WO2025114771A1 - A new class of aryl hydrocarbon receptor antagonists and their optimisation - Google Patents
A new class of aryl hydrocarbon receptor antagonists and their optimisation Download PDFInfo
- Publication number
- WO2025114771A1 WO2025114771A1 PCT/IB2024/059009 IB2024059009W WO2025114771A1 WO 2025114771 A1 WO2025114771 A1 WO 2025114771A1 IB 2024059009 W IB2024059009 W IB 2024059009W WO 2025114771 A1 WO2025114771 A1 WO 2025114771A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- alkyl
- arylc
- alkenyl
- heteroarylc
- ocf
- Prior art date
- Legal status (The legal status 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 status listed.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic 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
- C07D487/04—Ortho-condensed systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
Definitions
- a new class of aryl hydrocarbon receptor antagonists and their optimisation [001] The invention relates to novel AHR antagonists.
- the aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that mediates a broad spectrum of diverse biological processes from adaptive metabolism, acute toxicity, to normal physiology of the vascular and immune systems.
- the AHR was initially discovered to play a key role in mitigating the toxic effects of environmental pollutants; however, recent studies have shifted to its physiologic functions and its involvement in cell cytotoxicity, cell cycle regulation, neural development, and immune responses.
- the AHR belongs to basic helix-loop-helix Per-Arnt-Sim (bHLH-PAS) superfamily of regulatory proteins, and contains internal PAS domain with two similar repeats, PAS-A and PAS-B, an N-terminal bHLH domain, and a C-terminal transcriptional activation domain (TAD). 2–6
- the bHLH domain of the AHR participates in DNA binding, and together with the PAS-A domain dictates AHR–ARNT (AHR nuclear translocator) dimerisation.
- the PAS-B domain of the AHR is the ligand binding domain.
- Unliganded AHR is in the cytosol, where it is associated with the heat shock protein 90 (Hsp90) and co-chaperones like X-associated protein 2 (XAP2, also known as AHR-interacting protein/AHR-associated protein 9, AIP/ARA9) and p23 1,7,8 .
- Hsp90 heat shock protein 90
- XAP2 co-chaperones like X-associated protein 2
- AIP/ARA9 also known as AHR-interacting protein/AHR-associated protein 9, AIP/ARA9
- Ligand binding to the PAS-B domain induces conformational changes in AHR that trigger its translocation of the complex into the nucleus, where AHR is released and interacts with the ARNT to initiate what is defined as its canonical genomic signalling pathway.
- the newly formed AHR-ARNT heterodimer binds to a xenobiotic-response element (XRE) in their target gene promoter regions to regulate their expression.
- XRE xenobiotic
- AHR Due to AHR’s key regulatory role in the immune system and cancer immunology, it is now considered a promising drug target. 1,3,10 Studies have shown that AHR expression is upregulated in most tumour cells, and is associated with tumour proliferation, invasion, metastasis, and immune escape through increased checkpoint inhibitor expression. 7,11,12 As a result, AHR antagonists and immune checkpoint inhibitors are considered to have great potential in tumour immunotherapy. 8,13 [005] With the increasing understanding of AHR functions and signalling pathways, medicinal chemistry campaigns have emerged to identify novel and selective AHR antagonists.
- the AHR is activated by a wide range of structurally diverse compounds, including environmental pollutants such as non-halogenated polycyclic aromatic hydrocarbons (PAHs), halogenated aromatic hydrocarbons (HAHs) and polychlorinated biphenyls (PCBs), alkaloids, synthetic compounds, drugs, as well as intermediary and microbial metabolites of tryptophan, tetrapyrroles, eicosanoids.
- PHAs polycyclic aromatic hydrocarbons
- PCBs polychlorinated biphenyls
- AHR antagonists are in preclinical and clinical studies, such as StemRegenin 1 (1, IC50: 127 nM) 15 , the 1,3-diaryl-pyrazin-6-one-5-carboxamide based BAY2416964 (2, IC50: 22 nM) and BAY128 (3, IC 50 : 40 nM) developed by Bayer AG 16 , IK-175 (4, IC 50 : 91 nM) and KYN-101 (5, IC 50 : 23 nM) developed by Ikena Oncology (formerly Kyn Therapeutics) 17–19 , and GNF351 (6, IC 50 : 8.5 nM) 20 (Fig.1).
- AHR antagonists are not limited to 5,6-fused bicyclic heteroaromatic compound derivatives and analogues, and a comprehensive overview of the current state of AHR ligand development is described elsewhere. 14,21 Fig.1.
- AHR antagonists in preclinical and clinical trials.
- AHR inhibition activities (IC 50 ) in in- vitro assays is given below the compound molecular structure.
- SAR structure-activity relationships
- alkyl refers to a straight or branched hydrocarbon chain, containing the indicated number of carbon atoms.
- C 1 -C 12 alkyl indicates that the alkyl group may have from 1 to 12 (inclusive) carbon atoms.
- alkylene refers to a divalent alkyl, e.g., —CH 2 —, —CH 2 CH 2 —, —CH 2 CH 2 CH 2 — or —CH 2 CH(CH 3 )CH 2 —.
- An alkyl or alkylene may be optionally substituted.
- cycloalkyl refers to nonaromatic, saturated or partially unsaturated cyclic, bicyclic, tricyclic or polycyclic hydrocarbon groups having 3 to 12 carbons. Any ring atom can be substituted (e.g., with one or more substituents). Cycloalkyl groups can contain fused rings. Fused rings are rings that share one or more common carbon atoms.
- cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, methylcyclohexyl, adamantyl, norbornyl and norbornenyl.
- alkenyl refers to a straight or branched hydrocarbon chain having one or more double bonds. Examples of alkenyl groups include, but are not limited to, allyl, propenyl, 2-butenyl, 3-hexenyl and 3-octenyl groups.
- alkenylene refers to a divalent alkenyl, e.g., —CH ⁇ CH—, —CH ⁇ CH 2 CH 2 — or —CH ⁇ C ⁇ CH—.
- heterocyclyl refers to a nonaromatic, saturated or partially unsaturated 3-10 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, S, Si and P (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of O, N, S, Si and P if monocyclic, bicyclic, or tricyclic, respectively). Any ring atom can be substituted (e.g., with one or more substituents).
- Heterocyclyl groups can contain fused rings, which are rings that share one or more common atoms.
- heterocyclyl groups include, but are not limited to, radicals of tetrahydrofuran, tetrahydrothiophene, tetrahydropyran, piperidine, piperazine, morpholine, pyrroline, pyrimidine, pyrrolidine, indoline, tetrahydropyridine, dihydropyran, thianthrene, pyran, benzopyran, xanthene, phenoxathiin, phenothiazine, furazan, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like.
- cycle refers to any fully saturated, partially saturated or unsaturated carbocycles and heterocycles.
- Specific compounds of Formula I within the present invention include but are not limited to: DETAILED DESCRIPTION OF THE INVENTION [032] Human AHR PAS-B domain homology model was constructed using HIF-2 ⁇ (hypoxia inducible factor) PAS-B domain in complex with a tetrazole-containing antagonist as a template (PDB: 4XT2).
- the selected template has 26% identity and 51% similarity to human AHR PAS-B domain sequence, and homology modelling resulted in a model exhibiting a backbone atom rmsd of 1.8 ⁇ to the agonist-bound AHR structure that became available recently.
- Potential AHR antagonists were identified via high throughput virtual screening (HTVS) of MolPort’s commercially available compound library (6.3M comp., 2021) against the created AHR homology model.
- Top-ranked molecules were inspected for their ability to form hydrogen bonding and stacking interactions similar to those observed in available AHR homologue complex structures (e.g., stacking interactions with F295, F324 and/or F351; and hydrogen bonding with S365, H337 and/or H291; AHR numbering), and molecules with internal molecular strain or unsatisfied hydrogen bond donors/acceptors were deprioritised.
- a total of 52 compounds were purchased from MolPort and were experimentally tested in a luciferase reporter gene assay. Briefly, HuH-7 human hepatoma cells were transfected with Cyp1a1 regulated luciferase reporter gene. The next day cells were treated with 10 nM TCDD with and without test compounds for 20 h.
- luciferase activity was determine using the dual luciferase reporter gene system according to the manufacturer’s instructions (Promega, Madison WI USA). For full assay description see Methods section in SI. Of all the compounds tested, 9 compounds inhibited AHR activity by 50% or more at a 25 ⁇ M concentration, but only one compound showed activity comparable to that of BAY2416964. Three most active HTVS hits are shown in Fig.2 and inhibited AHR activity by 99%, 79% and 76%, respectively. Fig.2. AHR inhibitors identified. [034] The most active compound, 7, was selected for further optimisation. Hit compound optimisation was started by consecutive R 1 -R 10 substituent optimisation, avoiding changes to pyrazolopyrimidine core. Once optimal substituents for each position were identified, functional groups were combined into a single molecule. Exemplary compounds prepared and tested are summarised in Tables 1 and 2.
- the obtained mixture was then cooled to RT, diluted with DCM (40 ml) and washed with brine (10 ml). The water layer was additionally extracted with DCM (2 ⁇ 15 ml). The combined organics were dried over Na 2 SO 4 and concentrated in vacuo. The residue was dissolved in DCM (5 ml) and TFA (3 ml) and stirred for 18h at RT, then concentrated in vacuo. The obtained oil was basified by stirring with K 2 CO 3 (0.4 g, 5eq) in 90% aq MeOH for 2h. The obtained mixture was concentrated in vacuo, the residue was dissolved in mixture of DCM (40 ml) and water (20 ml). The organic layer was separated, dried over Na 2 SO 4 and concentrated in vacuo.
- N-(2-methoxyethyl)-5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-amine (int7b) was prepared from 7-chloro-5,6-dimethylpyrazolo[1,5-a]pyrimidine using the method described previously for the synthesis of N-(2-methoxyethyl)-5-methylpyrazolo[1,5-a]pyrimidin-7- amine.
- the isolated crude product was purified by column chromatography (10%-30% EtOAc/PE) to afford desired product as colorless oil (yield 78%).
- N-bromosuccinimide (1.2 g) was added to the crude tert-butyl (2-methoxyethyl)(5- methylpyrazolo[1,5-a]pyrimidin-7-yl)carbamate in MeCN (80 ml) at RT. After stirring the reaction mixture for 18h the solvent was evaporated in vacuo. The resulting semisolid was dissolved in DCM (50 ml). The obtained solution was washed with water (30 ml), dried over Na 2 SO 4 and concentrated in vacuo. The residue was subjected to column chromatography (10%-50% EtOAc/PE) to give brominated product as yellow solid (1.2 g, yield 40%).
- tert-Butyl (3-bromo-5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-yl)(2- methoxyethyl)carbamate (int9b) as white solid (yield 90%) was prepared by reacting tert- butyl (5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-yl)(2-methoxyethyl)carbamate with N- bromosuccinimide (1.2 eq) using the method described previously for the synthesis of int9a.
- tert-Butyl (5-isopropylpyrazolo[1,5-a]pyrimidin-7-yl)(2-methoxyethyl)carbamate (int8c) as colourless oil (yield 90%) was prepared from 5-iso-propyl-N-(2- methoxyethyl)pyrazolo[1,5-a]pyrimidin-7-amine using the method described previously for the synthesis of tert-butyl (5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-yl)(2- methoxyethyl)carbamate.
- N-(3-(1H-imidazol-1-yl)propyl)-3-(4-methoxyphenyl)-5,6-dimethylpyrazolo[1,5- a]pyrimidin-7-amine (15) was prepared from 7-chloro-3-(4-methoxyphenyl)-5,6- dimethylpyrazolo[1,5-a]pyrimidine. The purification was carried out using 20%-100% acetone/DCM as eluent to afford 15 as white solid (0.30 g, yield 39%, HPLC purity 99% at 254 nm, 99% at 210 nm).
- N-(3-(1H-imidazol-1-yl)propyl)-5-isopropyl-3-(4-methoxyphenyl)pyrazolo[1,5- a]pyrimidin-7-amine 10 was prepared from 7-chloro-5-isopropyl-3-(4- methoxyphenyl)pyrazolo[1,5-a]pyrimidine. After crystallization from MeOH 10 was isolated as white solid (0.3 g, yield 38%, HPLC purity 99% at 254 nm, 98% at 210 nm).
- pGudLuc 4.1 is an AHR-driven reporter construct containing a luciferase gene downstream of the Cyp1a1 promoter.
- the cells were treated with either DMSO, 10 nM TCDD alone or in combination with test compounds (0.001 to 10 mM).
- the cells were lysed, and the luciferase activity was measured using Dual luciferase assay and normalized to Renilla activity.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Immunology (AREA)
- Public Health (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The invention relates to a novel class of aryl hydrocarbon receptor (AHR) antagonists. More specifically, the invention relates to novel bicyclic heteroaromatic compounds as AHR antagonists and their use in the immunotherapy of malignant tumors.
Description
A new class of aryl hydrocarbon receptor antagonists and their optimisation [001] The invention relates to novel AHR antagonists. Background of the art [002] The aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that mediates a broad spectrum of diverse biological processes from adaptive metabolism, acute toxicity, to normal physiology of the vascular and immune systems. The AHR was initially discovered to play a key role in mitigating the toxic effects of environmental pollutants; however, recent studies have shifted to its physiologic functions and its involvement in cell cytotoxicity, cell cycle regulation, neural development, and immune responses.1 [003] The AHR belongs to basic helix-loop-helix Per-Arnt-Sim (bHLH-PAS) superfamily of regulatory proteins, and contains internal PAS domain with two similar repeats, PAS-A and PAS-B, an N-terminal bHLH domain, and a C-terminal transcriptional activation domain (TAD).2–6 The bHLH domain of the AHR participates in DNA binding, and together with the PAS-A domain dictates AHR–ARNT (AHR nuclear translocator) dimerisation. The PAS-B domain of the AHR is the ligand binding domain. Unliganded AHR is in the cytosol, where it is associated with the heat shock protein 90 (Hsp90) and co-chaperones like X-associated protein 2 (XAP2, also known as AHR-interacting protein/AHR-associated protein 9, AIP/ARA9) and p231,7,8. Ligand binding to the PAS-B domain induces conformational changes in AHR that trigger its translocation of the complex into the nucleus, where AHR is released and interacts with the ARNT to initiate what is defined as its canonical genomic signalling pathway. The newly formed AHR-ARNT heterodimer binds to a xenobiotic-response element (XRE) in their target gene promoter regions to regulate their expression.9 [004] Due to AHR’s key regulatory role in the immune system and cancer immunology, it is now considered a promising drug target.1,3,10 Studies have shown that AHR expression is upregulated in most tumour cells, and is associated with tumour proliferation, invasion, metastasis, and immune escape through increased checkpoint inhibitor expression.7,11,12 As a result, AHR antagonists and immune checkpoint inhibitors are considered to have great potential in tumour immunotherapy.8,13 [005] With the increasing understanding of AHR functions and signalling pathways, medicinal chemistry campaigns have emerged to identify novel and selective AHR antagonists. The AHR is activated by a wide range of structurally diverse compounds, including environmental pollutants such as non-halogenated polycyclic aromatic hydrocarbons (PAHs), halogenated aromatic hydrocarbons (HAHs) and polychlorinated biphenyls (PCBs), alkaloids,
synthetic compounds, drugs, as well as intermediary and microbial metabolites of tryptophan, tetrapyrroles, eicosanoids.14 [006] Several AHR antagonists are in preclinical and clinical studies, such as StemRegenin 1 (1, IC50: 127 nM)15, the 1,3-diaryl-pyrazin-6-one-5-carboxamide based BAY2416964 (2, IC50: 22 nM) and BAY128 (3, IC50: 40 nM) developed by Bayer AG16, IK-175 (4, IC50: 91 nM) and KYN-101 (5, IC50: 23 nM) developed by Ikena Oncology (formerly Kyn Therapeutics)17–19, and GNF351 (6, IC50: 8.5 nM)20 (Fig.1). AHR antagonists are not limited to 5,6-fused bicyclic heteroaromatic compound derivatives and analogues, and a comprehensive overview of the current state of AHR ligand development is described elsewhere.14,21
Fig.1. AHR antagonists in preclinical and clinical trials. AHR inhibition activities (IC50) in in- vitro assays is given below the compound molecular structure. [007] Despite the discovery of antagonists with high activity, the structure-activity relationships (SAR) of the AHR inhibitors are poorly understood. This is mostly due to the lack of structural information on the ligand binding PAS-B domain. Only recently, the crystal structures of the soluble PAS-B domain of Drosophila AHR3, agonist-bound cytosolic complex of human AHR (Hsp90-XAP2-AHR)6 and ligand-free cytosolic mouse AHR complex (Hsp90- AhR-p23 with or without bound XAP2)2 have been resolved. However, no antagonist-bound AHR structures have been determined thus far. [008] Here we report structure-based drug design (SBDD) of novel AHR antagonists starting from virtual screening against an AHR PAS-B domain homology model constructed based on human hypoxia inducible factor 2α (hHIF-2α). The virtual screening hits were validated
experimentally, and the most potent compound was optimised to achieve low nanomolar AHR inhibition activity. The AHR inhibitors presented here hold the potential to be used in medicine to treat cancer or other proliferative disorders. Summary of the invention [009] We disclose compounds selected from those of formula I
wherein: [010] R is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2- 5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L -OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N - C ( 1,
- - n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; [011] R1, R2, R3 R4 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1- 6alkyl, C2-6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2- n,
L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L
-OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N HR11, -NR11 2, -N(R11)R12, -L-NH2, -L-NHR11, -L-NR11 2, -L-N(R11)R12, -NH-L-NH2, -NH-L- - C ( 1,
each L, if present, is independently saturated aliphatic C1‑5alkylene; [012] R1 and R2 may form a cycle; R3 and R4 may form a cycle; R and R2 may form a cycle; R and R3 may form a cycle; [013] R5 is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L-NH2, -O-L -NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L-OR11, -O- L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -NHR11, -NR 11 - 1) - R
2, - - n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; [014] R6 is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L
-NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L -OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N HR11, -NR11 2, -N(R11)R12, -L-NH2, -L-NHR11, -L-NR11 2, -L-N(R11)R12, -NH-L-NH2, -NH-L- - C ( 1,
each L, if present, is independently saturated aliphatic C1‑5alkylene; [015] R5 and R6 may form a cycle; X, Y and Z may be independently substituted CR11R12, NR11, O; R11 and R12 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, aryl C5-8biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH,-O-L-OH, -OL-NH2, OCF3, -OCH2CF3, -OCF2CF2H, SCF3, -CN, -NO2, -NO2, -NH2, R11 and R12 may form a cycle; R5 and R11 may form a cycle; R6 and R11 may form a cycle; R5 and R12 may form a cycle; R6 and R12 may form a cycle; [016] W is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8 biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; V is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; U is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; [017] Specific compounds of Formula I within the present invention include a compound selected from general formula 2:
II [018] R is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2- 5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L -OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N - C ( 1,
n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; [019] R1, R2, R3 R4 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1- 6alkyl, C2-6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2- 6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L - N - C
( =O)OR12, -OC(=O)NH2, -OC(=O)NHR11, -OC(=O)N(R11)R12, -OC(=O)R11NR12, -OC(=O)R1
1, -C(=O)R11,-NHC(=O)NH2, -NHC(=O)NHR8, -NHC(=O)NR11 2, -NHC(=O)N(R11)R12, -NR1
each L, if present, is independently saturated aliphatic C1‑5alkylene; [020] R1 and R2 may form a cycle; R3 and R4 may form a cycle; R and R2 may form a cycle; R and R3 may form a cycle; [021] R5 is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, L - 11 - 1) - R 2,
n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; [022] R6, R7 R8, R9 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1- 6alkyl, C2-6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2- 6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L -OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N HR11, -NR11 2, -N(R11)R12, -L-NH2, -L-NHR11, -L-NR11 2, -L-N(R11)R12, -NH-L-NH2, -NH-L- NHR11, -NH-L-N(R11)R12, -NH-L-N(R11)R12, -NR11-L-NH2, -NR11-L-NHR11, -NR11-L-N(R11) R12, -NR11-L-N(R11)R12, -N(R11)R12, -C(=O)OH, -C(=O)OR11, -C(=O)NH2, -C(=O)NHR11, - C(=O)N(R11)12, -C(=O)N(R11)R12, -NHC(=O)R11, -NR11C(=O)R12, -NHC(=O)OR11, -NR11C(
=O)OR12, -OC(=O)NH2, -OC(=O)NHR11, -OC(=O)N(R11)R12, -OC(=O)R11NR12, -OC(=O)R1 1,
each L, if present, is independently saturated aliphatic C1‑5alkylene; [023] R10 is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, L - 11 - 1) - R 2,
n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; [024] R11 and R12 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2-6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH,-O-L-OH, -OL-NH2, OCF3, -OCH2CF3, -OCF2CF2H, SCF3, -CN, -NO2, -NO2, -NH2, - R11 and R12 may form a cycle; R11 and R12 may form a cycle; R5 and R11 may form a cycle; R6 and R11 may form a cycle; R5 and R12 may form a cycle; R6 and R12 may form a cycle; [025] W is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl;
V is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; U is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; Pairs of substituents selected from R5, R6, R7, R8, R9, R10 may form a cycle. [026] As used herein, the term “alkyl” refers to a straight or branched hydrocarbon chain, containing the indicated number of carbon atoms. For example, C1-C12 alkyl indicates that the alkyl group may have from 1 to 12 (inclusive) carbon atoms. The term “alkylene” refers to a divalent alkyl, e.g., —CH2—, —CH2CH2—, —CH2CH2CH2— or —CH2CH(CH3)CH2—. An alkyl or alkylene may be optionally substituted. [027] The term “cycloalkyl” as used herein refers to nonaromatic, saturated or partially unsaturated cyclic, bicyclic, tricyclic or polycyclic hydrocarbon groups having 3 to 12 carbons. Any ring atom can be substituted (e.g., with one or more substituents). Cycloalkyl groups can contain fused rings. Fused rings are rings that share one or more common carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, methylcyclohexyl, adamantyl, norbornyl and norbornenyl. [028] The term “alkenyl” refers to a straight or branched hydrocarbon chain having one or more double bonds. Examples of alkenyl groups include, but are not limited to, allyl, propenyl, 2-butenyl, 3-hexenyl and 3-octenyl groups. One of the double bond carbons may optionally be the point of attachment of the alkenyl substituent. The term “alkenylene” refers to a divalent alkenyl, e.g., —CH═CH—, —CH═CH2CH2— or —CH═C═CH—. [029] The term “heterocyclyl” as used herein refers to a nonaromatic, saturated or partially unsaturated 3-10 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, S, Si and P (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of O, N, S, Si and P if monocyclic, bicyclic, or tricyclic, respectively). Any ring atom can be substituted (e.g., with one or more substituents). Heterocyclyl groups can contain fused rings, which are rings that share one or more common atoms. Examples of heterocyclyl groups include, but are not limited to, radicals of tetrahydrofuran, tetrahydrothiophene, tetrahydropyran, piperidine, piperazine, morpholine, pyrroline, pyrimidine, pyrrolidine, indoline, tetrahydropyridine, dihydropyran, thianthrene,
pyran, benzopyran, xanthene, phenoxathiin, phenothiazine, furazan, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like. [030] The term “cycle” refers to any fully saturated, partially saturated or unsaturated carbocycles and heterocycles. [031] Specific compounds of Formula I within the present invention include but are not limited to:
DETAILED DESCRIPTION OF THE INVENTION [032] Human AHR PAS-B domain homology model was constructed using HIF-2α (hypoxia inducible factor) PAS-B domain in complex with a tetrazole-containing antagonist as a template (PDB: 4XT2).22 The selected template has 26% identity and 51% similarity to human AHR PAS-B domain sequence, and homology modelling resulted in a model exhibiting a backbone atom rmsd of 1.8 Å to the agonist-bound AHR structure that became available recently. [033] Potential AHR antagonists were identified via high throughput virtual screening (HTVS) of MolPort’s commercially available compound library (6.3M comp., 2021) against the created AHR homology model. Top-ranked molecules were inspected for their ability to form hydrogen bonding and stacking interactions similar to those observed in available AHR homologue complex structures (e.g., stacking interactions with F295, F324 and/or F351; and hydrogen bonding with S365, H337 and/or H291; AHR numbering), and molecules with internal molecular strain or unsatisfied hydrogen bond donors/acceptors were deprioritised. A total of 52 compounds were purchased from MolPort and were experimentally tested in a luciferase reporter gene assay. Briefly, HuH-7 human hepatoma cells were transfected with Cyp1a1 regulated luciferase reporter gene. The next day cells were treated with 10 nM TCDD with and without test compounds for 20 h. The amount of luciferase activity was determine using the dual luciferase reporter gene system according to the manufacturer’s instructions
(Promega, Madison WI USA). For full assay description see Methods section in SI. Of all the compounds tested, 9 compounds inhibited AHR activity by 50% or more at a 25 μM concentration, but only one compound showed activity comparable to that of BAY2416964. Three most active HTVS hits are shown in Fig.2 and inhibited AHR activity by 99%, 79% and 76%, respectively.
Fig.2. AHR inhibitors identified. [034] The most active compound, 7, was selected for further optimisation. Hit compound optimisation was started by consecutive R1-R10 substituent optimisation, avoiding changes to pyrazolopyrimidine core. Once optimal substituents for each position were identified, functional groups were combined into a single molecule. Exemplary compounds prepared and tested are summarised in Tables 1 and 2.
Table 1. Exemplary compounds of formula I Comp. IC50, 7 2 7a 0 7a 1 7b 3 7c 7 7d 0 7e 7 7f 2 7g 0 7h 4 7i 0 7j 0 7k 0 7l 8 7m 1 7n 8 7o 4 7p 3 7r 9 7s 0 7t 6 7u 6 7v 0 7w 5 7x 5 7y 7 7z 0 7a 5 7a 0 7a
0 7ae H H H H H H CH2CH2OH Me H CH(CH3)2 CH2 CH2 O 0.662
af H H H H H H CH2CH2OH H Me Me CH2 CH2 O 0.890a 4a a 9a a 0 a 0a 80a 7a 3a 7a 8 a 9a 1a a 80 51 82 43
64 H H OMe H H H Me H Me Me CH2 CH2 O 2.431
Table 2. Exemplary AHR inhibitors
EXPERIMENTAL METHODS [035] The NMR spectra were recorded on Varian 400-MR instruments (working frequencies 400 and 100 MHz for 1H and 13C, respectively) and 300-MR instruments (working frequencies 300 for 1H) with the residual signals of the protons of DMSO (2.50 ppm for 1H-NMR and 39.52 ppm for 13C-NMR), of CHCl3 (7.26 ppm for 1H-NMR and 77.16 ppm for 13C-NMR) and of MeOH (3.31 ppm for 1H-NMR and 49.00 ppm for 13C-NMR) as internal standards. Chemical shifts (^) are expressed in ppm and coupling constants (J) are given in Hz. The low-resolution mass spectra were obtained with an Acquity UPLC liquid chromato-mass spectrometer of the (Wasters)-Q-TOF (Micromass) system with an Acquity UPLC BEH C18 chromatographic column (1.7 μm, 2.1×50 mm), gradient elution MeOH–HCOOH (0.1%) in water, ESI ionization, for positive and negative ions. The purity of the compounds was determined by HPLC on a Waters Alliance 2695 liquid chromatograph with a Waters 2489 UV/Vis detector, an Apollo C18 column or Adamas column (5 μm, 4.6×150 mm), and gradient elution with MeCN–0.1% H3PO4 in water. Examples
[036] Preparation of the disclosed compounds in the present invention is described in the following examples, which are intended as an illustration of and not a limitation on the scope of the invention. [037] The synthesis of 2-(2-((3-aryl-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ols Scheme 1
[038] Synthesis of N-(2-(2-((tert-butyldimethylsilyl)oxy)ethoxy)ethyl)-5- phenylpyrazolo[1,5-a]pyrimidin-7-amine (int1) 7-Chloro-5-phenylpyrazolo[1,5-a]pyrimidine (1.2 g, 5.2 mmol) was suspended in the mixture of Dox (35 ml) and DMF (3 ml) followed by addition of DIPEA (3.6 ml, 4 eq) and 2-(2-((tert- butyldimethylsilyl)oxy)ethoxy)ethan-1-amine (1.7 g, 1.5 eq). The reaction mixture was stirred by reflux for 18h, and then cooled to RT. The solvent was removed to give an oily residue that was dissolved in EtOAc (50 ml). The obtained solution was washed with brine (2x15 ml), dried over Na2SO4 and the solvent was removed to give oil that was purified by column chromatography (EtOAc/PE 1:2) to afford the desired product as colourless oil (1.1 g, yield 50%). 1H-NMR (300 MHz, CDCl3) δ 8.06-7.98 (m, 3H), 7.52-7.41 (m, 3H), 6.67 (t, J = 5.4 Hz, 1H), 6.55 (d, J = 2.3 Hz, 1H), 6.38 (s, 1H), 3.85 (t, J= 5.4 Hz, 2H), 3.82-3.77 (m, 2H), 3.67 (t, J= 5.4 Hz, 2H), 3.64-3.59 (m, 2H), 0.88 (s, 9H), 0.06 (s, 6H). MS: 413.3 [M+H]+.
[039] Synthesis of tert-butyl (2-(2-((tert-butyldimethylsilyl)oxy)ethoxy)ethyl)(5- phenylpyrazolo[1,5-a]pyrimidin-7-yl)carbamate (int2) To a stirred solution of N-(2-(2-((tert-butyldimethylsilyl)oxy)ethoxy)ethyl)-5- phenylpyrazolo[1,5-a]pyrimidin-7-amine (4.0 g, 9.7 mmol) in DCM (100 ml) was added di- tert-butyl dicarbonate (4.2 g, 2 eq) followed by a catalytic amount of 4-dimethylaminopyridine (120 mg, 0.1 eq). The reaction mixture was stirred at RT for 18h, then washed with water (2^ 50 ml), dried over Na2SO4, concentrated in vacuo. The obtained colourless oil was used in the next step without purification. 1H-NMR (300 MHz, CDCl3) δ 8.14-8.06 (m, 3H), 7.55-7.43 (m, 3H), 7.39 (s, 1H), 6.75 (d, J = 2.3 Hz, 1H), 4.02 (t, J = 5.2 Hz, 2H), 3.73 (t, J = 5.2 Hz, 2H), 3.57 (t, J = 5.4 Hz, 2H), 3.42 (t, J = 5.4 Hz, 2H), 1.36 (s, 9H), 0.81 (s, 9H), 0.04 (s, 6H). MS: 513.4 [M+H]+. [040] Synthesis of tert-butyl (3-bromo-5-phenylpyrazolo[1,5-a]pyrimidin-7-yl)(2-(2- ((tert-butyldimethylsilyl)oxy)ethoxy)ethyl)carbamate (int3) N-bromosuccinimide (1.7 g) was added to crude tert-butyl (2-(2-((tert- butyldimethylsilyl)oxy)ethoxy)ethyl)(5-phenylpyrazolo[1,5-a]pyrimidin-7-yl)carbamate in MeCN (80 ml) at RT. The reaction mixture was stirred at RT for 5h, and then the solvent was evaporated in vacuo. The residue was dissolved in DCM (200 ml), the obtained solution was washed with water (3^50 ml), dried over Na2SO4 and concentrated in vacuo. The residue was subjected to column chromatography (EA/PE 1:2) to give brominated product as yellow oil (4.7 g, yield 82% calculated from N-(2-(2-((tert-butyldimethylsilyl)oxy)ethoxy)ethyl)-5- phenylpyrazolo[1,5-a]pyrimidin-7-amine). 1H-NMR (400 MHz, CDCl3) δ 8.22-8.15 (m, 2H), 8.09 (s, 1H), 7.55-7.49 (m, 3H), 7.47 (s, 1H), 4.00 (t, J = 5.1 Hz, 2H), 3.72 (t, J = 5.1 Hz, 2H), 3.56 (t, J = 5.3 Hz, 2H), 3.41 (t, J = 5.3 Hz, 2H), 1.36 (s, 9H), 0.81 (s, 9H), 0.04 (s, 6H). MS: 591.2 [M (79Br)+H]+, 593.2 [M (81Br)+H]+. [041] General method A for the synthesis of 2-(2-((3-aryl-5-phenylpyrazolo[1,5- a]pyrimidin-7-yl)amino)ethoxy)ethan-1-ols PdCl2(dffp) (20 mg, 5 mol-%) was added to a mixture of int3 (0.59 mmol), aryl boronic acid (1.5 eq) and K3PO4 (0.5 g, 4 eq) in Dox (9 ml) and water (3 ml) under argon. The reaction mixture was stirred and heated at 80^C. The course of the reaction was controlled by LC-MS. The obtained mixture was then cooled to RT, diluted with DCM (40 ml) and washed with brine (10 ml). The water layer was additionally extracted with DCM (2^15 ml). The combined organics were dried over Na2SO4 and concentrated in vacuo. The residue was dissolved in DCM (5 ml) and TFA (3 ml) and stirred for 18h at RT, then concentrated in vacuo. The obtained oil was basified by stirring with K2CO3 (0.4 g, 5eq) in 90% aq MeOH for 2h. The obtained mixture was concentrated in vacuo, the residue was dissolved in mixture of DCM (40 ml) and water (20
ml). The organic layer was separated, dried over Na2SO4 and concentrated in vacuo. The desired reaction product was isolated by purification via column chromatography. [042] 2-(2-((3,5-Diphenylpyrazolo[1,5-a]pyrimidin-7-yl)amino)ethoxy)ethan-1-ol (7): Instead of PdCl2(dffp), Pd(PPh3)4 was used as catalyst. The purification was carried out using MeOH/DCM (1:20) as eluent to afford 7 as yellow oil (40 mg, yield 20%, HPLC purity 99% at 254 nm, 97% at 210 nm).1H-NMR (400 MHz, Methanol-d4) δ 8.43 (s, 1H), 8.25-8.14 (m, 4H), 7.45-7.36 (m, 3H), 7.44-7.34 (m, 2H), 7.19 (t, J = 7.3 Hz, 1H), 6.72 (s, 1H), 3.87-3.80 (m, 2H), 3.79-3.74 (m, 2H), 3.74-3.67 (m, 2H), 3.65-3.60 (m, 2H); 13C-NMR (101 MHz, Methanol-d4) δ 158.7, 148.9, 146.8, 142.8, 140.1, 134.5, 130.8, 129.6, 129.5, 128.4, 126.9, 126.4, 110.2, 83.6, 73.7, 70.3, 62.2, 42.9; LC-MS: 375 [M+H]+;373 [M-H]-. [043] 2-(2-((5-Phenyl-3-(p-tolyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)ethoxy)ethan-1-ol (7i): The purification was carried out using MeOH/DCM (1:15) as eluent to afford 7i as white solid (100 mg, yield 44%, HPLC purity 97% at 254 nm, 97% at 210 nm). 1H-NMR (400 MHz, Methanol-d4) δ 8.39 (s, 1H), 8.24-8.19 (m, 2H), 8.08 (d, J = 8.9 Hz, 2H), 7.56-7.45 (m, 3H), 7.24 (d, J = 8.9 Hz, 2H), 6.72 (s, 1H), 3.87-3.81 (m, 2H), 3.80-3.75 (m, 2H), 3.73-3.68 (m, 2H), 3.66-3.61 (m, 2H), 2.37 (s, 3H); 13C-NMR (101 MHz, Methanol-d4) δ 158.5, 148.8, 146.6, 142.6, 140.1, 136.0, 131.5, 130.7, 130.1, 129.6, 128.4, 126.8, 110.3, 83.4, 73.7, 70.3, 62.2, 42.9, 21.2; LC-MS: 389.3 [M+H]+; 387.2 [M-H]-. [044] 2-(2-((5-Phenyl-3-(o-tolyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)ethoxy)ethan-1-ol (7m): The purification was carried out using MeOH/DCM (1:15) as eluent to afford 7m as white solid (100 mg, yield 44%, HPLC purity 91% at 254 nm, 92% at 210 nm). 1H-NMR (400 MHz, Methanol-d4) δ 8.09 (s, 1H), 8.08-8.04 (m, 2H), 7.58-7.53 (m, 1H), 7.49-7.41 (m, 3H), 7.32- 7.28 (m, 1H), 7.28-7.18 (m, 2H), 6.67 (s, 1H), 3.86-3.80 (m, 2H), 3.80-3.74 (m, 2H), 3.73-3.67 (m, 2H), 3.66-3.60 (m, 2H), 2.43 (s, 3H); 13C-NMR (101 MHz, Methanol-d4) δ 158.9, 149.0, 146.9, 145.0, 140.1, 138.0, 133.3, 131.9, ,131.3, 130.7, 129.6, 128.5, 127.9, 126.6, 111.3, 83.7, 73.7, 70.4, 62.3, 43.0, 21.3; LC-MS: 389.3[M+H]+; 387.2 [M-H]-. [045] 2-(2-((3-(4-Methoxyphenyl)-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7b): The purification was carried out using acetone/DCM (1:5) as eluent to afford 7b as yellow oil (86 mg, yield 36%, HPLC purity 92% at 254 nm, 93% at 210 nm). 1H-NMR (400 MHz, Methanol-d4) δ 8.36 (s, 1H), 8.18-8.24 (m, 2H), 8.11 (d, J = 8.9 Hz, 2H), 7.45-7.56 (m, 3H), 7.00 (d, J = 8.9 Hz, 2H), 6.71 (s, 1H), 3.84 (m, 3H), 3.80-3.88 (m, 2H),3.74-3.80 (m, 2H), 3.67- 3.73 (m, 2H), 3.60-3.66 (m, 2H); 13C-NMR (101 MHz, CDCl3) δ 157.9, 157.1, 147.1, 145.2,
141.4, 139.0, 129.9, 128.9, 127.5, 127.2, 125.7, 114.3, 109.7, 82.5, 72.7, 69.2, 62.0, 55.5, 42.2; LC-MS: 405.3[M+H]+. [046] 2-(2-((5-Phenyl-3-(m-tolyl)pyrazolo[1,5-a]pyrimidin-7-yl)amino)ethoxy)ethan-1- ol (7c): The purification was carried out using MeOH/DCM (1:15) as eluent to afford 7c as yellow solid (80 mg, yield 35%, HPLC purity 99% at 254 nm, 99% at 210 nm). 1H-NMR (400 MHz, Methanol-d4) δ 8.42 (s, 1H), 8.25-8.19 (m, 2H), 8.03-8.00 (m, 1H), 8.00-7.99 (m, 1H), 7.56- 7.46 (m, 3H), 7.30 (t, J = 7.8 Hz, 1H), 7.03 (d, J = 7.8 Hz, 1H), 6.73 (s, 1H), 3.88-3.81 (m, 2H), 3.81-3.74 (m, 2H), 3.74-3.67 (m, 2H), 3.66-3.60(m, 2H), 2.42 (s, 3H); 13C-NMR (101 MHz, Methanol-d4) δ 158.6, 148.8, 146.7, 142.8, 140.1, 139.0, 134.3, 130.8, 129.6, 129.4, 128.4, 127.5, 127.2, 124.1, 110.3, 83.5, 73.7, 70.3, 62.2, 42.9, 21.8; LC-MS: 389.3[M+H]+. [047] 2-(2-((3-(3-Methoxyphenyl)-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7n): The purification was carried out using acetone/DCM (1:5) as eluent to afford 7n as yellow semisolid (100 mg, yield 42%, HPLC purity 97% at 254 nm, 98% at 210 nm). 1H-NMR (400 MHz, Methanol-d4) δ 8.40 (s, 1H), 8.18-8.23 (m, 2H), 7.99 (dd, J =2.5;1.5 Hz, 1H), 7.66 (ddd, J = 8.2; 1.5; 0.9 Hz, 1H), 7.44-7.53 (m, 3H), 7.29 (t, J = 8.2 Hz, 1H), 6.76 (ddd, J = 8.2; 2.5; 0.9 Hz, 1H), 6.69 (s, 1H), 3.79-3.85 (m, 2H), 3.77-3.72(m, 2H), 3.72-3.68 (m, 2H), 3.65-3.60 (m, 2H), 3.89 (s, 3H).13C-NMR (101 MHz, Methanol-d4) δ 161.4, 158.6, 148.9, 146.8, 142.9, 140.0, 135.8, 130.8, 130.4, 129.6, 128.4, 119.0, 112.3, 112.2, 109.9, 83.6, 73.7, 70.4, 62.2, 55.6, 43.0; LC-MS: 405.2 [M+H]+. [048] 2-(2-((5-Phenyl-3-(4-(trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)-ethan-1-ol (7g): The purification was carried out using acetone/DCM (1:5) as eluent to afford 7g as yellow solid (70 mg, yield 28%, HPLC purity 91% at 254 nm, 95% at 210 nm). 1H-NMR (400 MHz, Methanol-d4) δ 8.42 (s, 1H), 8.33 (d, J = 8.2 Hz, 2H), 8.18-8.13 (m, 2H), 7.63 (d, J = 8.2 Hz, 2H), 7.51-7.42 (m, 3H), 6.64 (s, 1H), 3.80 (t, 2H, J = 5.2 Hz), 3.73-3.66 (m, 4H), 3.64-3.59 (m, 2H); 13C-NMR (101 MHz, Methanol-d4) δ 159.1, 148.9, 147.2, 143.1, 139.8, 138.6, 130.9, 129.6, 128.4, 127.6 (q, J = 32.3 Hz), 126.55, 126.3 (q, J = 3.8 Hz), 126.1(q, J = 270.0 Hz), 108.4, 84.1, 73.7, 70.3, 62.2, 43.0; LC-MS: 443.2 [M+H]+. [049] N-(3-(7-((2-(2-Hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5-a]pyrimidin-3- yl)phenyl)acetamide (7v): The purification was carried out using 10-50% acetone/DCM as eluent to afford 7v as yellow oil (65 mg, yield 25%, HPLC purity 99% at 254 nm, 99% at 210 nm). 1H-NMR (400 MHz, CDCl3) δ 8.36 (s, 1H), 8.24 (t, J = 1.3 Hz, 1H), 8.18-8.13 (m, 2H), 7.95 (dt, J = 7.9; 1.3 Hz,1H),
7.55-7.44 (m, 4H), 7.40 (t, J = 7.9 Hz, 1H), 7.23 (br s, 1H), 6.74 (t, J = 5.5 Hz, 1H), 6.46 |(s, 1H), 3.87 (t, J = 5.2 Hz, 2H), 3.84-3.79 (m, 2H), 3.73-3.66 (m, 4H), 2.30 (br s, 1H), 2.21 (s, 3H); 13C-NMR (101 MHz, CDCl3) δ 168.4, 157.7, 147.1, 145.6, 142.0, 139.0, 138.3, 133.8, 129.9, 129.4, 128.8, 127.6, 122.0, 117.3, 117.1, 109.2, 83.0, 72.8, 69.1, 62.0, 42.2, 24.9; LC- MS: 432.3 [M+H]+. [050] 2-(2-((3-(4-(Methylsulfonyl)phenyl)-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)-ethan-1-ol (7o): The purification was carried out using 10-40% acetone/DCM as eluent to afford 7o as yellow solid (180 mg, yield 67%, HPLC purity 96% at 254 nm, 97% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.39 (s, 1H), 8.38 (d, J = 8.6 Hz, 2H), 8.16-8.11 (m, 2H), 7.97 (d, J = 8.6 Hz, 2H), 7.56-7.47 (m, 3H), 6.80 (t, J= 5.5 Hz, 1H), 6.51 (s, 1H), 3.89 (t, J = 5.5 Hz, 2H), 3.84-3.79 (m, 2H), 3.72 (t, J = 5.5 Hz, 2H),3.70-3.67 (m, 2H), 3.07 (s, 3H), 2.24 (s, 1H); 13C-NMR (101 MHz, CDCl3) δ 158.4, 147.3, 146.3, 142.2, 139.0, 138.5, 136.5, 130.3, 129.0, 127.9, 127.5, 126.0, 107.8, 83.6, 72.7, 69.1, 62.0, 44.7, 42.3; LC-MS: 453.3 [M+H]+. [051] 2-(2-((3-(3-(Methylsulfonyl)phenyl)-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7p): The purification (carried out using acetone/DCM, 1:5, as eluent) followed by crystallization from MeOH afforded product 7p as yellow solid (80 mg, yield 30%, HPLC purity 98% at 254 nm, 99% at 210 nm). 1H-NMR (400 MHz, CDCl3) δ 8.79 (t, J = 1.7 Hz, 1H), 8.56 (ddd, J = 7.8; 1.7, 1.1 Hz, 1H), 8.41 (s, 1H), 8.19-8.15 (m, 2H), 7.76 (ddd, J = 7.8, 1.7, 1.1 Hz, 1H), 7.63 (t, J = 7.8 Hz, 1H), 7.57-7.47 (m, 3H), 6.79 (t, J = 5.8 Hz, 1H), 6.53 (s, 1H), 3.85-3.78 (m, 2H), 3.74 (t, J = 5.3 Hz, 2H), 3.71-3.68 (m, 2H), 3.89 (t, J = 5.3 Hz, 2H), 3.13 (s, 3H), 2.15 (t, J = 5.3 Hz, 1H); 13C-NMR (101 MHz, CDCl3) δ 158.0, 147.3, 141.7, 145.9, 140.9, 138.4, 134.8, 130.6, 130.3, 129.7, 128.9, 127.5, 124.2, 123.7, 107.6, 83.2, 72.8, 69.1, 62.0, 44.7, 42.3; LC- MS: 453.2 [M+H]+. [052] N-(4-(7-((2-(2-Hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5-a]pyrimidin-3- yl)phenyl)acetamide (7l): The purification was carried out using acetone/DCM (1:1.7) as eluent to afford 7l as yellow solid (170 mg, yield 66%, HPLC purity 98% at 254 nm, 98% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.32 (s, 1H), 8.17 (d, J = 8.6 Hz, 2H), 8.16-8.13 (m, 2H), 7.58 (d, J = 8.6 Hz, 2H), 7.55-7.44 (m, 3H), 7.16 (s, 1H), 6.73 (t, J = 5.5 Hz, 1H), 6.46 (s, 1H), 3.88 (t, J = 5.2 Hz, 2H), 3.84-3.77 (m, 2H), 3.71(t, J = 5.5 Hz, 2H), 3.70-3.66 (m, 2H), 2.20 (s, 3H), 1.26-1.23 (m, 1H); 13C-NMR (101 MHz, CDCl3) δ 168.1, 155.9, 147.3, 144.8, 141.3, 138.2, 136.7, 129.8, 128.7, 127.9, 127.2, 125.4, 119.2, 107.6, 82.4, 72.2, 68.7, 60.3, 41.4, 24.0; LC-MS: 432.3 [M+H]+.
[053] 2-(2-((3-(3-(Tert-butyl)phenyl)-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7ac): The purification was carried out using acetone/DCM (1:5) as eluent to afford 7ac as yellow solid (46 mg, yield 18%, HPLC purity 98% at 254 nm, 99% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.36 (s, 1H), 8.45 (t, J = 1.8 Hz, 1H), 8.22-8.17 (m, 2H), 7.88 (ddd, J = 7.8; 1.8; 1.2 Hz, 1H), 7.54 -7.44 (m, 3H), 7.39 (t, J = 7.8 Hz, 1H), 7.29 (ddd, J = 7.8, 1.8, 1.2 Hz, 1H), 6.47 (s, 1H), 6.74 (t, J = 5.5 Hz, 1H),3.87 (t, J = 5.4 Hz, 2H), 3.83-3.78 (m, 2H), 3.71 (t, J = 5.5 Hz, 2H), 3.69-3.66 (m, 2H), 1.44 (s, 9H); 13C-NMR (101 MHz, CDCl3) δ 157.1, 151.5, 147.2, 145.6, 141.9, 138.9, 132.6, 129.9, 128.7, 128.4, 127.4, 123.5, 123.1, 122.9, 110.3, 82.5, 72.8, 69.2, 62.0, 42.3, 35.0, 31.6; LC-MS: 431.4 [M+H]+; 430.2 [M-H]-. [054] 2-(2-((3-(2-Methoxyphenyl)-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7y): The purification was carried out using acetone/DCM (1:5) as eluent to afford 7y as yellow solid (24 mg, yield 10%, HPLC purity 94% at 254 nm, 96% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.68 (s, 1H), 8.70 (dd, J = 7.4; 1.8 Hz, 1H), 8.14-8.09 (m, 2H), 7.51-7.41 (m, 3H), 7.24 (ddd, J = 8.2; 7.4; 1.8 Hz, 1H), 7.14 (td, J = 7.4; 1.2 Hz, 1H), 6.81 (t, J = 5.5 Hz, 1H), 6.99 (dd, J = 8.2; 1.2 Hz, 1H), 6.39 (s, 1H), 3.92 (s, 1H), 3.83-3.75 (m, 4H), 3.66-3.58 (m, 4H); 13C-NMR (101 MHz, CDCl3) δ 157.0, 156.3, 147.0, 145.9, 145.1, 138.8, 129.7, 129.5, 128.7, 127.4, 126.7, 121.8, 121.0, 111.1, 105.9, 82.6, 72.7, 67.0, 61.8, 55.5, 42.1; LC-MS: 405.3 [M+H]+. [055] 2-(2-((5-Phenyl-3-(2-(trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7x): The purification was carried out using acetone/DCM (1:5) as eluent to afford 7x as white solid (10 mg, yield 4%, HPLC purity 97% at 254 nm, 96% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.21 (s, 1H), 8.06-8.02 (m, 2H), 7.49-7.39 (m, 4H), 7.98 (d, J = 7.8 Hz, 1H), 7.78 (d, J = 7.8 Hz, 1H), 7.63 (t, J = 7.8 Hz, 1H), 6.79 (t, J = 5.4 Hz, 1H), 6.40 (s, 1H), 3.90 (t, J = 5.2 Hz, 2H), 3.85-3.80 (m, 2H), 3.73 (t, J = 5.4 Hz, 2H), 3.71-3.68 (m, 2H), 2.14 (t, J = 5.8 Hz, 1H); 13C- NMR (101 MHz, CDCl3) δ 158.1, 147.4, 145.5, 144.4 (q, J = 4.0 Hz), 137.9, 133.6, 131.6, 130.7 (q, J = 1.7 Hz), 130.2, 128.8, 128.6 (q, J = 30.0 Hz), 127.6, 127.0, 126.5 (q, J = 5.5 Hz), 124.6(q, J =274.7 Hz), 107.6, 83.5, 72.8, 69.0, 61.9, 42.3; LC-MS: 443.3 [M+H]+. [056] 2-(2-((5-Phenyl-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7a): The purification was carried out using 0-30% acetone/DCM as eluent to afford 7a as yellow solid (70 mg, yield 27%, HPLC purity 97% at 254 nm, 98% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.53 (s, 1H), 8.39-8.34 (m, 2H), 8.18-8.12 (m, 2H), 7.56-7.42 (m, 5H), 6.77 (t, J = 5.6 Hz, 1H), 6.40 (s, 1H), 3.88 (t, J = 5.2 Hz, 2H), 3.82 (m, 2H), 3.71 (t, J = 5.4 Hz, 2H), 3.70-
3.67 (m, 2H), 2.39 (t, J = 5.7 Hz, 1H); 13C-NMR (101 MHz, CDCl3) δ 157.2, 147.2, 145.8, 141.7, 138.5, 133.9, 131.0 (q, J = 31.9 Hz), 130.2, 129.1, 128.9, 128.8, 127.4, 124.6 (q, J = 272.7 Hz), 122.5 (q, J = 4.0 Hz), 122.1 (q, J = 3.7 Hz), 108.3, 83.0, 72.8, 69.1, 62.0, 42.3; MS: 443.3 [M+H]+; 441.3 [M-H]-. [057] 2-(2-((3-(4-(Tert-butyl)phenyl)-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7t): The purification was carried out using 0-50% acetone/DCM as eluent to afford 7t as yellow solid (172 mg, yield 68%, HPLC purity 97% at 254 nm, 98% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.34 (s, 1H), 8.19-8.14 (m, 2H), 8.12 (d, J = 8.7 Hz, 2H), 7.54-7.42 (m, 5H), 6.73 (t, J = 5.7 Hz, 1H), 6.45 (s, 1H), 3.87 (t, J = 5.2 Hz, 2H), 3.83-3.78 (m, 2H), 3.71 (t, J = 5.4 Hz, 2H), 3.70-3.66 (m, 2H), 2.30 (br s, 1H), 1.37 (s, 9H); 13C-NMR (101 MHz, CDCl3) δ 157.2, 148.6, 147.1, 145.5, 141.8, 139.0, 130.1, 129.8, 128.8, 127.5, 125.8, 125.7, 109.9, 82.6, 72.7, 69.2, 62.0, 42.2, 34.6, 31.5; MS: 431.3 [M+H]+. [058] 2-(2-((5-Phenyl-3-(4-(trifluoromethoxy)phenyl)pyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7e): The purification was carried out using 15-40% acetone/DCM as eluent to afford 7e as white solid (70 mg, yield 26%, HPLC purity 98% at 254 nm, 99% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.33 (s, 1H), 8.22 (d, J = 8.7 Hz, 2H), 8.17-8.12 (m, 2H), 7.57-7.45 (m, 3H), 7.30 (d, J
(t, J = 5.6 Hz, 1H), 6.49 (s, 1H), 3.89 (t, J = 5.6 Hz, 2H), 3.85-3.78 (m, 2H), 3.73 (t, J = 5.3 Hz, 2H), 3.71-3.67 (m, 2H), 2.15 (t, J = 5.9 Hz, 1H); 13C-NMR (101 MHz, CDCl3) δ 157.8, 147.2, 147.1 (q, J = 1.8 Hz), 145.7, 141.8, 138.8, 132.0, 130.1, 128.9, 127.5, 127.1, 121.5, 120.8 (q, J = 256.7 Hz), 108.5, 83.0, 72.7, 69.2, 62.0, 42.3; MS: 459.2 [M+H]+. [059] 2-(2-((5-Phenyl-3-(3-(trifluoromethoxy)phenyl)pyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7h): The purification was carried out using 15-40% acetone/DCM as eluent to afford 7h as yellow semisolid (57 mg, yield 21%, HPLC purity 91% at 254 nm, 95% at 210 nm). 1H-NMR (400 MHz, CDCl3) δ 8.33 (s, 1H), 8.23 (m, 1H), 8.17-8.12 (m, 2H), 8.03 (ddd, J = 7.9; 1.5; 0.9 Hz, 1H), 7.55-7.43 (m, 3H), 7.43 (t, J = 7.9Hz, 1H), 7.05 (m, 1H), 6.76 (t, J = 5.6 Hz, 1H), 6.48 (s, 1H), 3.87 (t, J = 5.1 Hz, 2H), 3.84-3.78 (m, 2H), 3.72-3.65 (m, 4H), 2.42 (br s, 1H); 13C-NMR (101 MHz, CDCl3) δ 157.8, 149.9 (q, J = 1.8 Hz), 147.2, 145.8, 141.8, 138.5, 135.1, 130.1, 129.9, 128.8, 127.5, 123.8, 120.8 (q, J = 256.6 Hz), 118.2, 117.7, 108.3, 83.0, 72.8, 69.1, 62.0, 42.2; MS: 459.2 [M+H]+. [060] N-(4-(7-((2-(2-Hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5-a]pyrimidin-3- yl)phenyl)methanesulfonamide (7r):
The purification was carried out using 15-70% acetone/DCM as eluent to afford 7r as white semisolid (195 mg, yield 71%, HPLC purity 99% at 254 nm, 99% at 210 nm). 1H-NMR (400 MHz, CDCl3) δ 8.31 (s, 1H), 8.20-8.16 (m, 2H), 8.16-8.12 (m, 2H), 7.55-7.45 (m, 3H), 7.33- 7.28 (m, 2H), 6.75 (t, J = 5.5 Hz, 1H), 6.47 (s, 1H), 6.42 (br s, 1H),3.89 (t, J = 5.2 Hz, 2H), 3.85-3.79 (m, 2H), 3.72 (t, J = 5.4 Hz, 2H), 3.71-3.67 (m, 2H), 3.02 (s, 3H), 2.26 (t, J = 5.9 Hz, 1H); 13C-NMR (101 MHz, CDCl3) δ 157.7, 147.2, 145.6, 141.6, 138.8, 133.9, 131.0, 130.11, 129.0, 127.5, 127.1, 122.0, 108.8, 82.9, 72.7, 69.2, 62.0, 42.3, 39.3; MS: 468.3 [M+H]+. [061] N-(3-(7-((2-(2-Hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5-a]pyrimidin-3- yl)phenyl)methanesulfonamide (7w): The purification was carried out using 15-50% acetone/DCM as eluent, followed by repeated column chromatography using 70-90% EtOAc/PE as eluent, to afford 7w as yellow solid (115 mg, yield 42%, HPLC purity 99% at 254 nm, 99% at 210 nm). 1H-NMR (400 MHz, DMSO- d6) δ 9.79 (s, 1H), 8.59 (s, 1H), 8.39-8.34 (m, 2H), 8.31 (t, J = 1.8 Hz, 1H), 7.99-7.93 (m, 1H), 7.86 (ddd, J = 8.0;1.8;1.0 Hz, 1H), 7.58-7.48 (m, 3H), 7.38 (t, J = 8.0 Hz, 1H), 7.02 (ddd, J = 8.0; 1.8;1.0 Hz, 1H), 6.90 (s, 1H), 4.66-4.59 (m, 1H), 3.79-3.70 (m, 4H), 3.54-3.47 (m, 4H), 3.06 (s, 3H); 13C-NMR (101 MHz, DMSO-d6) δ 156.3, 147.4, 145.1, 141.7, 138.7, 138.0, 134.1, 129.9, 129.4, 128.6, 127.4, 120.7, 116.9, 116.5, 107.3, 82.6, 72.2, 68.7, 60.3, 41.4, one signal was overlapping with DMSO; MS: 468.3 [M+H]+. [062] 4-(7-((2-(2-Hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5-a]pyrimidin-3- yl)benzonitrile (7u): The purification was carried out using 20-40% acetone/DCM as eluent to afford 7u as yellow solid (47 mg, yield 20%, HPLC purity 98% at 254 nm, 99% at 210 nm).1H-NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 8.30 (d, J = 8.5 Hz, 2H), 8.15-8.10 (m, 2H), 7.68 (d, J = 8.5 Hz, 2H), 7.56-7.48 (m, 3H), 6.80 (t, J = 5.5 Hz, 1H), 6.51 (s, 1H), 3.89 (t, J = 5.1 Hz, 2H), 3.85-3.78 (m, 2H), 3.72 (t, J = 5.3 Hz, 2H), 3.71-3.67 (m, 2H), 2.27 (br s, 1H); 13C-NMR (101 MHz, DMSO- d6) δ 158.4, 147.3, 146.2, 142.1, 138.5, 138.0, 132.6, 130.3, 128.9, 127.5, 125.8, 119.8, 108.2, 107.8, 83.5, 72.8, 69.1, 62.0, 42.3; MS: 400.3 [M+H]+. [063] Methyl 4-(7-((2-(2-hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5- a]pyrimidin-3-yl)-benzoate (7f): The purification was carried out using 15-30% acetone/DCM as eluent to afford 7f as yellow solid (100 mg, yield 45%, HPLC purity 99% at 254 nm, 99% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.40 (s, 1H), 8.18-8.14 (m, 2H), 8.30 (d, J = 8.6 Hz, 2H), 8.11 (d, J = 8.6 Hz, 2H), 7.56-7.45 (m, 3H), 6.50 (s, 1H), 6.77 (t, J = 5.6 Hz, 1H), 3.93 (s, 3H), 3.88 (t, J = 5.2 Hz, 2H), 3.85-3.80 (m, 2H), 3.72 (t, J = 5.4 Hz, 2H), 3.70-3.67 (m, 2H), 2.22 (t, J = 5.1 Hz, 1H); 13C-
NMR (101 MHz, CDCl3) δ 167.4, 158.0, 147.2, 146.1, 142.2, 138.6, 138.0, 130.22, 130.20, 128.9, 127.5, 126.8, 125.3, 108.7, 83.2, 72.7, 69.2, 62.0, 52.0, 42.3; MS: 433.3 [M+H]+. [064] 3-(7-((2-(2-Hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5-a]pyrimidin-3- yl)benzonitrile (7d): The purification was carried out using 15-30% acetone/DCM as eluent to afford 7d as yellow solid (118 mg, yield 50%, HPLC purity 99% at 254 nm, 99% at 210 nm).1H-NMR (300 MHz, CDCl3) δ 8.47-8.43 (m, 1H), 8.36 (dt, J = 7.6;1.7 Hz, 1H), 8.28 (s, 1H), 8.12-8.07 (m, 2H), 7.55-7.41 (m, 5H), 6.78 (t, J = 5.5 Hz, 1H), 6.45 (s, 1H), 3.87 (t, J = 5.2 Hz, 2H), 3.85-3.79 (m, 2H), 3.73-3.66 (m, 4H), 2.56 (t, J= 5.5 Hz, 1H); 13C-NMR (101 MHz, CDCl3) δ 158.1, 147.2, 145.8, 141.6, 138.4, 134.4, 130.3, 129.7, 129.4, 129.0, 128.9, 128.7, 127.4, 119.6, 112.7, 107.3, 83.3, 72.8, 69.0, 61.9, 42.2; MS: 400.2 [M+H]+. [065] 2-(2-((5-Phenyl-3-(2-(trifluoromethoxy)phenyl)pyrazolo[1,5-a]pyrimidin-7- yl)amino)-ethoxy)ethan-1-ol (7aa): The purification was carried out using 15-30% acetone/DCM as eluent to afford 7aa as white solid (97 mg, yield 36%, HPLC purity 98% at 254 nm, 98% at 210 nm).1H-NMR (300 MHz, CDCl3) δ 8.72 (dd, J = 7.9; 1.4 Hz, 1H), 8.52 (s, 1H), 8.14-8.08 (m, 2H), 7.54-7.45 (m, 3H), 7.42 (ddd, J = 7.9;7.2;1.4Hz, 1H), 7.35 (m, 1H), 7.25 (ddd, J = 8.2;7.2;1.5 Hz, 1H, the signals overlapped with CDCl3), 6.48 (s, 1H), 6.83 (t, J = 5.4 Hz, 1H), 3.87 (t, J = 5.2 Hz, 2H), 3.84- 3.78 (m, 2H), 3.72-3.66 (m, 4H), 2.42 (br s, 1H); 13C-NMR (101 MHz, CDCl3) δ 157.9, 147.2, 146.3, 145.9, 144.3, 138.8, 130.5, 130.0, 128.8, 127.5, 127.1, 126.6, 126.3, 120.9, 120.8 (q, J = 257.5 Hz), 104.5, 83.1, 72.8, 69.2, 62.0, 42.2; MS: 459.3 [M+H]+. [066] Methyl 3-(7-((2-(2-hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5- a]pyrimidin-3-yl)benzoate (7k): The purification was carried out using 15-30% acetone/DCM as eluent to afford 7k as white solid (107 mg, yield 42%, HPLC purity 99% at 254 nm, 99% at 210 nm).1H-NMR (300 MHz, CDCl3) δ 8.87 (dd, J = 1.9;1.6 Hz, 1H), 8.51 (ddd, J = 7.8;1.9;1.2 Hz, 1H), 8.41 (s, 1H), 8.22- 8.16 (m, 2H), 7.90 (ddd, J = 7.7;1.6;1.2 Hz, 1H), 7.57-7.46 (m, 4H), 6.77 (t, J = 5.5 Hz, 1H), 6.50 (s, 1H), 3.98 (s, 3H), 3.89 (t, J = 5.2 Hz, 2H), 3.85-3.78 (m, 2H), 3.73 (t, J = 5.5 Hz, 2H), 3.72-3.66 (m, 2H), 2.20 (t, J = 5.2 Hz, 1H); 13C-NMR (101 MHz, CDCl3) δ 167.6, 157.6, 147.2, 145.8, 141.8, 138.7, 133.4, 130.5, 130.2, 130.1, 128.8 (2C), 127.5, 126.8, 126.6, 108.7, 82.8, 72.8, 69.2, 62.0, 52.2, 42.3; MS: 433.3 [M+H]+; 431.3 [M-H]- . [067] Methyl 2-(7-((2-(2-hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5- a]pyrimidin-3-yl)benzoate (7z):
The purification was carried out using 5-15% acetone/DCM as eluent to afford 7z as brown solid (10 mg, yield 5%).1H-NMR (400 MHz, CDCl3) δ 8.18 (s, 1H), 8.14-8.10 (m, 2H), 7.86 (ddd, J = 7.8; 1.4; 0.4 Hz, 1H), 7.70 (ddd, J = 7.8; 1.4; 0.4 Hz, 1H), 7.54 (td, J = 7.8; 1.4 Hz, 1H), 7.50-7.40 (m, 3H), 7.34 (td, J = 7.8; 1.4 Hz, 1H), 6.77 (t, J = 5.6 Hz, 1H), 6.46 (s, 1H), 3.87 (t, J = 5.2 Hz, 2H), 3.83-3.79 (m, 2H), 3.72-3.66 (m, 4H), 3.57 (s, 3H); 13C-NMR (101 MHz, CDCl3) δ 170.0, 157.3, 147.2, 145.9, 143.0, 138.4, 132.0, 131.4, 131.1, 130.9, 130.3, 130.0, 128.8, 127.4, 126.3, 110.0, 82.5, 72.7, 69.2, 62.0, 52.1, 42.3; MS: 433.3 [M+H]+; 431.1 [M-H]- . [068] N-(2-(7-((2-(2-hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5-a]pyrimidin-3- yl)phenyl)acetamide (7ad): The purification was carried out using 10-30% acetone/DCM as eluent to afford 7ad as yellow solid (80 mg, yield 36%, HPLC purity 98% at 254 nm, 98% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 11.32 (s, 1H), 8.14 (s, 1H), 8.11-8.06 (m, 2H), 8.04 (dd, J = 8.2; 1.1 Hz, 1H), 7.56- 7.47 (m, 4H), 7.35 (ddd, J = 8.2; 7.6; 1.6 Hz, 1H), 7.20 (td, J = 7.6; 1.1Hz, 1H), 6.99 (t, J = 5.5 Hz, 1H), 6.49 (s, 1H), 3.89 (t, J = 5.2 Hz, 2H), 3.84-3.79 (m, 2H), 3.72 (t, J = 5.5 Hz, 2H), 3.70- 3.67 (m, 2H), 1.73 (s, 3H); 13C-NMR (101 MHz, CDCl3) δ 169.0, 156.9, 147.8, 144.3, 144.2, 137.7, 135.5, 130.7, 129.5, 129.1, 127.4, 127.2, 125.2, 124.9, 124.7, 109.4, 82.2, 72.8, 69.1, 62.0, 42.4, 24.3; MS: 432.3 [M+H]+; 430.3 [M-H]-. [069] N-(2-(7-((2-(2-hydroxyethoxy)ethyl)amino)-5-phenylpyrazolo[1,5-a]pyrimidin-3- yl)phenyl)methanesulfonamide (7ab): The purification was carried out using acetone/DCM (1:5) as eluent to afford 7ab as white solid (33 mg, yield 12%).1H-NMR (400 MHz, DMSO-d6) δ 11.11 (s, 1H), 8.57 (s, 1H), 8.35 (t, J = 5.9 Hz, 1H), 8.25-8.18 (m, 2H), 7.80-7.74 (m, 1H), 7.59-7.47 (m, 4H), 7.38-7.30 (m, 2H), 6.93 (s, 1H), 4.63 (m, 1H), 3.82-3.71 (m, 4H), 3.51 (d, J = 2.7 Hz, 4H), 2.58 (s, 3H).13C-NMR (101 MHz, DMSO-d6) δ 156.5, 148.0, 143.9, 143.8, 137.3, 133.7, 130.3, 129.6, 128.8, 127.4, 127.2, 127.1, 126.5, 126.3, 106.6, 83.1, 72.2, 68.7, 60.3, 41.6. The signal of SO2Me is overlapped with DMSO-d 6 . MS: 468.3 [M+H]+; 466.2 [M-H]-. [070] 2-(2-((3-(1H-Indol-4-yl)-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7s): The purification was carried out using 5%-30% acetone/DCM as eluent to afford 7s as brown solid (22 mg, yield 9%).1H-NMR (300 MHz, CD3OD) δ 8.49 (s, 1H), 8.20-8.16 (m, 2H), 7.81 (dd, J = 7.3;0.8 Hz, 1H), 7.52-7.43 (m, 3H), 7.34 (dt, J = 8.1;0.8 Hz, 1H), 7.30 (d, J = 3.2 Hz, 1H), 7.23 (dd, J = 8.1;7.3 Hz, 1H), 6.82 (dd, J = 3.2;0.8 Hz, 1H), 6.73 (s, 1H), 3.89-3.83 (m, 2H), 3.82-3.77 (m, 2H), 3.74-3.69 (m, 2H), 3.67-3.62 (m, 2H).13C-NMR (101 MHz, CDCl3) δ
157.2, 147.2, 145.9, 143.7, 139.0, 136.6, 129.8, 128.7, 127.5, 126.1, 125.2, 124.0, 122.6, 120.0, 110.5, 109.3, 103.0, 82.7, 72.7, 69.2, 62.0, 42.3. MS: 414.3 [M+H]+. [071] 2-(2-((3-(3-(Hydroxymethyl)phenyl)-5-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ol (7j): The purification was carried out using 15%-40% acetone/DCM as eluent to afford 7j as yellow solid (114 mg, yield 48%, HPLC purity 99% at 254 nm, 99% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 8.16-8.06 (m, 4H), 7.53-7.44 (m, 3H), 7.40 (t, J = 7.7 Hz, 1H), 7.21 (dt, J = 7.7;1.3 Hz, 1H), 6.71 (t, J = 5.5 Hz, 1H), 6.39 (s, 1H), 4.75 (s, 2H), 3.86 (t, J = 5.2 Hz, 2H), 3.83-3.79 (m, 2H), 3.70-3.63 (m, 4H), 2.74 (br s, 1H), 2.31 (br s, 1H). 13C-NMR (101 MHz, CDCl3) δ 157.4, 147.1, 145.6, 141.8, 141.2, 138.8, 133.2, 130.0, 129.0, 128.8, 127.5, 125.2, 124.6, 124.5, 109.4, 82.8, 72.9, 69.0, 65.9, 62.0, 42.2. MS: 405.3 [M+H]+; 403.3 [M-H]- . [072] The synthesis of 2-(2-((5-aryl-3-phenylpyrazolo[1,5-a]pyrimidin-7- yl)amino)ethoxy)ethan-1-ols Scheme 2
[073] Synthesis of N-(2-(2-((tert-butyldimethylsilyl)oxy)ethoxy)ethyl)-5-chloro-3- phenylpyrazolo[1,5-a]pyrimidin-7-amine (int4) A mixture of 5,7-dichloro-3-phenylpyrazolo[1,5-a]pyrimidine (3.3 g, 12.5 mmol), 2-(2-((tert- butyldimethylsilyl)oxy)ethoxy)ethan-1-amine (4.1 g, 1.5 eq) and K2CO3 (8.6 g, 5 eq) in MeCN (100 ml) was stirred at RT for 18h. The solvent was removed to give an oily residue that was dissolved in EtOAc (50 ml). The obtained solution was washed with brine (2x15 ml), dried over Na2SO4 and the solvent was removed to give oil that was purified by column chromatography (5%-20% EtOAc/PE) to afford the desired product as colourless oil (5.0 g, yield 90%). 1H-NMR (300 MHz, CDCl3) δ 8.30 (s, 1H), 8.00 (d, J = 7.8 Hz, 2H), 7.43 (t, J = 7.8 Hz, 2H), 7.24 (t, J = 7.8 Hz, 1H, overlapped with CDCl3), 6.76 (t, J = 5.3 Hz, 1H), 6.00 (s, 1H), 3.86- 3.77 (m, 4H), 3.64-3.54 (m, 4H), 0.90 (s, 9H), 0.08 (s, 6H); MS: 447.3 [M (35Cl)+H]+, 449.2 [M (37Cl)+H]+.
[074] tert-Butyl (2-(2-((tert-butyldimethylsilyl)oxy)ethoxy)ethyl)(5-chloro-3- phenylpyrazolo[1,5-a]pyrimidin-7-yl)carbamate (int5) int5 was prepared using the method described previously for tert-butyl (2-(2-((tert- butyldimethylsilyl)oxy)ethoxy)ethyl)(5-phenylpyrazolo[1,5-a]pyrimidin-7-yl)carbamate (see Scheme 1). Isolated crude product was purified by column chromatography (10%-30% EtOAc/PE) to give the desired product as colourless oil (yield 90%). 1H-NMR (300 MHz, CDCl3) δ 8.42 (s, 1H), 8.04-7.98 (m, 2H), 7.49-7.41 (m, 2H), 7.32-7.24 (m, 1H, overlapped with CDCl3), 6.96 (s, 1H), 3.99 (t, J = 5.2 Hz, 2H), 3.71 (t, J = 5.2 Hz, 2H), 3.58-3.52 (m, 2H), 3.43-3.38 (m, 2H), 1.37 (s, 9H), 1.84 (s, 9H), 0.01 (s, 6H); MS: 547.3 [M (35Cl)+H]+, 549.3 [M (37Cl)+H]+. [075] 4-(7-((2-(2-Hydroxyethoxy)ethyl)amino)-3-phenylpyrazolo[1,5-a]pyrimidin-5- yl)phenol (7ak): 7ak was prepared from int5 and 4-hydroxyphenylboronic acid by following the procedure described in general method A. The purification was carried out using 10-30% acetone/DCM as eluent to afford 7ak as white solid (yield 42%, HPLC purity 98% at 254 nm, 97% at 210 nm).1H-NMR (400 MHz, DMSO-d6) δ 9.90 (s, 1H), 8.60 (s, 1H), 8.28-8.22 (m, 2H), 8.15 (d, J = 8.7 Hz, 2H), 7.82-7.76 (m, 1H), 7,43 (t, J = 7.7 Hz, 2H), 7.22-7.15 (m, 1H), 6.93 (d, J = 8.7 Hz, 2H), 6.76 (s, 1H), 4.69-4.62 (m, 1H), 3.77-3.68 (m, 4H), 3.56-3.48 (m, 4H).13C-NMR (101 MHz, DMSO-d6) δ 159.3, 156.3, 147.2, 145.2, 141.5, 133.2, 129.0, 128.8, 128.6, 125.1, 125.0, 115.4, 107.2, 81.6, 72.3, 68.7, 60.3, 41.3. MS: 391.3 [M+H]+; 389.2 [M-H]-. [076] 3-(7-((2-(2-Hydroxyethoxy)ethyl)amino)-3-phenylpyrazolo[1,5-a]pyrimidin-5- yl)phenol (7al): 7al was prepared from int5 and 3-hydroxyphenylboronic acid by following the procedure described in general method A. The purification was carried out using 10-30% acetone/DCM as eluent to afford 7al as white solid (yield 80%, HPLC purity 99% at 254 nm, 99% at 210 nm). 1H-NMR (400 MHz, DMSO-d6) δ 9.64 (s, 1H), 8.65 (s, 1H), 8.29-8.22 (m, 2H), 7.94-7.86 (m, 1H), 7.72-7.63 (m, 2H), 7.44 (t, J = 7.7 Hz, 2H), 7.34 (t, J = 7.9 Hz, 1H), 7.24-7.16 (m, 1H), 6.90 (dd, J = 7.9; 0.7 Hz, 1H), 6.77 (s, 1H), 4.67-4.60 (m, 1H), 3.73 (s, 4H), 3.53-3.48 (m, 4H). 13C-NMR (101 MHz, DMSO-d6) δ 157.6, 156.3, 147.3, 145.0, 141.7, 139.7, 133.1, 129.7, 128.6, 125.2 (2C), 118.1, 116.9, 114.0, 107.6, 82.6, 72.3, 68.7, 60.3, 41.4. MS: 391.3 [M+H]+; 389.2 [M-H]-. [077] Synthesis of 3,5-diphenylpyrazolo[1,5-a]pyrimidin-7-amines Scheme 3
[078] 7-Chloro-3,5-diphenylpyrazolo[1,5-a]pyrimidine (int6) was prepared by method described by Yin L., 2005, Synthesis of new calcineurin inhibitors via palladium catalysis cross coupling reactions, (20121004). [079] Synthesis of 5-((3,5-diphenylpyrazolo[1,5-a]pyrimidin-7-yl)amino)pentanoic acid (7az) A mixture of int6 (300 mg, 1.0 mmol), 5-aminopentanoic acid (172 mg, 1.5 eq) and DIPEA (0.9 ml, 5 eq) in DMAA (5 ml) was stirred at 80^C for 5h, then cooled to RT and poured on ice. The solid was filtered off and dried in air. The crude product was purified by column chromatography (acetone /DCM 1:5) to afford desired 7az as yellow solid (100 mg, yield 26%, HPLC purity 99% at 254 nm, 98% at 210 nm).1H-NMR (400 MHz, DMSO-d6) δ 12.04 (br s, 1H), 8.65 (s, 1H), 8.31-8.23 (m, 4H), 8.13 (t, J = 6.1 Hz, 1H), 7.59-7.48 (m, 3H), 7.47-7.40 (m, 2H), 7.19 (tt, J = 7.3; 1.1 Hz, 1H), 6.78 (s, 1H), 3.56 (m, 2H), 2.30 (t, J = 7.2 Hz, 2H), 1.79- 1.68 (m, 2H), 1.68-1.58 (m, 2H).13C-NMR (101 MHz, DMSO-d6) δ 174.5, 156.2, 147.2, 145.1, 141.7, 138.3, 133.1, 129.8, 128.7, 128.6, 127.2, 125.2, 125.2, 107.6, 82.2, 40.95, 33.3, 27.9, 21.9. MS: 387.3 [M+H]+; 385.2 [M-H]-. [080] Synthesis of N-(1H-imidazol-2-yl)-3,5-diphenylpyrazolo[1,5-a]pyrimidin-7-amine (7ax) In a flask were charged int6 (100 mg, 0.32 mmol), 2-amino-1-Boc-imidazole (88 mg, 1.5 eq), t-BuOK (90 mg, 2.5 eq), Pd(dba)2 (15 mg, 5mol-%) and dioxane (5 ml), then Xantphos (18 mg, 10 mol-%) was introduced under argon. The reaction mixture was heated and stirred at 100^C for 18h, then cooled to RT, diluted with water (10 ml) and extracted with EtOAc (3^10 ml). Combined organic extracts were dried over Na2SO4 and concentrated in vacuo. The obtained residue was subjected to column chromatography (10%-100% EtOAc/PE). Isolated 7ax-Boc was dissolved in DCM (5 ml) and TFA (1 ml) and stirred for 2h at RT, then concentrated in vacuo. The obtained oil was dissolved in EtOAc (20 ml) and washed with aq NaHCO3 (2^5 ml). EtOAc layer was dried over Na2SO4 and concentrated in vacuo. The residue was crystallized from toluene to give 7ax as yellow solid (25 mg, yield 22%, HPLC purity 99% at 254 nm, 98% at 210 nm); 1H-NMR (400 MHz, DMSO-d6) δ 11.02 (br s, 1H), 8.78 (s, 1H), 8.33-8.27 (m, 2H),
8.23-8.12 (m, 3H), 7.65-7.52 (m, 3H), 7.47 (t, J = 7.8 Hz , 2H), 7.26-7.19 (m, 1H), 6.97 (s, 2H). 13C-NMR (101 MHz, DMSO-d6) δ 156.5 (2C), 145.1, 143.2, 141.8, 141.5, 138.0, 132.8, 130.1, 129.0, 128.7, 127.0, 125.5, 125.4, 108.4, 87.2. MS: 353.2 [M+H]+; 351.2 [M-H]-. [081] Synthesis of N-(1H-imidazol-5-yl)-3,5-diphenylpyrazolo[1,5-a]pyrimidin-7-amine (7ay) A suspension of 1H-imidazol-4-amine hydrochloride (38 mg, 1.5 eq) and NaH (40 mg, 6 eq, 60% suspension in mineral oil) in DMAA (4 ml) was stirred for 1h at RT, then int6 (50 mg, 0.16 mmol) was added and the reaction mixture was stirred at 60^C for 2h. After cooling to RT, the mixture was quenched with 5% aq AcOH (5 ml) and extracted with EtOAc (3^15 ml). Combined organics were dried over Na2SO4, concentrated in vacuo. The obtained residue was subjected to column chromatography (0%-50% MeOH/DCM) to afford 7ay as a mixture of tautomers in ratio 1:1 (10 mg, yield 18%, HPLC purity 93% at 254 nm, 95% at 210 nm). 1H- NMR (400 MHz, DMSO-d6) δ 12.46 (s, 0.5H), 11.60 (s, 0.5H), 8.78 (s, 0.5H), 8.74 (s, 0.5H), 8.39-8.19 (m, 4H), 7.91 (s, 0.5H), 7.75 (s, 0.5H), 7.66-7.50 (m, 3.5 H), 7.51-7.42 (m, 2.5H), 7.24 (t, J = 7.4 Hz, 1H), 7.19 (s, 1H), 6.40 (s, 1H). 13C-NMR (101 MHz, CD3OD) δ 156.9 (2C), 147.3, 142.4, 139.0, 133.8, 131.0, 129.8, 129.6, 129.4, 129.1, 128.3, 127.0, 126.7, 126.2, 110.3, 100.1. MS: 353.2 [M+H]+. [082] Synthesis of N-(2-methoxyethyl)-3-arylpyrazolo[1,5-a]pyrimidin-7-amines Scheme 4
[083] Synthesis of N-(2-methoxyethyl)-5-methylpyrazolo[1,5-a]pyrimidin-7-amine (int7a)
A mixture of 7-chloro-5-methylpyrazolo[1,5-a]pyrimidine (1.5 g, 9.0 mmol), 2- methoxyethylamine (1.2 ml, 1.5 eq) and K2CO3 (6.2 g, 5 eq) in MeCN (40 ml) was refluxed for 18h, then cooled to RT and concentrated in vacuo. The resulting residue was dissolved in water (30 ml) and DCM (30 ml). The phases were separated, and the water layer was extracted with DCM (1^25 ml). Combined organics were dried over Na2SO4 and concentrated in vacuo. The desired product was isolated as colorless oil (1.6 g, 90 %).1H-NMR (300 MHz, CDCl3) δ 7.95 (d, J = 2.3 Hz, 1H), 6.49 (br s, 1H), 6.37 (d, J = 2.3 Hz, 1H), 5.82 (s, 1H), 3.70-3.66 (m, 2H), 3.57-3.51 (m, 2H), 3.42 (s, 3H), 2.50 (s, 3H); MS: 207.1 [M+H]+. [084] N-(2-methoxyethyl)-5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-amine (int7b) was prepared from 7-chloro-5,6-dimethylpyrazolo[1,5-a]pyrimidine using the method described previously for the synthesis of N-(2-methoxyethyl)-5-methylpyrazolo[1,5-a]pyrimidin-7- amine. The isolated crude product was purified by column chromatography (10%-30% EtOAc/PE) to afford desired product as colorless oil (yield 78%).1H-NMR (300 MHz, CDCl3) δ 7.90 (d, J = 2.3 Hz), 6.35 (d, J = 2.3 Hz), 6.28 (br s, 1H), 3.93-3.85 (m, 2H), 3.62 (t, J = 5.2 Hz, 2H), 3.41 (s, 3H), 2.50 (s, 3H), 2.31 (s, 3H); MS: 221.1 [M+H]+. [085] 5-iso-Propyl-N-(2-methoxyethyl)pyrazolo[1,5-a]pyrimidin-7-amine (int7c) was prepared in 90% yield from 7-chloro-5-isopropylpyrazolo[1,5-a]pyrimidine using the method described previously for the synthesis of N-(2-methoxyethyl)-5-methylpyrazolo[1,5- a]pyrimidin-7-amine.1H-NMR (300 MHz, CDCl3) δ 7.95 (d, J = 2.3 Hz, 1H), 6.50 (br s, 1H), 6.41 (d, J = 2.3 Hz, 1H), 5.83 (s, 1H), 3.71-3.64 (m, 2H), 3.59-3.52 (m, 2H), 3.41 (s, 3H), 2.98 (septet, J = 6.9 Hz, 1H), 1.31 (d, J = 6.9 Hz, 6H); MS: 235.1 [M+H]+. [086] Synthesis of tert-butyl (3-bromo-5-methylpyrazolo[1,5-a]pyrimidin-7-yl)(2- methoxyethyl)carbamate (int9a) To a stirred solution of N-(2-methoxyethyl)-5-methylpyrazolo[1,5-a]pyrimidin-7-amine (1.6 g, 7.8 mmol) in DCM (50 ml) was added di-tert-butyl dicarbonate (4.2 g, 2.5 eq) followed by 4-dimethylaminopyridine (94 mg, 0.1 eq).The reaction mixture was stirred at RT for 18h, then washed with water (2^50 ml), dried over Na2SO4, concentrated in vacuo to obtain crude tert- butyl (2-methoxyethyl)(5-methylpyrazolo[1,5-a]pyrimidin-7-yl)carbamate as colorless oil. N-bromosuccinimide (1.2 g) was added to the crude tert-butyl (2-methoxyethyl)(5- methylpyrazolo[1,5-a]pyrimidin-7-yl)carbamate in MeCN (80 ml) at RT. After stirring the reaction mixture for 18h the solvent was evaporated in vacuo. The resulting semisolid was dissolved in DCM (50 ml). The obtained solution was washed with water (30 ml), dried over Na2SO4 and concentrated in vacuo. The residue was subjected to column chromatography (10%-50% EtOAc/PE) to give brominated product as yellow solid (1.2 g, yield 40%).1H-NMR
(300 MHz, CDCl3) δ 8.04 (s, 1H), 6.77 (s, 1H), 3.92 (t, J = 5.2 Hz, 2H), 3.56 (t, J = 5.2 Hz, 2H), 3.20 (s, 3H), 2.66 (s, 3H), 1.34 (s, 9H); MS: 385.2 [M (79Br)+H]+, 387.2 [M (81Br)+H]+. [087] Synthesis of tert-butyl (5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-yl)(2- methoxyethyl)carbamate (int8b) A mixture of N-(2-methoxyethyl)-5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-amine (5.0 g, 22.7 mmol), di-tert-butyl dicarbonate (9.9 g, 2 eq), TEA (9.6 ml, 3eq) and 4-dimethylaminopyridine (280 mg, 0.1 eq) in DCM was stirred for 18h at RT. Additional amount (2 eq) of di-tert-butyl dicarbonate was added portionwise to reaction mixture and stirring was continued until all starting material was converted to the desired Boc-product (LC-MS control). After the usual workup (see previously) the isolated crude product was purified by column chromatography (10%-50% EtOAc/PE) to give the desired Boc-product as colourless oil 5.8 g (yield 78%).1H- NMR (400 MHz, CDCl3, mixture of two rotamers) δ 8.00-7.97 (m, 1H), 6.56 (d, J = 2.3 Hz, 1H), 4.18-4.12 (m, 0.3H), 4.12-4.03 (m, 0.7H), 3.84-3.73 (m, 1H), 3.65-3.58 (m, 0.7H), 3.58- 3.51 (m, 0.3H), 3.46-3.36 (m, 1H), 3.22 (s, 0.9H), 3.16 (s, 2.1 H), 2.59 (s, 2.1 H), 2.58 (s, 0.9H), 2.25 (s, 2.1 H), 2.23 (s, 0.9H), 1.56 (s, 2.7H), 1.28 (s, 6.3 H); MS: 321.2 [M+H]+. [088] tert-Butyl (3-bromo-5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-yl)(2- methoxyethyl)carbamate (int9b) as white solid (yield 90%) was prepared by reacting tert- butyl (5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-yl)(2-methoxyethyl)carbamate with N- bromosuccinimide (1.2 eq) using the method described previously for the synthesis of int9a. 1H-NMR (400 MHz, CDCl3) δ 7.98 (s, 0.7H), 7.97 (s, 0.3H), 4.10-4.06 (m, 0.3H), 4.06-3.97 (m, 0.7H), 3.82-3.74 (m, 0.7H), 3.74-3.70 (m, 0.3H), 3.64-3.57 (m, 0.7H), 3.57-3.51 (m, 0.3H), 3.45-3.38 (m, 0.7H), 3.38-3.33 (m, 0.3H), 3.20 (s, 0.9H), 3.13 (s, 2.1H), 2.65 (s, 2.1H), 2.64 (s, 0.9H), 2.26 (s, 2.1H), 2.24 (s, 0.9H), 1.55 (s, 2.7H), 1.28 (s, 6.3 H); MS: 399.2 [M (79Br)+H]+, 401.2 [M (81Br)+H]+. [089] tert-Butyl (5-isopropylpyrazolo[1,5-a]pyrimidin-7-yl)(2-methoxyethyl)carbamate (int8c) as colourless oil (yield 90%) was prepared from 5-iso-propyl-N-(2- methoxyethyl)pyrazolo[1,5-a]pyrimidin-7-amine using the method described previously for the synthesis of tert-butyl (5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-yl)(2- methoxyethyl)carbamate. The crude product was purified by column chromatography (0%- 20% acetone/DCM) 1H-NMR (400 MHz, CDCl3) δ 8.06 (d, J = 2.3 Hz, 1H), 6.76 (s, 1H), 6.63 (d, J = 2.3 Hz, 1H), 3.98 (t, J = 5.4 Hz, 2H), 3.57 (t, J = 5.4 Hz, 2H), 3.24 (s, 3H), 3.10 (septet, J = 6.9 Hz, 1H), 1.38-1.33 (m, 15H); MS: 335.2 [M+H]+. [090] tert-Butyl (3-bromo-5-isopropylpyrazolo[1,5-a]pyrimidin-7-yl)(2- methoxyethyl)carbamate (int9c) as brown oil (yield 68%) was prepared in reaction of tert- butyl (5-isopropylpyrazolo[1,5-a]pyrimidin-7-yl)(2-methoxyethyl)carbamate with N-
bromosuccinimide (1.2 eq) using the method described previously for the synthesis int9a.1H- NMR (400 MHz, CDCl3) δ 8.04 (s, 1H), 6.82 (s, 1H), 3.94 (t, J = 5.2 Hz, 2H), 3.55 (t, J = 5.2 Hz, 2H), 3.22 (s, 3H), 3.17 (septet, J = 6.9 Hz, 1H), 1.38-1.33 (m, 15H); MS: 413.1 [M (79Br)+H]+, 415.1 [M (81Br)+H]+. [091] N-(2-Methoxyethyl)-5-methyl-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5- a]pyrimidin-7-amine (12): 12 was prepared from int9 and (3-(trifluoromethyl)phenylboronic acid by following the procedure described in general method A. The purification was carried out using 0-10% acetone/DCM as eluent to afford 12 as brown solid (yield 28%, HPLC purity 99% at 254 nm, 99% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.33 (br s, 1H), 8.31 (d, J = 7.7 Hz , 1H), 8.30 (s, 1H), 7.51 (t, J = 7.7 Hz, 1H), 7.43 (d, J = 7.7 Hz, 1H), 6.53 (t, J = 5.4 Hz, 1H), 5.90 (s, 1H), 3.69 (t, J = 5.4 Hz, 2H), 3.56 (q, J = 5.4 Hz, 2H), 3.43 (s, 3H), 2.58 (s, 3H). 13C-NMR (101 MHz, CDCl3) δ 160.6, 146.5, 145.7, 141.4, 134.05, 131.0 (q, J = 31.9 Hz), 129.1, 129.0, 124.7 (q, J = 271.6 Hz), 122.4 (q, J = 3.8 Hz), 122.0 (q, J = 3.8 Hz), 107.2, 86.2, 70.3, 59.2, 41.9, 25.6. MS: 351.2 [M+H]+; 349.1 [M-H]-. [092] N-(2-Methoxyethyl)-3-(4-methoxyphenyl)-5-methylpyrazolo[1,5-a]pyrimidin-7- amine (17): 17 was prepared from int9 and 4-methoxyphenylboronic acid by following the procedure described in general method A. The purification was carried out using 0-10% acetone/DCM as eluent to afford 17 as white solid (yield 48%, HPLC purity 99% at 254 nm, 98% at 210 nm). 1H-NMR (400 MHz, CDCl3) δ 8.21 (s, 1H), 8.01-7.96 (m, 2H), 7.01-6.95 (m, 2H), 6.47 (t, J = 5.4 Hz, 1H), 5.85 (s, 1H), 3.84 (s, 3H), 3.69 (t, J = 5.5 Hz, 2H), 3.55 (q, J = 5.5 Hz, 2H), 3.43 (s, 3H), 2.56 (s, 3H).13C-NMR (101 MHz, CDCl3) δ 159.7, 157.8, 146.4, 145.1, 141.1, 127.3, 125.9, 114.3, 108.6, 85.6, 70.4, 59.32, 55.5, 41.9, 25.6. MS: 313.2 [M+H]+. [093] N-(2-Methoxyethyl)-5,6-dimethyl-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5- a]pyrimidin-7-amine (13): 13 was prepared from int9 and (3-(trifluoromethyl)phenylboronic acid by following the procedure described in general method A. The purification was carried out using 0-50% acetone/DCM as eluent to afford 13 as white solid (yield 16% after recrystallization from TBME, HPLC purity 99% at 254 nm, 99% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.35 (s, 1H), 8.29 (d, J = 7.8 Hz, 1H), 8.27 (s, 1H), 7.51 (t, J = 7.8 Hz, 1H), 7.42 (d, J = 7.8 Hz, 1H), 6.33 (t, J = 5.4 Hz, 1H), 3.93 (q, J = 5.4 Hz, 2H), 3.65 (t, J = 5.4 Hz, 2H), 3.43 (s, 3H), 2.59 (s, 3H), 2.36 (s, 3H).13C-NMR (101 MHz, DMSO) δ 160.7, 145.9, 144.2, 140.5, 134.2, 131.0 (q,
J = 31.7 Hz), 129.1, 128.8, 124.6 (q, J = 272.0 Hz), 122.3 (q, J = 3.9 Hz), 121.8 (q, J = 3.9 Hz), 106.8, 97.3, 71.7, 59.2, 45.5, 24.8, 13.6. MS: 365.2 [M+H]+; 363.1 [M-H]- [094] N-(2-Methoxyethyl)-3-(4-methoxyphenyl)-5,6-dimethylpyrazolo[1,5-a]pyrimidin- 7-amine (14): 14 was prepared from int9 and 4-methoxyphenylboronic acid by following the procedure described in general method A. The purification was carried out using 0-50% acetone/DCM as eluent to afford 14 as white solid (yield 61%, HPLC purity 99% at 254 nm, 99% at 210 nm). 1H-NMR (400 MHz, CDCl3) δ 8.18 (s, 1H), 8.01-7.96 (m, 2H), 7.00-6.95 (m, 2H), 6.28 (t, J = 5.6 Hz, 1H), 3.92-3.87 (m, 2H), 3.84 (s, 3H), 3.64 (t, J = 5.2 Hz, 2H), 3.43 (s, 3H), 2.57 (s, 3H), 2.34 (s, 3H).13C-NMR (101 MHz, CDCl3) δ 159.8, 157.7, 145.8, 143.7, 140.1, 127.1, 126.0, 114.3, 108.2, 96.8, 71.8, 59.2, 55.5, 45.5, 24.7, 13.6. MS: 327.2 [M+H]+. [095] 5-Isopropyl-N-(2-methoxyethyl)-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5- a]pyrimidin-7-amine (11): 11 was prepared from int9 and (3-(trifluoromethyl)phenylboronic acid by following the procedure described in general method A. The purification was carried out using 0-10% acetone/DCM as eluent to afford 11 as yellow solid (yield 82%, HPLC purity 98% at 254 nm, 99% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.55 (s, 1H), 8.33 (s, 1H), 8.30 (d, J = 7.8 Hz, 1H), 7.51 (t, J = 7.8 Hz, 1H), 7.42 (d, J = 7.8 Hz, 1H), 6.52 (t, J = 5.5 Hz, 1H), 5.93 (s, 1H), 3.72-3.68 (m, 2H), 3.61-3.55 (m, 2H), 3.44 (s, 3H), 3.05 (septet, J = 6.9 Hz, 1H), 1.38 (d, J = 6.9 Hz, 6H). 13C-NMR (101 MHz, CDCl3) δ 169.2, 146.9, 145.8, 141.2, 134.2, 130.9 (q, J = 31.7 Hz), 129.0, 128.5, 124.6 (q, J = 273.1 Hz), 122.4 (q, J = 4.0 Hz), 121.7 (q, J = 3.8 Hz), 107.1, 84.1, 70.3, 59.2, 42.0, 37.0, 22.3. MS: 379.2 [M+H]+; 377.2 [M+H]-. [096] 5-Isopropyl-N-(2-methoxyethyl)-3-(4-methoxyphenyl)pyrazolo[1,5-a]pyrimidin-7- amine (16): 16 was prepared from int9 and 4-methoxyphenylboronic acid by following the procedure described in general method A. The purification was carried out using 0-10% acetone/DCM as eluent to afford 16 as brown solid (yield 85%, HPLC purity 99% at 254 nm, 98% at 210 nm). 1H-NMR (400 MHz, CDCl3) δ 8.24 (s, 1H), 8.10-8.03 (m, 2H), 7.02-6.94 (m, 2H), 6.45 (t, J = 5.5 Hz, 1H), 5.87 (s, 1H), 3.85 (s, 3H), 3.71-3.67 (m, 2H), 3.60-3.54 (m, 2H), 3.43 (s, 3H), 3.04 (septet, J = 6.9 Hz, 1H), 1.36 (d, J = 6.9 Hz, 6H).13C-NMR (101 MHz, CDCl3) δ 168.3, 157.7, 146.8, 145.1, 140.9, 127.1, 126.1, 114.2, 108.4, 83.3, 70.4, 59.2, 55.5, 41.9, 37.1, 22.4. MS: 341.2 [M+H]+.
[097] Synthesis of N-(3-(1H-imidazol-1-yl)propyl)-3-arylpyrazolo[1,5-a]pyrimidin-7- amines Scheme 5
[098] General method B for the synthesis of 3-arylpyrazolo[1,5-a]pyrimidin-7-ols (int10) A mixture of 4-aryl-1H-pyrazol-5-amine (8.0 mmol) and corresponding β-keto ester (1.2 eq) in AcOH (10 ml) was stirred at 100^C. The obtained mixture was then cooled to RT and diluted with TBME (30 ml). The resulting white solid was filtered off and dried in air. [099] 3-(4-Methoxyphenyl)-5-methylpyrazolo[1,5-a]pyrimidin-7-ol (int10a) was prepared in 78% yield (1.6 g) from 4-(4-methoxyphenyl)-1H-pyrazol-5-amine and ethyl acetoacetate by heating for 4h. 1H-NMR (400 MHz, DMSO-d6) δ 11.81 (s, 1H), 8.02 (s, 1H), 7.47 (d, J= 8.8 Hz, 2H), 7.03 (d, J= 8.8 Hz, 2H), 5.63 (s, 1H), 3.80 (s, 3H), 2.33 (s, 3H). MS: 256.1 [M+H]+. [100] 5-Methyl-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidin-7-ol (int10b) was prepared in 43% yield (1.0 g) from 4-(3-(trifluoromethyl)phenyl)-1H-pyrazol-5-amine and ethyl acetoacetate by heating for 18h.1H-NMR (400 MHz, DMSO-d6) δ 12.02 (s, 1H), 8.21 (s, 1H), 7.90-7.82 (m, 2H), 7.74-7.63 (m, 2H), 5.70 (s, 1H), 2.35 (s, 3H). MS: 294.0 [M+H]+. [101] 3-(4-Methoxyphenyl)-5,6-dimethylpyrazolo[1,5-a]pyrimidin-7-ol (int10c) was prepared in 60% yield (1.2 g) from 4-(4-methoxyphenyl)-1H-pyrazol-5-amine and ethyl 2- methyl-3-oxobutanoate by heating for 2h.1H-NMR (400 MHz, DMSO-d6) δ 11.59 (s, 1H), 7.99 (s, 1H), 7.46 (d, J = 8.8 Hz, 2H), 7.02 (d, J = 8.8 Hz, 2H), 3.79 (s, 3H), 2.36 (s, 3H), 1.99 (s, 3H). MS: 270.0 [M+H]+. [102] 5,6-Dimethyl-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidin-7-ol (int10d) was prepared in 46% yield (1.1 g) from 4-(3-(trifluoromethyl)phenyl)-1H-pyrazol-5-amine and ethyl 2-methyl-3-oxobutanoate by heating for 2h.1H-NMR (300 MHz, DMSO-d6) δ 11.80 (s, 1H), 8.19 (s, 1H), 7.88-7.83 (m, 2H), 7.72-7.61 (m, 2H), 2.38 (s, 3H), 2.00 (s, 3H). MS: 308.1 [M+H]+. [103] 5-Isopropyl-3-(4-methoxyphenyl)pyrazolo[1,5-a]pyrimidin-7-ol (int10e) was prepared in 50% yield (1.1 g) from 4-(4-methoxyphenyl)-1H-pyrazol-5-amine and ethyl 4-
methyl-3-oxopentanoate by heating for 2h. 1H-NMR (300 MHz, DMSO-d6) δ 11.70 (s, 1H), 8.01 (s, 1H), 7.46 (d, J = 8.9 Hz, 2H), 7.04 (d, J = 8.9 Hz, 2H), 5.65 (s, 1H), 3.80 (s, 3H), 3.03 (septet, J = 6.9 Hz, 1H), 1.25 (d, J= 6.9 Hz, 6H). MS: 284.1 [M+H]+. [104] 5-Isopropyl-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidin-7-ol (int10f) was prepared in 60% yield (1.5 g) from 4-(3-(trifluoromethyl)phenyl)-1H-pyrazol-5-amine and ethyl 4-methyl-3-oxopentanoate by heating for 3h.1H-NMR (300 MHz, DMSO-d6) δ 11.88 (s, 1H), 8.20 (s, 1H), 7.90-7.78 (m, 2H), 7.75-7.63 (m, 2H), 5.72 (s, 1H), 3.00 (septet, J = 6.9 Hz, 1H), 1.26 (d, J = 6.9 Hz, 6H). [105] General method C for the synthesis of 3-aryl-7-chloropyrazolo[1,5-a]pyrimidines (int11) N,N-dimethylaniline (0.1 ml, 0.3 eq) was added to a suspension of int10 (3.0 mmol) in POCl3 (5 ml). The reaction mixture was stirred at 100^C. The course of the reaction was controlled by LC-MS. The obtained solution was cooled to RT and evaporated in vacuo. Obtained oily residue was dissolved in DCM (40 ml), washed with aq NaHCO3 (2^15 ml). Organic layer was dried over Na2SO4 and filtered through a SiO2 pad. The filtrate was concentrated to give the desired chloro compound int11 as yellow solid. [106] 7-Chloro-3-(4-methoxyphenyl)-5-methylpyrazolo[1,5-a]pyrimidine (int11a) was prepared in 78% yield (0.64 g) from 3-(4-methoxyphenyl)-5-methylpyrazolo[1,5-a]pyrimidin- 7-ol.1H-NMR (300 MHz, CDCl3) δ 8.41 (s, 1H), 7.97 (d, J = 8.9 Hz, 2H), 7.00 (d, J = 8.9 Hz, 2H), 6.85 (s, 1H), 3.86 (s, 3H), 2.65 (s, 3H). MS: 274.1 [M (35Cl)+H]+, 276.1 [M (37Cl)+H]+. [107] 7-Chloro-5-methyl-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidine (int11b) was prepared in 90% yield (0.84 g) from 5-methyl-3-(3- (trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidin-7-ol.1H-NMR (300 MHz, CDCl3) δ 8.51 (s, 1H), 8.34 (br s, 1H), 8.26 (d, J= 7.20 Hz, 1H), 7.60-7.48 (m, 2H), 6.93 (s, 1H), 2.69 (s, 3H). MS: 312.0 [M (35Cl)+H]+, 314.0 [M (37Cl)+H]+. [108] 7-Chloro-3-(4-methoxyphenyl)-5,6-dimethylpyrazolo[1,5-a]pyrimidine (int11c) was prepared in 77% yield (0.66 g) from 3-(4-methoxyphenyl)-5,6-dimethylpyrazolo[1,5- a]pyrimidin-7-ol. 1H-NMR (300 MHz, CDCl3) δ 8.34 (s, 1H), 7.97 (d, J = 8.9 Hz, 2H), 6.99 (d, J = 8.9 Hz, 2H), 3.85 (s, 3H), 2.65 (s, 3H), 2.44 (s, 3H). MS: 288.0 [M (35Cl)+H]+, 290.0 [M (37Cl)+H]+. [109] 7-Chloro-5,6-dimethyl-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidine (int11d) was prepared in 79% yield (0.77 g) from 5,6-dimethyl-3-(3- (trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidin-7-ol.1H-NMR (300 MHz, CDCl3) δ 8.44 (s,
1H), 8.36 (br s, 1H), 8.25 (d, J = 7.4 Hz, 1H), 7.59-7.46 (m, 2H), 2.63 (s, 3H), 2.47 (s, 3H). MS: 326.0 [M (35Cl)+H]+, 328.0 [M (37Cl)+H]+. [110] 7-Chloro-5-isopropyl-3-(4-methoxyphenyl)pyrazolo[1,5-a]pyrimidine (int11e) was prepared in 83% yield (0.75 g) from 5-iso-propyl-3-(4-methoxyphenyl)pyrazolo[1,5- a]pyrimidin-7-ol. 1H-NMR (300 MHz, CDCl3) δ 8.43 (s, 1H), 8.03 (d, J = 8.9 Hz, 2H), 7.01 (d, J = 8.9 Hz, 2H), 6.89 (s, 1H), 3.86 (s, 3H), 3.14 (septet, J = 6.9 Hz, 1H), 1.39 (d, J= 6.9 Hz, 6H). MS: 302.1 [M (35Cl)+H]+, 304.1 [M (37Cl)+H]+. [111] 7-Chloro-5-isopropyl-3-(3-(trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidine (int11f) was prepared in 44% yield (0.45 g) from 5-iso-propyl-3-(3- (trifluoromethyl)phenyl)pyrazolo[1,5-a]pyrimidin-7-ol.1H-NMR (300 MHz, CDCl3) δ 8.52 (s, 1H), 8.50 (br s, 1H), 8.23 (d, J = 7.3 Hz, 1H), 7.59-7.46 (m, 2H), 6.95 (s, 1H), 3.15 (septet, J = 6.9 Hz, 1H), 1.40 (d, J= 6.9 Hz, 6H). MS: 340.1 [M (35Cl)+H]+, 342.1 [M (37Cl)+H]+. [112] General method D for the synthesis of N-(3-(1H-imidazol-1- yl)propyl)pyrazolo[1,5-a]pyrimidin-7-amines (int12) A mixture of int11 (2.0 mmol), 3-(1H-imidazol-1-yl)propan-1-amine (0.36 ml, 1.5 eq) and K2CO3 (1.38 g, 5 eq) in MeCN (1 ml) was refluxed for 6h. The solvent was removed to give a residue that was dissolved in DCM (30 ml) and water (30 ml). Organic layer was collected, dried over Na2SO4 and the solvent was removed to give crude product, which was purified by crystallization or by column chromatography. [113] N-(3-(1H-imidazol-1-yl)propyl)-3-(4-methoxyphenyl)-5-methylpyrazolo[1,5- a]pyrimidin-7-amine (18) was prepared from 7-chloro-3-(4-methoxyphenyl)-5- methylpyrazolo[1,5-a]pyrimidine. After crystallization from MeOH 18 was isolated as white solid (0.3g, yield 46%, HPLC purity 99% at 254 nm, 99% at 210 nm). 1H-NMR (400 MHz, CDCl3) δ 8.21 (s, 1H), 8.00-7.95 (m, 2H), 7.51 (t, J = 1.1 Hz, 1H), 7.12 (t, J = 1.1 Hz, 1H), 7.00-6.96 (m, 2H), 6.95 (t, J = 1.1 Hz, 1H), 6.22 (t, J = 5.9 Hz, 1H), 5.72 (s, 1H), 4.13 (t, J = 6.7 Hz, 2H), 3.84 (s, 3H), 3.40-3.33 (m, 2H), 2.55 (s, 3H), 2.24 (pentet, J = 6.7 Hz, 2H).13C- NMR (101 MHz, CDCl3) δ 159.8, 157.9, 146.1, 145.0, 141.2, 137.3, 130.3, 127.3, 125.6, 118.8, 114.3, 108.9, 85.6, 55.5, 44.1, 38.8, 30.4, 25.6. MS: 363.2 [M+H]+ ; 361.1 [M+H]-. [114] N-(3-(1H-imidazol-1-yl)propyl)-3-(4-methoxyphenyl)-5,6-dimethylpyrazolo[1,5- a]pyrimidin-7-amine (15) was prepared from 7-chloro-3-(4-methoxyphenyl)-5,6- dimethylpyrazolo[1,5-a]pyrimidine. The purification was carried out using 20%-100% acetone/DCM as eluent to afford 15 as white solid (0.30 g, yield 39%, HPLC purity 99% at 254 nm, 99% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.16 (s, 1H), 7.99-7.94 (m, 2H), 7.46 (t, J = 1.1 Hz, 1H), 7.07 (t, J = 1.1 Hz, 1H), 6.99-6.94 (m, 2H), 6.88 (t, J = 1.1 Hz, 1H), 5.99 (t, J =
6.7 Hz, 1H), 4.07 (t, J = 6.7 Hz, 2H), 3.82 (s, 3H), 3.58 (q, J = 6.7 Hz, 2H), 2.54 (s, 3H), 2.20 (s, 3H), 2.14 (pentet, J = 6.7 Hz, 2H). 13C-NMR (101 MHz, CDCl3) δ 160.1, 157.8, 145.2, 143.4, 140.0, 137.2, 130.1, 127.1, 125.7, 118.8, 114.3, 108.6, 97.4, 55.4, 43.8, 42.3, 32.2, 24.6, 13.4. MS: 377.2 [M+H]+; 375.1 [M+H]-. [115] N-(3-(1H-imidazol-1-yl)propyl)-5-isopropyl-3-(4-methoxyphenyl)pyrazolo[1,5- a]pyrimidin-7-amine (10) was prepared from 7-chloro-5-isopropyl-3-(4- methoxyphenyl)pyrazolo[1,5-a]pyrimidine. After crystallization from MeOH 10 was isolated as white solid (0.3 g, yield 38%, HPLC purity 99% at 254 nm, 98% at 210 nm).1H-NMR (400 MHz, CDCl3) δ 8.24 (s, 1H), 8.09-8.03 (m, 2H), 7.51 (t, J = 1.1 Hz, 1H), 7.13 (t, J = 1.1 Hz, 1H), 7.00-6.97 (m, 2H), 6.94 (t, J = 1.1 Hz, 1H), 6.20 (t, J = 6.0 Hz, 1H), 5.74 (s, 1H), 4.13 (t, J = 6.7 Hz , 2H), 3.84 (s, 3H), 3.40-3.34 (m, 2H), 3.02 (septet, J = 6.9 Hz, 1H), 2.24 (pentet, J = 6.7 Hz, 2H), 1.35 (d, J = 6.9 Hz, 6H). 13C-NMR (101 MHz, CDCl3) δ 168.4, 157.8, 146.4, 145.0, 140.9, 137.3, 130.3, 127.1, 125.8, 118.8, 114.2, 108.8, 83.3, 55.5, 44.0, 38.8, 37.1, 30.5, 22.4. MS: 391.2 [M+H]+; 389.2 [M+H]-. [116] Luciferase Reporter Gene Assay. HuH-7 cells, human hepatoma cells, were plated on 24-well dishes at a density of 8.0×104 cells with 1 mL media per well. The following day, each well was transfected with 1 μL Lipofectamine 2000 and 1 µg DNA consisting of 350 ng pGudLuc 4.1, 100 ng pRenilla, and 50 ng pEGFP, pGudLuc 4.1 is an AHR-driven reporter construct containing a luciferase gene downstream of the Cyp1a1 promoter. After 6 hours, the cells were treated with either DMSO, 10 nM TCDD alone or in combination with test compounds (0.001 to 10 mM). The next morning, the cells were lysed, and the luciferase activity was measured using Dual luciferase assay and normalized to Renilla activity.
References (1) Stockinger, B.; Meglio, P. Di; Gialitakis, M.; Duarte, J. H. The Aryl Hydrocarbon Receptor: Multitasking in the Immune System. Annu Rev Immunol 2014, 32 (1), 403– 432. https://doi.org/10.1146/annurev-immunol-032713-120245. (2) Wen, Z.; Zhang, Y.; Zhang, B.; Hang, Y.; Xu, L.; Chen, Y.; Xie, Q.; Zhao, Q.; Zhang, L.; Li, G.; Zhao, B.; Sun, F.; Zhai, Y.; Zhu, Y. Cryo-EM Structure of the Cytosolic AhR Complex. Structure 2023, 31 (3), 295-308.e4. https://doi.org/10.1016/j.str.2022.12.013. (3) Dai, S.; Qu, L.; Li, J.; Zhang, Y.; Jiang, L.; Wei, H.; Guo, M.; Chen, X.; Chen, Y. Structural Insight into the Ligand Binding Mechanism of Aryl Hydrocarbon Receptor. Nat Commun 2022, 13 (1), 6234. https://doi.org/10.1038/s41467-022-33858-w. (4) Seok, S. H.; Lee, W.; Jiang, L.; Molugu, K.; Zheng, A.; Li, Y.; Park, S.; Bradfield, C. A.; Xing, Y. Structural Hierarchy Controlling Dimerization and Target DNA Recognition in the AHR Transcriptional Complex. Proc Natl Acad Sci U S A 2017, 114 (21), 5431–5436. https://doi.org/10.1073/pnas.1617035114. (5) Schulte, K. W.; Green, E.; Wilz, A.; Platten, M.; Daumke, O. Structural Basis for Aryl Hydrocarbon Receptor-Mediated Gene Activation. Structure 2017, 25 (7), 1025- 1033.e3. https://doi.org/10.1016/j.str.2017.05.008. (6) Gruszczyk, J.; Grandvuillemin, L.; Lai-Kee-Him, J.; Paloni, M.; Savva, C. G.; Germain, P.; Grimaldi, M.; Boulahtouf, A.; Kwong, H.-S.; Bous, J.; Ancelin, A.; Bechara, C.; Barducci, A.; Balaguer, P.; Bourguet, W. Cryo-EM Structure of the Agonist-Bound Hsp90-XAP2-AHR Cytosolic Complex. Nat Commun 2022, 13 (1), 7010. https://doi.org/10.1038/s41467-022-34773-w. (7) Murray, I. A.; Patterson, A. D.; Perdew, G. H. Aryl Hydrocarbon Receptor Ligands in Cancer: Friend and Foe. Nat Rev Cancer 2014, 14 (12), 801–814. https://doi.org/10.1038/nrc3846. (8) Rothhammer, V.; Quintana, F. J. The Aryl Hydrocarbon Receptor: An Environmental Sensor Integrating Immune Responses in Health and Disease. Nat Rev Immunol 2019, 19 (3), 184–197. https://doi.org/10.1038/s41577-019-0125-8. (9) Tompkins, L. M.; Li, H.; Li, L.; Lynch, C.; Xie, Y.; Nakanishi, T.; Ross, D. D.; Wang, H. A Novel Xenobiotic Responsive Element Regulated by Aryl Hydrocarbon Receptor Is Involved in the Induction of BCRP/ABCG2 in LS174T Cells. Biochem Pharmacol 2010, 80 (11), 1754–1761. https://doi.org/10.1016/j.bcp.2010.08.016. (10) Wilkinson, I. V. L.; Perkins, K. J.; Dugdale, H.; Moir, L.; Vuorinen, A.; Chatzopoulou, M.; Squire, S. E.; Monecke, S.; Lomow, A.; Geese, M.; Charles, P. D.; Burch, P.; Tinsley, J. M.; Wynne, G. M.; Davies, S. G.; Wilson, F. X.; Rastinejad, F.; Mohammed, S.; Davies, K. E.; Russell, A. J. Chemical Proteomics and Phenotypic Profiling Identifies the Aryl Hydrocarbon Receptor as a Molecular Target of the Utrophin Modulator Ezutromid. Angewandte Chemie - International Edition 2020, 59 (6), 2420–2428. https://doi.org/10.1002/anie.201912392. (11) Campesato, L. F.; Budhu, S.; Tchaicha, J.; Weng, C. H.; Gigoux, M.; Cohen, I. J.; Redmond, D.; Mangarin, L.; Pourpe, S.; Liu, C.; Zappasodi, R.; Zamarin, D.; Cavanaugh, J.; Castro, A. C.; Manfredi, M. G.; McGovern, K.; Merghoub, T.; Wolchok, J. D. Blockade of the AHR Restricts a Treg-Macrophage Suppressive Axis Induced by L-Kynurenine. Nat Commun 2020, 11 (1), 1–11. https://doi.org/10.1038/s41467-020-17750-z. (12) Gutiérrez-Vázquez, C.; Quintana, F. J. Regulation of the Immune Response by the Aryl Hydrocarbon Receptor. Immunity 2018, 48 (1), 19–33. https://doi.org/10.1016/j.immuni.2017.12.012.
(13) Cheong, J. E.; Sun, L. Targeting the IDO1/TDO2–KYN–AhR Pathway for Cancer Immunotherapy – Challenges and Opportunities. Trends Pharmacol Sci 2018, 39 (3), 307–325. https://doi.org/10.1016/j.tips.2017.11.007. (14) Lin, L.; Dai, Y.; Xia, Y. An Overview of Aryl Hydrocarbon Receptor Ligands in the Last Two Decades (2002–2022): A Medicinal Chemistry Perspective. Eur J Med Chem 2022, 244 (September), 114845. https://doi.org/10.1016/j.ejmech.2022.114845. (15) Boitano, A. E.; Wang, J.; Romeo, R.; Bouchez, L. C.; Parker, A. E.; Sutton, S. E.; Walker, J. R.; Flaveny, C. A.; Perdew, G. H.; Denison, M. S.; Schultz, P. G.; Cooke, M. P. Aryl Hydrocarbon Receptor Antagonists Promote the Expansion of Human Hematopoietic Stem Cells. Science (1979) 2010, 329 (5997), 1345–1348. https://doi.org/10.1126/science.1191536. (16) Schmees, N.; Gutcher, I.; Irlbacher, H.; Bader, B.; Zhao, N.; Platten, M.3-Oxo-2,6- Diphenyl-2,3-Dihydropyridazine-4-Carboxamides. WO2017202816A1, 2017. (17) Castro, A. C.; Evans, C. A. Indole AHR Inhibitors and Uses Thereof. WO2018195397A2, 2018. (18) Mcgovern, K.; Castro, A.; Cavanaugh, J.; Sanchez-Martin, M.; Tchaicha, J.; Syed, S.; Frosch, H.; Natarajan, P.; O’Callaghan, K.; Amidon, B.; Michelle Zhang, X.; Ecsedy, J.448 Discovery of Clinical Candidate IK-175, a Selective Orally Active AHR Antagonist. In Regular and young investigator award abstracts; BMJ Publishing Group Ltd, 2020; p A272.1-A272. https://doi.org/10.1136/jitc-2020-SITC2020.0448. (19) Castro, A. C. Indole AHR Inhibitors and Uses Thereof. WO2020081636, 2020. (20) Smith, K. J.; Murray, I. A.; Tanos, R.; Tellew, J.; Boitano, A. E.; Bisson, W. H.; Kolluri, S. K.; Cooke, M. P.; Perdew, G. H. Identification of a High-Affinity Ligand That Exhibits Complete Aryl Hydrocarbon Receptor Antagonism. Journal of Pharmacology and Experimental Therapeutics 2011, 338 (1), 318–327. https://doi.org/10.1124/jpet.110.178392. (21) Sun, L. Recent Advances in the Development of AHR Antagonists in Immuno- Oncology. RSC Med Chem 2021, 12 (6), 902–914. https://doi.org/10.1039/d1md00015b. (22) Scheuermann, T. H.; Stroud, D.; Sleet, C. E.; Bayeh, L.; Shokri, C.; Wang, H.; Caldwell, C. G.; Longgood, J.; MacMillan, J. B.; Bruick, R. K.; Gardner, K. H.; Tambar, U. K. Isoform-Selective and Stereoselective Inhibition of Hypoxia Inducible Factor-2. J Med Chem 2015, 58 (15), 5930–5941. https://doi.org/10.1021/acs.jmedchem.5b00529.
Claims
Claims 1. A compound with Formula I
wherein: R is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L -OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N - C ( 1,
n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; R1, R2, R3 R4 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L
-OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N HR11, -NR11 2, -N(R11)R12, -L-NH2, -L-NHR11, -L-NR11 2, -L-N(R11)R12, -NH-L-NH2, -NH-L- - C ( 1,
each L, if present, is independently saturated aliphatic C1‑5alkylene; R1 and R2 may form a cycle; R3 and R4 may form a cycle; R and R2 may form a cycle; R and R3 may form a cycle; R5 is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, L - 11 - 1) - R
2, - - n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; R6 is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L
-NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L -OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N HR11, -NR11 2, -N(R11)R12, -L-NH2, -L-NHR11, -L-NR11 2, -L-N(R11)R12, -NH-L-NH2, -NH-L- - C ( 1,
each L, if present, is independently saturated aliphatic C1‑5alkylene; R5 and R6 may form a cycle; X, Y and Z may be independently substituted CR11R12, NR11, O; R11 and R12 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH,-O-L-OH, -OL-NH2, OCF3, -OCH2CF3, -OCF2CF2H, SCF3, -CN, -NO2, -NO2, -NH2, R11 and R12 may form a cycle; R5 and R11 may form a cycle; R6 and R11 may form a cycle; R5 and R12 may form a cycle; R6 and R12 may form a cycle; W is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; V is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; U is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl. 2. The compound according to Claim 1,
II R is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L -OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N - C ( 1,
n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; R1, R2, R3 R4 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L -OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N HR11, -NR11 2, -N(R11)R12, -L-NH2, -L-NHR11, -L-NR11 2, -L-N(R11)R12, -NH-L-NH2, -NH-L- NHR11, -NH-L-N(R11)R12, -NH-L-N(R11)R12, -NR11-L-NH2, -NR11-L-NHR11, -NR11-L-N(R11) R12, -NR11-L-N(R11)R12, -N(R11)R12, -C(=O)OH, -C(=O)OR11, -C(=O)NH2, -C(=O)NHR11, - C(=O)N(R11)12, -C(=O)N(R11)R12, -NHC(=O)R11, -NR11C(=O)R12, -NHC(=O)OR11, -NR11C(
=O)OR12, -OC(=O)NH2, -OC(=O)NHR11, -OC(=O)N(R11)R12, -OC(=O)R11NR12, -OC(=O)R1 1,
each L, if present, is independently saturated aliphatic C1‑5alkylene; R1 and R2 may form a cycle; R3 and R4 may form a cycle; R and R2 may form a cycle; R and R3 may form a cycle; R5 is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L-NH2, -O-L -NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L-OR11, -O- L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -NHR11, -NR 11 - 1) - R 2,
n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; R6, R7 R8, R9 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L -NH2, -O-L-NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L -OR11, -O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -N HR11, -NR11 2, -N(R11)R12, -L-NH2, -L-NHR11, -L-NR11 2, -L-N(R11)R12, -NH-L-NH2, -NH-L- NHR11, -NH-L-N(R11)R12, -NH-L-N(R11)R12, -NR11-L-NH2, -NR11-L-NHR11, -NR11-L-N(R11) R12, -NR11-L-N(R11)R12, -N(R11)R12, -C(=O)OH, -C(=O)OR11, -C(=O)NH2, -C(=O)NHR11, -
C(=O)N(R11)12, -C(=O)N(R11)R12, -NHC(=O)R11, -NR11C(=O)R12, -NHC(=O)OR11, -NR11C( 1,
each L, if present, is independently saturated aliphatic C1‑5alkylene; R10 is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biarylC10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroarylC2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, R11O(CH2)n, R11S(CH2)n, R11OC(=O)(CH2)n, R11N(R12)C(=O)(CH2)n, R11N(R12)(CH2)n, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH, -O-L-OH, -OR11, -O-L-NH2, -O-L -NHR11, -O-L-N(R11)R12, -L-OR11,-O-L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -L-OR11, -O- L-OR11,-OCF3, -OCH2CF3, -OCF2CF2H, -SR11, -SCF3, -CN, -NO2, -NO2, -NH2, -NHR11, -NR 11 - 1) - R 2,
n is an integer number 1 to 3, each L, if present, is independently saturated aliphatic C1‑5alkylene; R11 and R12 are independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2- 6alkenyl, C2-6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl, F, -Br, -I, -CF3, -CH2CF3, -CF2CF2H, -OH, -L-OH,-O-L-OH, -OL-NH2, OCF3, -OCH2CF3, -OCF2CF2H, SCF3, -CN, -NO2, -NO2, -NH2, - R11 and R12 may form a cycle; R11 and R12 may form a cycle; R5 and R11 may form a cycle; R6 and R11 may form a cycle; R5 and R12 may form a cycle; R6 and R12 may form a cycle;
W is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; V is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; U is independently H, C1-6alkyl, cycloC3-12alkyl, cycloC3-12alkyl-C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, arylC5-8, biaryl C10-12, arylC1-6alkyl, arylC2-6alkenyl, arylC2-6alkynyl, heteroaryl C2-5, heteroarylC1-6alkyl, heteroarylC2-6alkenyl; pairs of substituents selected from R5, R6, R7, R8, R9, R10 may form a cycle. 3. The compound according to Claims 1 or 2, wherein the compound is:
4. The compound according to any one of claims 1 to 3 for use in medicine.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| LVLVP2023000127 | 2023-11-29 | ||
| LVP2023000127 | 2023-11-29 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025114771A1 true WO2025114771A1 (en) | 2025-06-05 |
Family
ID=89661277
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/IB2024/059009 Pending WO2025114771A1 (en) | 2023-11-29 | 2024-09-17 | A new class of aryl hydrocarbon receptor antagonists and their optimisation |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025114771A1 (en) |
Citations (6)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2001023387A2 (en) * | 1999-09-30 | 2001-04-05 | Neurogen Corporation | CERTAIN ALKYLENE DIAMINE-SUBSTITUTED PYRAZOLO[1,5,-a]-1,5-PYRIMIDINES AND PYRAZOLO[1,5-a]-1,3,5-TRIAZINES |
| WO2017202816A1 (en) | 2016-05-25 | 2017-11-30 | Bayer Pharma Aktiengesellschaft | 3-oxo-2,6-diphenyl-2,3-dihydropyridazine-4-carboxamides |
| WO2018195397A2 (en) | 2017-04-21 | 2018-10-25 | Kyn Therapeutics | Indole ahr inhibitors and uses thereof |
| WO2020081636A1 (en) | 2018-10-16 | 2020-04-23 | Ikena Oncology, Inc. | Indole ahr inhibitors and uses thereof |
| WO2021119343A1 (en) * | 2019-12-10 | 2021-06-17 | The Board Of Regents Of The University Of Texas System | Compositions and methods for substituted 7-(piperazin-1-yl)pyrazolo[1,5-a]pyrimidine analogs as inhibitors of kras |
| US11141418B1 (en) * | 2018-06-22 | 2021-10-12 | The Board Of Regents, The University Of Texas System | Methods and compositions for KRAS inhibitors |
-
2024
- 2024-09-17 WO PCT/IB2024/059009 patent/WO2025114771A1/en active Pending
Patent Citations (6)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2001023387A2 (en) * | 1999-09-30 | 2001-04-05 | Neurogen Corporation | CERTAIN ALKYLENE DIAMINE-SUBSTITUTED PYRAZOLO[1,5,-a]-1,5-PYRIMIDINES AND PYRAZOLO[1,5-a]-1,3,5-TRIAZINES |
| WO2017202816A1 (en) | 2016-05-25 | 2017-11-30 | Bayer Pharma Aktiengesellschaft | 3-oxo-2,6-diphenyl-2,3-dihydropyridazine-4-carboxamides |
| WO2018195397A2 (en) | 2017-04-21 | 2018-10-25 | Kyn Therapeutics | Indole ahr inhibitors and uses thereof |
| US11141418B1 (en) * | 2018-06-22 | 2021-10-12 | The Board Of Regents, The University Of Texas System | Methods and compositions for KRAS inhibitors |
| WO2020081636A1 (en) | 2018-10-16 | 2020-04-23 | Ikena Oncology, Inc. | Indole ahr inhibitors and uses thereof |
| WO2021119343A1 (en) * | 2019-12-10 | 2021-06-17 | The Board Of Regents Of The University Of Texas System | Compositions and methods for substituted 7-(piperazin-1-yl)pyrazolo[1,5-a]pyrimidine analogs as inhibitors of kras |
Non-Patent Citations (28)
| Title |
|---|
| BOITANO, A. E.; WANG, J.; ROMEO, R.; BOUCHEZ, L. C.; PARKER, A. E.; SUTTON, S. E.;WALKER, J. R.; FLAVENY, C. A.; PERDEW, G. H.; DE: "Hydrocarbon Receptor Antagonists Promote the Expansion of Human Hematopoietic Stem Cells", SCIENCE, vol. 329, no. 5997, 2010, pages 1345 - 1348, XP055558705, Retrieved from the Internet <URL:https://doi.org/10.1126/science.1191536> DOI: 10.1126/science.1191536 |
| CAMPESATO, L. F.; BUDHU, S.; TCHAICHA, J.; WENG, C. H.; GIGOUX, M.; COHEN, I. J.;REDMOND, D.; MANGARIN, L.; POURPE, S.; LIU, C.; Z: "Blockade of the AHR Restricts a Treg-Macrophage Suppressive Axis Induced by L-Kynurenine", NAT COMMUN, vol. 11, no. 1, 2020, pages 1 - 11, Retrieved from the Internet <URL:https://doi.org/10.1038/s41467-020-17750-z> |
| CHEONG, J. E.SUN, L: "Targeting the IDO1/TDO2-KYN-AhR Pathway for Cancer Immunotherapy - Challenges and Opportunities", TRENDS PHARMACOL SCI, vol. 39, no. 3, 2018, pages 307 - 325, XP055882712, Retrieved from the Internet <URL:https://doi.org/10.1016/j.tips.2017.11.007> DOI: 10.1016/j.tips.2017.11.007 |
| DA C ET AL: "Discovery of Mer kinase inhibitors by virtual screening using Structural Protein-Ligand Interaction Fingerpr", BIOORGANIC & MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 23, no. 5, 13 January 2015 (2015-01-13), pages 1096 - 1101, XP029199007, ISSN: 0968-0896, DOI: 10.1016/J.BMC.2015.01.001 * |
| DAI, S.; QU, L.; LI, J.; ZHANG, Y.; JIANG, L.; WEI, H.; GUO, M.; CHEN, X.; CHEN, Y.: "Structural Insight into the Ligand Binding Mechanism of Aryl Hydrocarbon Receptor", NAT CONINIIT, vol. 13, no. 1, 2022, pages 6234, Retrieved from the Internet <URL:https://doi.org/10.1038/s41467-022-33858-w> |
| DATABASE REGISTRY [online] Chemical Abstract Service, Columbus, Ohio, US; 17 August 2006 (2006-08-17), "902311-61-9", XP093164852, retrieved from https://stn.org/stn/# Database accession no. 902311-61-9 * |
| DATABASE REGISTRY [online] Chemical Abstract Service, Columbus, Ohio, US; 17 August 2006 (2006-08-17), "902322-71-8", XP093164851, retrieved from https://stn.org/stn/# Database accession no. 902322-71-8 * |
| DATABASE REGISTRY [online] Chemical Abstract Service, Columbus, Ohio, US; 17 August 2006 (2006-08-17), "902331-82-2", XP093164849, retrieved from https://stn.org/stn/# Database accession no. 902331-82-2 * |
| DATABASE REGISTRY [online] Chemical Abstract Service, Columbus, Ohio, US; 17 August 2006 (2006-08-17), H19 C21: "902326-81-2", XP093164850, retrieved from https://stn.org/stn/# Database accession no. 902326-81-2 * |
| DATABASE REGISTRY [online] Chemical Abstract Service, Columbus, Ohio, US; 27 January 2017 (2017-01-27), "2060525-78-0", XP093164848, retrieved from https://stn.org/stn/# Database accession no. 2060525-78-0 * |
| DATABASE REGISTRY [online] Chemical Abstract Service, Columbus, Ohio, US; 5 July 2006 (2006-07-05), "890624-46-1", XP093164854, retrieved from https://stn.org/stn/# Database accession no. 890624-46-1 * |
| GRUSZCZYK, J.GRANDVUILLEMIN, L.LAI-KEE-HIM, J.PALONI, M.SAVVA, C. G.GERMAIN, P.GRIMALDI, M.BOULAHTOUF, A.KWONG, H.-S.BOUS, J.: "Cryo-EM Structure of the Agonist-Bound Hsp90-XAP2-AHR Cytosolic Complex", NAT COMMUN, vol. 13, no. 1, 2022, pages 7010, Retrieved from the Internet <URL:https://doi.org/10.1038/s41467-022-34773-w> |
| GUTIÉRREZ-VÁZQUEZ, C.QUINTANA, F. J: "Regulation of the Immune Response by the Aryl Hydrocarbon Receptor", IMMUNITY, vol. 8, no. 1, 2018, pages 19 - 33, XP055686215, Retrieved from the Internet <URL:https://doi.org/10.1016/j.immuni.2017.12.012> DOI: 10.1016/j.immuni.2017.12.012 |
| LIN, L.DAI, Y.XIA, Y: "An Overview of Aryl Hydrocarbon Receptor Ligands in the Last Two Decades (2002-2022): A Medicinal Chemistry Perspective", EUR J MED CHEM, vol. 244, 2022, pages 114845 |
| MCGOVERN, K.; CASTRO, A.; CAVANAUGH, J.; SANCHEZ-MARTIN, M.; TCHAICHA, J.; SYED, S.;FROSCH, H.; NATARAJAN, P.; O'CALLAGHAN, K.; AM: "Regular and yoit g investigator award abstracts", 2020, BMJ PUBLISHING GROUP LTD, article "448 Discovery of Clinical Candidate IK-175, a Selective Orally Active AHR Antagonist" |
| MURRAY, I. A.PATTERSON, A. D.PERDEW, G. H: "Aryl Hydrocarbon Receptor Ligands in Cancer: Friend and Foe", NATREV CANCER, vol. 14, no. 12, 2014, pages 801 - 814, XP055356946, Retrieved from the Internet <URL:https://doi.org/10.1038/nrc3846> DOI: 10.1038/nrc3846 |
| ROTHHAMMER, V.QUINTANA, F. J: "The Aryl Hydrocarbon Receptor: An Environmental Sensor Integrating Immune Responses in Health and Disease", NAT REV IMMUNOL, vol. 19, no. 3, 2019, pages 184 - 197, XP036712991, Retrieved from the Internet <URL:https://doi.org/10.1038/s41577-019-0125-8> DOI: 10.1038/s41577-019-0125-8 |
| SCHEUERMANN, T. H.; STROUD, D.; SLEET, C. E.; BAYEH, L.; SHOKRI, C.; WANG, H.;CALDWELL, C. G.; LONGGOOD, J.; MACMILLAN, J. B.; BRU: "Isoform-Selective and Stereoselective Inhibition of Hypoxia Inducible Factor-2", J MED CHEM, vol. 58, no. 15, 2015, pages 5930 - 5941, XP055335881, Retrieved from the Internet <URL:https://do1.0org/10.1021/acs.jmedchem.5600529> DOI: 10.1021/acs.jmedchem.5b00529 |
| SCHULTE, K. W.GREEN, E.WILZ, A.PLATTEN, M.DAUMKE, O: "Structural Basis for Aryl Hydrocarbon Receptor-Mediated Gene Activation", STRUCTURE, vol. 25, no. 7, 2017, pages 1025 - 1033, XP085115767, Retrieved from the Internet <URL:https://doi.org/10.1016/j.str.2017.05.008> DOI: 10.1016/j.str.2017.05.008 |
| SEOK, S. H.; LEE, W.; JIANG, L.; MOLUGU, K.; ZHENG, A.; LI, Y.; PARK, S.; BRADFIELD, C.A.; XING, Y.: "Structural Hierarchy Controlling Dimerization and Target DNA Recognition in the AHR Transcriptional Complex", PROC NATL ACAD SCI USA, vol. 114, no. 21, 2017, pages 5431 - 5436, Retrieved from the Internet <URL:https://doi.org/10.1073/pnas.1617035114> |
| SMITH, K. J.MURRAY, I. A.TANOS, R.TELLEW, J.BOITANO, A. E.BISSON, W. H.KOLLURI, S. K.COOKE, M. P.PERDEW, G. H: "Identification of a High-Affinity Ligand That Exhibits Complete Aryl Hydrocarbon Receptor Antagonism", JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 338, no. 1, 2011, pages 318 - 327, XP055122740, Retrieved from the Internet <URL:https://doi.org/10.1124/jpet.110.178392> DOI: 10.1124/jpet.110.178392 |
| STOCKINGER, B.MEGLIO, P. DIGIALITAKIS, M.DUARTE, J. H: "The Aryl Hydrocarbon Receptor: Multitasking in the Immune System", ANNU REV IMMUNOL, vol. 32, no. 1, 2014, pages 403 - 432, XP093072307, Retrieved from the Internet <URL:https://doi.org/10.1146/annurev-immunol-032713-120245> DOI: 10.1146/annurev-immunol-032713-120245 |
| SUN, L: "Recent Advances in the Development of AHR Antagonists in Immuno-Oncology", RSC MED CHEM, vol. 12, no. 6, 2021, pages 902 - 914, Retrieved from the Internet <URL:https://doi.org/10.1039/d1md00015b> |
| SUTHERLAND HAMISH S. ET AL: "Synthesis and Structure-Activity Relationships for the Anti-Mycobacterial Activity of 3-Phenyl-N-(Pyridin-2-ylmethyl)Pyrazolo[1,5-a]Pyrimidin-7-Amines", PHARMACEUTICALS, vol. 15, no. 9, 8 September 2022 (2022-09-08), pages 1125, XP093039451, DOI: 10.3390/ph15091125 * |
| TOMPKINS, L. M.LI, H.LI, L.LYNCH, C.XIE, Y.NAKANISHI, T.ROSS, D. D.WANG, H: "A Novel Xenobiotic Responsive Element Regulated by Aryl Hydrocarbon Receptor Is Involved in the Induction of BCRP/ABCG2 in LS174T Cells", BIOCHEM PHARMACOL, vol. 80, no. 11, 2010, pages 1754 - 1761, Retrieved from the Internet <URL:https://doi.org/10.1016/j.bcp.2010.08.016> |
| WEN, Z.ZHANG, YZHANG, B.HANG, Y.XU, L.CHEN, Y.XIE, Q.ZHAO, Q.ZHANG, L.LI, G.: "Cryo-EM Structure of the Cytosolic AhR Complex", STRUCTURE, vol. 31, no. 3, 2023, pages 295 - 308, Retrieved from the Internet <URL:https://doi.org/10.1016/j.str.2022.12.013> |
| WILKINSON, I. V. L.PERKINS, K. J.DUGDALE, H.MOIR, L.VUORINEN, A.CHATZOPOULOU, M.SQUIRE, S. E.MONECKE, S.LOMOW, A.GEESE, M.: "Chemical Proteomics and Phenotypic Profiling Identifies the Aryl Hydrocarbon Receptor as a Molecular Target of the Utrophin Modulator Ezutromid", AFRGEWΑFRDTE CHEMIE - INTERNATIONAL EDITION, vol. 59, no. 6, 2020, pages 2420 - 2428, XP072104860, Retrieved from the Internet <URL:https://doi.org/10.1002/anie.201912392> DOI: 10.1002/anie.201912392 |
| YIN L., SYNTHESIS OF NEW CALCI EITRI INHIBITORS VIA PALLADIUM CATALYSIS CROSS COUPLING REACTIONS, 2005 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP5116660B2 (en) | HIV integrase inhibitor | |
| AU2019312393B2 (en) | Iminosulfanone compound as bromodomain protein inhibitor and pharmaceutical composition and medical use thereof | |
| US20090136499A1 (en) | RAF Inhibitors and Uses Thereof | |
| CA2693904A1 (en) | Heterocyclic compounds useful as mk2 inhibitors | |
| KR20150011838A (en) | Pyrrolopyrazone inhibitors of tankyrase | |
| WO2022135365A1 (en) | Disubstituted cyclopentane kinase inhibitors | |
| Bhattarai et al. | Design, synthesis, and biological evaluation of structurally modified isoindolinone and quinazolinone derivatives as hedgehog pathway inhibitors | |
| JP2010508288A (en) | Heterocyclic amide compounds useful as kinase inhibitors | |
| KR20150005711A (en) | Pyrrolopyrimidone and pyrrolopyridone inhibitors of tankyrase | |
| Cherukumalli et al. | Design and synthesis of novel urea derivatives of pyrimidine-pyrazoles as anticancer agents | |
| EP2258704A1 (en) | Synthesis of imidazooxazole and imidazothiazole inhibitors of p38 map kinase | |
| EP4612155A1 (en) | 1,5-naphthyridine derivatives as kras oncoprotein inhibitors | |
| JP4000268B2 (en) | 4- (Phenylamino)-[1,4] dioxano [2,3-g] quinazoline derivative and method for producing the same | |
| Perupogu et al. | Anticancer activity of newly synthesized 1, 2, 4-oxadiazole linked 4-(oxazolo [5, 4-d] pyrimidine derivatives | |
| Ashok et al. | Design, synthesis and biological evaluation of 1, 2, 4-oxadiazole linked 1, 2, 4-thiadiazole-pyrimidines as anticancer agents | |
| Naseer et al. | Studies on chromene based 2, 6-disubstituted-thiazolo [3, 2-B][1, 2, 4] triazole derivatives: Synthesis and biological evaluation | |
| CA3195127A1 (en) | Rev-erb agonists | |
| Tabibi et al. | Efficient and green synthesis of novel hexahydro-5 H-thiazolo [2', 3': 2, 3] pyrimido [4, 5-b] quinoline derivatives | |
| Meng et al. | Synthesis and Antitumor Activity Evaluation of 2, 4-Substituted Py-rimidine Derivatives Containing Trifluoromethyl | |
| Alapati et al. | Design, synthesis and biological various aryl derivatives of (pyridin-4-yl) imidazo [1, 5-a] pyridin-1-yl) oxazoles as anticancer agents | |
| JP2025534444A (en) | Phosphoramidate compounds and uses thereof | |
| Jin et al. | Synthesis and biological evaluation of 1-substituted-3 (5)-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl) pyrazoles as transforming growth factor-β type 1 receptor kinase inhibitors | |
| KR20090127272A (en) | Fused Pyrimidinone Compounds as MmZeruca R Ligands | |
| WO2025114771A1 (en) | A new class of aryl hydrocarbon receptor antagonists and their optimisation | |
| Zhang et al. | Discovery of novel pyrimidine molecules containing boronic acid as VCP/p97 Inhibitors |
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: 24786564 Country of ref document: EP Kind code of ref document: A1 |