiv.) in 1 ,4-dioxane (8 mL) and H2 O (2 mL),was added Cs2CO3 (0.29 g, 0.883 mmol, 1.5 equiv.) and Pd(dppf)CI2 (0.09 g, 0.118 mmol, 0.2 equiv.) in portions at 100°C under N2 atmosphere for overnight. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, silica gel; mobile phase, MeOH in DCM, 0% to 15% gradient in 30 min; detector, UV 254 nm.This resulted in the desired product (0.0389 g, 13.28%) as a off- white solid. LCMS (ESI, m/z): 553 [M+H]+1 H NMR (300 MHz, DMSO-d6) δ 12.98 (s, 1 H), 9.86 (s, 1 H), 8.63 (d, J = 4.4 Hz, 1 H), 8.25 (d, J = 2.0 Hz, 1 H), 8.10 - 7.77 (m, 3H), 7.59 - 7.40 (m, 2H), 7.14 (t, J = 5.7 Hz, 1 H), 3.84 (q, J = 6.2 Hz, 2H), 3.02 (t, J = 6.5 Hz, 2H), 2.57 (d, J = 15.7 Hz, 9H).
Compound 107 N-[2-chloro-4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-methoxypyridine-2-sulfonamide; trifluoroacetic acid This compound was prepared according to a procedure similar to the synthesis of Compound 84. The desired product was obtained as an off- white solid (51.4 mg, 16.59%) .LCMS: (ES, m/z): [M+H]+ = 517.1 H NMR (400 MHz, DMSO-d6) δ 13.20 (s, 1 H), 10.34 (s, 1 H), 9.56 (s, 1 H), 8.56 (d, J = 5.6 Hz, 1 H), 8.35 (d, J = 1.9 Hz, 1 H), 8.28 (dd, J =
8.5, 2.0 Hz, 1 H), 7.51 (d, J = 8.5 Hz, 1 H), 7.44 (d, J = 2.4 Hz, 1 H), 7.38 (q, J = 5.7 Hz, 1 H), 7.27 (dd, J = 5.6, 2.5 Hz, 1 H), 3.99 (q, J = 5.9 Hz, 2H), 3.91 (s, 2H), 2.89 (d, J = 4.4 Hz, 6H), 2.58 (s, 3H).
Compound 111 5-chloro-N-[2-chloro-4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2-fluorobenzenesulfonamide
This compound was prepared according to a procedure similar to the synthesis of Compound 84. The desired product was obtained as a white solid (0.0328 g, 12.83%). LCMS (ESI, m/z): 538 [M+H]+.1 H NMR (300 MHz, DMSO-d6) δ 12.94 (s, 1 H), 8.21 (d, J = 2.1 Hz, 1 H), 7.98 (d, J = 8.7 Hz, 1 H), 7.76 - 7.45 (m, 2H), 7.32 - 7.02 (m, 3H), 3.91 (d, J = 5.9 Hz, 2H), 2.85 - 2.67 (m, 6H), 2.54 (s, 5H). Compound 112 N-[2-chloro-4-(4-{[2-(dimethylamino)ethyl]amino}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-4-methylpyridine-2-sulfonamide; trifluoroacetic acid
This compound was prepared according to a procedure similar to the synthesis of Compound 84. The desired product was obtained as a (32.2 mg, 10.62%) as an off- white solid. LCMS: (ES, m/z): [M+H]+ = 5011 H NMR (400 MHz, DMSO-d6) δ 13.17 (s, 1 H), 10.28 (s, 1 H), 9.47 (s, 1 H), 8.58 (d, J = 2.1 Hz, 1 H), 8.33 (d, J = 2.0 Hz, 1 H), 8.27 (dd, J = 8.4, 2.0 Hz, 1 H), 7.90 (dd, J = 8.2, 2.2 Hz, 1 H), 7.84 (d, J = 8.0 Hz, 1 H), 7.50 (d, J = 8.5 Hz, 1 H), 7.36 (p, J = 5.5, 4.9 Hz, 1 H), 3.98 (q, J = 5.9 Hz, 2H), 3.42 (q, J = 5.7 Hz, 2H), 2.89 (d, J = 4.2 Hz, 6H), 2.58 (s, 3H), 2.41 (s, 3H).
Compound 126 2,5-difluoro-N-(2-fluoro-4-{3-methyl-4-[(1-methylpiperidin-4-yl)oxy]-1 H- pyrazolo[3,4-d]pyrimidin-6-yl}phenyl)benzenesulfonamide This compound was prepared according to a procedure similar to the synthesis of Compound 102. The desired product was obtained as a yellow solid (0.4049 g, 42.11%). LCMS (ESI, m/z): 533 [M+H]+.1H NMR (300 MHz, DMSO-d6) δ 13.41 (s, 1 H), 9.95 (s, 1 H), 8.10 - 7.61 (m, 2H), 7.65 - 7.04 (m, 5H), 5.71 (d, J = 36.7 Hz, 1 H), 3.21 - 3.09 (m, 4H), 2.73 (s, 3H), 2.42 (s, 3H), 2.16 (d, J = 44.1 Hz, 4H).
Compound 154 5-chloro-N-(4-(4-((2-(dimethylamino)ethyl)(methyl)amino)-3-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl)-2-fluorobenzenesulfonamide
This compound was prepared according to the procedure described in example 9. The desired product was obtained as an off-solid (5 mg, 4.9%). LCMS (ESI, m/z): 519.8 [M+H]+. 1H NMR (400 MHz, DMSO-d6): δ 11.05 (s, 1 H), 8.26 (d, J = 8.60, 2H), 7.87-7.73 (m, 2H), 7.50 (s, 1 H), 7.23 (d J = 8.79 Hz, 2H), 4.19 (m, 2H), 3.46 - 3.40 (m, 2H), 3.38 (s, 3H), 2.85 (d, J =4.49 Hz, 6H), 2.59 (s, 3H).
Compound 156 N-[2-chloro-4-(4-{[(3S,4R)-3-fluoropiperidin-4-yl]oxy}-3-methyl-1 H- pyrazolo[3,4-d]pyrimidin-6-yl)phenyl]-2,5-difluorobenzenesulfonamide hydrochloride
This compound was prepared according to a procedure similar to the synthesis of Compound 29. The desired product was obtained as an off-white solid (0.2211 g, 77.66%)
LCMS (ESI, m/z): 553 [M+H]+, 1H NMR (300 MHz, DMSO-d6) δ 13.67 (s, 1 H), 10.81 (s, 1 H), 9.30 (s, 1 H), 8.78 (s, 1 H), 8.46 - 8.27 (m, 2H), 7.72 - 7.44 (m, 4H), 5.90 (dd, J = 26.8, 9.3 Hz, 1 H), 5.46 (s, 1 H), 3.67 (s, 4H), 2.56 (s,3H) 2.21 (s, 2H).
Compounds 127-155 and 157 to 175 are prepared according to procedures similar to other procedures described herein.
Table A. Listing of compounds
Figure imgf000260_0001
EXAMPLE 2: Effect of the Compounds on SGK-1 Activity
• SGK1 assay
The ability of the synthesized compounds to inhibit SGK-1 was assessed in an enzymatic assay by determining their effect on the ability of the isolated SGK1 enzyme to catalyze the transfer of the phosphate from ATP to serine/threonine residues in a labeled substrate peptide and in cellular assay by using the NanoBRET target engagement assay kit.
Enzymatic activity assay. The compounds were tested for SGK-1 activity by measuring the ability of the compound to inhibit the transfer of phosphate from ATP by the isolated enzyme to serine/threonine residues in a fluorescein labeled substrate peptide, FLPeptide 6 (PerkinElmer, Waltham, USA, Cat. No: 760350). The enzymatic reaction was initiated by addition of 15 μL of solution 1 containing (in mM) 10 MgCI2, 0.010 % Brij-35, 2 DTT, 0.05 % BSA, 1 EGTA, 50 HEPE (pH7.5) and 0.665 nM SGKto 5 μL of solution 2 containing 10 MgCI2, 0.010 % of Brij-35, 2 DTT, 0.05 %BSA, 1 EGTA, 50 HEPES (pH7.5), 6 μM of FLPeptide and 80 μM of ATP. After incubating the plate at room temperature for 90 min, 75 μL of stopping buffer (containing 0.5 M EDTA) is added to terminate the reaction. The samples were analyzed using an EZ reader. For the determination of the compound dose response, stock solution of compound prepared in DMSO was diluted and tested in a 10 point, three-fold dilution series run in duplicate beginning at 10 μM final concentration.
Cellular enzymatic activity assay. The cellular SGK1 -kinase activity is determined using SGK1 NanoBRET™ target engagement (TE) intracellular kinase assay kit. The BRET methodology relies on the emission of an optical signal dependent on the spatial proximity of the luciferase-conjugated target protein and a fluorescent-labelled tracer molecule. The displacement of the tracer by a competitive inhibitor therefore diminishes the apparent BRET signal. HEK293 cells that were cultured in Dulbecco modified Eagle medium (DMEM), was transfected with 9 ug/mL Carrier DNA and 1 ug/mL SGK1-NanoLuc fusion vector and mix liplid: DNA complex with 1:20 (v:v) cell suspension. Transfected cells were seeded at 2000 cells/100 μL/well in 96-well plate and incubated overnight. After overnight incubation, 5 μL 20 x NanoBRET tracer was added per well (final 0.5 uM), followed by 10 μL 10x cpd solution per well, incubated for 2 hours at 37 °C, 5% CO2. 50 μL 3 x complete substrate solution was then added per well and the luminescence signal was read after 15-30 min equilibration at room temperature using Synergy 4 plate reader. Table B. IC50 values for inhibition of SGK-1 activity
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
• Kinase Selectivity
The following compounds were profiled against a 50-kinase Mixed panel at 10 μM using the KinaseSeeker™ assay.
Mixed Kinase panel:
AKT1 , AKT2, AKT3, PDK1/PDPK1, PKA/PRKACA, PKC-ε/PRKCE, PKG1/PRKG1 , PKX/PRKX, RPS6KA3/RSK2, RPS6KA4/MSK2, YANK2, AMPK-a1/AMPK, CAMK1 D, CAMK2D, DAPK3, MARK1 , MARK2, PIM1 , SNF1 LK/SIK1 , SNF1 LK2/SIK2/QIK, CLK2, CDK5, p38-a/MAPK14, AAK1 , AURKA, AURKB, AURKC, PLK4, SLK, TAOK1 , YSK1 , ABL1 , DDR1 , EPHA5, EPHB2, FLT1/VEGFR1 , FLT3, HCK, IGF1 R, ITK, KIT, MUSK, PDGFRB, PTK2B/PYK2, SRC, TNK1 , VEGFR2/KDR/FLK1 , ACVR2A/ACVR2, MLK2/MAP3K10, MLK3/MAP3K11 .
Assay Design KinaseSeeker is a homogeneous competition binding assay where the displacement of an active site dependent probe by an inhibitor is measured by a change in luminescence signal. Luminescence readout translates into a highly sensitive and robust assay with low background and minimal interference from test compounds.
Assay Method 10 mM stock of the compound was diluted in DMSO to a concentration of 250 μM. Prior to initiating a profiling campaign, the compound was evaluated for false positive against split-luciferase. The compound was then screened in duplicate against each of the kinases. For kinase assays, each Cfluc-Kinase was translated along with Fos- Nfluc using a cell-free system (cell lysate) at 30 °C for 90 min. 24 μL aliquot of this lysate containing either 1 μL of DMSO (for no inhibitor control) or compound solution in DMSO (10 μM final concentration) was incubated for 2 hours at room temperature in presence of a kinase specific probe. 80 μL of luciferin assay reagent was added to each solution and luminescence was immediately measured on a luminometer.
Figure imgf000287_0001
Profiling data for all kinases was plotted as % inhibition vs. kinases profiled. A heat map representing the effect of compounds on kinases was also generated.
Table C. Kinase Selectivity of the compounds
Figure imgf000287_0002
Figure imgf000288_0001
• hERG
Cell lines and cell culture hERG stably expressed HEK 293 cell line (Cat# K1236) was purchased from Invitrogen. The cells are cultured in 85% DM EM, 10% dialyzed FBS, 0.1 mM NEAA, 25 mM HEPES, 100 U/rnL Penicillin-Streptomycin and 5 pg/mL Blasticidin and 400 pg/mL Geneticin. Cells are split using TryμLE™ Express about three times a week, and maintained between -40% to -80% confluence. Before the assay, the cells were transferred onto the coverslips at 5 x 105 cells/per 6 cm cell culture dish and induced with doxycycline at 1 pg/mL for 48 hours.
Solution preparations Extracellular solution (in mM): 132 NaCI, 4 KCI, 3 CaCl2, 0.5 MgCl2, 11.1 glucose, and 10 HEPES (pH adjusted to 7.35 with NaOH). Intercellular solution (in mM): 140 KCI, 2 MgCI2, 10 EGTA, 10 HEPES and 5 MgATP (pH adjusted to 7.35 with KOH)
Working solution preparation for test compound Test compounds were initially prepared in DMSO with final concentration of 10 mM as stock solution according to SOP- ADMET-M AN-007. Then stock solution of each compound was serial-diluted by ratio of 1 :3 with DMSO to prepare additional 3 intermediate solutions including 3.33, 1.11 and 0.37 mM. Before hERG assay, the working solutions were prepared by dilution of 10, 3.33, 1.11- and 0.37-mM intermediate solutions in 1000 folds using extracellular solution, so that the final concentration of working solution was 10, 3.33, 1.11 and 0.37 mM, while 30 μM working solution was prepared by 333.333-folds dilution of 10 mM DMSO stock. The final DMSO concentration in working solutions was maintained in range of 0.1-0.3% (v/v). hERG current in presence of 5 doses including 30, 10, 3.33, 1.11 and 0.37 μM, was measured for IC50 determination. Data analysis Percent current inhibition was calculated using the following equation.
Peak current inhibition
Figure imgf000289_0001
The dose response curve of test compounds was plotted with %inhibition against the concentration of test compounds using Graphpad Prism 8.0, and fit the data to a sigmoid dose-response curve with a variable slope.
Table D. hERG values of the compounds
Figure imgf000289_0002
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Culture and treatment of PBMCs
Cryopreserved peripheral blood mononuclear cells (PBMCs) were ordered from ZenBio (catalogue number SER-PBMC-P-F, lot number PBMC031022A-H, 15-30 million cells per vial), quickly thawed in a 37°C water bath and the cell suspension was transferred to a 50 mL sterile polypropylene tube. The empty vial was rinsed with 1 mL of RPMI media containing 10% heat inactivated FBS and 1% penicillin/streptomycin and transferred drop- wise to the 50 mL tube containing the cell suspension. Ten milliliters of media were added drop-wise to the 50 mL tube, and the tube was centrifuged at 300g for 6 min. Supernatant was removed, and the PBMC pellet was gently resuspended by flicking the tube with the remaining media. Twelve milliliters of media was added to the tube, and 1.5 ml of the cell suspension was aliquoted in eight wells of 6-well plates to obtain a final seeding density of 3.9 million cells per well, and incubated at 37°C, 5% CO2 for 24 hours. PBMCs were then treated with either a serial dilution or various concentrations of Compound 84, Compound 79, or DMSO and incubated for 0-24 hours at 37°C, 5%CO2. The 6-well plates were then put on ice, and the PBMC suspension was transferred in a separate 15 mL centrifuge tube for each treatment. Each well of the 6-well plate was rinsed with 2 ml of cold 1X PBS, and added to the corresponding 15 mL centrifuge tube. PBMCs were centrifuged at 300g for 6 min, pellets were rinse once with 2 ml of cold PBS, centrifuged a second time (300g for 6 min) and the cell pellets were resuspended with 50 μL of RIPA buffer (Pierce catalogue number 89900) + 1% Protease/phosphatase inhibitors (Cell Signaling 5872s), and mixed by pipetting up and down and transferred in a pre-chilled 1.7 mL microtube. Samples were incubated for 10min on ice. Cell lysates were then centrifuged for 10min at 4°C at 10 000 rpm. Supernatants were collected and transferred in another new pre-chilled 1.7 mL microtube. Determination of cell lysate total protein concentration was done using a BCA assay kit (ThermoScientific catalogue #23227) by diluting the samples 1/10 (2.5μL sample + 22.5 μL 1X PBS). Protein lysates were stored at -80°C, until further use.
Western blot of PBMCs
Twenty micrograms of cell lysate samples were diluted in 4X Loading Buffer (800 μL 5X Loading buffer, BioLegend catalogue number 426311 + 200 μL B-Mercaptoethanol, Sigma catalogue number M3148-25ml), heated for 5min at 95°C, and loaded on Novex WedgeWell precast Tris-Glycine 8-16% gels (Invitrogen catalogue number XP08162BOX using 10X Tris/Glycine/SDS Running buffer (Biorad catalogue number 1610734). The gels were run at 120V for 90 min, and transferred on Hybond 0.45 PVDF membrane (Cytiva catalogue number 10600029) in 10X Tris/Glycine Transfer buffer (Biorad catalogue number 1610734) at 400mA for 1.5 hours at 4°C. Membranes were rinsed in TBST buffer (1X TBS +0.1%Tween™ 20 (Fisher catalogue number 0777-1 L)), and blocked for 1 hour with 5% BSA (Sigma catalogue number 10735086001) in TBST at RT. The membranes were then incubated overnight with primary antibodies (anti pNDRGI , Cell Signaling catalogue number 5482S at 1 :1000 dilution, anti-NDRG1 , Cell signaling catalogue number 5196S at 1 :1000 dilution, and anti-vinculin, Cell Signaling catalogue number 13901S at 1 : 5000 dilution in 1 % BSA in TBST. The next day, the membranes were washed three times 15minutes in TBST at room temperature, and incubated with anti-Rabbit HRP Antibody (Cell Signaling, 7074P2) at 1 :5,000 dilution for pNDRGI and NDRG1 , and 1 :50,000 dilution for vinculin in 1% BSA in TBST for 1 hour at room temperature. The membranes were washed 3x 15 minutes in TBST at room temperature, and revealed with ECL reagent (SuperSignal West Pico Plus chemiluminescent substrate, ThermoScientific catalogue number 34577 or SuperSignal West Pico Femto chemiluminescent Substrate, ThermoScientific catalogue number 34094) on Biorad ChemiDoc MP.
Fig. 4A shows that compound 79 inhibits phosphorylation of NDRG1 in PBMCs. Similarly, Fig. 4B shows that compound 79 inhibits phosphorylation of NDRG1 in BT-549 breast cancer cells in a dose-dependent manner. Fig. 5 shows that compound 84 inhibits phosphorylation of NDRG1 in ex vivo treated blood from healthy volunteers. Inhibition of NDRG1 phosphorylation ws observed in 5/5 healthy volunteers at 5 μM of compound 79. Inhibition of NDRG1 phosphorylation ws observed in 3/5 healthy volunteers at 0.5 μM of compound 79.
Fig. 6 shows that NDRG1 is a direct target of SGK1 phosphorylation. These data therefore suggest that compounds 84 and 79 directly inhibit SGK1 activity.
EXAMPLE 3: Determination of the efficacy of SGK1 inhibitors on LQT3 by studying its effect on the action potential duration (APD) of LQT-patient derived cardiomyocytes (iPSC-CMs)
SGK1 inhibition is suggested to decrease the APD of cardiomyocytes that exhibits the phenotype of LQT3 patients. Incubation with SGK1 inhibitors reduces the APD of the cardiomyocytes which can be investigated by imaging of cells using FluoVolt dye.
Material and methods for differentiation of cardiomyocytes: Stem cells derived from LQT-3 patients (iPSCs) were cultured in mTeSR™ 1 media (STEMCELL Tech., 85851) in 6-cm dishes pre-coated with Geltrex (Life Technology, A1413302) and incubated at 37°C and 5% CO2. At 85% confluence, iPSCs were disaggregated with ReLeSR™ (STEMCELL Tech., 05872), passaged into 24-well plates, and allowed to grow for 3-4 days to create a monolayer. The differentiation strategy used has been reported previously. For differentiation, the culture medium was changed to RPMI 1640 GlutaMAX™ plus 25mM HEPES supplemented with B27-minus Insulin (Gibco, A18956-01) containing CHIR99021 (TOCRIS, 4423, 6μM as working concentration) from days 0 to 2. On day 2, medium was changed to RPMI-B27-minus insulin containing IWP2 (TOCRIS, 3533, 5μM as working concentration) and incubated until day 4. On day 4, the medium was changed back to normal RPMI GlutaMAX™-B27-minus insulin and cells were maintained in this media until beating cardiomyocytes appeared, typically around day 10 or day 12. After beating was seen, iPSC-CMs were maintained in cardiomyocyte maintenance medium (DMEM, No phenol red, 2% charcoal stripped FBS). Cardiomyocyte differentiation and maintenance are done in a 24 well format. Prior to each experiment iPSC-CMs need to be re-plated onto a 35mm dish and allowed to stabilize for 1 week. Following stabilization of iPSC-CMs in a glass bottom 35 mm dish for 1 week, compounds are applied. Experiment design of APD measurement: iPSC-CMs are maintained in DMEM plus 2% FBS until replating. 3x105 cells are plated into each 35 mm glass dish in DMEM plus 20% FBS and maintained in DMEM plus 2% for 1 week for CM stabilization. Either DMSO, Mex (10uM), SGK1 Inhibitor Compound (3uM), or SGKI Inhibitor Compound (30uM) in DMEM plus 2% FBS are added to the plated cells. At 4 hours post drug administration, the media with the drug is washed out of the first set of 4 plates and replaced with a Tyrode solution containing the FluoVolt dye. Live imaging is taken of approximately 10-12 randomly selected “flashing” cells (see live imaging methods section). Cells are paced at 1 Hz. The raw data from live cell imaging is exported to Excel software (Microsoft, Redmond, WA) and then analyzed with an “in-lab” developed Excel-based program. The loading of the FluoVolt dye in the experiments is performed as follows: Before starting, pre-warm 6.5mL Tyrode’s solution to 37°C. Aspirate medium and rinse cells with 1mL Tyrode. Add 1.25μL PowerLoad and 0.125|JL FluoVolt to 0.5mL Tyrode’s and add to the center of the 35mm dish glass inset. Incubate 20min at 37°C. Rinse cells 3 times with 1mL Tyrode’s. Add 2mL Tyrode’s. Image cells within 2h, using GFP filter.
Live cell imaging for action potential duration (APD) measurement: iPSC-CMs were cultured on 35mm glass bottom dishes (MatTek, P35G-1.5-10-C) that was pre-coated with fibronectin solution at 10 pg/ml (Thermofisher, 3016015) at 37°C, 5% CO2. For imaging, cells were incubated at 37°C, 5% CO2 for 20 minutes in Tyrode solution containing a fluorescent voltage sensitive dye, FluoVolt (ThermoFisher, Cat#F10488, working concentration of 5 uM) and Pluronic ® F-127 (Thremofisher, P3000MP, working concentration of 0.05%). They were then washed three times in fresh Tyrode solution. During imaging, the dishes were kept in a heated 37°C stage-top environment chamber supplied with 5% CO2. Imaging of voltage-indicated cellular action potential duration (APD) was taken under a 40X-water objective using a Nikon Eclipse Ti light microscope. Time-lapse videos of multiple, individual beating iPSC-CMs, paced at 1 Hz were recorded at a speed of 20 milliseconds per frame for 20 seconds at 5% LED power. Single regions of interest were selected for every beating iPSC-CM captured in the recordings. The raw data was exported to Excel software (Microsoft, Redmond, WA) and then analyzed with an “in-lab” developed Excel-based program.
Table E. Effects of compounds on APD90 of 4-week-old P1332L-SCN5A iPSC-CMs (4- hour treatment) APD shortening and concentration
Figure imgf000296_0001
EXAMPLE 4: Solubility and Pharmacokinetic properties of the compounds Solubility
Buffer for preparing FeSSIF buffer was prepared by dissolving 4.040 g of NaOH, 8.650 g of glacial acetic acid and 11.874 g of NaCI in about 900 mL ultrapure water and the pH of the solution was adjusted to 5.0 with 1 N NaOH or 1 N HCI. Then the solution was diluted with ultrapure water to 1000 mL at room temperature. 11.200 g of FaSSIF, FeSSIF & FaSSGF Powder was added to about 500 mL of buffer. Stir until the powder was completely dissolved. Then the solution was diluted with the buffer to 1000 mL at room temperature. Ready to use within 48 hours at room temperature and 24 hours at 37°C.
Preparation of stock solutions the stock solutions of test compounds and control compound progesterone were prepared in DMSO at the concentration of 10 mM. Procedure for solubility determination 30 μL of stock solution (10 mM) of each sample was placed in order into its proper 96-well rack, 970 μL of FeSSIF or PBS pH 7.4 was added into each vial of the cap-less Solubility Sample plate. The assay was performed in duplicate. Add one stir stick to each vial and seal using a molded PTFE/Silicone plug. Then the Solubility Sample plate was transferred to the Eppendorf Thermomixer Comfort plate shaker and shaken at 25°C at 1100 RPM for 2 hours. After completion of the 2 hours, plugs were removed and the stir sticks were removed using a big magnet, the samples from the Solubility Sample plate were transferred into the filter plate. Using the Vacuum Manifold, all the samples were filtered. Aliquot of 5 μL was taken from the filtrate followed by addition of 5 μL DMSO and 490 μL of a mixture of H2 O and acetonitrile. The dilution factor was changed according to the solubility values and the LC-MS signal response.
Preparation of 3 μM standards (STD) From the 10 mM DMSO STD plate, 15 μL was transferred into the remaining empty plate, and then 485 μL of DMSO was added to that plate to have a STD concentration of 300 μM. From the 300 μM DMSO STD plate, 5 μL DMSO STD was transferred into the remaining empty plate, and then 5 μL FeSSIF or PBS pH 7.4 and 490 μL of a mixture of H2 O and acetonitrile was added to that plate to have a final STD concentration of 3 μM. The concentration of the standard samples was changed according to the LC-MS signal response.
Procedure for sample analysis the plate was placed into the well plate autosampler. The samples were evaluated by LC-MS/MS analysis (LC system: Shimadzu, MS analysis: Triple Quad™ 5500 instrument from AB Inc (Canada) with an ESI interface, method: temperature 40 °C, Injection volume: 1 μL or 2 μL, Column: XSelect™ Hss T3 2.5pm (2.1 x50 mm) Column XP, Mobile phase: 0.1% formic acid in water (A) and 0.1% formic acid in acetonitrile (B) and Elution rate: 1.0 mL/min)
Data analysis All calculations were carried out using Microsoft Excel. The filtrate was analyzed and quantified against a standard of known concentration using LC coupled with mass spectral peak identification and quantitation. Solubility values of the test compound and control compound were calculated as follows:
Figure imgf000297_0001
Any value of the compounds that was not within the specified limits was rejected and the experiment was repeated. Table F. Solubility of the compound
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
• Pharmacokinetic data
Protocol for PK study in CD1 mouse via ora/IV administration
All experimental procedures have been conducted in accordance to German Animal Protection Law, as well as according to international animal welfare legislation and rules.
■ PO (30 mg/kg, 10 mL/kg) dosing:
Three male CD1 mice (6-8 weeks, 20-30 g) were used for this study. Each mouse was given 30 mg/kg of the tested drug by oral route PO. The test compound was dissolved in 0.5% HEC, 0.4% Tween 80 in saline for oral PK. Following the administration of the test compound, 30 uL of blood was collected from each mouse at 0.25, 0.5, 1 , 2, 4, 8, 24 h post dosing to measure the concentration of test samples in the plasma. For the processing of collected blood samples, the collected blood sample is dissolved in heparin to prevent the coagulation of blood, following which it is centrifuged at 4000 g for 5 minutes. After centrifugation, the separated plasma will be stored in freezer at 75±15°C. A LC-MS/MS system is used to measure the concentration of the test sample in plasma. WinNonlin (Phoenix TM , version 6.1) will be used for the pharmacokinetic calculations and from the plasma concentration versus time data C max , T max , T 1/2 , AUC inf , AUG last , the number of points for regression are calculated.
■ IV (2 mg/kg, 5 mL/kg) Dosing
Three male CD1 mice (6-8 weeks, 20-30 g) were used for this study. Each mouse was given 2 mg/kg of the tested drug by IV route PO. The test compound was dissolved 5% DMSO in “20% SBE in PBS (pH 7.4)”. Following the administration of the test compound, 30 uL of blood was collected from each mouse at 0.25, 0.5, 1, 2, 4, 8, 24 h post dosing to measure the concentration of test samples in the plasma. For the processing of collected blood samples, the collected blood sample is dissolved in heparin to prevent the coagulation of blood, following which it is centrifuged at 4000 g for 5 minutes. After centrifugation, the separated plasma will be stored in freezer at 75±15°C. A LC-MS/MS system is used to measure the concentration of the test sample in plasma. WinNonlin (Phoenix TM , version 6.1) will be used for the pharmacokinetic calculations and from the plasma concentration versus time data C max , T max , T 1/2 , AUC inf , AUC last , the number of points for regression are calculated. Table G. Mouse Pharmacokinetic data
Figure imgf000301_0001
Figure imgf000302_0001
Protocol for PK study in SD rat via ora/IV administration
All experimental procedures have been conducted in accordance to German Animal Protection Law, as well as according to international animal welfare legislation and rules. ■ PO (30 mg/kg, 10 mL/kg) dosing:
Three male SD rat (6-8 weeks, 200-300 g) were used for this study. Each mouse was given 30 mg/kg of the tested drug by oral route PO. The test compound was dissolved in 0.5% HEC, 0.4% Tween 80 in saline for oral PK. Following the administration of the test compound, 200 uL of blood was collected from each rat at 0.25, 0.5, 1, 2, 4, 8, 24 h post dosing to measure the concentration of test samples in the plasma. For the processing of collected blood samples, the collected blood sample is dissolved in heparin to prevent the coagulation of blood, following which it is centrifuged at 4000 g for 5 minutes. After centrifugation, the separated plasma will be stored in freezer at 75±15°C. A LC-MS/MS system is used to measure the concentration of the test sample in plasma. WinNonlin (Phoenix TM , version 6.1) will be used for the pharmacokinetic calculations and from the plasma concentration versus time data C max , T max , T 1/2 , AUC inf , AUC last , the number of points for regression are calculated. ■ IV (2 mg/kg, 5 mL/kg) Dosing
Six male SD rat (6-8 weeks, 200-300 g) were used for this study. Each mouse was given 2 mg/kg of the tested drug by IV route PO. The test compound was dissolved 5% NMP, 5% solutol in “20% SBE in PBS (pH 7.4)”. Following the administration of the test compound, 200 uL of blood was collected from each rat at 0.25, 0.5, 1 , 2, 4, 8, 24 h post dosing to measure the concentration of test samples in the plasma. For the processing of collected blood samples, the collected blood sample is dissolved in heparin to prevent the coagulation of blood, following which it is centrifuged at 4000 g for 5 minutes. After centrifugation, the separated plasma will be stored in freezer at 75±15°C. A LC-MS/MS system is used to measure the concentration of the test sample in plasma. WinNonlin (Phoenix TM , version 6.1) will be used for the pharmacokinetic calculations and from the plasma concentration versus time data C max , T max , T 1/2 , AUC inf , AUC last , the number of points for regression are calculated.
Table H. Rat Pharmacokinetic data
Figure imgf000303_0001
Figure imgf000304_0001
• CYP inhibition
The master solution was prepared according to Table I, and then 1 μL of 2 mM of compound solution or 1 μL of DMSO was added to the above master solution. The final concentration of test compound and control compounds was 10 μM.
Table I. Preparation of master solution
Figure imgf000304_0002
For CYP1A2 inhibition, 1 μL of specific drug substrate (Phenacetin: 8 mM) was added at the final concentration of 40 μM to the above solution.
For CYP2B6 inhibition, 1 μL of specific drug substrate (Bupropion: 10 mM) was added at the final concentration of 50 μM to the above solution.
For CYP2C9 inhibition, 1 μL of specific drug substrate (Tolbutamide: 40 mM) was added at the final concentration of 200 μM to the above solution. For CYP2D6 inhibition, 1 μL of specific drug substrate (Dextromethorphan: 2 mM) was added at the final concentration of 10 μM to the above solution.
For CYP3A4/5 inhibition, 1 μL of specific drug substrate (Midazolam: 1 mM) was added at the final concentration of 5 μM to the above solution.
For CYP3A4/5 inhibition, 1 μL of specific drug substrate (Testosterone: 10 mM) was added at the final concentration of 50 μM to the above solution.
The mixture was pre-warmed at 37°C for 5 min. The reaction was started by the addition of 20 μL of 10 mM NADPH solution at the final concentration of 1 mM and carried out at 37°C.
The reaction was stopped by addition of 400 μL of cold quench solution (methanol containing internal standards (IS: 100 nM alprazolam, 500 nM labetalol and 2 μM ketoprofen)) at the designated time points (Phenacetin: 20 min; Bupropion: 20 min; Tolbutamide: 20 min; Dextromethorphan: 20 min; Midazolam: 5 min; Testosterone: 10 min). Samples were vortexed for 5 minutes and centrifuged at 3220 g for 40 minutes at 4°C. And then 100 μL of the supernatant was transferred to a new 96-well plate with 100 μL water for LC-MS/MS analysis. All experiments were performed in duplicate. The formation of metabolites was analyzed by using LC-MS/MS. A decrease in the formation of the metabolites in peak area ratios to vehicle control was used to calculate % inhibition values.
% Remaining activity = (average ratio of test compounds or inhibitor)/(average ratio of vehicle control)*100
% Inhibition = 100 - % Remaining activity
Table J. Inhibition percentages for test compound and known inhibitors against CYP1A2, CYP2B6, CYP2C9, CYP2D6 and CYP3A4
Figure imgf000305_0001
Figure imgf000306_0003
• Safety Margin
A safety margin can be obtained by calculating hERG I IC50, using the values IC50 (Whole cell) and hERG (IC50, nM) from tables B and E, respectively. A higher safety margin is desired. The safety margins of several compounds of the present application were compared with each other and with the safety margin of exemple compounds disclosed in patent application Pub No. WO 2014/140065, which is hereby incorporated by reference in its entirety. Some safety margin values are shown at Table K.
The safety margin of several compounds of the following Formula is shown at Table K, to evaluate the effect of the Z-R3 group on the safety margin.
Figure imgf000306_0001
where
Figure imgf000306_0002
Table K. Effect of Z-R3 on safety margin hERG / IC50
Figure imgf000307_0001
Figure imgf000308_0001
It was also found that when W1 is F or Cl, bioavailability is generally improved compared to when W1 is H, while generally maintaining an acceptable safety margin. This can be seen, for example, when comparing several compounds, as shown in Table L:
Table L. Effect of Wi on Bioavailability and hERG / IC50
Figure imgf000308_0002
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0001
Figure imgf000312_0001
Figure imgf000313_0001
EXAMPLE 5: Anticancer effect of the Compounds
In vitro Combination assay
Cell culture and seeding of cell lines'. Cell lines were ordered and cultivated according to the provider’s instructions as highlighted in Table N below.
Table N. Description and seeding density of cell lines
Figure imgf000313_0002
Figure imgf000314_0001
For example, JIMT-1 cells (Creative Bioarray catalogue number CSC-C0592) were grown in Dulbecco's Modified Eagle's Medium - high glucose, (DMEM, Sigma catalogue number D6429-500ml) containing 10% heat-inactivated fetal bovine serum (VWR catalogue number 97068-085) and penicillin/streptomycin (Corning, catalogue number 30-002-CL) and prophylactic plasmocin (InvivoGen, catalogue number ant-mpp) as per the cell line manufacturer’s instructions. When cells reached 70-90% conflucency in a T-75 or T-150 flask (Corning catalogue number 430641 U), media was removed from the cultured flask, and cells were gently rinsed with 10 mL of 1X PBS. 1.5 mL of trypsin (ThermoFisher Scientific catalogue number 25200072) was added and the flask was incubated for 10 min at 37°C, 5% CO2. 8.5 mL of culture media was added to inhibit the trypsin and the cell suspension was transferred into a 15 mL sterile polypropylene tube and centrifuged at 130 x g for 5 min. The supernatant was removed and the cell pellet was resuspended with 4 mL of DMEM. For counting, 10 μL of cell suspension was used and mixed with a Trypan Blue solution (ThermoFisher catalogue number T10282), which were added to a cell counting chamber slide (FisherScientific catalogue number C10228) and counted using a Countess counter (Invitrogen catalogue number C10281) or similar apparatus. Cells should have >85% viability to proceed.
Thirty-nine milliliters of DMEM was added to two 50 mL polypropylene sterile tubes, then sufficient cell suspension was added to each 50mL tube to reach a final concentration of 25,000 cells/mL. Cell suspension was gently mixed by inverting the tubes and 100 μL of cells were plated in each well of 96-well clear bottom white sterile tissue culture plates (Corning catalogue number 3610) to obtain 2,500 cells/well, except for wells A1 of each plate (no cell control). Cells were incubated overnight at 37°C, 5%CO2.
A six-by-six matrix approach was used to measure combination effects, where each compound was serially diluted two-fold by row (first compound) or column (second compound) in order to test different permutations of each compound. The AKT inhibitor dilutions at 1000X in DMSO were prepared ranging from 10 mM to 0.3125 mM using two- fold dilutions. For Compound 79, two-fold dilutions starting at 10 uM to 0.3125 uM were added directy to DMEM media in 96 deep well storage plates (FroggaBio, catalogue number 4TI-0126). Row G contained DMEM and 0.1 % DMSO, row G contained only DMEM. Columns seven and eight contained two-fold serial dilution of the first compound and the second compound alone, respectively, from row A to F. For each concentration of AKT inhibitor, one microliter of 1000X AKT inhibitor was added to the corresponding well already containing the final dilution of Compound 79 in DMEM media. An example of the compound dilution plate layout for Compound 79 and an AKT inhibitor is shown below at Table O.
Table O. Example of compound dilution plate layout for Compound 79
Figure imgf000315_0001
Figure imgf000316_0001
Cell treatment : Culture media was removed from the 96 well cell plate by aspiration and 100 μL of the drug combination dilutions was added to the cell plate, in triplicate for each treatment (written in simplicate in Table O below for easier read). A total of two 96-well plates were needed for each combination, as outlined below. Plates containing the cells with compounds were incubated at 37°C, 5% CO2 for 3 days. The compound dilution plate was stored at -20°C to re-use at Day 4 to re-treat the cells for an additional four days (total of 7 day incubation). Table P. Cell Plate Scheme:
Figure imgf000316_0002
PLATE #2
Figure imgf000317_0001
Figure imgf000318_0001
After 3 days, media was aspirated from each 96-well plate and compound treatment was repeated as done previously using the same compound dilution plate(s) made previously on Day 1.
Measurement of cell viability and calculation of Bliss synergy score: On Day 7, add 100 μL per well of pre-warmed Cell Titer Gio reagent (Promega, catalogue number G8462). Mix on shaker for 3 to 5min. Luminescence was recorded on a Synergy H4 reader (Biotek) using the manufacturer’s instructions. Background luminescence was removed by substracting the luminescence recorded for the well with no cells from each well. Percentage viability was calculated as a percentage of the DMSO control wells. To calculate the synergy scores, the data was uploaded on the SynergyFinder™ Version 2.0 web site following the site’s instructions choosing the options of viability readout and matrix configuration to calculate the Bliss synergy scores for each combination. The Bliss model assumes a stochastic process in which two drugs elicit their effects independently, and the expected combination effect can be calculated based on the probability of independent events. Generally, a score between -10 to less than 10 may be interpreted or predict that the two drugs are additive, and synergistic if the score is greater or equal to 10, if the two drugs are independent. In the case of two inhibitors acting in the same or redundant pathways, the synergy score might underestimate the combination synergy and other methods such as measurement of tumor growth inhibition in a mouse xenograft model might be more accurate.
In the case of SGK inhibition, Bliss synergy scores were used to identify compounds that were predictively additive and synergistic in vitro (e.g., on cancer cell lines) before testing in an in vivo model (e.g., mouse xenograft model).
The Bliss synergy scores for combinations of Compounds 79, 175, and 156 with different inhibitors of the PI3K, AKT, mTOR pathways in different cancer cell lines are shown at:
Figs. 1A and 1 B (combination of Compound 79 with AKT inhibitor Ipatasertib in JIMT-1 breast cancer cells) - Bliss synergy score of 26; Figs. 2A and 2B (combination of Compound 79 with AKT inhibitor Capivasertib in JIMT-1 breast cancer cells) - Bliss synergy score of 26;
Figs. 3A and 3B (combination of Compound 79 with AKT inhibitor MK-2206 (allosteric) in JIMT-1 breast cancer cells) - Bliss synergy score of 18;
Figs. 7A-7B and 8A-8B (combination of Compounds 156 and 175 with AKT inhibitor Ipatasertib or PIK3CA inhibitor Alpelisib in JIMT-1 breast cancer cells);
Figs. 9A-9C (combination of Compounds 79, 156 and 175 with PIK3CA inhibitor Alpelisib in BT-549 breast cancer cells);
Figs. 10A-10D (combination of Compound 79 with the mTOR inhibitor Everolimus, the HER2/EGFR inhibitor Lapatinib, the multi-AGC kinase inhibitor AT13148, or the mTOR/PI3K inhibitor Gedatolisib on the growth of JIMT-1 breast cancer cells); Figs. 11 A-11C (combination of Compound 79 with the mTOR inhibitor Everolimus, the PIK3a/b/g inhibitor BAY-1082439, or the MEK inhibitor GSK112212 on the growth of BT-549 breast cancer cells);
Figs. 12A-12C (combination of Compounds 79, 156, or 175 with the PIK3CA inhibitor Alpelisib in HT-29 colorectal cancer cells) - Bliss synergy score of 23.5 for compound 79;
Figs. 13A-13C (combination of Compounds 79, 175, or 156 with the BRAF inhibitor Dabrafenib on the growth of HT-29 colorectal cancer cells - Bliss synergy score of 4.06 for compound 79;
Figs. 14A-14D (combination of Compound 79 with the PI3K/mTOR inhibitor Gedatolisib, the MEK inhibitor Trametinib, the EGFR inhibitor Erlotinib, or the PDK1 inhibitor GSK2334470 on the growth of HT-29 colorectal cancer cells - Bliss synergy score of 8.25 for trametinib, and 2.57 for erlotinib;
Figs. 15A-15B (combination of Compound 79 with the BRAF inhibitor Dabrafenib or the MEK inhibitor Trametinib on the growth of 8305c anaplastic thyroid cancer cells) - Bliss synergy scores of -3.55 for Dabrafenib and -4.96 for Trametinib;
Figs. 16A-16D and 17A-17F (triple combination of Compound 79 with the BRAF inhibitor Dabrafenib and the MEK inhibitor Trametinib on the growth of 8305c anaplastic thyroid cancer cells) - Bliss synergy score of -3.03 for the triple combination;
Figs. 18A-18B (combination of Compound 79 with the BRAF inhibitor Dabrafenib or the MEK inhibitor Trametinib on the growth of A375 melanoma cancer cells - Bliss synergy scores of 2.6 for Dabrafenib and 4.99 for Trametinib; Figs. 19A-19B (the combination of Compound 79 with the M EK inhibitor Trametinib or the EGFR inhibitor Erlotinib on the growth of SW403 colorectal cancer cells - Bliss synergy score of 0.44 for Trametinib;
Figs. 20A-20B (combination of Compound 79 with the BRAF inhibitor Dabrafenib or the MEK inhibitor Trametinib on the growth of RKO colorectal cancer cells that are intrinsically resistant to Dabrafenib due to an NF1 mutation) - Bliss synergy score of 8.37 for Trametinib; and
Figs. 21 A-21S’ (treatment with compound 79 alone, the PIK3CA inhibitor Alpelisib alone, or in combination of 45 PIK3CA mutant cell lines). Screening of PIK3CA mutated cancer cell lines
Cell lines’, cell lines were grown in a 37°C tissue culture incubator with 5% CO2 in the media outlined in the table below. Cisplatin was used as the reference compound to ensure reproducibility from prior assays.
Table Q: PIK3CA mutated cell lines and culture medium used
Figure imgf000320_0001
Figure imgf000321_0001
Figure imgf000322_0001
Reagents and preparation: general cell culture reagents and plastic were stored and used according to the manufacturer’s instructions: fetal bovine serum (FBS, origin Australia, cat# FND500, ExCell Bio), 96- Well Flat Clear Bottom Black Polystyrene TC-Treated Microplates (Cat# 3340, Corning), CellTiter-Glo® Luminescent Cell Viability Assay (Cat# G7572, Promega), The CellTiter-Glo Buffer was thawed and equilibrated to room temperature prior to use. The lyophilized CellTiter-Glo Substrate was equilibrated to room temperature prior to use. One hundred milliliters of CellTiter-Glo Buffer was transfered into the amber bottle containing CellTiter-Glo Substrate to reconstitute the lyophilized enzyme/substrate mixture to form the CellTiter-Glo Reagent. The reagent was mixed gently by vortexing, swirling or by inverting the contents to obtain a homogeneous solution.
Test articles: the following test articles was used for the assay:
Table R: Test articles used
Figure imgf000322_0002
Table S: Reference Control
Figure imgf000323_0001
Equipment: EnVision™ Multi Label Reader 2104-001 OA, Perkin Elmer (USA), Equip ID: TAREA0020; Countstar™, Inno-Alliance Biotech (USA), Equip ID: BEANA0020/ BEANA0040; Forma™ Series II Water Jacket CO2 Incubator, Thermo Scientific (USA), Equip ID: BEINC0150/ BEINC0190/ BEINC0200/ BEINC0220/ BEINC0260/ BEINC0330; CO2 Incubator, SANYO (Japan), Equip ID: BEINC0060; Biological safety Cabinet, Thermo Scientific (USA), Equip ID: BABSC0160/ BEBSC0170/ BEBSC0180/ BEBSC0240/ BEBSC0250/ BEBSC0270/ BEBSC0320; Clean bench, HDL Apparatus (China), Equip ID: BACLB0390/ BECLB0580/ TACLE0550; Personal Pipettor, Apricot Designs (USA), Equip ID: TAAPR0030; Biomek FXP Laboratory Automation Workstation, Beckman Coulter (USA), Equip ID: BESTA0010; Inverted Microscope, Olympus CKX41SF (Japan), Equip ID: BEMIC0190/ BEMIC0200; Multidrop, Thermo Scientific (USA), Equip ID: BEPFL0010/ BEPFL0020.
Cell culture and plating: cells were recovered and maintained in appropriate culture media listed in Table Q. Cells were harvested during the logarithmic growth period and counted using Count-star. Cell concentrations were adjusted to 4.44x104 cells/mL with respective culture medium. Eighty microliters of cell suspensions were added to two 96-well plates (plates A and B) with the final cell density of 4x103 cells/well, and the plates were incubated overnight in humidified incubator at 37° C with 5% CO2.
Plate reading at TO, compound dilution and treatment: twenty microliters of culture medium were added to each well of plate A for time = 0 (TO) reading, and the plates were equilibrated at room temperature for approximately 30 min. Fifty microliters of CellTiter- Glo reagent were added to each well, and the content was mixed for 5 min on an orbital shaker to induce cell lysis, and further incubated at room temperature for 20 min to stabilize the luminescent signal. The TO luminescence was recorded using an EnVision Multi Label Reader. 10X solution of test article (Top working concentration: 30μM (Compound #79)/10μM (alpelisib)) of test articles were prepared in the appropriate culture media with 3.16-fold serial dilutions to achieve 9 dose levels, as highlighted in Tables T and an example of plate maps are shown in Table U. 10X reference control solutions of cisplatin (Top working concentration: 100 μM) was prepared in media with 3.16-fold serial dilutions. 10 μL of 10X drug solution of both test article and reference control were simultaneously dispensed in each well (triplicate for each drug concentration) of the plate B and incubated for 72 h in the humidified incubator at 37°C with 5% CO2. 70 μL of drug- containing media were aspirated with a liquid handling Biomek FXP, and replaced with 70μL of fresh culture media containing 1X drug solutions to each well, and incubated for another 72h in the humidified incubator at 37°C with 5% CO2.
Figure imgf000324_0001
Figure imgf000325_0001
Notes:
Volumel : Amount of DMSO drug solution left after a volume has been taken
Volume2: Volume of DMSO without drug Volume3: Amount of DMSO drug solution added from next higher concentration
Volume4: Total of volume after culture medium dilution
Volumes: Final volume in 96-well plate
Alpelisib dilution: top working concentration is 10μM with 3.16-fold serial dilutions to achieve 9 dose levels
Stock=1000xC1 3.16-fold serial dilutions in DMSO (C1-C9)
Figure imgf000325_0002
Figure imgf000326_0001
Notes:
Volumel : Amount of DMSO drug solution left after a volume has been taken
Volume2: Volume of DMSO without drug
Volume3: Amount of DMSO drug solution added from next higher concentration
Volume4: Total of volume after culture medium dilution
Volume5: Final volume in 96-well plate
Reference drug (Cisplatin)
Figure imgf000326_0002
Notes:
Volumel : Amount of culture medium drug solution left after a volume has been taken
Volume2: Volume of culture medium without drug
Volume3: Amount of culture medium drug solution added from next higher concentration Volume4: Final volume in 96-well plate
Table U: examples of plate maps
Figure imgf000327_0003
PlateB-1
Figure imgf000327_0001
PlateB-2
Figure imgf000327_0002
Figure imgf000328_0001
E: Empty well containing complete culture medium or PBS
M : Medium control (blank control)
N: Vehicle control with culture medium containing 0.2%DMSO or PBS
C1-C9: Nine concentration levels of test articles
Plate reading at T=144h and data analysis: plates B were equilibrated at room temperature for approximately 30 min. 50 μL of CellTiter-Glo reagent was added to each well, and the content was mixed for 5 min on an orbital shaker to induce cell lysis, and the plates were incubated at room temperature for 20 min to stabilize the luminescent signal. The T=144 luminescence was recorded using an EnVision Multi Label Reader. The data were displayed graphically using GraphPad™ Prism Version 9.5.1. In order to calculate absolute IC50, a dose-response curve were fitted using nonlinear regression model with a sigmoidal dose response. The formula for calculating surviving rate is shown below and the absolute IC50 were calculated according to the dose-response curve generated by GraphPad™ Prism.
The surviving rate (%) = (LumTest article-LumMedium control)/ (LumNone treated- LumMedium control)* 100%.
Combination indices were calculated using the CompuSyn™ software (V1.0), whereby a combination index of less than 1 indicated synergy between compounds, and a score of 1 indicated additivity. Cell lines were also grouped according to the difference in survival rates of the combination of Compound 79 and alpelisib. Group 1 had more than 10% decrease in viability of the combination at <_1 uM of Compound 79 compared to the viability of either single agents. Group 2 had between 5% and 10% decrease in viability of the combination at <_1 uM of Compound 79 compared to the viability of either single agents. Group 3 had less than 10% decrease in viability of the combination compared to single agents at any concentration tested. Group 4 had more than 10% decrease in viability of the combination at more than 1 uM of Compound #79. Groups 1 and 3 represented the cell lines with the best and weakest combination synergies between Compound 79 and alpelisib respectively. Table V summarizes the grouping of the different cells lines that was obtained. Table V: Grouping of the 45 PIKC3A mutant cell lines according to the combinatorial activity observed. Cell lines with available RNA-Seq data are highlighted in bold.
Figure imgf000329_0001
Geneset enrichment analysis (GSEA): Cell lines with available gene expression data from Group 1 (best synergy) vs. Group 3 (no synergy) were compared by GSEA. Transcript per million (TPM) values of the protein coding genes for DeμMap cell lines were downloaded from the Cancer Cell Line Encyclopedia (CCLE_expression, 22Q1). Genes with an expression value < 0.1 across all cell lines were removed from the dataset. GSEA was done comparing Group 1 versus Group 3 using the software version 4.3.2 downloaded from the Broad Institute using the default parameters. A false discovery rate (FDR) p < 0.05 was considered significant. Cell line mutation data was obtained from the DeμMap data portal (https://depmap.org/portal/) as well as the Cellosaurus database. GSEA plots were generated by the software. Graphical display and additional data analysis was done using GraphPad Prism Version 9.5.1 , and the Mann-Whitney statistical test for group comparisons.
Conversion of gene signatures into activity scores: For each gene of the signature, a Z- score for each cell line was determined using all the cell lines that were used in the screening, and for which the RNA-Seq data was available. For each cell line, the activity score was calculated by adding the Z-scores for all the genes.
Table V shows the RAS/MAPK pathway activating mutations identified in the PIK3CA mutant cell lines that were screened with he Compound 79 and alpelisib combination divided in Groups 1-4 according to their synergy activities.
Fig. 21 shows the proliferation results obtained from the 45 PIK3CA mutant cell lines screened with Compound 79, alpelisib, and the combination of Compound 79 and alpelisib. These data suggest that compound 79 can be used either alone or in combination with a PIK3CA inhibitor for the treatment of multiple cancers. For some cancers, compound 79 was more effective in inhibiting proliferation than alpelisib.
Fig. 22 shows that Compound 79 synergizes with alpelisib (PIK3CA inhibitor) in several cancer cell lines originating from different tissues defined by a combination index of less than 1 . A combination index could not be calculated for all cell lines. For colorectal cancer cell lines, 10/12 cell lines showed synergy, and 9/14 were classified in either Groups 1 or 2. For breast cancer cell lines expressing the estrogen receptor, HER2, or androgen receptor defining luminal breast cancer (lum. breast), 8/10 cell lines did not display synergy when Compound 79 and alpelisib were combined and were classified in Group 3.
Fig. 23 shows that cell lines with mutations activating the RAS/MAPK pathway (mutations in KRAS, HRAS, BRAF, NF1 , NF2, CIC, ARAF, MAP2K1 , SOS1 , or RASA2 genes) were significantly associated with synergy between Compound 79 and alpelisib in inhibiting proliferation (p-value = 0.003).
Figs. 24A-24D show that luminal breast cancer signatures were significantly enriched in cell lines from Group 3 that did not show synergy between Compound 79 and alpelisib when compared to cell lines from Group 1 .
Figs. 25A-25E shows that up-regulation of ribosomal signatures are enriched in cell lines from Group 1 that show synergy when Compound 79 and alpelisib are combined compared to cell lines from Group 3.
Figs. 26A-26B show that incubation of the HT-29 colorectal cancer cell line with either Compound 79 or alpelisib results in down-regulation of the phosphorylated S6 protein (pS6), and that their combination further reduces its expression at both 4 hours and 24 hours. Furthermore, incubation of HT-29 cells with Compound 79 results in a dose- dependent inhibition of the pS6.
Figs. 27A-27B show that the Kegg-Ribosome and Reactome_Translation_initiation activity scores are significantly higher in cell lines from Group 1 that is composed mainly of cell lines with RAS/MAPK activating mutation, and that the two cell lines with RAS/MAPK activating mutations in Group 3 have a lower activity scores for both signatures.
Figs. 28A-28D and Table W show the curated genesets from MsigDB that were significantly upregulated in Group 1 vs. Group 3 cell lines.
Table W: curated genesets from MsigDB that were significantly upregulated in
Group 1 vs. Group 3 cell lines
Figure imgf000332_0001
Figure imgf000333_0001
Table Z: oncogenic signatures genesets from MsigDB that were significantly upregulated in Group 1 vs. Group 3 cell lines
Figure imgf000334_0001
In vivo Mouse xenograft studies
8305C (CL-01029) cancer cells were maintained in vitro with M EM medium supplemented with 1% non-essential amino acids (NEAA), 10% FBS, Sodium pyruvate, 2mM Glutamine at 37°C in an atmosphere of 5% CO2 in the air. The cells in exponential growth phase were harvested and quantitated by cell counter before tumor inoculation. Each mouse was inoculated subcutaneously in the right upper flank region with 8305C tumor cells (5 x 105) in 0.1 ml of PBS for tumor development. Randomization started when the mean tumor size reached approximately 117 mm3. Thirty-two mice were enrolled in the study, randomly allocated to 4 study groups, 8 mice in each group. Randomization was performed based on “Matched distribution” method/ “Stratified” method (StudyDirector™ software, version 3.1.399.19)/randomized block design. The date of randomization was denoted as day 0. The treatment was initiated on the same day of randomization (day 0) per study design. After tumor cells inoculation, the animals were monitored daily for morbidity and mortality. During routine monitoring, the animals were monitored for any effects of tumor growth and treatments on behavior such as mobility, food and water consumption, body weight gain/loss (Body weights were measured twice per week after randomization), eye/hair matting and any other abnormalities. T umor volumes were measured twice per week after randomization in two dimensions using a caliper, and the volume was expressed in mm3 using the formula: “V = (L x W x W)/2, where V is tumor volume, L is tumor length (the longest tumor dimension) and W is tumor width (the longest tumor dimension perpendicular to L). Dosing as well as tumor and body weight measurements were conducted in a Laminar Flow Cabinet. The body weights and tumor volumes were recorded using StudyDirector™ software (version 3.1.399.19). No mortality or clinical signs related to dosing were observed. Figs. 29A-29D show the Anaplastic thyroid cancer (ATC) tumor xenograft model using 8305c cells treated with Compound 79 (45 mg/kg QD), BRAF inhibitor dabrafenib (30 mg/kg QD), MEK inhibitor trametinib (0.6 mg/kg QD) or the triple combination showing tumor volume (Fig. 29A), total body weight (Fig. 29B), tumor response (waterfall pit) (Fig, 29C), and tumor volumes in individual mice after treatment with the triple combination. These data suggest that the triple combination of Compound 79, dabrafenib, and trametinib decreased tumor volumes in comparison to dabrafenib and trametinib, and was shown to be well tolerated by the mice.
Fig. 30A shows the tumor volumes of the Anaplastic thyroid cancer (ATC) tumor xenograft model using 8305c cells, whereby mice were treated with BRAF inhibitor dabrafenib (30 mg/kg QD) and MEK inhibitor trametinib (0.6 mg/kg QD) at Day 0 and Compound 79 (45 mg/kg QD) at Day 13. Fig. 30B shows the tumor volumes of the same model as Fig. 30A whereby mice were treated with Compound 79 (45 mg/kg QD) at Day 0 and BRAF inhibitor dabrafenib (30 mg/kg QD) and MEK inhibitor trametinib (0.6 mg/kg QD) at Day 13. Fig. 30C shows the tumor waterfall of the different groups of the model of Figs. 30A and 30B.
These data further support the finding that compound 79 enhances the anticancer effect of the combination of dabrafenib and trametinib. These data suggest that the addition of Compound 79 causes tumor regression and delays resistance to a combination of dabrafenib and trametinib in a xenograft model of anaplastic thyroid cancer using 8305c cells.
EXAMPLE 6: Phosphoproteomic screen in PBMCs
Cell Thawing, count and seeding: Cryopreserved human peripheral blood mononuclear cells (PBMCs, ZenBio, SER-PBMC-200P-F) were taken from liquid nitrogen storage and immediately placed it into a 37°C water bath. The cells were quickly thawed by gently swirling the vial in the 37°C water bath and the whole content was pipetted into the pre- filled sterile tube with 20 ml pre-warmed cell culture medium (RPMI, ATCC 30-2001 , containing 10% heat-inactivated fetal bovine serum VWR 97068-085 and 1 % penicillin/streptomycin (100X), VWR K952). The suspension was centrifuged at 300 x g for 15 minutes and the cell pellet was resuspended in 30.0 mL of cell culture medium. Cells were counted using a Countess cell counter (Invitrogen, C10281) and trypan blue (ThermoFisher T10282) according to the manufacturer’s instructions and seeded at a density of 88 million cells (4 millions/ml) per 25 cm flask in culture medium and incubated overnight at 37°C.
Cell Treatment and Phosphoarray: 1.09 ul of Compound 79 20 mM stock was added in one T-25 flask containing PBMCs, and 1.09 μL of DMSO was added to a second T-25 flask as a vehicle control. The flasks were mixed by swirling 5-10 times, and incubated at 37°C, 5%CO2 for 4 hrs. The cell suspension was transferred in a 50 mL tubes (1 treatment per tube) and the flask was rinsed with 5 mL of cold 1X PBS and combine to the 50 ml Tube containing cells and centrifuged at 300 x g for 10 minutes. The cell pellet was resuspended with 10mL of cold 1X PBS and centrifuged 300 x g for an additional 10 minutes. The pellets were lysed and processed according to the C-Series Human Phosphorylation Multi- Pathway Profiling Array C55 (RayBiotech, AAH-PPP-1-4) instructions. One hundred and fifty micrograms of each PBMC extract sample was incubated for each membrane previously blocked in the provided buffer and incubated overnight at 4°C with gentle shaking as instructed by the manufacturer. Detection was done on a ChemiDoc™ MP imaging system (Bio-Rad, 12003154) and quantification was done according to the instructions and as detailed below.
Data extraction and analysis: The RayBiotech™ analysis software tools was used for automatic analysis of the results. Basically, the algorithm below was used to calculate and determine the signal fold expression between like analytes.
X(Ny) = X(y) * P1/P(y)
Where:
P1 = mean signal density of Positive Control spots on the reference array P(y) = mean signal density of Positive Control spots on Array "y" X(y) = mean signal density for spot "X" on Array "y" X(Ny)= normalized signal intensity for spot "X" on Array "y"
Quantified data was processed and graphed using GraphPad Prism version 9.5.1.
A phosphoproteomic screen was used to evaluate the potential targets for compound 79. Figs. 31A-31 B shows the phosphoproteomic array of peripheral blood mononuclear cells (PBMCs) incubated with 1 uM of Compound 79 for 4 hours. As shown, the phosphorylated protein expression of p38, HSP27, TP53, RSK1 , and PRAS40 kinases were increased by greater than 50% (outside grey shaded area) upon treatment with compound 79. These data suggest phosphorylation of specific kinases may be used to track the progress of the anticancer treatment by the compounds described herein.
EXAMPLE 7: Effect of the compounds on hyperglycemia
Blood glucose studies in mice
Animal maintenance: C57BL/6 mice were quarantined for at least 7 days before the study. The general health of the animals was evaluated by a veterinarian, and complete health checks were performed. Animals with abnormalities were excluded prior the study. General procedures for animal care and housing were in accordance with the standard, Commission on Life Sciences, National Research Council, Standard operating procedures (SOPs). The mice were kept in laminar flow rooms at constant temperature and humidity with two animals in each cage. Animals were housed in polycarbonate cages and in an environmentally monitored, well-ventilated room maintained at a temperature of (22 ± 3°C) and a relative humidity of 40%-80%. Fluorescent lighting provided illumination approximately 12 hours per day. The bedding material was soft wood, which was changed once per week. Analysis of bedding contamination was conducted annually per facility SOPs and the results of the analyses were retained in the facility records. Each animal was marked with an identification number on the tail of the animal with marker pen; the following identification method was applied. Each cage card was labeled with such information as study number, group, sex, dose, animal number, initiation date, study director and telephone number. Animals had free access to normal diet during the entire study period except for time periods specified by the protocol. Tap water was treated with water purification system and then autoclaved. Sterile drinking water in a bottle is available to all animals’ ad libitum during the quarantine and study periods. The bottle and the stopper with attached sipper tube was autoclaved prior to use. Samples of water from the animal facility were analyzed and results of water analysis were retained in the facility records and were reviewed by the veterinarian, or designee, to assure that no known contaminants are present that could interfere with or affect the outcome of studies. Groups and treatments: Based on the body weights, animals were randomly assigned to respective groups. The study groups and number of animals per group are shown in Table AA, BB and CC.
Table AA: Groups and Treatments Experiment 1
Figure imgf000338_0001
Table BB: Groups and Treatments Experiment 2
Figure imgf000338_0002
Table CC: Groups and Treatments Experiment 3
Figure imgf000338_0003
Figure imgf000339_0001
Formulations: Alpelisib (MedChemExpress, HY-1524): 0.5% carboxymethyl cellulose (Sigma, SLBS9559), MK-2206 (MedChemExpress, HY-108232): 30% captisol (Cydex, NC-04A-180185), Compound 79 (lot number EB2137150-056P1): 0.5% HEC (Sigma, 9004-62-0), 0.4% tween-80 (Sigma, BCCC9754) in 30 mM Citrate Buffer (pH 3.0±0.1).
Preparation of test article and vehicle are shown in Table DD.
Table DD: Formulation (Example)
Figure imgf000339_0002
Figure imgf000340_0001
Body Weight and clinical observations: Body weights of all animals were measured daily during the study.Clinical observation and cage side observation were performed and recorded daily pre- dose and after treatment. The detailed information is listed in Table EE.
Table EE: Clinical Observation
Figure imgf000340_0002
Figure imgf000341_0001
Figure imgf000342_0001
Measurement of fed glucose and blood collection: Fed blood glucose level of all animals were measured via tail vein on the last day of dosing by using Accu-Chek Performa System at 0, 30, 60, 90, 120, 150, 180, & 240 min. Blood samples were collected on the last day of dosing from study animals at 240 min post-dose and put into tubes without any anticoagulant. The blood samples were centrifuged at 4°C, 8000 g for 15 min, and serum samples were transferred into another sample tube and kept at -80°C for C- peptide, insulin, or glucagon analysis.
Statistical Analysis: All statistical analysis was conducted, and the level of significance was set at P<0.05. The group means, standard deviation was calculated for all measurement parameters as study designed. One way ANOVA comparisons among the groups were performed with GraphPad Prism 8.0.
In many anticancer standard of care treatments, such as the PIK3CA inhibitor Alpelisib or the AKT inhibitor MK-2206, patients typically develop hyperglycemia, hyperinsulinemia, increased ALT/AST, pyrexia, rash, diarrhea, nausea, fatigue, thrombocytopenia, anemia, dermatitis acneiform, QT elongation, and/or dyspnea, which may force a pause or change of treatment. Furthermore, patients with existing hyperglycemia or any of other conditions mentioned above, or risk thereof, may not be eligible for certain anticancer treatments. Therefore, there is a need for anticancer treatments that do not increase, or rather lower, blood glucose levels and hyperglycemia, as well as mitigate the toxicity/side effects of current anticancer treatments. Figs. 32A and 32B show the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof. Figs. 32C and 32D show the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof. Figs. 32E and 32F shows the results of mice blood glucose levels at different time points after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (5, 15, or 45 mg/kg mg/kg), or combinations thereof.
Figs. 33A-33B shows the results of mice blood C-peptide levels (Fig. 33A) and insulin levels (Fig. 33B) after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof. Fig. 33C shows the results of mice blood C-peptide after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof. Figs. 33D- 33E shows the results of mice blood C-peptide levels (Fig. 33D) and insulin levels (Fig. 33E) after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (5, 15, or 45 mg/kg), or combinations thereof. Fig. 33F shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (45 mg/kg), or combinations thereof. Fig. 33G shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (25, 50, or 100 mg/kg), or combinations thereof. Fig. 33H shows the results of mice blood glucagon levels after treatment with a single dose of the PIK3CA inhibitor Alpelisib (45 mg/kg), the AKT inhibitor MK-2206 (120 mg/kg), Compound 79 (5, 15, or 45 mg/kg), or combinations thereof.
These data suggest that the compounds described herein provide a further advantage other than directly decreasing tumor growth in that they can decrease or maintain a normal blood glucose level, and subsequently decrease the risk of developing hyperglycemia or other side effects caused by current anticancer treatments during treatment. All publications, patents and patent applications are incorporated by reference in their entirety, as though individually incorporated by reference. The invention has been described with reference to various specific and preferred embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the invention.

Claims

1. A compound of Formula VII:
Formula VII or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer or tumor or in the manufacture of a medicament for the treatment of cancer or tumor, wherein:
Y1 is H or F; Z1 and Z2 are independently from one another selected from the group consisting of halogen, (C1-C4)alkyl, -OH, -O-(C1-C4)alkyl, -CF3, and -CN;
W1 is selected from the group consisting of H and halogen; and
R35 is H or methyl.
2. The compound or pharmaceutically acceptable salt thereof for use of claim 1 , wherein Y1 is H.
3. The compound or pharmaceutically acceptable salt thereof for use of claim 1 , wherein Y1 is F.
4. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 3, wherein Z1 is F and Z2 is F.
5. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 3, wherein Z1 is Cl and Z2 is F.
6. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 5, wherein W1 is H.
7. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 5, wherein W1 is Cl.
8. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 5, wherein W1 is F.
9. The compound 1 to 8 or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 8, wherein R35 is H.
10. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 8, wherein R35 is methyl.
11. The compound or pharmaceutically acceptable salt thereof for use of claim 1 , which is a compound of Formula Vila: wherein:
Figure imgf000346_0001
Z1 is F or Cl;
Z2 is F; and
W1 is H, Cl or F.
12. The compound for use of claim 1, which is:
, or a pharmaceutically acceptable salt thereof.
Figure imgf000346_0002
13. The compound or pharmaceutically acceptable salt thereof for use of claim 1 , which is a compound of Formula VI lb:
Formula VI lb wherein: Z1 is F or Cl;
Z2 is F;
R35 is H or methyl; and
W1 is H, Cl or F.
14. The compound or pharmaceutically acceptable salt thereof for use of claim 13, wherein R35 is H.
15. The compound or pharmaceutically acceptable salt thereof for use of claim 13, wherein R35 is methyl.
16. The compound for use of claim 1 , which is: or a pharmaceutically acceptable salt thereof.
17. The compound for use of claim 1 , which is: or a pharmaceutically acceptable salt thereof.
18. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 17, in combination with at least one inhibitor of AKT, PI3K, and/or mTOR.
19. The compound or pharmaceutically acceptable salt thereof for use of claim 18, wherein the at least one inhibitor of AKT, PI3K, and/or mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI-587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP- BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK-1117, MLN-1117, TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC-907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-100- 115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
20. The compound or pharmaceutically acceptable salt thereof for use of claim 18 wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
21. The compound or pharmaceutically acceptable salt thereof for use of claim 18, wherein the at least one inhibitor of AKT, PI3K, and/or mTOR is selected from the group consisting of vistusertib (AZD2014), NU7441 (KU-57788), KU-0063794, TGX-221 , RLY- 2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223, ABTL0812, DCBCI0901, AZD-8055, sapanisertib, JR-AB2-011 , omipalisib, and torkinib (PP242), and PI-103.
22. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 21 , in combination with at least one inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK.
23. The compound or pharmaceutically acceptable salt thereof for use of claim 22, wherein the at least one inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK is selected from the group consisting of dabrafenib, trametinib (GSK1120212), encorafenib (LGX818), binimetinib, cobimetinib, naporafenib (LXH254), sorafenib (BAY 43-9006), vemurafenib (PLX4032), PLX-4720, regorafenib (BAY 73-4506), GDC_0879, RAF265 (CHIR-265), AZ 628, lifirafenib (BGB-283), NVP- BHG712, SB590885, ZM 336372, GW5074, TAK-632, agerafenib (RXDX-105), GNE- 9815, TBAP-001 , L-779450, belvarafenib (HM95573), AZ304, PLX8394, RAF709, CCT196969, BAW2881 (NVP-BAW2881), LY3009120, avutometinib (RO5126766, CH5126766), doramapimod (BIRB 796), MCP110, PLX7904, tovorafenib (MLN2480), selumetinib (AZD6244), mirdametinib (PD0325901), U0126, PD98059, PD184352 (CI- 1040), BIX 02189, pimasertib (AS-70326), pelitinib (EKB-569), BIX 02188, TAK-733, AZD8330, binimetinib (MEK162), SL-327, refametinib (RDEA119), zapnometinib (PD0184264), GDC-0623, BI-847325, cobimetinib (GDC_0973), PD318088, SCH772984, ASN007, MRTX-1257, AZD0634 (ATG-017), MK-8353 (SCH900353), temuterkib (LY3214996), VX-11e, ulixertinib (BVD-523), ravoxertinib (GDC-0994), exarafenib, ERAS- 601 , BDTX-4933, ADT007, HM99462, DCC-3084, PHI-501 , MRTX1133. Sotorasib (AMG510), lonafamib (SCH66336), ASP2453, BI-3406, BI-2852, BAY-293, KRpep-2d, ARS-853, adagrasib (MRTX849), adagrasib (MRTX849), ARS1620, divarasib (GDC- 6036), GDC-1971, IACS-13909, JAB-3068, RMC-4550, RMC-4630, SHP099, TNO155, GDC-1971 , HBI-2376, BBP-398 (IACS-15509), PF-07284892, selpercatinib (LOXO-292), pralsetinib (BLU-667), LOXO-260, TAS-0953/HM06, TPX-0046, EP0031 , APS03118, TP- 0903, BGB324 (R428), bosutinib (SKI-606), and gilteritinib (ASP2215).
24. The compound or pharmaceutically acceptable salt thereof for use of claim 22 or 23, wherein the at least one RTK inhibitor is selected from the group consisting of epidermal growth factor receptor (EGFR) inhibitors erlotinib, osimertinib, neratinib, gefitinib, cetuximab, panitumumab, dacomitinib, lapatinib, necitumumab, mobercitinib, vandetanib, sapitinib, afatinib, canertinib, CP-724714, CUDC-101 , mubritinib (TAK 165), OSI-420, pelitinib, irbinitinib (ARRY-380, ONT-380), varlitinib, TAK-285, BIBX 1382, PD168393, raciletinib, poziotinib, CL-387785, icotinib, CTX-2006, WHI-P154, AZ5104, AZD3759, Erb-B2 receptor tyrosine kinase 2 (HER2) such as neratinib, trastuzumab and its derivatives such as trastuzumab deruxtecan, lapatinib, dacomitinib, tucatinib, pertuzumab, margetuximab, afatinib, AZD8931 , AST1306, AEE788, canertinib, CP724.714, CUDC101, TAK285, dacomitinib, pelitinib, AC480, canertinib, sapitinib, mubritinib (TAK 165), BDTX-189, epertinib, TAS0728, poziotinib, multikinase inhibitors cabozantinib, imatinib, Lenvatinib, regorafenib, sorafenib, sunitinib, vandetinib, MET inhibitors crizotinib, cabozantinib, foretinib, PHA-665752, SU11274, SGX-523, BMS- 777607, tivantinib (ARQ 197), JNJ-38877605, PF-04217903, MGCD-265, capmatinib (INCB28060), BMS-754807, BMS-794833, AMG-208, MK-2461 , golvatinib (E7050), AMG-458, NVP-BVU972, XL092, UNC2025, elzovantinib (TPX-0022), AMG-1 , JNJ- 38877618 (OMO-1), altiratinib, SAR125844, glumetinib (SCC244), savolitinib (AZD6094), RXDX-106 (CEP-40783), S49076, merestinib (LY2801653), AMG 337, tepotinib, neurotrophic receptor tyrosine kinase 1 (NTRK1) inhibitors larotrectinib, entrectinib, danusertib, BMS-754807, GW441756, UNC2025, taletrectinib, altiratinib, selitrectinib (LOXO-195), CH7057288, BMS-935177, PF-06273340, sitravatinib (MGCD516), GNF- 5837, vascular endothelial growth factor receptor inhibitors bevacizumab, ramucirumab, SU5408, linifanib (ABT-869), axitinib (AG 013736), nintedanib (BIBF 1120), cediranib (AZD2171), motesanib (AMG-706), pazopanib (GW786034), vandetanib, sunitinib (SU11248), sorafenib (BAY 43-9006), brivanib (BMS-540215), vatalanib (PTK787), foretinib (GSK1363089), cabozantinib (BMS-907351), brivanib (BMS-582664), Lenvatinib (E7080), CYC116, regorafenib (BAY 73-4506), ENMD-2076, tivozanib, ponatinib (AP24534), apatinib (YN968D1), telatinib, PP121, pazopanib, dovitinib, SAR131675, semaxanib (SU5416), golvatinib (E7050), fibroblast growth factor receptor inhibitors ponatinib (AP24534), infigratinib (BGJ398), nintedanib (BIBF 1120), PD173074, AZD4547, dovitinib, pemigatinib, futibatinib, infigratinib, and RLY-4008.
25. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 24, wherein the cancer or tumor comprises cells having an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation.
26. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 25, wherein the cancer or tumor comprises cells having an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer.
27. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 26, wherein the cancer or tumor comprises cells having a downregulation of gene signatures associated with luminal breast cancer.
28. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 27, wherein the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway.
29. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 27, wherein the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIO.
30. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 28, wherein the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by one or more concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN.
31. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 30, wherein the cancer or tumor comprises cells having upregulation of a mitogen activated protein kinase (MAPK) gene expression.
32. The compound or pharmaceutically acceptable salt thereof for use of claim 31 , wherein the MAPK gene expression signature comprises CCND1 , DLISP4, DLISP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and SPRY4.
33. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 32, wherein said cancer or tumor is resistant to one or more anticancer therapies (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof).
34. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 33, wherein the cancer or tumor affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
35. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 34, wherein the cancer or tumor is a melanoma, liposarcoma, lung cancer (e.g., non-small cell lung cancer), breast cancer (e.g., ER+, ER-, HER2+, HER2-, PR-, PR+, triple negative, luminal, basal), Luminal androgen receptor (LAR)) prostate cancer, leukemia, kidney cancer, esophageal cancer, thyroid cancer (e.g., Anaplastic Thyroid Cancer (ATC)), ovarian cancer, endometrial cancer, Head and Neck Squamous Cell Carcinoma, brain cancer, lymphoma, rectal cancer, colon cancer, uterine cancer, cervical cancer, Hepatocellular carcinoma, Papillary thyroid carcinoma (PTC), or colorectal cancer.
36. The compound or pharmaceutically acceptable salt thereof for use of any one of claims 1 to 35, wherein the cancer or tumor is prostate cancer, colorectal cancer or breast cancer.
37. A pharmaceutical composition comprising one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, for use in the treatment of cancer or tumor or in the manufacture of a medicament for the treatment of cancer or tumor.
38. The pharmaceutical composition for use of claim 37, further comprising at least one inhibitor of AKT/PI3K/mTOR.
39. The pharmaceutical composition for use of claim 38, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI-587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK-1117.MLN- 1117.TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC-907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-100-115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
40. The pharmaceutical composition for use of claim 38,, wherein the at least one inhibitor of AKT.PI3K, and/or mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
41. The pharmaceutical composition for use of claim 38, wherein the at least one inhibitor of AKT, PI3K, and/or mTOR is selected from the group consisting of vistusertib (AZD2014), NU7441 (KU-57788), KU-0063794, TGX-221 , RLY-2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223, ABTL0812, DCBCI0901 , AZD-8055, sapanisertib, JR- AB2-011 , omipalisib, and torkinibm PI-103.
42. The pharmaceutical composition for use of any one of claims 37 to 41 , further comprising at least one inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK.
43. The pharmaceutical composition for use of claim 42, wherein the at least one inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK is selected from the group consisting of dabrafenib, trametinib (GSK1120212), encorafenib (LGX818), binimetinib, cobimetinib, naporafenib (LXH254), sorafenib (BAY 43-9006), vemurafenib (PLX4032), PLX-4720, regorafenib (BAY 73-4506), GDC_0879, RAF265 (CHIR-265), AZ 628, lifirafenib (BGB-283), NVP-BHG712, SB590885, ZM 336372, GW5074, TAK-632, agerafenib (RXDX-105), GNE-9815, TBAP-001 , L-779450, belvarafenib (HM95573), AZ304, PLX8394, RAF709, CCT196969, BAW2881 (NVP- BAW2881), LY3009120, avutometinib (RO5126766, CH5126766), doramapimod (BIRB 796), MCP110, PLX7904, tovorafenib (MLN2480), selumetinib (AZD6244), mirdametinib (PD0325901), U0126, PD98059, PD184352 (CI-1040), BIX 02189, pimasertib (AS- 70326), pelitinib (EKB-569), BIX 02188, TAK-733, AZD8330, binimetinib (MEK162), SL- 327, refametinib (RDEA119), zapnometinib (PD0184264), GDC-0623, BI-847325, cobimetinib (GDC_0973), PD318088, SCH772984, ASN007, MRTX-1257, AZD0634 (ATG-017), MK-8353 (SCH900353), temuterkib (LY3214996), VX-11e, ulixertinib (BVD- 523), ravoxertinib (GDC-0994), exarafenib, ERAS-601 , BDTX-4933, ADT007, HM99462, DCC-3084, PHI-501 , MRTX1133. Sotorasib (AMG510), lonafamib (SCH66336), ASP2453, BI-3406, BI-2852, BAY-293, KRpep-2d, ARS-853, adagrasib (MRTX849), adagrasib (MRTX849), ARS1620, divarasib (GDC-6036), GDC-1971 , IACS-13909, JAB- 3068, RMC-4550, RMC-4630, SHP099, TNO155, GDC-1971 , HBI-2376, BBP-398 (IACS- 15509), and PF-07284892.
44. The pharmaceutical composition for use of claim 42 or 43, wherein the at least one RTK inhibitor is selected from the group consisting of epidermal growth factor receptor (EGFR) inhibitors erlotinib, osimertinib, neratinib, gefitinib, cetuximab, panitumumab, dacomitinib, lapatinib, necitumumab, mobercitinib, vandetanib, sapitinib, afatinib, canertinib, CP-724714, CUDC-101 , mubritinib (TAK 165), OSI-420, pelitinib, irbinitinib (ARRY-380, ONT-380), varlitinib, TAK-285, BIBX 1382, PD168393, raciletinib, poziotinib, CL-387785, icotinib, CTX-2006, WHI-P154, AZ5104, AZD3759, Erb-B2 receptor tyrosine kinase 2 (HER2) such as neratinib, trastuzumab and its derivatives such as trastuzumab deruxtecan, lapatinib, dacomitinib, tucatinib, pertuzumab, margetuximab, afatinib, AZD8931 , AST1306, AEE788, canertinib, CP724.714, CUDC101 , TAK285, dacomitinib, pelitinib, AC480, canertinib, sapitinib, mubritinib (TAK 165), BDTX-189, epertinib, TAS0728, poziotinib, multikinase inhibitors cabozantinib, imatinib, Lenvatinib, regorafenib, sorafenib, sunitinib, vandetinib, MET inhibitors crizotinib, cabozantinib, foretinib, PHA- 665752, SU11274, SGX-523, BMS-777607, tivantinib (ARQ 197), JNJ-38877605, PF- 04217903, MGCD-265, capmatinib (INCB28060), BMS-754807, BMS-794833, AMG-208, MK-2461 , golvatinib (E7050), AMG-458, NVP-BVU972, XL092, UNC2025, elzovantinib (TPX-0022), AMG-1 , JNJ-38877618 (OMO-1), altiratinib, SAR125844, glumetinib (SCC244), savolitinib (AZD6094), RXDX-106 (CEP-40783), S49076, merestinib (LY2801653), AMG 337, tepotinib, neurotrophic receptor tyrosine kinase 1 (NTRK1) inhibitors larotrectinib, entrectinib, danusertib, BMS-754807, GW441756, UNC2025, taletrectinib, altiratinib, selitrectinib (LOXO-195), CH7057288, BMS-935177, PF- 06273340, sitravatinib (MGCD516), GNF-5837, vascular endothelial growth factor receptor inhibitors bevacizumab, ramucirumab, SU5408, linifanib (ABT-869), axitinib (AG 013736), nintedanib (BIBF 1120), cediranib (AZD2171), motesanib (AMG-706), pazopanib (GW786034), vandetanib, sunitinib (SU11248), sorafenib (BAY 43-9006), brivanib (BMS- 540215), vatalanib (PTK787), foretinib (GSK1363089), cabozantinib (BMS-907351), brivanib (BMS-582664), Lenvatinib (E7080), CYC116, regorafenib (BAY 73-4506), ENMD-2076, tivozanib, ponatinib (AP24534), apatinib (YN968D1), telatinib, PP121 , pazopanib, dovitinib, SAR131675, semaxanib (SU5416), golvatinib (E7050), fibroblast growth factor receptor inhibitors ponatinib (AP24534), infigratinib (BGJ398), nintedanib (BIBF 1120), PD173074, AZD4547, dovitinib, pemigatinib, futibatinib, infigratinib, and RLY-4008.
45. A combination comprising one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17 and at least one inhibitor of AKT, PI3K, and/or mTOR.
46. The combination of claim 45, wherein the at least one inhibitor of AKT, PI3K, and/or mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC- 0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI-587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK-1117, MLN-1117, TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC-907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-100- 115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
47. The combination of claim 45, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
48. The combination of claim 45, wherein the at least one inhibitor of AKT, PI3K, and/or mTOR is selected from the group consisting of vistusertib (AZD2014), NU7441 (KU- 57788), KU-0063794, TGX-221 , RLY-2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223, ABTL0812, DCBCI0901 , AZD-8055, sapanisertib, JR-AB2-011 , omipalisib, and torkinibm PI-103.
49. A combination comprising one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17 and at least one inhibitor of AKT, PI3K, mTOR, PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK.
50. The combination of claim 49, wherein the at least one inhibitor of AKT, PI3K, and/or mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC- 0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI-587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK-1117, MLN-1117, TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC-907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-1 OO- 115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, rapamycin, (AZD2014), NU7441 (KU-57788), KU-0063794, TGX-221 , RLY- 2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223, ABTL0812, DCBCI0901 , AZD-8055, sapanisertib, JR-AB2-011 , omipalisib, and torkinibm PI-103.
51. The combination of claim 49, wherein the at least one inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK is selected from the group consisting of dabrafenib, trametinib (GSK1120212), encorafenib (LGX818), binimetinib, cobimetinib, naporafenib (LXH254), sorafenib (BAY 43-9006), vemurafenib (PLX4032), PLX-4720, regorafenib (BAY 73-4506), GDC_0879, RAF265 (CHIR-265), AZ 628, lifirafenib (BGB-283), NVP-BHG712, SB590885, ZM 336372, GW5074, TAK-632, agerafenib (RXDX-105), GNE-9815, TBAP-001 , L-779450, belvarafenib (HM95573), AZ304, PLX8394, RAF709, CCT196969, BAW2881 (NVP-BAW2881), LY3009120, avutometinib (RO5126766, CH5126766), doramapimod (BIRB 796), MCP110, PLX7904, tovorafenib (MLN2480), selumetinib (AZD6244), mirdametinib (PD0325901), 110126, PD98059, PD184352 (CI-1040), BIX 02189, pimasertib (AS-70326), pelitinib (EKB-569), BIX 02188, TAK-733, AZD8330, binimetinib (MEK162), SL-327, refametinib (RDEA119), zapnometinib (PD0184264), GDC-0623, BI-847325, cobimetinib (GDC_0973), PD318088, SCH772984, ASN007, MRTX-1257, AZD0634 (ATG-017), MK-8353 (SCH900353), temuterkib (LY3214996), VX-11e, ulixertinib (BVD-523), ravoxertinib (GDC-0994), exarafenib, ERAS-601 , BDTX-4933, ADT007, HM99462, DCC-3084, PHI- 501 , MRTX1133. Sotorasib (AMG510), lonafamib (SCH66336), ASP2453, BI-3406, Bl- 2852, BAY-293, KRpep-2d, ARS-853, adagrasib (MRTX849), adagrasib (MRTX849), ARS1620, divarasib (GDC-6036), GDC-1971 , IACS-13909, JAB-3068, RMC-4550, RMC- 4630, SHP099, TNO155, GDC-1971 , HBI-2376, BBP-398 (IACS-15509), and PF- 07284892
52. The combination of claim 49 or 51 , wherein the at least one RTK inhibitor is selected from the group consisting of epidermal growth factor receptor (EGFR) inhibitors erlotinib, osimertinib, neratinib, gefitinib, cetuximab, panitumumab, dacomitinib, lapatinib, necitumumab, mobercitinib, vandetanib, sapitinib, afatinib, canertinib, CP-724714, CUDC- 101 , mubritinib (TAK 165), OSI-420, pelitinib, irbinitinib (ARRY-380, ONT-380), varlitinib, TAK-285, BIBX 1382, PD168393, raciletinib, poziotinib, CL-387785, icotinib, CTX-2006, WHI-P154, AZ5104, AZD3759, Erb-B2 receptor tyrosine kinase 2 (HER2) such as neratinib, trastuzumab and its derivatives such as trastuzumab deruxtecan, lapatinib, dacomitinib, tucatinib, pertuzumab, margetuximab, afatinib, AZD8931 , AST1306, AEE788, canertinib, CP724.714, CUDC101 , TAK285, dacomitinib, pelitinib, AC480, canertinib, sapitinib, mubritinib (TAK 165), BDTX-189, epertinib, TAS0728, poziotinib, multikinase inhibitors cabozantinib, imatinib, Lenvatinib, regorafenib, sorafenib, sunitinib, vandetinib, MET inhibitors crizotinib, cabozantinib, foretinib, PHA-665752, SU 11274, SGX-523, BMS-777607, tivantinib (ARQ 197), JNJ-38877605, PF-04217903, MGCD-265, capmatinib (INCB28060), BMS-754807, BMS-794833, AMG-208, MK-2461 , golvatinib (E7050), AMG-458, NVP-BVU972, XL092, UNC2025, elzovantinib (TPX-0022), AMG-1 , JNJ-38877618 (OMO-1), altiratinib, SAR125844, glumetinib (SCC244), savolitinib (AZD6094), RXDX-106 (CEP-40783), S49076, merestinib (LY2801653), AMG 337, tepotinib, neurotrophic receptor tyrosine kinase 1 (NTRK1) inhibitors larotrectinib, entrectinib, danusertib, BMS-754807, GW441756, UNC2025, taletrectinib, altiratinib, selitrectinib (LOXO-195), CH7057288, BMS-935177, PF-06273340, sitravatinib (MGCD516), GNF-5837, vascular endothelial growth factor receptor inhibitors bevacizumab, ramucirumab, SU5408, linifanib (ABT-869), axitinib (AG 013736), nintedanib (BIBF 1120), cediranib (AZD2171), motesanib (AMG-706), pazopanib (GW786034), vandetanib, sunitinib (SU11248), sorafenib (BAY 43-9006), brivanib (BMS- 540215), vatalanib (PTK787), foretinib (GSK1363089), cabozantinib (BMS-907351), brivanib (BMS-582664), Lenvatinib (E7080), CYC116, regorafenib (BAY 73-4506), ENMD-2076, tivozanib, ponatinib (AP24534), apatinib (YN968D1), telatinib, PP121 , pazopanib, dovitinib, SAR131675, semaxanib (SU5416), golvatinib (E7050), fibroblast growth factor receptor inhibitors ponatinib (AP24534), infigratinib (BGJ398), nintedanib (BIBF 1120), PD173074, AZD4547, dovitinib, pemigatinib, futibatinib, infigratinib, and RLY-4008.
53. A combination comprising one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17 and ipatasertib, alpelisib, everolimus, lapatinib, AT13148, gedatolisib, BAY-1082439, GSK112212, dabrafenib, trametinib, erlotinib, and/or GSK2334470.
54. A combination comprising at least two compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17.
55. Use of the pharmaceutical composition of any one of claims 37 to 44 or the combination of any one of claims 45 to 54 for the treatment of cancer or tumor or for the manufacture of a medicament for the treatment of cancer or tumor.
56. The use of claim 55, wherein the cancer or tumor comprises cells having an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation.
57. The use of claim 54 or 55, wherein the cancer or tumor comprises cells having an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer.
58. The use of any one of claims 55 to 57, wherein the cancer or tumor comprises cells having a downregulation of gene signatures associated with luminal breast cancer.
59. The use of any one of claims 55 to 58, wherein the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway.
60. The use of any one of claims 55 to 59, wherein the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIO.
61. The use of any one of claims 55 to 60, wherein the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by one or more concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN.
62. The use of any one of claims 55 to 61 , wherein the cancer or tumor comprises cells having upregulation of a mitogen activated protein kinase (MAPK) gene expression.
63. The use of claim 62, wherein the MAPK gene expression signature comprises CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and SPRY4.
64. The use of any one of claims 55 to 63, wherein said cancer or tumor is resistant to one or more anticancer therapies (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof).
65. The use of any one of claims 55 to 64, wherein the cancer or tumor affects tissues comprising cancerous cells in at least one of the breast, prostate, brain, blood, bone marrow, liver, pancreas, skin, kidney, colon, intestine, endometrium, ovary, lung, testicle, penis, thyroid, parathyroid, pituitary, thymus, retina, uvea, conjunctiva, spleen, head, neck, trachea, gall bladder, rectum, salivary gland, adrenal gland, throat, esophagus, lymph nodes, sweat glands, sebaceous glands, muscle, heart, bone, and stomach.
66. The use of any one of claims 55 to 65, wherein the cancer or tumor is a melanoma, liposarcoma, lung cancer (e.g., non-small cell lung cancer), breast cancer (e.g., ER+, ER- , HER2+, HER2-, PR-, PR+, triple negative, luminal, basal), Luminal androgen receptor (LAR)) prostate cancer, leukemia, kidney cancer, esophageal cancer, thyroid cancer (e.g., Anaplastic Thyroid Cancer (ATC)), ovarian cancer, endometrial cancer, Head and Neck Squamous Cell Carcinoma, brain cancer, lymphoma, rectal cancer, colon cancer, uterine cancer, cervical cancer, Hepatocellular carcinoma, Papillary thyroid carcinoma (PTC), or colorectal cancer.
67. The use of any one of claims 55 to 66, wherein the cancer or tumor is prostate cancer, colorectal cancer or breast cancer.
68. A method for the treatment of cancer, comprising administering to a subject in need thereof one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17, the pharmaceutical composition of any one of claims 37 to 44, or the combination of any one of claims 45 to 54.
69. The method of claim 68, wherein the compound of any one of claims 1 to 17 or a pharmaceutically acceptable salt thereof, is administered to the subject in combination with at least one inhibitor of AKT/PI3K/mTOR.
70. The method of claim 69, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of NVP-BEZ235 (BEZ235, Dactolisib), GDC-0084 (RG7666), GDC-0980 (Apitolisib, RG7422), LY3023414, PF-05212384 (Gedatolisib, PKI- 587), PQR309 (Bimiralisib), P7170, SF-1126, Copanlisib (BAY 80-6946), Buparlisib (BKM120 NVP-BKM120), IPI-145 (Duvelisib), RP6530 (Tenalisib), GDC-0032 (Taselisib), KA2237, BYL719 (Alpelisib), CAL-101 (GS-1101 , Idelalisib), GSK2636771 , INCB050465 (Parsaclisib), Serabelisib (INK-1117, MLN-1117, TAK-117), ME401 (PWT-143), Umbralisib (RP5264, TGR-1202), CUDC-907 (Fimepinostat), Rigosertib (ON-01910), samotolisib, paxalisib, voxtalisib, CH5132799, pilaralisib, ZSTK474, sonolisib, pictilisib, B591 , TG-100- 115, RIDR-PI-103, zandelisib, AMG319, linperlisib, leniolisib, eganelisib, AZD8186, AZD8835, MK-2206, ipatasertib, GSK690693, capivasertib, PF-04691502, AT7867, MAY1125976, TAS117, Afuresertib, Uprosertib, AT13148, everolimus, temsirolimus, ridaforolimus, sirolimus, umirolimus, zotarolimus, ICSN3250, LY3023414, OSU-53, AZD8055, and rapamycin.
71. The method of claim 69, wherein the at least one inhibitor of AKT/PI3K/mTOR is selected from the group consisting of capivasertib, ipatasertib and MK-2206.
72. The method of claim 69, wherein the at least one inhibitor of AKT, PI3K, and/or mTOR is selected from the group consisting of vistusertib (AZD2014), NU7441 (KU- 57788), KU-0063794, TGX-221 , RLY-2608, CYH33, STX-478, LOXO-783, GSK2334470, CC-223, ABTL0812, DCBCI0901 , AZD-8055, sapanisertib, JR-AB2-011 , omipalisib, and torkinibm PI-103.
73. The method of any one of claims 69 to 72, wherein the compound is in combination with at least one inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK.
74. The method of claim 73, wherein the at least one inhibitor of PDK1 , RAS, RTK, RAF, MEK, ERK, HER2, EGFR, SOS1 , SOS2, SHP1 , and/or MAPK is selected from the group consisting of dabrafenib, trametinib (GSK1120212), encorafenib (LGX818), binimetinib, cobimetinib, naporafenib (LXH254), sorafenib (BAY 43-9006), vemurafenib (PLX4032), PLX-4720, regorafenib (BAY 73-4506), GDC_0879, RAF265 (CHIR-265), AZ 628, lifirafenib (BGB-283), NVP-BHG712, SB590885, ZM 336372, GW5074, TAK-632, agerafenib (RXDX-105), GNE-9815, TBAP-001 , L-779450, belvarafenib (HM95573), AZ304, PLX8394, RAF709, CCT196969, BAW2881 (NVP-BAW2881), LY3009120, avutometinib (RO5126766, CH5126766), doramapimod (BIRB 796), MCP110, PLX7904, tovorafenib (MLN2480), selumetinib (AZD6244), mirdametinib (PD0325901), 110126, PD98059, PD184352 (CI-1040), BIX 02189, pimasertib (AS-70326), pelitinib (EKB-569), BIX 02188, TAK-733, AZD8330, binimetinib (MEK162), SL-327, refametinib (RDEA119), zapnometinib (PD0184264), GDC-0623, BI-847325, cobimetinib (GDC_0973), PD318088, SCH772984, ASN007, MRTX-1257, AZD0634 (ATG-017), MK-8353 (SCH900353), temuterkib (LY3214996), VX-11e, ulixertinib (BVD-523), ravoxertinib (GDC-0994), exarafenib, ERAS-601 , BDTX-4933, ADT007, HM99462, DCC-3084, PHI- 501 , MRTX1133. Sotorasib (AMG510), lonafamib (SCH66336), ASP2453, BI-3406, Bl- 2852, BAY-293, KRpep-2d, ARS-853, adagrasib (MRTX849), adagrasib (MRTX849), ARS1620, divarasib (GDC-6036), GDC-1971 , IACS-13909, JAB-3068, RMC-4550, RMC- 4630, SHP099, TNO155, GDC-1971 , HBI-2376, BBP-398 (IACS-15509), and PF- 07284892.
75. The method of claim 73 or 74, wherein the at least one RTK inhibitor is selected from the group consisting of epidermal growth factor receptor (EGFR) inhibitors erlotinib, osimertinib, neratinib, gefitinib, cetuximab, panitumumab, dacomitinib, lapatinib, necitumumab, mobercitinib, vandetanib, sapitinib, afatinib, canertinib, CP-724714, CUDC- 101 , mubritinib (TAK 165), OSI-420, pelitinib, irbinitinib (ARRY-380, ONT-380), varlitinib, TAK-285, BIBX 1382, PD168393, raciletinib, poziotinib, CL-387785, icotinib, CTX-2006, WHI-P154, AZ5104, AZD3759, Erb-B2 receptor tyrosine kinase 2 (HER2) such as neratinib, trastuzumab and its derivatives such as trastuzumab deruxtecan, lapatinib, dacomitinib, tucatinib, pertuzumab, margetuximab, afatinib, AZD8931 , AST1306, AEE788, canertinib, CP724.714, CUDC101 , TAK285, dacomitinib, pelitinib, AC480, canertinib, sapitinib, mubritinib (TAK 165), BDTX-189, epertinib, TAS0728, poziotinib, multikinase inhibitors cabozantinib, imatinib, Lenvatinib, regorafenib, sorafenib, sunitinib, vandetinib, MET inhibitors crizotinib, cabozantinib, foretinib, PHA-665752, SU 11274, SGX-523, BMS-777607, tivantinib (ARQ 197), JNJ-38877605, PF-04217903, MGCD-265, capmatinib (INCB28060), BMS-754807, BMS-794833, AMG-208, MK-2461 , golvatinib (E7050), AMG-458, NVP-BVU972, XL092, UNC2025, elzovantinib (TPX-0022), AMG-1 , JNJ-38877618 (OMO-1), altiratinib, SAR125844, glumetinib (SCC244), savolitinib (AZD6094), RXDX-106 (CEP-40783), S49076, merestinib (LY2801653), AMG 337, tepotinib, neurotrophic receptor tyrosine kinase 1 (NTRK1) inhibitors larotrectinib, entrectinib, danusertib, BMS-754807, GW441756, UNC2025, taletrectinib, altiratinib, selitrectinib (LOXO-195), CH7057288, BMS-935177, PF-06273340, sitravatinib (MGCD516), GNF-5837, vascular endothelial growth factor receptor inhibitors bevacizumab, ramucirumab, SU5408, linifanib (ABT-869), axitinib (AG 013736), nintedanib (BIBF 1120), cediranib (AZD2171), motesanib (AMG-706), pazopanib (GW786034), vandetanib, sunitinib (SU11248), sorafenib (BAY 43-9006), brivanib (BMS- 540215), vatalanib (PTK787), foretinib (GSK1363089), cabozantinib (BMS-907351), brivanib (BMS-582664), Lenvatinib (E7080), CYC116, regorafenib (BAY 73-4506), ENMD-2076, tivozanib, ponatinib (AP24534), apatinib (YN968D1), telatinib, PP121 , pazopanib, dovitinib, SAR131675, semaxanib (SU5416), golvatinib (E7050), fibroblast growth factor receptor inhibitors ponatinib (AP24534), infigratinib (BGJ398), nintedanib (BIBF 1120), PD173074, AZD4547, dovitinib, pemigatinib, futibatinib, infigratinib, and RLY-4008.
76. The method of any one of claims 68 to 75, wherein the cancer or tumor is defined in any one of claims 25 to 36.
77. A method for the treatment of cancer or tumor in a subject, comprising: i. determining that cells from said cancer or tumor (e.g., cancer or tumor sample or biopsy): have an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; have an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; have a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; have an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIO; have an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by one or more concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN; have an upregulation of a mitogen activated protein kinase (MAPK) gene expression (e.g., CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and SPRY4); and/or are resistant to one or more anticancer therapies (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof); and ii. administering to the subject one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17, the pharmaceutical composition of any one of claims 37 to 44, or the combination of any one of claims 45 to 54.
78. Use of one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17, the pharmaceutical composition of any one of claims 37 to 44, or the combination of any one of claims 45 to 54, for the treatment of cancer or tumor or for the manufacture of a medicament for the treatment of cancer or tumor, wherein the cancer or tumor (e.g., cancer or tumor sample or biopsy) has been predetermined as: having an upregulation in expression of genes associated with ribosomal protein synthesis, ribosome biogenesis, protein translation, translation initiation, and/or translation elongation; having an upregulation of gene signatures associated with basal and/or mesenchymal breast cancer; having a downregulation of gene signatures associated with luminal breast cancer the cancer or tumor comprises cells having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway; having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by at least one mutation in KRAS, HRAS, NRAS, NF1 , BRAF, ARAF1 , NF2, MAP2K1 , RRAS2, and/or CIO; having an activation of a RAF, MEK, ERK, PI3K, AKT, and/or mTOR pathway that is mediated by one or more concomitant mutations in KRAS/PIK3CA, HRAS/PIK3CA, NRAS/PIK3CA, NF1/BRAF/PIK3CA, BRAF/PIK3CA, ARAF1/PIK3CA, NF2/PIK3CA, MAP2K1/PIK3CA, BRAF/AKT1 , and/or BRAF/PTEN; having an upregulation of a mitogen activated protein kinase (MAPK) gene expression (e.g., CCND1 , DUSP4, DUSP6, PHLDA1 , SPRY2, SPRY4, EPHA2, EPHA4, ETV4, and ETV, or DUSP6, ETV4, ETV5 and SPRY4); and/or being resistant to one or more anticancer therapies (e.g., BRAF inhibitors, MEK inhibitors, EGFR inhibitors, or combinations thereof).
79. Use of one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17, the pharmaceutical composition of any one of claims 37 to 44, or the combination of any one of claims 45 to 54, for the treatment of one or more side effects or toxicities caused by an anticancer therapy in a subject undergoing said anticancer therapy.
80. The use of claim 79, wherein the one or more side effects or toxicities caused by the anticancer therapy is selected from the group consisting of: hyperglycemia and hyperinsulinemia, increased ALT/AST, pyrexia, rash, diarrhea, nausea, fatigue, thrombocytopenia, anemia, dermatitis acneiform, QT elongation, and dyspnea.
81. A method for treating one or more side effects or toxicities caused by an anticancer therapy, comprising administering to a subject undergoing said anticancer therapy one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17, the pharmaceutical composition of any one of claims 37 to 44, or the combination of any one of claims 45 to 54.
82. The method of claim 81 , wherein the one or more side effects or toxicities caused by the anticancer therapy is selected from the group consisting of: hyperglycemia and hyperinsulinemia, increased ALT/AST, pyrexia, rash, diarrhea, nausea, fatigue, thrombocytopenia, anemia, dermatitis acneiform, QT elongation, and dyspnea.
83. A method for monitoring the progression of an anticancer therapy in a subject that was previously administered with one or more compounds or pharmaceutically acceptable salts thereof as defined in any one of claims 1 to 17, the pharmaceutical composition of any one of claims 37 to 44, or the combination of any one of claims 45 to 54, wherein the method comprises determining in a sample from said subject: a loss of phosphorylation of NDRG1 ; an increase in phosphorylation of MAPK14 (i.e. , p38); an increase in phosphorylation of MAPK14 (i.e., p38); an increase in phosphorylation of heat shock protein family B1 (i.e., HSPB1 or HSP27); an increase in phosphorylation of tumor suppressor p53 (TP53); an increase in phosphorylation of ribosomal protein S6 kinase A1 (RSK1); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERKI); an increase in phosphorylation of mitogen-activated protein kinase 3 (i.e., MAPK3 or ERK1); and/or an increase in phosphorylation of AKT1 substrate 1 (PRAS40).
84. The method of claim 83, wherein: said loss of phosphorylation of NDRG1 is at threonine 346 of the NDRG1 ; said increase in phosphorylation of MAPK14 (i.e., p38) is at threonine 180 of the MAPK14; said increase in phosphorylation of MAPK14 (i.e., p38) is at tyrosine 182 of the MAPK14; said increase in phosphorylation of heat shock protein family B1 (i.e., HSPB1 or HSP27) at serine 82 of the HSPB1 ; said increase in phosphorylation of tumor suppressor p53 (TP53) is at serine 15 of the TP53; said increase in phosphorylation of ribosomal protein S6 kinase A1 (RSK1) is at serine 380 of the RSK1 ; said increase in phosphorylation of mitogen-activated protein kinase 1 (i.e., MAPK1 or ERK2) is at tyrosine 185 of the MAPK1 ; said increase in phosphorylation of mitogen-activated protein kinase 1 (i.e.,
MAPK1 or ERK2) is at tyrosine 187 of the MAPK1 ; said increase in phosphorylation of mitogen-activated protein kinase 3 (i.e.,
MAPK3 or ERK1) is at threonine 202 of the MAPK3; said increase in phosphorylation of mitogen-activated protein kinase 3 (i.e.,
MAPK3 or ERK1) is at tyrosine 204 of the MAPK3; and/or said increase in phosphorylation of AKT 1 substrate 1 (PRAS40) is at threonine 246 of the AKT1 substrate 1.
85. The method of claim 83 or 84, wherein said loss or increase in phosphorylation is in comparison to a sample from a healthy subject or a subject that is not undergoing said anticancer therapy.
PCT/CA2023/050681 2022-07-13 2023-05-17 Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1 and treatment of cancer WO2024011307A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
USPCT/US2022/036956 2022-07-13
PCT/US2022/036956 WO2024015055A1 (en) 2022-07-13 2022-07-13 Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1 and treatment of cancer

Publications (1)

Publication Number Publication Date
WO2024011307A1 true WO2024011307A1 (en) 2024-01-18

Family

ID=89535091

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2022/036956 WO2024015055A1 (en) 2022-07-13 2022-07-13 Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1 and treatment of cancer
PCT/CA2023/050681 WO2024011307A1 (en) 2022-07-13 2023-05-17 Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1 and treatment of cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US2022/036956 WO2024015055A1 (en) 2022-07-13 2022-07-13 Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1 and treatment of cancer

Country Status (1)

Country Link
WO (2) WO2024015055A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014140065A1 (en) * 2013-03-13 2014-09-18 Sanofi N-(4-(azaindazol-6-yl)-phenyl)-sulfonamides and their use as pharmaceuticals
WO2022150911A1 (en) * 2021-01-13 2022-07-21 Therapeutique Qtl Inc. Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102007002717A1 (en) * 2007-01-18 2008-07-24 Merck Patent Gmbh Heterocyclic indazole derivatives
PT2570415E (en) * 2011-09-19 2015-11-23 Sanofi Sa N-[4-(1h-pyrazolo[3,4-b]pyrazin-6-yl)-phenyl]-sulfonamides and their use as pharmaceuticals

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014140065A1 (en) * 2013-03-13 2014-09-18 Sanofi N-(4-(azaindazol-6-yl)-phenyl)-sulfonamides and their use as pharmaceuticals
WO2022150911A1 (en) * 2021-01-13 2022-07-21 Therapeutique Qtl Inc. Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HALLAND, NIS ET AL.: "Rational Design of Highly Potent, Selective, and Bioavailable SGK1 Protein Kinase Inhibitors for the Treatment of Osteoarthritis", JOURNAL OF MEDICINAL CHEMISTRY, vol. 65, 21 December 2021 (2021-12-21), pages 1567 - 1584, XP055929134, DOI: 10.1021/acs.jmedchem.1c01601 *
ORLACCHIO ARTURO, RANIERI MICHELA, BRAVE MARTINA, ARCIUCH VALERIA ANTICO, FORDE TONI, DE MARTINO DANIELA, ANDERSON KAREN E., HAWKI: "SGK1 Is a Critical Component of an AKT-Independent Pathway Essential for PI3K-Mediated Tumor Development and Maintenance", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 77, no. 24, 15 December 2017 (2017-12-15), US, pages 6914 - 6926, XP093130616, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-17-2105 *

Also Published As

Publication number Publication date
WO2024015055A1 (en) 2024-01-18

Similar Documents

Publication Publication Date Title
EP3237418B1 (en) Triazolopyrimidine compounds and uses thereof
JP6140170B2 (en) Substituted benzylindazoles for use as BUB1 kinase inhibitors in the treatment of hyperproliferative diseases
AU2018200536B2 (en) Amino pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
US20220106281A1 (en) Modulators of the integrated stress pathway
RU2537945C2 (en) Triazine, pyrimidine and pyridine analogues and use thereof as therapeutic agents and diagnostic samples
RU2470936C2 (en) Phosphoinositide3-kinase inhibiting compounds and methods for use thereof
CN109790166A (en) Imidazopyridine is used for treating cancer
JP2020502246A (en) 2-benzopyrazinyl-N-heteroaryl-2-phenyl-acetamide compound
HUE026041T2 (en) Pyrrolotriazinone derivatives as pi3k inhibitors
JP6751212B2 (en) Antitumor agent
CN109906224A (en) Triazole pyridine compounds and its application
WO2013113669A1 (en) Isoquinoline and naphthyridine derivatives
TW202116753A (en) Cinnolines as inhibitors of hpk 1
JP2020536920A (en) Methods of Inhibiting Pyrrolotriazine Compounds and TAM Kinases
WO2022150911A1 (en) Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1
EP3885348A1 (en) Novel tricyclic compound as irak4 inhibitor
EP3134396B1 (en) Amino pyridine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2024011307A1 (en) Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1 and treatment of cancer
CN118201896A (en) PI 3K-alpha inhibitors and methods of use thereof
CN117062817A (en) Pyrazolo [3,4-D ] pyrimidin-6-yl-sulfonamide derivatives for SGK-1 inhibition

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23838351

Country of ref document: EP

Kind code of ref document: A1