The largest database of trusted experimental protocols

Piperidine

Piperidine is a heterocyclic organic compound consisting of a six-membered ring with one nitrogen atom.
It is a colorless, flammable liquid with a distinctive fishy odor.
Piperidine and its derivatives are widely used in organic synthesis, pharmaceuticals, and as industrial solvents.
They are also found in various natural products, such as alkaloids.
Piperidine's chemical structure and properties make it a versatile building block for the development of diverse compounds, including many biologically active molecules.
Researchers in the field of piperidine chemistry can leverage PubCompare.ai's AI-driven platform to locate the best protocols from literature, preprints, and patents, enhancing their research reproducibility and accuaracy.

Most cited protocols related to «Piperidine»

BMDM were seeded at 5 ×0 105/ml or 1 × 106/ml, HMDM at 5 × 105/ml and PBMC at 2 × 106/ml or 5 ×0 106/ml in 96 well plates. The following day the overnight medium was replaced and cells were stimulated with 10 ng/ml LPS from Escherichia coli serotype EH100 (ra) TLRgrade™ (Alexis Biochemicals) for 3 h. Medium was removed and replaced with serum free medium (SFM) containing DMSO (1:1,000), MCC950 (0.001–10 µM), glyburide (200 µM) (Sigma Aldrich), parthenolide (10 µM) (Enzo Life Sciences) or Bayer cysteinyl leukotriene receptor antagonist 1-(5-carboxy-2{3-[4-(3-cyclohexylpropoxy)phenyl]propoxy}benzoyl)piperidine-4-carboxylic acid (40 µM) (Amgen Inc., Thousand Oaks, CA, USA) for 30 min. Cells were then stimulated with inflammasome activators: 5 mM adenosine 5’-triphosphate disodium salt hydrate (ATP) (1 h), 1 µg/ml Poly(deoxyadenylic-thymidylic) acid sodium salt (Poly dA:dT) (Sigma Aldrich) transfected with Lipofectamine 2000™ (Invitrogen) (3–4 h), 200 µg/ml MSU (overnight) and 10 µM nigericin (Invivogen) (1 h) or S. typhimurium UK-1 strain (M.O.I. 20) obtained from Dr. Sinead Corr, Trinity College Dublin, Ireland (2 h). Cells were also stimulated with 25 µg/ml Polyadenylic-polyuridylic acid (Invivogen) (4 h). For non-canonical inflammasome activation cells were primed with 100 ng/ml Pam3CSK4 (Invivogen) for 4 h, medium was removed and replaced with SFM containing DMSO or MCC950 and 2 µg/ml LPS was transfected using 0.25% FuGENE® (Promega) for 16 h. Supernatants were removed and analysed using ELISA kits according to the manufacturer’s instructions (DuoSet® R&D Systems or ReadySetGo!® eBioscience). LDH release was measured using the CytoTox96® non-radioactive cytotoxicity assay (Promega)
Publication 2015
Adenosine Triphosphate Biological Assay Carboxylic Acids Cells cysteinyl leukotriene receptor Cytotoxin Enzyme-Linked Immunosorbent Assay Escherichia coli FuGene Glyburide Inflammasomes Leukotriene Antagonists lipofectamine 2000 MCC-950 Nigericin parthenolide piperidine poly(dA) Poly A Poly A-U Promega Quercus Radioactivity Serum Sodium Sodium Chloride Strains Sulfoxide, Dimethyl Thymidine Monophosphate

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2009
1-hydroxybenzotriazole Cholic Acid Common Cold Dendrites Esters Ethers Lysine piperidine Polylysine Powder Vacuum

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2010
acrylate Amines Anabolism butanediol diacrylate Desiccants Gel Chromatography M 320 Molar piperidine Poly A Polymers Solvents Sulfoxide, Dimethyl
A reaction mixture of 6-methoxy-2-naphthol (1) (0.01 mol), different aromatic aldehydes (2a-2q) (0.01 mol), malononitrile (3) (0.01 mol), and piperidine (0.5 ml) in ethanol (25 ml) was heated under microwave irradiation conditions at 140°C for 2 min. After completion of the reaction, the reaction mixture was cooled to room temperature, and the precipitated solid was filtered off, washed with methanol, and recrystallized from suitable solvent. The physical and spectral data of compounds 4a–4q are as follows:
3-Amino-1-(4-fluorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4a). Compound 4a was prepared according to the literature procedure (Ahmed et al., 2018 (link)).
3-Amino-1-(2-chlorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4b). Colorless needles from ethanol; yield 82%; m.p. 265–266°C; IR (KBr) υ (cm−1): 3,407, 3,330, 3,210 (NH2), 2,200 (CN); 1H-NMR δ: 7.87–6.99 (m, 9H, aromatic), 7.00 (bs, 2H, NH2), 5.67 (s, 1H, H-1), 3.82 (s, 3H, OCH3); 13C-NMR δ: 159.96 (C-3), 156.49 (C-8), 145.64 (C-4a), 130.97 (C-6a), 129.50 (C-10a), 128.15 (C-6), 125.01 (C-10), 119.87 (C-10b), 119.35 (C-9), 117.09 (C-7), 114.92 (CN), 107.57 (C-5), 56.11 (C-2), 55.21 (CH3), 35.17 (C-1), 142.66, 132.14, 130.00, 128.58, 128.44, 124.09 (aromatic); MS m/z (%): 364 (M+ + 2, 4.65), 362 (M+, 13.54) with a base peak at 251 (100); Anal. Calcd for C21H15ClN2O2 (362.81): C, 69.52; H, 4.17; N, 7.72. Found: C, 69.47; H, 4.13; N, 7.68%.
3-Amino-1-(3-chlorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4c). Colorless needles from ethanol; yield 86%; m.p. 215–216°C; IR (KBr) υ (cm−1): 3,468, 3,324, 3,198 (NH2), 2,197 (CN); 1H-NMR δ: 7.87–7.11 (m, 9H, aromatic), 7.03 (bs, 2H, NH2), 5.36 (s, 1H, H-1), 3.83 (s, 3H, OCH3); 13C-NMR δ: 159.94 (C-3), 156.52 (C-8), 148.19 (C-4a), 132.21 (C-6a), 128.48 (C-10a), 126.63 (C-6), 124.99 (C-10), 120.31 (C-10b), 119.21 (C-9), 117.11 (C-7), 115.11 (CN), 107.35 (C-5), 57.18 (C-2), 55.20 (CH3), 37.60 (C-1), 145.36, 133.18, 130.66, 126.61, 125.65 (aromatic); MS m/z (%): 364 (M++ 2, 1.24), 362 (M+, 3.58) with a base peak at 208 (100); Anal. Calcd for C21H15ClN2O2 (362.81): C, 69.52; H, 4.17; N, 7.72. Found: C, 69.57; H, 4.23; N, 7.77%.
3-Amino-1-(4-chlorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4d). Compound 4d was prepared according to the literature procedure (Ahmed et al., 2018 (link)).
3-Amino-1-(4-bromophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4e). Compound 4e was prepared according to the literature procedure (Ahmed et al., 2018 (link)).
3-Amino-1-(4-iodophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4f). Pale yellow needles from ethanol; yield 89%; m.p. 227–228°C; IR (KBr) υ (cm−1): 3,455, 3,322, 3,190 (NH2), 2,201 (CN); 1H-NMR δ: 7.85–6.99 (m, 9H, aromatic), 6.98 (bs, 2H, NH2), 5.27 (s, 1H, H-1), 3.82 (s, 3H, OCH3); 13C-NMR δ: 159.80 (C-3), 156.48 (C-8), 145.31 (C-4a), 132.20 (C-6a), 128.37 (C-10a), 125.01 (C-6), 120.37 (C-10), 119.11 (C-10b), 117.08 (C-7,9), 115.22 (CN), 107.35 (C-5), 57.29 (C-2), 55.21 (CH3), 37.69 (C-1), 145.54, 137.43, 129.32, 92.40 (aromatic); MS m/z (%): 454 (M+, 100); Anal. Calcd for C21H15IN2O2 (454.26): C, 55.52; H, 3.33; N, 6.17. Found: C, 55.65; H, 3.46; N, 6.34%.
3-Amino-1-(2,4-difluorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4g). Colorless crystals from ethanol; yield 81%; m.p. 301–302°C; IR (KBr) υ (cm−1): 3,476, 3,335, 3,291 (NH2), 2,201 (CN); 1H-NMR δ: 7.83–7.05 (m, 8H, aromatic), 7.09 (bs, 2H, NH2), 5.67 (s, 1H, H-1), 3.82 (s, 3H, OCH3); 13C-NMR δ: 160.71 (C-3), 149.61 (C-8), 145.61 (C-4a), 131.44 (C-6a), 129.11 (C-10a), 128.39 (C-6), 123.52 (C-10), 120.18 (C-9), 120.04 (C-7), 119.92 (C-10b), 116.94 (CN), 107.57 (C-5), 55.19 (C-2), 53.58 (CH3), 28.39 (C-1), 161.40, 156.14, 129.39, 125.59, 116.64, 112.82 (aromatic); MS m/z (%): 364 (M+, 61.46) with a base peak at 251 (100); Anal. Calcd for C21H14F2N2O2 (364.34): C, 69.23; H, 3.87; N, 7.69. Found: C, 69.29; H, 3.92; N, 7.73%.
3-Amino-1-(2,6-difluorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4h). Compound 4h was prepared according to the literature procedure (Halawa et al., 2017 (link)).
3-Amino-1-(2,3-dichlorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4i). Colorless needles from ethanol; yield 87%; m.p. 255–256°C; IR (KBr) υ (cm−1): 3,423, 3,335, 3,206 (NH2), 2,196 (CN); 1H-NMR δ: 7.88–6.96 (m, 8H, aromatic), 7.07 (bs, 2H, NH2), 5.74 (s, 1H, H-1), 3.83 (s, 3H, OCH3); 13C-NMR δ: 160.08 (C-3), 156.53 (C-8), 145.23 (C-4a), 131.98 (C-6a), 129.16 (C-10a), 128.79 (C-6), 124.92 (C-10), 119.77 (C-10b), 119.50 (C-9), 117.09 (C-7), 114.44 (CN), 107.65 (C-5), 55.59 (C-2), 55.21 (CH3), 36.22 (C-1), 145.63, 132.16, 129.01, 128.93, 128.64, 123.95 (aromatic); MS m/z (%): 400 (M++ 4, 10.10), 398 (M++ 2, 3.37), 396 (M+, 12.08) with a base peak at 110 (100); Anal. Calcd for C21H14Cl2N2O2 (397.25): C, 63.49; H, 3.55; N, 7.05. Found: C, 63.54; H, 3.60; N, 7.10%.
3-Amino-1-(2,4-dichlorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4j). Compound 4j was prepared according to the literature procedure (Okasha et al., 2017 (link)).
3-Amino-1-(2,5-dichlorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4k). Compound 4k was prepared according to the literature procedure (El Gaafary et al., 2021 (link)).
3-Amino-1-(2,6-dichlorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4l). Colorless crystals from ethanol; yield 85%; m.p. 314–315°C; IR (KBr) υ (cm−1): 3,466, 3,318, 3,198 (NH2), 2,193 (CN); 1H-NMR δ: 7.84–7.09 (m, 8H, aromatic), 7.05 (bs, 2H, NH2), 6.09 (s, 1H, H-1), 3.82 (s, 3H, OCH3); 13C-NMR δ: 160.41 (C-3), 156.22 (C-8), 146.39 (C-4a), 134.33 (C-6a), 132.01 (C-10a), 128.86 (C-6), 125.22 (C-10), 123.83 (C-10b), 119.56 (C-9), 119.18 (C-7), 116.78 (CN), 107.69 (C-5), 55.18 (C-2), 52.59 (CH3), 35.19 (C-1), 137.45, 135.04, 130.96, 129.57 (aromatic); MS m/z (%): 400 (M++ 4, 6.19), 398 (M++ 2, 2.09), 396 (M+, 7.88) with a base peak at 251 (100); Anal. Calcd for C21H14Cl2N2O2 (397.25): C, 63.49; H, 3.55; N, 7.05. Found: C, 63.46; H, 3.52; N, 7.02%.
3-Amino-1-(3,4-dichlorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4m). Colorless crystals from ethanol/benzene; yield 85%; m.p. 240–241°C; IR (KBr) υ (cm−1): 3,412, 3,328, 3,200 (NH2), 2,198 (CN); 1H-NMR δ: 7.88–6.96 (m, 8H, aromatic), 7.08 (bs, 2H, NH2), 5.76 (s, 1H, H-1), 3.84 (s, 3H, OCH3); 13C-NMR δ: 160.08 (C-3), 156.53 (C-8), 145.63 (C-4a), 129.01 (C-6a), 128.79 (C-10a), 128.64 (C-6), 123.95 (C-10), 119.77 (C-10b), 119.50 (C-9), 117.09 (C-5), 114.44 (CN), 107.65 (C-7), 55.59 (C-2), 55.21 (CH3), 36.23 (C-1), 145.23, 132.16, 131.98, 129.15, 128.93, 128.79, 124.92 (aromatic); MS m/z (%): 400 (M++ 4, 1.49), 398 (M++ 2, 0.47), 396 (M+, 1.79) with a base peak at 208 (100); Anal. Calcd for C21H14Cl2N2O2 (397.25): C, 63.49; H, 3.55; N, 7.05. Found: C, 63.46; H, 3.52; N, 7.02%.
3-Amino-1-(3,5-dibromo-2-methoxyphenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4n). Compound 4n was prepared according to the literature procedure (Amr et al., 2017 (link)).
3-Amino-1-(2,3,4-trimethoxyphenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4o). Colorless crystals from ethanol; yield 84%; m.p. 239–240°C; IR (KBr) υ (cm−1): 3,424, 3,381, 3,322 (NH2), 2,192 (CN); 1H-NMR δ: 7.77–6.55 (m, 7H, aromatic), 6.94 (bs, 2H, NH2), 5.31 (s, 1H, H-1), 3.80 (s, 3H, OCH3), 3.78 (s, 6H, 2OCH3), 3.71 (s, 3H, OCH3); 13C-NMR δ: 159.98 (C-3), 156.79 (C-8), 145.65 (C-4a), 131.83 (C-10a), 128.25 (C-6), 125.74 (C-6a), 125.03 (C-10), 121.54 (C-10b), 119.39 (C-7), 117.59 (C-9), 116.88 (CN), 107.62 (C-5), 60.91 (CH3), 60.54 (C-2), 57.53 (C-2), 55.98 (CH3), 55.41 (CH3), 33.01 (C-1), 152.86, 140.92, 136.11, 112.64, 104.62 (aromatic); MS m/z (%): 418 (M+, 77.17) with a base peak at 387 (100); Anal. Calcd for C24H22N2O5 (418.44): C, 68.89; H, 5.30; N, 6.69. Found: C, 68.81; H, 5.29; N, 6.61%.
3-Amino-1-(3,4,5-trimethoxyphenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4p). Colorless crystals from ethanol; yield 84%; m.p. 259–260°C; IR (KBr) υ (cm−1): 3,469, 3,350, 3,225 (NH2), 2,197 (CN); 1H-NMR δ: 7.78–6.60 (m, 7H, aromatic), 6.92 (bs, 2H, NH2), 5.35 (s, 1H, H-1), 3.81 (s, 3H, OCH3), 3.79 (s, 3H, OCH3), 3.72 (s, 3H, OCH3), 3.66 (s, 3H, OCH3); 13C-NMR δ: 160.60 (C-3), 156.80 (C-8), 145.70 (C-4a), 131.85 (C-10a), 128.23 (C-6), 125.75 (C-6a), 125.00 (C-10), 121.53 (C-10b), 119.46 (C-7), 117.55 (C-9), 116.86 (CN), 107.60 (C-5), 61.75 (CH3), 60.62 (CH3), 57.62 (C-2), 56.05 (CH3), 55.57 (CH3), 33.04 (C-1), 152.49, 150.24, 141.71, 132.45, 123.80, 108.77 (aromatic); 13C-NMR-DEPT spectrum at 135°: CH, CH3 [positive (up)] and CH2 [negative (down)], revealed the following signals at δ: 128.23 (C-6 ↑), 125.00 (C-10 ↑), 123.80 (aromatic ↑), 119.46 (C-7 ↑), 117.55 (C-9 ↑), 108.77 (aromatic ↑), 107.60 (C-5 ↑), 61.75 (CH3 ↑), 60.62 (CH3 ↑), 56.05 (CH3 ↑), 55.57 (CH3 ↑), 33.04 (C-1 ↑); MS m/z (%): 418 (M+, 13.93) with a base peak at 40 (100); Anal. Calcd for C24H22N2O5 (418.44): C, 68.89; H, 5.30; N, 6.69. Found: C, 68.95; H, 5.37; N, 6.76%.
3-Amino-1-(2,3,5-trichlorophenyl)-8-methoxy-1H-benzo[f]chromene-2-carbonitrile (4q). Colorless crystals from ethanol; yield 84%; m.p. 259–260°C; IR (KBr) υ (cm−1): 3,441, 3,333, 3,210 (NH2), 2,183 (CN); MS m/z (%): 436 (M++ 6, 3.91), 434 (M++ 4, 33.80), 432 (M++ 2, 96.27), 430 (M+, 100); Anal. Calcd for C21H13Cl3N2O2 (431.70): C, 58.43; H, 3.04; N, 6.49. Found: C, 58.49; H, 3.10; N, 6.54%.
Full text: Click here
Publication 2021

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2010
4-methylmorpholine acetonitrile Amino Acids Biotin Ethyl Ether High-Performance Liquid Chromatographies Histone H2a Histone H3 Ninhydrin Peptides piperidine Resins, Plant Rink amide resin Solvents Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization Trifluoroacetic Acid

Most recents protocols related to «Piperidine»

Targeted pyrrole/piperidine derivatives (126) based on benzimidazole were afforded via several steps. 2-Marcaptobenzimidazole (I, 1 equivalent) was treated with variously substituted phenacyl bromides (1 equivalent) in ethanol (10 mL) and triethylamine (catalyst), and the resulting reaction mixture was then refluxed for 4 h to offer S-substituted benzimidazole (II, 1 equivalent), which was then further redissolved in hydrazine hydrate (5 mL) solution while being agitated in acetic acid (10 mL). The solvent was evaporated after 3 h of refluxing, and the resultant solid residue (III) underwent two distinct reactions. Substrate (III, 1 equivalent) was first placed in ethanol (10 mL) and triethylamine (catalyst), and then 1H-pyrrole-3-carbaldehyde (1 equivalent) was added. For seven hours, the reaction mixture was refluxed and stirred to form the desired pyrrole derivatives (113) based on benzimidazole. Additionally, for the benzimidazole-based piperidine derivatives (1426), the intermediate (III, 1 equivalent) was again reacted with piperidine-1-carbaldehyde (1 equivalent) in ethanol (10 mL) along with catalytic amounts of triethylamine.
Full text: Click here
Publication 2024

Example 2

[Figure (not displayed)]
[Figure (not displayed)]
[Figure (not displayed)]
[Figure (not displayed)]

Example 4

[Figure (not displayed)]

Compound 4a can be prepared according to the procedure described in Example 223 of U.S. Pat. No. 9,382,248.

Biological Studies

peIF4E Signaling Cellular Assay

Phosphorylated eIF4E is assayed using the CisBio peIF4E HTRF® assay kit (CisBio, catalogue No. 64EF4PEG). Cells are plated in 96-well tissue-culture treated plate in appropriate growth medium (90 μL). Compounds (10×) are diluted using 3-fold serial dilutions in cell culture medium and added to cells. Plates are incubated for 2 hrs at 37° C. The cell supernatant is carefully removed either by aspirating supernatant or by flicking the plate. Immediately 50 μL of supplemented lysis buffer (1×) is added and incubated for at least 30 minutes at room temperature under shaking. After homogenization by pipeting up and down, 16 μL of cell lysate is transferred from the 96-well cell-culture plate to a 384-well small volume white plate. 4 μL of premixed antibody solutions (vol/vol) is prepared in the detection buffer and added. The plate is covered with a plate sealer and incubated overnight at room temperature. The fluorescence emissions at two different wavelengths are read (665 nm and 620 nm) on a Wallac Victor2. Emission ratios are converted into percent inhibitions and imported into GraphPad Prism software. The concentration of compound necessary to achieve inhibition of enzyme activity by 50% (IC50) is calculated using concentrations ranging from 20 μM to 0.1 nM (12-point curve). IC50 values are determined using a nonlinear regression model available in GraphPad Prism 5.

The results of these assays are set forth in Table 1 below. To this end, IC50 values of less than 0.001 μM are labelled as “+++”, from 0.001 to 0.01 μM are labelled as “++”, and greater than 0.01 μM are labelled as “+” (NA means “not available”).

MNK Inhibition Decreases Expression of Immune Checkpoint Receptors and Ligands

Upon activation of T cell receptor (TCR) signaling, T cells proliferate, produce cytokines (e.g., IL-2) and induce the expression of immune checkpoint receptors. Programmed death 1 (PD-1) is an inhibitory checkpoint receptor expressed on the surface of activated T cells, as well as on myeloid cells. The ligand for PD-1, programmed death ligand-1 (PD-L1, B7-H1/CD274) is not expressed by T cells or normal epithelial cells, but is expressed by antigen presenting cells and overexpressed in several cancers. Interaction of PD-1 with PD-L1 results in an anti-proliferative effect on T cells and ultimately T cell exhaustion and apoptosis. To study the role of MNK in activated T cells and tumor cells, the effect of a MNK inhibitor on molecules of immune checkpoint control was examined.

PD-1 (CD279) Expression

To examine the effect of MNK inhibitors on PD-1 expression, Jurkat cells (Clone E6.1, ATCC, transformed T cells) were used, which express PD-1 when activated through T cell receptor (TCR) signaling. Briefly, Jurkat cells were grown in 1×RPMI with 1×Pen/Strep, and 10% FBS, then about 3×106 Jurkat cells were activated in presence of 1 μg/mL PHA (Sigma) and 50 ng/mL PMA (Sigma). Test Cells were treated simultaneously with various concentrations of an MNK inhibitor (0, 0.01, 0.1, 1, 3 and 10 μM). After 48 hours, culture supernatants were harvested and examined via sandwich ELISA for the presence of IL-2 using human IL-2 ELISA DuoSet (R&D Systems, Minneapolis, Minn.). The level of PD-1 on Jurkat cells was examined by incubating with human FcR block, then contacted with allophycocyanin (APC) conjugated anti-PD-1 antibody (5 μl per 100 μl volume of test, Biolegend, San Diego, CA) for 25 minutes at 4° C., without washing the cells, fixable dead cell stain (1:10,000; BD Biosciences, San Jose, CA) was added and incubated further for 10 minutes at 4° C. Cells were washed two times with flow buffer, and finally cells were fixed with fixation buffer for 15 minutes at 4° C. After fixation, cells were washed twice with flow buffer and re-suspended in flow buffer and assessed for fluorescence using BD Accuri C6 flow cytometer. Data were analyzed using the C6 cytometer software (BD Biosciences, San Jose, CA) or Attune Nxt Cytometer (Invitrogen, Carlsbad, CA).

Activation of Jurkat T cells with PHA and PMA induced the expression of PD-1 on the cell surface of about 25-30% of the stimulated Jurkat cells as compared to uninduced cells (Unstim) and induced a 1,000-fold increase in IL-2 cytokine production, respectively. Treatment of PHA/PMA activated Jurkat T cells with the MNK inhibitor resulted in a concentration dependent decrease in the expression of the immune inhibitory receptor PD-1, up to a 50% reduction at the highest concentration as compared to control. In addition, this reduction of PD-1 was not due to a block in Jurkat T cell activation per se since MNK inhibition by an MNK inhibitor did not alter cytokine production as measured by IL-2 levels. Lastly, MNK inhibition by various different MNK inhibitors had no effect on cell viability. In fact, various different MNK inhibitors in the Jurkat T cell assay showed the ability to downregulate immune checkpoint inhibitors without affecting cell viability. The results of these assays are set forth in Table 1 below. To this end, percentage of inhibition of PD-1 positive cells (10 μM) of more than 50% are labelled as “+++”, from 10% to 50% are labelled as “++”, and less than 10% are labelled as “+” (NA means “not available”).

Aqueous Solubility

Solubility, the phenomenon of dissolution of solute in solvent to give a homogenous system, is one of the important parameters to achieve a desired concentration of a compound in systemic circulation for desired (anticipated) pharmacological response. Compounds having good aqueous solubility are desirable because they result in good in vivo bioavailability owing to their high dissolution rate following administration to a subject. Compounds having good aqueous solubility also contribute to the ease of formulation development, formulation manufacture and stability of the formulation.

High Throughput Thermodynamic Solubility Procedure

In a 96-well plate, 10 mM DMSO stocks (50-100 uL) of each compound were dried under heated nitrogen flow using a SPE-96 plate dryer (upper flow rate=50 L/min, temperature=60° C., lower flow rate=20 L/min, temperature=80° C.). After DMSO had been completely removed, remaining materials were dissolved in test solvents including deionized water, Fasted-State Simulated Intestinal Fluid (FaSSIF, pH 6.5) and Fasted-State Simulated Gastric Fluid (FaSSGF, pH 1.6). Compounds prepared as the free base were assessed in conditions with and without one equivalent of TFA added. The theoretical maximum concentrations of the aqueous solutions were 10 mM. Each well was capped and incubated for a period of 18 hours at room temperature or 37° C. Mixing of the solutions during the incubation period was performed by either shaking the plate at 750 rpm or adding StirStix (stainless steel capillaries) and agitating the mixture using a rotary magnetic tumble stirrer. After the incubation period, aliquots from each well were filtered. Solubility of all samples were quantified following comparison to standards of known concentration by HPLC-UV.

The results of the aqueous solubility are set forth in Table 1 below.

TABLE 1
pEIF4E signaling activity, PD-1 inhibition activity and
aqueous solubility
Aqueous
pEI4IEPD-1Solubility
Cpd.signalinginhibitionFaSSGFFaSSIF
No.activityactivityDI H2O(pH 1.2)(pH 6.5)
3-piperidine-substituted compounds
3A+NA
3B+
3C+++++2.64.10.256
3D++3.34.40.11
rac-1F++++4.39.80.24
3E+
3F++0.240.7<1
3G++
3H+
3I+++
3J++
3K++++1.531.680.158
3L+++++3.23.10.07
3M
3N++
3O+++
3P+++
3Q++
3R+++>2.090.068
3S+++++4.2100.6
3T+++10100.5
3U+
rac-2F++++1.840.079
3V+1.820.107
3W++1.40.593
3X+++
3Y+++
1Fa+++>1.851.530.125
1Fb++++>1.85>1.850.088
2Fa++++1.841.320.077
2Fb+++1.771.770.151
3II
3JJ
3KK
3LL
3MM
3NN
3OO
3PP
3QQ
3RR
3SS
3TT
Cycloalkyl-substituted compounds
4A++++++0.00030.260.045
4-piperidine-substituted compounds
3Z++
3AA+++4.43.20.03
3BB++++
3CC++++0.771.90.04
3DD++++++2.130.13
3EE+++++
3FF+++++
3GG++3.6
3HH+++2.40.018

The inventors have unexpectedly found that the dissolution rate of an Mnk inhibitor compound can be remarkably improved over comparative compound 4a by: (i) substituting the imidazopyridine in the compound with 3- or 4-piperidine; and (ii) by substituting the pyrimidine in the compound with lower alkyl, halogen or cyano. Such substitutions or structural features induce chirality in the molecules.

The improvement in the aqueous solubility of the compounds of the invention is especially significant in low pH environments, such as in gastric or stomach fluid. For example, as shown in Table 1, compounds 3D, 3K, 3L, 3X, 3Y, rac-1F, 1Fa, 1Fb, rac-2F, 2Fa and 2Fb exhibited excellent solubility in fasted state simulated gastric fluid (FaSSGF), which has a pH value of 1.6.

Importantly, in addition to improved aqueous solubility, the 3- and 4-piperidine-substituted compounds have also retained their anti-cancer potency in the form of their ability to inhibit pEIF4E signaling. In some cases, the anti-cancer potency is further manifested in the ability of these 3- and 4-piperidine-substituted compounds to inhibit PD-1, as can be seen in Table 1.

The various embodiments described above can be combined to provide further embodiments. All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, applications and publications to provide yet further embodiments.

These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.

Full text: Click here
Patent 2024
In an oven-dried round bottom flask was dissolved tert-butyl 4-(azidomethyl)piperidine-1-carboxylate (15.2 g, 63.3 mmol, 1.00 equiv.) in methanol (210 mL, C ∼ 0.2 M). The solution was cooled to 0°C and acetyl chloride (54.2 mL, 760 mmol, 12.0 equiv.) was added dropwise through a syringe over 30 minutes. The reaction was allowed to warm to room temperature and stirred for 18 h until the complete consumption of the starting material (monitored by TLC). Excess of acetyl chloride and methanol were removed under reduced pressure and the residue was poured into diethyl ether and stirred at room temperature for 1 h. The resulting precipitate was filtered off, washed with cold diethyl ether (3 x 100 mL) and dried under reduced pressure to provide 4-(azidomethyl)piperidine hydrochloride as a white cristalline solid (11.0 g, 98% yield).
Full text: Click here
Publication 2024

Example 161

[Figure (not displayed)]
Step 1.

tert-butyl 5-(4-methyl-3-quinolyl)spiro[3H-benzofuran-2,4′-piperidine]-1′-carboxylate was synthesized using tert-butyl 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)spiro[3H-benzofuran-2,4′-piperidine]-1′-carboxylate and 3-bromo-4-methyl-quinoline Analysis: LCMS m/z=431 (M+1); 1H NMR (400 MHz, DMSO-d6) δ: 8.71 (s, 1H), 8.18 (dd, J=8.4, 0.9 Hz, 1H), 8.03 (dd, J=8.3, 1.0 Hz, 1H), 7.76 (ddd, J=8.3, 6.9, 1.4 Hz, 1H), 7.71-7.64 (m, J=8.3, 6.5, 1.4 Hz, 1H), 7.32 (d, J=1.5 Hz, 1H), 7.19 (dd, J=8.3, 2.0 Hz, 1H), 6.91 (d, J=8.0 Hz, 1H), 3.62-3.50 (m, 2H), 3.48-3.36 (m, 2H), 3.13 (s, 2H), 2.62 (s, 3H), 1.91-1.68 (m, 4H), 1.43 (s, 9H).

Step 2.

5-(4-Methyl-3-quinolyl)spiro[3H-benzofuran-2,4′-piperidine] 2HCl was synthesized using tert-butyl 5-(4-methyl-3-quinolyl)spiro[3H-benzofuran-2,4′-piperidine]-1′-carboxylate and 2N HCL in dioxane. Analysis: LCMS m/z=331 (M+1); 1H NMR (400 MHz, DMSO-d6) δ: 9.24 (br s, 1H), 9.06 (s, 1H), 8.45 (d, J=8.3 Hz, 1H), 8.35 (d, J=8.3 Hz, 1H), 8.13-8.03 (m, 1H), 7.97-7.88 (m, 1H), 7.44 (d, J=1.5 Hz, 1H), 7.32 (dd, J=8.3, 2.0 Hz, 1H), 7.00 (d, J=8.0 Hz, 1H), 3.22 (s, 6H), 2.81 (s, 3H), 2.14-2.02 (m, 4H).

Step 3.

5-(4-Methyl-3-quinolyl)-N-tetrahydropyran-2-yloxy-spiro[3H-benzofuran-2,4′-piperidine]-1′-carboxamide was synthesize using o-(tetrahydro-2H-pyran-2-yl)hydroxylamine and 5-(4-methyl-3-quinolyl)spiro[3H-benzofuran-2,4′-piperidine] 2HCl. LCMS m/z=474 (M+1).

Step 4.

5-(4-Methyl-3-quinolyl)spiro[3H-benzofuran-2,4′-piperidine]-1′-carbohydroxamic acid was synthesized using 5-(4-methyl-3-quinolyl)-N-tetrahydropyran-2-yloxy-spiro[3H-benzofuran-2,4′-piperidine]-1′-carboxamide and TFA. Analysis: LCMS m/z=390 (M+1); 1H NMR (400 MHz, DMSO-d6) δ: 9.13 (s, 1H), 8.71 (s, 1H), 8.18 (dd, J=8.5, 0.8 Hz, 1H), 8.03 (dd, J=8.3, 1.0 Hz, 2H), 7.76 (ddd, J=8.3, 6.9, 1.4 Hz, 1H), 7.71-7.63 (m, 1H), 7.31 (d, J=1.5 Hz, 1H), 7.19 (dd, J=8.2, 1.9 Hz, 1H), 6.91 (d, J=8.0 Hz, 1H), 3.53-3.38 (m, 4H), 3.13 (s, 2H), 2.62 (s, 3H), 1.87-1.69 (m, 4H).

Full text: Click here
Patent 2024

Example 175

[Figure (not displayed)]
Step 1.

tert-Butyl 6-(8-methoxy-7-quinolyl)spiro[chromane-2,4′-piperidine]-1′-carboxylate. A solution of tert-butyl 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)spiro[chromane-2,4′-piperidine]-1′-carboxylate (0.90 g, 2.1 mmol), 7-bromo-8-methoxy-quinoline (0.50 g, 2.1 mmol), palladium(II) acetate (0.024 g, 0.11 mmol), triphenylphosphine (0.11 g, 0.42 mmol), 1,4-dioxane (30 mL), and DMF (50 mL) was added aq. Na2CO3 (0.5 M) (8.0 mL, 4.0 mmol). The mixture was vacuum degassed then heated at 85° C. overnight. The mixture was treated with water (120 mL) then cooled to RT and extracted with EtOAc (3×70 mL). The organic extract was washed with a mixture of water:brine (9:1, 100 mL) then with brine (100 mL), dried over Na2SO4, filtered and concentrated in vacuo. The residue was dried overnight under vacuum then the residue was dissolved in DCM, applied to a silica gel loading cartridge (25 g) and purified on silica gel (40 g, 0-40% ethyl acetate:hexane) to afford tert-butyl 6-(8-methoxy-7-quinolyl)-spiro[chromane-2,4′-piperidine]-1′-carboxylate as a white solid. Analysis: LCMS m/z 461 (M+1); 1H NMR (400 MHz, DMSO-d6) δ: 8.94 (dd, J=4.3, 1.8 Hz, 1H), 8.38 (dd, J=8.4, 1.6 Hz, 1H), 7.75 (d, J=8.5 Hz, 1H), 7.58 (d, J=8.5 Hz, 1H), 7.56-7.52 (m, 1H), 7.42-7.36 (m, 2H), 6.90 (d, J=8.3 Hz, 1H), 3.93 (s, 3H), 3.78-3.68 (m, 2H), 3.29-3.11 (m, 2H), 2.82 (t, J=6.7 Hz, 2H), 1.85 (t, J=6.8 Hz, 2H), 1.79-1.70 (m, 2H), 1.62-1.52 (m, 2H), 1.42 (s, 9H).

Step 2.

6-(8-Methoxy-7-quinolyl)spiro[chromane-2,4′-piperidine] 2HCl. tert-Butyl 6-(8-methoxy-7-quinolyl)spiro[chromane-2,4′-piperidine]-1′-carboxylate was dissolved in HCl (2M in 1,4-dioxane) (6.0 mL, 12 mmol) and after 5 min a precipitate formed. The reaction was diluted with ethanol (6.0 mL), stirred at RT overnight, then concentrated in vacuo to afford 6-(8-methoxy-7-quinolyl)spiro[chromane-2,4′-piperidine] 2HCl. Analysis: LCMS 361 (M+1); 1H NMR (400 MHz, DMSO-d6) δ: 9.16-9.02 (m, 2H), 9.00-8.89 (m, 1H), 8.87-8.77 (m, 1H), 7.98 (br d, J=8.8 Hz, 1H), 7.88-7.72 (m, 2H), 7.52-7.48 (m, 2H), 7.01 (d, J=8.3 Hz, 1H), 3.80 (s, 3H), 3.27-3.19 (m, 2H), 3.18-3.07 (m, 2H), 2.87 (br t, J=6.7 Hz, 2H), 2.00-1.86 (m, 6H).

Step 3.

6-(8-Methoxy-7-quinolyl)spiro[chromane-2,4′-piperidine]-1′-carboxamide. A suspension of 6-(8-methoxy-7-quinolyl)spiro[chromane-2,4′-piperidine] 2HCl (0.101 g, 0.233 mmol) in THF (2.0 mL) was treated with DIPEA (0.11 g, 0.15 mL, 0.86 mmol). After stirring for 2 min, a white precipitate formed. ACN (1.0 mL) was added, followed by DMF (2.0 mL) to give a homogenous solution. Trimethylsilyl isocyanate (0.085 g, 0.10 mL, 0.63 mmol) was then added to the mixture. After 90 min, water (11 mL) was added to the mixture and was aged at RT then at 4° C. overnight. The fine precipitate was collected on a Hirsch funnel, washed with water and dried under vacuum to afford crude product. The solids were dissolved in DMSO and purified by preparative HPLC (5-50% ACN: water, containing 0.1% TFA) to afford 6-(8-methoxy-7-quinolyl)spiro[chromane-2,4′-piperidine]-1′-carboxamide (0.064 g, 0.16 mmol, 68%). The pure fractions (freebased) treated with aq. Na2CO3 (10 mL) then extracted with DCM (2×30 mL), dried, concentrated and reconcentrated from ethanol and further dried. Analysis: LCMS 404 (M+1); 1H NMR (400 MHz, DMSO-d6) δ: 8.94 (dd, J=4.0, 1.8 Hz, 1H), 8.38 (dd, J=8.3, 1.8 Hz, 1H), 7.75 (d, J=8.5 Hz, 1H), 7.58 (d, J=8.5 Hz, 1H), 7.56-7.52 (m, 1H), 7.41-7.35 (m, 2H), 6.90 (d, J=8.3 Hz, 1H), 5.96 (s, 2H), 3.93 (s, 3H), 3.74-3.65 (m, 2H), 3.22-3.12 (m, 2H), 2.83 (t, J=6.7 Hz, 2H), 1.85 (t, J=6.8 Hz, 2H), 1.75-1.66 (m, 2H), 1.60-1.51 (m, 2H).

Full text: Click here
Patent 2024

Top products related to «Piperidine»

Sourced in United States, Germany, Australia, Italy, France, Hungary, Canada, Poland, Sao Tome and Principe
Piperidine is a colorless, flammable liquid organic compound with the chemical formula C₅H₁₁N. It is a heterocyclic amine that is widely used as a building block in the synthesis of various pharmaceutical and industrial chemicals.
Sourced in United States, Germany, Australia, Hungary, Canada, France
Triisopropylsilane is a silicon-based organic compound. It is a colorless, volatile liquid with a mild odor. Triisopropylsilane is commonly used as a protecting group in organic synthesis, particularly in the protection of hydroxyl groups.
Sourced in United States, Germany, United Kingdom, France, Spain, Italy, Australia, China, India, Sao Tome and Principe, Poland, Canada, Switzerland, Macao, Belgium, Netherlands, Czechia, Japan, Austria, Brazil, Denmark, Ireland
Trifluoroacetic acid is a colorless, corrosive liquid commonly used as a reagent in organic synthesis and analytical chemistry. It has the chemical formula CF3COOH.
Sourced in Germany, United States, Italy, India, China, United Kingdom, France, Poland, Spain, Switzerland, Australia, Canada, Brazil, Sao Tome and Principe, Ireland, Belgium, Macao, Japan, Singapore, Mexico, Austria, Czechia, Bulgaria, Hungary, Egypt, Denmark, Chile, Malaysia, Israel, Croatia, Portugal, New Zealand, Romania, Norway, Sweden, Indonesia
Acetonitrile is a colorless, volatile, flammable liquid. It is a commonly used solvent in various analytical and chemical applications, including liquid chromatography, gas chromatography, and other laboratory procedures. Acetonitrile is known for its high polarity and ability to dissolve a wide range of organic compounds.
Sourced in United States, Germany, United Kingdom, China, Italy, Canada, India, Australia, Japan, Spain, Singapore, Sao Tome and Principe, Poland, France, Switzerland, Macao, Chile, Belgium, Czechia, Hungary, Netherlands
N,N-dimethylformamide is a clear, colorless liquid organic compound with the chemical formula (CH3)2NC(O)H. It is a common laboratory solvent used in various chemical reactions and processes.
Sourced in United States, Germany, Italy, Canada, France, Sao Tome and Principe, Czechia
N,N-diisopropylethylamine is a basic, organic compound commonly used as a reagent in chemical synthesis and laboratory procedures. It functions as a base and serves as a proton acceptor. The compound is colorless and has a characteristic amine-like odor.
Sourced in United States, Germany, Italy, India, United Kingdom, Spain, France, Belgium, Switzerland, Poland, China, Australia, Japan, Sao Tome and Principe, Canada
Diethyl ether is a colorless, volatile, and highly flammable liquid. It is commonly used as a laboratory solvent and reagent in various chemical processes and experiments.
Sourced in United States, Germany, United Kingdom, France, China, Italy, Switzerland, Sao Tome and Principe, Spain, Australia, Poland, Macao, Belgium, Sweden, Hungary, Netherlands, Ireland
Trifluoroacetic acid (TFA) is a colorless, corrosive liquid used in various laboratory applications. It is a strong organic acid with a chemical formula of CF3COOH. TFA is commonly utilized as a reagent or solvent in various chemical processes, including protein and peptide synthesis, sample preparation, and chromatographic techniques.
Sourced in United States
Piperidine is a versatile chemical compound commonly used in laboratory settings. It functions as a basic, cyclic amine with a variety of applications in organic synthesis and chemical analysis.
Sourced in Germany, United States, United Kingdom, Italy, India, France, Spain, China, Australia, Poland, Sweden, Sao Tome and Principe, Switzerland, Belgium, Denmark, Canada, Macao, Brazil, Portugal, Pakistan, Singapore, Ireland, Czechia, Mexico
Dichloromethane is a clear, colorless, and volatile liquid commonly used as a laboratory solvent. It has a molecular formula of CH2Cl2 and a molar mass of 84.93 g/mol. Dichloromethane is known for its high solvent power and low boiling point, making it suitable for various laboratory applications where a versatile and efficient solvent is required.

More about "Piperidine"

Piperidine is a versatile heterocyclic compound with a wide range of applications in organic synthesis, pharmaceuticals, and industrial solvents.
This six-membered nitrogen-containing ring structure is found in numerous natural products, including alkaloids.
Piperidine's unique chemical properties make it a valuable building block for the development of diverse biologically active molecules.
Researchers in the field of piperidine chemistry can leverage powerful AI-driven platforms like PubCompare.ai to optimize their research workflows.
These cutting-edge tools enable researchers to locate the best protocols from literature, preprints, and patents through intelligent comparisons, enhancing their research reproducibility and accuracy.
Beyond piperidine, other key reagents and solvents commonly used in piperidine synthesis and derivatization include triisopropylsilane, trifluoroacetic acid (TFA), acetonitrile, N,N-dimethylformamide (DMF), N,N-diisopropylethylamine (DIPEA), and dichloromethane.
These versatile compounds play crucial roles in various organic transformations, protecting group strategies, and purification techniques.
By harnessing the capabilities of AI-powered platforms like PubCompare.ai, piperidine researchers can streamline their workflows, stay up-to-date with the latest advancements, and enhance the reproducibility and accuracy of their work.
This enables them to push the boundaries of piperidine chemistry and unlock new avenues for the development of innovative pharmaceuticals, materials, and other valuable products.