WO2024125662A1 - 新型联芳环化合物作为免疫调节剂的制备和应用 - Google Patents

新型联芳环化合物作为免疫调节剂的制备和应用 Download PDF

Info

Publication number
WO2024125662A1
WO2024125662A1 PCT/CN2023/139583 CN2023139583W WO2024125662A1 WO 2024125662 A1 WO2024125662 A1 WO 2024125662A1 CN 2023139583 W CN2023139583 W CN 2023139583W WO 2024125662 A1 WO2024125662 A1 WO 2024125662A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
group
cancer
heteroatoms
Prior art date
Application number
PCT/CN2023/139583
Other languages
English (en)
French (fr)
Inventor
王喆
潜安然
李德亮
Original Assignee
上海长森药业有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 上海长森药业有限公司 filed Critical 上海长森药业有限公司
Publication of WO2024125662A1 publication Critical patent/WO2024125662A1/zh

Links

Definitions

  • the present invention relates to the field of small molecule drugs. Specifically, the present invention provides a small molecule compound that can be used to treat diseases related to the PD-1/PD-L1 signaling pathway.
  • the immune system plays a vital role in controlling and curing many diseases, such as various cancers and diseases caused by viruses.
  • cancer cells can often evade or suppress the immune system in some ways, thereby multiplying rapidly.
  • One of the ways is to change the activation and inhibition molecules expressed on immune cells.
  • Blocking inhibitory immune checkpoints, such as PD-1, has proven to be a very effective way to inhibit cancer cells.
  • PD-1 is programmed cell death protein-1, also known as CD279. It is mainly expressed in activated T cells and B cells, and its function is to inhibit cell activation, which is a normal self-stabilizing mechanism of the immune system. Because excessive T/B cell activation can cause autoimmune diseases, PD-1 is a protective wall for our body.
  • PD-1 is a type I transmembrane glycoprotein composed of 268 amino acids. Its structure mainly includes the outer immunoglobulin variable region, the hydrophobic transmembrane region, and the intracellular region.
  • the intracellular region contains two phosphorylation sites, which are located in the immunoreceptor tyrosine inhibitory motif and the immunoreceptor tyrosine conversion motif, which also proves that PD-1 can reversely regulate T cell receptor-mediated signals.
  • PD-1 has two ligands, PD-L1 and PD-L2, which are expressed differently.
  • PD-L1 is upregulated in a variety of tumor cells. It binds to PD-1 on T cells, inhibits T cell proliferation and activation, and makes T cells inactive, ultimately inducing immune escape.
  • PD-1/PD-L1 plays a reverse immunomodulatory role.
  • PD-1 binds to PD-L1, it can cause tyrosine polyphosphorylation in the tyrosine conversion motif domain of the T cell immune receptor, and the phosphorylated tyrosine can bind to the phosphatase protein tyrosinase 2 and protein tyrosinase 1.
  • This can hinder the activation of extracellular signal-regulated kinases, and can also block the activation of phosphatidylinositol 3-kinase (PI3K) and serine-threonine protein kinase (Akt), thereby inhibiting T lymphocyte proliferation and the secretion of related cytokines.
  • PI3K phosphatidylinositol 3-kinase
  • Akt serine-threonine protein kinase
  • PD-1/PD-L1 signals inhibit T cell activation and proliferation, they can also cause the secretion of cytokines interleukin 2, interferon ⁇ , and IL-10.
  • PD-1/PD-L1 signals also have similar immune functions on B cells.
  • the PD-1 cytoplasmic region interacts with tyrosinase containing the protein tyrosinase 2 binding site, thereby hindering the activation of B cells.
  • PD-1/PD-L1-based immunotherapy is a new generation of immunotherapy that has attracted much attention.
  • PD-1/PD-L1 inhibitors have strong anti-tumor activity against a variety of tumors.
  • PD-1/PD-L1 antibody modulators include BMS's Ninolumab, Merck's Lambrolizumab, and Roche's Atezolizumab.
  • PD-1/PD-L1 antibody modulators under development, including Cure Tech's Pidilizumab, GSK's AMP-224, and AstraZeneca's MEDI-4736.
  • tumor immunotherapy is considered to be a new generation of revolution in cancer treatment after targeted therapy
  • the PD-1 monoclonal antibodies currently on the market and under development have their own defects, including only being injected and not orally administered, being unstable in the body, being easily decomposed by proteases, being prone to immune cross-reactions, being difficult to purify and having high production costs, etc. Therefore, small molecule regulators of the PD-1/PD-L1 interaction are a better choice for tumor immunotherapy.
  • the purpose of the present invention is to provide a novel small molecule regulator of PD-1/PD-L1 interaction.
  • the first aspect of the present invention is a compound as shown in the following formula I, or an optical isomer, hydrate, solvate, or pharmaceutically acceptable salt thereof:
  • n 0, 1, 2, 3, 4 or 5;
  • n, v and p are each independently 0, 1, 2, 3 or 4;
  • Each is independently selected from the following group: a substituted or unsubstituted 5-12 membered heteroaryl, a substituted or unsubstituted C6-C10 aryl, a substituted or unsubstituted 5-12 membered heterocyclic group, a substituted or unsubstituted 5-12 membered C3-C12 (preferably C5-C12) cyclic group, wherein the heterocyclic group has 1-3 heteroatoms; or D is
  • X1 , X2 , X3 , X4 , X5 and X6 are each independently selected from the group consisting of N, O, S, SO, SO2 , C(R) 2 , CHR, NR; wherein each R may be optionally Replacement;
  • Y 1 , Y 2 , Y 3 , Y 4 , Y 5 and Y 6 are each independently selected from the group consisting of N, CH, C;
  • L 3 is selected from the group consisting of a chemical bond, a substituted or unsubstituted C 1 -C 7 alkylene group, a substituted or unsubstituted C 2 -C 4 alkenylene group, a substituted or unsubstituted C 2 -C 4 alkynylene group, -S-, -O-, a substituted or unsubstituted -NH-, -S(O)-, or -S(O) 2 -;
  • E is selected from the group consisting of a substituted or unsubstituted C6-C12 arylene group, a substituted or unsubstituted 5-12-membered heteroarylene group having 1-3 heteroatoms, a substituted or unsubstituted C3-C8 cycloalkylene group, and a substituted or unsubstituted 3-10-membered heterocyclylene group having 1-3 heteroatoms;
  • r is 1, 2, 3, 4, 5, 6;
  • s 0, 1, or 2;
  • a is 1, 2, 3, 4, 5 or 6;
  • R4 is selected from the following group:
  • Each is independently selected from the following group: a substituted or unsubstituted C6-C10 arylene group, or a substituted or unsubstituted 5-12-membered (preferably 5-7-membered) heteroarylene group having 1-3 heteroatoms, a substituted or unsubstituted 5-12-membered heterocyclylene group, or a substituted or unsubstituted 5-12-membered C3-C12 (preferably C5-C12) cycloalkylene group; or does not exist;
  • L1 and L2 are each independently selected from the group consisting of a chemical bond, a substituted or unsubstituted C1-C4 alkylene group, a substituted or unsubstituted C2-C4 alkenylene group, a substituted or unsubstituted C2-C4 alkynylene group, -S-, -O-, a substituted or unsubstituted -NH-, -S(O)-, -S(O) 2- , a substituted or unsubstituted -NHC(O)NH-, Substituted or unsubstituted Substituted or unsubstituted Substituted or unsubstituted Substituted or unsubstituted Substituted or unsubstituted
  • Each L 1a is independently a group selected from the group consisting of a chemical bond, a substituted or unsubstituted C 1 -C 7 alkylene group, a substituted or unsubstituted C 2 -C 4 alkenylene group, a substituted or unsubstituted C 2 -C 4 alkynylene group, -S-, -O-, a substituted or unsubstituted -NH-, -S(O)-, or -S(O) 2 -;
  • L 2a is selected from the group consisting of a substituted or unsubstituted C6-C12 arylene group, a substituted or unsubstituted 5-12-membered heteroarylene group having 1-3 heteroatoms, a substituted or unsubstituted C3-C8 cycloalkylene group, and a substituted or unsubstituted 3-10-membered heterocyclylene group having 1-3 heteroatoms;
  • L 3a is selected from the group consisting of substituted or unsubstituted C1-C10 alkyl, C1-C10 aryl, -CN, hydroxy, amino, carboxyl, -CO-NH-SO 2 -R g , -NH-SO 2 -R g , -SO 2 -NH-CO-R g , -OR g , -N(R g ) 2 -CO 2 R g , -CON(R g ) 2 , -CONHCOR g , NR g -CO-N(R g ) 2 , -NR g -SO 2 -N(R g ) 2 ;
  • r is 1, 2, 3, 4, 5, 6;
  • s 0, 1, and 2 respectively;
  • R d , Re and R g are each independently selected from the following group: H, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 6 -C 10 aryl; or R d and Re together form a substituted or unsubstituted 4-10 (preferably 5-10) membered heterocyclic group having 1-3 heteroatoms selected from N, S and O;
  • any of the heteroatoms is selected from the group consisting of B, P, N, S and O.
  • the Each is independently a divalent group formed by a ring selected from the group consisting of: Wherein, the bonding position of the ring can be N or C; more preferably, the Each independently
  • the It has the structure shown below:
  • X 6 , X 7 , X 8 , X 9 , X 10 and X 11 are each independently selected from the group consisting of N, CR;
  • the It has the structure shown below: Wherein, the bonding position of the ring can be N or C.
  • the A ring or D ring has a structure selected from the following group:
  • the E ring is selected from the following group: a substituted or unsubstituted benzene ring, a substituted or unsubstituted 5-6-membered heteroaromatic ring, a substituted or unsubstituted C 3 -C 6 carbon ring (including saturated or partially unsaturated), a substituted or unsubstituted 3-7-membered heterocycle (including saturated or partially unsaturated); any CH, CH 2 or NH position of the ring can lose a H atom to form a chemical bond.
  • the A ring or D ring has a structure selected from the following group:
  • Z 2 , Z 3 , and Z 4 are each independently selected from the group consisting of NH, N, CH, CH 2 , NO, SO, or SO 2 ;
  • X 1 and X 2 are each independently selected from the group consisting of CH, NH, N, NO, O, and S;
  • the D ring has a structure selected from the following group: (Bonding position can be N or C) Preferably, it is a structure selected from the following group:
  • the X5 is CR, and the R is Alternative.
  • the D ring is selected from the following group:
  • At least one R is
  • the compound has a structure selected from the following group:
  • the F ring is a group selected from the following group: substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl;
  • R 6 is selected from the following group: H, halogen, C1-C4 alkyl.
  • the R 4 has a structure selected from the following group:
  • the compound has a structure selected from the following group:
  • ring A is:
  • X 6 , X 7 , X 8 , X 9 , X 10 and X 11 are each independently selected from the group consisting of N, CR; preferably, X 10 is C(CHF 2 );
  • R6 is selected from the group consisting of H, halogen, unsubstituted or halogenated C1-C6 alkyl;
  • X1 , X2 , X3 , X4 , X5 and X6 are each independently selected from the group consisting of N, O, S, SO, SO2 , C(R) 2 , CHR, NR; wherein each R may be optionally Substitution; wherein r and s are 1; L 3 is a chemical bond, a substituted or unsubstituted C 1 -C 7 alkylene group; and E is a 5-7 membered nitrogen heterocyclic group;
  • R4 is selected from the following group:
  • the second aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising (1) a compound as described in the first aspect of the present invention or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof; and (2) a pharmaceutically acceptable carrier.
  • the third aspect of the present invention provides a use of the compound as described in the first aspect of the present invention or its stereoisomer or tautomer, or its pharmaceutically acceptable salt, hydrate or solvate, or the pharmaceutical composition as described in the third aspect of the present invention, characterized in that it is used to prepare a pharmaceutical composition for preventing and/or treating diseases related to the activity or expression of PD-1/PD-L1 (HBV, HCV, solid tumors, hematological tumors, etc.).
  • diseases related to the activity or expression of PD-1/PD-L1 HBV, HCV, solid tumors, hematological tumors, etc.
  • the fourth aspect of the present invention provides a PD-1/PD-L1 modulator, which comprises the compound described in the first aspect of the present invention, or its stereoisomer or tautomer, or its pharmaceutically acceptable salt, hydrate or solvate.
  • the pharmaceutical composition is used to treat a disease selected from the following group: cancer, infectious diseases, and autoimmune diseases.
  • the cancer is selected from the following group: pancreatic cancer, bladder cancer, colorectal cancer, breast cancer, prostate cancer, kidney cancer, hepatocellular carcinoma, lung cancer, ovarian cancer, cervical cancer, gastric cancer, esophageal cancer, melanoma, neuroendocrine cancer, central nervous system cancer, brain cancer, bone cancer, soft tissue sarcoma, non-small cell lung cancer, small cell lung cancer or colon cancer, skin cancer, lung cancer, urinary system tumors, blood tumors, gliomas, digestive system tumors, reproductive system tumors, lymphomas, nervous system tumors, brain tumors, head and neck cancer.
  • the cancer is selected from the following group: acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), myelodysplastic syndrome (MDS), myeloproliferative disease (MPD), chronic myeloid leukemia (CML), multiple myeloma (MM), non-Hodgkin's lymphoma (NHL), mantle cell lymphoma (MCL), follicular lymphoma, Waldestrom macroglobulinemia (WM), T cell lymphoma, B cell lymphoma or diffuse large B cell lymphoma (DLBCL).
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • MDS myelodysplastic syndrome
  • MDS myeloproliferative disease
  • the infectious disease is selected from bacterial infection and viral infection.
  • the autoimmune disease is selected from organ-specific autoimmune disease, systemic autoimmune disease Epidemic.
  • the organ-specific autoimmune diseases include chronic lymphocytic thyroiditis, hyperthyroidism, insulin-dependent diabetes mellitus, myasthenia gravis, ulcerative colitis, pernicious anemia with chronic atrophic gastritis, Goodpasture's syndrome, primary biliary cirrhosis, multiple sclerosis, and acute idiopathic polyneuritis.
  • systemic autoimmune diseases include rheumatoid arthritis, systemic lupus erythematosus, systemic vasculitis, scleroderma, pemphigus, dermatomyositis, mixed connective tissue disease, and autoimmune hemolytic anemia.
  • the pharmaceutical composition is also used to improve T cell function in patients with chronic hepatitis B (CHB).
  • CHB chronic hepatitis B
  • the inhibitor further comprises at least one therapeutic agent selected from the group consisting of nivolumab, pembrolizumab, atezolizumab, or ipilimumab.
  • the sixth aspect of the present invention provides a method for regulating PD-1/PD-L1 interaction in vivo, characterized in that it comprises the steps of: contacting the compound described in the first aspect of the present invention, or its stereoisomer or tautomer, or its pharmaceutically acceptable salt, hydrate or solvate with PD-L1 protein.
  • alkyl includes linear or branched alkyl groups.
  • C1 - C8 alkyl groups represent linear or branched alkyl groups having 1 to 8 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, and the like.
  • alkenyl includes straight or branched alkenyl groups.
  • C2 - C6 alkenyl refers to straight or branched alkenyl groups having 2 to 6 carbon atoms, such as vinyl, allyl, 1-propenyl, isopropenyl, 1-butenyl, 2-butenyl, or the like.
  • alkynyl includes straight or branched alkynyl groups.
  • C2 - C6 alkynyl refers to a straight or branched alkynyl group having 2 to 6 carbon atoms, such as ethynyl, propynyl, butynyl, or the like.
  • C 3 -C 10 cycloalkyl refers to a cycloalkyl group having 3 to 10 carbon atoms. It may be a monocyclic ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or the like. It may also be a bicyclic ring, such as a bridged ring or a spiro ring.
  • C 1 -C 8 alkylamino refers to an amine group substituted by a C 1 -C 8 alkyl group, which may be monosubstituted or disubstituted; for example, methylamino, ethylamino, propylamino, isopropylamino, butylamino, isobutylamino, tert-butylamino, dimethylamino, diethylamino, dipropylamino, diisopropylamino, dibutylamino, diisobutylamino, di-tert-butylamino and the like.
  • C 1 -C 8 alkoxy refers to a straight or branched alkoxy group having 1 to 8 carbon atoms; for example, methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tert-butoxy and the like.
  • the term "3-10 membered heterocycloalkyl having 1-3 heteroatoms selected from the group consisting of N, S and O” refers to a saturated or partially saturated cyclic group having 3-10 atoms, wherein 1-3 atoms are heteroatoms selected from the group consisting of N, S and O. It may be a monocyclic or bicyclic form, such as a bridged ring or a spirocyclic form. Specific examples may include oxetane, azetidine, tetrahydro-2H-pyranyl, piperidinyl, tetrahydrofuranyl, morpholinyl and pyrrolidinyl, etc.
  • C 6 -C 10 aryl group refers to an aryl group having 6 to 10 carbon atoms, for example, phenyl or naphthyl and the like.
  • the term "5-10 membered heteroaryl having 1-3 heteroatoms selected from the group consisting of N, S and O” refers to a cyclic aromatic group having 5-10 atoms, wherein 1-3 atoms are heteroatoms selected from the group consisting of N, S and O.
  • the ring may also be a condensed ring.
  • Specific examples may include pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3)-triazolyl and (1,2,4)-triazolyl, tetrazolyl, furanyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, and the like.
  • the groups described in the present invention may be substituted by substituents selected from the following groups: halogen, nitrile, nitro, hydroxyl, amino, C 1 -C 6 alkyl-amine, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxy, halogenated C 1 -C 6 alkyl, halogenated C 2 -C 6 alkenyl, halogenated C 2 -C 6 alkynyl, halogenated C 1 -C 6 alkoxy, allyl, benzyl, C 6 -C 12 aryl, C 1 -C 6 alkoxy-C 1 -C 6 alkyl, C 1 -C 6 alkoxy-carbonyl, phenoxycarbonyl, C 2 -C 6 alkynyl-carbonyl, C 2 -C 6
  • halogen or halogen atom refers to F, Cl, Br, and I. More preferably, the halogen or halogen atom is selected from F, Cl and Br. "Halogenated” means substituted with an atom selected from F, Cl, Br, and I.
  • the structural formulas described in the present invention are intended to include all isomeric forms (such as enantiomers, diastereomers and geometric isomers (or conformational isomers)): for example, R, S configurations containing asymmetric centers, (Z), (E) isomers of double bonds, etc. Therefore, single stereochemical isomers of the compounds of the present invention or mixtures of their enantiomers, diastereomers or geometric isomers (or conformational isomers) are all within the scope of the present invention.
  • tautomer means that structural isomers with different energies can interconvert across a low energy barrier.
  • proton tautomers i.e., prototropic
  • Valence tautomers include interconversion via reorganization of some bonding electrons.
  • solvate refers to a complex in which the compound of the present invention is coordinated with solvent molecules to form a specific ratio.
  • hydrate refers to a complex formed by coordination of a compound of the present invention with water.
  • the compounds described in the present invention include possible other derivative compounds that can be converted into the compounds of the present invention in vivo, such as forming esters at any site (such as carboxyl, hydroxyl, thiol and other functional groups in the molecule) or
  • compounds of the present invention refers to compounds of Formula I, and also includes various crystalline forms, pharmaceutically acceptable salts or esters, hydrates or solvates of the compounds of Formula I.
  • Preferred compounds of the present invention include compounds 1-143 (including various R-configuration and/or S-configuration stereoisomers of each compound, and/or E-/Z-cis-trans isomers).
  • the pharmaceutically acceptable salts include salts formed by combining with inorganic acids, organic acids, alkali metal ions, alkaline earth metal ions or organic bases that can provide physiologically acceptable cations, and ammonium salts.
  • the inorganic acid is selected from hydrochloric acid, hydrobromic acid, phosphoric acid or sulfuric acid;
  • the organic acid is selected from methanesulfonic acid, p-toluenesulfonic acid, trifluoroacetic acid, citric acid, maleic tartaric acid, fumaric acid, citric acid or lactic acid;
  • the alkali metal ion is selected from lithium ion, sodium ion, potassium ion;
  • the alkaline earth metal ion is selected from calcium ion and magnesium ion;
  • the organic base capable of providing physiologically acceptable cations is selected from methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris(2-hydroxyethyl)amine.
  • the starting materials and intermediates in the preparation method of the present invention are easily available, and each step of the reaction can be easily synthesized according to the reported literature or by conventional methods in organic synthesis for those skilled in the art. It can exist in the form of solvates or unsolvates. Crystallization using different solvents may result in different solvates.
  • the compound of general formula I of the present invention can be obtained by the following synthetic route.
  • Compound 1-1 is used as a raw material to form chiral alcohol 1-2 under the action of a chiral auxiliary (such as R/S-CBS) and a reducing agent (such as borane);
  • a chiral auxiliary such as R/S-CBS
  • a reducing agent such as borane
  • Compound 2-5 is used as a raw material and reacted with a nitrating agent (such as concentrated sulfuric acid/NaNO3, concentrated sulfuric acid/fuming nitric acid, etc.) to form intermediate 2-6;
  • a nitrating agent such as concentrated sulfuric acid/NaNO3, concentrated sulfuric acid/fuming nitric acid, etc.
  • the R 4 is: for: a precursor form containing a carbonyl group or a carbonyl group on the D ring; t is selected from 0, 1, 2, 3; X 1 , X 2 , X 3 , X 4 , X 5 are each independently selected from C, N, O, S, P, B.
  • the starting materials and intermediates in the above reactions are easily available, and each step of the reaction can be easily synthesized according to the reported literature or by conventional methods in organic synthesis for those skilled in the art.
  • the compound described in general formula I may exist in the form of a solvate or an unsolvate, and different solvates may be obtained by crystallization using different solvents.
  • compositions and methods of administration are provided.
  • the compounds of the present invention have excellent PD-1/PD-L1 interaction regulating activity
  • the compounds of the present invention and their various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates, and pharmaceutical compositions containing the compounds of the present invention as the main active ingredient can be used to prevent and/or treat (stabilize, alleviate or cure) PD-1/PD-L1 interaction.
  • Interaction-related diseases e.g., cancer, infectious diseases, autoimmune diseases).
  • the pharmaceutical composition of the present invention comprises a safe and effective amount of the compound of the present invention and a pharmaceutically acceptable excipient or carrier.
  • safe and effective amount means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical composition contains 1-2000 mg of the compound of the present invention per dose, and more preferably, contains 10-200 mg of the compound of the present invention per dose.
  • the "one dose” is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” refers to: one or more compatible solid or liquid fillers or gel substances, which are suitable for human use and must have sufficient purity and sufficiently low toxicity. "Compatibility” here means that the components in the composition can be mixed with the compounds of the present invention and with each other without significantly reducing the efficacy of the compounds.
  • Some examples of pharmaceutically acceptable carriers include cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers (such as Tween ), wetting agents (such as sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • cellulose and its derivatives such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.
  • gelatin such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.
  • compositions of the present invention include (but are not limited to): oral administration, parenteral administration (intravenous administration, intramuscular administration or subcutaneous administration).
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrators, for example, agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) solubilizers, for example, paraffin; (f) absorption accelerators, for example, quaternary ammonium compounds; (g) wetting agents, for example,
  • Solid dosage forms such as tablets, pills, capsules, pills and granules can be prepared using coatings and shell materials, such as enteric coatings and other materials known in the art. They may contain opacifiers, and the release of the active compound or compounds in such compositions can be delayed in a certain part of the digestive tract. Examples of embedding components that can be used are polymeric substances and waxes. If necessary, the active compound can also be formed into microcapsules with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • the liquid dosage form may contain an inert diluent conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butylene glycol, dimethylformamide and oils, in particular cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances.
  • an inert diluent conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butylene glycol, dimethylformamide and oils, in particular cottons
  • composition may also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions in addition to the active compounds, may contain suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methanol and agar, or mixtures of these substances, and the like.
  • suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methanol and agar, or mixtures of these substances, and the like.
  • compositions for parenteral injection may include physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • the compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds (eg, other anticancer agents).
  • other pharmaceutically acceptable compounds eg, other anticancer agents.
  • the pharmaceutical composition When administered in combination, the pharmaceutical composition also includes one or more (2, 3, 4, or more) other pharmaceutically acceptable compounds.
  • One or more (2, 3, 4, or more) of the other pharmaceutically acceptable compounds can be used simultaneously, separately or sequentially with the compounds of the present invention to prevent and/or treat diseases related to PD-1/PD-L1 interaction.
  • a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, wherein the dosage during administration is a pharmaceutically effective dosage, and for a person weighing 60 kg, the daily dosage is usually 1 to 2000 mg, preferably 20 to 500 mg.
  • the specific dosage should also take into account factors such as the route of administration and the health status of the patient, which are all within the skill of a skilled physician.
  • the compounds of the present invention have high regulatory activity on the PD-1/PD-L1 interaction, have strong binding ability with the PD-L1 protein, and have the ability to relieve the IFN ⁇ inhibition of PD-L1.
  • the compounds of the present invention have better solubility and very low toxicity to normal cells, and thus can be applied to therapeutic subjects within a wider dosage range.
  • the compounds of the present invention have better solubility and thus have good drugability. Compared with the existing compounds, the compounds of the present invention show good bioavailability in in vivo experiments. In addition, compared with the existing compounds, the compounds of the present invention are very easy to prepare into pharmaceutically acceptable salts, thus facilitating the further formation of preparations.
  • Step 1-2
  • Step 2-1
  • Step 2-2
  • Step 3-2
  • Step 5-2
  • Step 6-2
  • Step 3-2
  • 134-02 (800 mg) was dissolved in DCM (10 mL), trifluoroacetic acid (2 mL) was added, and the mixture was stirred at room temperature for 3 hours. The reaction was monitored by TLC, and the solvent was concentrated under reduced pressure to obtain 134-03.
  • Example A PD-1/PD-L1 Homogeneous Time-Resolved Fluorescence (HTRF) Binding Assay
  • the assay was performed in a standard black 384-well polystyrene plate in a final volume of 20 ⁇ L.
  • the inhibitor was first serially diluted in DMSO and added to the wells before the other reaction components were added.
  • the final concentration of DMSO in the assay was 1%.
  • the assay was performed in PBS buffer (pH 7.4) containing 0.05% Tween-20 and 0.1% BSA at 25°C.
  • Recombinant human PD-L1 protein (19-238) with a His tag at the C-terminus was purchased from AcroBiosystems (PD1-H5229).
  • Recombinant human PD-1 protein (25-167) with an Fc tag at the C-terminus was also purchased from AcroBiosystems (PD1-H5257).
  • PD-L1 and PD-1 proteins were diluted in assay buffer and 0.1 ⁇ l of the solution was extracted and added to the wells. The plate was centrifuged and the proteins were preincubated with the inhibitors for 40 minutes. After incubation, 0.1 ⁇ l of HTRF detection buffer containing europium-blocked anti-human IgG (PerkinElmer-AD0212) Fc-specific and anti-His -Allophycocyanin (APC, PerkinElmer-AD0059H) conjugated antibody. After centrifugation, the well plate was incubated at 25 ° C for 60 minutes. The data (665nm/620nm ratio) was read in a PHERAstar FS plate reader.
  • the final concentrations in the assay were ⁇ 3nM PD1, 10nM PD-L1, 1nM europium anti-human IgG, and 20nM anti-His-allophycocyanin.
  • the IC50 values of the inhibitors were obtained by fitting the activity data using GraphPad Prism5.0 software.
  • the data of the example compounds obtained using the PD-1/PD-L1 homogeneous time-resolved fluorescence (HTRF) binding assay described in Example A are provided in Table 1.
  • Example B Pharmacokinetics in ICR mice after single intravenous and oral administration
  • Preparation method for intravenous administration accurately weigh about 2.5 mg of the compound into a glass bottle, add a certain volume of 20% HP- ⁇ -CD aqueous solution, vortex and oscillate to mix evenly, adjust the pH to about 3.0 (if necessary), ultrasonically dissolve the solid fully, filter, and obtain a group A intravenous administration preparation solution with a concentration of 1 mg/mL.
  • Preparation method for oral administration accurately weigh about 10 mg of the compound into a glass bottle, add a certain volume of 20% HP- ⁇ -CD aqueous solution, vortex and oscillate to mix evenly, adjust the pH to about 3.0 (if necessary), and sonicate to fully dissolve the solid to obtain a concentration of 2 mg/mL of Group B oral administration preparation solution.
  • the experimental animals were kept in an animal room equipped with an air-conditioning system, which was well ventilated and maintained at a room temperature of 20 ⁇ 26°C, humidity maintained in the range of 40% ⁇ 70%. Artificial lighting was used in the animal room, with 12 hours of light and dark (except for experimental operation, cleaning, etc., when working lighting was turned on), and experimental animals were free to eat and drink. Animals were fed normally for at least 5 days after purchase (for acute termination experiments lasting no more than 48 hours, mice were fed for at least 3 days). Animals with good physical signs were selected for this experiment after veterinary examination, and each mouse was marked with a tail number.
  • the animals in this experiment were given the prescribed doses by intravenous injection or oral gavage.
  • Sample collection About 100 ⁇ L of blood was collected from the eye socket at 0.25 h, 0.5 h, 1 h, 2 h, 4 h, 6 h, 8 h and 24 h after administration. The collected whole blood was placed in an EDTA-K 2 anticoagulant tube, inverted several times to mix thoroughly, stored on wet ice, and centrifuged (1500-1600 g) for 10 min within 30 min to separate the plasma. The obtained plasma samples were stored at -40 to -20 °C for biological sample analysis.
  • mice were euthanized by carbon dioxide asphyxiation or cervical dislocation.
  • the samples are returned to the biological sample room, which will transfer the samples to a -40 to -20°C environment for sealing according to the SOP.

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明提供了一种新型联芳环化合物作为免疫调节剂的制备和应用,具体地,本发明提供了一种如下式(I)所示的化合物,或其光学异构体、水合物、溶剂合物,或其药学上可接受的盐;其中,各基团的定义如说明书中所述。所述的式I化合物可以用于治疗与PD-1/PD-L1信号通路有关的疾病。

Description

新型联芳环化合物作为免疫调节剂的制备和应用 技术领域
本发明涉及小分子药物领域,具体地,本发明提供了一种可以用于治疗与PD-1/PD-L1信号通路有关的疾病的小分子化合物。
发明背景
免疫系统对控制及根治很多疾病起着至关重要的作用,像各种癌症、病毒引起的疾病等。但癌症细胞经常能通过一些途径躲避或抑制免疫系统,从而快速繁殖。其中一个方式就是改变免疫细胞上表达的激活和抑制分子。阻断抑制免疫检查点,像PD-1,证明是一个非常有效的抑制癌细胞的方法。
PD-1是程序性细胞死亡蛋白-1,也称之为CD279。它主要在激活的T细胞和B细胞中表达,功能是抑制细胞的激活,这是免疫系统的一种正常的自稳机制,因为过度的T/B细胞激活会引起自身免疫病,所以PD-1是我们人体的一道保护墙。PD-1是由268个氨基酸组成的I型跨膜糖蛋白,它的结构主要包括外免疫球蛋白可变区、疏水跨膜区以及胞内区。胞内区包含两个磷酸化位点,分别位于免疫受体酷氨酸抑制基序和免疫受体酷氨酸转换基序,这也证明PD-1能够反向调节T细胞受体介导的信号。PD-1有两个配体,PD-L1和PD-L2,它们在表达方式上不同。PD-L1在多种肿瘤细胞中均有上调表达,它与T细胞上的PD-1结合,抑制T细胞增殖和活化,使T细胞处于失活状态,最终诱导免疫逃逸。
PD-1/PD-L1发挥着反向免疫调节作用。当PD-1与PD-L1结合后,可致使T细胞的免疫受体酷氨酸转换基序结构域的酪氨酸多磷酸化,磷酸化的酪氨酸可结合磷酸酶蛋白酪氨酸酶2和蛋白酪氨酸酶1。这样即可阻碍细胞外信号调节激酶的活化,还可阻断磷脂肌醇3-激酶(PI3K)和丝氨酸-苏氨蛋白激酶(Akt)的激活,从而抑制T淋巴细胞增殖和相关细胞因子的分沁。PD-1/PD-L1信号抑制T细胞活化和增殖的同时,还可使细胞因子白细胞介素2、干扰素γ和IL-10的分泌。此外,PD-1/PD-L1信号对B细胞也有类似的免疫功能,当PD-1与B细胞抗原受体结合后,PD-1细胞质区与含有蛋白酪氨酸酶2结合位点的酪氨酸酶发生作用,从而阻碍B细胞的活化。
基于PD-1/PD-L1的免疫疗法是新一代备受关注的免疫疗法。最近几年,一系列令人惊喜的研究结果证实PD-1/PD-L1抑制剂对多种肿瘤具有强大的抗肿瘤活性。目前已经上市的PD-1/PD-L1抗体调节剂有BMS的Ninolumab、Merck的Lambrolizumab以及Roche的Atezolizumab。除此之外还有很多在研的PD-1/PD-L1抗体调节剂,包括Cure Tech的Pidilizumab、GSK的AMP-224和AstraZeneca的MEDI-4736。
虽然肿瘤免疫治疗被认为是靶向治疗后癌症治疗的新一代革命。但是目前上市和在研的PD-1单抗药有其自身的缺陷,包括只能注射给药,不能口服,在体内不稳定,易被蛋白酶分解,易产生免疫交叉反应,纯化比较困难且生产成本高等。所以PD-1/PD-L1相互作用的小分子调节剂是肿瘤免疫治疗的更好选择。
综上所述,本领域迫切需要开发新型PD-1/PD-L1相互作用的小分子调节剂。
发明内容
本发明的目的是提供一种新型PD-1/PD-L1相互作用的小分子调节剂。
本发明的第一方面,一种如下式I所示的化合物,或其光学异构体、水合物、溶剂合物,或其药学上可接受的盐:
其中,m为0、1、2、3,4或5;
n、v和p各自独立地为0、1、2、3或4;
各自独立地选自下组:取代或未取代的5-12元杂芳基、取代或未取代的C6-C10芳基、取代或未取代的5-12元杂环基、取代或未取代的5-12元的C3-C12(优选为C5-C12)环基,其中,所述的杂环基具有1-3个杂原子;或D为
其中,X1、X2、X3、X4、X5和X6各自独立地选自下组:N、O、S、SO、SO2、C(R)2、CHR、NR;其中,各个R可以任选地被替代;
Y1、Y2、Y3、Y4、Y5和Y6各自独立地选自下组:N、CH、C;
其中,X3、X4、X5的碳原子上的氢(如存在的情况下)均可各自独立的被氘替换;
L3选自下组:化学键、取代或未取代的C1-C7亚烷基、取代或未取代的C2-C4亚烯基、取代或未取代的C2-C4亚炔基、-S-、-O-、取代或未取代的-NH-、-S(O)-、或-S(O)2-;
E选自下组:取代或未取代的C6-C12亚芳基、取代或未取代的具有1-3个杂原子的5-12元亚杂芳基、取代或未取代的C3-C8亚环烷基、取代或未取代的具有1-3个杂原子的3-10元亚杂环基;
r为1、2、3、4、5、6;
s为0、1、2;
a为1、2、3、4、5或6;
R4选自下组:
R5选自下组:H、-CN、三氟甲基、-CHF2、-OCF3、-OCHF2、磺酰氨基、硝基、羟基、卤素、取代或未取代的C1-C6烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、-CN、羟基、
各自独立地选自下组:取代或未取代的C6-C10亚芳基、或取代或未取代的具有1-3个杂原子的5-12元(优选5-7元)亚杂芳基、取代或未取代的5-12元亚杂环基、取代或未取代的5-12元的C3-C12(优选为C5-C12)亚环烷基;或不存在;
L1和L2各自独立地选自下组:化学键、取代或未取代的C1-C4亚烷基、取代或未取代的C2-C4亚烯基、取代或未取代的C2-C4亚炔基、-S-、-O-、取代或未取代的-NH-、-S(O)-、-S(O)2-、取代或未取代的-NHC(O)NH-、取代或未取代的取代或未取代的取代或未取代的
表示单键或双键;
R、R1、R2和R3各自独立地选自下组:H、-CN、三氟甲基、-CHF2、-OCF3、-OCHF2、磺酰氨基、硝基、羟基、卤素、-S-R8、-S(O)-R8、-S(O)2-R8、取代或未取代的C1-C10烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、-CN、羟基、NRdRe(例如氨基)、取代或未取代的C1-C6胺基、取代或未取代的-(C1-C6亚烷基)-NH-(C1-C6亚烷基)、羧基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个杂原子的5-12元杂芳基、取代或未取代的具有1-4个杂原子的3-12元杂环基,其中,取代或未取代的C1-C10烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、羟基、NRdRe(例如氨基)、取代或未取代的C1-C6胺基、取代或未取代的-(C1-C6亚烷基)-NH-(C1-C6亚烷基)、羧基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个杂原子的5-12元杂芳基、取代或未取代的具有1-4个杂原子的3-12元杂环基的碳原子上的氢均可各自独立的被氘替换;取代或未取代的取代或未取代的取代或未 取代的其中,Rb和Rc各自独立地选自下组:H、取代或取代的C1-C8烷基;或所述的Rb和Rc与相邻的N原子共同构成取代或未取代的具有1-3个选自N、S和O的杂原子的3-10元杂环基,或所述的Rb和Rc与相邻的N原子共同构成取代或未取代的4-10元内酰胺,所述取代基包括但不限于:羟基、羧基、巯基、氨基、F、Cl,其中,Rb、Rc和Rd的碳原子上的氢均可各自独立地被氘替换;或-(L1a)r-(L2a)s-(L3a)s-;-C0-8-O-R8,-C0-8-C(O)OR8,-C0-8-OC(O)OR8,-C0-8-NR8R9,-C0-8-N(R8)C(O)R9,-C0-8-C(O)NR8R9
R8和R9各自独立地选自下组:H、羟基,取代或未取代的C1-C10烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、-CN、羟基、NRdRe(例如氨基)、取代或未取代的C1-C6胺基、取代或未取代的-(C1-C6亚烷基)-NH-(C1-C6亚烷基)、羧基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个杂原子的5-12元杂芳基、取代或未取代的具有1-4个杂原子的3-12元杂环基,取代或未取代的取代或未取代的取代或未取代的或-(L1a)r-(L2a)s-(L3a)s-;
各个L1a各自独立地为选自下组的基团;化学键、取代或未取代的C1-C7亚烷基、取代或未取代的C2-C4亚烯基、取代或未取代的C2-C4亚炔基、-S-、-O-、取代或未取代的-NH-、-S(O)-、或-S(O)2-;
L2a选自下组:取代或未取代的C6-C12亚芳基、取代或未取代的具有1-3个杂原子的5-12元亚杂芳基、取代或未取代的C3-C8亚环烷基、取代或未取代的具有1-3个杂原子的3-10元亚杂环基;
L3a选自下组:取代或未取代的C1-C10烷基、C1-C10芳基、-CN、羟基、氨基、羧基、-CO-NH-SO2-Rg、-NH-SO2-Rg、-SO2-NH-CO-Rg、-ORg、-N(Rg)2-CO2Rg、-CON(Rg)2、-CONHCORg、NRg-CO-N(Rg)2、-NRg-SO2-N(Rg)2
r为1、2、3、4、5、6;
s分别为0、1、2;
Rd、Re和Rg各自独立地选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C10环烷基、取代或未取代的C6-C10芳基;或Rd和Re共同形成取代或未取代的具有1-3个选自N、S和O的杂原子的4-10(优选为5-10)元杂环基;
所述的Rf选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C6-C10芳基、取代或未取代的5-10元杂芳基、氰基、-C(=O)-NRdRe、-C(=O)-取代或未取代的C1-C6烷氧基、-C(=O)-取代或未取代的C1-C6烷基、-C(=O)-取代或未取代的C3-C10环烷基、-C(=O)-取代或未取代的C2-C6烯基、-C(=O)-取代或未取代的C2-C6炔基;
除非特别说明,所述的“取代”是指被选自下组的一个或多个(例如2个、3个、4个等)取代基所取代:卤素(包括-F、-Cl、-Br、-I)、-CH2Cl、-CHCl2、-CCl3、-CH2F、-CHF2、-CF3、氧代(=O)、-CN、羟基、氨基、C1-C6烷胺基、羧基、-NHAc、 未取代或被一个或多个取代基取代的选自下组的基团:C1-C6烷基、C1-C6烷氧基、C6-C10芳基、C3-C8环烷基、卤代的C6-C10芳基、具有1-3个选自N、S和O的杂原子的5-10元杂芳基、具有1-3个选自N、S和O的杂原子的5-10元杂环基;所述的取代基选自下组:卤素、羟基、羧基、氧基、氰基、C1-C6烷氧基、C1-C6烷胺基;
上述各式中,任一所述杂原子选自下组:B、P、N、S和O。
在另一优选例中,所述的各自独立地为选自下组的环形成的二价基团: 其中,所述的环的成键位置可以为N或C;更佳地,所述的各自独立地为
在另一优选例中,所述的具有如下式所示的结构:
其中,
X6、X7、X8、X9、X10和X11各自独立地选自下组:N、CR;
R6选自下组:H、卤素、取代或未取代的C1-C6烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、-CN、羟基、NRdRe(例如氨基)、取代或未取代的C1-C6胺基、取代或未取代的-(C1-C6亚烷基)-NH-(C1-C6亚烷基)、羧基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个杂原子的5-12元杂芳基、取代或未取代的具有1-4个杂原子的5-12元杂环基,取代或未取代的取代或未取代的取代或未取代的或-(L1a)r-(L2a)s-(L3a)s-,-C0-8-O-R8,-C0-8-C(O)OR8,-C0- 8-OC(O)OR8,-C0-8-NR8R9,-C0-8-N(R8)C(O)R9,-C0-8-C(O)NR8R9
在另一优选例中,所述的具有如下式所示的结构: 其中,所述的环的成键位置可以为N或C。
在另一优选例中,所述的A环或D环具有选自下组的结构:
其中,E环选自下组:取代或未取代的苯环、取代或未取代的5-6元的杂芳环、取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-7元的杂环(包括饱和或部分不饱和的情况);所述环的任意CH、CH2或NH位置可以失去一个H原子从而形成化学键。
在另一优选例中,所述的A环或D环具有选自下组的结构:
其中,
Z1选自下组:O、S、NRf、N-O-Rf;其中,所述的Rf选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C6-C10芳基、取代或未取代的C6-C10杂芳基、氰基、-C(=O)-NRdRe、-C(=O)-取代或未取代的C1-C6烷氧基、-C(=O)-取代或未取代的C1-C6烷基、-C(=O)-取代或未取代的C3-C10环烷基、-C(=O)-取代或未取代的C2-C6烯基、-C(=O)-取代 或未取代的C2-C6炔基;
Z2、Z3、Z4各自独立地选自下组:NH、N、CH、CH2、N-O、SO或SO2
X1、X2各自独立地选自下组:CH、NH、N、N-O、O、S;
为单键或双键;
为芳香性或非芳香性片段。
在另一优选例中,所述的D环具有选自下组的结构: (成键位置可以为N或C) 较佳地,为选自下组的结构:
在另一优选例中,所述的X5为CR,且所述的R被替代。
在另一优选例中,所述的D环选自下组:
其中,至少一个R为
在另一优选例中,所述的化合物具有选自下组的结构:
其中,F环为选自下组的基团:取代或未取代的苯基、取代或未取代的吡啶基;R6选自下组:H、卤素、C1-C4烷基。
在另一优选例中,所述的R4具有选自下组的结构:
在另一优选例中,所述的化合物具有选自下组的结构:
其中,所述的环A为:
其中,
X6、X7、X8、X9、X10和X11各自独立地选自下组:N、CR;较佳地,X10为C(CHF2);
R6选自下组:H、卤素、未取代或卤代的C1-C6烷基;
D为其中,X1、X2、X3、X4、X5和X6各自独立地选自下组:N、O、S、SO、SO2、C(R)2、CHR、NR;其中,各个R可以任选地被替代;其中,r和s为1;L3为化学键、取代或未取代的C1-C7亚烷基;且E为5-7元的亚氮杂环基;
R4选自下组:
本发明的第二方面,提供了一种药物组合物,包含(1)如本发明第一方面所述的化合物或其立体异构体或互变异构体,或其药学上可接受的盐、水合物或溶剂化物;(2)药学上可接受的载体。
本发明的第三方面,提供了一种如本发明第以方面所述的化合物或其立体异构体或互变异构体,或其药学上可接受的盐、水合物或溶剂化物或如本发明第三方面所述的药物组合物的用途,其特征在于,用于制备预防和/或治疗与PD-1/PD-L1的活性或表达量相关的疾病(HBV、HCV、实体瘤、血液瘤等)的药物组合物。
本发明的第四方面,提供了一种PD-1/PD-L1调节剂,所述调节剂包含本发明第一方面所述的化合物、或其立体异构体或互变异构体、或其药学上可接受的盐、水合物或溶剂化物。
在另一优选例中,所述的药物组合物被用于治疗选自下组的疾病:癌症、感染性疾病、自身免疫性疾病。
在另一优选例中,所述的癌症选自下组:胰腺癌,膀胱癌,结肠直肠癌,乳腺癌,前列腺癌,肾癌,肝细胞癌,肺癌,卵巢癌,宫颈癌,胃癌,食道癌,黑色素瘤,神经内分泌癌,中枢神经系统癌,脑癌,骨癌,软组织肉瘤,非小细胞肺癌癌症,小细胞肺癌或结肠癌、皮肤癌、肺癌、泌尿系肿瘤、血液肿瘤、胶质瘤、消化系统肿瘤、生殖系统肿瘤、淋巴瘤、神经系统肿瘤、脑瘤、头颈癌。
在另一优选例中,所述癌症选自下组:急性淋巴细胞白血病(ALL),急性髓性白血病(AML),慢性淋巴细胞白血病(CLL),小淋巴细胞性淋巴瘤(SLL),骨髓增生异常综合征(MDS),骨髓增生性疾病(MPD),慢性粒细胞白血病(CML),多发性骨髓瘤(MM),非霍奇金淋巴瘤(NHL),套细胞淋巴瘤(MCL),滤泡性淋巴瘤,Waldestrom巨球蛋白血症(WM),T细胞淋巴瘤,B细胞淋巴瘤或弥漫性大B细胞淋巴瘤(DLBCL)。
在另一优选例中,所述的感染性疾病选自细菌感染、病毒感染。
在另一优选例中,所述的自身免疫性疾病选自器官特异性自身免疫病、系统性自身免 疫病。
在另一优选例中,所述的器官特异性自身免疫病包括慢性淋巴细胞性甲状腺炎、甲状腺功能亢进、胰岛素依赖型糖尿病、重症肌无力、溃疡性结肠炎、恶性贫血伴慢性萎缩性胃炎、肺出血肾炎综合症、原发性胆汁性肝硬化、多发性脑脊髓硬化症、急性特发性多神经炎。
在另一优选例中,所述的系统性自身免疫病包括类风湿关节炎、系统性红斑狼疮、系统性血管炎、硬皮病、天疱疮、皮肌炎、混合性结缔组织病、自身免疫性溶血性贫血。
在另一优选例中,所述的药物组合物还用于改善慢性乙型肝炎(CHB)患者T细胞功能。
在另一优选例中,所述的抑制剂还包括至少一种选自下组的治疗剂:纳武单抗,派姆单抗,atezolizumab,或伊匹单抗。
本发明的第六方面,提供了一种体内调节PD-1/PD-L1相互作用的方法,其特征在于,包括步骤:将本发明第一方面所述的化合物、或其立体异构体或互变异构体、或其药学上可接受的盐、水合物或溶剂化物与PD-L1蛋白接触。
具体实施方式
本发明人经过广泛而深入的研究,发现了一类具有优异的调节效果的PD-1/PD-L1相互作用调节剂。在此基础上,发明人完成了本发明。
定义
如本文所用,术语“烷基”包括直链或支链的烷基。例如C1-C8烷基表示具有1-8个碳原子的直链或支链的烷基,例如甲基、乙基、丙基、异丙基、丁基、异丁基、叔丁基等。
如本文所用,术语“烯基”包括直链或支链的烯基。例如C2-C6烯基指具有2-6个碳原子的直链或支链的烯基,例如乙烯基、烯丙基、1-丙烯基、异丙烯基、1-丁烯基、2-丁烯基、或类似基团。
如本文所用,术语“炔基”包括直链或支链的炔基。例如C2-C6炔基是指具有2-6个碳原子的直链或支链的炔基,例如乙炔基、丙炔基、丁炔基、或类似基团。
如本文所用,术语“C3-C10环烷基”指具有3-10个碳原子的环烷基。其可以是单环,例如环丙基、环丁基、环戊基、环己基、或类似基团。也可以是双环形式,例如桥环或螺环形式。
如本文所用,术语“C1-C8烷胺基”是指被C1-C8烷基所取代的胺基,可以是单取代或双取代的;例如,甲胺基、乙胺基、丙胺基、异丙胺基、丁胺基、异丁胺基、叔丁胺基、二甲胺基、二乙胺基、二丙胺基、二异丙胺基、二丁胺基、二异丁胺基、二叔丁胺基等。
如本文所用,术语“C1-C8烷氧基”是指具有1-8个碳原子的直链或支链的烷氧基;例如,甲氧基、乙氧基、丙氧基、异丙氧基、丁氧基、异丁氧基、叔丁氧基等。
如本文所用,术语“具有1-3个选自下组N、S和O的杂原子的3-10元杂环烷基”是指具有3-10个原子的且其中1-3个原子为选自下组N、S和O的杂原子的饱和或部分饱和的环状基团。其可以是单环,也可以是双环形式,例如桥环或螺环形式。具体的实例可以为氧杂环丁烷、氮杂环丁烷、四氢-2H-吡喃基、哌啶基、四氢呋喃基、吗啉基和吡咯烷基等。
如本文所用,术语“C6-C10芳基”是指具有6-10个碳原子的芳基,例如,苯基或萘基等类似基团。
如本文所用,术语“具有1-3个选自下组N、S和O的杂原子的5-10元杂芳基”指具有5-10个原子的且其中1-3个原子为选自下组N、S和O的杂原子的环状芳香基团。其可以是单 环,也可以是稠环形式。具体的实例可以为吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、吡咯基、吡唑基、咪唑基、(1,2,3)-三唑基以及(1,2,4)-三唑基、四唑基、呋喃基、噻吩基、异恶唑基、噻唑基、恶唑基等。
本发明所述的基团除非特别说明是“取代的或未取代的”,否则本发明的基团均可被选自下组的取代基所取代:卤素、腈基、硝基、羟基、氨基、C1-C6烷基-胺基、C1-C6烷基、C2-C6烯基、C2-C6炔基、C1-C6烷氧基、卤代C1-C6烷基、卤代C2-C6烯基、卤代C2-C6炔基、卤代C1-C6烷氧基、烯丙基、苄基、C6-C12芳基、C1-C6烷氧基-C1-C6烷基、C1-C6烷氧基-羰基、苯氧羰基、C2-C6炔基-羰基、C2-C6烯基-羰基、C3-C6环烷基-羰基、C1-C6烷基-磺酰基等。
如本文所用,“卤素”或“卤原子”指F、Cl、Br、和I。更佳地,卤素或卤原子选自F、Cl和Br。“卤代的”是指被选自F、Cl、Br、和I的原子所取代。
除非特别说明,本发明所描述的结构式意在包括所有的同分异构形式(如对映异构,非对映异构和几何异构体(或构象异构体)):例如含有不对称中心的R、S构型,双键的(Z)、(E)异构体等。因此,本发明化合物的单个立体化学异构体或其对映异构体、非对映异构体或几何异构体(或构象异构体)的混合物都属于本发明的范围。
除非特别说明,本发明所描述的结构式意在包括所有可能的氘代衍生物(即,分子中的一个或多个氢原子被D取代)。
如本文所用,术语“互变异构体”表示具有不同能量的结构同分异构体可以超过低能垒,从而互相转化。比如,质子互变异构体(即质子移变)包括通过质子迁移进行互变,如1H-吲唑与2H-吲唑。化合价互变异构体包括通过一些成键电子重组而进行互变。
如本文所用,术语“溶剂合物”是指本发明化合物与溶剂分子配位形成特定比例的配合物。
如本文所用,术语“水合物”是指本发明化合物与水进行配位形成的配合物。
应理解,在本发明中所述的化合物已在包括可能的,在生物体内可以转化为本发明化合物的其他衍生化合物,例如在任意位点形成酯(例如分子内的羧基、羟基、巯基等官能团)或
活性成分
如本文所用,“本发明化合物”指式I所示的化合物,并且还包括及式I化合物的各种晶型形式、药学上可接受的盐或酯、水合物或溶剂合物。
优选的本发明化合物包括化合物1-143(包括各化合物的各类R构型和/或S构型的立体异构体,和/或E-/Z-的顺反异构体)。









在另一优选例中,所述的药用盐包括与无机酸、有机酸、碱金属离子、碱土金属离子或能提供生理上可接受的阳离子的有机碱结合形成的盐以及铵盐。
在另一优选例中,所述的无机酸选自盐酸、氢溴酸、磷酸或硫酸;所述的有机酸选自甲磺酸、对甲苯磺酸、三氟乙酸、枸杞酸、马来酸酒石酸、富马酸、柠檬酸或乳酸;所述的碱金属离子选自锂离子、钠离子、钾离子;所述的碱土金属离子选自钙离子、镁离子;所述的能提供生理上可接受的阳离子的有机碱选自甲胺、二甲胺、三甲胺、哌啶、吗啉或三(2-羟乙基)胺。
在本发明范围内的所有这些盐都可采用常规方法制备。在所述的通式I化合物及其溶剂化物和其盐的制备过程中,不同的结晶条件可能出现多晶或共晶。
本发明制备方法中的起始原料及中间体容易得到,各步反应可依据已报道的文献或对本领域熟练技术人员来说可以用有机合成中的常规方法很容易合成。通式I所述化合物 可以溶剂化物或非溶剂化物的形式存在,利用不同的溶剂进行结晶可能得到不同的溶剂化物。
式I化合物的制备
为了制备本发明通式I所述的化合物,依据通式I的结构,本发明制备通式I化合物可以通过以下合成路线获得。
(a)以中间体1和2(或2’)为原料,通过合适的钯催化剂催化的Suzuki偶联反应得到目标产物I或中间体II;
(b)以中间体II和中间体4为原料,在酸性或碱性的条件下,用还原剂(如氰基硼氢化钠、三乙酰氧基硼氢化钠或硼氢化钠等)来发生还原氨化反应得到目标产物I;
其中,中间体1、2和3的制备方法(参考WO 2018195321、WO2018005374和WO2019023575)如下:
方法1:
(a)以化合物1-1为原料,在手性辅剂(如R/S-CBS)和还原剂(如硼烷)的作用下,形成手性醇1-2;
(b)以1-2(或1-5)和1-3为原料,在合适的钯催化剂和配体的条件下,发生偶联反应(如Suzuki,Buchwald等),得到中间体1-4(或1-6);
(c)以化合物1-1和R/S-叔丁基磺酰亚胺为原料,在Lwesis酸(如四钛酸乙酯)和还原试剂(如四氢锂铝,硼氢化钠等)的作用下,形成保护的手性氨基化合物,而后在酸性条件下脱除保护基,得到手性氨基化合物1-5;
(d)以1-4(或1-6)和联硼酸频哪酯为原料,在合适的钯催化剂和配体的条件下,发生Suzuki偶联反应,得到中间体1a;
方法2:
(a)以化合物1-7和1-8为原料,在酸性或者碱性条件条,发生亲和取代反应,形成中间体1-9;
(b)以1-9和联硼酸频哪酯为原料,在合适的钯催化剂和配体的条件下,发生Suzuki偶联反应,得到中间体1b;
方法3:
(a)以化合物2-1和2-2为原料,在脱水剂(如浓硫酸,PPA等)的作用下,形成中间体2;
方法4:
(a)以化合物2-5为原料,与硝化剂反应(如浓硫酸/NaNO3,浓硫酸/发烟硝酸等),形成中间体2-6;
(b)以2-6为原料,在还原条件下(Pd-C/H2;锌粉/氯化铵;铁粉/醋酸等),发生还原反应,形成中间体2-7;
(c)以2-7和2-8为原料,在缩水剂(如PPA,Eaton试剂,PPh3/DDQ,PPh3/DIAD等)条件下,发生环化反应,得到中间体2c;
所述的R4为:为:含羰基的前体形式或D环上的羰基;t选自0、1、2、3;X1、X2、X3、X4、X5各自独立地选自C、N、O、S、P、B。
另外,上述反应中的起始原料及中间体容易得到,各步反应可依据已报道的文献或对本领域熟练技术人员来说可以用有机合成中的常规方法很容易合成。通式I所述化合物可以溶剂化物或非溶剂化物的形式存在,利用不同的溶剂进行结晶可能得到不同的溶剂化物。
药物组合物和施用方法
由于本发明化合物具有优异的PD-1/PD-L1相互作用的调节活性,因此本发明化合物及其各种晶型,药学上可接受的无机或有机盐,水合物或溶剂合物,以及含有本发明化合物为主要活性成分的药物组合物可用于预防和/或治疗(稳定、减轻或治愈)PD-1/PD-L1相 互作用相关疾病(例如,癌症、感染性疾病、自身免疫性疾病)。
本发明的药物组合物包含安全有效量范围内的本发明化合物及药学上可以接受的赋形剂或载体。其中“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明化合物/剂,更佳地,含有10-200mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。
“药学上可接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如吐温)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
本发明化合物或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、肠胃外(静脉内、肌肉内或皮下)。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。
除了活性化合物外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。
本发明化合物可以单独给药,或者与其他药学上可接受的化合物(例如其他抗癌制剂)联合给药。
联合给药时,所述药物组合物还包括与一种或多种(2种,3种,4种,或更多种)其他药学上可接受的化合物。该其他药学上可接受的化合物中的一种或多种(2种,3种,4种,或更多种)可与本发明的化合物同时、分开或顺序地用于预防和/或治疗PD-1/PD-L1相互作用相关疾病。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选20~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
本发明的主要优点包括:
(1)本发明化合物对PD-1/PD-L1相互作用具有很高的调节活性,与PD-L1蛋白具有很强的结合能力,并具有解除PD-L1抑制IFNγ的能力。
(2)本发明的化合物具有更好的溶解性较好;对正常细胞的毒性非常低,因而可以在较大的剂量范围内应用于治疗对象。
(3)相较于现有技术的化合物,本发明的化合物具有更好的溶解性,因此具有良好的成药性,相较于现有化合物而言,本发明化合物在体内实验之中表现出良好的生物利用度,除此之外,相较于现有化合物,本发明的化合物极易制成药学上可接受的盐,因而有助于进一步形成制剂。
(4)体内药效研究表明,本发明化合物无论从肿瘤体积还是重量上,都可显著的抑制皮下肿瘤的生长,并可明显增加小鼠血液中、脾脏中各淋巴细胞数量。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。
实施例1化合物1的合成
步骤1-1:
将1-01(7.6g,0.0152mol,WO2021047553),1-02(5.56g,0.0152mol,WO2021047553),XantPhos-PdCl2(1.15g,0.00152mol)和碳酸钠(4.8g,0.0455mol)依次加入1,4-二氧六环/水(4:1,100mL)中,氮气保护,油浴95度加热搅拌,TLC检测反应。反应过夜,加硅藻土过滤,滤液悬干,加乙酸乙酯(100mL)溶解残留物,依 次用水(10mL x 1)和饱和食盐水(10mL x 2)洗涤,无水硫酸钠干燥有机层,拌样,柱层析,得灰色固体2.4g。MS(APCI):681[M+H]+
步骤1-2:
将1-03(530mg,0.8mmol),1-04(223mg,1.6mmol)和氰基硼氢化钠(503mg,8mmol)依次加入DMAc(6mL)中,油浴90度搅拌过夜,LC-MS检测反应。反应完全后,加水搅拌,取固体,反相柱制备得白色固体100mg。MS(APCI):782[M-H]-
实施例2化合物102的合成
步骤2-1:
将102-01(350mg,0.816mmol,按照WO2019023575类似的方法合成),R-3-羟基吡咯盐酸盐(201mg,1.632mmol)加入DCM(10mL)中室温搅拌1小时,而后往反应液中加入三乙酰氧基硼氢化钠(519mg,2.448mmol),TLC检测反应,室温搅拌过夜。反应完全后,加水淬灭,DCM萃取,无水硫酸钠干燥,柱层析,得油状物300mg。MS(APCI):500[M+H]+
步骤2-2:
以102-02(200mg)和1-02(180mg)为原料,按照步骤1-1的合成方法,得到灰色固体150mg。MS(APCI):660[M+H]+
步骤2-3:
以102-03(150mg)和102-04(70mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体42mg。MS(APCI):781[M-H]-
实施例3化合物40的合成
步骤3-1:
以40-01(200mg,WO2019191707)和40-02(180mg,WO2021047553)为原料,按照步骤1-1的合成方法,得到灰色固体120mg。MS(APCI):692[M+H]+
步骤3-2:
以40-03(120mg)和102-04(56mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体21mg。MS(APCI):813[M-H]-
实施例4化合物47的合成
步骤4-1:
以47-01(200mg)和1-02(152mg)为原料,按照步骤1-1的合成方法,得到灰色固体115mg。MS(APCI):621[M+H]+
步骤4-2:
以47-02(115mg)和1-04(52mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体30mg。MS(APCI):742[M-H]-
实施例5化合物49的合成
步骤5-1:
以47-01(200mg)和40-02(160mg)为原料,按照步骤1-1的合成方法,得到灰色固体122mg。MS(APCI):641[M+H]+
步骤5-2:
以49-01(122mg)和102-04(56mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体30mg。MS(APCI):762[M-H]-
实施例6化合物55的合成
步骤6-1:
以55-01(200mg,WO2021047547)和1-02(143mg)为原料,按照步骤1-1的合成方法,得到灰色固体151mg。MS(APCI):586[M+H]+
步骤6-2:
以55-02(151mg)和1-04(63mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体25mg。MS(APCI):707[M-H]-
实施例7化合物104的合成
步骤7-1:
以1-01(200mg)和40-02(160mg)为原料,按照步骤1-1的合成方法,得到灰色固体131mg。MS(APCI):661[M+H]+
步骤7-2:
以104-01(131mg)和102-04(59mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体26mg。MS(APCI):802[M-H]-
实施例8化合物56的合成
步骤8-1:
以56-01(200mg,WO2021047547)和1-02(153mg)为原料,按照步骤1-1的合成方法,得到灰色固体142mg。MS(APCI):606[M+H]+
步骤8-2:
以55-02(142mg)和102-04(60mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体25mg。MS(APCI):727[M-H]-
实施例9化合物105的合成
步骤9-1:
以102-02(200mg)和105-01(180mg,WO2021047547)为原料,按照步骤1-1的合成方法,反相柱制备得到白色固体31mg。MS(APCI):759[M+H]+
实施例10化合物106的合成
步骤10-1:
以102-01(200mg)和(S)-5-氨基甲基吡咯烷-2-酮盐酸盐(120mg)为原料,按照步骤2-1的合成方法,得到油状物130mg。MS(APCI):527[M+H]+
步骤10-2:
以106-01(130mg)和105-01(102mg)为原料,按照步骤1-1的合成方法,反相柱制备得到得白色固体42mg。MS(APCI):786[M+H]+
实施例11化合物107的合成
步骤11-1:
以107-01(100mg)和1-02(153mg)为原料,按照步骤1-1的合成方法,得到灰色固体81mg。MS(APCI):423[M+H]+
步骤11-2:
以107-02(81mg)和102-04(42mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体23mg。MS(APCI):546[M-H]-
实施例12化合物108的合成
步骤12-1:
以107-01(100mg)和1-02(170mg)为原料,按照步骤1-1的合成方法,得到灰色固体85mg。MS(APCI):443[M+H]+
步骤12-2:
以108-01(85mg)和102-04(40mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体22mg。MS(APCI):566[M-H]-
实施例13化合物37的合成
步骤13-1:
以37-01(200mg,WO2018119266)和102-04(40mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体31mg。MS(APCI):716[M-H]-
实施例14化合物131的合成
步骤14-1:
以47-02(110mg)和104-02(50mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体23mg。MS(APCI):742[M-H]-
实施例15化合物132的合成
步骤3-1:
以40-01(200mg,WO2019191707)和1-02(180mg,WO2021047553)为原料,按照步骤1-1的合成方法,得到灰色固体101mg。MS(APCI):672[M+H]+
步骤3-2:
以132-01(101mg)和102-04(51mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体21mg。MS(APCI):793[M-H]-
实施例16化合物133的合成
步骤16-1:
以1-03(122mg)和102-04(65mg)为原料,按照步骤1-2的合成方法,反相柱制备得白色固体23mg。MS(APCI):782[M-H]-
实施例17化合物134的合成
步骤17-1:
以40-03(1.0g)和134-01(500mg)为原料,按照步骤1-2的合成方法,反应完全后,加水淬灭,DCM萃取,无水硫酸钠干燥,柱层析得白色固体860mg。MS(APCI):847[M+H]+
步骤17-2:
将134-02(800mg)溶于DCM(10mL)中加入三氟乙酸(2mL)室温搅拌3小时,TLC检测反应,减压浓缩溶剂得到134-03。
步骤17-3:
0℃内将EDCI(29mg)加到134-03(100mg)/DMAP(34mg)/DCM(5ml)+甲醇(0.5ml)中,加完室温搅拌过夜,HPLC/TLC检测反应完全。反应液水洗,饱和食盐水洗,加入Amberlist 120(H)树脂室温搅拌15min,干燥浓缩得浅黄色油状物,制备HPLC纯化(氯化铵溶液),收集相应组分冻干得到化合物134。MS(APCI):805[M+H]+
实施例18:化合物135~140的合成
参照步骤17-3,将甲醇换成乙醇,分别得到化合物135。MS(APCI):819[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.41(s,1H),8.98(s,1H),8.20(m,2H),8.09(s,1H),7.81(d,J=7.9Hz,1H),7.74(t,J=7.7Hz,1H),7.66(d,J=7.2Hz,1H),7.44(t,J=7.8Hz,1H),7.20(d,J=7.5Hz,1H),6.76(t,J=54.5Hz,1H),4.81(d,J=4.4Hz,1H),4.38(t,J=5.6Hz,1H),4.26(br,1H),4.07(q,J=6.9Hz,2H),3.89(q,J=14.0Hz,2H),3.26(m,1H),3.05(m,12.6Hz,2H),2.88(t,J=8.6Hz,1H),2.73(m,5H),2.49(m,1H),2.44(dd,J=9.5,3.1Hz,1H),2.25(m,2H),2.07(m,6H),1.62(br,1H),1.19(t,J=7.1Hz,3H).
参照步骤17-3,将甲醇换成异丙醇,分别得到化合物136。MS(APCI):833[M+H]+
参照步骤17-3,将甲醇换成正丁醇,分别得到化合物137。MS(APCI):847[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.36(s,1H),8.94(d,J=1.8Hz,1H),8.15(d,J=4.2Hz,2H),8.03(s,1H),7.80(d,J=8.0Hz,1H),7.69(t,J=7.7Hz,1H),7.62(d,J=7.5 Hz,1H),7.41(t,J=7.8Hz,1H),7.16(d,J=7.6Hz,1H),6.72(t,J=54.5Hz,1H),4.78(d,J=4.5Hz,1H),4.30(t,J=5.8Hz,1H),4.22(tt,J=7.6,3.7Hz,1H),3.98(t,J=6.5Hz,2H),3.85(q,J=14.0Hz,2H),3.20(dt,J=15.8,7.5Hz,1H),3.01(ddd,J=23.9,11.2,4.6Hz,2H),2.82(t,J=8.6Hz,1H),2.71(tt,J=14.8,7.0Hz,3H),2.59(p,J=8.3,7.4Hz,2H),2.48(dd,J=10.9,5.0Hz,1H),2.41(dd,J=9.6,3.5Hz,1H),2.22(q,J=6.9Hz,2H),2.04(d,J=2.8Hz,3H),1.59(qt,J=8.2,3.9Hz,1H),1.53–1.42(m,2H),1.25(q,J=7.4Hz,2H),0.82(t,J=7.4Hz,3H).
参照步骤17-3,将甲醇换成异丁醇,分别得到化合物138。MS(APCI):847[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.42(s,1H),8.98(s,1H),8.20(d,J=4.8Hz,2H),8.11(s,1H),7.80(d,J=8.0Hz,1H),7.74(t,J=7.7Hz,1H),7.66(d,J=6.9Hz,1H),7.44(t,J=7.8Hz,1H),7.20(d,J=7.5Hz,1H),6.75(t,J=54.5Hz,1H),4.79(d,J=4.5Hz,1H),4.36(t,J=5.5Hz,1H),4.26(s,1H),3.99–3.78(m,4H),3.26(dt,J=15.9,7.7Hz,1H),3.16–3.00(m,2H),2.87(t,J=8.6Hz,1H),2.81–2.73(m,2H),2.71(t,J=7.4Hz,1H),2.64(d,J=6.5Hz,2H),2.49(m,1H),2.44(dd,J=9.5,3.1Hz,1H),2.27(q,J=6.3Hz,2H),2.13–1.93(m,6H),1.93–1.79(m,1H),1.58(br,1H),1.24(s,4H),0.86(d,J=6.7Hz,6H).
参照步骤17-3,将甲醇换成正己醇,分别得到化合物139。MS(APCI):875[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.40(s,1H),8.96(s,1H),8.18(d,J=5.5Hz,2H),8.09(s,1H),7.78(d,J=8.0Hz,1H),7.72(t,J=7.7Hz,1H),7.63(d,J=7.0Hz,1H),7.42(t,J=7.8Hz,1H),7.18(d,J=7.4Hz,1H),6.73(t,J=54.5Hz,1H),4.77(s,1H),4.34(t,J=5.4Hz,1H),4.28–4.19(m,1H),3.99(t,J=5.6Hz,2H),3.87(q,J=13.9Hz,2H),3.29–3.17(m,2H),3.05(ddd,J=27.4,13.5,6.3Hz,2H),2.84(t,J=8.6Hz,1H),2.79–2.58(m,5H),2.42(m,2H),2.23(m,2H),2.02(d,J=12.4Hz,6H),1.52(br,4H),1.22(s,8H),0.81(d,J=6.3Hz,3H).
参照步骤17-3,将甲醇换成正辛醇,分别得到化合物140。MS(APCI):903[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.42(s,1H),8.98(s,1H),8.20(s,2H),8.10(s,1H),7.81(d,J=8.0Hz,1H),7.74(t,J=7.6Hz,1H),7.65(d,J=7.3Hz,1H),7.44(t,J=7.7Hz,1H),7.20(d,J=7.4Hz,1H),6.75(t,J=54.5Hz,1H),4.80(d,J=4.4Hz,1H),4.36(t,J=5.4Hz,1H),4.26(s,1H),4.06–3.97(m,2H),3.90(q,J=13.9Hz,2H),3.31–3.20(m,1H),3.06(ddd,J=35.5,14.2,7.4Hz,2H),2.86(t,J=8.6Hz,1H),2.81–2.69(m,3H),2.68–2.61(m,2H),2.52–2.40(m,2H),2.32–2.19(m,2H),2.10–1.91(m,7H),1.62(br,1H),1.57–1.48(m,2H),1.29–1.13(m,12H),0.82(t,J=6.1Hz,3H).
实施例19:化合物141~143的合成
0℃内将EDCI(29mg)加到141-01(100mg)/DMAP(34mg)/DCM(5ml)+甲醇(0.5ml)中,加完室温搅拌过夜,HPLC/TLC检测反应完全。反应液水洗,饱和食盐水洗,加入Amberlist 120(H)树脂室温搅拌15min,干燥浓缩得浅黄色油状物,制备HPLC纯化(氯化铵溶液),收集相应组分冻干得到化合物141。MS(APCI):774[M+H]+
参照上述步骤将甲醇换成乙醇,得到化合物142。MS(APCI):788[M+H]+
参照上述步骤将甲醇换成正丁醇,得到化合物143。MS(APCI):816[M+H]+
按照实施例1-12的合成方法,以相应的原料和和试剂合成了下列表中的化合物:

生物测试
实施例A:PD-1/PD-L1均相时间分辨荧光(HTRF)结合测定
测定在标准黑色384孔聚苯乙烯板中进行,终体积为20μL。首先将抑制剂用DMSO连续稀释后加入板孔中,再加入其他反应组分。测定中DMSO的最终浓度为1%。测定是在25℃下含有0.05%Tween-20和0.1%BSA的PBS缓冲液(pH7.4)中进行的。在C端带有His标记的重组人PD-L1蛋白(19-238)购自AcroBiosystems公司(PD1-H5229)。在C端带有Fc标记的重组人PD-1蛋白(25-167)也购自AcroBiosystems公司(PD1-H5257)。将PD-L1和PD-1蛋白在测定缓冲液中稀释然后提取0.1μl溶液加入到板孔中。离心平板并将蛋白质与抑制剂预孵育40分钟。孵育后加入0.1μl HTRF检测缓冲液含有铕封闭标记的抗人IgG(PerkinElmer-AD0212)Fc专属的和抗His的-别藻蓝蛋白(APC,PerkinElmer-AD0059H)缀合的抗体。离心后,将孔板在25℃下孵育60分钟。置于PHERAstar FS读板器中读取数据(665nm/620nm比率)。测定中的最终浓度为~3nM PD1、10nM PD-L1、1nM铕抗人IgG和20nM抗-His-别藻蓝蛋白。使用GraphPad Prism5.0软件拟合活性数据得出抑制剂的IC50值。实施例中举例说明的化合物IC50值以下列方式表示:IC50:+=≤100nM;++=100~1000nM;+++=>1000nM。使用实施例A中描述的PD-1/PD-L1均相时间分辨荧光(HTRF)结合测定获得的实施例化合物的数据提供于表1中。
表1.

实施例B:ICR小鼠体内单次静注和灌胃给药后的药代动力学
1.处方配制
静注给药制剂配制方法:准确称量约2.5mg化合物至玻璃瓶中,加入一定体积的20%HP-β-CD水溶液,涡旋振荡混合均匀后,调节pH至3.0左右(如需要),超声使固体充分溶解,过滤,得浓度为1mg/mL的A组静注给药制剂溶液。
灌胃给药制剂配制方法:准确称量约10mg化合物至玻璃瓶中,加入一定体积的20%HP-β-CD水溶液,涡旋振荡混合均匀后,调节PH至3.0左右(如需要),超声使固体充分溶解,得浓度为2mg/mL的B组灌胃给药制剂溶液。
2.储备液配制
准确称量2mg左右化合物至5mL玻璃瓶中,精密加入所需体积的DMSO,配制成浓度为1mg/mL的储备液,于2~8℃储存备用。
3.实验动物
实验动物饲养在动物房内,动物房装备空调系统,通风良好,室内温度维持在20 ~26℃范围,湿度维持在40%~70%范围内。动物房内采用人工照明,明暗各12小时(因实验操作、清洁需开启工作照明等情况除外),实验动物自由采食和饮水。动物购入后正常喂养至少5天(用于急性终止性实验持续时间不超过48小时的小鼠喂养至少3天),经兽医检验,体征状况良好的动物入选本实验,每只小鼠均用尾部编号标记。
4.实验设计
本实验的动物静注、灌胃按规定的剂量进行给药。
给药前,检查给药制剂状态,以涡旋或快速摇匀的方式确保制剂均匀性,依照下述公式计算每只动物的理论给药量。
样品采集:按照给药后0.25h、0.5h、1h、2h、4h、6h、8h和24h时间点,由眼眶采血约100μL,将采集的全血置于EDTA-K2的抗凝管中,颠倒数次充分混匀,存放于湿冰上,并于30min之内,离心(1500~1600g)10min,分离血浆,将所得血浆样品保存于-40~-20℃环境中,用于生物样品分析。
实验结束后,采用二氧化碳窒息法或者颈椎脱臼法对小鼠实施安乐死。
分析结束后样品返还生物样品间,样品间根据SOP将样品转移至-40~-20℃环境中进行封存。
5.生物分析
根据液质联用法生物样品分析方法的要求,建立测定小鼠血浆中化合物及酯基水解代谢物浓度的LC-MS/MS分析方法,并用于测定本实验获得的生物样品中化合物的浓度。用于本实验获得的生物样品的浓度测定。采用Phoenix WinNonlin 8.3中的非房室模型计算相应的药代动力学参数。数据分析结果见表2~5:
表2 5mg化合物134-03灌胃给药情况下的药代动力学实验数据
表3 5mg化合物134和40静脉滴注给药下的药代动力学实验数据

表4不同给药方式下各个化合物的药代动力学实验数据
表5不同给药方式下各个化合物的药代动力学实验数据
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (15)

  1. 一种如下式I所示的化合物,或其光学异构体、水合物、溶剂合物,或其药学上可接受的盐:
    其中,m为0、1、2、3,4或5;
    n、v和p各自独立地为0、1、2、3或4;
    各自独立地选自下组:取代或未取代的5-12元杂芳基、取代或未取代的C6-C10芳基、取代或未取代的5-12元杂环基、取代或未取代的5-12元的C3-C12(优选为C5-C12)环基,其中,所述的杂环基具有1-3个杂原子;或D为
    其中,X1、X2、X3、X4、X5和X6各自独立地选自下组:N、O、S、SO、SO2、C(R)2、CHR、NR;其中,各个R可以任选地被替代;
    Y1、Y2、Y3、Y4、Y5和Y6各自独立地选自下组:N、CH、C;
    其中,X3、X4、X5的碳原子上的氢(如存在的情况下)均可各自独立的被氘替换;
    L3选自下组:化学键、取代或未取代的C1-C7亚烷基、取代或未取代的C2-C4亚烯基、取代或未取代的C2-C4亚炔基、-S-、-O-、取代或未取代的-NH-、-S(O)-、或-S(O)2-;
    E选自下组:取代或未取代的C6-C12亚芳基、取代或未取代的具有1-3个杂原子的5-12元亚杂芳基、取代或未取代的C3-C8亚环烷基、取代或未取代的具有1-3个杂原子的3-10元亚杂环基;
    r为1、2、3、4、5、6;
    s为0、1、2;
    a为1、2、3、4、5或6;
    R4选自下组:
    R5选自下组:H、-CN、三氟甲基、-CHF2、-OCF3、-OCHF2、磺酰氨基、硝基、羟基、卤素、取代或未取代的C1-C6烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、-CN、羟基、
    各自独立地选自下组:取代或未取代的C6-C10亚芳基、或取代或未取代的具有1-3个杂原子的5-12元(优选5-7元)亚杂芳基、取代或未取代的5-12元亚杂环基、取代或未取代的5-12元的C3-C12(优选为C5-C12)亚环烷基;或不存在;
    L1和L2各自独立地选自下组:化学键、取代或未取代的C1-C4亚烷基、取代或未取代的C2-C4亚烯基、取代或未取代的C2-C4亚炔基、-S-、-O-、取代或未取代的-NH-、-S(O)-、-S(O)2-、取代或未取代的-NHC(O)NH-、取代或未取代的取代或未取代的取代或未取代的
    表示单键或双键;
    R、R1、R2和R3各自独立地选自下组:H、-CN、三氟甲基、-CHF2、-OCF3、-OCHF2、磺酰氨基、硝基、羟基、卤素、-S-R8、-S(O)-R8、-S(O)2-R8、取代或未取代的C1-C10烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、-CN、羟基、NRdRe(例如氨基)、取代或未取代的C1-C6胺基、取代或未取代的-(C1-C6亚烷基)-NH-(C1-C6亚烷基)、羧基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个杂原子的5-12元杂芳基、取代或未取代的具有1-4个杂原子的3-12元杂环基,其中,取代或未取代的C1-C10烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、羟基、NRdRe(例如氨基)、取代或未取代的C1-C6胺基、取代或未取代的-(C1-C6亚烷基)-NH-(C1-C6亚烷基)、羧基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个杂原子的5-12元杂芳基、取代或未取代的具有1-4个杂原子的3-12元杂环基的碳原子上的氢均可各自独立的被氘替换;取代或未取代的取代或未取代的取代或未 取代的其中,Rb和Rc各自独立地选自下组:H、取代或取代的C1-C8烷基;或所述的Rb和Rc与相邻的N原子共同构成取代或未取代的具有1-3个选自N、S和O的杂原子的3-10元杂环基,或所述的Rb和Rc与相邻的N原子共同构成取代或未取代的4-10元内酰胺,所述取代基包括但不限于:羟基、羧基、巯基、氨基、F、Cl,其中,Rb、Rc和Rd的碳原子上的氢均可各自独立地被氘替换;或-(L1a)r-(L2a)s-(L3a)s-;-C0-8-O-R8,-C0-8-C(O)OR8,-C0-8-OC(O)OR8,-C0-8-NR8R9,-C0-8-N(R8)C(O)R9,-C0-8-C(O)NR8R9
    R8和R9各自独立地选自下组:H、羟基,取代或未取代的C1-C10烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、-CN、羟基、NRdRe(例如氨基)、取代或未取代的C1-C6胺基、取代或未取代的-(C1-C6亚烷基)-NH-(C1-C6亚烷基)、羧基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个杂原子的5-12元杂芳基、取代或未取代的具有1-4个杂原子的3-12元杂环基,取代或未取代的取代或未取代的取代或未取代的或-(L1a)r-(L2a)s-(L3a)s-;
    各个L1a各自独立地为选自下组的基团;化学键、取代或未取代的C1-C7亚烷基、取代或未取代的C2-C4亚烯基、取代或未取代的C2-C4亚炔基、-S-、-O-、取代或未取代的-NH-、-S(O)-、或-S(O)2-;
    L2a选自下组:取代或未取代的C6-C12亚芳基、取代或未取代的具有1-3个杂原子的5-12元亚杂芳基、取代或未取代的C3-C8亚环烷基、取代或未取代的具有1-3个杂原子的3-10元亚杂环基;
    L3a选自下组:取代或未取代的C1-C10烷基、C1-C10芳基、-CN、羟基、氨基、羧基、-CO-NH-SO2-Rg、-NH-SO2-Rg、-SO2-NH-CO-Rg、-ORg、-N(Rg)2-CO2Rg、-CON(Rg)2、-CONHCORg、NRg-CO-N(Rg)2、-NRg-SO2-N(Rg)2
    r为1、2、3、4、5、6;
    s分别为0、1、2;
    Rd、Re和Rg各自独立地选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C10环烷基、取代或未取代的C6-C10芳基;或Rd和Re共同形成取代或未取代的具有1-3个选自N、S和O的杂原子的4-10(优选为5-10)元杂环基;
    所述的Rf选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C6-C10芳基、取代或未取代的5-10元杂芳基、氰基、-C(=O)-NRdRe、-C(=O)-取代或未取代的C1-C6烷氧基、-C(=O)-取代或未取代的C1-C6烷基、-C(=O)-取代或未取代的C3-C10环烷基、-C(=O)-取代或未取代的C2-C6烯基、-C(=O)-取代或未取代的C2-C6炔基;
    除非特别说明,所述的“取代”是指被选自下组的一个或多个(例如2个、3个、4个等)取代基所取代:卤素(包括-F、-Cl、-Br、-I)、-CH2Cl、-CHCl2、-CCl3、-CH2F、-CHF2、-CF3、氧代(=O)、-CN、羟基、氨基、C1-C6烷胺基、羧基、-NHAc、 未取代或被一个或多个取代基取代的选自下组的基团:C1-C6烷基、C1-C6烷氧基、C6-C10芳基、C3-C8环烷基、卤代的C6-C10芳基、具有1-3个选自N、S和O的杂原子的5-10元杂芳基、具有1-3个选自N、S和O的杂原子的5-10元杂环基;所述的取代基选自下组:卤素、羟基、羧基、氧基、氰基、C1-C6烷氧基、C1-C6烷胺基;
    上述各式中,任一所述杂原子选自下组:B、P、N、S和O。
  2. 如权利要求1所述的化合物,或其异构体、光学异构体、水合物、溶剂合物,或其药学上可接受的盐,其特征在于,所述的各自独立地为选自下组的环形成的二价基团: 其中,所述的环的成键位置可以为N或C;更佳地,所述的各自独立地为
  3. 如权利要求1所述的化合物,其特征在于,所述的具有如下式所示的结构:
    其中,
    X6、X7、X8、X9、X10和X11各自独立地选自下组:N、CR;
    R6选自下组:H、卤素、取代或未取代的C1-C6烷基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、氧代(即=O)、=NRf、-CN、羟基、NRdRe(例如氨基)、取代或未取代的C1-C6胺基、取代或未取代的-(C1-C6亚烷基)-NH-(C1-C6亚烷基)、羧基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个杂原子的5-12元杂芳基、取代或未取代的具有1-4个杂原子的5-12元杂环基,取代或未取代的取代或未取代的取代或未取代的或-(L1a)r-(L2a)s-(L3a)s-,-C0-8-O-R8,-C0-8-C(O)OR8,-C0- 8-OC(O)OR8,-C0-8-NR8R9,-C0-8-N(R8)C(O)R9,-C0-8-C(O)NR8R9
  4. 如权利要求1所述的化合物,或其光学异构体、水合物、溶剂合物,或其药学上可接受的盐,其特征在于,所述的具有如下式所示的结构: 其中,所述的环的成键位置可以为N或C。
  5. 如权利要求1所述的化合物,其特征在于,所述的D环具有选自下组的结构: (成键位置可以为N或C) 较佳地,为选自下组的结构:
  6. 如权利要求1所述的化合物,或其光学异构体、水合物、溶剂合物,或其药学上可接受的盐,其特征在于,所述的化合物具有选自下组的结构:
    其中,F环为选自下组的基团:取代或未取代的苯基、取代或未取代的吡啶基;R6选自下组:H、卤素、C1-C4烷基。
  7. 如权利要求1所述的化合物,或其光学异构体、水合物、溶剂合物,或其药学上可接受的盐,其特征在于,所述的R4具有选自下组的结构:
  8. 如权利要求1所述的化合物,或其光学异构体、水合物、溶剂合物,或其药学上可接受的盐,其特征在于,所述的化合物具有选自下组的结构:
    其中,所述的环A为:
    其中,
    X6、X7、X8、X9、X10和X11各自独立地选自下组:N、CR;较佳地,X10为C(CHF2);
    R6选自下组:H、卤素、未取代或卤代的C1-C6烷基;
    D为其中,X1、X2、X3、X4、X5和X6各自独立地选自下组:N、O、S、SO、SO2、C(R)2、CHR、NR;其中,各个R可以任选地被替代;其中,r和s为1;L3为化学键、取代或未取代的C1-C7亚烷基;且E为5-7元的亚氮杂环基;
    R4选自下组:
  9. 如权利要求1所述的化合物,或其光学异构体、水合物、溶剂合物,或其药学上可接受的盐,其特征在于,所述的化合物选自下列化合物:









  10. 一种药物组合物,其特征在于,包含(1)如权利要求1所述的化合物或其立体异构体或互变异构体,或其药学上可接受的盐、水合物或溶剂化物;(2)药学上可接受的载体。
  11. 如权利要求1所述的化合物或其立体异构体或互变异构体,或其药学上可接受的盐、水合物或溶剂化物或如权利要求10所述的药物组合物的用途,其特征在于,用于制备预防和/或治疗与PD-1/PD-L1的活性或表达量相关的疾病的药物组合物;较佳地,所述的药物组合物用于治疗选自下组的疾病:癌症、感染性疾病、自身免疫性疾病。
  12. 如权利要求11所述的用途,其特征在于,所述的癌症选自下组:胰腺癌,膀胱癌,结肠直肠癌,乳腺癌,前列腺癌,肾癌,肝细胞癌,肺癌,卵巢癌,宫颈癌,胃癌,食道癌,黑色素瘤,神经内分泌癌,中枢神经系统癌,脑癌,骨癌,软组织肉瘤,非小细胞肺癌癌症,小细胞肺癌或结肠癌、皮肤癌、肺癌、泌尿系肿瘤、血液肿瘤、胶质瘤、消化系统肿瘤、生殖系统肿瘤、淋巴瘤、神经系统肿瘤、脑瘤、头颈癌。
  13. 如权利要求11所述的用途,其特征在于,所述的感染性疾病选自细菌感染、病毒感染。
  14. 如权利要求11所述的用途,其特征在于,所述的自身免疫性疾病选自器官特异性自身免疫病、系统性自身免疫病。
  15. 如权利要求11所述的用途,其特征在于,所述的药物组合物还包括至少一种选自下组的治疗剂:纳武单抗,派姆单抗,atezolizumab,或伊匹单抗。
PCT/CN2023/139583 2022-12-16 2023-12-18 新型联芳环化合物作为免疫调节剂的制备和应用 WO2024125662A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211627914.1 2022-12-16
CN202211627914 2022-12-16

Publications (1)

Publication Number Publication Date
WO2024125662A1 true WO2024125662A1 (zh) 2024-06-20

Family

ID=91484404

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/139583 WO2024125662A1 (zh) 2022-12-16 2023-12-18 新型联芳环化合物作为免疫调节剂的制备和应用

Country Status (1)

Country Link
WO (1) WO2024125662A1 (zh)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110799509A (zh) * 2017-04-20 2020-02-14 吉利德科学公司 Pd-1/pd-l1抑制剂
WO2021047555A1 (zh) * 2019-09-09 2021-03-18 上海长森药业有限公司 芳香杂环衍生物作为免疫调节剂的制备及其应用
WO2021047547A1 (zh) * 2019-09-09 2021-03-18 上海长森药业有限公司 新型三环芳香杂环化合物,及其制备方法、药物组合物和应用
WO2021047553A1 (zh) * 2019-09-09 2021-03-18 上海长森药业有限公司 含三环结构的芳香杂环化合物,及其制备方法和应用
CN113637013A (zh) * 2020-05-11 2021-11-12 上海长森药业有限公司 联芳环链接芳杂环衍生物作为免疫调节剂的制备及其应用
CN113717165A (zh) * 2020-05-22 2021-11-30 上海长森药业有限公司 新型三环芳香杂环化合物,及其制备方法、药物组合物和应用

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110799509A (zh) * 2017-04-20 2020-02-14 吉利德科学公司 Pd-1/pd-l1抑制剂
WO2021047555A1 (zh) * 2019-09-09 2021-03-18 上海长森药业有限公司 芳香杂环衍生物作为免疫调节剂的制备及其应用
WO2021047547A1 (zh) * 2019-09-09 2021-03-18 上海长森药业有限公司 新型三环芳香杂环化合物,及其制备方法、药物组合物和应用
WO2021047553A1 (zh) * 2019-09-09 2021-03-18 上海长森药业有限公司 含三环结构的芳香杂环化合物,及其制备方法和应用
CN113637013A (zh) * 2020-05-11 2021-11-12 上海长森药业有限公司 联芳环链接芳杂环衍生物作为免疫调节剂的制备及其应用
CN113717165A (zh) * 2020-05-22 2021-11-30 上海长森药业有限公司 新型三环芳香杂环化合物,及其制备方法、药物组合物和应用

Similar Documents

Publication Publication Date Title
US11407749B2 (en) Heterocyclic compounds as immunomodulators
US11613536B2 (en) Heterocyclic compounds as immunomodulators
CN111039942B (zh) 含氮杂环类化合物,及其制备方法、药物组合物和应用
CN108699001B (zh) 作为免疫调节剂的杂环化合物
CN108349981B (zh) 新型的吡唑并[3,4-d]嘧啶化合物或其盐
KR102215172B1 (ko) 이미다조피라진아민 페닐 유도체 및 그의 용도
US20180179201A1 (en) Heterocyclic compounds as immunomodulators
CN112457308B (zh) 新型三环芳香杂环化合物,及其制备方法、药物组合物和应用
CN112292374B (zh) 一种新型磷酸肌醇3-激酶抑制剂及其制备方法和用途
WO2021228000A1 (zh) 联芳环链接芳杂环衍生物作为免疫调节剂的制备及其应用
WO2021047553A1 (zh) 含三环结构的芳香杂环化合物,及其制备方法和应用
CN110003204B (zh) 一种bet蛋白抑制剂、其制备方法及用途
WO2021047555A1 (zh) 芳香杂环衍生物作为免疫调节剂的制备及其应用
WO2023208174A1 (zh) 去泛素化酶抑制剂及其应用
WO2021233454A1 (zh) 新型三环芳香杂环化合物,及其制备方法、药物组合物和应用
WO2024125662A1 (zh) 新型联芳环化合物作为免疫调节剂的制备和应用
CN111978325B (zh) 咪唑并哒嗪类mnk1/mnk2激酶抑制剂及其制备方法和应用
WO2021238818A1 (zh) 一种大环jak抑制剂及其应用
WO2023016511A1 (zh) 氮杂芳基化合物、其制备方法及应用
KR20240021239A (ko) Cdk 키나아제 억제제로 사용되는 화합물 및 이의 용도
WO2024006776A1 (en) Estrogen receptor alpha degraders and medical use thereof
WO2022117012A1 (zh) 螺环jak抑制剂、含其的药物组合物及其应用