WO2020205538A1 - Cd73 inhibitors and therapeutic uses thereof - Google Patents

Cd73 inhibitors and therapeutic uses thereof Download PDF

Info

Publication number
WO2020205538A1
WO2020205538A1 PCT/US2020/025269 US2020025269W WO2020205538A1 WO 2020205538 A1 WO2020205538 A1 WO 2020205538A1 US 2020025269 W US2020025269 W US 2020025269W WO 2020205538 A1 WO2020205538 A1 WO 2020205538A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
cancer
heterocyclyl
hydrogen
Prior art date
Application number
PCT/US2020/025269
Other languages
French (fr)
Inventor
Jian Liu
Heping Wu
Linghang Zhuang
Suxing Liu
Rumin Zhang
Jing Li
Feng He
Weikang Tao
Original Assignee
Eternity Bioscience Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eternity Bioscience Inc. filed Critical Eternity Bioscience Inc.
Priority to CN202080024912.6A priority Critical patent/CN113905743B/en
Publication of WO2020205538A1 publication Critical patent/WO2020205538A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/167Purine radicals with ribosyl as the saccharide radical

Definitions

  • the present invention relates to novel nucleoside and nucleotide analogs, pharmaceutical compositions containing these compounds useful as CD73 inhibitors for the treatment of cancer and other diseases mediated by CD73, and methods of preparing these compounds and compositions.
  • CD73 (also known as ecto-5’ -nucleotidase) is a cell surface enzyme through a glycosyl phosphatidylinositol linkage to anchor onto the cell membrane and is expressed in different tissues, especially in the colon, kidney, brain, liver, heart, lung, spleen, lymph nodes, and bone marrow (Antonioli, L., et al., Trends Mol. Med., 2013, 19, 355-367).
  • the enzymatic activity of CD73 is to catalyze the extracellular dephosphorylation of nucleoside monophosphates to their corresponding nucleosides (e.g., 5-AMP to adenosine).
  • CD73 exerts physiological influences mainly via its enzymatic nucleoside products, particularly adenosine in extracellular space, including epithelial ion and fluid transportation, tissue barrier function control, adaptation to hypoxia, ischemic preconditioning, anti-inflammation, and immune suppression signaling (Colgan, S.P., et al., Purinergic Signaling, 2006, 2, 351-360).
  • adenosine produces a broad range of physiological responses in the human body via interaction with adenosine receptors (receptor subtypes: Al , A2A , A2B , and A3), including the vasodilation and atrioventricular conduction suppression properties in the cardiovascular system; the sedative, local neuronal excitability inhibition, anticonvulsant, and neuroprotective effects in the central nervous system (Dunwiddie, T.V., et al., Annu. Rev. Neurosci.
  • CD73 is broadly expressed in many cancer types (Antonioli, L. et al., Trends in Cancer, 2016, 2(2), 95-109) and associated with many cancer types’ poor prognosis (Allard, D., et al., Immunotherapy, 2016, 8(2), 145-163). CD73 promotes cancer metastasis (Yang, Q., et al., Pathol. Oncol.
  • CD73 is found on the surface of macrophages, lymphocytes, regulatory T cells, myeloid-derived suppressor cells (MDSCs), and dendritic cells.
  • the extracellular adenosine mainly produced by CD73, can chronically accumulate in tumor microenvironment, activating adenosine receptors, promoting tumor- inducing mononuclear phagocytes, deregulating anti-tumor T cell response, expanding MDSCs population, triggering immune suppression and favoring the escape of cancer cells from immune surveillance, hence promoting cancer transformation and growth (Antonioli, L., et al., Nature Reviews Cancer, 2013, 13, 842-857).
  • CD73 inhibitors can be used to enhance immune response and treat adenosine and adenosine receptor related diseases or disorders, including neurological, neurodegenerative and CNS disorders and diseases, depression and Parkinson’s disease, cerebral and cardiac ischaemic diseases, sleep disorders, fibrosis, immune and inflammatory disease, and cancer.
  • small-molecule drug candidates as CD73 inhibitors are mostly at the discovery stage, which are represented in several published patent applications, such as WO2015164573, WO2017098421, W02017120508, WO2017153952, US20170044203,
  • the present invention in one aspect, provides a compound of formula (I) having the following structure:
  • W is selected from the group consisting of O, S, NH, NR a and C(R b )2, wherein R a is alkyl, and R b at each occurrence is independently selected from the group consisting of hydrogen, halogen, alkyl and alkenyl ;
  • G 1 and G 2 are each independently N or CR C , wherein R c is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, hydroxy, amino, nitro, cyano, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • G 3 and G 4 are each independently selected from the group consisting of C, CH, CH 2 , N, NH, O, S and S0 2 ;
  • ring A is selected from the group consisting of Cvxcycloalkyl, 5 to 8-member heterocyclyl, aryl fused Cs-scycloalkyl, heteroaryl fused Cs-scycloalkyl, aryl fused 5 to 8- member heterocyclyl, heteroaryl fused 5 to 8-member heterocyclyl;
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of hydroxy, hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxyalkyl, cyano, amino, azide group and OR 10 ;
  • R 1 and R 2 together with the carbon atom they attached to form a cycloalkyl or heterocyclyl, wherein the heterocyclyl contains 1 to 2 heteroatoms which are the same or different from N, O and S, and wherein the cycloalkyl and heterocyclyl are each optionally substituted by one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl and heterocyclyl;
  • R 5 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro and azide;
  • R 6 is selected from the group consisting of hydrogen and optionally substituted Ci-G, alkyl;
  • R 7 at each occurrence is independently selected from the group consisting of hydrogen, alkyl, aryl, -C(R m R n )-aryl, -C(R m R n )-0-C(0)0R d , -C(R m R n )-0-C(0)R d , - C(R m R n )C(0)OR d , cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl is optionally substituted by one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of cycloalkyl, heterocyclyl, aryl and heteroaryl; optionally, two R 7 are combined to form a 5- to 7- membered heterocyclic ring;
  • R m and R n are each independently selected from the group consisting of H, D, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydro xyalkyl and amino;
  • R d is selected from the group consisting of hydrogen, alkyl and alkoxy
  • R 8 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, azide, cycloalkyl and heterocyclyl;
  • R 9 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, azide, cycloalkyl and heterocyclyl;
  • R 10 is selected from the group consisting of -C(0)R n , -C(0)OR n , -S(0) 2 R n , and - P(0)(0R 7 ) 2 ;
  • R 11 is selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, hydroxyl and hydroxyalkyl;
  • X is O
  • m 1 , 2 or 3 ;
  • s 0, 1, 2, 3 or 4.
  • the present invention provides a pharmaceutical composition, comprising a compound of formula (I), in any embodiment disclosed herein, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the present invention provides a method of treating or preventing a CD73 -mediated disease or condition, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I), in any embodiment disclosed herein, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof.
  • a compound of formula (I) in any embodiment disclosed herein, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof.
  • the present invention relates to use of a compound of formula (I), in any embodiment disclosed herein, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for treatment or prevention of a CD73 -mediated disease or condition.
  • the CD73-mediated diseases or conditions include, without limitation, tumors, cancers, immune-related diseases, inflammatory-related diseases, nervous system diseases, neurodegenerative and central nervous system diseases, depression, Parkinson's disease, ischemic diseases of the brain and heart, sleep disorders, and fibrosis, or the like.
  • the present invention relates to novel nucleoside and nucleotide analogs useful as
  • the present invention provides a compound of formula (I) having the following structure, as defined above:
  • the compound of formula (I) is selected from a compound of formula (II):
  • the compound of formula (I) is selected from a compound of formula (III) :
  • G 1 , G 2 , G 3 , G 4 , ring A, R 1 , R 3 , R 7 to R 9 , m and s are as defined in formula (I).
  • the compound of formula (I) is selected from a compound of formula
  • G 3 , G 4 , ring A, R 7 to R 9 , m and s are as defined in formula (I).
  • the invention provides a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof,
  • R 9 and s are as defined in formula (I).
  • the invention provides a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R 7 is selected from the group consisting of hydrogen, alkyl, and -C(R m R n )-0-C(0)0R d .
  • the invention provides a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R 8 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl and haloalkoxy.
  • the invention provides a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R 9 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl and haloalkoxy.
  • R 9 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl and haloalkoxy.
  • Exemplified compounds of the invention include, but are not limited to:
  • the present invention also provides a pharmaceutical composition, comprising a therapeutically effective amount of a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, together with one or more pharmaceutically acceptable carriers, diluents or excipients.
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, together with one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present invention also relates to a method for inhibiting CD73, comprising contacting a biological sample comprising CD73 with a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof.
  • a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof.
  • the present invention also relates to a method for treating a CD73 -mediated disease or condition, comprising a step of administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof.
  • a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof.
  • the present invention also relates to a method for treating a disease or condition, comprising administering to a subject in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof, wherein the disease or condition is selected from the group consisting of tumor, cancer, immune-related disease, inflammatory-related diseases, nervous system, neurodegenerative and central nervous system diseases, depression, Parkinson's disease, ischemic diseases of the brain and heart, sleep disorders and fibrosis.
  • the cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, gastric cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, metrocarcinoma, endometriosis, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder cancer, gallbladder cancer, cholangiocarcinoma, chorionic epithelial cancer, and pediatric tumor.
  • the present invention also relates to use of a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof, in the preparation of a medicament for treating a CD73 -mediated disease or condition.
  • the CD73-mediated disease or condition includes, but is not limited to, tumor, cancer, immune-related disease, inflammatory-related diseases, nervous system, neurodegenerative and central nervous system diseases, depression, Parkinson's disease, ischemic diseases of the brain and heart, sleep disorders and fibrosis.
  • compositions of this invention can be formulated by conventional methods using one or more pharmaceutically acceptable carriers.
  • the active compounds of this invention can be formulated as various dosage forms for oral, buccal, intranasal, parenteral (e.g., intravenous, intramuscular or subcutaneous), rectal administration, inhalation or insufflation administration.
  • the compounds of this invention can also be formulated as sustained release dosage forms.
  • Suitable dosage forms include, but are not limited to, a tablet, troche, lozenge, aqueous or oily suspension, dispersible powder or granule, emulsion, hard or soft capsule, or syrup or elixir.
  • Oral compositions can be prepared according to any known method in the art for the preparation of pharmaceutical compositions. Such compositions can contain one or more additives selected from the group consisting of sweeteners, flavoring agents, colorants and preservatives, in order to provide a pleasing and palatable pharmaceutical preparation. Tablets contain the active ingredient and nontoxic pharmaceutically acceptable excipients suitable for the manufacture of tablets. These excipients can be inert excipients, granulating agents, disintegrating agents, and lubricants.
  • the tablet can be uncoated or coated by means of a known technique to mask the taste of the drug or delay the disintegration and absorption of the drug in the gastrointestinal tract, thereby providing sustained release over an extended period.
  • a known technique to mask the taste of the drug or delay the disintegration and absorption of the drug in the gastrointestinal tract, thereby providing sustained release over an extended period.
  • water soluble taste masking materials can be used.
  • Oral formulations can also be provided as soft gelatin capsules in which the active ingredient is mixed with an inert solid diluent, or the active ingredient is mixed with a water soluble carrier.
  • An aqueous suspension contains the active ingredient in admixture with excipients suitable for the manufacture of an aqueous suspension.
  • excipients are suspending agents, dispersants or humectants, and can be naturally occurring phospholipids.
  • the aqueous suspension can also contain one or more preservatives, one or more colorants, one or more flavoring agents, and one or more sweeteners.
  • An oil suspension can be formulated by suspending the active ingredient in a vegetable oil, or in a mineral oil.
  • the oil suspension can contain a thickener.
  • the aforementioned sweeteners and flavoring agents can be added to provide a palatable preparation. These compositions can be preserved by adding an antioxidant.
  • the active ingredient and the dispersants or wetting agents, suspending agent or one or more preservatives can be prepared as a dispersible powder or granule suitable for the preparation of an aqueous suspension by adding water. Suitable dispersants or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, such as sweeteners, flavoring agents and colorants, can also be added. These compositions can be preserved by adding an antioxidant such as ascorbic acid.
  • the present pharmaceutical composition can also be in the form of an oil-in-water emulsion.
  • the oil phase can be a vegetable oil, or a mineral oil, or mixture thereof.
  • Suitable emulsifying agents can be naturally occurring phospholipids. Sweeteners can be used.
  • Such formulations can also contain moderators, preservatives, colorants and antioxidants.
  • the pharmaceutical composition can be in the form of a sterile injectable aqueous solution.
  • the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • the sterile injectable preparation can also be a sterile injectable oil-in-water microemulsion in which the active ingredient is dissolved in the oil phase.
  • the injectable solution or microemulsion can be introduced into an individual’s bloodstream by local bolus injection. Alternatively, it can be advantageous to administer the solution or microemulsion in such a way as to maintain a constant circulating concentration of the present compound. In order to maintain such a constant concentration, a continuous intravenous delivery device can be utilized. An example of such a device is Deltec CADD- PLUS. TM.
  • the pharmaceutical composition can be in the form of a sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration ⁇ Such a suspension can be formulated with suitable dispersants or wetting agents and suspending agents as described above according to known techniques.
  • the sterile injectable preparation can also be a sterile injectable solution or suspension prepared in a nontoxic parenterally acceptable diluent or solvent.
  • sterile fixed oils can easily be used as a solvent or suspending medium, and fatty acids can also be used to prepare injections.
  • the present compound can be administered in the form of a suppository for rectal administration ⁇
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures, but liquid in the rectum, thereby melting in the rectum to release the drug.
  • compositions can be formulated as tablets or lozenges by conventional means.
  • the active compounds of the present invention are conveniently delivered in the form of a solution or suspension released from a pump spray container that is squeezed or pumped by the patient, or as an aerosol spray released from a pressurized container or nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit can be determined by providing a valve to deliver a metered amount.
  • the pressurized container or nebulizer can contain a solution or suspension of the active compound.
  • Capsules or cartridges for example, made from gelatin
  • for use in an inhaler or insufflator can be formulated
  • the dosage of a drug depends on a variety of factors, including but not limited to, the following factors: activity of the specific compound, age, weight, general health, behavior, diet of the patient, administration time, administration route, excretion rate, drug combination and the like.
  • the best treatment such as treatment mode, daily dose of the compound of formula (I) or the type of pharmaceutically acceptable salt thereof can be verified by traditional therapeutic regimens.
  • Alkyl refers to a saturated aliphatic hydrocarbon group including C1-C20 straight chain and branched chain groups.
  • an alkyl group is an alkyl having 1 to 12 carbon atoms.
  • Representative examples include, but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1 -dimethyl propyl, 1,2-dimethyl propyl, 2,2-dimethyl propyl, 1-ethyl propyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl- 2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2- dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-
  • an alkyl group is a lower alkyl having 1 to 6 carbon atoms.
  • Representative examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n- pentyl, 1,1- dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3- methylbutyl, n-hexyl, l-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2- dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3- methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, etc.
  • the alkyl group can be substituted or unsubstituted.
  • the substituent group(s) can be substituted at any available connection point, preferably the substituent group(s) is one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group.
  • Alkenyl refers to an alkyl defined as above that has at least two carbon atoms and at least one carbon-carbon double bond, for example, vinyl, 1-propenyl, 2- propenyl, 1-, 2-, or
  • the alkenyl group can be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group.
  • Alkynyl refers to an alkyl defined as above that has at least two carbon atoms and at least one carbon-carbon triple bond, for example, ethynyl, 1-propynyl, 2-propynyl, 1-, 2-, or 3-butynyl etc., preferably C2-20 alkynyl, more preferably C2-12 alkynyl, and most preferably C2-6 alkynyl.
  • the alkynyl group can be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxo group.
  • Alkylene refers to a saturated linear or branched aliphatic hydrocarbon group, wherein having 2 residues derived by removing two hydrogen atoms from the same carbon atom of the parent alkane or two different carbon atoms.
  • the straight or branched chain group containing 1 to 20 carbon atoms preferably has 1 to 12 carbon atoms, more preferably 1 to 6 carbon atoms.
  • alkylene groups include, but are not limited to, methylene (-CH2-), 1,1-ethylene (-CH(CH3)-), 1,2-ethylene (-CH2CH2)-, 1,1-propylene (- CH(CH 2 CH 3 )-), 1,2- propylene (-CH 2 CH(CH3)-), 1,3-propylene (-CH2CH2CH2-), 1,4- butylidene (-CH2CH2CH2CH2-) etc.
  • the alkylene group can be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of selected from alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxo group.
  • Alkenylene refers to an alkylene defined as above that has at least two carbon atoms and at least one carbon-carbon double bond, preferably C2-20 alkenylene, more preferably C2- 12 alkenylene, and most preferably C2-6 alkenylene.
  • the alkenylene group can be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of selected from alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxo group.
  • Cycloalkyl refers to a saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group having 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably 3 to 8 carbon atoms, and most preferably 5 to 8 carbon atoms or 5 to 6 carbon atoms.
  • Representative examples of monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, etc.
  • Polycyclic cycloalkyl includes a cycloalkyl having a spiro ring, fused ring or bridged ring.
  • “Spiro Cycloalkyl” refers to a 5 to 20 membered polycyclic group with rings connected through one common carbon atom (called a spiro atom), wherein one or more rings can contain one or more, preferably one to three, double bonds, it can be aryl and heteroaryl.
  • a spiro cycloalkyl is 6 to 14 membered, and more preferably 8 to 10 membered.
  • a spiro cycloalkyl is divided into mono-spiro cycloalkyl, di-spiro cycloalkyl, or poly-spiro cycloalkyl, and preferably refers to a mono-spiro cycloalkyl or di-spiro cycloalkyl, more preferably 4-membered/4- membered, 4-membered/5-membered, 4-membered/6-membered, 5 -membered/5 -membered, or 5-membered/6-membered mono-spiro cycloalkyl.
  • Representative examples of spiro cycloalkyl include, but are not limited to the following groups:
  • “Fused Cycloalkyl” refers to a polycyclic group, which is a cycloalkyl attached together with one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from cycloalkyl, heterocyclyl, aryl and heteroaryl in a fused manner. Wherein cycloalkyl, heterocyclyl, aryl and heteroaryl are as defined in the present invention.
  • fused cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic fused cycloalkyl, and preferably refers to a bicyclic or tricyclic fused cycloalkyl, more preferably refers to aryl fused Cvxcycloalkyl, heteroaryl fused Cvxcycloalkyl.
  • fused cycloalkyls include, but are not limited to, the following groups:
  • “Bridged Cycloalkyl” refers to a 5 to 20 membered polycyclic hydrocarbon group, wherein every two rings in the system share two disconnected carbon atoms.
  • the rings can have one or more, preferably one to three, double bonds, but have no completely conjugated pi-electron system.
  • a bridged cycloalkyl is 6 to 14 membered, and more preferably 7 to 10 membered.
  • bridged cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl, and preferably refers to a bicyclic, tricyclic or tetracyclic bridged cycloalkyl, more preferably a bicyclic or tricyclic bridged cycloalkyl.
  • Representative examples of bridged cycloalkyls include, but are not limited to, the following groups:
  • the cycloalkyl can be fused to the ring of an aryl, heteroaryl or heterocyclic alkyl, wherein the ring bound to the parent structure is cycloalkyl.
  • Representative examples include, but are not limited to indanylacetic, tetrahydronaphthalene, benzocycloheptyl and so on.
  • the cycloalkyl is optionally substituted or unsubstituted.
  • the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group.
  • Heterocyclyl refers to a 3 to 20 membered saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group having one or more, preferably one to five, and sometimes more preferably one to three, heteroatoms selected from the group consisting of N, O, and S(0)m (wherein m is 0,1, or 2) as ring atoms, but excluding -0-0-, -O-S- or -S-S- in the ring, the remaining ring atoms being C.
  • heterocyclyl is a 3 to 12 membered having 1 to 4 heteroatoms; more preferably a 3 to 10 membered having 1 to 3 heteroatoms; most preferably a 5 to 8 membered having 1 to 2 heteroatoms.
  • monocyclic heterocyclyls include, but are not limited to, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, sulfo-morpholinyl, homopiperazinyl, and so on.
  • Polycyclic heterocyclyl includes the heterocyclyl having a spiro ring, fused ring or bridged ring.
  • “Spiro heterocyclyl” refers to a 5 to 20 membered polycyclic heterocyclyl with rings connected through one common carbon atom (called a spiro atom), wherein said rings have one or more, preferably one to five, and sometimes more preferably one to three, heteroatoms selected from the group consisting of N, O, and S(0)m (wherein m is 0,1 or 2) as ring atoms, the remaining ring atoms being C, wherein one or more rings can be attached together with one or more, preferably one to three, group(s) selected from cycloalkyl, heterocyclyl, aryl and heteroaryl in a fused manner.
  • cycloalkyl, heterocyclyl, aryl and heteroaryl are as defined in the present invention.
  • a spiro heterocyclyl is 6 to 14 membered.
  • Representative examples of spiro heterocyclyl include, but are not limited to the following groups:
  • “Fused Heterocyclyl” refers to a polycyclic group, which is a heterocyclyl attached together with one or more, preferably one to three, group(s) selected from cycloalkyl, heterocyclyl, aryl and heteroaryl in a fused manner. Wherein cycloalkyl, heterocyclyl, aryl and heteroaryl are as defined in the present invention.
  • fused heterocyclyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclyl, and preferably refers to a bicyclic or tricyclic fused cycloalkyl, more preferably refers to aryl fused 5 to 8-member heterocyclyl, heteroaryl fused 5 to 8-member heterocyclyl.
  • Representative examples of lused heterocyclyl include, but are not limited to, the following groups:
  • “Bridged Heterocyclyl” refers to a 5 to 14 membered polycyclic heterocyclic alkyl group, wherein every two rings in the system share two disconnected atoms, the rings can have one or more, preferably one to three, double bonds, but have no completely conjugated pi-electron system, and the rings have one or more, preferably one to five, and sometimes more preferably one to three, heteroatoms independently selected from the group consisting of N, O, and S (O)m (wherein m is 0, 1, or 2) as ring atoms, the remaining ring atoms being C.
  • a bridged heterocyclyl is 6 to 14 membered, and more preferably 7 to 10 membered.
  • bridged heterocyclyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclyl, and preferably refers to bicyclic, tricyclic or tetracyclic bridged heterocyclyl, more preferably bicyclic or tricyclic bridged heterocyclyl.
  • Representative examples of bridged heterocyclyl include, but are not limited to, the following groups:
  • the ring of said heterocyclyl can be fused to the ring of an aryl, heteroaryl or cycloalkyl, wherein the ring bound to the parent structure is heterocyclyl.
  • Representative examples include, but are not limited to the following groups:
  • the heterocyclyl is optionally substituted or unsubstituted.
  • the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxy group.
  • Aryl refers to a 6 to 14 membered all-carbon monocyclic ring or a polycyclic fused ring (a "fused" ring system means that each ring in the system shares an adjacent pair of carbon atoms with another ring in the system) group, and has a completely conjugated pi- electron system.
  • aryl is 6 to 10 membered, such as phenyl and naphthyl, most preferably phenyl.
  • the aryl can be lused to the ring of heteroaryl, heterocyclyl or cycloalkyl, wherein the ring bound to parent structure is aryl. Representative examples include, but are not limited to, the following groups:
  • group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxy group.
  • Heteroaryl refers to an aryl system having 1 to 4 heteroatoms selected from the group consisting of O, S and N as ring atoms and having 5 to 14 annular atoms.
  • a heteroaryl is 5- to 10- membered, more preferably 5- or 6- membered, for example, thiadiazolyl, pyrazolyl, oxazolyl, oxadiazolyl, imidazolyl, triazolyl, thiazolyl, furyl, thienyl, pyridyl, pyrrolyl, N-alkyl pyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, and the like.
  • the heteroaryl can be fused with the ring of an aryl, heterocyclyl or cycloalkyl, wherein the ring bound to parent structure is heteroaryl. Representative examples include, but are not limited to, the following groups:
  • the heteroaryl group can be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxy group.
  • Alkoxy refers to both an -O-(alkyl) and an -0-(unsubstituted cycloalkyl) group, wherein the alkyl is defined as above. Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
  • the alkoxyl can be substituted or unsubstituted.
  • the substituent is preferably one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxy group.
  • “Bond” refers to a covalent bond using a sign of“—”.
  • Hydroalkyl refers to an alkyl group substituted by a hydroxy group, wherein alkyl is as defined above.
  • Haldroxy refers to an -OH group.
  • Halogen refers to fluoro, chloro, bromo or iodo atoms.
  • Amino refers to a -NH2 group.
  • Cyano refers to a -CN group.
  • Niro refers to a -NO2 group.
  • Carboxyl refers to a -C(0)0H group.
  • “Optional” or “optionally” means that the event or circumstance described subsequently can, but need not, occur, and the description includes the instances in which the event or circumstance may or may not occur.
  • “the heterocyclic group optionally substituted by an alkyl” means that an alkyl group can be, but need not be, present, and the description includes the case of the heterocyclic group being substituted with an alkyl and the heterocyclic group being not substituted with an alkyl.
  • “Substituted” refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3 hydrogen atoms, independently substituted with a corresponding number of substituents. It goes without saying that the substituents exist in their only possible chemical position. The person skilled in the art is able to determine if the substitution is possible or impossible without paying excessive efforts by experiment or theory. For example, the combination of amino or hydroxyl group having free hydrogen and carbon atoms having unsaturated bonds (such as olefinic) may be unstable.
  • A“pharmaceutical composition” refers to a mixture of one or more of the compounds described in the present invention or physiologically/pharmaceutically acceptable salts or prodrugs thereof and other chemical components such as physiologically/pharmaceutically acceptable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism, which is conducive to the absorption of the active ingredient and thus displaying biological activity.
  • “Pharmaceutically acceptable salts” refer to salts of the compounds of the invention, such salts being safe and effective when used in a mammal and have corresponding biological activity.
  • the present invention provides compounds which are in a prodrug form.
  • Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention.
  • prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
  • the compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
  • Unnatural proportions of an isotope may be defined as ranging from the amount found in nature to an amount consisting of 100% of the atom in question.
  • the compounds may incorporate radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 1) or carbon- 14( 14 C), or non-radioactive isotopes, such as deuterium (D) or carbon-13( 13 C).
  • radioactive isotopes such as for example tritium ( 3 H), iodine-125 ( 125 1) or carbon- 14( 14 C), or non-radioactive isotopes, such as deuterium (D) or carbon-13( 13 C).
  • isotopic variations can provide additional utilities to those described elsewhere within this application.
  • isotopic variants of the compounds of the invention may find additional utility, including but not limited to, as diagnostic
  • therapeutically effective amount refers to the administration of an agent to a subject, either alone or as part of a pharmaceutical composition and either in a single dose or as part of a series of doses, in an amount capable of having any detectable, positive effect on any symptom, aspect, or characteristic of a disease, disorder or condition when administered to the subject.
  • the therapeutically effective amount can be ascertained by measuring relevant physiological effects, and it can be adjusted in connection with the dosing regimen and diagnostic analysis of the subject’s condition, and the like.
  • measurement of the serum level of a CD73 inhibitor (or, e.g., a metabolite thereof) at a particular time post- administration may be indicative of whether a therapeutically effective amount has been used.
  • solvate means a physical association of a compound of this invention with one or more, preferably one to three, solvent molecules, whether organic orinorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example, when one or more, preferably one to three, solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are generally known in the art.
  • Prodrug refers to compounds that can be transformed in vivo to yield the active parent compound under physiological conditions, such as through hydrolysis in blood.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
  • the term “treat”, “treating”, “treatment”, or the like refers to: (i) inhibiting the disease, disorder, or condition, i.e., arresting its development; and (ii) relieving the disease, disorder, orcondition, i.e., causing regression of the disease, disorder, and/or condition.
  • thecompounds of present invention may be used for their prophylactic effects in preventing a disease, disorder or condition from occurring in a subject that may be predisposed to the disease, disorder, and/or condition but has not yet been diagnosed as having it.
  • subject or“patient” refers to a mammalian animal.
  • mammal or“mammalian animal” includes, but is not limited to, humans, dogs, cats, horses, pigs, cows, monkeys, rabbits and mice.
  • the preferred mammals are humans.
  • Step 1 reacting with formula (IV- 1) and formula (IV-2) under alkaline condition (such as DBU) to obtain formula (IV-3);
  • Step 2 reacting with formula (IV-3) and 2,2-dimethoxypropane under acidic condition (such as p-toluenesulfonic acid), then neutralize the reaction mixture to obtain formula (IV-4);
  • acidic condition such as p-toluenesulfonic acid
  • Step 3 reacting with formula (IV-4) and methyl enebis(phosphonic dichloride) with the additive (such as N-methylimidazole) to obtain formula (IV-5);
  • Step 4 reacting to remove the protecting group of acetal with a compound of formula (IV-5) under acidic condition (such as TFA) to obtain the compound of formula (IV-6);
  • X is halogen, preferably chlorine
  • the additive includes, but is not limited to, pyridine, 2,4,6-collidine, 1,4- diazabicyclo[2.2.2]octane (DABCO), 4-(dimethylamino)pyridine, N-methylimidazole.
  • DABCO 1,4- diazabicyclo[2.2.2]octane
  • 4-(dimethylamino)pyridine N-methylimidazole.
  • Agents that provide acidic conditions include, but are not limited to, hydrogen chloride, hydrogen chloride 1,4-dioxane solution, trimethylsilyl bromide (TMSBr), ammonium chloride, trifluoro acetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, -toluenesul Ionic acid, p- toluenesulfonic acid monohydrate and TMSOTf.
  • TMSBr trimethylsilyl bromide
  • reaction is preferably in solvent, wherein solvent used herein includes, but is not limited to, acetic acid, methanol, ethanol, toluene, acetone, tetrahydrofuran, dichloromethane, dimethylsulfoxide, 1,4-dioxane, water, N, V-dimethylformamide, trimethylphosphate, methyl tert-butyl ether and the mixture thereof.
  • solvent used herein includes, but is not limited to, acetic acid, methanol, ethanol, toluene, acetone, tetrahydrofuran, dichloromethane, dimethylsulfoxide, 1,4-dioxane, water, N, V-dimethylformamide, trimethylphosphate, methyl tert-butyl ether and the mixture thereof.
  • the structure of a compound is determined by mass spectrometry (MS) and or nuclear magnetic resonance (NMR). NMR shift (d) is given in units of 10 6 (ppm).
  • MS mass spectrum
  • the NMR measurement was performed on a Bruker AVANCE-400 and 500 Ultrashield nuclear magnetic resonance spectrometer.
  • the solvents were deuterated dimethylsulfoxide ( D SO-iL), deuterated chloroform (CDCE) and deuterated methanol (CD 3 OD) Silane (TMS).
  • HPLC was performed using a Shimadzu OPTION BOX-L high pressure liquid phase Chromatograph (Gimini 5um NX-C18 100x21.2mm column).
  • TLC Thin-layer chromatography
  • Known starting materials of the present invention may be synthesized according to methods known in the art or may be purchased from Acros Organics, Sigma-Aldrich Chemical Company, AstaTech and other companies. Unless otherwise specified in the examples, the reaction can be carried out under an argon atmosphere or a nitrogen atmosphere.
  • Argon or nitrogen atmosphere refers to the reaction flask connected to a volume of about 1 L argon or nitrogen balloon.
  • Hydrogen atmosphere refers to the reaction bottle connected to a volume of about 1 L hydrogen balloon.
  • Hydrogenation reaction is usually evacuated, filled with hydrogen, repeated 3 times.
  • the microwave reaction used a CEM Disco ver-S 908860 microwave reactor.
  • reaction temperature is room temperature and is 20 0 C to 30 0 C.
  • TLC thin layer chromatography
  • developing solvent for the reaction a column chromatography eluent for purifying compound
  • developing system for thin-layer chromatography include: A: dichloromethane / methanol system, B: n- hexane / ethyl acetate system, C: dichloromethane / ethyl acetate system.
  • the volume ratio of the solvents is adjusted according to the polarity of the compound. A small amount of triethylamine and acetic acid and other alkaline or acidic reagents can be used for adjustment.
  • TEA is triethylamine
  • DBU is l,8-diazabicyclo[5.4.0]undec-7-ene
  • TMSOTf is trimethylsilyl trifluoromethanesulfonate
  • THF is tetrahydrofuran
  • DCM is dichloromathene
  • MTBE is methyl tert-butyl ether
  • TEAC triethylammonium bicarbonate
  • NMI N-methylimidazole
  • DMSO dimethyl sulfoxide
  • Compound examples 2-9 can be prepared with the similar method to the compound example 1.
  • CD73 enzyme an ecto-5 '-Nucleotidase, converts extracellular nucleoside-5 '-monophosphates to nucleosides, with AMP or CMP as the preferred substrate.
  • recombinant human CD73 expressed from a Chinese hamster ovary cell line (R&D Systems) was used to convert cytidine monophosphate (CMP) to cytidine and phosphate.
  • CD73 enzyme was pre-incubated with compounds for 2 hours. The amount of phosphate was then measured by Malachite Green Phosphate Detection Kit. The experimental method is summarized as follows:
  • the highest concentration of compound was 125 pM and the DMSO concentration was 1.25%.
  • 3 pL of CMP dissolved in assay buffer was added to each reaction.
  • the final CMP concentration was 45 pM.
  • the reaction was incubated at 37°C for 15 minutes.
  • 3 pL of Malachite Green Reagent A was added to each reaction. Spin the plate briefly in centrifuge for 30 seconds. After incubation for additional 10 minutes at room temperature, 3 pL of malachite green Reagent B was added to each reaction. Spin the plate briefly in centrifuge for 30 seconds. After 20 minutes of incubation at room temperature, the signal was read on a TEC AN reader at OD620.
  • reaction containing CD73 enzyme, substrate CMP and DMSO (no compound) is used as an assay positive control while the reaction containing substrate CMP and DMSO without the CD73 enzyme as an assay negative control.
  • IC50 values were calculated by plotting the logarithm of the compound concentration and the percent inhibition using the appropriate program in GraphPad Prism.
  • biochemical inhibitions of CD73 enzymatic activities by compound 1 of the present invention was determined by the assay described above, and the resulting IC50 value was 0.56 nM.
  • the compounds of the present invention have a significant inhibitory effect on CD73 enzyme activity in vitro.
  • CD73 enzyme an ecto-5 '-Nucleotidase, converts extracellular nucleoside-5 '-monophosphates to nucleosides, with AMP or CMP as the preferred substrate.
  • membrane-bound CD73 enzyme activity on the surface of human melanoma A375 cells was used to convert cytidine monophosphate (CMP) to cytidine and phosphate in presence of compounds and CMP.
  • CMP cytidine monophosphate
  • the amount of phosphate was then measured by Malachite Green Phosphate Detection Kit. The experimental method is summarized as follows:
  • A375 cell line (ATCC, Cat#, CRL-1619)
  • A375 cells are maintained using DMEM medium containing 10% FBS and 1% Penicillin-Streptomycin. A day before the assay, use trypsin to harvest A375 cells and perform a cell count. Seed cells to each well of the 96- well plate in 100 pL media (2500 cells/well). Next day, prepare for assay buffer containing 20 mM HEPES, 137 mM NaCl, 5.4 mM KC1, 1.3 mM CaCh, 4.2mM NaHC03 and 1 mg/mL glucose. Warm the buffer in 37°C water bath.
  • the compounds of the present invention have a significant inhibitory effect on CD73 enzyme activity in A375 cells.
  • PBMCs human peripheral blood mononuclear cells
  • Lymphocyte culture medium (Zenbio, Cat# LYMPH- 1)
  • CD3 and CD28 antibody beads (Fisher Scientific, Cat# 1 161D)
  • HTRF human IFNy Cytokine kit (Cisbio, human IFNy Cat# 62HIFNGPEH)
  • Lymphocyte medium and TexMACS medium were incubated in a water bath at 37 0 C. 10 mL of incubated lymphocyte medium was added to a 50 mL conical tube. The cells were quickly thawed in a 37 0 C water bath and transferred to a 50 ml tube and the tube gently swirled. The cell suspension was centrifuged at 1100 rpm for 10 minutes at room temperature. The supernatant was removed, and the cell pellet gently resuspended in 10 mL of TexMACS medium. The cells were counted to make 5 x 10 5 cells / ml.
  • PBMCs seeds 100 pL of 5 x 10 5 cells / ml of PBMCs seeds, inserted into a 96-well plate (cell density of 50,000 cells / well), and the most outer edge wells of the 96-well plate were filled with water and were not used for the test.
  • the CD73 substrate AMP was added to human PBMC cell culture in order to generate more adenosine via CD73 enzyme.
  • Compounds were diluted to 40 pM in TexMACS media.
  • the compound of the invention can stimulate the production of IFNy cytokines and thus have a significant modulating effect on the cellular immune functions.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This application discloses CD73 inhibitors of the general formula (I) and analogs thereof, pharmaceutical compositions containing these compounds, methods of preparing them, and use of these compounds as therapeutic agents for the treatment of diseases or conditions associated with CD73 activity, such as various cancers. Formula (I).

Description

CD73 INHIBITORS AND THERAPEUTIC USES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority under 35 U.S.C. § 119(e) to United States Provisional Patent Application No. 62/826,441, filed on March 29, 2019, the disclosure of which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
The present invention relates to novel nucleoside and nucleotide analogs, pharmaceutical compositions containing these compounds useful as CD73 inhibitors for the treatment of cancer and other diseases mediated by CD73, and methods of preparing these compounds and compositions.
BACKGROUND OF THE INVENTION
CD73 (also known as ecto-5’ -nucleotidase) is a cell surface enzyme through a glycosyl phosphatidylinositol linkage to anchor onto the cell membrane and is expressed in different tissues, especially in the colon, kidney, brain, liver, heart, lung, spleen, lymph nodes, and bone marrow (Antonioli, L., et al., Trends Mol. Med., 2013, 19, 355-367). The enzymatic activity of CD73 is to catalyze the extracellular dephosphorylation of nucleoside monophosphates to their corresponding nucleosides (e.g., 5-AMP to adenosine). CD73 exerts physiological influences mainly via its enzymatic nucleoside products, particularly adenosine in extracellular space, including epithelial ion and fluid transportation, tissue barrier function control, adaptation to hypoxia, ischemic preconditioning, anti-inflammation, and immune suppression signaling (Colgan, S.P., et al., Purinergic Signaling, 2006, 2, 351-360).
As a ubiquitous extracellular signaling molecule with neuromodulating properties, adenosine produces a broad range of physiological responses in the human body via interaction with adenosine receptors (receptor subtypes: Al , A2A , A2B , and A3), including the vasodilation and atrioventricular conduction suppression properties in the cardiovascular system; the sedative, local neuronal excitability inhibition, anticonvulsant, and neuroprotective effects in the central nervous system (Dunwiddie, T.V., et al., Annu. Rev. Neurosci. 2001, 24, 31-55); the bronchoconstriction effects in the respiratory system (Pauwels, R., et al., Drug Development Research, 1993, 28, 318-321); and the mediation of immune/inflammatory responses in the immune system (Hasko, G, et al.,“A Key Link between Metabolism and Brain Activity”, 2013, 233-251). CD73 is broadly expressed in many cancer types (Antonioli, L. et al., Trends in Cancer, 2016, 2(2), 95-109) and associated with many cancer types’ poor prognosis (Allard, D., et al., Immunotherapy, 2016, 8(2), 145-163). CD73 promotes cancer metastasis (Yang, Q., et al., Pathol. Oncol. Res., 2013, 19, 811-814) and chemoresistance (Loi, S., et al., PNAS, 2013, 110(27), 11091-11096). In the immune system, CD73 is found on the surface of macrophages, lymphocytes, regulatory T cells, myeloid-derived suppressor cells (MDSCs), and dendritic cells. The extracellular adenosine, mainly produced by CD73, can chronically accumulate in tumor microenvironment, activating adenosine receptors, promoting tumor- inducing mononuclear phagocytes, deregulating anti-tumor T cell response, expanding MDSCs population, triggering immune suppression and favoring the escape of cancer cells from immune surveillance, hence promoting cancer transformation and growth (Antonioli, L., et al., Nature Reviews Cancer, 2013, 13, 842-857).
Because of a wide range of physiological functions of adenosine in human body, CD73 inhibitors can be used to enhance immune response and treat adenosine and adenosine receptor related diseases or disorders, including neurological, neurodegenerative and CNS disorders and diseases, depression and Parkinson’s disease, cerebral and cardiac ischaemic diseases, sleep disorders, fibrosis, immune and inflammatory disease, and cancer.
At present, small-molecule drug candidates as CD73 inhibitors are mostly at the discovery stage, which are represented in several published patent applications, such as WO2015164573, WO2017098421, W02017120508, WO2017153952, US20170044203,
WO2018049145, WO2018067424, and WO2018094148, but no small-molecule compounds have been put into clinical research yet. Therefore, there is a need to identify and develop CD73 new inhibitors that will provide new therapeutic approaches to limit tumor progression and metastasis, increase the efficacy of anti-cancer therapy, and treat cancer by decreasing extracellular adenosine level in tumor microenvironment to resume immune cells effective response against cancer cells.
SUMMARY OF THE INVENTION
The present invention, in one aspect, provides a compound of formula (I) having the following structure:
Figure imgf000004_0001
or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof,
wherein:
W is selected from the group consisting of O, S, NH, NRa and C(Rb)2, wherein Ra is alkyl, and Rb at each occurrence is independently selected from the group consisting of hydrogen, halogen, alkyl and alkenyl ;
G1 and G2 are each independently N or CRC, wherein Rc is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, hydroxy, amino, nitro, cyano, cycloalkyl, heterocyclyl, aryl and heteroaryl;
G3 and G4 are each independently selected from the group consisting of C, CH, CH2, N, NH, O, S and S02;
ring A is selected from the group consisting of Cvxcycloalkyl, 5 to 8-member heterocyclyl, aryl fused Cs-scycloalkyl, heteroaryl fused Cs-scycloalkyl, aryl fused 5 to 8- member heterocyclyl, heteroaryl fused 5 to 8-member heterocyclyl;
R1, R2, R3 and R4 are each independently selected from the group consisting of hydroxy, hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxyalkyl, cyano, amino, azide group and OR10;
or R1 and R2 together with the carbon atom they attached to form a cycloalkyl or heterocyclyl, wherein the heterocyclyl contains 1 to 2 heteroatoms which are the same or different from N, O and S, and wherein the cycloalkyl and heterocyclyl are each optionally substituted by one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl and heterocyclyl;
R5 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro and azide; R6 is selected from the group consisting of hydrogen and optionally substituted Ci-G, alkyl;
R7 at each occurrence is independently selected from the group consisting of hydrogen, alkyl, aryl, -C(RmRn)-aryl, -C(RmRn)-0-C(0)0Rd, -C(RmRn)-0-C(0)Rd, - C(RmRn)C(0)ORd, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl is optionally substituted by one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of cycloalkyl, heterocyclyl, aryl and heteroaryl; optionally, two R7 are combined to form a 5- to 7- membered heterocyclic ring;
Rm and Rn are each independently selected from the group consisting of H, D, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydro xyalkyl and amino;
Rd is selected from the group consisting of hydrogen, alkyl and alkoxy;
R8 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, azide, cycloalkyl and heterocyclyl;
R9 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, azide, cycloalkyl and heterocyclyl;
R10 is selected from the group consisting of -C(0)Rn, -C(0)ORn, -S(0)2Rn, and - P(0)(0R7)2;
R11 is selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, hydroxyl and hydroxyalkyl;
X is O;
m is 1 , 2 or 3 ; and
s is 0, 1, 2, 3 or 4.
In another aspect, the present invention provides a pharmaceutical composition, comprising a compound of formula (I), in any embodiment disclosed herein, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier.
In another aspect, the present invention provides a method of treating or preventing a CD73 -mediated disease or condition, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I), in any embodiment disclosed herein, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof.
In another aspect, the present invention relates to use of a compound of formula (I), in any embodiment disclosed herein, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for treatment or prevention of a CD73 -mediated disease or condition.
The CD73-mediated diseases or conditions include, without limitation, tumors, cancers, immune-related diseases, inflammatory-related diseases, nervous system diseases, neurodegenerative and central nervous system diseases, depression, Parkinson's disease, ischemic diseases of the brain and heart, sleep disorders, and fibrosis, or the like.
Other aspects and advantages of the present invention will be better appreciated in view of the following detailed description, examples, and claims.
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to novel nucleoside and nucleotide analogs useful as
CD73 inhibitors. In one aspect, the present invention provides a compound of formula (I) having the following structure, as defined above:
Figure imgf000006_0001
or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof.
In one embodiment of the invention, the compound of formula (I) is selected from a compound of formula (II):
Figure imgf000007_0001
or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof,
wherein:
X, W, G1, G2, G3, G4, ring A, R1 to R9, m and s are as defined in formula (I).
In one embodiment of the invention, the compound of formula (I) is selected from a compound of formula (III) :
Figure imgf000007_0002
or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof,
wherein:
G1, G2, G3, G4, ring A, R1, R3, R7 to R9, m and s are as defined in formula (I).
In one embodiment of the invention, the compound of formula (I) is selected from a compound of formula
Figure imgf000007_0003
or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof,
wherein:
G3, G4, ring A, R7 to R9, m and s are as defined in formula (I).
In one embodiment, the invention provides a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof,
Figure imgf000008_0001
R9 and s are as defined in formula (I).
In one embodiment, the invention provides a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R7 is selected from the group consisting of hydrogen, alkyl, and -C(RmRn)-0-C(0)0Rd.
In one embodiment, the invention provides a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R8 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl and haloalkoxy.
In one embodiment, the invention provides a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein R9 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl and haloalkoxy. Exemplified compounds of the invention include, but are not limited to:
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
The present invention also provides a pharmaceutical composition, comprising a therapeutically effective amount of a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, together with one or more pharmaceutically acceptable carriers, diluents or excipients.
The present invention also relates to a method for inhibiting CD73, comprising contacting a biological sample comprising CD73 with a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof.
The present invention also relates to a method for treating a CD73 -mediated disease or condition, comprising a step of administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof.
The present invention also relates to a method for treating a disease or condition, comprising administering to a subject in need thereof a therapeutically effective amount of the compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof, wherein the disease or condition is selected from the group consisting of tumor, cancer, immune-related disease, inflammatory-related diseases, nervous system, neurodegenerative and central nervous system diseases, depression, Parkinson's disease, ischemic diseases of the brain and heart, sleep disorders and fibrosis.
The cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, gastric cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, metrocarcinoma, endometriosis, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder cancer, gallbladder cancer, cholangiocarcinoma, chorionic epithelial cancer, and pediatric tumor.
In another aspect, the present invention also relates to use of a compound of formula (I), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition thereof, in the preparation of a medicament for treating a CD73 -mediated disease or condition.
The CD73-mediated disease or condition includes, but is not limited to, tumor, cancer, immune-related disease, inflammatory-related diseases, nervous system, neurodegenerative and central nervous system diseases, depression, Parkinson's disease, ischemic diseases of the brain and heart, sleep disorders and fibrosis.
The compositions of this invention can be formulated by conventional methods using one or more pharmaceutically acceptable carriers. Thus, the active compounds of this invention can be formulated as various dosage forms for oral, buccal, intranasal, parenteral (e.g., intravenous, intramuscular or subcutaneous), rectal administration, inhalation or insufflation administration. The compounds of this invention can also be formulated as sustained release dosage forms.
Suitable dosage forms include, but are not limited to, a tablet, troche, lozenge, aqueous or oily suspension, dispersible powder or granule, emulsion, hard or soft capsule, or syrup or elixir. Oral compositions can be prepared according to any known method in the art for the preparation of pharmaceutical compositions. Such compositions can contain one or more additives selected from the group consisting of sweeteners, flavoring agents, colorants and preservatives, in order to provide a pleasing and palatable pharmaceutical preparation. Tablets contain the active ingredient and nontoxic pharmaceutically acceptable excipients suitable for the manufacture of tablets. These excipients can be inert excipients, granulating agents, disintegrating agents, and lubricants. The tablet can be uncoated or coated by means of a known technique to mask the taste of the drug or delay the disintegration and absorption of the drug in the gastrointestinal tract, thereby providing sustained release over an extended period. For example, water soluble taste masking materials can be used.
Oral formulations can also be provided as soft gelatin capsules in which the active ingredient is mixed with an inert solid diluent, or the active ingredient is mixed with a water soluble carrier.
An aqueous suspension contains the active ingredient in admixture with excipients suitable for the manufacture of an aqueous suspension. Such excipients are suspending agents, dispersants or humectants, and can be naturally occurring phospholipids. The aqueous suspension can also contain one or more preservatives, one or more colorants, one or more flavoring agents, and one or more sweeteners.
An oil suspension can be formulated by suspending the active ingredient in a vegetable oil, or in a mineral oil. The oil suspension can contain a thickener. The aforementioned sweeteners and flavoring agents can be added to provide a palatable preparation. These compositions can be preserved by adding an antioxidant.
The active ingredient and the dispersants or wetting agents, suspending agent or one or more preservatives can be prepared as a dispersible powder or granule suitable for the preparation of an aqueous suspension by adding water. Suitable dispersants or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, such as sweeteners, flavoring agents and colorants, can also be added. These compositions can be preserved by adding an antioxidant such as ascorbic acid.
The present pharmaceutical composition can also be in the form of an oil-in-water emulsion. The oil phase can be a vegetable oil, or a mineral oil, or mixture thereof. Suitable emulsifying agents can be naturally occurring phospholipids. Sweeteners can be used. Such formulations can also contain moderators, preservatives, colorants and antioxidants.
The pharmaceutical composition can be in the form of a sterile injectable aqueous solution. The acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. The sterile injectable preparation can also be a sterile injectable oil-in-water microemulsion in which the active ingredient is dissolved in the oil phase. The injectable solution or microemulsion can be introduced into an individual’s bloodstream by local bolus injection. Alternatively, it can be advantageous to administer the solution or microemulsion in such a way as to maintain a constant circulating concentration of the present compound. In order to maintain such a constant concentration, a continuous intravenous delivery device can be utilized. An example of such a device is Deltec CADD- PLUS. TM. 5400 intravenous injection pump. The pharmaceutical composition can be in the form of a sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration· Such a suspension can be formulated with suitable dispersants or wetting agents and suspending agents as described above according to known techniques. The sterile injectable preparation can also be a sterile injectable solution or suspension prepared in a nontoxic parenterally acceptable diluent or solvent. Moreover, sterile fixed oils can easily be used as a solvent or suspending medium, and fatty acids can also be used to prepare injections.
The present compound can be administered in the form of a suppository for rectal administration· These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures, but liquid in the rectum, thereby melting in the rectum to release the drug.
For buccal administration, the compositions can be formulated as tablets or lozenges by conventional means.
For intranasal administration or administration by inhalation, the active compounds of the present invention are conveniently delivered in the form of a solution or suspension released from a pump spray container that is squeezed or pumped by the patient, or as an aerosol spray released from a pressurized container or nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit can be determined by providing a valve to deliver a metered amount. The pressurized container or nebulizer can contain a solution or suspension of the active compound. Capsules or cartridges (for example, made from gelatin) for use in an inhaler or insufflator can be formulated containing a powder mix of the present invention and a suitable powder base such as lactose or starch.
It is well known to those skilled in the art that the dosage of a drug depends on a variety of factors, including but not limited to, the following factors: activity of the specific compound, age, weight, general health, behavior, diet of the patient, administration time, administration route, excretion rate, drug combination and the like. In addition, the best treatment, such as treatment mode, daily dose of the compound of formula (I) or the type of pharmaceutically acceptable salt thereof can be verified by traditional therapeutic regimens.
Unless otherwise stated, the terms used in the specification and claims have the meanings described below.
“Alkyl” refers to a saturated aliphatic hydrocarbon group including C1-C20 straight chain and branched chain groups. Preferably an alkyl group is an alkyl having 1 to 12 carbon atoms. Representative examples include, but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1 -dimethyl propyl, 1,2-dimethyl propyl, 2,2-dimethyl propyl, 1-ethyl propyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl- 2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2- dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4- methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5- methylhexyl, 2,3-dimethylpentyl, 2,4-dimethylpentyl, 2,2-dimethylpentyl, 3,3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2,3-dimethylhexyl, 2,4-dimethylhexyl, 2,5- dimethylhexyl, 2,2-dimethylhexyl, 3,3-dimethylhexyl, 4,4-dimethylhexyl, 2-ethylhexyl, 3- ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-
2-ethylhexyl, 2-methyl-3-ethylhexyl, 2,2-diethylpentyl, n-decyl, 3,3-diethylhexyl, 2,2- diethylhexyl, and the isomers of branched chain thereof. More preferably an alkyl group is a lower alkyl having 1 to 6 carbon atoms. Representative examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n- pentyl, 1,1- dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3- methylbutyl, n-hexyl, l-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2- dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3- methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, etc. The alkyl group can be substituted or unsubstituted. When substituted, the substituent group(s) can be substituted at any available connection point, preferably the substituent group(s) is one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group.
“Alkenyl” refers to an alkyl defined as above that has at least two carbon atoms and at least one carbon-carbon double bond, for example, vinyl, 1-propenyl, 2- propenyl, 1-, 2-, or
3-butenyl, etc. preferably C2-20 alkenyl, more preferably C2-12 alkenyl, and most preferably C2-6 alkenyl. The alkenyl group can be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group. “Alkynyl” refers to an alkyl defined as above that has at least two carbon atoms and at least one carbon-carbon triple bond, for example, ethynyl, 1-propynyl, 2-propynyl, 1-, 2-, or 3-butynyl etc., preferably C2-20 alkynyl, more preferably C2-12 alkynyl, and most preferably C2-6 alkynyl. The alkynyl group can be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxo group.
“Alkylene” refers to a saturated linear or branched aliphatic hydrocarbon group, wherein having 2 residues derived by removing two hydrogen atoms from the same carbon atom of the parent alkane or two different carbon atoms. The straight or branched chain group containing 1 to 20 carbon atoms, preferably has 1 to 12 carbon atoms, more preferably 1 to 6 carbon atoms. Non-limiting examples of alkylene groups include, but are not limited to, methylene (-CH2-), 1,1-ethylene (-CH(CH3)-), 1,2-ethylene (-CH2CH2)-, 1,1-propylene (- CH(CH2CH3)-), 1,2- propylene (-CH2CH(CH3)-), 1,3-propylene (-CH2CH2CH2-), 1,4- butylidene (-CH2CH2CH2CH2-) etc. The alkylene group can be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of selected from alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxo group.
“Alkenylene” refers to an alkylene defined as above that has at least two carbon atoms and at least one carbon-carbon double bond, preferably C2-20 alkenylene, more preferably C2- 12 alkenylene, and most preferably C2-6 alkenylene. Non-limiting examples of alkenylene groups include, but are not limited to, -CH=CH-, -CH=CHCH2-, -CItCHCtbCtb-, - CH2CH=CHCH2- etc. The alkenylene group can be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of selected from alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxo group.
“Cycloalkyl” refers to a saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group having 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably 3 to 8 carbon atoms, and most preferably 5 to 8 carbon atoms or 5 to 6 carbon atoms. Representative examples of monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, etc. Polycyclic cycloalkyl includes a cycloalkyl having a spiro ring, fused ring or bridged ring.
“Spiro Cycloalkyl” refers to a 5 to 20 membered polycyclic group with rings connected through one common carbon atom (called a spiro atom), wherein one or more rings can contain one or more, preferably one to three, double bonds, it can be aryl and heteroaryl. Preferably a spiro cycloalkyl is 6 to 14 membered, and more preferably 8 to 10 membered. According to the number of common spiro atoms, a spiro cycloalkyl is divided into mono-spiro cycloalkyl, di-spiro cycloalkyl, or poly-spiro cycloalkyl, and preferably refers to a mono-spiro cycloalkyl or di-spiro cycloalkyl, more preferably 4-membered/4- membered, 4-membered/5-membered, 4-membered/6-membered, 5 -membered/5 -membered, or 5-membered/6-membered mono-spiro cycloalkyl. Representative examples of spiro cycloalkyl include, but are not limited to the following groups:
Figure imgf000018_0001
“Fused Cycloalkyl” refers to a polycyclic group, which is a cycloalkyl attached together with one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from cycloalkyl, heterocyclyl, aryl and heteroaryl in a fused manner. Wherein cycloalkyl, heterocyclyl, aryl and heteroaryl are as defined in the present invention. According to the number of membered rings, fused cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic fused cycloalkyl, and preferably refers to a bicyclic or tricyclic fused cycloalkyl, more preferably refers to aryl fused Cvxcycloalkyl, heteroaryl fused Cvxcycloalkyl. 4-membered heterocyclyl fused C5-8 cycloalkyl, 5 -membered heterocyclyl fused C5-8 cycloalkyl, Ce cycloalkyl fused C5-8 cycloalkyl or C5 cycloalkyl fused C5-8 cycloalkyl, Representative examples of fused cycloalkyls include, but are not limited to, the following groups:
Figure imgf000019_0001
“Bridged Cycloalkyl” refers to a 5 to 20 membered polycyclic hydrocarbon group, wherein every two rings in the system share two disconnected carbon atoms. The rings can have one or more, preferably one to three, double bonds, but have no completely conjugated pi-electron system. Preferably, a bridged cycloalkyl is 6 to 14 membered, and more preferably 7 to 10 membered. According to the number of membered rings, bridged cycloalkyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl, and preferably refers to a bicyclic, tricyclic or tetracyclic bridged cycloalkyl, more preferably a bicyclic or tricyclic bridged cycloalkyl. Representative examples of bridged cycloalkyls include, but are not limited to, the following groups:
Figure imgf000019_0002
The cycloalkyl can be fused to the ring of an aryl, heteroaryl or heterocyclic alkyl, wherein the ring bound to the parent structure is cycloalkyl. Representative examples include, but are not limited to indanylacetic, tetrahydronaphthalene, benzocycloheptyl and so on. The cycloalkyl is optionally substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, halogen, alkoxy, alkenyl, alkynyl, alkylsulfo, alkylamino, thiol, hydroxy, nitro, cyano, amino, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic, cycloalkylthio, heterocylic alkylthio and oxo group.
“Heterocyclyl” refers to a 3 to 20 membered saturated and/or partially unsaturated monocyclic or polycyclic hydrocarbon group having one or more, preferably one to five, and sometimes more preferably one to three, heteroatoms selected from the group consisting of N, O, and S(0)m (wherein m is 0,1, or 2) as ring atoms, but excluding -0-0-, -O-S- or -S-S- in the ring, the remaining ring atoms being C. Preferably, heterocyclyl is a 3 to 12 membered having 1 to 4 heteroatoms; more preferably a 3 to 10 membered having 1 to 3 heteroatoms; most preferably a 5 to 8 membered having 1 to 2 heteroatoms. Representative examples of monocyclic heterocyclyls include, but are not limited to, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, sulfo-morpholinyl, homopiperazinyl, and so on. Polycyclic heterocyclyl includes the heterocyclyl having a spiro ring, fused ring or bridged ring.
“Spiro heterocyclyl” refers to a 5 to 20 membered polycyclic heterocyclyl with rings connected through one common carbon atom (called a spiro atom), wherein said rings have one or more, preferably one to five, and sometimes more preferably one to three, heteroatoms selected from the group consisting of N, O, and S(0)m (wherein m is 0,1 or 2) as ring atoms, the remaining ring atoms being C, wherein one or more rings can be attached together with one or more, preferably one to three, group(s) selected from cycloalkyl, heterocyclyl, aryl and heteroaryl in a fused manner. Wherein cycloalkyl, heterocyclyl, aryl and heteroaryl are as defined in the present invention. Preferably a spiro heterocyclyl is 6 to 14 membered. Representative examples of spiro heterocyclyl include, but are not limited to the following groups:
Figure imgf000020_0001
“Fused Heterocyclyl” refers to a polycyclic group, which is a heterocyclyl attached together with one or more, preferably one to three, group(s) selected from cycloalkyl, heterocyclyl, aryl and heteroaryl in a fused manner. Wherein cycloalkyl, heterocyclyl, aryl and heteroaryl are as defined in the present invention. According to the number of membered rings, fused heterocyclyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclyl, and preferably refers to a bicyclic or tricyclic fused cycloalkyl, more preferably refers to aryl fused 5 to 8-member heterocyclyl, heteroaryl fused 5 to 8-member heterocyclyl. C5-8 cycloalkyl fused 4-membered heterocyclyl, C5-8 cycloalkyl fused 5 -membered heterocyclyl, C5-8 cycloalkyl fused 6-member heterocyclyl. Representative examples of lused heterocyclyl include, but are not limited to, the following groups:
Figure imgf000021_0001
“Bridged Heterocyclyl” refers to a 5 to 14 membered polycyclic heterocyclic alkyl group, wherein every two rings in the system share two disconnected atoms, the rings can have one or more, preferably one to three, double bonds, but have no completely conjugated pi-electron system, and the rings have one or more, preferably one to five, and sometimes more preferably one to three, heteroatoms independently selected from the group consisting of N, O, and S (O)m (wherein m is 0, 1, or 2) as ring atoms, the remaining ring atoms being C. Preferably a bridged heterocyclyl is 6 to 14 membered, and more preferably 7 to 10 membered. According to the number of membered rings, bridged heterocyclyl is divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclyl, and preferably refers to bicyclic, tricyclic or tetracyclic bridged heterocyclyl, more preferably bicyclic or tricyclic bridged heterocyclyl. Representative examples of bridged heterocyclyl include, but are not limited to, the following groups:
Figure imgf000021_0002
The ring of said heterocyclyl can be fused to the ring of an aryl, heteroaryl or cycloalkyl, wherein the ring bound to the parent structure is heterocyclyl. Representative examples include, but are not limited to the following groups:
Figure imgf000021_0003
The heterocyclyl is optionally substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, group(s) independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxy group.
“Aryl” refers to a 6 to 14 membered all-carbon monocyclic ring or a polycyclic fused ring (a "fused" ring system means that each ring in the system shares an adjacent pair of carbon atoms with another ring in the system) group, and has a completely conjugated pi- electron system. Preferably aryl is 6 to 10 membered, such as phenyl and naphthyl, most preferably phenyl. The aryl can be lused to the ring of heteroaryl, heterocyclyl or cycloalkyl, wherein the ring bound to parent structure is aryl. Representative examples include, but are not limited to, the following groups:
Figure imgf000022_0001
group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxy group.
“Heteroaryl” refers to an aryl system having 1 to 4 heteroatoms selected from the group consisting of O, S and N as ring atoms and having 5 to 14 annular atoms. Preferably a heteroaryl is 5- to 10- membered, more preferably 5- or 6- membered, for example, thiadiazolyl, pyrazolyl, oxazolyl, oxadiazolyl, imidazolyl, triazolyl, thiazolyl, furyl, thienyl, pyridyl, pyrrolyl, N-alkyl pyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, and the like. The heteroaryl can be fused with the ring of an aryl, heterocyclyl or cycloalkyl, wherein the ring bound to parent structure is heteroaryl. Representative examples include, but are not limited to, the following groups:
Figure imgf000023_0001
The heteroaryl group can be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxy group.
“Alkoxy” refers to both an -O-(alkyl) and an -0-(unsubstituted cycloalkyl) group, wherein the alkyl is defined as above. Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like. The alkoxyl can be substituted or unsubstituted. When substituted, the substituent is preferably one or more, preferably one to five, and sometimes more preferably one to three, groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylsulfo, alkylamino, halogen, thiol, hydroxy, nitro, cyano, cycloalkyl, heterocyclic alkyl, aryl, heteroaryl, cycloalkoxyl, heterocylic alkoxyl, cycloalkylthio, heterocylic alkylthio and oxy group.
“Bond” refers to a covalent bond using a sign of“—”.
"Hydroxyalkyl" refers to an alkyl group substituted by a hydroxy group, wherein alkyl is as defined above.
“Hydroxy” refers to an -OH group.
“Halogen” refers to fluoro, chloro, bromo or iodo atoms.
‘Amino” refers to a -NH2 group.
‘Cyano” refers to a -CN group.
‘Nitro” refers to a -NO2 group.
Oxo group” refers to a =0 group.
‘Carboxyl” refers to a -C(0)0H group.
“Optional” or “optionally” means that the event or circumstance described subsequently can, but need not, occur, and the description includes the instances in which the event or circumstance may or may not occur. For example,“the heterocyclic group optionally substituted by an alkyl” means that an alkyl group can be, but need not be, present, and the description includes the case of the heterocyclic group being substituted with an alkyl and the heterocyclic group being not substituted with an alkyl.
“Substituted” refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3 hydrogen atoms, independently substituted with a corresponding number of substituents. It goes without saying that the substituents exist in their only possible chemical position. The person skilled in the art is able to determine if the substitution is possible or impossible without paying excessive efforts by experiment or theory. For example, the combination of amino or hydroxyl group having free hydrogen and carbon atoms having unsaturated bonds (such as olefinic) may be unstable.
A“pharmaceutical composition” refers to a mixture of one or more of the compounds described in the present invention or physiologically/pharmaceutically acceptable salts or prodrugs thereof and other chemical components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism, which is conducive to the absorption of the active ingredient and thus displaying biological activity.
“Pharmaceutically acceptable salts” refer to salts of the compounds of the invention, such salts being safe and effective when used in a mammal and have corresponding biological activity.
In addition to salt forms, the present invention provides compounds which are in a prodrug form. Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention. Additionally, prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
The compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. Unnatural proportions of an isotope may be defined as ranging from the amount found in nature to an amount consisting of 100% of the atom in question. For example, the compounds may incorporate radioactive isotopes, such as for example tritium (3H), iodine-125 (1251) or carbon- 14(14C), or non-radioactive isotopes, such as deuterium (D) or carbon-13(13C). Such isotopic variations can provide additional utilities to those described elsewhere within this application. For instance, isotopic variants of the compounds of the invention may find additional utility, including but not limited to, as diagnostic and/or imaging reagents, or as cytotoxic/radiotoxic therapeutic agents.
The phrase“therapeutically effective amount" refers to the administration of an agent to a subject, either alone or as part of a pharmaceutical composition and either in a single dose or as part of a series of doses, in an amount capable of having any detectable, positive effect on any symptom, aspect, or characteristic of a disease, disorder or condition when administered to the subject. The therapeutically effective amount can be ascertained by measuring relevant physiological effects, and it can be adjusted in connection with the dosing regimen and diagnostic analysis of the subject’s condition, and the like. By way of example, measurement of the serum level of a CD73 inhibitor (or, e.g., a metabolite thereof) at a particular time post- administration may be indicative of whether a therapeutically effective amount has been used.
The term“solvate,” as used herein, means a physical association of a compound of this invention with one or more, preferably one to three, solvent molecules, whether organic orinorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example, when one or more, preferably one to three, solvent molecules are incorporated in the crystal lattice of the crystalline solid. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are generally known in the art.
“Prodrug” refers to compounds that can be transformed in vivo to yield the active parent compound under physiological conditions, such as through hydrolysis in blood.
The term“pharmaceutically acceptable,” as used herein, refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and are effective for their intended use. The term “treat”, “treating”, “treatment”, or the like, refers to: (i) inhibiting the disease, disorder, or condition, i.e., arresting its development; and (ii) relieving the disease, disorder, orcondition, i.e., causing regression of the disease, disorder, and/or condition. In addition, thecompounds of present invention may be used for their prophylactic effects in preventing a disease, disorder or condition from occurring in a subject that may be predisposed to the disease, disorder, and/or condition but has not yet been diagnosed as having it.
The term“subject” or“patient” refers to a mammalian animal.
The term "mammal" or“mammalian animal” includes, but is not limited to, humans, dogs, cats, horses, pigs, cows, monkeys, rabbits and mice. The preferred mammals are humans.
As used herein, the singular forms“a”,“an”, and“the” include plural reference, and vice versa, unless the context clearly dictates otherwise.
SYNTHESIS METHOD
The compounds and processes of the present invention will be better understood in connection with the following synthetic schemes that illustrate the methods by which the compounds of the invention may be prepared. Other reaction schemes could be readily devised by those skilled in the art based on the present disclosure.
Figure imgf000026_0001
Step 1, reacting with formula (IV- 1) and formula (IV-2) under alkaline condition (such as DBU) to obtain formula (IV-3);
Step 2, reacting with formula (IV-3) and 2,2-dimethoxypropane under acidic condition (such as p-toluenesulfonic acid), then neutralize the reaction mixture to obtain formula (IV-4);
Step 3, reacting with formula (IV-4) and methyl enebis(phosphonic dichloride) with the additive (such as N-methylimidazole) to obtain formula (IV-5);
Step 4, reacting to remove the protecting group of acetal with a compound of formula (IV-5) under acidic condition (such as TFA) to obtain the compound of formula (IV-6);
wherein:
X is halogen, preferably chlorine;
and ring A, G3, G4, R8, R9, m and s are as defined in formula (IV).
The additive includes, but is not limited to, pyridine, 2,4,6-collidine, 1,4- diazabicyclo[2.2.2]octane (DABCO), 4-(dimethylamino)pyridine, N-methylimidazole.
Agents that provide acidic conditions include, but are not limited to, hydrogen chloride, hydrogen chloride 1,4-dioxane solution, trimethylsilyl bromide (TMSBr), ammonium chloride, trifluoro acetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, -toluenesul Ionic acid, p- toluenesulfonic acid monohydrate and TMSOTf.
The reaction is preferably in solvent, wherein solvent used herein includes, but is not limited to, acetic acid, methanol, ethanol, toluene, acetone, tetrahydrofuran, dichloromethane, dimethylsulfoxide, 1,4-dioxane, water, N, V-dimethylformamide, trimethylphosphate, methyl tert-butyl ether and the mixture thereof.
EXAMPLES
The structure of a compound is determined by mass spectrometry (MS) and or nuclear magnetic resonance (NMR). NMR shift (d) is given in units of 10 6 (ppm).
The mass spectrum (MS) was determined using a Shimadzu LCMS-2020 liquid chromatography-mass spectrometer.
The NMR measurement was performed on a Bruker AVANCE-400 and 500 Ultrashield nuclear magnetic resonance spectrometer. The solvents were deuterated dimethylsulfoxide ( D SO-iL), deuterated chloroform (CDCE) and deuterated methanol (CD3OD) Silane (TMS). HPLC was performed using a Shimadzu OPTION BOX-L high pressure liquid phase Chromatograph (Gimini 5um NX-C18 100x21.2mm column).
Thin-layer chromatography (TLC) silica gel plates used were Agela Technologies T- CSF10050-M silica gel plate with size of 50 mm,
Column chromatography was commonly done using CombiFlash Rf+ Automated Flash Chromatography System (TELEDYNE ISCO) with Agela Technologies Flash Column Silica - CS prepacked columns.
Known starting materials of the present invention may be synthesized according to methods known in the art or may be purchased from Acros Organics, Sigma-Aldrich Chemical Company, AstaTech and other companies. Unless otherwise specified in the examples, the reaction can be carried out under an argon atmosphere or a nitrogen atmosphere.
Argon or nitrogen atmosphere refers to the reaction flask connected to a volume of about 1 L argon or nitrogen balloon.
Hydrogen atmosphere refers to the reaction bottle connected to a volume of about 1 L hydrogen balloon.
Hydrogenation reaction is usually evacuated, filled with hydrogen, repeated 3 times.
The microwave reaction used a CEM Disco ver-S 908860 microwave reactor.
Unless otherwise specified in the examples, the reaction temperature is room temperature and is 20 0 C to 30 0 C.
The progress of the reaction in the examples was monitored using thin layer chromatography (TLC), developing solvent for the reaction, a column chromatography eluent for purifying compound, and developing system for thin-layer chromatography include: A: dichloromethane / methanol system, B: n- hexane / ethyl acetate system, C: dichloromethane / ethyl acetate system. The volume ratio of the solvents is adjusted according to the polarity of the compound. A small amount of triethylamine and acetic acid and other alkaline or acidic reagents can be used for adjustment.
TEA is triethylamine,
DBU is l,8-diazabicyclo[5.4.0]undec-7-ene,
EtOAc is ethyl acetate,
TMSOTf is trimethylsilyl trifluoromethanesulfonate,
THF is tetrahydrofuran,
DCM is dichloromathene,
AcOH is acetic acid, DMF is N,N-dimethylformamide,
MTBE is methyl tert-butyl ether,
TEAC is triethylammonium bicarbonate,
NMI is N-methylimidazole,
DMSO is dimethyl sulfoxide, and
MS is mass spectroscopy with (+) referring to the positive mode which generally gives a M+l (or M+H) absorption where M = the molecular mass.
Example 1 (Compound 1)
Figure imgf000029_0001
(((((2R,3S,4R,5R)-5-(6-chloro-4-(2',3'-dihydrospiro[azetidine-3,r-inden]-l-yl)-lH- pyrazolo[3,4-d]pyrimidin- l-yl)-3, 4-dihydro xytetrahydro furan-2- yl)methoxy)(hydroxy)phosphoryl)methyl)phosphonic acid 1
Figure imgf000029_0002
Step 1
Methyl 2,3-dihydro- 1 //-indene- 1 -carhoxylate lb
A dried round-bottom flask was charged with 2,3-dihydro- 1 /-indene- 1 -carboxylic acid la (10.0 g, 61.66 mmol) and DCM (100 mL), followed by addition of SOCh (6.7 mL, 92.49 mmol) at room temperature. The reaction mixture was stirred at room temperature for lh, then concentrated to half volume by rotavapor. To the reaction mixture above was added MeOH (100 mL) slowly and the reaction was continued to stir at room temperature for 3h. Then the mixture was concentrated by rotavapor and purified by column chromatography on silica-gel with 0-25% EtOAc in hexane as eluent on the TELEDYNE ISCO system to give the desired product lb as colorless oil (9.85 g, yield: 91%), MS (ESI): m/z =177 [M + 1].
Step 2
1-ethyl 1-methyl 2, 3-dihydro-177-indene-l,l-dicarboxylate lc A dried round-bottom flask was charged with methyl 2,3-dihydro- 1 //-indene- 1 - carhoxylate lb (3.80 g, 21.6 mmol) and THF (50 mL), then the solution was cooled to -78 °C. To this solution was added 1M lithium diisopropylamide in hexane/THF (24 mL, 24 mmol) and stirred for lh. Then ethyl cyanoformate (2.97 g, 30 mmol) was added and the reaction mixture was stirred overnight while allowing the temperature to warm up to room temperature slowly. The reaction was quenched with sat. NH4CI aq. and allowed to stir for 15 min. Then the mixture was extracted with EtOAc (100 mL), dried over MgS04, filtered and concentrated by rotavapor. The residue was purified by column chromatography on silica-gel with 0-30% EtOAc in hexane as eluent on the TELEDYNE ISCO system to give lc as colorless oil (4.5 g, yield: 90%), MS (ESI): m/z = 249 [M + 1].
Step 3
(2,3 -dihydro- 177- indene- 1 , 1 -diyl)dimethanol Id
A dried round-bottom flask was charged with 1-ethyl 1-methyl 2,3-dihydro- 1/7- indene-l,l-dicarboxylate lc (3.00 g, 12.00 mmol) and THF (15 mL), followed by cooling the solution down to 0 °C. To this solution was added 1M lithium aluminum hydride in THF (96 mL, 96 mmol) dropwise and the mixture was stirred overnight to allow the temperature to rise to room temperature. The mixture was quenched with sat. NH4CI aq. and 4M HC1 respectively at 0 °C to get a clear solution, then extracted with EtOAc (3 X 100 mL). The combined organic phases were dried over MgS04, filtered and concentrated by rotavapor. The resulted residue was purified by column chromatography on silica-gel with 0-50% EtOAc in hexane as eluent on the TELEDYNE ISCO system to give Id as white solids (1.6 g, yield: 75%), MS (ESI): m/z = 179 [M + 1] Step 4
(2,3-dihydro- 1/7- indene- 1 , 1 -diyl)bis(methylene) dimethanesulfonate le
A dried round-bottom flask was charged with (2,3-dihydro- 1 //-indene- 1 , 1 -diyl) dimethanol Id (1.00 g, 5.61 mmol), TEA (2.72 mL, 19.64 mmol) and DCM (15 mL), followed by cooling the solution down to 0 °C. To this solution was added methanesulfonyl chloride (1.30 mL, 16.83 mmol) and stirred at 0 °C for 2h. Then the mixture was diluted with EtOAc (25 mL), washed with water. The organic phase was separated and the aqueous phase was extracted with EtOAc (2 X 25 mL). The combined organic phases were dried over MgS04, filtered, and concentrated by rotavapor. The residue was purified by column chromatography on silica-gel with 0-30% EtOAc in hexane as eluent on the TELEDYNE ISCO system to give le as white solids (1.73 g, yield: 92%), MS (ESI): m/z = 335 [M + 1].
Step 5
1 -benzyl-2', 3'-dihydrospiro[azetidine-3,r-indene] If
A vial was charged with (2, 3 -dihydro- 177- indene- l,l-diyl)bis(methylene) dimethanesulfonate le (1.50 g, 4.49 mmol) and benzylamine (10 mL), followed by stirring at 110 °C for 15h. After cooling, removal of volatile on rotavapor, the residue was diluted with EtOAc (50 mL) and quenched with sat. NaHC03 aq. The organic phase was separated and the aqueous phase was extracted with EtOAc (50 mL). The combined organic phases were dried over MgS04, filtered and concentrated by rotavapor. The residue was purified by column chromatography on silica-gel with 0-60% EtOAc in hexane as eluent on the TELEDYNEISCO system to give If as white solids (0.26 g, yield: 23%), MS (ESI): m/z = 250 [M + 1]
Step 6
2',3'-dihydrospiro[azetidine-3,r-indene] acetate lg
A round-bottom flask was charged with 1 -benzyl-2’, 3’-dihydrospirao[azetidine-3,r- indene] If (0.26 g, 1.04 mmol), 20% Pd/C (100 mg), AcOH (0.1 mL) and methanol (15 mL). The flask was capped with septum and exchanged air with nitrogen gas by vacuum and refilling nitrogen gas. The mixture was then stirred under 1 atm ¾ atmosphere at room temperature for 15 h. The solid was removed by filtration and washed with methanol. The filtrate was concentrated on rotavapor and the residue was dried in vacuum to give the crude lg, which was used directly for next reaction without further purification, MS (ESI): m/z =160 [M + 1] Step 7
(2/?,3/?,4/?,5/?)-2-(acetoxymethyl)-5-(4,6-dichloro- 17/-pyrazolo|3 ,4-d]pyrimidin- 1 - yl)tetrahydrofuran-3,4-diyl diacetate lj
To a dried round-bottom flask (500 mL) was charged with 4,6-dichloro-lH- pyrazolo[3,4-d] pyrimidine li (12.5 g, 66.1 mmol), ammonium sulfate (0.1 g, 0.75 mmol) and hexamethyldisilazane (75 mL). After 3h refluxing, the mixture was cooled to room temperature, and then concentrated to dryness in high vacuum. The resulting residue was taken up into acetonitrile (150 mL) followed by addition of (2S,3R,4R,5R)-5- (acetoxymethyl)-tetrahydrofuran-2,3,4-triyl triacetate lh (25.3 g, 79.5 mmol). The mixture was then cooled down to 0 °C, followed by addition of TMSOTf (13.5 mL, 72.5 mmol) dropwise.
After 18h stirring with the temperature slowly warmed up to room temperature, the reaction mixture was concentrated by rotavapor and the residue was taken up into EtOAc (150 mL), washed with sat. aq. NaHCCL solution and brine in sequence. The organic phase was dried over anhydrous MgSCL, filtered, concentrated by rotavapor and the residue was purified by column chromatography on silica-gel with 0-40% EtOAc in hexane as eluent on the TELEDYNE ISCO system to give lj as yellowish sticky oil (22.4 g, yield: 76%), MS (ESI): m/z = 447 [M + 1]
Step 8
(2/?,3/?,4.V,5/?)-2-(6-chloro-4-(2,,3'-dihydrospiro|azetidine-3, 1 '-inden|- 1 -yl)- 17/ pyrazolo|3,4- djpyrimidin- 1 -yl)-5 -(hydro xymethyl)tetrahydrofuran-3 ,4-diol lk
A round-bottom flask was charged with the crude 2’,3’-dihydrospirao[azetidine-3,l’- indene] lg (0.26 g, 1.20 mmol), TEA (0.5 mL, 3.6 mmol) and methanol (10 mL), which was then cooled down to 0 °C. To this solution was added a solution of (2R RAR,5R)-2- (acetoxymethyl)-5-(4,6-dichloro-177-pyrazolo[3,4-d]pyrimidin-l-yl)tetrahydrofuran-3,4-diyl diacetate lj (0.54 g, 1.2 mmol) in methanol (2 mL) and the reaction mixture was stirred at 0 °C for lh and room temperature for lh respectively. The reaction mixture was then re-cooled down to 0 °C, followed by addition of DBU (0.51 mL, 3.6 mmol). The reaction was run for 2h while allowing the temperature warm to room temperature. The mixture was concentrated by rotavapor and the residue was purified by column chromatography on silica-gel with 0- 10% methanol in dichloromethane as eluent on the TELEDYNE ISCO system to give lk as white solids (0.33 g, yield: 62%), MS (ESI): m/z = 444 [M + 1]. Step 9
((3aR,4R,6R,6aR)-6-(6-chloro-4-(2,,3,-dihydrospiro|azetidine-3, 1 '-inden |- 1 -yl)- 1 H- pyrazolo[3,4-d]pyrimidin-l-yl)-2,2-dimethyltetrahydrofuro[3,4-d][l,3]dioxol-4-yl)methanol
11
A vial was charged with (2/?,3/?,4.V,5 ?)-2-(6-chloro-4-(2,,3,-dihydrospiro|azetidine- 3 , G-inden]- 1 -yl)- 17/-pyrazolo|3 ,4-d]pyrimidin- 1 -yl)-5 -(hydro xymethyl)tetrahydrofuran-3 ,4- diol lk (0.33 g, 0.74 mmol), 2,2-dimethoxypropane (1.12 mL, 8.88 mmol) and acetone (5 mL). To this solution was added -toluenesul Ionic acid monohydrate (0.13 g, 0.68 mmol) and the reaction was stirred at room temperature for 4h. The reaction mixture was concentrated by rotavapor and the residue was taken into EtOAc (25 mL), followed by quenching with sat. NaHCCb aq. and extracting with EtOAc (2 X 25 mL). The combined organic phases were washed with brine, dried over anhydrous MgS04, filtered and concentrated by rotavapor. The residue was purified by column chromatography on silica-gel with 0-40% EtOAc in DCM as eluent on the TELEDYNE ISCO system to give 11 as white solids (0.18 g, yield: 50%), MS (ESI): m/z = 484 [M + 1]
Step 10
(((((3 aR,4R,6R,6aR)-6-(6-chloro-4-(2',3'-dihydrospiro[azetidine-3 , 1’-inden]- 1 -yl)- 1H- pyrazolo[3,4-d]pyrimidin-l-yl)-2,2-dimethyltetrahydrofuro[3,4-d][l,3]dioxol-4- yl)methoxy)(hydroxy)phosphoryl)methyl)phosphonic acid lm
To a suspension of ((3aR,4R,6R,6aR)-6-(6-chloro-4-(2',3'-dihydrospiro[azetidine-3,r- inden]- 1 -yl)- 17/-pyrazolo|3 ,4-d]pyrimidin- 1 -yl)-2,2-dimethyltetrahydrofuro[3,4- d][l,3]dioxol-4-yl)methanol 11 (145mg, 0.3mmol) in trimethylphosphate (lmL) and NMI (O.lmL) was added a solution of methylenebis(phosphonic dichloride) (195 mg, 0.78mmol) in trimethylphosphate (lmL) dropwise at room temperature. After 15 min stirring at room temperature, the reaction mixture was quenched with 1M TEAC, extracted with MTBE, and the aqueous layer was purified by HPLC with 10-70% methanol in H2O + 0.5% NH4HCO3 to give lm as white solids (124mg, yield: 64%). MS (ESI): m/z = 642 [M + 1].
Step 11
(((((2R,3S,4R,5R)-5-(6-chloro-4-(2’,3’-dihydrospiro[azetidine-3,l’-inden]-l-yl)-lH- pyrazolo[3,4-d]pyrimidin- l-yl)-3, 4-dihydro xytetrahydro furan-2- yl)methoxy)(hydroxy)phosphoryl)methyl)phosphonic acid 1
To a solution of (((((3aR,4R,6R,6aR)-6-(6-chloro-4-(2',3'-dihydrospiro[azetidine-3,r- inden]- 1 -yl)- lH-pyrazolo[3 ,4-d]pyrimidin- 1 -yl)-2,2-dimethyltetrahydrofuro[3,4- d][l,3]dioxol-4-yl)methoxy)(hydroxy)phosphoryl)methyl)phosphonic acid lm (30mg, 0.047mmol) in DCM (lmL) was added TFA (5% water, 0.5mL) at 0 degree dropwise. After 30 min stirring at 0 degree, to the reaction mixture was added a solution of sodium carbonate (0.69 g) in H2O (5 mL) under stirring. After 30 min, the suspended mixture was mixed with acetonitrile and water to get a clear solution, which was used for the purification by HPLC with 10-70% methanol in H2O + 0.5% NH4HCO3 to give 1 as white solids (16 mg, yield: 56%). MS (ESI): m/z = 602 [M + l].1!! NMR (400 MHz, Methanol-cL) d 8.06 (s, 1H), 7.47- 7.56 (m, 1H), 7.21-7.30 (m, 3H), 6.23 (d, J = 3.5 Hz,IH), 4.56-4.78 (m, 4H), 4.38-4.50 (m,2H), 4.18-4.26 (m, 1H), 4.01-4.13 (m, 2H), 2.97-3.01 (m, 2H), 2.47-2.58 (m, 2H), 2.07- 2.25 (m, 2H); 31P NMR (162.4 MHz, Methanol-iA) d 17.78, 15.73.
Example 2
Compound examples 2-9 can be prepared with the similar method to the compound example 1.
Example 3
BIOLOGICAL ASSAYS
The present invention will be further described with reference to the following test examples, but the examples should not be considered as limiting the scope of the invention.
Test of the compounds of the present invention for their inhibition on CD73 enzyme activity in vitro.
CD73 enzyme, an ecto-5 '-Nucleotidase, converts extracellular nucleoside-5 '-monophosphates to nucleosides, with AMP or CMP as the preferred substrate. In this assay, recombinant human CD73 expressed from a Chinese hamster ovary cell line (R&D Systems) was used to convert cytidine monophosphate (CMP) to cytidine and phosphate. Before adding substrate, CD73 enzyme was pre-incubated with compounds for 2 hours. The amount of phosphate was then measured by Malachite Green Phosphate Detection Kit. The experimental method is summarized as follows:
I. Experimental Materials and Equipment
1. Malachite green phosphate detection kit (R & D Systems, Cat#, DY996)
2. Recombinant human 5 '-nucleotidase (CD73) (R & D Systems, Cat#, 5795-EN)
3. HEPES buffer (Gibco, Cat#, 15630-080)
4. CMP (Sigma, Cat#, C1006)
5. DMSO (Fisher Chemical, Cat#, D128-1)
6. NaCl 5M (Boston Bioproducts, Cat#, BM-244)
7. 384-well plate (Fisher, Cat#, 5795-EN)
8. TEC AN plate reader (TEC AN) II. Experimental Procedure
Compounds are first dissolved in DMSO to 10 mM as a stock solution. When determining the IC50 of the compound, prepare 3 -fold serial dilutions with a highest concentration of 125 mM for a total of 12 concentration points and ensure each dilution containing equal amount of DMSO. In each well of 384-well plate, 0.34 nM of recombinant human 5'-nucleotidase (CD73) was pre-incubated at 37°C for 2 hours with the compounds tested in assay buffer containing 20 mM HEPES buffer (pH 7.4), 137 mM NaCl, 0.001% Tween 20. The final reaction volume of the reaction in each well was 12 pL. The highest concentration of compound was 125 pM and the DMSO concentration was 1.25%. After pre incubation, 3 pL of CMP dissolved in assay buffer was added to each reaction. The final CMP concentration was 45 pM. The reaction was incubated at 37°C for 15 minutes. Then 3 pL of Malachite Green Reagent A was added to each reaction. Spin the plate briefly in centrifuge for 30 seconds. After incubation for additional 10 minutes at room temperature, 3 pL of malachite green Reagent B was added to each reaction. Spin the plate briefly in centrifuge for 30 seconds. After 20 minutes of incubation at room temperature, the signal was read on a TEC AN reader at OD620. The reaction containing CD73 enzyme, substrate CMP and DMSO (no compound) is used as an assay positive control while the reaction containing substrate CMP and DMSO without the CD73 enzyme as an assay negative control. IC50 values were calculated by plotting the logarithm of the compound concentration and the percent inhibition using the appropriate program in GraphPad Prism.
The biochemical inhibitions of CD73 enzymatic activities by compound 1 of the present invention was determined by the assay described above, and the resulting IC50 value was 0.56 nM.
Conclusion: The compounds of the present invention have a significant inhibitory effect on CD73 enzyme activity in vitro.
Example 4
Test of the compounds of the present invention for their inhibition of membrane- bound CD73 enzyme activity on human melanoma A375 cells.
CD73 enzyme, an ecto-5 '-Nucleotidase, converts extracellular nucleoside-5 '-monophosphates to nucleosides, with AMP or CMP as the preferred substrate. In this assay, membrane-bound CD73 enzyme activity on the surface of human melanoma A375 cells (ATCC® CRL-1619) was used to convert cytidine monophosphate (CMP) to cytidine and phosphate in presence of compounds and CMP. The amount of phosphate was then measured by Malachite Green Phosphate Detection Kit. The experimental method is summarized as follows:
I. Experimental Materials and Equipment
1. Malachite green phosphate detection kit (R & D Systems, Cat#, DY996)
2. A375 cell line (ATCC, Cat#, CRL-1619)
3. DMEM (ATCC, Cat#, 30-2002)
4. Trypsin-EDTA 0.25% (Gibco, Cat#, 25-200-056)
5. FBS (Gibco, Cat#, 16-140-071)
6. Penicillin-Streptomycin (Gibco, Cat#, 15-140-122)
7. CMP (Sigma, Cat#, C1006)
8. 96-well plate for cell culture (Corning, Cat#, 3595)
9. 2ml 96-well block (Costar, Cat#, 3960)
10. Transparent flat-bottom 96-well plate (Thermo Scientific, Cat#, 260836)
11. DMSO (Fisher Chemical, Cat#, D128-1)
12. HEPES (Gibco, Cat#, 15630-080)
13. NaCl 5M (Boston Bioproducts, Cat#, BM-244)
14. KC1 2M (Ambion, Cat#, AM9640G)
15. CaCl2 2M (Fisher, Cat#, BP9742)
16. NaHCOs 7.5% (Gibco, Cat#, 25080-094)
17. Glucose (Gibco, Cat#, A2494001)
18. TECAN plate reader (TECAN)
II. Experimental Procedure
A375 cells are maintained using DMEM medium containing 10% FBS and 1% Penicillin-Streptomycin. A day before the assay, use trypsin to harvest A375 cells and perform a cell count. Seed cells to each well of the 96- well plate in 100 pL media (2500 cells/well). Next day, prepare for assay buffer containing 20 mM HEPES, 137 mM NaCl, 5.4 mM KC1, 1.3 mM CaCh, 4.2mM NaHC03 and 1 mg/mL glucose. Warm the buffer in 37°C water bath. Make 3.16-fold serial dilutions of the compounds, starting with a highest concentration of 10 mM for a total of 12 concentration points, in assay buffer containing 50 pM CMP and ensure each dilution containing equal amount of DMSO (0.1%).
Remove the medium gently from the cell culture plate, gently wash the cell layer with assay buffer once, and then transfer 200 pL of serial diluted compound solutions into the corresponding wells. Add 200 pL assay buffer into negative control wells and 200 pL DMSO/CMP/assay buffer into positive control wells.
After 4-hour incubation at 37°C with 5% CO2, transfer 100 pL of supernatant from each well to a transparent flat-bottom 96-well plate. Add 20 pL Reagent A from the Malachite Green Phosphate Detection Kit to each well. Incubate for 10 minutes at RT. Add 10 pL Reagent B to each well. Tap the plate to help mixing. Incubate for 20 minutes at RT. Then read at OD620 using Tecan plate reader.
III. Data analysis:
Inhibition % values were calculated
Figure imgf000037_0001
by plotting the logarithm of the compound concentration and the percent inhibition using the appropriate program in GraphPad Prism.
The biochemical inhibition of CD73 enzymatic activities by compound 1 of the present invention was determined by the assay described above, and the resulting IC50 value was 0.013 nM.
IV. Conclusion
The compounds of the present invention have a significant inhibitory effect on CD73 enzyme activity in A375 cells.
Example 5
Determination of the compounds of the Present Invention Modulates Cellular Immune Function by IFNy Cytokine Production
I, Experimental Materials and Equipment
1. Cells: Cryopreserved human peripheral blood mononuclear cells (PBMCs)
(Stemcell, Cat # 70025).1, 15 to 25 million cells per bottle
2. Lymphocyte culture medium (Zenbio, Cat# LYMPH- 1)
3. TexMACS medium (Miltenyi, Cat# 130-097-196)
4. CD3 and CD28 antibody beads (Fisher Scientific, Cat# 1 161D)
5. HTRF human IFNy Cytokine kit (Cisbio, human IFNy Cat# 62HIFNGPEH)
6. PHERAstar FSX Multilabel Reader (BMG Labtech)
II. Experimental Procedure
Lymphocyte medium and TexMACS medium were incubated in a water bath at 37 0 C. 10 mL of incubated lymphocyte medium was added to a 50 mL conical tube. The cells were quickly thawed in a 37 0 C water bath and transferred to a 50 ml tube and the tube gently swirled. The cell suspension was centrifuged at 1100 rpm for 10 minutes at room temperature. The supernatant was removed, and the cell pellet gently resuspended in 10 mL of TexMACS medium. The cells were counted to make 5 x 105 cells / ml. 100 pL of 5 x 105 cells / ml of PBMCs seeds, inserted into a 96-well plate (cell density of 50,000 cells / well), and the most outer edge wells of the 96-well plate were filled with water and were not used for the test. To mimic tumor microenvironment in human PBMC from normal donor and measure inhibition activity of the compound against CD73 enzyme, the CD73 substrate AMP was added to human PBMC cell culture in order to generate more adenosine via CD73 enzyme. Added 50 pL of 200 pM AMP (Sigma A2252) to each of the other wells. The final AMP concentration was 50 pM. Compounds were diluted to 40 pM in TexMACS media. In addition to the“AMP only” wells, 50 pL of compound was added to each well. The final compound concentration was 10 pM in 0.1%DMSO. A 0.4% DMSO was prepared in TexMACS medium and 50 pL was added to AMP control wells. Gently tapped the board to mix it evenly. After 2 hours of incubation with 5% CO2 at 37°C, CD3 and CD28 antibody beads were washed twice with TexMACS media and a magnet rack. Added 2 pL of CD3 and CD28 antibody beads to each well. The final antibody beads and cells ratio was 1 :1. Pipetted cells and antibody beads several times to homogenize. Incubated at 37°C for 72 hours in a 5% CO2 humidified incubator. After incubation, the cells were spun (1000 rpm for 5 minutes), 90 pL of cell culture supernatant was carefully collected, and the culture supernatant was stored with HTRF IFNy cytokine reagent and assayed immediately or frozen at -80°C. In a sample diluted in TexMACS medium (25 and 100 times), 12.8 pL of sample was added to each well of a 384-well plate (Proxiplate-384plus). The IFN Cryptate was mixed with XL (1:1) and 3.2 pL of the mixed solution was added to each well in the plate. Swirl swiftly spinned down. Protected from light at room temperature. Read the values of 665 nm and 620 nm on a PHERAstar FSX Multilabel Reader.
III. Data analysis:
Calculated ratio = (signal at 665 nm / signal at 620 nm) x 104, which reflected the “raw signal”. Delta Ratio reflected the specific signal. Delta Ratio = Standard or Sample Ratio - Standard 0 Ratio, where Standard 0 was negative control and was used as an internal assay control. A standard curve was drawn using ratios with a quadratic curve fit method following vendor’s instruction. The concentration of IFNy cytokine of each sample corresponding to the standard curve was calculated. Response rate of IFNy cytokines by a compound (fold) = (sample- AMP only) / (DMSO- AMP only), where sample = IFNy produced by 10 pM compound in 0.1% DMOS and 50 pM AMP; DMSO = IFNy produced by 0.1%DMSO (vehicle control) and 50 mM AMP; AMP only = IFNy produced by 50 mM AMP (as background).
The production of IFNy cytokines by compound 1 of the present invention was measured by the above test with the result as IFNy Induction ECso nM / Emax % = 2.2 nM / 106%.
IV. Conclusion
The compound of the invention can stimulate the production of IFNy cytokines and thus have a significant modulating effect on the cellular immune functions.
The foregoing embodiments and examples are provided for illustration only and are not intended to limit the scope of the invention. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art based on the present disclosure, and such changes and modifications may be made without departure from the spirit and scope of the present invention.

Claims

CLAIMS What is claimed is:
1. A compound of formula (I):
Figure imgf000040_0001
or a tautomer, mesomer, racemate, enantiomer, diastereomer thereof, or mixture thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof,
wherein:
W is selected from the group consisting of O, S, NH, NRa and C(Rb)2, wherein Ra is alkyl, and Rb at each occurrence is independently selected from the group consisting of hydrogen, halogen, alkyl and alkenyl;
G1 and G2 are each independently N or CRC, wherein Rc is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, hydroxy, amino, nitro, cyano, cycloalkyl, heterocyclyl, aryl and heteroaryl;
G3 and G4 are each independently selected from the group consisting of C, CH, CH2, N, NH, O, S and S02;
ring A is selected from the group consisting of Cs-scycloalkyl, 5 to 8-member heterocyclyl, aryl fused Cs-scycloalkyl, heteroaryl fused Cs-scycloalkyl, aryl fused 5 to 8- member heterocyclyl, heteroaryl fused 5 to 8-member heterocyclyl;
R1, R2, R3 and R4 are each independently selected from the group consisting of hydroxy, hydrogen, halogen, alkyl, alkoxy, haloalkyl, hydroxyalkyl, cyano, amino, azide group and OR10;
or R1 and R2 together with the carbon atom they are attached to form a cycloalkyl or heterocyclyl, wherein the heterocyclyl contains 1 to 2 heteroatoms which are the same or different from N, O and S, and wherein the cycloalkyl and heterocyclyl are each optionally substituted by one or more groups selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl and heterocyclyl;
R5 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro and azide;
R6 is selected from the group consisting of hydrogen and optionally substituted Ci-Ce alkyl;
R7 at each occurrence is independently selected from the group consisting of hydrogen, alkyl, aryl, -C(RmRn)-aryl, -C(RmRn)-0-C(0)0Rd, -C(RmRn)-0-C(0)Rd, - C(RmRn)C(0)ORd, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl is optionally substituted by one or more groups selected from the group consisting of cycloalkyl, heterocyclyl, aryl and heteroaryl; optionally, two R7 are combined to form a 5- to 7-membered heterocyclic ring;
Rm and Rn are each independently selected from the group consisting of H, D, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl and amino;
Rd is selected from the group consisting of hydrogen, alkyl and alkoxy;
R8 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, azide, cycloalkyl and heterocyclyl;
R9 is selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, azide, cycloalkyl and heterocyclyl;
R10 is selected from the group consisting of -C(0)Rn, -C(0)ORn, -S(0)2Rn, and - P(0)(0R7)2;
R11 is selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, hydroxyl and hydroxyalkyl;
X is O;
m is 1 , 2 or 3 ; and
s is 0, 1, 2, 3 or 4.
2. The compound of claim 1, being a compound of formula (II):
Figure imgf000042_0001
or a tautomer, or a pharmaceutically acceptable salt, solvate, or prodrug thereof,
wherein:
X, W, G1, G2, G3, G4, ring A, R1 to R9, m and s are as defined in claim 1.
3. The compound of claim 1 or 2, being a compound of formula (III):
Figure imgf000042_0002
or a tautomer, or a pharmaceutically acceptable salt, solvate, or prodrug thereof,
wherein:
G1, G2, G3, G4, ring A, R1, R3, R7 to R9, m and s are as defined in claim 1.
4. The compound of claim 1 or 2, wherein R2 and R4 are each hydrogen; and R1 and R3 are each independently selected from the group consisting of hydroxy, hydrogen, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, and hydroxyalkyl.
5. The compound according to any one of claims 1 to 4, being a compound of formula (IV):
Figure imgf000042_0003
or a tautomer, a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein:
G3, G4, ring A, R7 to R9, m and s are as defined in claim 1.
6. The compound according to any one of claims 1 to 5, wherein
Figure imgf000043_0001
selected from the group consisting of
Figure imgf000043_0002
R9 and s are as defined in claim 1.
7. The compound according to any one of claims 1 to 6, wherein R7 is selected from the group consisting of hydrogen, alkyl, and -C(RmRn)-0-C(0)0Rd.
8. The compound according to any one of claims 1 to 7, wherein R8 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, and haloalkoxy.
9. The compound according to any one of claims 1 to 8, wherein R9 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, and haloalkoxy.
10. The compound according to any one of claims 1 to 9, wherein the compound is selected from the group consisting of:
Figure imgf000044_0001
Figure imgf000045_0001
11. A pharmaceutical composition, comprising a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and one or more pharmaceutically acceptable carriers, diluents and/or excipients.
12. A method for inhibiting CD73, comprising contacting a biological sample containing CD73 with a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, or a pharmaceutical composition according to claim 11.
13. A method for treating an adenosine or adenosine receptor related disease or disorder, comprising administering to a subject in need thereof a therapeutically effective amount of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, or the pharmaceutical composition according to claim 11.
14. A method for treating a disease or condition mediated by CD73, comprising
administering to a subject in need thereof a therapeutically effective amount of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, or the pharmaceutical composition according to claim 11, wherein the disease or condition is selected from the group consisting of tumor, cancer, immune-related diseases, inflammatory-related diseases, nervous system, neurodegenerative and central nervous system diseases, depression, Parkinson's disease, ischemic diseases of the brain and heart, sleep disorders, and fibrosis.
15. The method according to claim 14, wherein cancer is selected from the group consisting of melanoma, brain tumor, esophageal cancer, gastric cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, metrocarcinoma, endometriosis, prostate cancer, skin cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, lymphoma, polycythemia vera, leukemia, thyroid tumor, ureteral tumor, bladder cancer, gallbladder cancer, cholangiocarcinoma, chorionic epithelial cancer, and pediatric tumor.
16. Use of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, or a pharmaceutical composition according to claim 11, in the manufacture of a medicament for treating an adenosine and adenosine receptor related disease or disorder.
PCT/US2020/025269 2019-03-29 2020-03-27 Cd73 inhibitors and therapeutic uses thereof WO2020205538A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080024912.6A CN113905743B (en) 2019-03-29 2020-03-27 CD73 inhibitors and therapeutic uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962826441P 2019-03-29 2019-03-29
US62/826,441 2019-03-29

Publications (1)

Publication Number Publication Date
WO2020205538A1 true WO2020205538A1 (en) 2020-10-08

Family

ID=72667468

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/025269 WO2020205538A1 (en) 2019-03-29 2020-03-27 Cd73 inhibitors and therapeutic uses thereof

Country Status (2)

Country Link
CN (1) CN113905743B (en)
WO (1) WO2020205538A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022237747A1 (en) * 2021-05-11 2022-11-17 四川海思科制药有限公司 Small molecular cd73 antagonist and use thereof
WO2023201267A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170044203A1 (en) * 2014-04-25 2017-02-16 Vitae Pharmaceuticals, Inc. Purine derivatives as cd73 inhibitors for the treatment of cancer
WO2017120508A1 (en) * 2016-01-08 2017-07-13 Arcus Biosciences, Inc. Modulators of 5'-nucleotidase, ecto and the use thereof
WO2018049145A1 (en) * 2016-09-09 2018-03-15 Calithera Biosciences, Inc. Ectonucleotidase inhibitors and methods of use thereof
WO2020047082A1 (en) * 2018-08-28 2020-03-05 Eternity Bioscience Inc. Cd73 inhibitors and therapeutic uses thereof
US20200078388A1 (en) * 2018-09-11 2020-03-12 Risen (Suzhou) Pharma Tech Co. Ltd. Cd73 inhibitors and pharmaceutical uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170044203A1 (en) * 2014-04-25 2017-02-16 Vitae Pharmaceuticals, Inc. Purine derivatives as cd73 inhibitors for the treatment of cancer
WO2017120508A1 (en) * 2016-01-08 2017-07-13 Arcus Biosciences, Inc. Modulators of 5'-nucleotidase, ecto and the use thereof
WO2018049145A1 (en) * 2016-09-09 2018-03-15 Calithera Biosciences, Inc. Ectonucleotidase inhibitors and methods of use thereof
WO2020047082A1 (en) * 2018-08-28 2020-03-05 Eternity Bioscience Inc. Cd73 inhibitors and therapeutic uses thereof
US20200078388A1 (en) * 2018-09-11 2020-03-12 Risen (Suzhou) Pharma Tech Co. Ltd. Cd73 inhibitors and pharmaceutical uses thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022237747A1 (en) * 2021-05-11 2022-11-17 四川海思科制药有限公司 Small molecular cd73 antagonist and use thereof
WO2023201267A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers

Also Published As

Publication number Publication date
CN113905743B (en) 2024-04-16
CN113905743A (en) 2022-01-07

Similar Documents

Publication Publication Date Title
JP5808869B2 (en) Bicyclic piperazine compounds
ES2619125T3 (en) Heterocyclic compounds fused as protein kinase inhibitors
JP5842004B2 (en) Pyridazinone, its production method and its use
KR101829940B1 (en) Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof
JP5770751B2 (en) Pyrido [3,2-d] pyrimidine-based PI3K delta inhibitor compounds and methods of use
KR20210025016A (en) Conjugated tricyclic heterocycle compound and its therapeutic use
WO2022247816A1 (en) Nitrogen-containing heterocyclic compound, preparation method therefor, and application thereof in medicines
WO2020205538A1 (en) Cd73 inhibitors and therapeutic uses thereof
JP2023528925A (en) Pyridone pyrimidine derivative, preparation method thereof and pharmaceutical application thereof
US11332492B2 (en) CD73 inhibitors and therapeutic uses thereof
CN111094314B (en) Prodrug containing glucuronide derivative JAK inhibitor and preparation method and application thereof
CN116669740A (en) Pyrimidopyridine compound, preparation method and medical application thereof
JP2023145548A (en) Cd73 inhibitor, preparation method therefor and application thereof
WO2020151707A1 (en) Novel small molecule cd73 inhibitor, preparation method therefor and pharmaceutical use thereof
KR20220154722A (en) Fused bicyclic derivatives, methods for their preparation, and pharmaceutical uses thereof
WO2022002243A1 (en) Imidazopyrimidine derivative, preparation method therefor and medical use thereof
RU2787428C1 (en) Cd73 inhibitor, its production method and its use
CN113135942B (en) Condensed pyrimidine derivative, preparation method and medical application thereof
CN113912608B (en) Pyrimidopyrimidinone derivatives, preparation method thereof and application thereof in medicines
TW202140474A (en) Fused heteroaryl derivatives, preparation method and medical use thereof
US20220135539A1 (en) Biaryl amide compounds, preparation methods and medical applications thereof

Legal Events

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

Ref document number: 20783105

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20783105

Country of ref document: EP

Kind code of ref document: A1