WO2017161349A1 - Cyclic di-nucleotide compounds and methods of use - Google Patents

Cyclic di-nucleotide compounds and methods of use Download PDF

Info

Publication number
WO2017161349A1
WO2017161349A1 PCT/US2017/023093 US2017023093W WO2017161349A1 WO 2017161349 A1 WO2017161349 A1 WO 2017161349A1 US 2017023093 W US2017023093 W US 2017023093W WO 2017161349 A1 WO2017161349 A1 WO 2017161349A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
halogen
thiol
carbonyl
carboxyl
Prior art date
Application number
PCT/US2017/023093
Other languages
French (fr)
Inventor
Boyu Zhong
Lijun Sun
Qi Wei
Yuanwei DAI
Chuo Chen
Zhijian Chen
Original Assignee
Immune Sensor, Llc
The Board Of Regents Of The University Of Texas System
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
Priority to NZ746112A priority Critical patent/NZ746112A/en
Priority to ES17767683T priority patent/ES2929628T3/en
Priority to SG11201807660QA priority patent/SG11201807660QA/en
Priority to CA3017524A priority patent/CA3017524A1/en
Priority to LTEPPCT/US2017/023093T priority patent/LT3429596T/en
Priority to CN202210558244.6A priority patent/CN114751950A/en
Priority to CN202111504995.1A priority patent/CN114230625A/en
Priority to DK17767683.0T priority patent/DK3429596T3/en
Priority to EA201892095A priority patent/EA037513B1/en
Priority to AU2017233068A priority patent/AU2017233068C1/en
Priority to CN201780030376.9A priority patent/CN109475570B/en
Priority to EP19218894.4A priority patent/EP3692996A1/en
Application filed by Immune Sensor, Llc, The Board Of Regents Of The University Of Texas System filed Critical Immune Sensor, Llc
Priority to BR112018068748-0A priority patent/BR112018068748B1/en
Priority to IL280430A priority patent/IL280430B2/en
Priority to KR1020187029283A priority patent/KR102530488B1/en
Priority to JP2018568181A priority patent/JP6980198B2/en
Priority to EP17767683.0A priority patent/EP3429596B1/en
Priority to HRP20221263TT priority patent/HRP20221263T1/en
Priority to PL17767683.0T priority patent/PL3429596T3/en
Publication of WO2017161349A1 publication Critical patent/WO2017161349A1/en
Priority to US15/953,492 priority patent/US10519188B2/en
Priority to ZA2018/06074A priority patent/ZA201806074B/en
Priority to IL261827A priority patent/IL261827B/en
Priority to US16/438,153 priority patent/US11299512B2/en
Priority to US17/697,247 priority patent/US20220340613A1/en

Links

Classifications

    • 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
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • C07H19/213Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids containing cyclic phosphate
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7084Compounds having two nucleosides or nucleotides, e.g. nicotinamide-adenine dinucleotide, flavine-adenine dinucleotide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical

Definitions

  • the present invention provides no el cyclic di -nucleotide cGAMP analogs, pharmaceutical compositions thereof their synthetic methods and their use in medical therapy.
  • the compounds of the invention enhance the body's immune responses by acti vating STING (Stimulator of Interferon Genes) and are useful for the immunotherap of cancer, infectious diseases and immune disorders.
  • STING Stimulator of Interferon Genes
  • the compounds are also useful as adjuvants for developing vaccines against cancer and infectious diseases.
  • Cytosolic DNA induces type-I interferons and other cytokines that are important for immune defense against microbial infections and malignant cells but can also result in autoimmunity.
  • This DNA signaling pathway requires the adaptor protein STING (Stimulator of Interferon Genes) and the transcription factor IRF3, but the mechanism of DNA sensing was unclear until recently, WO 2014099824 to The University of Texas disclosed that mammalian cytosolic extracts synthesized cyclic-GMP-AMP (cGAMP) in vitro from ATP and GTP in the presence of DNA but not RNA.
  • STING Stimulator of Interferon Genes
  • cGAMP represents the first cyclic di-nucleotide in metazoa and it functions as an endogenous second messenger that triggers interferon production in response to cytosolic DNA.
  • cGAS cGAMP synthase
  • the inventors on WO 2014099824 also determined that the second messenger cGAMP they isolated and synthesized contains two phosphodiester linkages, one between the 2'-OH of GMP and 5" -phosphate of AMP, and the othe between the 3 ' -OH of AMP and 5 '-phosphate of GMP; this molecule is referred to as 2 ' 3'-cGAMP.
  • CDN cyclic-di- nucleotide
  • WO 2015077354 Al to The University of Chicago discloses Methods and compositions for treating cancer by intratumorally administering a stimulator of interferon genes (STING) agonist.
  • STING interferon genes
  • compositions and methods concerning methods for treating cancer in a subject comprising administering to the subject an effective amount of a stimulator of interferon genes (STING) agonist, wherein the STING agonist is administered intratumorally.
  • W 2015161762 to Fudan University discloses the use of cyclic dmucleotide cGAMP for preparing antitumor drugs, wherein the tumor is gastric cancer, lung cancer, colon cancer, liver cancer, prostate cancer or pancreatic cancer, cGAMP was shown to inhibit the growth of human tumor cell lines in immun e compromised mice.
  • WO 2015185565 to GlaxoSmithKiine discloses a class of cyclic dmucleotide compounds, or a pharmaceutically acceptable salt and tautomers thereof, compositions, combinations and medicaments containing said compounds and processes for their preparation.
  • the invention also relates to the use of said compounds, combinations, compositions and medicaments, in the treatment of diseases and conditions m which modulation of STING is beneficial, for example inflammation, allergic and autoimmune diseases, infectious diseases, cancer and as vaccine adjuvants.
  • WO 2014179335 to Memorial Sloan Kettering Cancer Center discloses compositions, methods, kits, and assays related to the use and/or exploitation of isomers of cGAMP as well as the structure of the enzyme cGAS.
  • cGAMP analogues with better potency are still needed.
  • Formula 1 encompasses Formula la - li.
  • the present invention provides a compound of Formula la
  • Formula la wherein: a and b are independently 0 or 1 and a + b :::: 1 , when a is 1, b is 0 and R 5 is not present; and when a is 0, b is 1 and R 4 is not present;
  • X 1 and X 2 are independently 0, S or Se in a five-membered ring;
  • L ! starting from the carbon alpha to X 1
  • L 2 starting from the carbon alpha to X 2
  • L ! are independently -C3 ⁇ 40-P(0)R 6 -0-,-CH20-P(S)R -0-, -C(Y ⁇ (Y 2 )0-P(0)R 6 -C(Y 3 )(Y 4 )-, - CH2NHSO2NH-, -CI !Ai ii (OSNH-.
  • Y 1 , Y 2 , Y 3 , and Y 4 are independently H or F;
  • R 6 is hydroxy!, thiol, Ci-ealk l, Ci-ealkyl selectively functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi, carbonyioxyl Ci-ealkoxy, d- &hydroxyalkoxy, amino, Ci-ealkylamino, di(Cwalk>'l)amino, or azido groups, Ci- eaikoxy, Ci-6aikoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyi, carbonyioxyl, d-ehydroxyalkoxy, amino, Ci- ealky!amino, di(C :-6alkyl)amino, or azido groups, Cs-salkenyl-O-, C3.5alkyn.yl-O-, oli
  • R ? and R 8 are independently hydrogen, Ci-ealkyl.
  • Ci-ea kyl selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyi, carbonyioxyl, Ci -ealkoxy, d-Aydroxyalkoxy, amino, Ci-ealkylamino, di(Ci- 6alkyl)amiiio, or azido groups, cyclic -(Ci-salkyi)-, cyclic -(Ci-ealkyl)- seiectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi carbonyioxyl, Ci-ealkoxy, d-ehydroxyalkoxy, amino, Ci- ealkylammo, or di(Ci-6alkyl)amino groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl
  • R 1 and R 2 are independently aromatic rings or heieroaromatic rings with the following genera! structure including its tautomeric forms:
  • g and h are independently 0 or 1;
  • W 1 and W 2 are independently hydrogen, halogen, hydroxyl, Ci-ealkyl Ci- ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyi, carbonyioxyl, Ci-ealkoxy, d-diydroxyalkoxy, amino, di(Ci 6a!kyl)amino, or azido groups, Ci-6a!koxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl carboxyi, carbonyioxyl, Ci- shydroxyalkoxy, amino, Ci-6aikylamino, di(Ci-6alkyi)aniino, or azido groups, ( ' - salkenyl-O-, C3-5alkynyl-0-, oligo(ethyiene glycol), polyiethylene glycol), azido, or - NR 7 R 8 ;
  • Z " Z 2 , Z 3 , Z 4 , Z 5 , and Z b are independently CH or N; if present, Z 7 , Z 8 , Z 9 , Z i0 , and Z 11 are independently CH or N, and then W ! is O i or V. and
  • R 3 , R 4 , and R 5 are independently hydrogen, halogen, hydroxy], ammo, Ci-ealkyl, Ci- 6alkyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyi, carboxyl, carbonyloxyl, Ci-eaikoxy, Ci-shydroxyalkoxy, amino, Ci-6alkylamino, di(Ci- 6alkyl)amino, or azido groups, Ci-6alkoxy, C :-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxvl, carbonyi, carboxyl, carbonyloxyl, Ci-6hydroxyalkoxy, amino, Ci-ealkylamino, di(C!-6alkyl)amino, or azido groups, C3-5alkenyl-0-, Cs-salkynyl-O-, oligo(ethyiene glycol), poly(eth lene glycol), azido, or
  • the present invention provides a compound of f ormula lb
  • X 1 and X 2 are independently 0, S or Se;
  • Z 12 , Z ! 3 , Z 14 , v Z 16 , and / " are independently CH or N;
  • L l starting from the carbon alpha to X s
  • L 2 starting from the carbon alpha to X independently -CH 2 0-P(0)R 6 -0-,-CH 2 0-P(S)R 6 -0-, -C(Y 3 )(Y 2 )0-P(0)R 6 -C(Y 3 )(Y 4 )-, CH 2 NHS0 2 NH-, ⁇ ( ⁇ ⁇ ⁇ 1( * ! ⁇ ) ⁇ ⁇ ⁇ .
  • -CH 2 NHC(S)NH- -CH 2 NHC(NH)NH- CH 2 NHC(0)CH 2 -, -CH2NHSO2CH2- -CH 2 CH 2 C(0)NH-, -CH2CH2SO2NH-
  • Y ⁇ Y 2 , Y 3 , and Y 4 are independently H or F,
  • R 6 is hydroxy!, thiol, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl Ci-ealkoxy, Ci- ehydroxyalkoxy, amino, Chalky laniino, di(Ci-6alkyi)amino, or azido groups, Ci- ealkoxy, Ci-salkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- 6alkylamino, or azido groups, Cs-salkenyl-O-, Ca-salkynyl-O-, oligo(ethylene glycol), polyfethylene glycol), borano (-BH3 " ), or -NR 7 R 8 ;
  • R ? and R s are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-6alkoxy, Cwhydroxyalkoxy, amino, Ci-ea!kylamino, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Ci-6alkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cusalkoxy, Ci-e.hydroxyalkoxy, amino, Ci- ealkylamino, or di(Ci -6alkyl)amino groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more
  • R 3 and R 4 are independently hydrogen, halogen, hvdroxyl, amino, Cj- 6 alkyl, C 1 -ealkyl selectively functionalized with one or more halogen, thiol, hydroxy 1, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, di(Ci-6a!kyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-fthydroxyalkoxy, amino, G- 6alkylamino, di(G-6aiky!)amino, or azido groups, G-salkenyl-O-, G-sa!kynyi-O-, oligo(ethylene glycol), poly(ethy!ene glycol), azido, or -NR 7 R S ; or
  • the present invention provides a compound of
  • zi 2j Z Z H Z _ and Z i ? are independently CH or N;
  • R 3 and R 4 are independently hydrogen, halogen, hydroxvl, amino, Cwalk l, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonyloxvl, Ci-ehydroxyalkoxy, amino, Ci-6aikyiamino, di(Ci- 6 alkyl)amino, or azido groups, Ci-ealkoxy, Ci-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxvl, carbonyl, carboxyl carbonyloxvl, Ci-bhydroxyalkoxy, ammo, Ci- ealkyiammo, di(Ci-6alk ⁇ 'i)amino, or azido groups, Cs-salkenyl-O, C3-5alkynyl-0-, oligo(ethyiene glycol), poly(ethylene glycol), azido, or -NR 7 R 8 ;
  • R 7 and R 8 are independently hydrogen, Ci-ealkyl, Ci-ealkyi selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, C i ⁇ hydrox alkoxy, amino, Cwalkylamino, difCi- 6aikyl)amino, or azido groups, cyclic -(Cwaikyl)-, cyclic -(Ci-ealkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyi, Ci-eaikoxy, C ⁇ hydroxy aikoxy, amino, Ci-ealkylamino, or di(Ci- 6alkyl)amino groups, cyclic -(Ci-eoxaalkyl)-, or cyclic -(Ci-eoxaalkyl)
  • R v and R 10 are independently hydroxy!, thiol Ci-ealkyl, Ci-6alkyl selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonvl, carboxyl, carbonvloxyl Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci-6alkylamino, di(Ci-6alkyi)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively fundi on ali zed with one or more halogen, thiol, hydroxyl, carbonvl, carboxyl, carbonvloxyl, Ci-ehydroxyalkoxy, amino, Ci-ealkylamino.
  • tetrahydrofuranyl rings of Formula lc is replaced by a sulfur or a selenium atom.
  • the present invention provides a compound of Formula Id
  • X *! and X 2 are independently 0, S or Se;
  • L 1 starting from the carbon alpha to X 1 , and L% starting from the carbon alpha to X 2 , are independently -CH20-P(0)R 6 -0-,-CH 2 0-P(S)R 6 -0-, -C(Y i )(Y )0-P(0)R 6 -C(Y 3 )(Y 4 )-, - CH2NHSO2NH-, -CH_NHC(0)NH-, -CH_NHC(S)NH-, -CH 2 NHC(NH)NH- 5 - CH 2 NHC(0)CH 2 -, -CH2NHSO2CH2- -CH 2 CH 2 C(0)NH-, -CH 2 CH 2 SQ2NH-, -CH 2 NH(3,4-
  • Y ] , Y", Y- and Y 4 are independently H or F;
  • R ' ' is hydroxy], thiol, Ci-6alkyl, Ci-ealkyl selectively fundi on alized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi, carbonyloxyl, Ci-ealkoxy, Ci- bh droxyalkoxy, amino, Ci-ealkylamino, di(Ci-6alkyi)amino, or azido groups, Ci- ealkoxy, Ci-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi, carbonyloxyl, Ct-shydroxyalkoxy, amino, Ci- 6alkylamino, di(Ci-6alkyl)amino, or azido groups, C3-5alkenyl-0-, Cs-salkynyl-O-, oligo(ethy!ene glycol), polyCethyiene glycol), borano (-BH 3 "
  • R 7 and R s are independently hydrogen, Ci-ealkyl , Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi, carbonyloxyl, Ci-ealkoxy, Cwhydroxyalkoxy, amino, Cwalkylamino, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Ci-f.alkyl)- selective!y functionalized with one or more halogen, thiol, hydroxy!, carbonyl carboxyi, carbonyloxyl, Ci-ealkoxy, Cwhydroxyalkoxy, amino, Ci- balkylammo, or groups, cyclic -(Cwoxaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl,
  • R 3 and R 4 are independently hydrogen, halogen, hydroxy!, ammo, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyi, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Cwalkylamino, di(Ci- 6 alkyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi.
  • R n and R 12 are independently selected from the group consisting of:
  • 7I2 7i3 7I4 is 7I6 717 7I8 7I 720 721 722 23 724 725 26 727 72S
  • W 3 , W 4 , W 5 , W 6 , W 7 , W 8 , and W 9 are independently hydrogen, halogen, hydroxyl, Ci-6alkyl, Cj-ealkyl selectively function alized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci-ealkylamino, di(Ci-&alkyl)arriino, or azido groups, Ci-6alkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cj-ehydroxyalkoxy, amino, Chalky lamino, di(Ci- 6alkyl)
  • the present invention provides a compound of Formula ie
  • X 3 , X 4 , X 5 , and X 6 are independently 0, NH, C3 ⁇ 4 CHF, or CF;?;
  • R and R i4 are independently selected from the roup consisting of:
  • 7l2 ⁇ 14 is 7I6 717 7I8 7I 720 21 722 723 724 725 726 727
  • n l i 72! are each independently CH o ;
  • W 3 , W 4 , W 5 , W 6 , and W 7 are independently hydrogen, halogen, hydroxy!, Ci- 6alkyl, Chalky 1 selectively functionalized with one or more halogen, thiol, liydroxyl, carbonyl, carboxyl, carbonyioxyl, Ci-ealkoxy, Ci- 6 hydroxyalkoxy, amino, Ci- ealkylarnino, di(Ci-6alkyl)amino, or azido groups, Ci-ealkoxy, Ci-ea!koxy selectively functionalized with one or more halogen, thiol, liydroxyl, carbonyl, carboxyl, carbonyioxyl, Ci-bhydroxyalkoxy, amino, Ci-eaikylamino, di(Ci-6alkyl)amino, or azido groups, Cs-salkenyl-O-, Cs-saikynyl-O-, oiigo(
  • R 3 and R 4 are independently hydrogen, halogen, hydroxyl, amino, d ⁇ alkyl, C 3 -ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, d-ehydroxyalkoxy, amino, d ⁇ alkylamino, di(d-6alkyl)amino, or azido groups, Ci-6alkoxy, d- ⁇ alkoxy selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl carbonyloxyl, d-ohydroxyaikoxy, amino, Ci- ealkyiammo, di(d- 6 alkyl)amino, or azido groups, d-salkenyl-O-, d-salkynyl-O-, oligo(ethylene glycol), polyiethylene glycol), azido, or -NR 7 R 8 ;
  • R 15 and R 16 are independently hydroxyl, thiol, methoxy, ethoxy, amino, N- methylamino, AyV-dimethylamino, N-ethylamino, N,N-diethylammo, N-morpholino, or borano ( ⁇ Bi h ⁇ ; or a pharmaceutically acceptable salt thereof.
  • tetrahydrofuranyl rings of Formula le is replaced by a sulfur or a selenium atom.
  • the compound is:
  • the compound is:
  • the compound is:
  • the compound is:
  • the present invention provides a compound of Formula If
  • X 1 and X 2 are independently 0, S or Se;
  • Z n , IP, Z u , Z 15 , Z 36 , and Z 17 are independently CH or N;
  • L 1 starting from the carbon alpha to X 1
  • L 2 starting from the carbon alpha to X 2
  • L 1 is independently -CH20-P(0)R 6 -0- CH20-P(S)R 6 -0-, -C(Y i )(Y 2 )0-P(0)R 6 -C(Y 3 )(Y 4 )-, - CH2NHSO2NH-, ⁇ i ! ⁇ ⁇ ) ⁇ ! ⁇ -.
  • c is 0, 1 , or 2; d, e, and f are independently 0 or 1 ;
  • Y 1 , Y 2 , Y 3 , and Y 4 are independently H or F;
  • R 6 is hydroxyl, thiol.
  • Ci-6alkyL Ci-ealk l selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl.
  • thiol hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- ealkylamino, di(Ci-6alkyl)amino, or azido groups, C3-5alk.enyl.-O-, Ci-salkyriyl-O-, oligo(ethylene glycol), poly(eihylene glycol), borano ⁇ - ⁇ ; ⁇ .. or -NR 7 R 8 ;
  • R 7 and R 8 are independently hydrogen, Ci-ealk l, Cr-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci-ealkylamino, di(Ci- 6a3kyl)amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Cwalkyi)- seiectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Cwhydroxyalkoxy, amino, Ci- 6alkylamino, or di(Ci-6alkyl)amino groups, cyclic -(Ci-eoxaalkyi)-, or cyclic - (Ci»60xaalkyl)- selectively functionalized with one or
  • R 3 and R 5 are independently hydrogen, halogen, hydroxyl amino, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cwalkoxy, Ci-ehydroxyaikoxy, amino, Cwalkylamino, di(Cwalkyl)amino, or azido groups, Ci-ealkoxy, selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-6hydroxyalkoxy, ammo, Ci- 6alkyiamino, di(Ci -6 alkyl)amino, or azido groups, Ci-salkenyl-O-, Ci-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR 7 R S ; or a pharmaceutically acceptable salt thereof.
  • the present invention provides a compound of Formula Ig
  • Z 12 , Z 13 , Z 1 , Z 15 , Z 16 , and Z" are independently CH or N;
  • R 3 and R 5 are independently hy drogen, halogen, hydroxyl, ammo, Ci- 6 alkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy], carbonvl, carboxyi, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci ⁇ alkyiamino.
  • Ci-ealkoxy Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyi, carbonyloxyl, Ci- 6 hydroxyalkoxy, amino, Ci- ealkylamino, di(Ci- 6 alkyl)amino, or azido groups, Cs-salkenyl-O-, C3-5alkynyl-0-, oligo(ethyiene glycol), poly(ethylene glycol), azido, or -NR 7 R 8 :
  • R 7 and R 8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy], carbonvl, carboxyi, carbonyloxyl, Cwalkoxy, Ci-chydroxyaikoxy, amino, Ci-ealkylamino, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Chalky!)-, cyclic -(Ci-ealkyi)- selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyi, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci-6alkylamino, or di(Ci- 6alkyl)amino groups, cyclic -(Ci-eoxaalkyl)-, or cyclic -(Ci-eoxaalkyl)- selectively functionalized with one or more halogen,
  • tetrahydrofuranyl rings of Formula Ig is replaced by a sulfur or a selenium atom
  • the present invention provides a compound of Formula Ih
  • X ! and X 2 are independently 0, S or Se;
  • L ! starting from the carbon alpha to X 1
  • LA starting from the carbon alpha to X 2
  • c is 0, 1 , or 2; d, e, and f are independently 0 or 1 ;
  • Y 1 , Y 2 , Y 3 , and Y 4 are independently H or F;
  • R 6 is hydroxy!, thiol, Ci-ealk l, Ci-ealkyl selectively functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl Ci-ealkoxy, Ci- ehydroxyalkoxy, amino, Ci-ealkylarnino, di(Cwalk>'l)amino, or azido groups, Ci- ealkoxy, Cwalkoxy selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- ealkw!amino, di(C :-6alkyl)amino, or azido groups, Cs-salkenyl-O-, C3.5alkyn.yl-O-, oligo
  • R ? and R 8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Cwhydroxyalkoxy, amino, Ci-ealkylamino, di(G- 6alkyl)amino, or azido groups, cyclic -(Ci-salkyi)-, cyclic -(Ci-ealkyl)- seiective!y functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-e.hydroxyalkoxy, amino, Ci- ea!kylammo, or di(Ci-6alkyl)amino groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively function
  • R 3 and R 5 are independently hydrogen, halogen, hydroxyl, amino, Cwalkyl, C 3 -ealkyl selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cwalkoxy, Ci-ehydroxyalkoxy, amino, Ci-6aikyiamino, di(Ci-6a3kyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cwhydroxyalkoxy, amino, Ci- 6alkylamino, di(Ci-6aiky!amino, or azido groups, C saikenyl-Q-, Cs-salkynyl-O-, oligo(ethy!ene glycol), poly(ethy!ene glycol), azido
  • R 11 and R 12 are independently selected from the group consisting of:
  • 212 2 13 ⁇ ⁇ 4 Z 15 Z 16 Z 17 Z 18 Z 19 Z 20 Z 2i Z 22 Z 23 Z 24 Z 25 Z 26 Z 27 Z 28 Z 29 , Z 30 , Z 31 , 7? Z? 7? Z 35 , Z 36 , and Z 37 are each independently CH or N; and ⁇ - W 4 , W 5 , W 6 , W 7 , W 8 , and W 9 are independently hydrogen, halogen, hydroxy!, Cwalkyl, Ci-ealkyl selectively funciionaiized with one or more halogen, thiol hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cwalkoxy, Ci-ehydroxyalkoxy, amino, Ci-aalkylamino, di(Ci-5alkyl)amino, or azido groups, Ci-6alkoxy, Ci-ealkoxy selectively funciionaiized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbon
  • the present invention provides a compound of Formula Ii
  • X 3 , X '5 , X 6 , and X 7 are independently 0, NH, CH>, CHF, or CF 2 ;
  • R 13 and R 14 are independently selected from the group consisting of:
  • Z 12 , Z i3 , Z 14 , Z 15 , Z 16 , Z 17 , 7 ⁇ Z 19 , ZP, Z 2 IP, Z 23 , Z 24 , Z 25 , Z 26 , Z 27 , and Z 2S are each independently CH or N; and
  • W 3 , W 4 , W 5 , W 6 , and W 7 are independently hydrogen, halogen, hydroxy!, Ci- ealkyl, selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyioxyl, Ci-ealkoxy, Ci ⁇ hydroxy alkoxy, amino, Ci- 6aikylamino, di(Ci-6aikyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, cai ' bonyloxyl, Ci-ehydroxyalkoxy, amino, Ci-salkylamino, di(Ci-6alkyl)amino, or azido groups, Ci-salkenyi-O-, C 3 -5aIkynyl-0-, oligo( ethylene glycol), poly(ethylene glycol), azido,
  • R 7 and R 8 are independently hydrogen, Cwalkyl, Ci-ealk l selectively functionalized with one or more halogen, thiol, hvdroxyl, carbonyl, carboxyl, carbonyloxyl, Ci- 6 alkoxy, Cwhydroxyalkoxy, amino, Ca ⁇ alkylamino, di(Ci- 6alky])amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Ci-f.alkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-6alkoxy, Ci-6hydroxyalkoxy, amino, Ci- 6alkylamino, or groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thiol, hvdr
  • R 3 and R 5 are independently hydrogen, halogen, hydroxyl, amino, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-&aikoxy, Ci-shydroxyalkoxy, amino, Ci-ealkylamino, di(Ci-6alkyl)amino, or azido groups, Ci-ealkoxy, Cwaikoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- ealkyiamino, di(Ci-6alkyi)amino, or azido groups, C ; .-Uilkcn ! ⁇ () ⁇ .
  • R 15 and R 16 are independently hydroxyl, thiol, methoxy, ethoxy, amino, N- methylamino, AyY-dimetlrylamino, N-ethylamino, N/V-diethylamino, N-rnorpholino, or borano (-BH 3 ⁇ ); or a pharmaceutically acceptable salt thereof.
  • tetrahydrofuratiyl rings of Formula li is replaced by a sulfur or a selenium atom
  • the compound is:
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.
  • the present invention provides a method of treating a disease or condition in which modulation of STING is beneficial comprising: administering to a patient in need thereof a compound of Formula I or a pharrnaceutically acceptable salt thereof.
  • the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease or condition in which modulation of STING is beneficial.
  • the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in therapy.
  • the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
  • the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, or a pharmaceutically composition thereof, such as a nanoparticle or a deliver ⁇ 7 vehicles that enhances the cellular uptake, stability and efficacy of a compound of Formula I for use in the treatment of cancer.
  • the present invention provides a method of treating cancer comprising: administering a therapeutically effective amount of a compound of Formula L or a pharmaceutically acceptable salt thereof, to a patient in need thereof.
  • the present invention provides the use of a compound of Formula 1, or a pharmaceutically acceptable salt thereof, in the manufacture of a medication for the treatment of cancer.
  • the present invention provides pharmaceutical composition
  • a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising; a compound of Formula I, or a pharmaceutically acceptable salt thereof, at least one further therapeutic agent, and one or more of pharmaceutically acceptable excipients.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in therapy.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising; a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in the treatment of a disease or condition for which modulation of STING is beneficial.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in the treatment of cancer,
  • the present invention provides a method for treating a disease or condition for which modulation of STING is beneficial comprising: administering to a patient in need thereof a therapeutically effective amount of a combination comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof and at least one further therapeutic agent.
  • the present invention provi des a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and ax least one further therapeutic agent for use in the treatment of cancer.
  • the therapeutic agent includes but is not limited to immune checkpoint inhibitors, such as humanized antibodies against PD1, PD-LL CTLA4 and other molecules that block effective anti-tumor immune responses,
  • the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent.
  • the therapeutic agent includes but is not limited to immune checkpoint inhibitors, such as humanized antibodies against PD1, PD-L1, CTLA4 and other molecules that block effective anti-tumor immune responses.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in the treatment of cancer.
  • the therapeutic agent includes radiation, such as high-dose radiation, which directly kills tumor cells, enhances presentation of tumor antigens and activates the STING pathway.
  • the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent.
  • the therapeutic agent includes radiation, such as high-dose radiation, which directly kills tumor cells, enhances presentation of tumor antigens and activates the STING pathway.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in the treatment of cancer.
  • the therapeutic agent includes another chemotherapeutic agent that selectively kills tumor cells and enhances presentation of tumor antigens,
  • the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent.
  • the therapeutic agent includes another chemotherapeutic agent that selectively kills tumor cells and enhances presentation of tumor antigens.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, a pharmaceutical formulation including a nanoparticie, and at least one further therapeutic agent for use in the treatment of cancer.
  • the therapeutic agent includes radiation and/or another chemotherapeutic agent,
  • the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula ⁇ , or a pharmaceutically acceptable salt thereof, a pharmaceutical formulation including a nanoparticie, and at least one further therapeutic agent for use in the treatment of cancer.
  • a pharmaceutical composition comprising: a compound of Formula ⁇ , or a pharmaceutically acceptable salt thereof, a pharmaceutical formulation including a nanoparticie, and at least one further therapeutic agent for use in the treatment of cancer.
  • the therapeutic agent includes radiation and/or another chemotherapeutic agent,
  • the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, a pharmaceutical formulation including a nanoparticie, and at least one further therapeutic agent for use in the treatment of cancer.
  • the compoun d of Formula L may be injected directly to tumors, or system! caliy, including injection into muscles (intramuscular), skins (subcutaneous and intra-dermal), peritoneal (intraperitoneal), lymph nodes (mtralymphatic) or veins (intravenous).
  • the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use as a vaccine adjuvant.
  • the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, such as a nanoparticie or a deliver ⁇ 7 vehicles that enhances the cellular uptake, stability and efficacy of a compound of Formula I, for use as a vaccine adjuvant.
  • the pharmaceutical composition is a vaccine.
  • the present invention provides a method of inducing or promoting an immune response comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical compositio comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, as an adjuvant and a tumor antigen.
  • the present invention provides a method of inducing or promoting an immune response comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I, or a pharmaceutical composition thereof, as an adjuvant, a tumor antigen, or a pharmaceutical composition thereof, such as a nanoparticle or a delivery vehicles that enhances the cellular uptake of the adjuvant and tumor antigen,
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, as an adjuvant and an immunogen for a target pathogen,
  • the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use as a vaccine adjuvant.
  • the present invention provides a method of inducing or promoting an immune response comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, as an adjuvant and an immunogen for a target pathogen,
  • the present invention provides a vaccine adjuvant comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof.
  • the present invention provides an immunogenic composition
  • an immunogenic composition comprising: an antigen or antigen composition and a compound of Formula I, or a pharmaceutically acceptable salt thereof.
  • the present invention provides an immunogenic composition
  • an immunogenic composition comprising: an antigen or antigen composition and a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a disease, including cancer and infectious diseases.
  • the present invention provides the use of a compound of Formula ⁇ , or a pharmaceutically acceptable salt thereof, for the manufacture of an immunogenic composition comprising an antigen or antigen composition, for the treatment or prevention of a disease, including cancer and infectious diseases.
  • the present invention pro vides a method of treating or preventing a disease comprising: administering to a patient suffering from or susceptible to the disease, an immunogenic composition comprising an antigen or antigen composition and a compound of Formula L or a pharmaceutically acceptable salt thereof.
  • the present invention provides a vaccine composition
  • a vaccine composition comprising: an antigen or antigen composition and a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a disease, including cancer and infectious diseases.
  • the present invention provides the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof, for the manufacture of a vaccine composition comprising an antigen or antigen composition for the treatment or pre vention of a disease, including cancer and infectious diseases.
  • the present invention provides a method of treating or preventing disease comprising the administration to a patient suffering from or susceptible to the disease, a vaccine composition comprising an antigen or antigen composition and a compound of Formula I, or a pharmaceutically acceptable salt thereof.
  • the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment of immune disorders, including autoimmune and autoinflammatory diseases.
  • the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, or a pharmaceutically composition thereof, such as a nanoparticie or a deliver ⁇ ' vehicles that enhances the cellular uptake, stability and efficacy of a compound of Formula I, for use in the treatment of immune disorders, including autoimmune and autoinflammatory diseases.
  • the present invention provides a method of treating immune disorders comprising: administering a therapeutically effective amount of a compound of Formula 1, or a pharmaceutically acceptable salt thereof, to a patient in need thereof.
  • the present invention provides the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof, in the manufacture of a medication for the treatment of immune disorders, including autoimmune and autoin ilammaton ' ' diseases,
  • the present, invention provides novel cGAMP analogs, pharmaceutical compositions thereof, and uses thereof in therapy.
  • 2'3'-cGAMP is an endogenous second messenger produced by mammalian cells, it is a high affinity ligand for STING, inducing conformational changes therein, and a potent inducer of type-I interferons.
  • cGAS and the cGAS-cGAMP pathway is important for triggering intlaimmatory responses to self and foreign DMA. As such, cGAS is important for immune defense against microbial pathogens that contain DNA and require DNA in their life cycles. These pathogens include DNA viruses, retroviruses including HI V, bacteria including mycobacterium tuberculosis, fungi and parasites.
  • cGAS can also detect tumor DNA and is important for the body's intrinsic immunity against malignant cells. Activation of the cG AS-cG AMP-STING pathway is importa t for cancer immunotherapy.
  • cGAMP As a potent inducer of type-1 interferons, cGAMP (and hence the cGAMP analogs of the present invention) provides a rational immune adjuvant.
  • a compound of Formula I or a pharmaceutically acceptable salt thereof may be used as a vaccine adjuvant, particularly with mucosal vaccines, and may be formulated with immunogens and delivered as have been cyclic-di-GMP and c-di-AMP as vaccine adjuvants (see, e.g.
  • the invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
  • the pharmaceutical composition is a compound of Formula 1.
  • the pharmaceutical composition is a compound of Formula I in a pharmaceutical formulation including ananoparticle or another delivery vehicle.
  • the pharmaceutical composition is a compound of Formula I in combination with at least one further therapeutic agent, which includes but is not limited to immune checkpoint inhibitors such as antibodies against PD-1, PD-L1 or CTLA-4,
  • the therapeutic agent used in combination with a compound of Formula I also includes radiation of tumors or a chemotherapeutic agent that targets tumor cells.
  • the invention provides a pharmaceutical composition comprising a compound of Formula 1 or a pharmaceutically acceptable salt thereof, as an adj uvant and an immunogen for a target pathogen, in one embodiment, the pharmaceutical composition is a vaccine.
  • the present invention provides a method of inducing or promoting an immune response comprising: administering to a patient in need thereof an effective amount a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, as an adjuvant and an immunogen for a target pathogen.
  • halo and halogen', alone or in combination with other groups, refers to fluoro-, chloro-, bromo- and iodo ⁇ .
  • Ci-e alkyi refers to monovalent, linear chain or branched chain alkyl groups containing from 1 to 6 carbon atoms.
  • Exemplar' Ci-e alkyl groups include but not limited to metheyl, ethyl, w-propyi, wo-propyl, n-butyl, sec-butyl and fer/-butyl groups. More preferred are Ci4 alkyls.
  • Cu- alkoxy refers to, alone or in combination with other groups, ! " ⁇ ( ⁇ . where R " is Ci-6 alkyl.
  • haloCi-ealkyF refers to a Ci-6 alkyl group subsituted with one or more halo suctsitutents, for example CF 3 and CH2CF3.
  • a compound of the invention or "a compound of Formula ⁇ includes ail solvates, complexes, polymorphs, radiolabeled derivatives, lautomers, stereoisomers, and optical isomers of the compoimcls of Formula I and salts thereof, unless otherwise specified.
  • the term "effective amount” means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician.
  • terapéuticaally effective amount means any amount which, as compared to a corresponding subject who Iras not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder.
  • the term also includes within its scope amounts effective to enhance normal physiological function.
  • prophylaxis includes prevention and refers to a measure or procedure which is to prevent rather than cure or treat a disease. Preventing refers to a reduction in risk of acquiring or developing a. disease causing at least one clinical symptom of the disease not to developing a subject that may be exposed to a disease-causing agent or a subject predisposed to the disease in advance of disease outset
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and dosage forms which are. within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable excipients includes all diluents, earners, binders, glidants, and other components of pharmaceutical formul ti ns with which the compound of the invention is administered.
  • the compounds of the invention may exist in solid or liquid form.
  • compound of the invention may exist in a continuum of solid states ranging from fully amorphous to fully crystalline.
  • the term 'amorphous' refers to a state in which the material lacks long range order at the molecular level and, depending upon the temperature, may exhibit the physical properties of a solid or a liquid. Typically, such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid. Upon heating, a change from solid to liquid properties occurs which is characterized by a change of state, typically second order ('glass transition').
  • 'crystalline' refers to a solid phase in which the material has a regular ordered internal structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterized by a phase change, typically first order ('melting point').
  • the compounds of the invention may have the ability to crystallize in more than one form a characteristic, which is known as polymorphism, and it is understood that such polymorphic forms (“polymorphs ' ') are within the scope of the invention.
  • Polymorphism generally can occur as a response to changes in temperature or pressure or both and can also result from variations in the crystallization process.
  • Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility and melting point
  • the compound of Formula I may exist in solvated and unsolvated forms.
  • solvate refers to a complex of variable stoichiometry formed by a solute (in. this invention, a compound of Formula 1 or a salt) and a solvent. Such solvents for the purpose of the invention may not interfere with the biological activity of the solute.
  • pharmaceutically acceptable solvates may be formed for crystalline compounds wherein solvent molecules are incorporated into the crystalline lattice during cry stallization.
  • the incorporated solvent molecules may be water molecules or nonaqueous such as ethanol, isopropanol, DMSO, acetic acid, eiharsolamrae, and ethyl acetate molecules. Crystalline lattice incorporated with water molecules are typically referred to as "hydrates " . Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. The present invention includes ail such solvates.
  • tautomers refer to compounds that are interchangeable forms of a particuiar compound structure, and that van' in the displacement of hydrogen atoms and electrons. Thus, two structures may be in equilibrium through the movement of re electrons and an atom (usually H).
  • enols and ketones are tautomers because they are rapidly intercom- erted by treatment with either acid or base. It is understood that all tautomers and mixtures of tautomers of the compounds of the present invention are included within the scope of the compounds of the present invention.
  • the compounds of Formula I may be in tire form of a salt.
  • the salts of the present invention are pharmaceutically acceptable salts.
  • Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention.
  • suitable pharmaceutically acceptable salts can include acid addition salts.
  • a pharmaceutically acceptable acid addition salt can be formed by reaction of a compound of Formula I with a suitable inorganic or organic acid (such as hydrobromic, hydrochloric, sulfuric, nitric, phosphor!
  • a pharmaceutically acceptable acid addition salt of a compound of Formula 1 can be, for example, a hydrobromide. hydrochloride, sulfate, nitrate, phosphate, p- ioluenesulibnate, benzenesulfonate. methanesulfonate, ethanesulibnate, or
  • naphthalenesulfonate e.g. 2-naphthalenesulforiate
  • Other non-phannaceutically acceptable salts e.g. trifluoroacetates, may be used, for example in the isolation of compounds of the invention, and are included within the scope of this invention.
  • the invention includes within its scope all possible stoichiometric and non- stoichiometric forms of the compounds of Formula I.
  • the compound of the invention may be administered as the raw chemical, it is possible to present the compound of the invention as the active ingredient in a pharmaceutical composition.
  • Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
  • t invention further provides pharmaceutical compositions comprising a compound of the invention and one or more pharmaceutically acceptable excipients.
  • the excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof.
  • a process for the preparation of a pharmaceutical composition including the agent, or pharmaceutically acceptable salts thereof, with one or more pharmaceutically acceptable excipients.
  • the pharmaceutical composition can be for use in the treatment and'or prophylaxis of any of the conditions described herein.
  • the compound of the invention is administered in a
  • compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical exeipient, vehicle or carrier. Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • Preferred unit dosage compositions are those containing a daily dose or sub- dose, or an appropriate fraction thereof, of an active ingredient. Such unit doses may therefore be administered once or more than once a day. Such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art.
  • compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, inhaled, intranasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route.
  • Such compositions may be prepared by any meihod known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
  • compositions adapted for oral administrat on may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in- oil liquid emulsions.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert excipient such as ethanol, glycerol, water and the like.
  • an oral, non-toxic pharmaceutically acceptable inert excipient such as ethanol, glycerol, water and the like.
  • Powders are prepared by reducing the compound to a suitable fine size and mixing with a similarly prepared pharmaceutical excipient such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
  • Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths. Excipients including glidams and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation. A disintegrating or soiubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
  • Excipients including glidams and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation.
  • a disintegrating or soiubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
  • excipients including suitable binders, glidams, lubricants, sweetening agents, flavors, disintegrating agents and coloring agents can also be incorporated into the mixture.
  • suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, com sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymelhylcel] ulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets.
  • a powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymeihylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/ ' or an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • the powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cel!ulosic or polymeric materials and forcing through a screen.
  • the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules.
  • the granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate sail, talc or mineral oil.
  • the lubricated mixture is then compressed into tablets.
  • the compounds of the present invention can also be combined with a. free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps.
  • a clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
  • Oral fluids such as solution, suspensions, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound.
  • Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle.
  • Suspensions can be formulated by dispersing the compound in a non-toxic vehicle.
  • Solubiiizers and emuisifiers such as ethoxylated isosiear l alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
  • dosage unit compositions for oral administration can be microencapsulated,
  • the composition can also be prepared to prolong or sustam the release as for example by coating or embedding particulate material in polymers, wax or the like.
  • the compounds of the invention may also be administered in the form of liposome delivery sy tems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • Pharmaceutical compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils,
  • compositions are preferably applied as a. topical ointment or cream
  • topical ointment or cream When formulated in an ointment, the active ingredient may be employed with either a paraffmic or a water-mi scibie ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil -in- water cream base or a water-in-oil base.
  • Pharmaceutical compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
  • compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
  • compositions adapted for rectal administration may be presented as suppositories or as enemas.
  • Dosage forms tor nasal or inhaled administration may conveniently be formulated as aerosols, solutions, suspension drops, gels or dry powders.
  • compositions for intranasal administration include aqueous compositions administered to the nose by drops or by pressurised pump. Suitable compositions contain water as the diluent or carrier for this purpose.
  • Compositions for administration to the lung or nose may contain one or more excipients, for example one or more suspending agents, one or more preservatives, one or more surfactants, one or more tonicity adjusting agents, one or more co-solvents, and may include components to control the pH of the composition, for example a buffer system. Further, the compositions may contain other excipients such as antioxidants, for example sodium metabisulphite, and taste-masking agents.
  • compositions may also be administered to the nose or other regions of the respiratory traci by nebulization.
  • intranasal compositions may permit the compound(s) of Formula I or (a) pharmaceutically acceptable sait(s) thereof to be delivered to all areas of the nasal cavities (the target tissue) and further, may permit the compound(s) of Formula ⁇ or (a) pharmaceutically acceptable salt(s) thereof to remain in contact with the target tissue for longer periods of time.
  • a suitable dosmg regime for intranasal compositions would be for the patient to inhale slowly through the nose subsequent to the nasal cavity being cleared. During inhalation, the composition would be administered to one nostril while the other is manually compressed. This procedure would then be repeated for the other nostril.
  • one or two sprays per nostril would be administered by the above procedure one, two, or three tim.es each day, ideally once daily.
  • intranasal compositions suitable for once- daily administration are particularly useful for once- daily administration.
  • the suspending agent(s), if included, will typically be present in an amount of from 0.1 to 5% (w/w), such as from 1.5% to 2.4% (w/w), based on the total weight of the composition.
  • pharmaceutically acceptable suspending agents include, but are not limited to, Avicef (niiciOerystallme cellulose and carboxymethylceliulose sodium), carboxymethyicelluiose sodium, veegum, tragacantk bentonite, me!hylcelmlose, xantban gum, carbopol and polyethylene glycols.
  • compositions for administration to the king or nose may contain one or more excipients may be protected from microbial or fungal contamination and growth by inclusion of one or more preservatives.
  • pharmaceutically acceptable ami -microbial agents or preservatives include, but are not limited to, quaternary ammonium compounds (for example benzalkonium chloride, benzethonium chloride, cetrimide, cetylpyridmium chloride, lauralkonujm chloride and myristyl picolinium chloride), mercurial agents (for example phenyimercuric nitrate, phenylmercuric acetate and thimerosai), alcoholic agents (for example chlorobutanol, phenviethyi alcohol and benzyl alcohol), aniibacienai esters (for example esters of para-hydroxybenzoic acid), chelating agents such as di sodium edetate (EDTA) and other anti -microbial agents such as chlorhexidine,
  • compositions may include one or more surfactants which functions to facilitate dissolution of the medicament particles in the aqueous phase of the composition.
  • the amount of surfactant used is an amount which will not cause foaming during mixing.
  • Examples of pharmaceutically acceptable surfactants include fatty alcohols, esters and ethers, such as polyox ethylene (20) sorbitan monooleate (Poiysorbate 80), macrogol ethers, and poloxamers.
  • the surfactant may be present in an amount of between about 0.01 to 10% (w/w), such as from 0.01 to 0.75% (w/w), for example about 0.5% (w/w), based on the total weight of the composit n.
  • One or more tonicity-adjusting agent(s) may be included to achieve tonicity with body fluids e.g. fluids of the nasal cavity, resulting in reduced levels of irritancy.
  • Examples of pharmace tically acceptable tonicity-adjusting agents include, but are not limited to, sodium chloride, dextrose, xylitol, calcium chloride, glucose, glycerine and sorbitol.
  • a tonicity-adjusting agent, if present, may be included in an amount of from 0.1 to 10% (vv/w), such as from 4.5 to 5.5% (w/w), for example about 5.0% (w/w), based on the total weight of the composition.
  • compositions of the invention may be buffered by the addition of suitable buffering agents such as sodium citrate, citric acid, trometamol, phosphates such as disodium phosphate (for example the dodecahydrate, heptahydrate, dihydrate and anhydrous forms), or sodium phosphate and mixtures thereof [0139]
  • a buffering agent, if present, may be included in an amount of from 0.1 to 5% (w w), for example I to 3% (w/w) based on the total weight of the compositi n.
  • taste-masking agents include sucraiose. sucrose, saccharin or a salt thereof, fructose, dextrose, glycerol, corn syrup, aspartame, acesulfan e- . xylitoi, sorbitol, er thritol, ammonium glycyrrhizinate, triaumatm, neotanje, mannitol, menthol, eucalyptus oil camphor, a natural flavouring agent, an artificial flavouring agent, and combinations thereof.
  • One or more eo-sumble(s) may be included to aid solubility of the medicament cornpound(s) and/or other excipients.
  • pharmaceutically acceptable co-solvents include, but are not limited to, propylene glycol, dipropylene glycol, ethylene glycol glycerol, ethanol, polyethylene glycols (for example PEG300 or PEG400), and methanol.
  • the co-solvent is propylene glycol .
  • Co-sol vent(s), if present, may be included in an amount of from 0.05 to 30% (w/w), such as from 1 to 25% (w/w), for example from 1 to 10% (w/w) based on the total weight of the composition.
  • compositions for inhaled administration include aqueous, organic or aqueous/organic mixtures, dry powder or crystalline compositions admini tered to the respiratory tract by pressurised pump or inhaler, for example, reservoir dry powder inhalers, unit-dose dry- powder inhalers, pre-metered multi-dose dry powder inhalers, nasal inhalers or pressurised aerosol inhalers, nebulisers or insufflators.
  • Suitable compositions contain water as the diluent or carrier for this purpose and may be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like.
  • Aqueous compositions may also be administered to the nose and other regions of the respiratory tract by nebulisation.
  • Such compositions may be aqueous solutions or suspensions or aerosols delivered from pressurised packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant.
  • compositions for admini tration topically to the nose (for example, for the treatment of rhinitis) or to the lung include pressurised aerosol, compositions and aqueous compositions delivered to the nasal cavities by pressurised pump.
  • Compositions which are non- pressurised and are suitable for administration topically to the nasal cavity are of particular interest Suitable compositions contain water as the diluent or carrier for this purpose.
  • Aqueous compositions for administrati n to the lung or nose may be provided with conventional excipients such as buffering agents, tomcit -modifying agents and the like.
  • Aqueous compositions may also be administered to the nose by nebulisation.
  • a iluid dispenser may typically be used to deliver a fluid composition to the nasal cavities.
  • the iluid composition may be aqueous or non-aqueous, but typically aqueous.
  • Such a fluid dispenser may have a dispensing nozzle or dispensing orifice through which a metered dose of the fluid composition is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser.
  • Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid composition the doses being dispensable upon sequential pump actuations.
  • the dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid composition into the nasal cavity.
  • Dry powder compositions for topical delivery to the Sung by inhalation may. for example, be presented in capsules and cartridges of for example gelatine, or blisters of er example laminated aluminium foil, for use in an inhaler or insufflator.
  • Powder blend compositions generally contain a powder mix for inhalation of the compound of Formula ⁇ or a pharmaceutically acceptable salt thereof and a suitable powder base
  • carrier/dil uent/excipient substance such as mono-, di-, or polysaccharides (for example lactose or starch).
  • Dry powder compositions may also include, in addition to die drug and carrier, a further excipient (for example a ternary agent such as a sugar ester for example ceilobiose octaacetate, calcium stearate, or magnesium siearate.
  • a further excipient for example a ternary agent such as a sugar ester for example ceilobiose octaacetate, calcium stearate, or magnesium siearate.
  • compositions adapted for parental administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacienosiats and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • compositions may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include flavoring agents.
  • compositions may contain antibody(ies) or antibody fragment(s) or an antigenic component including but not limited to protein, DNA, live or dead bacteria and/or viruses or virus-like particles, together with one or more components with adjuvant activity including but not limited to aluminium salts, oil and water emulsions, heat shock proteins, lipid A preparations and derivatives, glycolipids, other TLR agonists such as CpG DNA or similar agents, cytokines such as GM-CSF or IL-12 or similar agents.
  • antibody(ies) or antibody fragment(s) or an antigenic component including but not limited to protein, DNA, live or dead bacteria and/or viruses or virus-like particles, together with one or more components with adjuvant activity including but not limited to aluminium salts, oil and water emulsions, heat shock proteins, lipid A preparations and derivatives, glycolipids, other TLR agonists such as CpG DNA or similar agents, cytokines such as GM-CSF or IL-12 or similar agents.
  • a therapeutically effective amount of the agent will depend upon a number of factors including, for example, the age and weight of the subject, the precise condition requiring treatment and its seventy, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian.
  • the subject to be treated is a mammal, particularly a human.
  • the agent may be administered in a daily dose. This amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or six) of sub- doses per day such that the total daily dose is the same.
  • the amount of the compound of the invention administered according to the present invention w ll be an amount selected from about 0.01 mg to about 1 g per day (calculated as the free or unsalted compound).
  • the compounds of Formula I and pharmaceutically acceptable salts thereof may be employed alone or in combination with other therapeutic agents.
  • the compounds of Formula I and pharmaceutically acceptable salts thereof and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately, administration may occur simultaneously or sequentially, in any order, by any convenient route in separate or combined pharmaceutical compositions.
  • the amounts of the compound(s) of Formula I or pharmaceutically acceptable salt(s) thereof and the other pharmaceutically active agent(s) and the relati ve timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • the compound(s) of Formula 1 or pharmaceutically acceptable salt(s) thereof and further therapeutic agent(s) may be emploved in combination by administration simultaneously in a unitary pharmaceutical composition including both compounds.
  • the combination may be administered separately in separate pharmaceutical compositions, each including one of the compounds in a sequential manner wherein, for example, the compound of the invention is administered first and the other second and vice versa, Such sequential administration may be close in time (e.g.
  • the compounds are administered in the same dosage form, e.g. one compound may be administered topically and the other compound may be administered orally. Suitably, both compounds are administered orally.
  • the combinations may be presented as a combination kit.
  • kit or kit of parts
  • the pharmaceutical composition or compositions that are used to administer the combination according to the invention When both compounds are administered simultaneously, the combination kit ca contain both compounds in a single pharmaceutical composition, such as a tablet, or in separate pharmaceutical compositions. When the compounds are not administered simultaneously, the combination kit will contain each compound in separate pharmaceutical compositions either in a single package or in separate pharmaceutical compositions in separate packages.
  • the combination kit can also be provided by instruction, such as dosage and administration instructions.
  • dosage and administration instructions can be of the kind that are provided to a doctor, for example by a drug product label, or they can be of the kind that are provided by a doctor, such as instructions to a patient.
  • sequential administration may be close in time or remote in time.
  • administration of the other agent several minutes to several dozen minutes after the administration of the first agent, and administration of the other agent several hours to several days after the administration of the first agent are included, wherein the lapse of time is not limited.
  • one agent may be administered once a day, and the other agent may be administered 2 or 3 times a day, or one agent may be administered once a week, and the other agent may be administered once a day and the like.
  • the other therapeutic ingredients(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alky] esters, or as solvates, for example hydrates, to optimize the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic ingredient. It will be clear also that, where appropriate, the therapeutic ingredients may be used m optically pure form.
  • the two compounds must be stable and compatible with each other and the other components of the composition and may be formulated for administration. When formulated separately they may be provided in any convenient composition, conveniently, in such a manner as known for such compounds in the ail.
  • the dose of each compound may differ from that when the compound is used alone. Appropri ate doses will be readily appreciated by those skilled in the art.
  • the patient in the methods and uses of the present invention is a mammal. In another embodiment, the patient is a human.
  • the compounds of the invention are useful in the treatment of diseases and conditions in which modulation of STING is beneficial, including cancer. As modulators of the immune response, the compounds of Formula I and pharmaceutically acceptable salts thereof may also be useful, as stand-alone, in combination or as adjuvants, in the treatment of diseases and conditions in which modulation of STING is beneficial.
  • the disease or condition to be treated is cancer.
  • cancer diseases and conditions in which a compound of Formula 1 or pharmaceutically acceptable salt thereof, may have potentially beneficial anti-tumor effects include cancers of the lung, bone, pancreas, skin, head, neck, uterus, ovaries, stomach, colon, breast, esophagus, small intestine, bowel, endocrine system, thyroid gland, parathyroid gland, adrenal gland, urethra, prostate, penis, testes, ureter, bladder, kidney or liver; rectal cancer; cancer of the anal region; carcinomas of the fallopian tubes, endometrium, cervix, vagina, vulva, renal pelvis, renal cell; sarcoma of soft tissue; myxoma; rhabdomyoma; fibroma, lipoma; teratoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hemagioma;
  • the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
  • the present invention provides a method of treating cancer comprising administering to a patient in need thereof a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof.
  • the present invention provides the use of a compound of Formula I or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
  • a compound of the invention may be employed with other therapeutic methods of cancer treatment, e.g., in anti-neoplastic therapy, combination therapy with immune checkpoint inhibitors, other chemotherapeutic, hormonal, antibody agents as well as surgical and/or radiation treatments.
  • Immune checkpoint inhibitors such as humanized antibodies against PD-1 , PD-L1 and CTLA4, have recently been shown to be highly successful in treating several types of metastatic cancer, including melanoma, non-small cell lung cancers, renal cell carcinoma and bladder cancer (Sharma and Allison, 2015, Science 348, 56).
  • checkporat inhibitor therapies in part because insufficient number of anti-tumor immune cells, such as CDS T cells, are generated and/or infiltrated into the tumors.
  • Activation of the cGAS-STING pathway activates anti -tumor immunity, including the production and infiltration of tumor-specific CDS T cells. Therefore, cGAMP analogues are expected to function synergistically with immune checkpoint inhibitors and the combination therapies are likely to bring therapeutic benefits to a larger percentage of cancer patients.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one immune checkpoint inhibitor.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one immune checkpoint inhibitor for use in therapy.
  • the present invention provides a pharmaceutical composition comprising a compound of Formula I or pharmaceutically acceptable salt thereof, and at least one immune checkpoint inhibitor for use in treating cancer.
  • the present invention provides the use of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one immune checkpoint inhibitor in the manufacture of a medicament for the treatment of cancer.
  • the present invention provides a method of treating cancer, comprising; administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least immune checkpoint inhibitor.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, at least one immune checkpoint inhibitor, and one or more of pharmaceutically acceptable carriers, diluents and excipients.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with radiation therapy such as SBRT.
  • the present invention provides a pharmaceutical composition comprising a compound of Formula I or pharmaceutically acceptable salt thereof, in combination with radiation therapy such as SBRT for use in treating cancer.
  • the present invention provides the use of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with radiation therapy such as SBRT in the manufacture of a medicament for the treatment of cancer.
  • the present invention provides a method of treating cancer, comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with radiation therapy such as SBRT.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and one or more of pharmaceutically acceptable carriers, diluents and excipients, in combination with radiation therapy such as SBRT for the treatment of cancer.
  • Anti-neoplastic agents include chemical compounds and antibodies that kill tumor ceils by inhibiting ceil cycle, signal transduction, DNA metabolism and angiogenesis and/or by promoting DNA damage, apoptosis and necrosis. These agents comprise that largest class of molecules currently used for cancer therapies. Anti-neoplastic agents selectively kill tumor cells, although many of them al so kill normal cel ls, thereby generating severe side effects. Processing of dead tumor cell associated antigens by antigen presenting cells leads to the generation of tumor-specific cytotoxic T cells. Tins process can be enhanced by cGAMP analogues. Therefore, combination of cGAMP analogues with anti-neoplastic agents are likely to generate synergistic effects that benefit a larger percentage of patients,
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula 1 or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent for use in therapy.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent for use in treating cancer.
  • the present invention provides the use of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent in the manufacture of a medicament for the treatment of cancer.
  • the present invention provides a method of treating cancer, comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula 1 or a pharmaceutically acceptable salt thereof, at least one anti-neoplastic agent, and one or more of pharmaceutically acceptable carriers, diluents and excipients,
  • anti-neoplastic agent that has activity versus a susceptible tumor being treated may be utilized in the combination.
  • Typical anti-neoplastic agents useful include anti- microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazapliosphorines, alkylsulfonates, nitrosoureas, and tnazenes; antibiotic agents such as anthracyclins, actinomycms and bleomycins; topoisoraerase ⁇ inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and anti -folate compounds; topoisoraerase I inhibitors such as camptoihecins: hormones and hormonal analogues; signal transduction pathway inhibitors; non-receptor tyrosine angiogenesis inhibitors: immunotherapeutic agents; proapoptotic agents; and cell cycle signaling
  • anti-microtubule agents examples include diterpenoids and vinca alkaloids.
  • Diterpenoids which are derived from natural sources, are phase specific anticancer agents that operate at the G2/M phases of the cell cycle. It is believed that the diterpenoids stabilize the ⁇ -tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include paclitaxel and its analog docetaxei.
  • Vinca alkaloids are phase specific an ti -neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include vinblastine, vincristine, and vmorelbine.
  • Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA.
  • the platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor.
  • Examples of platinum coordination complexes include oxaiiplatin, cisplatin and carboplatin.
  • Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleopliilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxy!, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death. Examples of alkylating agents include nitrogen mustards such as cyclophosphamide, melphalan. and chlorambucil; alky! sulfonates such as busu!fan;
  • nitrosoureas such as carmustine
  • triazenes such as dacarbazine
  • Antibiotic anti-neoplastics are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to ceil death.
  • antibiotic anti-neoplastic agents include actinomycins such as dactinomycin, anthrocyclins such as daunorubicin and doxorubicin; and bleomycins.
  • Topoisomerase ⁇ inhibitors include epipodophyllotoxins.
  • Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DN A causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include etoposide and teniposide.
  • Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows.
  • Examples of antimetabolite anti-neoplastic agents include fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine.
  • Camptothecins including, caraptothecin and camptothecin derivatives are available or under development as Topoisomerase I inhibitors
  • Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity.
  • camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4- methy Ipiperazmo-methylene)- 10, 11 -ethylenedioxy-20-camptothecin described below.
  • Hormones and hormonal analogues are useful compounds for treating cancers in which there is a relationship between the hormone(s) and growth and/or lack of growth of the cancer.
  • hormones and hormonal analogues useful in cancer treatment include adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lymphoma and acute leukemia in children; aminoglutetihimide and other aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane useful in the treatment of adrenocortical carcinoma and hormone dependent breast carcinoma containing estrogen receptors; progestins such as megestroi acetate useful in the treatment of hormone dependent breast cancer and endometria 1 carcinoma; estrogens, and anti-estrogens such as fulvestrant, flutamide.
  • adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lympho
  • nilutamide, bicaiutamide, cyproterone acetate and 5a- reductases such as finasteride and dutasteride, useful in the treatment of prostatic carcinoma and benign prostatic hypertrophy; anti-estrogens such as tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene, as well as selective estrogen receptor modulators (SERMS) useful in the treatment of hormone dependent breast carcinoma and other susceptible cancers; and gonadotropin- releasing hormone (GnRH) and analogues thereof which stimulate the release of leutini ing hormone (LH) and/or follicle stimulating hormone (FSH) for the treatment prostatic carcinoma, for instance, LHRH agonists and antagagonists such as goserelin acetate and luprolide.
  • LHRH agonists and antagagonists such as goserelin acetate and luprolide.
  • Signal transduction pathway inhibitors are those inhibitors, which block or inhibit a chemical process which evokes a intracellular change. As used herein this change is cell proliferation or differentiation.
  • Signal transduction inhibitors useful in the present invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3domam blockers, serine/threonine kinases, phosphotidyl inosito!-3 kinases, myoinositol signaling, and Ras oncogenes.
  • protein tyrosine kinases catalyze the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth.
  • protein tyrosine kinases can be broadly classified as receptor or non-receptor kinases.
  • Receptor tyrosine kinases are transmembrane proteins having an extracellular ligand binding domain, a transmembrane domain, and a tyrosine kinase domain. Receptor tyrosine kinases are involved in the regulation of cell growth and are generally termed growth factor receptors. Inappropriate or uncontrolled activation of many of these kinases, i.e.
  • Growth factor receptors include, for example, epidermal growth factor receptor (EGFr), platelet derived growth factor receptor (PDGFr), erbB2, erbB4, ret, vascular endothelial growth factor receptor (VEGFr).
  • TIE-2 immunoglobulin-like and epidermal growth factor homology domains
  • TGFI insulin growlh factor -I
  • cfms macrophage colony stimulating factor
  • BTK ckit
  • cmet fibroblast growth factor
  • Trk receptors TrkA, TrkB, and TrkC
  • ephrin (eph) receptors eph receptors
  • RET protooncogene RET protooncogene.
  • inhibitors of growth receptors are under development and include ligand antagonists, antibodies, tyrosine kinase inhibitors and anti-sense oligonucleotides.
  • Growth factor receptors and agents that inhibit growth factor receptor function are described, e.g., in Kaih, John ( ' . Exp. Opin. Ther. Patents (2000) 10(6):803-818; Shawver et al. DDT Vol 2, No. 2 February 1997; and Lofts, F. J. et al in "Growth factor receptors as targets", New Molecular Targets for Cancer Chemotherapy, ed. Workman, Paul and Kerr, David, CRC press 1994, London.
  • Tyrosine kinases which are not growth factor receptor kinases are termed nonreceptor tyrosine kinases.
  • Non-receptor tyrosine kinases useful in the present invention include cSrc, Lck, Fyn, Yes, Jak, cAbl, FAK (Focal adhesion kinase). Brutons tyrosine kinase, and Bcr-Abl.
  • SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including PI3-K p85 subunit, Src family kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP.
  • SH2/SH3 domains as targets for anti-cancer drugs are discussed in Smithgall, T.E. (1995), Journal of Pharmacological and Toxicoiogical Methods. 34(3) 125-32,
  • Inhibitors of Serine/Threonine Kinases include MAP kinase cascade blockers which include blockers of Raf kinases (rafk), Mitogen or Extracellular Regulated Kinase (M EKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family member blockers including blockers of PKCs (alpha, beta, gamma, epsilon, mu, lambda, iota, zeta), I kB kinase family (I KKa, I KKb), PKB family kinases, akt kinase family members, and TGF beta receptor kinases.
  • PKCs alpha, beta, gamma, epsilon, mu, lambda, iota, zeta
  • I kB kinase family I kB kinase family
  • I KKa, I KKb PKB family kin
  • Serine/Threonine kinases and inhibitors thereof are described, e.g., in Yamamoto, T. et ah, (1999), Journal of Biochemistry, 126 (5) 799-803; Brodt, P et al. (2000), Biochemical Pharmacol ogy, 60. 1101 -1 107; Massague, J., and Weis- Garcia, F. (1996) Cancer Surveys. 27:41-64; Philip, P.A.; and Harris, A.L. (1995), Cancer Treatment and Research. 78: 3-27; Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10). 2000, 223-226; U.S. Patent No. 6,268,391 ; and Martinez-lacaci, L, ei al, Int. J. Cancer (2000), 88(1), 44-52.
  • Inhibitors of Phosphotidyl inositol-3 Kinase family members including blockers of PT3-kinase, ATM, DNA-PK, and Ku are also useful in the present invention.
  • Such kinases are discussed, e.g., in Abraham, R.T. (1996), Current Opinion in Immunology. 8 (3) 412-8; Canman, C.E., Lim, D.S. (1998), Oncogene 17 (25) 3301-3308; Jackson, S.P. (1997), International Journal of Biochemistry and Cell Biology. 29 (7):935-8; and Zhong, H. et al. Cancer Res., (2000) 60(6), 1541 -1545.
  • Myo-inositol signaling inhibitors such as phospholipase C blockers and Myoinositol analogues.
  • signal inhibitors are described, e.g., in Powis, G,, and Kozikowski A., (1994) New Molecular Targets for Cancer
  • Another group of signal transduction pathway inhibitors are inhibitors of Ras Oncogene.
  • Such inhibitors include inhibitors of farnesyltransferase, geranyl-geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy.
  • Such inhibitors have been shown to block ras activation in cells containing wild type mutant ras, thereby acting as antiprohferation agents.
  • Ras oncogene inhibition is discussed in Scharovsky, O.G., et al. (2000), Journal of Biomedical Science. 7(4) 292-8; Ashby, M.N. (1998), Current Opinion in Lipidoiogy. 9 (2) 99 - 102; and BioChim. Biophys. Acta, (19899) 1423(3): 19-30.
  • Antibody antagonists to receptor kinase ligand binding may also serve as signal transduction inhibitors.
  • This group of signal transduction pathway inhibitors includes the use of humanized antibodies to the extracellular ligand binding domain of receptor tyrosine kinases. Examples include Imclone C225 EGFR specific antibody (see Green, M.C. et al, Monoclonal Antibody Therapy for Solid Tumors, Cancer Treat.
  • Herceptin ⁇ f> erbB2 antibody see Tyrosine Kinase Signalling in Breast cancer: erbB Family Receptor Tyrosine Kinases, Breast cancer Res., 2000, 2(3), 176- 183
  • 2CB VEGFR2 specific antibody see Brekken, R. A. et al, Selective inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks tumor growth in mice, Cancer Res. (2000) 60, 5117-5124).
  • Anti-angiogenic agents such as non-receptor MEK. angiogenesis inhibitors may also be useful, as well as those which inhibit the effects of vascular endothelial growth factor (e.g., the anti-vascular endothelial cell growth factor antibody bevacizumab
  • vasiinTM compounds that work by other mechanisms (e.g., linomide, inhibitors of integrin ⁇ 3 function, endostatin and angiostatin).
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti -neoplastic agent which is an anti-mi crotubule agent, platinum coordination complex, alkylating agent, antibiotic agent, topoisomerase 11 inhibitor, antimetabolite, topoisomerase I inhibitor, hormones and hormonal analogue, signal transduction path way- inhibitor, non-receptor tyrosine MEK angiogenesis inhibitor, immunotherapeutic agent, proapoptotic agent, or cell cycle signaling inhibitor.
  • an anti-neoplastic agent which is an anti-mi crotubule agent, platinum coordination complex, alkylating agent, antibiotic agent, topoisomerase 11 inhibitor, antimetabolite, topoisomerase I inhibitor, hormones and hormonal analogue, signal transduction path way- inhibitor, non-receptor tyrosine MEK angiogenesis inhibitor, immunotherapeutic agent, proapoptotic agent, or cell cycle signaling inhibitor.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent selected from diterpenoids and vinca alkaloids,
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti -neoplastic agent which is a platinum coordination complex.
  • at least one anti-neoplastic agent is paclitaxel, carbop!atin, or vinorelbine.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent which is a signal transduction pathway inhibitor
  • the signal transduction pathway inhibitor is an inhibitor of a growth factor receptor kinase VEGFR2, TIE2, PDGFR, BTK, erbB2, EGFr, IGFR-i , TrkA, TrkB, TrkC, or c-fms.
  • the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase rafk, akt, or P C-zeta.
  • the signal transduction pathway inhibitor is an inhibitor of a non- receptor tyrosine kinase selected from the src family of kinases. In another embodiment, the signal transduction pathway inhibitor is an inhibitor of c-src. In another embodiment, the signal transduction pathway inhibitor is an inhibitor of Ras oncogene selected from inhibitors of farnesyl transferase and geranylgeranyl transferase. In another embodiment, the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase selected from the group consisting of PI3K.
  • the signal transduction pathway inhibitor is a dual EGFr/erhB2 inhibitor, for example N- ⁇ 3-Chloro-4-[(3-fluorobenzy]) oxy]phenyl ⁇ -6-[5- ( ⁇ [2- (methanesulphonyl) ethyl]amino ⁇ methyl)-2-fur ⁇ 'l]-4-quinazolinamine.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti -neoplastic agent which is a cell cycle signaling inhibitor.
  • the cell cycle signaling inhibitor is an inhibitor of CDK2. CDK4 or CDK6.
  • Compounds of Formula I may be prepared by methods known in the art of organic synthesis as set forth in the schemes below and/or the specific Examples described below. In all of the methods, it is well understood that protecting groups for sensitive or reactive groups may be employed where necessary in accordance with general principles of chemistry . Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Green and P. G. M. Wuts (1.999) Protective Groups in Organic Synthesis, 3 ⁇ edition, John Wiley & Sons). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection of processes as well as the reaction conditions and order of their execution shall be consistent with the preparation of compounds of Formula 1.
  • TFA is tfifiuofoacetic acid
  • TFE is 2,2,2-trifluoroethyl
  • THE is tetrahydrofuran
  • TIPS is triisopropylsilyl
  • TEC is thin-layer chromatography
  • TMS is trimethylsiiyl: TMSOTf is trimethylsilyi trifluoromethanesulfonate: Tol is -toluoyl; Tr is tniyj.
  • Step 5 azaaderaiise 12
  • Step 2 die! 71
  • cmde Gl obtained from 187 nig of G2-TEA salt, 0.2 mmol, containing Py » DCA salt
  • MeCN 0.5 mL
  • A2 0.26 g, 0.26 mmol
  • MeC 0.2 mL
  • DDTT 46 mg, 0.22 mmol
  • the residue is dissolved in DCM (4.8 mL) and water (0.036 mL) and DCA (6% in DCM, 4.8 mL) are added.
  • Step 2 CC S
  • Step 1 b ranop ospi iiate 87
  • TBHP 5.5 M in decane, 0.164 mL, 0.9 mmol
  • sodium bisulfite aqueous solution 33%, 0.15 mL
  • the mixture is then concentrated and the residue is dissolved m DCM (4.8 mL) followed by addition of water (0.054 mL) and dichloroacetic acid (6% in methylene chloride, 4.8 mL).
  • Step 3 EB1 and EB2
  • Sena! dilutions of cGAMP analog compounds in phosphate buffer saline (PBS) are mixed with THP l luciferase reporter cells in a 96-weli plate at 0.2x lO b /well 3 in the presence or absence of 1 nM of Perfrmgolysm 0 (PFO), which can facilitate compound uptake by forming open channels on the plasma membrane.
  • PFO Perfrmgolysm 0
  • Table 6 Formulation of cyclic dinucleotides and analogs THPI -iSG-Liic THP-!SG-Liic THPI-iSG-Luc THP-!SG-Li!C
  • cGAMP analogues pyrophosphatase/phosphodiesterase (ENPP1) which is present in fetal bovine serum (FBS) (Li et al., 2015, Nat Chern Biol, 1 1 , 235),
  • FBS fetal bovine serum
  • EPP1 pyrophosphatase/phosphodiesterase 1
  • FBS fetal bovine serum
  • 5 uL of of synthetic cGAMP analogues 100 ⁇ stock
  • 10 uL of the reaction mixture was taken out and mixed with 10 ,uL phosphate buffered saline (PBS), then heated at 95 °C to denature proteins, which were removed by centrifugation at 13000 g for 5 minutes.
  • PBS phosphate buffered saline
  • the supernatants were delivered to THPl-ISG-luciferase cell line in the presence of PFO to measui ' e the activity of remaining cGAMP analogues, as described above.
  • Category A indicates less than 10% decrease of activity after 4-hour incubation
  • B indicates 10-75% decrease of activity after 4-hour incubation
  • C indicates more than 75% loss of activity after 4-hour incubation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Cephalosporin Compounds (AREA)

Abstract

Disclosed are cyclic-di-nucleotide cGAMP analogs, methods of synthesizing the compounds, pharmaceutical compositions comprising the compounds thereof, and use of compounds and compositions in medical therapy.

Description

CYCLIC DI-NUCLEOTIDE COMPOUNDS AND METHODS OF USE CROSS-REFERENCE O RELATED APPLICATIONS
[001] This application claims the benefit of U.S. Provisional Application No.
62/310,364, filed March 18, 2016, U.S. Provisional Application No. 62/355,382, filed June 28, 2016, and U.S. Provisional Application No. 62/396,140, filed September 17, 2016, the entire conten ts of each of which is hereby incorporated by reference herein.
TECHNICAL FIELD
[002] The present invention provides no el cyclic di -nucleotide cGAMP analogs, pharmaceutical compositions thereof their synthetic methods and their use in medical therapy. In particular, the compounds of the invention enhance the body's immune responses by acti vating STING (Stimulator of Interferon Genes) and are useful for the immunotherap of cancer, infectious diseases and immune disorders. The compounds are also useful as adjuvants for developing vaccines against cancer and infectious diseases.
BACKGROUND
[003] Cytosolic DNA induces type-I interferons and other cytokines that are important for immune defense against microbial infections and malignant cells but can also result in autoimmunity. This DNA signaling pathway requires the adaptor protein STING (Stimulator of Interferon Genes) and the transcription factor IRF3, but the mechanism of DNA sensing was unclear until recently, WO 2014099824 to The University of Texas disclosed that mammalian cytosolic extracts synthesized cyclic-GMP-AMP (cGAMP) in vitro from ATP and GTP in the presence of DNA but not RNA. DNA transfection or DN A virus infection of mammalian cells also triggered cGAMP production, cGAMP bound to STING, lead to the activati on of IRF3 an d induction of type-I interferons including interferon-β (IFN-β). Thus, cGAMP represents the first cyclic di-nucleotide in metazoa and it functions as an endogenous second messenger that triggers interferon production in response to cytosolic DNA.
[004] Through biochemical fractionation and quantitative mass spectrometry, the inventors on WO 2014099824 also identified a cGAMP synthase (cGAS), which belongs to the nucleotidyltransferase family. Overexpression of cGAS activated the transcription factor IRF3 and induced IFN in a STING- dependent manner. Knockdown of cGAS inhibited IRF3 activation and IFN induction by DNA transfeciion or DNA virus infection. cGAS bound to DN A in the cytoplasm and catalyzed cGAMP synthesis. These results indicate that cGAS is a cytosolic DNA sensor that induces interferons by producing the second messenger cGAMP. The inventors on WO 2014099824 also determined that the second messenger cGAMP they isolated and synthesized contains two phosphodiester linkages, one between the 2'-OH of GMP and 5" -phosphate of AMP, and the othe between the 3' -OH of AMP and 5 '-phosphate of GMP; this molecule is referred to as 2'3'-cGAMP.
[005] Several additional patents applications in this field have henceforth published:
[006] US20140205653 and US 20140341976 to Aduro Biotech disclose cyclic-di- nucleotide (CDN) compounds that activate and inhibit STING, respectively. In particular, the CDNs of the invention are provided in the form of a composition comprising one or more cyclic purine dinucleotides which activate or inhibit STING-dependent TBK1 activation and the resulting production of type 1 interferon.
[007] WO 2015077354 Al to The University of Chicago discloses Methods and compositions for treating cancer by intratumorally administering a stimulator of interferon genes (STING) agonist. In some embodiments, there are provided compositions and methods concerning methods for treating cancer in a subject comprising administering to the subject an effective amount of a stimulator of interferon genes (STING) agonist, wherein the STING agonist is administered intratumorally.
[008] W 2015161762 to Fudan University discloses the use of cyclic dmucleotide cGAMP for preparing antitumor drugs, wherein the tumor is gastric cancer, lung cancer, colon cancer, liver cancer, prostate cancer or pancreatic cancer, cGAMP was shown to inhibit the growth of human tumor cell lines in immun e compromised mice.
[009] WO 2015185565 to GlaxoSmithKiine discloses a class of cyclic dmucleotide compounds, or a pharmaceutically acceptable salt and tautomers thereof, compositions, combinations and medicaments containing said compounds and processes for their preparation. The invention also relates to the use of said compounds, combinations, compositions and medicaments, in the treatment of diseases and conditions m which modulation of STING is beneficial, for example inflammation, allergic and autoimmune diseases, infectious diseases, cancer and as vaccine adjuvants.
[010] WO 2014179335 to Memorial Sloan Kettering Cancer Center discloses compositions, methods, kits, and assays related to the use and/or exploitation of isomers of cGAMP as well as the structure of the enzyme cGAS. [Oil] There is still a need for the discover)' and development of new cyclic di- nucleotide cGAMP analogs for use in medical therapy. Specifically, cGAMP analogues with better potency . stability and specificity than endogenous cGAMP are still needed. cGAMP analogues with superior safety and efficacy in animal models of human diseases, including cancer and infectious diseases, have yet to be developed.
Figure imgf000004_0001
[012] Formula 1 encompasses Formula la - li.
[013] In one aspect, the present invention provides a compound of Formula la
Figure imgf000004_0002
Formula la wherein: a and b are independently 0 or 1 and a + b :::: 1 , when a is 1, b is 0 and R5 is not present; and when a is 0, b is 1 and R4 is not present;
X1 and X2 are independently 0, S or Se in a five-membered ring;
L!, starting from the carbon alpha to X1, and L2, starting from the carbon alpha to X2, are independently -C¾0-P(0)R6-0-,-CH20-P(S)R -0-, -C(Y^(Y2)0-P(0)R6-C(Y3)(Y4)-, - CH2NHSO2NH-, -CI !Ai ii (OSNH-. ~CH2NHC(S)NH-, -CH2NHC(NH)NH-, - CH2NHC(0)CH2-, -CH2NHSO2CH2- -CH?.CH2C(0)NH-, -CH2CH2SO2NH-, -CI Ι ΛΊ 1(3.4-
dioxocy clobuten- 1 ,2-diyl) H-,
Figure imgf000004_0003
c is 0, 1, or 2; d, e, and f are independently 0 or 1 ;
Y1, Y2, Y3, and Y4 are independently H or F; R6 is hydroxy!, thiol, Ci-ealk l, Ci-ealkyl selectively functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi, carbonyioxyl Ci-ealkoxy, d- &hydroxyalkoxy, amino, Ci-ealkylamino, di(Cwalk>'l)amino, or azido groups, Ci- eaikoxy, Ci-6aikoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyi, carbonyioxyl, d-ehydroxyalkoxy, amino, Ci- ealky!amino, di(C :-6alkyl)amino, or azido groups, Cs-salkenyl-O-, C3.5alkyn.yl-O-, oligo(ethylene glycol), poly(ethylene glycol), borano (-BH3"), or -NR7R8;
R? and R8 are independently hydrogen, Ci-ealkyl. Ci-ea kyl selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyi, carbonyioxyl, Ci -ealkoxy, d-Aydroxyalkoxy, amino, Ci-ealkylamino, di(Ci- 6alkyl)amiiio, or azido groups, cyclic -(Ci-salkyi)-, cyclic -(Ci-ealkyl)- seiectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi carbonyioxyl, Ci-ealkoxy, d-ehydroxyalkoxy, amino, Ci- ealkylammo, or di(Ci-6alkyl)amino groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thio!, hydroxy], carbonyl, carboxyi, carbonyioxyl Ci-ehydroxya!koxy, amino, Ci- 6alkylamino, or di(Ci-6alkyl)amino groups;
R1 and R2 are independently aromatic rings or heieroaromatic rings with the following genera! structure including its tautomeric forms:
Figure imgf000005_0001
g and h are independently 0 or 1;
W1 and W2 are independently hydrogen, halogen, hydroxyl, Ci-ealkyl Ci- ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyi, carbonyioxyl, Ci-ealkoxy, d-diydroxyalkoxy, amino,
Figure imgf000005_0002
di(Ci 6a!kyl)amino, or azido groups, Ci-6a!koxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl carboxyi, carbonyioxyl, Ci- shydroxyalkoxy, amino, Ci-6aikylamino, di(Ci-6alkyi)aniino, or azido groups, (' - salkenyl-O-, C3-5alkynyl-0-, oligo(ethyiene glycol), polyiethylene glycol), azido, or - NR7R8;
Z " Z2, Z3, Z4, Z5, and Zb are independently CH or N; if present, Z7, Z8, Z9, Zi0, and Z11 are independently CH or N, and then W! is O i or V. and
R3, R4, and R5 are independently hydrogen, halogen, hydroxy], ammo, Ci-ealkyl, Ci- 6alkyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyi, carboxyl, carbonyloxyl, Ci-eaikoxy, Ci-shydroxyalkoxy, amino, Ci-6alkylamino, di(Ci- 6alkyl)amino, or azido groups, Ci-6alkoxy, C :-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxvl, carbonyi, carboxyl, carbonyloxyl, Ci-6hydroxyalkoxy, amino, Ci-ealkylamino, di(C!-6alkyl)amino, or azido groups, C3-5alkenyl-0-, Cs-salkynyl-O-, oligo(ethyiene glycol), poly(eth lene glycol), azido, or -NR R8; or a pharmaceutically acceptable salt thereof.
[014] In one embodiment, the present invention provides a compound of f ormula lb
Figure imgf000006_0001
Formula lb wherein:
X1 and X2 are independently 0, S or Se;
Z12, Z! 3, Z14, v Z16, and / " are independently CH or N;
Ll, starting from the carbon alpha to Xs, and L2, starting from the carbon alpha to X independently -CH20-P(0)R6-0-,-CH20-P(S)R6-0-, -C(Y3)(Y2)0-P(0)R6-C(Y3)(Y4)-, CH2NHS02NH-, ··( Ι Ι ··Μ 1(*! ί)}Ν Ι ·. -CH2NHC(S)NH-, -CH2NHC(NH)NH- CH2NHC(0)CH2-, -CH2NHSO2CH2- -CH2CH2C(0)NH-, -CH2CH2SO2NH-
dioxocy clobuten- 1 ,2-diyl)NH-,
Figure imgf000007_0001
c is 0, 1, or 2; d, e, and f are independently 0 or 1 ;
Y\ Y2, Y3, and Y4 are independently H or F,
R6 is hydroxy!, thiol, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl Ci-ealkoxy, Ci- ehydroxyalkoxy, amino, Chalky laniino, di(Ci-6alkyi)amino, or azido groups, Ci- ealkoxy, Ci-salkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- 6alkylamino,
Figure imgf000007_0002
or azido groups, Cs-salkenyl-O-, Ca-salkynyl-O-, oligo(ethylene glycol), polyfethylene glycol), borano (-BH3"), or -NR7R8;
R? and Rs are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-6alkoxy, Cwhydroxyalkoxy, amino, Ci-ea!kylamino, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Ci-6alkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cusalkoxy, Ci-e.hydroxyalkoxy, amino, Ci- ealkylamino, or di(Ci -6alkyl)amino groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- 6a!ky!amino, or di(Ci-6aikyl)arnino groups; and
R3 and R4 are independently hydrogen, halogen, hvdroxyl, amino, Cj-6alkyl, C 1 -ealkyl selectively functionalized with one or more halogen, thiol, hydroxy 1, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino,
Figure imgf000007_0003
di(Ci-6a!kyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-fthydroxyalkoxy, amino, G- 6alkylamino, di(G-6aiky!)amino, or azido groups, G-salkenyl-O-, G-sa!kynyi-O-, oligo(ethylene glycol), poly(ethy!ene glycol), azido, or -NR7RS; or a pharmaceutically acceptable salt thereof.
[015] In another embodiment, the present invention provides a compound of
Formula ic
Figure imgf000008_0001
Formula k wherein:
zi 2j Z Z H Z _ and Zi? are independently CH or N;
R3 and R4 are independently hydrogen, halogen, hydroxvl, amino, Cwalk l, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonyloxvl,
Figure imgf000008_0002
Ci-ehydroxyalkoxy, amino, Ci-6aikyiamino, di(Ci-6alkyl)amino, or azido groups, Ci-ealkoxy, Ci-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxvl, carbonyl, carboxyl carbonyloxvl, Ci-bhydroxyalkoxy, ammo, Ci- ealkyiammo, di(Ci-6alk}'i)amino, or azido groups, Cs-salkenyl-O, C3-5alkynyl-0-, oligo(ethyiene glycol), poly(ethylene glycol), azido, or -NR7R8;
R7 and R8 are independently hydrogen, Ci-ealkyl, Ci-ealkyi selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, C i ^hydrox alkoxy, amino, Cwalkylamino, difCi- 6aikyl)amino, or azido groups, cyclic -(Cwaikyl)-, cyclic -(Ci-ealkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyi, Ci-eaikoxy, C ^hydroxy aikoxy, amino, Ci-ealkylamino, or di(Ci- 6alkyl)amino groups, cyclic -(Ci-eoxaalkyl)-, or cyclic -(Ci-eoxaalkyl)- selectively functionalized with one or more halogen, thiol, hvdroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino,
Figure imgf000009_0001
or di(Ci-6alkyi)amino groups; and
Rv and R10 are independently hydroxy!, thiol Ci-ealkyl, Ci-6alkyl selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonvl, carboxyl, carbonvloxyl Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci-6alkylamino, di(Ci-6alkyi)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively fundi on ali zed with one or more halogen, thiol, hydroxyl, carbonvl, carboxyl, carbonvloxyl, Ci-ehydroxyalkoxy, amino, Ci-ealkylamino. di(Ci- 6alkyl)amino, or azido groups, C3-salkenyl-0-, Cs-iaikynyi-O-, oligo(ethylene glycol), polyiethylene glycol), borano (-BH3"), or -NR7RS; or a pharmaceutically acceptable salt thereof.
[016] In another embodiment, the oxygen atom in one or both of the
tetrahydrofuranyl rings of Formula lc is replaced by a sulfur or a selenium atom.
[017] In another embodiment, the present invention provides a compound of Formula Id
Figure imgf000009_0002
Formula Id wherein:
X*! and X2 are independently 0, S or Se;
L1, starting from the carbon alpha to X1, and L% starting from the carbon alpha to X2, are independently -CH20-P(0)R6-0-,-CH20-P(S)R6-0-, -C(Yi)(Y )0-P(0)R6-C(Y3)(Y4)-, - CH2NHSO2NH-, -CH_NHC(0)NH-, -CH_NHC(S)NH-, -CH2NHC(NH)NH-5 - CH2NHC(0)CH2-, -CH2NHSO2CH2- -CH2CH2C(0)NH-, -CH2CH2SQ2NH-, -CH2NH(3,4-
dioxocy clobuten- 1 ,2-diyl)NH-,
Figure imgf000009_0003
c is 0, 1, or 2; d, e, and f are independently 0 or 1 ;
Y], Y", Y- and Y4 are independently H or F;
R'' is hydroxy], thiol, Ci-6alkyl, Ci-ealkyl selectively fundi on alized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi, carbonyloxyl, Ci-ealkoxy, Ci- bh droxyalkoxy, amino, Ci-ealkylamino, di(Ci-6alkyi)amino, or azido groups, Ci- ealkoxy, Ci-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi, carbonyloxyl, Ct-shydroxyalkoxy, amino, Ci- 6alkylamino, di(Ci-6alkyl)amino, or azido groups, C3-5alkenyl-0-, Cs-salkynyl-O-, oligo(ethy!ene glycol), polyCethyiene glycol), borano (-BH3 "), or -NR7R8;
R7 and Rs are independently hydrogen, Ci-ealkyl , Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi, carbonyloxyl, Ci-ealkoxy, Cwhydroxyalkoxy, amino, Cwalkylamino, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Ci-f.alkyl)- selective!y functionalized with one or more halogen, thiol, hydroxy!, carbonyl carboxyi, carbonyloxyl, Ci-ealkoxy, Cwhydroxyalkoxy, amino, Ci- balkylammo, or
Figure imgf000010_0001
groups, cyclic -(Cwoxaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyi, carbonyloxyl, C i-ehydroxy alkoxy , amino, Ci- 6alkylamino, or di(Ci-6a!iiyl)ammo groups;
R3 and R4 are independently hydrogen, halogen, hydroxy!, ammo, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyi, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Cwalkylamino, di(Ci-6alkyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyi. carbonyloxyl, Cwhydroxyalkoxy, amino, Ci- 6a!kylamino, di(Ci-6alkyl)amino, or azido groups, Cs-salkeny!-O, Ci-salkynyl-O-, oligo(ethyiene glycol), poly(ethylene glycol), azido, or - R7R8;
Rn and R12 are independently selected from the group consisting of:
Figure imgf000011_0001
7I2 7i3 7I4 is 7I6 717 7I8 7I 720 721 722 23 724 725 26 727 72S
Z29, Z30, Z33, Z32, Z33, Z34, Z35, Z36, and Z37 axe each independent!}' CH or N; and W3, W4, W5, W6, W7, W8, and W9 are independently hydrogen, halogen, hydroxyl, Ci-6alkyl, Cj-ealkyl selectively function alized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci-ealkylamino, di(Ci-&alkyl)arriino, or azido groups, Ci-6alkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cj-ehydroxyalkoxy, amino, Chalky lamino, di(Ci- 6alkyl)aniino, or azido groups, Cs-salkenyl-O-, Cs-salkynyl-O-, oligo(ethy1ene glycol), polyethylene glycol), azido, or -NR7R8; or a pharmaceutically acceptable salt thereof.
[018] In another embodiment, the present invention provides a compound of Formula ie
Figure imgf000012_0001
ormula ' wherein:
X3, X4, X5, and X6 are independently 0, NH, C¾ CHF, or CF;?;
R and Ri4 are independently selected from the roup consisting of:
Figure imgf000012_0002
7l2 Ύίί 14 is 7I6 717 7I8 7I 720 21 722 723 724 725 726 727 „n li 72! are each independently CH o ; and
W3, W4, W5, W6, and W7 are independently hydrogen, halogen, hydroxy!, Ci- 6alkyl, Chalky 1 selectively functionalized with one or more halogen, thiol, liydroxyl, carbonyl, carboxyl, carbonyioxyl, Ci-ealkoxy, Ci-6hydroxyalkoxy, amino, Ci- ealkylarnino, di(Ci-6alkyl)amino, or azido groups, Ci-ealkoxy, Ci-ea!koxy selectively functionalized with one or more halogen, thiol, liydroxyl, carbonyl, carboxyl, carbonyioxyl, Ci-bhydroxyalkoxy, amino, Ci-eaikylamino, di(Ci-6alkyl)amino, or azido groups, Cs-salkenyl-O-, Cs-saikynyl-O-, oiigo(ethy3ene glycol), poly(ethylene glycol), azido, or -NR7R8; 7 and R8 are independently hydrogen, Ci-ealkyl, Ci-eaikyl selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl, carbonyloxyl, d-ealkoxy, Ci-fthydroxyalkoxy, amino, d-ealkyl amino, di(Ci- 6alkyl)amino, or azido groups, cy clic -(Ci-ealkyl)-, cyclic -(Ci-ealkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl carbonyloxyl, Cj.-6alkoxy, d-ehydroxyalkoxy, amino, d- ealkylamino, or di(Ci-6alk>d)amino groups, cyclic -(Cwoxaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonyloxyl, d-ehydroxyalkoxy, amino, Ci- 6alkylamino, or di(d-6alkyl)amino groups;
R3 and R4 are independently hydrogen, halogen, hydroxyl, amino, d^alkyl, C 3 -ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, d-ehydroxyalkoxy, amino, d^alkylamino, di(d-6alkyl)amino, or azido groups, Ci-6alkoxy, d-^alkoxy selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl carbonyloxyl, d-ohydroxyaikoxy, amino, Ci- ealkyiammo, di(d-6alkyl)amino, or azido groups, d-salkenyl-O-, d-salkynyl-O-, oligo(ethylene glycol), polyiethylene glycol), azido, or -NR7R8; and
R15 and R16 are independently hydroxyl, thiol, methoxy, ethoxy, amino, N- methylamino, AyV-dimethylamino, N-ethylamino, N,N-diethylammo, N-morpholino, or borano (~Bi h }; or a pharmaceutically acceptable salt thereof.
[019] In another embodiment, the oxygen atom in one or both of the
tetrahydrofuranyl rings of Formula le is replaced by a sulfur or a selenium atom.
[020] In another embodiment, the compound is:
Figure imgf000014_0001
13
Figure imgf000015_0001
14
Figure imgf000016_0001
[026] n another embodiment, the compound is:
Figure imgf000017_0001
[028] n another embodiment, the compound is:
Figure imgf000018_0001
17
Figure imgf000019_0001
18
Figure imgf000020_0001
19
Figure imgf000021_0001
Figure imgf000021_0002
[036] In another embodiment, the compound is:
Figure imgf000022_0001
21
Figure imgf000023_0001
22
Figure imgf000024_0001
] In one embodiment, the present invention provides a compound of Formula If
Figure imgf000024_0002
Formula if wherein:
X1 and X2 are independently 0, S or Se;
Zn, IP, Zu, Z15, Z36, and Z17 are independently CH or N;
L1, starting from the carbon alpha to X1, and L2, starting from the carbon alpha to X2, are independently -CH20-P(0)R6-0- CH20-P(S)R6-0-, -C(Yi)(Y2)0-P(0)R6-C(Y3)(Y4)-, - CH2NHSO2NH-, ~i ! ί ·\ΠΠΟ)\! ί-. ~CH2NHC(S)NHk -CH2NHC(NH)NH-5 - CH2NHC(0)CH2-, -CH2NHSO2CH2- -CH2CH?.C(0)NH-, -CH2CH2SO2INH-, CH -M 1(3.4-
dioxocy clobuten- 1 ,2~diyl)NH-,
Figure imgf000025_0001
c is 0, 1 , or 2; d, e, and f are independently 0 or 1 ;
Y1, Y2, Y3, and Y4 are independently H or F;
R6 is hydroxyl, thiol. Ci-6alkyL Ci-ealk l selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl. Ci-salkoxy, Ci- ehydroxyalkoxy, amino, Ci-6alkyla.mino, di(Ci-6alkyl)amino, or azido groups, Ci- ealkoxy, Ci-ealkoxy selectivelv functionalized with one or more haloeen. thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- ealkylamino, di(Ci-6alkyl)amino, or azido groups, C3-5alk.enyl.-O-, Ci-salkyriyl-O-, oligo(ethylene glycol), poly(eihylene glycol), borano ί-ΒΠ ; }.. or -NR7R8;
R7 and R8 are independently hydrogen, Ci-ealk l, Cr-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci-ealkylamino, di(Ci- 6a3kyl)amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Cwalkyi)- seiectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Cwhydroxyalkoxy, amino, Ci- 6alkylamino, or di(Ci-6alkyl)amino groups, cyclic -(Ci-eoxaalkyi)-, or cyclic - (Ci»60xaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-6hydroxyalkoxy, amino, Ci- 6alkylamino, or di(Ci-6alkyl)amino groups; and
R3 and R5 are independently hydrogen, halogen, hydroxyl amino, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cwalkoxy, Ci-ehydroxyaikoxy, amino, Cwalkylamino, di(Cwalkyl)amino, or azido groups, Ci-ealkoxy,
Figure imgf000025_0002
selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-6hydroxyalkoxy, ammo, Ci- 6alkyiamino, di(Ci-6alkyl)amino, or azido groups, Ci-salkenyl-O-, Ci-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7RS; or a pharmaceutically acceptable salt thereof.
[042] In one embodiment, the present invention provides a compound of Formula Ig
Figure imgf000026_0001
Formula ig wherein:
Z12, Z13, Z1 , Z15, Z16, and Z" are independently CH or N;
R3 and R5 are independently hy drogen, halogen, hydroxyl, ammo, Ci-6alkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy], carbonvl, carboxyi, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci^alkyiamino. di(Cwa]kyl)ainino( or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyi, carbonyloxyl, Ci-6hydroxyalkoxy, amino, Ci- ealkylamino, di(Ci-6alkyl)amino, or azido groups, Cs-salkenyl-O-, C3-5alkynyl-0-, oligo(ethyiene glycol), poly(ethylene glycol), azido, or -NR7R8:
R7 and R8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxy], carbonvl, carboxyi, carbonyloxyl, Cwalkoxy, Ci-chydroxyaikoxy, amino, Ci-ealkylamino, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Chalky!)-, cyclic -(Ci-ealkyi)- selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyi, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci-6alkylamino, or di(Ci- 6alkyl)amino groups, cyclic -(Ci-eoxaalkyl)-, or cyclic -(Ci-eoxaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyi, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci-eaikyiamino, or di(Ci-6alkyl)amino groups; and R9 and R1C are independently hydroxy!, thiol, Ci-salkyl, Ci-eaikyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonvl, carboxyl, carbonvloxyl, Ci-6alkoxy, C i-ehydroxyalkoxy , amino, Ci-salkylamino, di(Ci.6alkyl)amino, or azido groups, Ci-ealkoxy, Ci-6aikoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonvl, carboxyl, carbonvloxyl, Ci-ehydroxyalkoxy, amino, Ci-6alkyiamino, di(Ci- 6alkyl)amino, or azido groups, C3-salkenyl-Q~, C3 -salkynyl-Q-, oligo(ethylene glycol).
poly(ethylene glycol), borano (-BHr ), or -NR7R8; or a pharmaceutically acceptable salt thereof.
[043] In another embodiment, the oxygen atom in one or both of the
tetrahydrofuranyl rings of Formula Ig is replaced by a sulfur or a selenium atom,
[044] In another embodiment, the present invention provides a compound of Formula Ih
Figure imgf000027_0001
Formula Ih wherein:
X! and X2 are independently 0, S or Se;
L!, starting from the carbon alpha to X1, and LA starting from the carbon alpha to X2, are independently -CH20-P(0)R6-0-,-CH20-P(S)R6-0-, -C(Y1)(Y2)0-P(0)R6-C(Y:i)(Y )-, - CH2NHSO2NH-, -CH2NHC(Q)NH-, ~CH2NHC(S)NH-, -CH2NHC( H)NH-, - CH2NHC(0)CH2-, -CH2NHS02CH2- -CH2CH2C(0)NH-, -CH2CH2SO2NH-, -CH2NH(3,4-
dioxocy clobuten- 1 ,2-diyl)NH-,
Figure imgf000027_0002
c is 0, 1 , or 2; d, e, and f are independently 0 or 1 ;
Y1, Y2, Y3, and Y4 are independently H or F; R6 is hydroxy!, thiol, Ci-ealk l, Ci-ealkyl selectively functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl Ci-ealkoxy, Ci- ehydroxyalkoxy, amino, Ci-ealkylarnino, di(Cwalk>'l)amino, or azido groups, Ci- ealkoxy, Cwalkoxy selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- ealkw!amino, di(C :-6alkyl)amino, or azido groups, Cs-salkenyl-O-, C3.5alkyn.yl-O-, oligo(ethylene glycol), poly(ethylene glycol), borano (-BH3"), or -NR7R8;
R? and R8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Cwhydroxyalkoxy, amino, Ci-ealkylamino, di(G- 6alkyl)amino, or azido groups, cyclic -(Ci-salkyi)-, cyclic -(Ci-ealkyl)- seiective!y functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-e.hydroxyalkoxy, amino, Ci- ea!kylammo, or di(Ci-6alkyl)amino groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionahzed with one or more halogen, thio!, hydroxy], carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxya!koxy, amino, Ci- 6alkylarnino, or di(Ci-6alkyl)amino groups;
R3 and R5 are independently hydrogen, halogen, hydroxyl, amino, Cwalkyl, C 3 -ealkyl selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cwalkoxy, Ci-ehydroxyalkoxy, amino, Ci-6aikyiamino, di(Ci-6a3kyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cwhydroxyalkoxy, amino, Ci- 6alkylamino, di(Ci-6aiky!)amino, or azido groups, C saikenyl-Q-, Cs-salkynyl-O-, oligo(ethy!ene glycol), poly(ethy!ene glycol), azido, or -NR7R8;
R11 and R12 are independently selected from the group consisting of:
Figure imgf000029_0001
, with at least one of R11 and RL being
Figure imgf000029_0002
212 213 Ζί4 Z15 Z16 Z17 Z18 Z19 Z20 Z2i Z22 Z23 Z24 Z25 Z26 Z27 Z28 Z29, Z30, Z31, 7? Z? 7? Z35, Z36, and Z37 are each independently CH or N; and ψ- W4, W5, W6, W7, W8, and W9 are independently hydrogen, halogen, hydroxy!, Cwalkyl, Ci-ealkyl selectively funciionaiized with one or more halogen, thiol hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cwalkoxy, Ci-ehydroxyalkoxy, amino, Ci-aalkylamino, di(Ci-5alkyl)amino, or azido groups, Ci-6alkoxy, Ci-ealkoxy selectively funciionaiized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci-salkylamino, di(Ci- 6alkyl)amino, or azido groups, Cs-salkenyl-O-, C - alk> m ! ·()·. oligoiethyiene glycol), poly(ethylene glycol), azido, or - R7R8; or a pharmaceutically acceptable salt thereof.
[045] In another embodiment, the present invention provides a compound of Formula Ii
Figure imgf000030_0001
Formula H wherein:
X3, X'5, X6, and X7 are independently 0, NH, CH>, CHF, or CF2;
R13 and R14 are independently selected from the group consisting of:
Figure imgf000030_0002
Z12, Zi3, Z14, Z15, Z16, Z17, 7}\ Z19, ZP, Z2 IP, Z23, Z24, Z25, Z26, Z27, and Z2S are each independently CH or N; and
W3, W4, W5, W6, and W7 are independently hydrogen, halogen, hydroxy!, Ci- ealkyl,
Figure imgf000030_0003
selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyioxyl, Ci-ealkoxy, Ci ^hydroxy alkoxy, amino, Ci- 6aikylamino, di(Ci-6aikyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, cai'bonyloxyl, Ci-ehydroxyalkoxy, amino, Ci-salkylamino, di(Ci-6alkyl)amino, or azido groups, Ci-salkenyi-O-, C3-5aIkynyl-0-, oligo( ethylene glycol), poly(ethylene glycol), azido, or ~NR7RS;
R7 and R8 are independently hydrogen, Cwalkyl, Ci-ealk l selectively functionalized with one or more halogen, thiol, hvdroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-6alkoxy, Cwhydroxyalkoxy, amino, Ca^alkylamino, di(Ci- 6alky])amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Ci-f.alkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-6alkoxy, Ci-6hydroxyalkoxy, amino, Ci- 6alkylamino, or
Figure imgf000031_0001
groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thiol, hvdroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- 6alkylamino, or di(Ci-6alkyl)amino groups;
R3 and R5 are independently hydrogen, halogen, hydroxyl, amino, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-&aikoxy, Ci-shydroxyalkoxy, amino, Ci-ealkylamino, di(Ci-6alkyl)amino, or azido groups, Ci-ealkoxy, Cwaikoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- ealkyiamino, di(Ci-6alkyi)amino, or azido groups, C ; .-Uilkcn ! ·()·. Cs-salkynyl-O-, oligo(ethyiene glycol), poly(ethylene glycol), azido, or -NR7R8; and
R15 and R16 are independently hydroxyl, thiol, methoxy, ethoxy, amino, N- methylamino, AyY-dimetlrylamino, N-ethylamino, N/V-diethylamino, N-rnorpholino, or borano (-BH3 ~); or a pharmaceutically acceptable salt thereof.
[046] In another embodiment, the oxygen atom in one or both of the
tetrahydrofuratiyl rings of Formula li is replaced by a sulfur or a selenium atom
[047] In another embodiment, the compound is:
Figure imgf000032_0001
In another embodiment, the compound
Figure imgf000032_0002
[049] n another aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.
[050] in another aspect, the present invention provides a method of treating a disease or condition in which modulation of STING is beneficial comprising: administering to a patient in need thereof a compound of Formula I or a pharrnaceutically acceptable salt thereof.
[051] In another aspect, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease or condition in which modulation of STING is beneficial. [052] In another aspect, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in therapy.
[053] In another aspect, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
[054] In another aspect, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, or a pharmaceutically composition thereof, such as a nanoparticle or a deliver}7 vehicles that enhances the cellular uptake, stability and efficacy of a compound of Formula I for use in the treatment of cancer.
[055] In another aspect, the present invention provides a method of treating cancer comprising: administering a therapeutically effective amount of a compound of Formula L or a pharmaceutically acceptable salt thereof, to a patient in need thereof.
[056] In another aspect, the present invention provides the use of a compound of Formula 1, or a pharmaceutically acceptable salt thereof, in the manufacture of a medication for the treatment of cancer.
[057] In another aspect, the present invention provides pharmaceutical composition comprising; a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent.
[058] In another aspect, the present invention provides a pharmaceutical composition comprising; a compound of Formula I, or a pharmaceutically acceptable salt thereof, at least one further therapeutic agent, and one or more of pharmaceutically acceptable excipients.
[059] In another aspect, the present invention provides a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in therapy.
[060] In another aspect, the present invention provides a pharmaceutical composition comprising; a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in the treatment of a disease or condition for which modulation of STING is beneficial.
[061] In another aspect, the present invention provides a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in the treatment of cancer,
[062] In another aspect, the present invention provides a method for treating a disease or condition for which modulation of STING is beneficial comprising: administering to a patient in need thereof a therapeutically effective amount of a combination comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof and at least one further therapeutic agent.
[063] In another aspect, the present invention provi des a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent.
[064] In another aspect, the present invention provides a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and ax least one further therapeutic agent for use in the treatment of cancer. The therapeutic agent includes but is not limited to immune checkpoint inhibitors, such as humanized antibodies against PD1, PD-LL CTLA4 and other molecules that block effective anti-tumor immune responses,
[065] In another aspect, the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent. The therapeutic agent includes but is not limited to immune checkpoint inhibitors, such as humanized antibodies against PD1, PD-L1, CTLA4 and other molecules that block effective anti-tumor immune responses.
[066] In another aspect, the present invention provides a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in the treatment of cancer. The therapeutic agent includes radiation, such as high-dose radiation, which directly kills tumor cells, enhances presentation of tumor antigens and activates the STING pathway.
[067] In another aspect, the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent. The therapeutic agent includes radiation, such as high-dose radiation, which directly kills tumor cells, enhances presentation of tumor antigens and activates the STING pathway.
[068] In another aspect, the present invention provides a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent for use in the treatment of cancer. The therapeutic agent includes another chemotherapeutic agent that selectively kills tumor cells and enhances presentation of tumor antigens,
[069] In another aspect, the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent. The therapeutic agent includes another chemotherapeutic agent that selectively kills tumor cells and enhances presentation of tumor antigens.
[070] In another aspect, the present invention provides a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, a pharmaceutical formulation including a nanoparticie, and at least one further therapeutic agent for use in the treatment of cancer. The therapeutic agent includes radiation and/or another chemotherapeutic agent,
[071] in another aspect, the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula Ϊ, or a pharmaceutically acceptable salt thereof, a pharmaceutical formulation including a nanoparticie, and at least one further therapeutic agent for use in the treatment of cancer. The therapeutic agent includes radiation and/or another chemotherapeutic agent,
[072] In another aspect, the present invention provides a method of treating cancer comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, a pharmaceutical formulation including a nanoparticie, and at least one further therapeutic agent for use in the treatment of cancer. The compoun d of Formula L may be injected directly to tumors, or system! caliy, including injection into muscles (intramuscular), skins (subcutaneous and intra-dermal), peritoneal (intraperitoneal), lymph nodes (mtralymphatic) or veins (intravenous).
[073] in another aspect, the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use as a vaccine adjuvant.
[074] In another aspect, the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, such as a nanoparticie or a deliver}7 vehicles that enhances the cellular uptake, stability and efficacy of a compound of Formula I, for use as a vaccine adjuvant.
[075] In one embodiment, the pharmaceutical composition is a vaccine. [076] In another embodiment, the present invention provides a method of inducing or promoting an immune response comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical compositio comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, as an adjuvant and a tumor antigen.
[077] In another embodiment, the present invention provides a method of inducing or promoting an immune response comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I, or a pharmaceutical composition thereof, as an adjuvant, a tumor antigen, or a pharmaceutical composition thereof, such as a nanoparticle or a delivery vehicles that enhances the cellular uptake of the adjuvant and tumor antigen,
[078] In another aspect, the present invention provides a pharmaceutical composition comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof, as an adjuvant and an immunogen for a target pathogen,
[079] in another aspect, the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use as a vaccine adjuvant.
[080] In another embodiment, the present invention provides a method of inducing or promoting an immune response comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I, or a pharmaceutically acceptable salt thereof, as an adjuvant and an immunogen for a target pathogen,
[081] In another aspect, the present invention provides a vaccine adjuvant comprising: a compound of Formula I, or a pharmaceutically acceptable salt thereof.
[082] In another aspect, the present invention provides an immunogenic composition comprising: an antigen or antigen composition and a compound of Formula I, or a pharmaceutically acceptable salt thereof.
[083] In another aspect, the present invention provides an immunogenic composition comprising: an antigen or antigen composition and a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a disease, including cancer and infectious diseases.
[084] In another aspect, the present invention provides the use of a compound of Formula Ϊ, or a pharmaceutically acceptable salt thereof, for the manufacture of an immunogenic composition comprising an antigen or antigen composition, for the treatment or prevention of a disease, including cancer and infectious diseases. [085] In another aspect, the present invention pro vides a method of treating or preventing a disease comprising: administering to a patient suffering from or susceptible to the disease, an immunogenic composition comprising an antigen or antigen composition and a compound of Formula L or a pharmaceutically acceptable salt thereof.
[086] In another aspect, the present invention provides a vaccine composition comprising: an antigen or antigen composition and a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a disease, including cancer and infectious diseases.
[087] In another aspect, the present invention provides the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof, for the manufacture of a vaccine composition comprising an antigen or antigen composition for the treatment or pre vention of a disease, including cancer and infectious diseases.
[088] In another aspect, the present invention provides a method of treating or preventing disease comprising the administration to a patient suffering from or susceptible to the disease, a vaccine composition comprising an antigen or antigen composition and a compound of Formula I, or a pharmaceutically acceptable salt thereof.
[089] In another aspect, the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment of immune disorders, including autoimmune and autoinflammatory diseases.
[090] In another aspect, the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, or a pharmaceutically composition thereof, such as a nanoparticie or a deliver}' vehicles that enhances the cellular uptake, stability and efficacy of a compound of Formula I, for use in the treatment of immune disorders, including autoimmune and autoinflammatory diseases.
[091] In another aspect, the present invention provides a method of treating immune disorders comprising: administering a therapeutically effective amount of a compound of Formula 1, or a pharmaceutically acceptable salt thereof, to a patient in need thereof.
[092] in another aspect, the present invention provides the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof, in the manufacture of a medication for the treatment of immune disorders, including autoimmune and autoin ilammaton'' diseases,
[093] It will be appreciated that all combinations of the above aspects/embodiments, and other aspects/embodiments disclosed elsewhere herein, are contemplated and are further embodiments of the invention. DETAILED DESCRIPTION
[094] The present, invention provides novel cGAMP analogs, pharmaceutical compositions thereof, and uses thereof in therapy. 2'3'-cGAMP is an endogenous second messenger produced by mammalian cells, it is a high affinity ligand for STING, inducing conformational changes therein, and a potent inducer of type-I interferons. cGAS and the cGAS-cGAMP pathway is important for triggering intlaimmatory responses to self and foreign DMA. As such, cGAS is important for immune defense against microbial pathogens that contain DNA and require DNA in their life cycles. These pathogens include DNA viruses, retroviruses including HI V, bacteria including mycobacterium tuberculosis, fungi and parasites. cGAS can also detect tumor DNA and is important for the body's intrinsic immunity against malignant cells. Activation of the cG AS-cG AMP-STING pathway is importa t for cancer immunotherapy.
[095] As a potent inducer of type-1 interferons, cGAMP (and hence the cGAMP analogs of the present invention) provides a rational immune adjuvant. As such, a compound of Formula I or a pharmaceutically acceptable salt thereof, may be used as a vaccine adjuvant, particularly with mucosal vaccines, and may be formulated with immunogens and delivered as have been cyclic-di-GMP and c-di-AMP as vaccine adjuvants (see, e.g.
Pedersen, et al PLoS ONE. Nov 201 1 , 6, 1 1. e26973: Ebensen et u! . Vaccine 29, 2011. 5210-5220; Chen et ah, Vaccine 28, 2010, 3080-3085). In fact, such adj uvants are often more effective because cGAMP (and the c AMP analogs of the present invention) is more potent than c-di-GMP in inducing interferons.
[096] In one aspect, the invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer. In one embodiment, the pharmaceutical composition is a compound of Formula 1. In another embodiment, the pharmaceutical composition is a compound of Formula I in a pharmaceutical formulation including ananoparticle or another delivery vehicle. In another embodiment, the pharmaceutical composition is a compound of Formula I in combination with at least one further therapeutic agent, which includes but is not limited to immune checkpoint inhibitors such as antibodies against PD-1, PD-L1 or CTLA-4, The therapeutic agent used in combination with a compound of Formula I also includes radiation of tumors or a chemotherapeutic agent that targets tumor cells.
[097] In another aspect, the invention provides a pharmaceutical composition comprising a compound of Formula 1 or a pharmaceutically acceptable salt thereof, as an adj uvant and an immunogen for a target pathogen, in one embodiment, the pharmaceutical composition is a vaccine. In another embodiment, the present invention provides a method of inducing or promoting an immune response comprising: administering to a patient in need thereof an effective amount a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, as an adjuvant and an immunogen for a target pathogen.
[098 S As used herein:
[099] The terms "halo" and "halogen', alone or in combination with other groups, refers to fluoro-, chloro-, bromo- and iodo~.
[0100] The term "Ci-e alkyi", alone or in combination with other groups, refers to monovalent, linear chain or branched chain alkyl groups containing from 1 to 6 carbon atoms. Exemplar' Ci-e alkyl groups include but not limited to metheyl, ethyl, w-propyi, wo-propyl, n-butyl, sec-butyl and fer/-butyl groups. More preferred are Ci4 alkyls.
[0101] The term "Cu- alkoxy"'refers to, alone or in combination with other groups, ! " ·(}·. where R" is Ci-6 alkyl.
[0102| The term "haloCi-ealkyF", alone or in combination with other groups, refers to a Ci-6 alkyl group subsituted with one or more halo suctsitutents, for example CF3 and CH2CF3.
[0103] The term "a compound of the invention" or "a compound of Formula Γ includes ail solvates, complexes, polymorphs, radiolabeled derivatives, lautomers, stereoisomers, and optical isomers of the compoimcls of Formula I and salts thereof, unless otherwise specified.
[0104] The term "effective amount" means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician.
[0105] The term "therapeutically effective amount" means any amount which, as compared to a corresponding subject who Iras not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. The term also includes within its scope amounts effective to enhance normal physiological function.
[0106] The term "prophylaxis" includes prevention and refers to a measure or procedure which is to prevent rather than cure or treat a disease. Preventing refers to a reduction in risk of acquiring or developing a. disease causing at least one clinical symptom of the disease not to developing a subject that may be exposed to a disease-causing agent or a subject predisposed to the disease in advance of disease outset
[0107] The term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and dosage forms which are. within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[0108] The term "pharmaceutically acceptable excipients" includes all diluents, earners, binders, glidants, and other components of pharmaceutical formul ti ns with which the compound of the invention is administered.
[0109] The compounds of the invention may exist in solid or liquid form. In solid form, compound of the invention may exist in a continuum of solid states ranging from fully amorphous to fully crystalline.
[ 110] The term 'amorphous' refers to a state in which the material lacks long range order at the molecular level and, depending upon the temperature, may exhibit the physical properties of a solid or a liquid. Typically, such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid. Upon heating, a change from solid to liquid properties occurs which is characterized by a change of state, typically second order ('glass transition').
[0111] The term 'crystalline' refers to a solid phase in which the material has a regular ordered internal structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterized by a phase change, typically first order ('melting point').
[0112] The compounds of the invention may have the ability to crystallize in more than one form a characteristic, which is known as polymorphism, and it is understood that such polymorphic forms ("polymorphs'') are within the scope of the invention. Polymorphism generally can occur as a response to changes in temperature or pressure or both and can also result from variations in the crystallization process. Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility and melting point
[0113] The compound of Formula I may exist in solvated and unsolvated forms. As used herein, the term "solvate" refers to a complex of variable stoichiometry formed by a solute (in. this invention, a compound of Formula 1 or a salt) and a solvent. Such solvents for the purpose of the invention may not interfere with the biological activity of the solute. The skilled artisan will appreciate that pharmaceutically acceptable solvates may be formed for crystalline compounds wherein solvent molecules are incorporated into the crystalline lattice during cry stallization. The incorporated solvent molecules may be water molecules or nonaqueous such as ethanol, isopropanol, DMSO, acetic acid, eiharsolamrae, and ethyl acetate molecules. Crystalline lattice incorporated with water molecules are typically referred to as "hydrates". Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. The present invention includes ail such solvates.
[0114] It is also noted that some compounds may form tautomers. 'Tautomers' refer to compounds that are interchangeable forms of a particuiar compound structure, and that van' in the displacement of hydrogen atoms and electrons. Thus, two structures may be in equilibrium through the movement of re electrons and an atom (usually H). For example, enols and ketones are tautomers because they are rapidly intercom- erted by treatment with either acid or base. It is understood that all tautomers and mixtures of tautomers of the compounds of the present invention are included within the scope of the compounds of the present invention.
[01 IS] The compounds of Formula I may be in tire form of a salt. Typically, the salts of the present invention are pharmaceutically acceptable salts. Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention. For a review on suitable salts, see e.g., Berge et al, J. Pharm. Sci. 1977, 66, 1-19. Suitable pharmaceutically acceptable salts can include acid addition salts. A pharmaceutically acceptable acid addition salt can be formed by reaction of a compound of Formula I with a suitable inorganic or organic acid (such as hydrobromic, hydrochloric, sulfuric, nitric, phosphor! c, p-to\ uenesulf oni c, benzenesulfonic, methanesulfonic, ethanesulfonic, naphthalenesulfonic such as 2-naphthalenesuifonic), optionally m a suitable solvent such as an organic solvent, to give the salt which is usually isolated for example by crystallization and filtration. A pharmaceutically acceptable acid addition salt of a compound of Formula 1 can be, for example, a hydrobromide. hydrochloride, sulfate, nitrate, phosphate, p- ioluenesulibnate, benzenesulfonate. methanesulfonate, ethanesulibnate, or
naphthalenesulfonate (e.g. 2-naphthalenesulforiate) salt. Other non-phannaceutically acceptable salts, e.g. trifluoroacetates, may be used, for example in the isolation of compounds of the invention, and are included within the scope of this invention.
[0116] The invention includes within its scope all possible stoichiometric and non- stoichiometric forms of the compounds of Formula I. [0117] While it is possible thai, for use in therapy, the compound of the invention may be administered as the raw chemical, it is possible to present the compound of the invention as the active ingredient in a pharmaceutical composition. Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound. Accordingly, t invention further provides pharmaceutical compositions comprising a compound of the invention and one or more pharmaceutically acceptable excipients. The excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof. In accordance with another aspect of the invention there is also provided a process for the preparation of a pharmaceutical composition including the agent, or pharmaceutically acceptable salts thereof, with one or more pharmaceutically acceptable excipients. The pharmaceutical composition can be for use in the treatment and'or prophylaxis of any of the conditions described herein.
[ 118] Generally, the compound of the invention is administered in a
pharmaceutically effective amount. The amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be trea ted, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like. Pharmaceutical compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical exeipient, vehicle or carrier. Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
[0119] Preferred unit dosage compositions are those containing a daily dose or sub- dose, or an appropriate fraction thereof, of an active ingredient. Such unit doses may therefore be administered once or more than once a day. Such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art.
[0120] Pharmaceutical compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, inhaled, intranasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route. Such compositions may be prepared by any meihod known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
[0121] Pharmaceutical compositions adapted for oral administrat on may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in- oil liquid emulsions.
[0122] For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert excipient such as ethanol, glycerol, water and the like. Powders are prepared by reducing the compound to a suitable fine size and mixing with a similarly prepared pharmaceutical excipient such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
[01.23] Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths. Excipients including glidams and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation. A disintegrating or soiubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
[01.24] Moreover, when desired or necessary, excipients including suitable binders, glidams, lubricants, sweetening agents, flavors, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, com sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymelhylcel] ulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets. A powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymeihylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/'or an absorption agent such as bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cel!ulosic or polymeric materials and forcing through a screen. As an alternative to granulating, the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules. 'The granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate sail, talc or mineral oil. The lubricated mixture is then compressed into tablets. The compounds of the present invention can also be combined with a. free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps. A clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
[0125] Oral fluids such as solution, suspensions, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound. Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersing the compound in a non-toxic vehicle. Solubiiizers and emuisifiers such as ethoxylated isosiear l alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
[0126] Where appropriate, dosage unit compositions for oral administration can be microencapsulated, The composition can also be prepared to prolong or sustam the release as for example by coating or embedding particulate material in polymers, wax or the like.
[0127] The compounds of the invention may also be administered in the form of liposome delivery sy tems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines. Pharmaceutical compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
[0128] Pharmaceutical compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils,
[0129] For treatments of the eye or other external tissues, for example mouth and skm, the compositions are preferably applied as a. topical ointment or cream When formulated in an ointment, the active ingredient may be employed with either a paraffmic or a water-mi scibie ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil -in- water cream base or a water-in-oil base. [0130] Pharmaceutical compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
[0131] Pharmaceutical compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
[0132] Pharmaceutical compositions adapted for rectal administration may be presented as suppositories or as enemas.
[0133] Dosage forms tor nasal or inhaled administration may conveniently be formulated as aerosols, solutions, suspension drops, gels or dry powders.
[0134] Compositions for intranasal administration include aqueous compositions administered to the nose by drops or by pressurised pump. Suitable compositions contain water as the diluent or carrier for this purpose. Compositions for administration to the lung or nose may contain one or more excipients, for example one or more suspending agents, one or more preservatives, one or more surfactants, one or more tonicity adjusting agents, one or more co-solvents, and may include components to control the pH of the composition, for example a buffer system. Further, the compositions may contain other excipients such as antioxidants, for example sodium metabisulphite, and taste-masking agents. Compositions may also be administered to the nose or other regions of the respiratory traci by nebulization. intranasal compositions may permit the compound(s) of Formula I or (a) pharmaceutically acceptable sait(s) thereof to be delivered to all areas of the nasal cavities (the target tissue) and further, may permit the compound(s) of Formula Ϊ or (a) pharmaceutically acceptable salt(s) thereof to remain in contact with the target tissue for longer periods of time. A suitable dosmg regime for intranasal compositions would be for the patient to inhale slowly through the nose subsequent to the nasal cavity being cleared. During inhalation, the composition would be administered to one nostril while the other is manually compressed. This procedure would then be repeated for the other nostril. Typically, one or two sprays per nostril would be administered by the above procedure one, two, or three tim.es each day, ideally once daily. Of particular interest are intranasal compositions suitable for once- daily administration.
[0135] The suspending agent(s), if included, will typically be present in an amount of from 0.1 to 5% (w/w), such as from 1.5% to 2.4% (w/w), based on the total weight of the composition. Examples of pharmaceutically acceptable suspending agents include, but are not limited to, Avicef (niiciOerystallme cellulose and carboxymethylceliulose sodium), carboxymethyicelluiose sodium, veegum, tragacantk bentonite, me!hylcelmlose, xantban gum, carbopol and polyethylene glycols. [0136] Compositions for administration to the king or nose may contain one or more excipients may be protected from microbial or fungal contamination and growth by inclusion of one or more preservatives. Examples of pharmaceutically acceptable ami -microbial agents or preservatives include, but are not limited to, quaternary ammonium compounds (for example benzalkonium chloride, benzethonium chloride, cetrimide, cetylpyridmium chloride, lauralkonujm chloride and myristyl picolinium chloride), mercurial agents (for example phenyimercuric nitrate, phenylmercuric acetate and thimerosai), alcoholic agents (for example chlorobutanol, phenviethyi alcohol and benzyl alcohol), aniibacienai esters (for example esters of para-hydroxybenzoic acid), chelating agents such as di sodium edetate (EDTA) and other anti -microbial agents such as chlorhexidine, chlorocresol, sorbic acid and its salts (such as potassium sorbate) atid polymyxin. Examples of pharmaceutically acceptable anti-fungal agents or preservatives include, but are not limited to, sodium benzoate, sorbic acid, sodium propionate, methylparaben, ethylparaben, propylparaben and butyiparaben. The preservative(s), if included, may be present in an amount of from 0.001 to 1 % (vv/w), such as from 0.015% to 0.5% (w/w) based on the total weight of the composition. Compositions (for example wherein at least one compound is in suspension) may include one or more surfactants which functions to facilitate dissolution of the medicament particles in the aqueous phase of the composition. For example, the amount of surfactant used is an amount which will not cause foaming during mixing. Examples of pharmaceutically acceptable surfactants include fatty alcohols, esters and ethers, such as polyox ethylene (20) sorbitan monooleate (Poiysorbate 80), macrogol ethers, and poloxamers. The surfactant may be present in an amount of between about 0.01 to 10% (w/w), such as from 0.01 to 0.75% (w/w), for example about 0.5% (w/w), based on the total weight of the composit n.
[0137] One or more tonicity-adjusting agent(s) may be included to achieve tonicity with body fluids e.g. fluids of the nasal cavity, resulting in reduced levels of irritancy.
Examples of pharmace tically acceptable tonicity-adjusting agents include, but are not limited to, sodium chloride, dextrose, xylitol, calcium chloride, glucose, glycerine and sorbitol. A tonicity-adjusting agent, if present, may be included in an amount of from 0.1 to 10% (vv/w), such as from 4.5 to 5.5% (w/w), for example about 5.0% (w/w), based on the total weight of the composition.
[0138] The compositions of the invention may be buffered by the addition of suitable buffering agents such as sodium citrate, citric acid, trometamol, phosphates such as disodium phosphate (for example the dodecahydrate, heptahydrate, dihydrate and anhydrous forms), or sodium phosphate and mixtures thereof [0139] A buffering agent, if present, may be included in an amount of from 0.1 to 5% (w w), for example I to 3% (w/w) based on the total weight of the compositi n.
[0140] Examples of taste-masking agents include sucraiose. sucrose, saccharin or a salt thereof, fructose, dextrose, glycerol, corn syrup, aspartame, acesulfan e- . xylitoi, sorbitol, er thritol, ammonium glycyrrhizinate, triaumatm, neotanje, mannitol, menthol, eucalyptus oil camphor, a natural flavouring agent, an artificial flavouring agent, and combinations thereof.
[0141] One or more eo-soivent(s) may be included to aid solubility of the medicament cornpound(s) and/or other excipients. Examples of pharmaceutically acceptable co-solvents include, but are not limited to, propylene glycol, dipropylene glycol, ethylene glycol glycerol, ethanol, polyethylene glycols (for example PEG300 or PEG400), and methanol. In one embodiment, the co-solvent is propylene glycol .
[0142] Co-sol vent(s), if present, may be included in an amount of from 0.05 to 30% (w/w), such as from 1 to 25% (w/w), for example from 1 to 10% (w/w) based on the total weight of the composition.
[0143] Compositions for inhaled administration include aqueous, organic or aqueous/organic mixtures, dry powder or crystalline compositions admini tered to the respiratory tract by pressurised pump or inhaler, for example, reservoir dry powder inhalers, unit-dose dry- powder inhalers, pre-metered multi-dose dry powder inhalers, nasal inhalers or pressurised aerosol inhalers, nebulisers or insufflators. Suitable compositions contain water as the diluent or carrier for this purpose and may be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like. Aqueous compositions may also be administered to the nose and other regions of the respiratory tract by nebulisation. Such compositions may be aqueous solutions or suspensions or aerosols delivered from pressurised packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant.
[0144] Compositions for admini tration topically to the nose (for example, for the treatment of rhinitis) or to the lung, include pressurised aerosol, compositions and aqueous compositions delivered to the nasal cavities by pressurised pump. Compositions which are non- pressurised and are suitable for administration topically to the nasal cavity are of particular interest Suitable compositions contain water as the diluent or carrier for this purpose. Aqueous compositions for administrati n to the lung or nose may be provided with conventional excipients such as buffering agents, tomcit -modifying agents and the like. Aqueous compositions may also be administered to the nose by nebulisation. [0145] A iluid dispenser may typically be used to deliver a fluid composition to the nasal cavities. The iluid composition may be aqueous or non-aqueous, but typically aqueous. Such a fluid dispenser may have a dispensing nozzle or dispensing orifice through which a metered dose of the fluid composition is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser. Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid composition the doses being dispensable upon sequential pump actuations. The dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid composition into the nasal cavity.
[0146] Dry powder compositions for topical delivery to the Sung by inhalation may. for example, be presented in capsules and cartridges of for example gelatine, or blisters of er example laminated aluminium foil, for use in an inhaler or insufflator. Powder blend compositions generally contain a powder mix for inhalation of the compound of Formula ί or a pharmaceutically acceptable salt thereof and a suitable powder base
(carrier/dil uent/excipient substance) such as mono-, di-, or polysaccharides (for example lactose or starch). Dry powder compositions may also include, in addition to die drug and carrier, a further excipient (for example a ternary agent such as a sugar ester for example ceilobiose octaacetate, calcium stearate, or magnesium siearate.
[0147] Pharmaceutical compositions adapted for parental administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacienosiats and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
[0148] It should be understood thai in addition to the ingredients particularly mentioned above, the compositions may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include flavoring agents.
[0149] The compounds of Formula 1 and pharmaceutically acceptable salts thereof may also be formulated with other adjuvants to modulate their activity. Such compositions may contain antibody(ies) or antibody fragment(s) or an antigenic component including but not limited to protein, DNA, live or dead bacteria and/or viruses or virus-like particles, together with one or more components with adjuvant activity including but not limited to aluminium salts, oil and water emulsions, heat shock proteins, lipid A preparations and derivatives, glycolipids, other TLR agonists such as CpG DNA or similar agents, cytokines such as GM-CSF or IL-12 or similar agents.
[0150] A therapeutically effective amount of the agent will depend upon a number of factors including, for example, the age and weight of the subject, the precise condition requiring treatment and its seventy, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian. In particular, the subject to be treated is a mammal, particularly a human.
[0151] The agent may be administered in a daily dose. This amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or six) of sub- doses per day such that the total daily dose is the same.
[0152] Suitably, the amount of the compound of the invention administered according to the present invention w ll be an amount selected from about 0.01 mg to about 1 g per day (calculated as the free or unsalted compound).
[0153] The compounds of Formula I and pharmaceutically acceptable salts thereof may be employed alone or in combination with other therapeutic agents. The compounds of Formula I and pharmaceutically acceptable salts thereof and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately, administration may occur simultaneously or sequentially, in any order, by any convenient route in separate or combined pharmaceutical compositions. The amounts of the compound(s) of Formula I or pharmaceutically acceptable salt(s) thereof and the other pharmaceutically active agent(s) and the relati ve timings of administration will be selected in order to achieve the desired combined therapeutic effect. The compound(s) of Formula 1 or pharmaceutically acceptable salt(s) thereof and further therapeutic agent(s) may be emploved in combination by administration simultaneously in a unitary pharmaceutical composition including both compounds. Alternatively, the combination may be administered separately in separate pharmaceutical compositions, each including one of the compounds in a sequential manner wherein, for example, the compound of the invention is administered first and the other second and vice versa, Such sequential administration may be close in time (e.g.
simultaneously) or remote in time. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered topically and the other compound may be administered orally. Suitably, both compounds are administered orally.
[0154] The combinations may be presented as a combination kit. By the term "combination kit" "or kit of parts" as used herein is meant the pharmaceutical composition or compositions that are used to administer the combination according to the invention. When both compounds are administered simultaneously, the combination kit ca contain both compounds in a single pharmaceutical composition, such as a tablet, or in separate pharmaceutical compositions. When the compounds are not administered simultaneously, the combination kit will contain each compound in separate pharmaceutical compositions either in a single package or in separate pharmaceutical compositions in separate packages. The combination kit can also be provided by instruction, such as dosage and administration instructions. Such dosage and administration instructions can be of the kind that are provided to a doctor, for example by a drug product label, or they can be of the kind that are provided by a doctor, such as instructions to a patient.
[0155] When the combination is administered separately in a sequential manner wherein one is administered first and the other second or vice versa, such sequential administration may be close in time or remote in time. For example, administration of the other agent several minutes to several dozen minutes after the administration of the first agent, and administration of the other agent several hours to several days after the administration of the first agent are included, wherein the lapse of time is not limited. For example, one agent may be administered once a day, and the other agent may be administered 2 or 3 times a day, or one agent may be administered once a week, and the other agent may be administered once a day and the like. It will be clear to a person skilled in the art that, where appropriate, the other therapeutic ingredients(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alky] esters, or as solvates, for example hydrates, to optimize the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic ingredient. It will be clear also that, where appropriate, the therapeutic ingredients may be used m optically pure form.
[0156] Mien combined in the same composition it will be appreciated that the two compounds must be stable and compatible with each other and the other components of the composition and may be formulated for administration. When formulated separately they may be provided in any convenient composition, conveniently, in such a manner as known for such compounds in the ail. [0157] When the compound of Formula I is used in combination with a second therapeutic agent active against the same disease, condition or disorder, the dose of each compound may differ from that when the compound is used alone. Appropri ate doses will be readily appreciated by those skilled in the art.
[0158] In one embodiment, the patient in the methods and uses of the present invention is a mammal. In another embodiment, the patient is a human. The compounds of the invention are useful in the treatment of diseases and conditions in which modulation of STING is beneficial, including cancer. As modulators of the immune response, the compounds of Formula I and pharmaceutically acceptable salts thereof may also be useful, as stand-alone, in combination or as adjuvants, in the treatment of diseases and conditions in which modulation of STING is beneficial.
[0159] In one aspect, the disease or condition to be treated is cancer. Examples of cancer diseases and conditions in which a compound of Formula 1 or pharmaceutically acceptable salt thereof, may have potentially beneficial anti-tumor effects include cancers of the lung, bone, pancreas, skin, head, neck, uterus, ovaries, stomach, colon, breast, esophagus, small intestine, bowel, endocrine system, thyroid gland, parathyroid gland, adrenal gland, urethra, prostate, penis, testes, ureter, bladder, kidney or liver; rectal cancer; cancer of the anal region; carcinomas of the fallopian tubes, endometrium, cervix, vagina, vulva, renal pelvis, renal cell; sarcoma of soft tissue; myxoma; rhabdomyoma; fibroma, lipoma; teratoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hemagioma; hepatoma; fibrosarcoma; chondrosarcoma; myeloma; chronic or acute leukemia; lymphocytic lymphomas; primary CNS lymphoma; neoplasms of the CNS; spinal axis tumours; squamous cell carcinomas; synovial sarcoma: malignant pleural mesotheliomas; brain stem glioma; pituitary adenoma; bronchial adenoma; chondromatous hanlartoma; mesothelioma; Hodgkin's Disease; or a combination of one or more of the foregoing cancers.
[0160] In a further aspect, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
[0161] in a further aspect, the present invention provides a method of treating cancer comprising administering to a patient in need thereof a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof.
[0162] In a further aspect, the present invention provides the use of a compound of Formula I or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer. [0163] A compound of the invention may be employed with other therapeutic methods of cancer treatment, e.g., in anti-neoplastic therapy, combination therapy with immune checkpoint inhibitors, other chemotherapeutic, hormonal, antibody agents as well as surgical and/or radiation treatments.
[0164] Immune checkpoint inhibitors, such as humanized antibodies against PD-1 , PD-L1 and CTLA4, have recently been shown to be highly successful in treating several types of metastatic cancer, including melanoma, non-small cell lung cancers, renal cell carcinoma and bladder cancer (Sharma and Allison, 2015, Science 348, 56). However, still only a small percentage of cancer patients benefit from the checkporat inhibitor therapies, in part because insufficient number of anti-tumor immune cells, such as CDS T cells, are generated and/or infiltrated into the tumors. Activation of the cGAS-STING pathway activates anti -tumor immunity, including the production and infiltration of tumor-specific CDS T cells. Therefore, cGAMP analogues are expected to function synergistically with immune checkpoint inhibitors and the combination therapies are likely to bring therapeutic benefits to a larger percentage of cancer patients.
[0165] In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one immune checkpoint inhibitor.
[0166] In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one immune checkpoint inhibitor for use in therapy.
[0167] In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or pharmaceutically acceptable salt thereof, and at least one immune checkpoint inhibitor for use in treating cancer.
[0168] In a further aspect, the present invention provides the use of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one immune checkpoint inhibitor in the manufacture of a medicament for the treatment of cancer.
[0169] In a further aspect, the present invention provides a method of treating cancer, comprising; administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least immune checkpoint inhibitor.
[0170] In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, at least one immune checkpoint inhibitor, and one or more of pharmaceutically acceptable carriers, diluents and excipients.
[0171] Radiation of tumors, especially high-dose radiation such as stereotatic body radiation therapy (SBRT), kills tumor cells with a high degree of precision. Dead tumor cells not only provide tumor antigens to generate tumor-specific cytotoxic T cells, but also release tumor DNA into antigen presenting cells to activate the cGAS-STING pathway (Deng et aL 2014, Immunity 41, 843). Therefore, cGAMP analogues are expected to function synergistically with radiation therapies to benefit a larger percentage of cancer patients.
[0172] In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with radiation therapy such as SBRT.
[0173] In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or pharmaceutically acceptable salt thereof, in combination with radiation therapy such as SBRT for use in treating cancer.
[0174] In a further aspect, the present invention provides the use of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with radiation therapy such as SBRT in the manufacture of a medicament for the treatment of cancer.
[0175] In a further aspect, the present invention provides a method of treating cancer, comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, in combination with radiation therapy such as SBRT.
[0176] In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and one or more of pharmaceutically acceptable carriers, diluents and excipients, in combination with radiation therapy such as SBRT for the treatment of cancer.
[0177] Anti-neoplastic agents include chemical compounds and antibodies that kill tumor ceils by inhibiting ceil cycle, signal transduction, DNA metabolism and angiogenesis and/or by promoting DNA damage, apoptosis and necrosis. These agents comprise that largest class of molecules currently used for cancer therapies. Anti-neoplastic agents selectively kill tumor cells, although many of them al so kill normal cel ls, thereby generating severe side effects. Processing of dead tumor cell associated antigens by antigen presenting cells leads to the generation of tumor-specific cytotoxic T cells. Tins process can be enhanced by cGAMP analogues. Therefore, combination of cGAMP analogues with anti-neoplastic agents are likely to generate synergistic effects that benefit a larger percentage of patients,
[0178] in a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent.
[0179] In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula 1 or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent for use in therapy.
[0180] In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent for use in treating cancer.
[0181] In a further aspect, the present invention provides the use of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent in the manufacture of a medicament for the treatment of cancer.
[0182] In a further aspect, the present invention provides a method of treating cancer, comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent.
[0183] in a further aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula 1 or a pharmaceutically acceptable salt thereof, at least one anti-neoplastic agent, and one or more of pharmaceutically acceptable carriers, diluents and excipients,
[0184] Any anti-neoplastic agent that has activity versus a susceptible tumor being treated may be utilized in the combination. Typical anti-neoplastic agents useful include anti- microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazapliosphorines, alkylsulfonates, nitrosoureas, and tnazenes; antibiotic agents such as anthracyclins, actinomycms and bleomycins; topoisoraerase Π inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and anti -folate compounds; topoisoraerase I inhibitors such as camptoihecins: hormones and hormonal analogues; signal transduction pathway inhibitors; non-receptor tyrosine angiogenesis inhibitors: immunotherapeutic agents; proapoptotic agents; and cell cycle signaling inhibitors. [G185] Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle.
Examples of anti-microtubule agents include diterpenoids and vinca alkaloids.
[0186] Diterpenoids, which are derived from natural sources, are phase specific anticancer agents that operate at the G2/M phases of the cell cycle. It is believed that the diterpenoids stabilize the β-tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include paclitaxel and its analog docetaxei.
[0187] Vinca alkaloids are phase specific an ti -neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include vinblastine, vincristine, and vmorelbine.
[0188] Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA. The platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor. Examples of platinum coordination complexes include oxaiiplatin, cisplatin and carboplatin.
[0189] Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleopliilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxy!, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death. Examples of alkylating agents include nitrogen mustards such as cyclophosphamide, melphalan. and chlorambucil; alky! sulfonates such as busu!fan;
nitrosoureas such as carmustine; and triazenes such as dacarbazine.
[0190] Antibiotic anti-neoplastics are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to ceil death.
Examples of antibiotic anti-neoplastic agents include actinomycins such as dactinomycin, anthrocyclins such as daunorubicin and doxorubicin; and bleomycins.
[0191] Topoisomerase ΪΙ inhibitors include epipodophyllotoxins.
Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DN A causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include etoposide and teniposide.
[0192] Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows. Examples of antimetabolite anti-neoplastic agents include fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine.
[0193] Camptothecins, including, caraptothecin and camptothecin derivatives are available or under development as Topoisomerase I inhibitors, Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4- methy Ipiperazmo-methylene)- 10, 11 -ethylenedioxy-20-camptothecin described below.
[0194] Hormones and hormonal analogues are useful compounds for treating cancers in which there is a relationship between the hormone(s) and growth and/or lack of growth of the cancer. Examples of hormones and hormonal analogues useful in cancer treatment include adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lymphoma and acute leukemia in children; aminoglutetihimide and other aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane useful in the treatment of adrenocortical carcinoma and hormone dependent breast carcinoma containing estrogen receptors; progestins such as megestroi acetate useful in the treatment of hormone dependent breast cancer and endometria 1 carcinoma; estrogens, and anti-estrogens such as fulvestrant, flutamide. nilutamide, bicaiutamide, cyproterone acetate and 5a- reductases such as finasteride and dutasteride, useful in the treatment of prostatic carcinoma and benign prostatic hypertrophy; anti-estrogens such as tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene, as well as selective estrogen receptor modulators (SERMS) useful in the treatment of hormone dependent breast carcinoma and other susceptible cancers; and gonadotropin- releasing hormone (GnRH) and analogues thereof which stimulate the release of leutini ing hormone (LH) and/or follicle stimulating hormone (FSH) for the treatment prostatic carcinoma, for instance, LHRH agonists and antagagonists such as goserelin acetate and luprolide.
[0195] Signal transduction pathway inhibitors are those inhibitors, which block or inhibit a chemical process which evokes a intracellular change. As used herein this change is cell proliferation or differentiation. Signal transduction inhibitors useful in the present invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3domam blockers, serine/threonine kinases, phosphotidyl inosito!-3 kinases, myoinositol signaling, and Ras oncogenes.
[0196] Several protein tyrosine kinases catalyze the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth. Such protein tyrosine kinases can be broadly classified as receptor or non-receptor kinases.
[0197] Receptor tyrosine kinases are transmembrane proteins having an extracellular ligand binding domain, a transmembrane domain, and a tyrosine kinase domain. Receptor tyrosine kinases are involved in the regulation of cell growth and are generally termed growth factor receptors. Inappropriate or uncontrolled activation of many of these kinases, i.e.
aberrant kinase growth factor receptor activity, for example by over-expression or mutation, has been shown to result in uncontrolled cell growlh. Accordingly, the aberrant activity of such kinases has been linked to malignant tissue growlh. Consequently, inhibitors of such kinases could provide cancer treatment methods. Growth factor receptors include, for example, epidermal growth factor receptor (EGFr), platelet derived growth factor receptor (PDGFr), erbB2, erbB4, ret, vascular endothelial growth factor receptor (VEGFr). tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (TIE-2), insulin growlh factor -I (TGFI) receptor, macrophage colony stimulating factor (cfms), BTK, ckit, cmet, fibroblast growth factor (FGF) receptors, Trk receptors (TrkA, TrkB, and TrkC), ephrin (eph) receptors, and the RET protooncogene. Several inhibitors of growth receptors are under development and include ligand antagonists, antibodies, tyrosine kinase inhibitors and anti-sense oligonucleotides. Growth factor receptors and agents that inhibit growth factor receptor function are described, e.g., in Kaih, John ( '. Exp. Opin. Ther. Patents (2000) 10(6):803-818; Shawver et al. DDT Vol 2, No. 2 February 1997; and Lofts, F. J. et al in "Growth factor receptors as targets", New Molecular Targets for Cancer Chemotherapy, ed. Workman, Paul and Kerr, David, CRC press 1994, London.
[0198] Tyrosine kinases which are not growth factor receptor kinases are termed nonreceptor tyrosine kinases. Non-receptor tyrosine kinases useful in the present invention, which are targets or potential targets of anti-cancer drugs, include cSrc, Lck, Fyn, Yes, Jak, cAbl, FAK (Focal adhesion kinase). Brutons tyrosine kinase, and Bcr-Abl. Such non-receptor kinases and agents which inhibit non-receptor tyrosine kinase function are described, e.g., in Sinh, S, and Corey, S.J., (1999) Journal of Hematotherapy and Stem Cell Research 8 (5): 465 - 80; and Bolen, IB,, Brugge, IS., (1997) Annual review of Immunology. 15: 371-404. [0199] SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including PI3-K p85 subunit, Src family kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP. SH2/SH3 domains as targets for anti-cancer drugs are discussed in Smithgall, T.E. (1995), Journal of Pharmacological and Toxicoiogical Methods. 34(3) 125-32,
[0200] Inhibitors of Serine/Threonine Kinases include MAP kinase cascade blockers which include blockers of Raf kinases (rafk), Mitogen or Extracellular Regulated Kinase (M EKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family member blockers including blockers of PKCs (alpha, beta, gamma, epsilon, mu, lambda, iota, zeta), I kB kinase family (I KKa, I KKb), PKB family kinases, akt kinase family members, and TGF beta receptor kinases. Such Serine/Threonine kinases and inhibitors thereof are described, e.g., in Yamamoto, T. et ah, (1999), Journal of Biochemistry, 126 (5) 799-803; Brodt, P et al. (2000), Biochemical Pharmacol ogy, 60. 1101 -1 107; Massague, J., and Weis- Garcia, F. (1996) Cancer Surveys. 27:41-64; Philip, P.A.; and Harris, A.L. (1995), Cancer Treatment and Research. 78: 3-27; Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10). 2000, 223-226; U.S. Patent No. 6,268,391 ; and Martinez-lacaci, L, ei al, Int. J. Cancer (2000), 88(1), 44-52.
[0201] Inhibitors of Phosphotidyl inositol-3 Kinase family members including blockers of PT3-kinase, ATM, DNA-PK, and Ku are also useful in the present invention. Such kinases are discussed, e.g., in Abraham, R.T. (1996), Current Opinion in Immunology. 8 (3) 412-8; Canman, C.E., Lim, D.S. (1998), Oncogene 17 (25) 3301-3308; Jackson, S.P. (1997), International Journal of Biochemistry and Cell Biology. 29 (7):935-8; and Zhong, H. et al. Cancer Res., (2000) 60(6), 1541 -1545.
[0202] Also useful in the present invention are Myo-inositol signaling inhibitors such as phospholipase C blockers and Myoinositol analogues. Such signal inhibitors are described, e.g., in Powis, G,, and Kozikowski A., (1994) New Molecular Targets for Cancer
Chemotherapy ed, , Paul Workman and David Kerr, CRC press 1994, London.
[0203] Another group of signal transduction pathway inhibitors are inhibitors of Ras Oncogene. Such inhibitors include inhibitors of farnesyltransferase, geranyl-geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy. Such inhibitors have been shown to block ras activation in cells containing wild type mutant ras, thereby acting as antiprohferation agents. Ras oncogene inhibition is discussed in Scharovsky, O.G., et al. (2000), Journal of Biomedical Science. 7(4) 292-8; Ashby, M.N. (1998), Current Opinion in Lipidoiogy. 9 (2) 99 - 102; and BioChim. Biophys. Acta, (19899) 1423(3): 19-30.
[0204] Antibody antagonists to receptor kinase ligand binding may also serve as signal transduction inhibitors. This group of signal transduction pathway inhibitors includes the use of humanized antibodies to the extracellular ligand binding domain of receptor tyrosine kinases. Examples include Imclone C225 EGFR specific antibody (see Green, M.C. et al, Monoclonal Antibody Therapy for Solid Tumors, Cancer Treat. Rev., (2000), 26(4), 269-286); Herceptin <f> erbB2 antibody (see Tyrosine Kinase Signalling in Breast cancer: erbB Family Receptor Tyrosine Kinases, Breast cancer Res., 2000, 2(3), 176- 183); and 2CB VEGFR2 specific antibody (see Brekken, R. A. et al, Selective inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks tumor growth in mice, Cancer Res. (2000) 60, 5117-5124).
[0205] Anti-angiogenic agents such as non-receptor MEK. angiogenesis inhibitors may also be useful, as well as those which inhibit the effects of vascular endothelial growth factor (e.g., the anti-vascular endothelial cell growth factor antibody bevacizumab
[Avasiin™]), and compounds that work by other mechanisms (e.g., linomide, inhibitors of integrin ανβ3 function, endostatin and angiostatin).
[0206] In one aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti -neoplastic agent which is an anti-mi crotubule agent, platinum coordination complex, alkylating agent, antibiotic agent, topoisomerase 11 inhibitor, antimetabolite, topoisomerase I inhibitor, hormones and hormonal analogue, signal transduction path way- inhibitor, non-receptor tyrosine MEK angiogenesis inhibitor, immunotherapeutic agent, proapoptotic agent, or cell cycle signaling inhibitor.
[0207] In another aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent selected from diterpenoids and vinca alkaloids,
[0208] in another aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti -neoplastic agent which is a platinum coordination complex. In one embodiment, at least one anti-neoplastic agent is paclitaxel, carbop!atin, or vinorelbine.
[0209] In another aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent which is a signal transduction pathway inhibitor, in one embodiment, the signal transduction pathway inhibitor is an inhibitor of a growth factor receptor kinase VEGFR2, TIE2, PDGFR, BTK, erbB2, EGFr, IGFR-i , TrkA, TrkB, TrkC, or c-fms. In another embodiment, the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase rafk, akt, or P C-zeta. In another embodiment, the signal transduction pathway inhibitor is an inhibitor of a non- receptor tyrosine kinase selected from the src family of kinases. In another embodiment, the signal transduction pathway inhibitor is an inhibitor of c-src. In another embodiment, the signal transduction pathway inhibitor is an inhibitor of Ras oncogene selected from inhibitors of farnesyl transferase and geranylgeranyl transferase. In another embodiment, the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase selected from the group consisting of PI3K. In another embodiment, the signal transduction pathway inhibitor is a dual EGFr/erhB2 inhibitor, for example N-{3-Chloro-4-[(3-fluorobenzy]) oxy]phenyl}-6-[5- ({[2- (methanesulphonyl) ethyl]amino}methyl)-2-fur\'l]-4-quinazolinamine.
[0210] in another aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and at least one anti -neoplastic agent which is a cell cycle signaling inhibitor. In one embodiment, the cell cycle signaling inhibitor is an inhibitor of CDK2. CDK4 or CDK6.
[0211] Compounds of Formula I may be prepared by methods known in the art of organic synthesis as set forth in the schemes below and/or the specific Examples described below. In all of the methods, it is well understood that protecting groups for sensitive or reactive groups may be employed where necessary in accordance with general principles of chemistry . Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Green and P. G. M. Wuts (1.999) Protective Groups in Organic Synthesis, 3~ edition, John Wiley & Sons). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection of processes as well as the reaction conditions and order of their execution shall be consistent with the preparation of compounds of Formula 1.
[0212] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in fight thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims.
[0213] The following list provides definitions of certain abbreviations as used herein. It will be appreciated that the list is not exhaustive, but the meaning of those abbreviations not herein below defined will be readily apparent to those skilled in the art: Ac is acetyl; AcOI-I is acetic acid; Ac?.0 is acetic anhydride; AIBN is 2,2'-az.obisisobulyronilriIe; Bn is benzyl; BSA is N,0-bis(trimethylsilyl)acetamide; BSTFA is N,0- bis(trimethylsilyl)trifluoroacetamide; Bu is butyl; Bz is benzoyl; CAN is eerie ammonium nitrate; CE is 2-cyanoethyl; DCA is dichloroacetic acid, DCM is diehloromethane; DDTT is 1 ,2,4-dithiazole-5-thi one; DEAD is diethyl azodicarboxylate; DIAD is diisopropyl azodicarboxylate; D1PEA is Ayv'~diisopropy!eihy!amine; DMAP is 4- idimethylaminojpyridine; DMT is AyY-dimethylformamide; DMOCP is 2-chIoro-5,5 dimetiiyl-l ,3,2-dioxaphosphorinane 2-oxide; DM SO is dimethylsulphoxide; DMTr is 4,4'- dirnethoxytTilyl; EtOAc is ethyl acetate; EtOH is ethanol; HMPT is hexaraethvlphosphofoas triamide; HPLC is bigli performance liquid chromatography: /bu is isobutyryi; I.BX is 2- iodoxy benzoic acid; I mid is imidazole; 'Pr is isopropyi; KOB is potassium hydroxide; Me is methyl; MeCN is acetomtrile; MeOH is methanol; MTBE is methyl ter(-butyl ether; Ms is methanesulfonyl; Pd/C is palladium on activated charcoal; NTS is N-iodosuccinimide; NPE is 2-(4-nitrophenyl)ethyl; PE is petroleum ether; Ph is phenyl; PMB is />methoxybenzyl: PPlr, is triphenylphosphine; Py is pyridine: TBAF is tetra^-butylammonium fluoride; TBAI is tetrabutylammonium iodide; TBDPS is feri-butyldiphenylsilyl; TBHP is lerl-Butyl hydroperoxide; TBS is /err-butyldimethylsily ; TCDi is i , i '-thiocarbonyldiimidazole, TDA-1 is tris[2-(2-methoxyethoxy)e !]amitte; TEA is triethylaraine; Tf is
trifluoromethanesulfonyl; TFA is tfifiuofoacetic acid; TFE is 2,2,2-trifluoroethyl ; THE is tetrahydrofuran; TIPS is triisopropylsilyl; TEC is thin-layer chromatography; TMS is trimethylsiiyl: TMSOTf is trimethylsilyi trifluoromethanesulfonate: Tol is -toluoyl; Tr is tniyj.
INTERMEDIATE PREPARATIONS
Figure imgf000062_0001
Step 1 : acetonide 2
Figure imgf000063_0001
[0214] To a suspension of D-Ribose (1) (160 g, 1.07 mol) in acetone (2.0 L) is added concentrated sulfuric aad (10.7 g, 107 mmol, 5.8 niL) at 27 °C dropwise. After stirring for 12 hours, solid Sodium bicarbonate (100 g) is added. The mixture is then filtered and the filtrate is concentrated to give crude 2 (215.0 g).
Step 2: sil l ether 3
Figure imgf000063_0002
[0215] To a solution of crude 2 (215 g, 1.13 mol) in DCM (1.5 L) is added TBSC1 (170 g, 1.13 mol) and TEA (172 g, 1.69 mol) at 0 °C. After stirring at 27 °C for 12 hours, the mixture is filtered, concenirated, and purified by silica gel column chromatography (EA/PE = 1/100 to 1/50) to give 3 as a colorless oil (285 g, 83% yield).
Step 3: deazapurisie 4
Figure imgf000063_0003
[0216] To a solution of 3 (60.0 g, 197.08 mmol) and carbon tetrachloride (67.3 g, 438 mmol, 42 mL) in THF (1.2 L) is added HMPT (63.0 g. 386 mmol, 70 mL) dropwise at - 78 °C and stirred at 27 °C for 2 hours. To another solution of 6-chloro-7-deazapurine (24.2 g, 158 mmol) and OH (16.6 g, 296 mmol) in MeCN (1.2 L) is added TDA-1 (6.37 g, 19.7 mmol) at 27 °C followed by the THF solution obtained above. After stirring at 27 °C for 12 hours, the reaction mixture is filtered, concentrated, and purified by silica gel column chromatography (EA/PE = 1/50 to 1/10) to give 4 as a yellow oil (15.3 g, 18% yield).
Step 4: adenine 5
Figure imgf000064_0001
[0217] A solution of 4 (28.6 g, 64.9 mmol) in dioxane (150 mL) and ammonium hydroxide aqueous solution (500 ml.) is stirred at 120 °C for 30 hours in a sealed autoclave. The volatiles are then removed and the aqueous solution is extracted with EA (300 mL χ 3). The combined organic layers are washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and partially purified by Silica gel column chromatography (EA DCM = 1/1) to give a yellow foam (9.65 g). This residue is then dissolved in THF (50 mL) and treated with TBAF trihydate (10.9 g, 34.4 mmol) at 27 °C. After stirring for 2 hours, the mixture is concentrated and purified by silica gel column chromatography (EA/DCM = 1/1 to 7/1) to give 5 (5.56 g, 79% yield) as yellow solid.
Step 5: benzoate 6
Figure imgf000065_0001
[02181 To a solution of 5 (7.26 g, 23.7 mmol) in DCM (60 mL) is added Imid (4.84 g, 71.1 mmol) and TBSC1 (5.36 g, 35.6 mmol) at 27 °C. After stirring at 27 °C for 1.5 hours, water (100 mL) is added and the mixture is extracted with DCM (200 mL). The organic layer is washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered, concentrated to give crude TBS-5. To a solution of the crude TBS-5 obtained above in DCM (100 mL) is added benzoyl chloride (5.14 g, 36.6 mmol) at 27 °C. After stirring for 12 hours, water (200 mL) is added the mixture is extracted with DCM (500 mL). The organic layer is dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/PE = 1/50 to 1/10) to give 6 as a yellow foam (8.12 g, 64% yield).
Step 6: Bl
Figure imgf000065_0002
[0219] A solution of 6 (15,2 g, 28.9 mmol) in TFA (90 mL) and DCM (20 mL) is stirred at 27 °C for 12 hours. The volatiles are then removed and the residue is purified by silica gel column chromatography (MeOH/DCM = 1/100 to 1/10) to Bl as a yello solid (10.16 g, 95% yield),
Figure imgf000066_0001
65 Step 1 : an !ffle 8
Figure imgf000067_0001
[0220] To a solution of 5-arnino-4,6-dichloropyrimidine (63.0 g, 384 mmol) in n- BuOH (300.0 mL) is added -methoxybenz lamine (58.0 g, 423 mmol, 55 mL) and DIPEA (99.3 g, 768 mmol, 134 mL). After stirring at 100-110 °C for 15 hours, the volatiles are removed before MTBE (100 mL) is added. The solid is collected by filtration and washed with EA to give 8 as an off-white solid (55.0 g, 54% yield). (MS: | \ I 1 1 j 265.0)
Step 2: azapurine 9
Figure imgf000067_0002
[0221] To a solution of 8 (10.0 g, 37.8 mmol) in a mixture of DCM (200 mL), AcOH (100 mL), and water ( 100 mL) is added sodium nitrite (2.87 g, 41.6 mmol, 2.3 mL) at 0 °C, After stirnng at 0-25 °C for 1 hour, DCM (30 mL) and saturated sodium bicarbonate aqueous solution (30 mL) are added. The layers are then separated and the aqueous phase is extracted with DCM (150 mL x 3). The combined organic phases are dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA-'PE = 1/3) to give 9 as a light yellow solid (6.0 g, 88% yield). (MS: [M+H]+ 276,0)
Step 3: azaadenine 10
Figure imgf000067_0003
[0222] To a solution of 9 (6.0 g, 21,8 mmol) in 1,4-dioxane (30 mL) is added ammonium hydroxide aqueous solution (30 mL). After stirnng at 30-40 °C for 5 hours, the solid is collected by filtration to give 10 as a white solid (4.0 g, 70% yield). (MS: {M+fi 257.1)
Step 4; azaadenine 11
Figure imgf000068_0001
[0223] To a solution of 10 (17.0 g, 66,3 niraol) in Py (100 mL) is added DMAP (8.92 g, 73.0 nimol), Imid (13.6 g, 199 mmol) and benzoyl chloride (14.0 g, 99.5 mmol, 11.6 mL). After stirring at 110-120 °C for 18 hours, the volatiles are removed and DCM (300 mL) and water (300 mL) are added. The layers are separated and the organic phase is dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/PE = 1 : 1) to give 11 as an off-white solid (17.0 g, 68% yield). (MS: [M+H]+ 361.2)
Step 5: azaaderaiise 12
Figure imgf000068_0002
[0224] To a suspension of 11 (6.40 g, 17.8 nimol) in MeCN (60 mL) is added a solution of CAN (29.2 g, 53.3 mmol) and sodium bicarbonate (1.49 g, 17.76 mmol) in water (60 mL) at 0 °C. After stirring at 0-25 °C for 12 hours, the mixture is neuiralized with sodium bicarbonate to ~pH 7. The solid is collected by filtration to give 12 (2.6 g, 57% yield). (MS: [M+H]+ 241.1)
Step 6; azaadenosine 14
Figure imgf000069_0001
[0225] To a solution of 12 (9.30 g, 38.7 mmol) and 13 (20.5 g, 40.7 mmol) in MeCN (350 mL) is added tin(iV) chlonde (30.3 g, 1 16 mmol, 13.6 mL) at 0 °C, After stimng at 0- 25 °C for 24 hours, the reaction mixture is poured into saturated sodium bicarbonate aqueous solution (300 mL). The solid is filtered off and washed with water (100 mL). The filtrate is extracted with DCM (150 mL x 4) and the combined organic layers are dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA-'DCM = 1/10) to give 14 as an off-white gum (6.10 g, 21% yield). (MS: [M+H]+ 684.9)
Step It B2
Figure imgf000069_0002
[0226] To a solution of 14 (6.1 g, 8.9 mmol) in a mixture of THF (35 mL) and MeOH (28 mL) is added lithium hydroxide aqueous solution (1M, 16.0 mL) at 0 °C After stirring at 0-25 °C for 3 hours, the mixture is neutralized with citric acid aqueous solution (1M) to ~pH 7 and then concentrated and purified by silica gel column chromatography (MeOH/DCM = 1/20) to give B2 as an off-white solid (2.9 g, 87% ield). (MS: | V1 H | 373.1)
Figure imgf000070_0001
69 Step 1 : dio! 16
Figure imgf000071_0001
[0227] To a solution of IS (880 mg, 2.5 mmoi) in Py (10 mL) is added a solution of DMTrCl (940 mg, 2.6 mmoi) in Py (5 mL). After stirring for 3 hours, the mixture is concentrated and purified by silica gel column chromatography (MeOH/DCM = 1/20 to 1/10) to provide 16 as a white foam (1.23 g, 75% yield), (MS: [M+Hf 656.2)
Step 2; alcohol 17
Figure imgf000071_0002
[0228] To a solution of 16 (900 mg, 1.4 mmoi) and Imid (280 mg, 4.15 mmoi) in Py (15 mL) is added TBSCl (310 mg, 2.05 mmoi). After stirring for 4 hours, the volatiles are removed and the residue is dissolved in DCM (50 mL), washed with saturated sodium bicarbonate aqueous solution and brine, dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column chromatography (EA/toluene :::: 1/3 to 2/3) to provide 17 as a white solid (480 mg, 45% yield). (MS: [M+H]+ 770.2)
Step 3: thiocarbamate 18
Figure imgf000072_0001
[0229] To a solution of 17 (500 mg, 0,65 mmol) in DMF (6 mL) is added TCDI (350 mg, 1.94 mmol). After stirring for 2 days, EA (40 mL) and water (25 mL) are added and the layers are separated. The aqueous layer is extracted with ethyl acetate (25 mL χ 3). The combined organic layers are washed with water (20 mL), brine (20 mL * 2), dried over anhydrous sodium, sulfate, and concentrated to give crude 18. (MS: [M+H]+ 880.2)
Step 4; silyl ether 19
Figure imgf000072_0002
[0230] To a degassed solution of crude 18 in toluene (10 mL) at 1 10 °C is added a degassed solution of AIBN (57 mg, 0.34 mmol), tributyltin hydnde (0.51 mL, 1.94 mmol) in toluene (3 mL) over 30 minutes. After stirring at 110 °C for 6 hours, the mixture is cooled to room temperature, concentrated, and purified by silica gel column chromatography (EA hexanes = 1/5 to 2/1) to give 19 as a yellow oil (195 mg, 40% yield over two steps). (MS: i I f ! I 754.2)
Step 5: DMTr-B3
Figure imgf000073_0001
[02311 To a solution of 19 (190 rag, 0.252 mmol) in THF (5 mL) is added TBAF (1 M in THF, 0.50 mL). After stirring at room temperature for 2 hours, water (5 mL) is added and the mixture is extracted with EA (8 mL χ 3), dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column chromatography (MeOH/DCM = 1/20) to give DMTr-B3 as a white solid (132 mg, 82% yield). (MS: j \M lj 640.2)
Figure imgf000074_0001
73 Step 1 : alcohol 21
Figure imgf000075_0001
[0232| To a solution of 20 (12.8 g, 67.0 mmol) in Py (300 mL) is added TBDPSCi (21.0 mL, 80.4 mmol). After stirring tor 3 h, MeOH (25 mL) is added and the mixture is concentrated. The residue is dissolved in diethyl ether (200 mL), washes with sodium bicarbonate aqueous solution (10%, 100 mL) and water (100 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (diethyl ether/PE = 1/2) to give 21 as a white solid (27.2 g, 95% yield). (MS: |M+Na]+ 451.2)
Step 2: alcohol 22
Figure imgf000075_0002
[0233] A solution of 21 (27.2 g, 63.7 mmol) in DMSO (200 mL.) and Ac?.*) (50 mL) is stirred for 16 hour before pouring into ice water (200 mL). The mixture is extracted with diethyl ether (100 mL χ 3) and the combined organic layers are washed with sodium bicarbonate aqueous solution (10%, 100 mL) and water (100 mL), and concentrated. The residue is then dissolved in MeOH (250 mL) and DCM (250 mL) at 0 °C followed by addition of sodium borohydride (12.0 g) in 10 portions. After stirring for 5 minutes, water (100 mL) is added and the layers are separated. The organic layer is then concentrated and purified by silica gel column chromatography (diethyl ether/PE = 1/2) to give 22 as a white solid (20.4 g, 75% yield over two steps), (MS: [M+Na]+ 451.2)
Step 3; methyl ether 23
Figure imgf000076_0001
[0234] To a solution of 22 (4.0 g, 9.33 mmol) in DMF (45 mL) is added sodium hydride (484 mg, 12.1 mmol) at 0 °C and stirred for 30 minutes before methyl iodide (0.64 mL, 10.3 mmol) is added slowly. After stirring for 3 hours, water (3 mL) is added and the volatiles are removed and purified by silica gel column chromatography (EA/PE :::: 1/10) to give 23 as a white solid (3.8 g, 92% yield), (MS: [M+Na]+ 465.2)
Step 4: benzoate 24
Figure imgf000076_0002
[0235] To a solution of 23 (3.1 g, 7.0 mmol) in THF (50 mL) is added TBAF (8.4 mL, 8.4 mmol) at 0°C. After stirring for 4 h at room temperature, water (5 mL) and EA are added. The layers are separated and the organic layer is washed with water and brine, concentrated, and the resulting residue is dissolved in DCM followed by addition of TEA (4.9 mL, 35 mmol) and benzoyl chloride (0.98 mL, 8.4 mmol). After stirring for 1 hour, water (3 mL) is added and the volatiles are removed. The residue is purified by silica gel column chromatography (EA/PE = 1/5) to give 24 as a white solid (1.9 g, 88% yield). (MS: [M+ Na]+ 331.0)
Figure imgf000076_0003
[0236] A solution of 24 (0.71 g, 2.3 mmol) in HO Ac (14 mL) and water (6 mL) is heated under reflux for 30 minutes. After cooling to room temperature, the mixture is co- evaporated with toluene (10 mL x 4) and the resulting residue is dissolved in Pv/Ac20 (10/1 v/v, 10 mL) followed by addition of DMAP (50 rag, 0.46 mmol). After stirnng for 4 hours, the mixture is concentrated and purified by silica gel column chromatography (EA/PE := 1 /3) to give 25 as a white solid (0.75 g, 92% yield), (MS: [M+Na]+ 375.0)
Step 6: guanosine 27
Figure imgf000077_0001
[0237] To a suspension of 25 (500 mg, 1.42 mmol) and A^-isobutyrylguanme (500 mg, 2.13 mmol) in DCM (20 mL) at 80 °C is added BSA (1.8 mL, 7.4 mmol) and stirred for 1 hour before addition of TMSOTf (0.77 mL, 4.26 mmol). After stirring at 80 °C for 3 hours, the mixture is cooled to room temperature before sodium bicarbonate aqueous solution (50 mL) is added. The mixture is then extracted with DCM (50 mL χ 3). The combined organic layers are dried over anhy drous sodium sulfate, concentrated, and purified by silica gel column chromatography (MeOH/DCM = 1/20 to 1/10) to give 26 as a white powder (624 mg, 85% yield). (MS: | M ! l | 514.2)
Step 7: B4
Figure imgf000077_0002
[0238] To a solution of 27 (0.49 g, 0,96 mmol) in MeOH/THF/waier (4/5/1 v/v/v, 20 mL) is added sodium hydroxide aqueous solution (10 M, 0.25 mL, 2.5 mmol) at 0 °C. After stirring for 30 minutes, HO Ac is added and the mixture is concentrated and purified by silica gel column chromatography (MeOH/DCM ===: 1/10 to 1/5) to give B4 as an oil (322 mg, 92% yield). (MS: [M+Hf 368,2)
Figure imgf000078_0001
77 Step 1 : acetate 28
Figure imgf000079_0001
[0239] To a solution of 15 (7.0 g, 20 mmol) in MeCN (100 mL) is added DMAP (1.2 g, 10 mmol) and AC2O (7.5 mL, 80 mmol) at 0 °C. After stirring at room temperature overnight, the mixture is concentrated and purified by silica gel flash chromatography (MeOH/DCM = 1/20 to 1/10) to give 27 as a white solid (8.77 g, 92% yield), (MS: [M+H]+ 480.0)
Step2: propargyl ether 29
Figure imgf000079_0002
[0240] To a solution of 27 (480 mg, 1.0 mmol) in 1,4-dioxane (3 mL) is added PPh?. (656 mg, 2.5 mmol), propargyl bromide (0.15 mL, 2 mmol) and a solution of DEAD (0.49 mL, 2.5 mmol) in dioxane (1 mL) at 0 °C. After stirring for 2 hours, the mixture is concentrated and purified by silica gel column chromatography (MeOH/DCM :::: 1/50 to 1/20) to give 28 as a white solid (440 mg, 47% yield). (MS: [M+H]+ 518.2)
Siep3: BS
Figure imgf000080_0001
[0241] To a solulion of 28 (150 rag, 0.37 mmol) in THF (4,5 mL) and MeOH (0.5 mL) is added sodium hydroxide aqueous solution (1 M, 0,5 mL) at 0 °C. After stirring for 1 hour, HO Ac (0, 1 mL) is added and the mixture is concentrated and purified by silica gel column chromatography (MeOH/DCM = 1/20 to 1/10) to give BS as a white solid (40 mg, 64% yield), (MS: [M+Hf 392.0)
Figure imgf000081_0001
80 Step 1 : deazapurisie 29
Figure imgf000082_0001
[0242] To a solution of 3 (40,0 g, 131 mmol) and carbon tetrachloride (33.6 g, 218 mmol, 21 mL) in THF (500 mL) at -78 °C is added HMPT (22,5 g, 138 mmol, 25 mL) over 15 min. After stirring for 2 hours with brief periods of slight warming to prevent gel formation, the mixture is concentrated to about 70 mL.. To a suspension of KOH (25.8 g, 460 mmol) in MeCN (600 mL) is added TDA-1 (4.25 g, 13.14 mmol, 4.2 mL). After stirrmg at 25 °C for 10 minutes, 2-amino-6-cliloro-7-deazapurine (22.2 g, 131 mmol) is added. The mixture is stirred for another 10 minutes before the THF solution obtained above is added. After stirring for 2 hours, the mixture is filtered, concentrated, and purified by silica gel column chromatography (EA/PE = 3/17) to give 29 (9,20 g, 15% yield). (MS: [M+H]+ 455.3)
Step 2: alcohol 30
Figure imgf000082_0002
[0243] To a mixture of 29 (13.7 g, 30.1 mmol) in dioxane (10 mL) is added a solution of sodium hydroxide (11.7 g, 291 mmol) in water (100 mL) at 25 °C. After stirring at 80 °C for 64 hours, the mixture is cooled to 0 °C, neutralized with AcOH to ~pH 7, and extracted with EtOAc (100 mL χ 3). The combined organic layers are dried over anhydrous sodium sulfate, filtered, and concentrated to give crude 30. (MS: [M+l]+ 323, 1) Step 3: silyl ether 31
Figure imgf000083_0001
[0244] To a solution of crude 30 (9.7 g, 30.1 mmol) and Imid (4.1 g, 60.3 mmol) in DCM (10 mL) is added TBSC1 (9.08 g, 60.3 mmol) at 25 °C. After stirring for 16 hours, the mixture is diluted with DCM (100 mL), washed with brine (80 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA-'PE = 1/4 So l I then MeOH/DCM = 1/50) to give 3i (9.0 g, 68% yield) as a solid. (MS: } I 1 11 437.2)
Step 4: isobut rate 32
Figure imgf000083_0002
[0245] To a solution of 31 (9.0 g, 20.6 mmol) and TEA (4.2 g, 41.2 mmol) in DCM (80 mL) is added isobutyryl chloride (3.29 g, 30.9 mmol) at 0 °C. After stirring at 25 °C for 16 hours, the mixture is diluted with DCM (100 mL), washed with saturated sodium bicarbonate aqueous solution (50 mL x 2) and brine (50 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA PE = 1 /5 to 1 /2) to give 32 as a white solid (4.2 g, 40% yield), (MS: | .v! ! i j 507.2)
Step 5: B6
Figure imgf000084_0001
[0246] A solution of 32 (4,2 g, 8.29 mmol) in DCM (6 mL) and TFA (24 mL) is stirred at 25 °C for 1 hour before concentrated. The residue is then treated with hydrogen chloride (4M in MeOH, 10 mL) at 0 °C. After stirring at 25 °C for 10 minutes, the mixture is concentrated to give crude B6 as a white solid (2.92 g, 99% yield), (MS: [M+H]+ 353.0)
Figure imgf000085_0001
84 Step 1 : proparyl efter 34
Figure imgf000086_0001
[0247] To a solution of adenosine (33) (5.0 g, 18,7 mraol) in DMF (200 mL) at 0 °C is added sodium hydride (60% dispersion in mineral oil, 1.0 g, 25 mmol) followed by the TBAI (1.5 g, 4.06 mmol) and propargyl bromide (2.12 mL, 20.9 mmol). After stirring at 55 °C for 2 days, the mixture is purified by silica gel column chromatography (MeOH/DCM = 7/93) followed by re-crystallization from ethanol to give 34 as a pale yellow solid (2,56 g, 45%).
Figure imgf000086_0002
[0248] To a solution of 34 (1 ,4 g, 4.59 mmol co-evaporated twice with Py) in Py (20 mL) is added TMSC1 (2.4 mL, 18.9 mmol). After stirring for 30 minutes, benzoyl chloride (0.7 mL, 6.0 mmol) is added and the mixture is stirred for 3 hours before addition of water (10 mL) and ammonium hydroxide aqueous solutioin (15 mL) at 0 °C. After stirring for 20 minutes at room temperature, the mixture is extracted with DCM (25 mL χ 3). The combined organic layers are dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column chromatography (MeOH/DCM = 5:95) to give BA1 as a white foam (1.73 g, 92%), (MS: [M+H]+ 410.2)
Figure imgf000087_0001
86 Step 1: pyrazolopyriimdine 36
Figure imgf000088_0001
36
[0249] To a solution of 35 (10.0 g, 56,5 mmol) in THF (80 mL) is added DIPEA (7,3 g, .56.5 mmol, 9.9 mL). After stirring at 0 °C for 10 minutes, a solution of hydrazine (1.81 g, 56.5 mmol, 2.0 mL) in THF (20 mL) is added. The mixture is then stirred at 20 °C for 2 hours before concentrated. After addition of DCM (100 mL) and H?.0 (300 mL) to the residue, the layers are separated and the aqueous layer is extracted with DCM (100 mL χ 3). The combined organic layers are washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/'PE :=: 1/15 to 1/8) to give 36 as a yellow solid (3.10 g, 35% yield). (MS: [M+H]+ 155.1 )
Step 2; pyrazolopyrimidine 37
Figure imgf000088_0002
37
[0250] To a solution of 36 (200 mg, 1.29 mmol) in THF (2.0 mL) is added ammonium hydroxide (2,0 mL). After stirring at 20-30 °C for 2 hours, the mixture is concentrated, triturated with MeCN (0.5 mL), and collected by filteration to give 37 as a red solid (100 mg, 57% yield).
Step 3:trsbesizoate 38
Figure imgf000089_0001
[0251] To a suspension of 37 (20.0 g, 148 mmol) and 13 (101 g, 200 mmol) in MeCN (1.2 L) is added boron trifluoride diethyl etherate (30.5 g, 215 mmol, 26.5 mL). After stirring at 75-85 °C for 2 hours, the mixture is concentrated and purified by silica gel column chromatography (EA/PE = 1 /5 to 2/1) to give 38 as a yellow solid (35.0 g, 40% yield). (MS: [M+H]+ 580.3)
Step 4; benzamide 39
Figure imgf000089_0002
[0252] To a solution of 38 (10.0 g. 17.3 mmol) in DCM (100 mL) is added DMAP (421 mg, 3.45 mmol) and TEA (5.24 g, 51.8 mmol, 7.2 mL) followed by benzoyl chloride (2.91 g, 20.7 mmol, 2.4 mL) dropwise. After stirring at 20-25 °C for 8 hours, the mixture is concentrated and purified by silica gel column chromatography (EA/PE :::: 1/5 to 11/') to give 39 as a white solid (9.0 u. 76% yield). (MS: [M+H]+ 684.1)
Step 5: BA2
Figure imgf000090_0001
[02531 To a solution of 39 (1.0 g, 1.46 mmol) in THF (1.5 mL), MeOH (1.2 mL) and
H2O (0.3 mL) is added lithium hydroxide aqueous solution (5 M, 0.53 mL), After stirring at 0-25 °C for 2 hours, the mixture is neutralized with citric acid (1 M) to ~pH 7 before removal of the volatiles. The solid in the aqueous solution is then collected by filtration to give BA2 as an off-white solid (300 nig, 54% yield). (MS: [M+Hf 372.2)
Figure imgf000091_0001
90 Step 1: trifluoroethyl ether 40
Figure imgf000092_0001
[0254] To a solution of 22 (0.4 g, 0.93 mmol) in DMF (8 mL) is added sodium hydride (48 rng, 1.12 mmol) at 0 °C and the mixture is stirred for 30 minutes before 2,2,2- trifluoroethyl trifluoromethanesulfonate (0.165 mL, 1.12 mmol) is added slowly. After stirring at 0 °C for 3 hours, water (3 mL) is added the mixture is concentrated and purified by silica gel column chromatography (EA/hexanes =: 1/10) to give 40 as a white solid (218 mg, 46% yield).
Step 2; bezoaie 41
Figure imgf000092_0002
[0255] To a solution of 40 (1.4 g, 2.74 mmol) in THF (25 mL) is added TBAF (3,3 mL, 3.3 mmol) at 0 °C. After stirring at room temperature for 4 hours, water (2 mL) is added and the mixture is extracted with EA. The organic layer is washed with water and brine, and concentrated. The residue is then dissolved in DCM followed by addition of and TEA (1.92 mL, 13.8 mmol) benzoyl chloride (0.42 mL, 3.6 mmol). After stirring for 1 hour, water (1 mL) is added and the mixture is concentrated and purified by silica gel column
chromatography (EA/PE = 1 /5) to give 41 as a white solid (0.795 g, 77% yield).
Step 3: acetate 42
Figure imgf000092_0003
42 [0256] A solution of 41 (0.79 g, 2.1 mmol) in HO Ac (17.5 mL) and water (7.5 mL) is stirred at reflux for 30 minutes before cooled to room temperature, and co-evaporated with toluene (10 mL x 4), The residue is then dissolved in Py (12 mL) followed by addition of Ac?.0 (0.8 mL, 8.4 mmol). After stirring for 6 hours, the mixture is concentrated and purified by silica gel column chromatography (EA/PE = 1 /4) to give 42 as a white solid (0.82 g, 91% yield).
Step 4; guanosine 43
[0257] To a suspensi
Figure imgf000093_0001
(633 mg, 2.86 mmol) in dichloroethane (25 mL) at 80 °C is added BSA (2.74 mL, 10.1 mmol) and stirred for 1 hour before addition of TMSOTf (1.03 mL, 5.7 mmol). After stirring for 3 hours at 100 °C, the mixture is poured into sodium bicarbonate aqueous solution (60 mL) and extracted with DCM (60 mL χ 3), The combined organic layers are dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column chromatography
(MeOH/DCM = 1/20 to 1/10) to give 43 as a white solid (938 mg, 85% yield).
Step Si BA3
Figure imgf000093_0002
[0258] To a solution of 43 (0.5 g, 0.86 mmol) in MeOH (8 mL), THF (10 mL) and water (2 mL) is added sodium hydroxide aqueous solution (10 M, 0,34 mL) at 0 °C. After stirring for 30 minutes, HO Ac is added and the mixture is concentrated and purified by silica gei column chromatography (MeOH/DCM =1/10 to 1 /5) to afford BA3 as an oil (348 mg, 93% yield).
Figure imgf000095_0001
94 Step 1: pyrazole 44
Figure imgf000096_0001
[0259] To a solution of 2 (75 g, 395 mmol) in MeOH (600 mL) is added hydrazine hydrate (120 mL) at 25 °C. After stirring for 2 hours, the mixture is concentrated and the residue is dissolved in EtOH (600 mL) before addition of (ethoxymethylene)malononitrile (110 g, 901 ramol), After stirring at 78 °C for 30 minutes, the rmxtiire is concentrated and purified by silica gel column chromatography (EA/PE = 1/20 to 1/15) to give 44 as a pale yellow solid (42 g, 38% yield).
Step 2: thiourea 45
Figure imgf000096_0002
[0260] To a solution of 44 (16.0 g, 57 mmol) in MeOH (7.5 ml.) and water (25 mL) is added ammonium hydroxide (280 mL) and hydrogen peroxide (150 mL). After stirring at 25 °C for 16 hours, the mixture is poured into sodium sulfite aqueous solution (2 L) and then extracted with EA (700mL χ 3). The combined organic layers are dried over anhydrous sodium sulfate, filtered, and concentrated. The residue is then dissolved in acetone (90 mL) before benzoyl isothiocyanate (6.96 g, 42.7 mmol, 5.75 mL) is added at 25 °C. After stirring at 60 °C for 4 hours, the mixture is concentrated to give crude 45 as a yellow solid.
Step 3: pyrazolopyrimidinone 46
Figure imgf000097_0001
[0261] To a solution of the crude 45 o btained above in MeOH (150 mL) is added sodium hydroxide aqueous solution (0.7 M, 80 mL) followed by methyl iodide (6.8 g, 47,9 mmol, 3.0 mL-). After stirring at 20 °C for 2 hours, the mixture is neutralized with HO Ac to ~pH 6 followed by addition of water (80 mL), and extracted with EA (100 mL χ 3). The combined organic layers are dried over anhydrous sodium sulfate, filtered, and concentrated. The residue is then dissolved in MeOH (30 mL) and sodium hydroxide aqueous solution (1.4 M, 250 mL) is added. After stirrins at 100 °C for 2 hours, the mixture is concentrated and the residue is co-evaporated with toluene (200 mL χ 3) and dissolved in DCM (500 mL). Imid (18.5 g, 271 mmol), DMAP (1.66 g, 13,6 mmol), and TBSC1 (40. 9 g, 271 mmol) are then added. After stirring at 25 °C for 18 hours, saturated sodium bicarbonate aqueous solution (1 L) is added and the mixture is extracted with EA (500 mL * 3). The combined organic layers are dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (MeOH DCM = 1/60 to 1/30) to give 46 as a white solid (8.50 g, 34% yield over five steps).
Step 4; isobutyrate 47
Figure imgf000097_0002
[0262] To a solution of 46 (23.4 g, 53.5 mmol) in Py (120 mL) is added isobutyryl chloride (11.4 g, 107 mmol, 11.2 mL) at 25 °C. After stirring at 25 °C for 16 hours, ammonium hydroxide (0.5 mL) is added and the mixture is stirred for 30 minutes before concentrated. The residue is then dissolved in EtOAc (1 ,5 L), washed with saturated ammonium hydroxide aqueous solution (500 mL * 3) and brine (.500 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/PE = 1/30 to 1/3) to give 47 as a light yellow solid (24.0 g, 88% yield), (MS: [M+Naf 530.1)
Step 5: BBl
Figure imgf000098_0001
BB1
[0263] A solution of 47 (10.0 g, 19,7 mmol) in HOAc (6 mL) and water (3 mL) is stirred at 65 °C for 5 hours. The reaction mixture is then concentrated and triturated with DCM (15 mL). The solid is collected by Alteration to give curde BBl as a white solid (4.0 g). (MS: | Yt H i 354.0)
Figure imgf000099_0001
98 Step 1 : aceiosiide 49
Figure imgf000100_0001
[0264] To a solution of 48 (50 g, 135 nimol) in acetone (500 mi.) is added 2,2- dimethoxypropane (85 g, 816 mmol, 100 mL) and concentrated sulfuric acid (1.32 g, 13.5 mmol, 0.72 mL.). After stirring at 25 °C for 30 minute, saturated sodium bicarbonate aqueous solution (30 mL) is added. The solution is filtered, concentrated, and purified by silica gel column chromatography (MeOH/DCM = 1/200 to 1/50) to give 49 as a white solid (35 g, 63% yield). (MS: \ \\ \ \ \ 4 12. 1 }
Step 2: azide SO
Figure imgf000100_0002
[0265] To a solution of 49 (5.0 g, 12.2 mmol) in Py (50 mL) is added
methanesuifonyl chloride (2.1 g, 18 mmol, 1.4 mL) at 0 °C. After stirring at 25 °C for 1 hour, DCM (200 mL) is added, and the solution is washed with saturated sodium bicarbonate aqueous solution followed by brine, dried over anhydrous sodium sulfate, and concentrated. The residue is then dissolved in DMF (50 mL) followed by addition of sodium azide (3.4 g. 52.3 mmol). After stirring at 50 °C for 16 hours, DCM (400 mL) is added. The mixture is washed with water (300 mL), bnne, dried over sodium sulfate, concentrate, and purified by- silica gel column chromatography (EA/PE = 1/1) to give 50 as a light yellow solid (4.0 g). (MS: | M f i r 437. 1 ) Step 3: BO
Figure imgf000101_0001
[0266] A solution of 50 (50 g, 115 rnmoi) in TFA (125 niL) and water (125 mL) is stirred at 25 °C for 5 hours before concentrated, co-evaporated twice with toluene, and dissolved in MeOH (50 mi,). The mixture is then neutralized by sodium bicarbonate aqueous solution (1%) and triturated with MTBE. The solid is collected, washed with MTBE, dried, and dissolved in DMF (400 mL). Pd/C (10% w/w, 10 g) is then added and the mixture is stirred under an atmosphere of hydrogen (15 psi) at 25 CC for 6 hours before filtered and concentrated to give crude BC1 as a yellow oil (39 g). (MS: | Vi H i 371. 1)
Figure imgf000102_0001
101 Step 1 : indole 52
Figure imgf000103_0001
52
[0267] To a solution of indole (305 mg, 2.6 mmol) in MeCN (10 mL) is added sodium hydride (160 mg, 4.0 mmol) at 0 °C and stirred for 30 minutes before 51 (1,0 g, 2.6 mmol) is added. After stirring for 1 hour, saturated sodium bicarbonate aqueous solution (5 mL) is added and the mixture is extracted with EA (20 mL χ 3). The combined organic layers are washed with saturated sodium bicarbonate aqueous solution and brine, concentrated, and purified by silica gel column chromatography (EA/hexanes = 1/4) to give 52 as a yellow oil (886 mg, 71% yield). (MS: [M+H]+ 470.2)
Step 2: BC2
Figure imgf000103_0002
[0268] To a solution of 52 (610 mg, 1.3 mmol) in MeOH (9 mL) is added sodium methoxide (5.4 M in methanol, 0.54 mL). After stirring for 1 hour, hydrochloric acid (5 M, 0.5 mL) is added at 0 °C and the solution is stirred for 10 minutes before concentrated and purified by silica gel column chromatography (MeOH/DCM = 1/9) to give the desired product as a white solid (197 mg, 92% yield). (MS: [ X i 1 1 ! 234.1)
Figure imgf000104_0001
103 Step 1: pyrene 54
Figure imgf000105_0001
54
[0269] To a solution of 53 (560 mg, 2 mmol) in THF is added magnesium (54 mg, 2.3 mmol) followed by a small amount of iodine. After stirring at 55 °C for 3 hours, copper(I) iodide (213 mg, 1.1 mmol) is added at 0 °C. The mixture is stirred at room temperature for 45 minutes before 51 (367 mg, 0.98 mmol) is added at 40 °C. After stirring for 2 hours, saturated ammonium chloride aqueous solution (2 mL) and DCM (20 mL) are added. The layers are separated and the organic layer is washed by saturated sodium bicarbonate aqueous solution and brine, dried anhydride sodium sulfate, concentrated, and purified by silica gel column chromatography (EA/hexanes = 1/10 to 1/5) to give 54 as a white solid (52 mg, 10%).
Step 2: BC3
Figure imgf000105_0002
[0270] To a solution of 54 (230 mg, 0.4 mmol) in MeOH (5 mL) is added sodium methoxide (30% in MeOH, 0.23 mL, 1.2 mmol) at room temperature. After stirring for 1 hour, saturated ammonium chloride (5 mL) is added and the mixture is extracted by EA (10 mL x 3). The organic layers are washed with brine, dried over anhydrous sodium sulfate, and concentrated to give crude BC3 as a white solid (150 mg).
Figure imgf000106_0001
105 Step 1 : alcohol
Figure imgf000107_0001
[0271] To a solution of 55 (91.4 g, 481 mmol) in Py (600 mL) is added triiyl chloride (160.7 g, 577 mmol). After stirrmg at 60 °C for 16 hours, the mixture is concentrated and co- evaporated with toluene for three times. The residue is partitioned between DCM (400 mL) and saturated sodium bicarbonate aqueous solution (750 mL). The layers are separated and the aqueous phase is extracted with DCM (400 mL χ 2). The combined organic layers are dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/PE :::: 1/10 to 1/5) to afford 56 as a white solid (180.5 g, 87% yield).
Step 2; ketone 57
Figure imgf000107_0002
[0272] To a solution of 56 (176 g, 407 mmol) in MeCN (1.0 L) is added 1BX (228 g, 814 mmol). After stirring at 90 °C for 6 hours, the mixture is filtered and concentrated to give crude 57 as a light yellow oil (175 g). (MS: [M+Naf 453.0)
Step 3: ester 58
Figure imgf000107_0003
[0273] To a solution of sodium hydride (20.1 g, 502 mmol) in THF (1.0 L) is added methyl 2-dimemoxyphosphoiylacetate (96.3 g, 529 mmol, 76.5 mL) at 0 °C dropwise over 15 minutes. After stirring for 60 minutes, crude 57 (175 g) obtained above in THF (500 mL) is added dropwise at 0 °C. After stirring at 25 °C for 16 hours, water (50 mL) is added at 0 °C and the volatiles are removed and brine (500 mL) is added. The mixture is then extracted with DCM (500 ml. x 3). The combined organic layers are dried over anhydrous sodium sulfate, filtered, and concentrated. The residue (198 g of 216 g obtained above) is then dissolved in EA (500 mL) and Pd-'C (10% w/w, 10 g) is added. After stirring under a hydrogen atmosphere (20 psi) at 25 °C for 16 hours, the mixture is filtered and the filtrate is concentrated and purified by silica gel column chromatography (EA/PE = 1/15 to 1/10) to give 58 as a white soli d ( 120 g, 66% yield) (MS : M+Naj 5 1 1. i )
Step 4: alcohol 59
Figure imgf000108_0001
[0274] To a solution of lithium aluminum hydride (6.21 g, 164 mmol) in THF (200 mL) is added 58 (20.0 g, 40.9 mmol) in THF (50 mL) slowly at 0 °C. After stirring at 25 °C for 16 hours, the reaction is quenched by sequential addition of water (6.2 mL), sodium hydroxide aqueous solution (15%, 6.2 mL), and water (18.6 mL). The mixture is then dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/PE = 1/10 to 1/4) to give 59 as a white solid. (MS: [M+Na]+ 483.2)
Step 5: alcohol 60
Figure imgf000108_0002
[0275] To a solution of sodium hydride (60% w/w, 6.95 g, 174 mmol) in THF (200 mL) is added 59 (20.0 g, 43.4 mmol) in THF (80 mL) at -20 °C dropwise over 5 minutes. After stirring at 25 °C for 2 hours, benzyl bromide (22.3 g, 130 mmol, 15.5 mL.) is added dropwise and the mixture is stirred at 80 °C for 16 hours before water (2 mL) is added at 0 °C. The mixture is diluted with water (200 mL) and extracted with DCM (200 mL x 3). The combined organic layers are dried over anhydrous sodium sulfate, filtered and concentrated. The residue is then dissolved in DCM (200 mL) and DCA (5.48 g. 42.5 mmol, 12.0 mL) is added. After stirring at 25 °C for 3 hours, saturated sodium bicarbonate aqueous solution is added at 0 °C. The mixture is then extracted with DCM (150 mL χ 3). The combined organic solvent are dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/PE = 1/5 to 1/2) to give 60 as a yellow oil (12.1 g, 90% yield).
Step 6; beuzoate 61
Figure imgf000109_0001
[02761 To a solution of 60 (24.0 g, 78 mmol) in DCM (500 mL) is added benzoyl chloride (16,4 g, 116.7 mmol, 13,6 mL) and TEA (23.6 g, 233,5 mmol, 32,4 mL). After stirring at 25 °C for 16 hours, the mixture is concentrated and purified by silica gel column chromatography (EA/PE = 1/15 to 1/10) to give 61 as a light yellow oil (30.0 g, 93% yield). (MS: [M+Na]+ 435.1)
Step 7: acetate 62
Figure imgf000109_0002
[0277] A mixture of 61 (29.0 g, 70.3 mmol) and water (3.0 mL) in HO Ac (220 mL) is stirred at 70 °C for 16 hours before saturated sodium bicarbonate aqueous solution is added. Hie mixture is then extracted with DCM (400 mL χ 3). The combined organic layers are concentrated and the residue is dissolved in Py (30 mL) followed by addition of AciQ (28.5 g, 280 mmol, 26 mL). After stirring at 20 °C for 16 hours, saturated sodium bicarbonate aqueous solution is added and the mixture is then extracted with DCM (500 mL χ 3). The combined organic layers are concentrated and purified by silica gel column chromatography (EA/PE - 1/10 to 1/5) to give 62 as a white solid (31.1 g, 97% yield). (MS: [M+Na] + 479.1) Step 8: purine 63
Figure imgf000110_0001
[0278] To a suspension of 06-diphenylcarbanioyl-A¾-isobuiyr 7iguanine (5.47 g, 13.1 mmol) in MeCN (150 raL) is added BSA (11.6 g, 57.0 mmol, 14.1 mL) at 20 °C. After stirring at 63 °C for 30 minutes, the volatiles are removed and the residue is dissolved in MeCN (200 mL) before 62 (5.00 g, 11.0 mmol) in MeCN (50 mL) and TMSOTf (3.65 g, 16.4 mmol, 3.0 mL) are added at -15 °C. After stirring at 63 °C for 50 minutes, the mixture is cooled to 0 CC, poured into saturated sodium bicarbonate aqueous solution and extracted with EA (150 mL x 3). The combined organic layers are dried over anhydrous sodium sulfate, and concentrated, and purified by silica gel flash chromatography (EA/PE = 1/3 to 1/1) to give 63 as a white solid, (MS: [M+H]+ 813.1)
Figure imgf000110_0002
[0279] A solution of 63 (16.2 g, 19,9 mmol) in 90% TFA aqueous solution (60 mL) is stirred at 20 °C for 30 minutes before poured into saturated sodium bicarbonate aqueous solution at 0 °C and extracted with EA (100 mL χ 4). The combined organic layers are dried over anhydrous sodium sulfate, concentrated, and purified by silica gel flash chromatography (EA/PE = 1/1 to 1/0) to give 64 as a white solid (1 1.4 g, 93% yield), (MS: [ Y!-H | 618.1) Step 10: si!y! ether 65
Figure imgf000111_0001
[0280| To a solution of 64 (15.0 g, 24.3 mmol) in EtOH (500 niL) is added Pd/C (10% w/'w, 2.0 g) and concentrated hydrochloric acid (10 drops). After stirring at 50 °C under an atmosphere of hydrogen (45 psi) for 15 hours, the mixture is filtered and solid is washed with EtOH (100 mL χ 3). The filtrate is concentrated and one-third of the residue is dissolved in DMF (60 mL) followed by addition of Imid (1.57 g, 23,0 mmol), DMAP (46.9 mg, 0.38 mmol) and trii soprop Isily 1 chloride (2.22 g, 11 .5 mmol, 2.5 mL), After stirring at 20 °C for 16 hours, saturated sodium bicarbonate aqueous solution (20 mL) and water (100 mL) are added. The mixture is then extracted with EA (100 mL χ 2). The combined organic layers are dried over anhydrous sodium sulfate, filtered, concentrated, and purified by siiica gel column chromatography (EA/PE = 1/5 to 2/1) to give 65 as a white solid (4.52 g, 86% yield). (MS: [M+H| 684.4)
Step 11: BC4
Figure imgf000111_0002
[0281] To a solution of 65 (3.0 g, 4.4 mmol) in EtOH (30 mL) is added sodium hydroxide aqueous solution (2 M, 31 mL.) at 0 °C. After stirring at 0 °C for 30 minute, the mix lure is neutralized by addition of hydrochloric acid solution (1 N) and HOAc at 0 °C. Toluene (30 mL) is then added and mixture is concentraled to give crude BC4 as a white solid (3.0 g), {MS: | V! H i 538.2)
Figure imgf000113_0001
112 Step 1 : alcohol 66
Figure imgf000114_0001
[0282] To a solution of 15 (2.0 g, 5.66 mmol) in Py (56 mL) at 0 °C is added 1,3- dichloro-l,l ,3,3-tetraisopropyldisiloxane (1.79 g, 5.66 mmol, 1.8 mL) slowly. After stirring at 0 °C for 30 minutes and 25 °C for 12 hours, the solution is concentrated and purified by silica gel column chromatography (MeOH/DCM = 1/20) to give 66 (1.6 g, 47% yield). (MS:
[M+H]+ 596.3)
Step 2: purine 67
Figure imgf000114_0002
[0283] To a solution of 66 (8.0 g, 13.5 mmol) and 2-(4-nitrophenyl)ethanol (3.37 g, 20,2 mmol) in THF (100 mL) is added DIAD (6.81 g, 33,7 mmol, 6.6 mL) and PPh3 (8.83 g, 33,7 mmol) at 25 °C slowly. After stirring at 25 °C for 12 hours, water (5 mL) is added and the mixture is concentrated and purified by silica gel column chromatography (MeOH/DCM = 1/20) to give 67 as a pale yellow solid (4.5 g, 44% yield), (MS: | \M ! j 745.3)
Step 3: alcohol 68
Figure imgf000115_0001
[0284] To a solution of 67 (4.20 g, 5,64 mmol) in MeCN (40 mL) at 25 °C is added 1BX (3.16 g, 11.3 mmol). After stirring at 80 °C for 12 hours, the mixture is filtered and concentrated, and dissolved in THF (50 mL). Sodium triacetoxyborohydride (5.7 g, 27.0 mmol) is then added at 0 °C slowly. After stirring at 25 °C for 6 hours, water (5 mL) is added and the mixture is concentrated and purified by silica gel column chromatography
(MeOH/DCM = 1/20) to give 68 as a pale yellow oil (1.0 g, 33% yield). (MS: [M+H]+ 745.3)
Step 4: azide 69
Figure imgf000115_0002
[0285] To a solution of 68 (2.2 g, 2.95 mmol) and DMAP (1.44 g, 11.8 mmol) in DCM (140 mL) and Py (10 mL) is added trifluoromethanesulfonic anhydride (1.33 g, 4.72 mmol, 0.78 ml.) at 0 °C slowly. After stirring at 0 °C for 1.5 hours, the mixture is concentrated. The residue is then dissolved in DMF (10 mL) and sodium azide (0.49 g, 7.53 mmol) is added. After stirring at 60 °C for 6 hours, the solution is concentrated and purified by preparative HPLC (MeOH/water with 0.1% HCOOH: 40-100%) to give 69 as a pale yellow solid (1.50 g, 79% yield). (MS: [M+H] ' 770.4)
Step 5: BCS
Figure imgf000116_0001
[0286] To a solution of 69 (2.50 g, 3,25 mmol) in THF (12 mL) is added TBAF (13.1 g. 50.1 mmol) and HOAc (1.50 g, 25.0 mmol, 1.43 mL) at 15 °C slowly. After stirring at 15 °C for 12 hours, the mixture is concentrated. The residue is then dissolved in DCM (20 mL), washed with water (5 mL χ 2), dried over anhydrous sodium sulfate, filtered, and purified by silica gel column, chromatography (MeOH/DCM = 1/20) to give BCS as a pale yellow solid (900 mg, 53% yield). (MS: j \! i l l 528.2)
[0287] The following compounds are prepared essentially by the method for Intermediates BCS and BC4 above.
Table 1. Intermediates BC6 and BC7
Figure imgf000116_0002
Figure imgf000117_0001
116 Step 1 : beiizoate 70
Figure imgf000118_0001
[0288] To a solution of 33 (120 g, 449 mmol) in Py (1,0 L) is added TMSC1 (390 g, 3.59 mol, 454 mL). After stimng at 0 °C for 2 hours, benzoyl chloride (316 g. 2.25 mol, 261 mL) is added dropwise and the mixture is stirred at 25 °C for 14 hours before cooled to 0 °C Water (240 mL) is then added and the mixture is stirred at 25 °C for 30 minutes before ammonium hydroxide (460 mL) is added at 0 °C. After stirring for 2 hours, the mixture is concentrated to give 70 as a white solid (150 g, 90% yield).
Step 2: die! 71
Figure imgf000118_0002
[0289] To a solution of 70 (150 g, 404 mmol) in Py (500 mL) is added DMTrCl (274 g, 808 mmol), TEA (81.8 g, 808 mmol 112 mL) and DMAP (4.93 g, 40,4 mmol) at 0 °C. After stimng at 25 °C for 16 hours, saturated sodium bicarbonate aqueous solution (1 L) is added and the mixture is extracted with EtOAc (600 mL x 3). The combined organic layers are dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/PE 1/4 to 1/2 then MeOH/DCM 1/100 to 1/20) to give 71 as a white solid (65,0 g, 24% yield).
Step 3: silyl ether 72
Figure imgf000119_0001
[0290] To a solution of 71 (65.0 g, 96.5 mmol) in Py (500 mL) is added silver nitrate (32.8 g, 193 mmol) and TBSC1 (29. 1 g, 193 mmol) at 25 °C. After stirring at 25 °C for 1 hour, saturated sodium bicarbonate aqueous solution (1 L) is added and the mixture is extracted with EtOAc (600 mL χ 3). The combined organic layers are dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/PE - 1/4 to 1/1) to give 72 as a white solid (20.0 g, 26% yield).
Figure imgf000119_0002
[0291] To a solution of 72 (12.0 g, 15,2 mmol) in DIPEA (15 mL) and DCM (30 mL) is added DMAP (744 mg, 6.09 mmol) and 2-cyanoethyl N- diisopropylchlorophosphoramidite (5.41 g, 22.9 mmol) at 25 °C. After stirring for 2 hours, the mixture is purified directly by basic silica gel column chromatography (EA PE = 1/4 tol/1) to give A2 as a white solid (13.0 g, 86% yield).
Step 5: Al
Figure imgf000120_0001
[0292] To a solution of A2 (3.8 g, 3.9 mmol) in MeCN (20 mL) is added water (0.1 mL) and pyridinium trifluoroacetate (1.1 g, 5.8 mmol) at 25 °C and stirred for 5 minutes before terf-butylamine (14.0 g, 0.19 mmol) is added. After stirring for 15 minutes, the volatiles are removed and the residue is dissolved in DCM (20 mL). A solution of DCA (1.9 g, 14.6 mmol) in DCM (20 mL) is then added. After stirring for 30 minutes, TEA (3 mL.) is added and the mixture is concentrated and purified by reverse-phase silica gel column chromatography (MeCN with 0.1% TEA/water = 0% to 100%) to give Α ΜΈΑ salt as a white solid (1.5 g; 71% yield). (MS: [M÷H 549.2)
Figure imgf000121_0001
120 [0293] To a solution of A2 (494 mg, 0.5 mmol) and triphenylphosphine (197 mg, 0.75 mmol). and 2-( eTi-buiyldimeih silyloxy)ethano] (132 mg, 0.75 mmol) in THF (5 mL) is added DIAD (0.15 mL, 0.75 mmol). After stirring at room temperature for 5 hours, the mixture is concentrated and purified by silica gel column chromatography (EA/nexanes :::: 1/9 to 1/4) to give AB1 as a white solid (230 mg. 40% yield).
Figure imgf000123_0001
122 Step 1 : diol 73
Figure imgf000124_0001
[0294] To a solution of crude BC1 (39 g) in Py (40 mL) is added DMTrCl (35.9 g, 106 mmol) at 0 °C. After stirring at 25 °C for 16 hours, MeOH (50 mL) is added and the mixture is concentrated. The residue is then dissolved in DCM (600 mL), washed with saturated sodium bicarbonate aqueous solution and brine, concentrated, and purified by silica gel column chromatography (MeOH/DCM = 1/100 to 1/50) to give 73 as a pale yellow solid (34.0 g, 48% yield over two steps). (MS: j \ I i i i ' 673.2)
Step 2: silyl ether 74
Figure imgf000124_0002
[0295] To a solution of 73 (1.0 g, 1.49 mmol) in Py (10 mL) is added silver nitrate (380 mg, 2.24 mmol, 0.38 mL) at 0 °C After stirrmg for 15 minutes, TBSC1 (270 mg, 1.79 mmol) is added and the mixture is stirred at 25 °C for 2 hours before saturated sodium bicarbonate aqueous solution is added, The mixture is then extracted with EA (10 mL) and the organic layer is washed with brine, concentrated, and purified by silica gel column chromatography (EA/PE = 1 /5) to give 74 as a pale yellow solid (400 mg, 34% yield). (MS: [M+H]+ 787.3)
Step 3: AC1
Figure imgf000125_0001
[0296] To a solution of 74 (1.0 g, 1.27 mmol) in Py (10 mL) is added diphenyl phosphite (80%, 744 mg, 2.54 mmol, 0.61 mL). After stirring at 20 °C for 1 hour, EA (2 mL) and saturated sodium bicarbonate aqueous solution (2 mL) are added and the mixture is stirred for 1 hour. The layers are separated and the organic layer is concentrated. The residue is then dissolved in DCM (1.0 mL.) and DCA (164 mg, 1.27 mmol, 0.1 mL) is added. After stirring at 25 °C for 30 minutes, TEA (1 mL) is added and the solution is concentrated and purified by reverse-phase silica gel column chromatography (MeCN with 0.1% TEA'water = 0% to 100%) to give AC ! as a white solid (500 mg, 72% yield). (MS: \ \\ I I I 549.2)
Figure imgf000126_0001
125 Step 1 : alcohol 75
Figure imgf000127_0001
[0297] To a solution of BC2 (348 mg, 1.49 mmol) in Py (15 mL) is added DMAP (18 mg, 0.15 mmol) and DMTrCl (0.66 g, 1.94 mmol). After stirring overnight, MeOH (3 ml.) is added and the mixture is concentrated and purified by silica gel column chi matography (EAnexanes = 1/4) to give 75 (662 mg, 83% yield). (MS: [M+H]+ 536.2)
Step 2: AC2
Figure imgf000127_0002
[0298] To a solution of 75 (0.2 g, 0.37 mmol) in DCM (4 mL) is added D1EPA (0.15 g, 1.2 mmol, 0.2 mL) and 2-cyatioethy3 A,r,N-diisopropylchlorophosphoraimdite (0.14 g, 0.56 mmol, 0.13 raL). After stirring for 4 hours, the mixture is concentrated and purified by silica gel column chromatography (EA'hexanes with 1% TEA = 1/4) to give AC2 (232 mg, 85% yield). (MS: [M-NiPn+H20]+ 653.2)
Figure imgf000128_0001
127 Step 1 : silyl ether 76
Figure imgf000129_0001
76
[0299] To a solution of 16 (11.0 g. 16.8 mmol) in DCM (80 ml .} is added TBSC1 (7.59 g, 50.3 mniol) and Imid (3.43 g, 50.3 mmol). After stirring at 25 °C for 16 hours, sodium bicarbonate aqueous solution (5%, 30 mL) is added and the mixture is extracted with DCM (60 mL χ 3). The combined organic layers are washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by basic silica gel column chromatography (EA/PE = 1/5 to 1/1) to 76 as a white solid (2.1 g, 16% yield).
Step 2: G2
Figure imgf000129_0002
[0300| To a solution of 76 (900 mg, 1.17 mmol) in THF (4, 0 mL) and DIEPA (4.0 mL) is added DMAP (14.3 mg, 0.12 mmol) followed by 2-cyarioethyl N,N- diisopropylchlorophosphoramidite (415 mg, 1,76 mmol) dropwise at 0 °C. After stirring at 20-25 °C for 2 hours, sodium bicarbonate aqueous solution (5%, 15 mL) is added at 0 °C. The mixture is then diluted with water (15 mL) and extracted with EA (15 mL χ 3). The combined organic layers are washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (acetone/PE = 1/10 to 1/3) to give G2 as a white solid (600 mg, 53% yield). Step 3; Gl
Figure imgf000130_0001
[0301] To a solution of Gl (7,0 g, 7, 22 mmol) in MeCN (30 mL) is added water (0.11 mL) and pyridinium trifluoroacetate (4.18 g, 21.7 mmol) at 25 °C. After stirring at 25 °C for 15 minutes, fc/ -butylamme (37 mL) is added and the mixture is stirred at 25 °C for 45 minutes before concentrated. The residue is then dissolved in DCM (30 mL) and a solution of DCA in DCM (6% v/v, 30 mL) is added dropwise. After stirring at 20-25 °C for 30 minutes, DCM (30 mL) and TEA (4 mL) are added. The mixture is then concentrated, dissolved in a mixture of MeCN (5 mL) and water (5 mL), and purified by CI 8 reverse-phase medium pressure liquid chromatography (MeCN with 0.1% TEA/water = 0% to 60%) to give G1»TEA salt as ayellow solid (2,30 g, 56% yield). (MS: [M+H]+ 532.3)
Figure imgf000131_0001
130 Step 1 : diol 77
Figure imgf000132_0001
[0302] To a solution of two batches of the crude BB1 (8.0 g) obtained above in Py (50 mL) is added DMTrCl (9.2 g, 27.2 mmol). After stirring at 20-30 °C for 1 hour, MeOH (10 mL) is added and the mixture is concentrated and purified by silica gel column chromatography (EA/PE = 1 /5 to MeOH/DCM := 1/20) to give 77 as a yellow solid (11.0 g, 30% yield over two steps). (MS: [M+Naf 678.2)
Step 2; si!y! ether 78
Figure imgf000132_0002
[0303] To a solution of 77 (9,0 g, 14.0 mmol) in Py (50 mL) is added TBSC1 (2,48 g, 16.5 mmol) and silver nitrate (5.83 g, 34,3 mmol). After stirring at 25-30 °C for 30 minutes, saturated sodium bicarbonate aqueous solution is added. The mixture is then extracted with DCM (200 mL χ 2), and the combined organic layers are washed with water (50 mL), brine (50 mL), dned over anhydrous sodium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA;'PE = 1/10 to 1/5 to 1/3) to give 78 as a white foam (1.50 g, 14% yield). (MS: [M+Na]+ 792.2)
Step 3: GB1
Figure imgf000133_0001
[0304] To a solution of 78 (2.50 g, 3.3 mmol) in DIEPA (5 mL) and DCM (5 mL) is added DMAP (200 mg, 1.62 mmol) and 2-cyanoethyl N,N-diisopropylchlorophosphoramidite (1.0 g, 4.22 mmol). After stirring at 20-25 °C for 2 hours, the mixture is concentrated and purified by silica gel column chromatography (EA/PE - 1/10 to 1/4) to give GUI as a white foam (2,30 g, 73% yield).
Step 4; GB2
Figure imgf000133_0002
[0305] To a solution of GB1 (2.30 g, 2.4 mmol) in MeCN (2.0 mL.) is added water (0.11 mL, 6.1 mmol) and pyridinium trifluoroacetate (687 mg, 3.56 mmol) at 25 °C. After stirring at 25-30 °C for 30 minutes, the mixture is concentrated and the residue is dissolved in MeCN (20 mL) before /er/-butylamine (10.5 g, 144 mmol, 15.0 mL) is added. After stirring at 25-30 °C for 30 minutes, the mixture is concentrated and DCM (20 mL) followed by addition of a solution of DC A in DCM (6% v/v, 18.2 mL). The mixture is stirred at 25-30 °C for 30 minutes before neutralized by TEA to ~pH 7, concentrated, and purified by CI 8 reverse-phase silica gel column chromatography (MeCN with 0.1% TE A/water =: 0% to 40%) to give GB2 as a while solid (800 mg, 63% yield). (MS; | M - l l j 532.0)
Figure imgf000134_0001
133 Step 1 : alcohol 79
Figure imgf000135_0001
[0306| To a solution of BC4 (1.97 g, 2.4 mmol) in Py (20 mL) is added DMTrCl (984 mg, 2.90 mmol) at 25 °C. After stirring for 3 hours, MeOH (30 mL) is added and the mixture is concentrated, and purified by basic silica gel flash chromatography (EA/PE = 1/5 to 4/1) to give 79 as a light yellow powder (1.65 g, 82% yield). (MS: j \i · H | 840.2)
Step 2; GO
Figure imgf000135_0002
[0307] To a solution of 79 (2.0 g, 2.38 mmol) in Py (15 mL) is added diphenyl phosphite (1.7 g, 7.1 mmol, 1.4 mL). After stirring at 20 °C for 30 minutes, saturated sodium bicarbonate aqueous solution (30 mL) is added and the mixture is stirred for 1 hour. The mixture is then extracted with EA (30 mL χ 3). The combined organic layers are washed with saturated sodium bicarbonate aqueous solution (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, and concentrated. The residue is then dissolved in DCM (40 mL) followed by addition of water (0.4 mL) and DCA (6% v/v in DCM, 40 mL). After stirring at 20 °C for 15 minutes, the mixture is neutralized to ~pH 7 by TEA, concentrated, and purified by reverse-phase silica gel column chromatography (MeCN/water = 25% to 90%) to give GC1 as a white solid (1.3 g, 90% yield). (MS: ! V! l i | 602, 1 )
Figure imgf000136_0001
135 Step 1: alcohol 80
Figure imgf000137_0001
[0308] To a solution of BC5 (900 mg, 1.71 mmol) in Py (10 mL) is added DMTrCl (809 mg, 2.39 mmol) at 15 °C. After stirring at 15 °C for 12 hours, MeOH (0.5 mL) is added and the mixture is concentrated and purified by silica gel column chromatography
(MeOH/DCM - 1/50) to give 80 as ayellow oil (1.0 g, 71% yield). (MS: [M+H]+ 528.2)
Figure imgf000137_0002
[0309] To a solution of 80 (2.05 g, 2.47 mmol) in DMF (5.0 mL) is added Imid (673 mg, 9.88 mmol) and TBSC1 (745 mg. 4.94 mmol, 0.61 mL) at 15 °C. After stirring at 15 °C for 12 hours, the mixture is concentrated and the residue is triturated with water (10 mL). The solid is then collected and washed with water (10 mL χ 2). PE (10 mL χ 2), dried, and dissolved in THF (18 mL) before PPh:, (1.11 g, 4.24 mmol) is added at 15 °C. After stirring at 15 °C for 2.5 hours, water (0.16 mL) is added and the mixture is stirred at 50 °C for 12 hours. The solution is then concentrated and purified by reverse-phase preparative-HPLC (MeOH with 0.1% TEA'water = 20% to 80%), to give GC2 as a white solid (900 mg, 58% yield). (MS: [M+H]+ 918.1)
[0310] The following compounds are prepared essentially by the method for intermediates Al, A2, AC1, AC2, Gl, G2, GO, and GC2 above.
Figure imgf000138_0001
137
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
142
Figure imgf000144_0001
143
Figure imgf000145_0001
Figure imgf000146_0001
145
Figure imgf000147_0001
146
Figure imgf000148_0001
147
Figure imgf000149_0001
148
Figure imgf000150_0001
149
Figure imgf000151_0001
150
Figure imgf000152_0001
151
152"
Figure imgf000154_0001
153 [0311] To a solution of cmde Gl (obtained from 187 nig of G2-TEA salt, 0.2 mmol, containing Py»DCA salt) in MeCN (0.5 mL) is added a solution of A2 (0.26 g, 0.26 mmol) in MeC (0.2 mL). After stirring for 30 minutes, DDTT (46 mg, 0.22 mmol) is added and the mixture is stirred for 1 hour before concentrated. The residue is dissolved in DCM (4.8 mL) and water (0.036 mL) and DCA (6% in DCM, 4.8 mL) are added. After stirring for 10 minutes, Py (1 mL) is added and the mixture is concentrated and purified by silica gel column chromatography (MeOH/DCM = 1/10 to 1/5) to give Ci«Py salt as a white solid (86 mg, 35% yield). (MS: [M+H]+ 1148.0)
Figure imgf000156_0001
155 [0312] To a mixture of Αί (1.0 g, 1.82 mmol, co-evaporated MeCN 20 mL * 3) and G2 (2.3 g, 2,37 mmol, co-evaporated with MeCN 20 mL χ 3) is added ietrazole (0.45 M in MeCN, 10 mL) at 25 °C and stirred for 1 hour before elemental sulfur (1.75 g, 6.84 mmol) is added. After stirring for 1 hour, MeCN (20 mL) is added and the mixture is filtered and concentrated. The residue is dissolved in DCM (100 ml.) and DCA (1.96 g, 15.2 mmol, 1.25 mL) is added. After stirring at 25 °C for 2 hours, saturated sodium bicarbonate aqueous solution (100 mL) is added. The layers are separated and the aqueous layer is extracted with EA (100 mL x 3). The combined organic layers are dried over anhydrous sodium sulfate, filtered, concentrated, and purified by reverse-phase silica gel column chromatography (MeCN with 0.1 % TEA/water = 0% to 100%) to gi ve C2»TEA salt as a white solid (100 mg, 5% yield). (MS: {[M+2H]2+}/2 574.6)
Figure imgf000158_0001
157 [0313] To a solution of A2 (510 mg, 0.52 mmol, co-evaporated with MeCN 5 mL χ 3) in MeCN (1 mL) treated with 3A MS (100 mg) for 30 minutes is added a mixture of Gl (250 mg, 0.47 mmol, co-evaporated with MeCN .5 mL χ 3) and pyridinium trifluoroacetate (109 mg, 0.56 mmol, co-evaporated with MeCN 5 mL χ 3) in MeCN (1.5 mL) treated with 3 A MS (50 mg) for 30 minutes. After stirring for 4 hours, TBHP (5.5 M in decane, 0.26 ml.) is added and the mixture is stirred for 30 minutes before sodium bisulfite aqueous solution (33%, 0.24 mL) is added at 0 °C. The mixture is then stirred at room temperature for 10 minutes before filtered and concentrated. The residue is dissolved in DCM (6.2 mL) followed by addition of water (0.09 mL) and DCA (0.37 ml.) in DCM (6.2 mL). After stirring at room temperature for 10 minutes, Py (0,73 mL, 9.05 mmol) and DCM (35 mL) are added. The mixture is washed with water (10 mL χ 2) and the combined aqueous layers are extracted by dichloromethane (10 mL x 2). The combined organic layers are dried over anhydrous sodium sulfate, concentrated, and purified by Silica gel column chromatography fvieOH ί'Η ί'Ί.' Py = 10:89.5:0.5 to 25:74.5:0.5) to give C3 as a white solid (250 mg, 47%). (MS: [M+H]+ 1132.2)
Figure imgf000160_0001
159 [0314] To a solution of Gl (500 nig, 0.94 mmol, co-evaporated with MeCN 10 mL 3) and pyndimum trifluoroacetate (218 mg, 1.13 mmol. co-evaporation with MeCN 10 mL x 3) in MeCN (3 mL) treated with 3 A MS (100 mg) for 30 minutes is added a solution of A2 (976 mg, 0.99 mmol, co-evaporated with MeCN 10 mL χ 3) in MeCN (2 mL) treated with 3 A MS (200 mg) for 30 minutes. After stirring at room temperature for 2,5 hours, the mixture is concentrated and co-evaporated with MeCN (10 mL χ 2). The residue is then dissolved in DCM (20 mL) followed by addition of borane dimethyl sulfide complex (2 M in THF, 0.94 mL, 1.88 mmol) dropwise. After stirring at room temperature for 1 hour, MeOH (0.17 mL) is added at 0 °C and stirred for 20 minutes before concentrated to give crude CA1.
Figure imgf000162_0001
161 Step 1 : squaramide 81
Figure imgf000163_0001
[0315] To a solution of GC2 (802 rng, 0.87 ramol) in DMF (5 mL) is added 3,4- dimethoxy-3-cyclobutene-l,2-dione (186 mg, 1.31 mmol) at 15 °C slowly. After stirring at 15 °C for 2 hours, the mixture is concentrated and purified by reverse-phase preparative HPLC (MeCN with 0.1% TEA/water = 0% to 100%) to give a mixture of the desired product and an unidentified byproduct (0.7 g, ca. 84% purity). (MS: [M+H]+ 1028,4)
Step 2: CC S
Figure imgf000163_0002
[0316] To a solutio of 81 (ca. 84% pure, 0.6 g) obtained above and AO (0,64 g, 1.17 ramol) in DMF (5.0 mL) is added TEA (177 mg, 1.75 ramol. 0.24 mL) at 15 °C. After stirring at 15 °C for 12 hours, the mixture is concentrated and the residue is dissolved in DCM (5.0 mL) before DCA (470 mg, 3.65 mmol, 0.3 mL) is added. The mixture is then stirred at 15 °C for 15 minutes before concentrated. The residue is purified by reverse-phase preparative HPLC (MeCN with 0. 1% TEA/water = 0% to 100%) to give CO as a while solid (0.54 L;. 40% yield over two steps). (MS: I \1 1 11 1242.3)
Figure imgf000165_0001
164 Step 1: sulfamate
Figure imgf000166_0001
[0317] To a solution of 4-nitrophenyl chlorosulfate (0.31 g, 1.31 mmol) in DCM (1.0 mL) is added a solution of of GC2 (0.40 g, 0.44 mmol), 4-nitrophenol (0.61 g, 4.4 mmol) and TEA (0.73 mL, 5.23 mmol) in DCM (5 mL) at -78 °C. After stirring for 30 minutes, the mixture is warmed to room temperature, diluted with DCM (20 mL), and washed with water (20 ml, x 3). The combined aqueous layers are extracted with DCM (20 mL x 2) and the combined organic layers are dried over anhydrous magnesium sulfate, filtered, concentrated, and purified by silica gel column chromatography (EA/PE with 1% TEA = 1/5 to 1/2) to give 82 (0.30 g, 59 % yield) as a white solid. (MS: | XI I I I 1 1 18.9)
Step 2: CC2
Figure imgf000166_0002
[0318] To a solution of 82 (0.2 g, 0.18 mmol) in THF (1.0 mL), 4A MS (0.05 g) and TEA (0.12 mL, 0.89 mmol) is added AC1 (0.12 g, 0.21 mmol). After stirring for 12 hours, the mixture is diluted with THF (2 mL), filtered, and concentrated. The residue is then dissolved in DCM (5.0 mL) before water (0.1 mL) and a solution of DCA (0.46 mL) in DCM (5.0 mL) are added. After siirrmg for 15 minutes, the mixture is neutralized with TEA to ~pH 7 before concentrated and purified by reversed-phase C18 silica gel column chromatography (MeCN with 0.5"·.. TEA/water - 0% to 40%) to give CC2 (0.11 g, 44% yield) as a white solid. (MS: [M+H]+ 1226,0)
Figure imgf000168_0001
167 Step l: C4
Figure imgf000169_0001
[0319] To a solution of A4 (1.0 g, 1.01 mmol) and Gl (1.07 g, 2.02 mn ol) in MeCN (10 mL) is added Py»DCA (420 mg, 2.02 mmol). After stirring at 20 °C for 2 hours, TBHP (70% in water, 0.65 mL, 5.05 mmol) is added and the mixture is stirred for 1 hour before sodium bicarbonate aqueous solution (50 mL) is added, The mixture is extracted with ethyl acetate (100 mL) and the organic layer is washed with brine (30 mL). dried with anhydrous sodmm sulfate, filtered, and concentrated. The residue is then dissolved in a mixture of DCM (20 mL), TFA (1.0 mL) and triethylsilane (5.0 mL). After stirring for 2 hours, the mixture is neutralized with solid sodium bicarbonate to ~pH 7. The mixture is then filtered and the solid is washed with EA (50 mL χ 3). The filtrate is concentrate and purified by preparative HPLC (MeCN with 0.1% TEA/water = 0% to 30%) to give C4»TEA as a white solid (620 mg, 49% yield). (MS: [M+H]+ 1 131.1)
Step 2: phosphodiester 83
Figure imgf000170_0001
[0320] To a solution of C4 (583 mg, 0.515 mmol) in Py (10 mL) is added DMOCP (583 mg, 3.16 mmol) at 20 C. After stirring for 2 hours, iodine (654 mg, 2.58 mmol) is added and the mixture is stirred for 1 hour before saturated sodium sulfate aqueous solution (30 mL) and saturated sodium bicarbonate aqueous solution (30 mL) is adde. The mixture is then extracted with EA (100 mL), washed with brine (60 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by reversed-phase silica gel column
chromatography (MeCN with 0.1% TEA/water = 20% to 40%) to give 83·ΤΕΑ as a white solid (172.0 mg, 31% yield), (MS: j .\ H ! | 1076.1)
Step 3: bisphosphodiester 84
Figure imgf000170_0002
[0321] A solution of 83 (100 mg, 0.093 mmol) in MeOH (1,0 mL) and ammonium hydroxide (1.0 mL) is stirred at 50 °C for 12 hours. The mixture is then concentrated and purified by reverse-phase silica gel column chromatography (MeCN with 0.1% TEAVater :=: 20% to 40%) to give 84 as a yellow solid (27.0 nig, 32% yield), (MS: [M+Hf 902.5)
Step 4: E5
Figure imgf000171_0001
[0322] A solution of 84 (27 mg, 0.030 mmol) in TEA»3HF (10 mL) is stirred at 50 °C for 3 hours. The mixture is then neutralized with cold triethylammonium bicarbonate to ~pH 7, concentrated, and purified by a C 18 reverse-phase silica gel column chromatography (MeCN with 0.1% TEA'water = 0% to 20%) to give Ε5·ΤΕΑ as a white solid (5.2 mg, 26%
Figure imgf000172_0001
171 Step l : C5
Figure imgf000173_0001
[0323] To a mixture of A4 (500 mg, 0.50 mmol, co-evaporated with MeCN 5 mL χ 1 and toluene 10 mL 2) and Gl (322 mg, 0.61 mmol, co-evaporated with MeCN 5 mL χ 1 and toluene 10 mL χ 2) is added tetrazole (0.45 M in MeCN, 4.0 mL). After stirring at 25 °C for 2 hours, DDTT (240 mg, 1.2 mmol) is added and the mixture is stirred for 16 hour before filtered and concentrated. The residue is then dissolved in DCM (10 mL) followed by addition of water (0.1 mL) and DC A (0.21 mL). After stirring for 10 minutes, TEA (1 mL) is added and the mixture is concentrate and purified by reverse-phase silica ge! column chromatography (MeCN with 0.1% TEA/water = 0% to 100%) to give C5»TEA as a white solid (250 mg, 35% yield). (MS: {[M+2H]2+)/2 574.6)
Step 2: phosphorothioate 85
Figure imgf000173_0002
[0324] To a solution of CS (600 mg, 0.423 mmol, co-evaporated with Py 3 mL 2) in Py (5.0 mL) is added DMOCP (313 mg, 1.69 mmol) at 25 °C After stirring for 2 hours, 3H- l,2~benzodithiol-3-one (142 mg, 0.85 mmol) is added and the mixture is stirred for 2 hours before sodium bicarbonate aqueous solution (5%, 10 mL) is added. The mixture is then extracted with EA (10 mL χ 3). The combined organic layers are dried with anhydrous sodium sulfate, filtered, concentrated, and purified by reverse-phase HPLC (MeCN with 0.1% TEA/water = 0 to 100%) to give four diastereomers of 85·ΤΕΑ as white solids. Isomer 1 (28 mg) (MS: [M+H]+ 1160.9), Isomer 2 (25 mg) (MS; ] M 1 11 1160.9); somer 3 (50 mg) (MS: I M · I ! Γ 1 160.9); Isomer 4 (52 mg) (MS: i V! ! j ' 1160.9)
Step 3: E1S-E18
Figure imgf000174_0001
[0325] Each of the isomers of 85·ΤΕΑ (25 mg, 0.022 mmol) in ammonium hydroxide (5.6 mL) and MeOH (4.0 mL) is stirred at 50 °C for 16 hours. The mixture is then purged with nitrogen at room temperature for 5 rainutes before concentrated. The residue is dissolved in TEA (0.5 mL) and Py (0.2 mL), and TEA-3HF (0.7 mL) is added. After stirring at 50 °C for 24 hours, triethylammonium bicarbonate aqueous solution (IM, 5 mL) is added and the mixture is purified by a reverse-phase silica gel column chromatorgraphy (MeCN with 0.1% TEA/water - 0% to 30%) to give E15-E18 as white solids.
Figure imgf000175_0001
174 Step 1 : phosphoramidate 44
Figure imgf000176_0001
[0326] To a solution of C3 (16 mg, 0.0.14 mmol, co-evaporation with P 1 mL χ 3) in Py (0.5 mL) is added DMOCP (10.4 mg, 0,056 mmol). After stirring for 15 mmutes, N!S (4.1 mg, 0,0183 mmol) and morpholine (0.012 mL, 0.141 mmol) are added and the mixture is stirred for 1 hour before sodium bisulfite aqueous solution (0.14%, 1 mL) and sodium bicarbonate (80 mg) is added. The mixture is then extracted with DCM (5 mL χ 3), dried over anhydrous sodium sulfate, and concentrated. The residue is then stirred in MeCN (0.5 mL) and f-butylamine (0.5 mL) at room temperature for 15 minutes before concenirated. The resulting residue is then co-evaporated with MeCN (1 mL χ 3) and purified by HPLC to give 86 as a white solid (2.4 mg, 15%). (MS: [M+H]'1" 1046.2)
Step 2: E24
Figure imgf000177_0001
[0327] To 86 (2,4 mg, 0.0021 mmol) is added methylamine (33% in EtOH, 0.3 mL). After stirring at room temperature for 16 hours, the mixture is concentration and the residue is stirred in a mixture of TEA and TEA«3HF in THF (0,036 mL/0.018 mL/0,3 mL) at 35 °C for 18 hours. MeCN (1.0 mL) is then added and the solid is collected by centnfugation, washed with MeCN (1 mL χ 2) to give E24 as a white solid (0.6 mg, 38% yield). (MS: |M+Hj+ 744.0)
Figure imgf000178_0001
177 Step 1 : b ranop ospi iiate 87
Figure imgf000179_0001
[0328] To a solution of C3 (100 mg, 0.088 mmol, co-evaporated with Py 4 mL >< 3) in Py (3 mL) is added DMOCP (57 mg, 0.337 mmol). After stirring for 15 minutes, BSTFA (0.10 mL, 0.371 mmol) is added dropwise and the mixture is stirred for 20 minutes before borane N,N-diisopropylethylamine complex (0.092 mL, 0.530 mmol) is added. The mixtue is then stirred for 3 hours before concentrated and pundied by silica gel column
chromatography (MeOH/DCM = 1/19 to 1/9) to give semi-pure CE-protected 87 as a yellow solid. The semi-pure CE-protected 87 obtained above is stirred in a mixture of MeCN (1 mL) and f-butylamine (0.5 mL) for 10 minutes before concentrated. The residue is then co- evaporated with MeCN (4 mL 3) and purified by reverse-phase HPLC (MeCN with 0.1% TEA/water = 40% to 90%) to give 87 as a white solid (11 mg, 12% over two steps). (MS: [M] 1073.2)
Step 2: E2S
Figure imgf000180_0001
[0329] To 87 (5.7 mg, 0.0053 mmol) is added methylarnine (33% in EtOH, 1 mL).
After stirring at room temperature for 18 hours, the mixture is concentration and the residue is stirred in a mixture of TEA (0.08 mL) and TEA-3HF (0.04 mL.) in THF (0.5 mL.) at 35 °C for 18 hours. MeCN (1.2 mL) is then added and the solid is collected by centrifugation, purified by reverse-phase HPLC (MeCN with 0.1% TF A/water = 0% to 20%) to give E25 as a white solid (2.5 mg, 61% yield) (MS: [M]~ 671.2)
Figure imgf000181_0001
180 Step l. CBl
Figure imgf000182_0001
[0330] To a solution of GB3 (160 nig, 0.25 mniol, co-evaporated with MeCN 1 mL χ 3) and pyridinium trifluoroacetate (35 mg, 0.39 mmol, co-evaporation with MeCN 1 mL 3) in MeCN (1 mL) treated with 3 A MS (500 mg) for 30 minutes is added a solution of A.4 (355 mg, 0.36 mmol, co-evaporated with MeCN 1 mL χ 3) in MeCN (1 mL) treated with 3 A MS (700 mg) for 30 minutes. After stirring at room temperature for 2 hours, TBHP (5.5 M in decane, 0.164 mL, 0.9 mmol) is added and the mixture is stirred for 30 minutes before sodium bisulfite aqueous solution (33%, 0.15 mL) is added at 0 °C. The mixture is then concentrated and the residue is dissolved m DCM (4.8 mL) followed by addition of water (0.054 mL) and dichloroacetic acid (6% in methylene chloride, 4.8 mL). After stirring for 10 min, the Py (1.5 mL) is added and the mixture is concentrated and purified by silica gel column chromatography (MeOH/DCM = 1/10 to 1/4 with 1% Py) to give CBl»Py as a white solid (213 mg, 66% yield).
Step 2: phosphorothioate 88
Figure imgf000183_0001
[0331| To a solution of CB1 (60 mg, 53 μιηοΐ) in Py (1 mL) is added DMOCP (30 mg, 162.5 umol). After stirring for 10 minutes, water (0.027 mL) and 3H-l,2-benzodithiol-3- one (13 mg, 0.077 mmol) are added. The mixture is stirred for 5 minutes before pouring into a solution of sodium bicarbonate (210 mg) in water (7.5 mL). After stirring for 5 minutes, the mixture is extracted by EA/diethyl ether (1: 1, 10 mL- χ 3). The combined organic layers are concentrated to give a yellow solid (100 mg). To a solution of the yellow solid obtained above in MeCN (0,5 mL) is added ie/ -butylarrsine (0.5 mL). After stirring for 10 minutes, the mixture is concentrated and purified by HPLC (MeCN/water with 0.1% TFA: 50% to 100%) to give two diastereomers of 88. Isomer 1 (7 mg) ( M+H]+ 977.0); Isomer 2 (16 mg) (MS: [M+H]+ 977.0)
Step 3: EB1 and EB2
Figure imgf000183_0002
[0332] To the isomer 1 of 88 (7 mg) is added methyl amine (33% in ethanoi, 1 mL). After stirring at room temperature for 12 hours, the mixture is concentrated and the residue is dissolved in TFA aqueous solution (3% v/v, 1 mL). After stirring for 2 hours, the mixture is concentrated and purified by HPLC (MeCN/water with 0.1% TFA, 0% to 45%) to give EB1 as a white solid (2.5 mg, 57% yield), (MS: j \i H i 689.0)
[0333] To the Isomer 2 of 63 (16 mg) is added methylamine (33% in ethanoi, 2 mL) at 0 °C. After stirring at room temperature for 12 hours, the mixture is concentrated and the residue is co-evaporated with a mixture of Py/TEA (5 iriL/2 mL χ 3) before dissolved in Py (0.04 mL). TEA (0.25 mL) and TEA«3HF (0.15 mL are then added. After stirring at 55 °C for 3 hours, acetone (2 mL) is added. The solid is collected (10 mg) by filtration and purified by HPLC (MeCN/water with 0.1% TFA, 0% to 30%) to give EB2 as a white solid (5 mg, 45% yield). (MS: [M+H]+ 689.0, j V! ! i | 687.0)
Example F: Synthesis of EOS
Figure imgf000185_0001
[0334] To a solution of ΕΙ·ΤΕΑ salt (10 mg, 0.0114 mmol) in water (0.3 tnL) is added 2-chloroacetaldehyde (0.015 mL. 0.118 mmol) and sodium hydroxide aqueous solution (1 M, 0.012 mL, 0.012 mmol). After stirring at 37 °C for 18 hours, the mixture is concentrated and purified by reverse-phase HPLC (MeCN/waier with 0.1% TFA = 0% to 30%) to give F.C'26 as a white solid. (MS: j M i 697.1)
[033SJ The following compounds are prepared essentially by the methods above.
Figure imgf000186_0001
185
Figure imgf000187_0001
186
Figure imgf000188_0001
187
Figure imgf000189_0001
188
Figure imgf000190_0001
189
Figure imgf000191_0001
190
Figure imgf000192_0001
191
Figure imgf000193_0001
192
Figure imgf000194_0001
193
Figure imgf000195_0001
194
Figure imgf000196_0001
195
Figure imgf000197_0001
196
Figure imgf000198_0001
197
Figure imgf000199_0001
198
Figure imgf000200_0001
199
Figure imgf000201_0001
200
Figure imgf000202_0001
201
Figure imgf000203_0001
202
Figure imgf000204_0001
203
Figure imgf000205_0001
204
Figure imgf000206_0001
Figure imgf000207_0001
206
Figure imgf000208_0001
207
Figure imgf000209_0001
208
Figure imgf000210_0001
209
Figure imgf000211_0001
210
Figure imgf000212_0001
211
Figure imgf000213_0001
212
Figure imgf000214_0001
213
Figure imgf000215_0001
214
Figure imgf000216_0001
215
Figure imgf000217_0001
216
Figure imgf000218_0001
217
Figure imgf000219_0001
218 [0336] Selected physical data of the example compounds are summaried below.
Figure imgf000221_0001
220
Figure imgf000222_0001
221
Figure imgf000223_0001
222
Figure imgf000224_0001
223
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
228
Figure imgf000230_0001
229
Figure imgf000231_0001
230
Figure imgf000232_0001
231
Figure imgf000233_0001
232
Figure imgf000234_0001
233
Figure imgf000235_0001
234
Figure imgf000236_0001
Figure imgf000237_0001
236
Figure imgf000238_0001
Figure imgf000240_0001
239
Figure imgf000241_0001
240
Figure imgf000242_0001
241
Figure imgf000243_0001
242
Figure imgf000244_0001
243
Figure imgf000245_0001
244
Figure imgf000246_0001
245
Figure imgf000247_0001
246
Figure imgf000248_0001
Figure imgf000249_0001
248
Figure imgf000250_0001
249
Figure imgf000251_0001
250
Figure imgf000252_0001
251
Figure imgf000253_0001
252
Figure imgf000254_0001
Figure imgf000255_0001
254
Figure imgf000256_0001
255 [0337] Stereochemical information of represented compounds is give s below. Table 5, -Cosifi ur fion of Examples E23 arsd EB3
Figure imgf000257_0001
BIOLOGICAL TESTING
[0338] Sena! dilutions of cGAMP analog compounds in phosphate buffer saline (PBS) are mixed with THP l luciferase reporter cells in a 96-weli plate at 0.2x lOb/well3 in the presence or absence of 1 nM of Perfrmgolysm 0 (PFO), which can facilitate compound uptake by forming open channels on the plasma membrane. 16 hours later, 15 μΙ_, of the media from each well is transferred to a new plate, and luminescence is measured. Fold increase in luminescence compared to PBS stimulated cells is plotted against logarithm of concentrations of each compound, and ECso is calculated using Graphpad.
Table 6„ Activity of cyclic dinucleotides and analogs THPI -iSG-Liic THP-!SG-Liic THPI-iSG-Luc THP-!SG-Li!C
Examp!e with PFO, without PFO, Examp!e with PFO, without PFO,
ECso, nM ECso, μΜ ECr,o, nM ECso, Μ
E1 A A EA10 A A
E2 A A EA11 A A
E3 A A EB1 C B
E4 A A EB2 A A
E5 C C EB3 A A
E6 A A EB4 C B
E7 A A EB5 B C
E8 B C EB6 C C
E9 A A EB7 C c
£10 C C EC1 A A
El l B C EC2 B B
E12 A A EI03 B A
E13 B C EC4 C C
£14 B C ECS B B
E15 C C ECS C C
£16 A A EC7 C C
E17 C C ECS C C
E18 A A EC9 C c
E19 C C EC10 C c
E20 B A EC11 A c
E21 B A EC12 A A
£22 B A EC13 B A
E23 A A EC14 C C
£24 B A EC15 B A
E25 A A EC16 C C
EA1 B C EC17 C C
EA2 A A EC18 C C
EA3 C C EC19 C c
EA4 B A EC20 c c
EA5 C C EC21 c c
EAS c C E C .12 c c
EA7 B A EC23 c c
EA8 B A EC24 A A
EA9 A A EC26 B c
A <100 nM A < 30 μΜ A 100 nM A < 30 μΜ
Activity B 100-1000 Activity
B 30-100 μΜ B 100-1000 nM B 30-100 μΜ Code n Code
O1000 nM O100 \M C >1000 nM C >100 μΜ [0339] 2'3'-cGAMP can be degraded by the enzyme ecto-nucleotide
pyrophosphatase/phosphodiesterase (ENPP1) which is present in fetal bovine serum (FBS) (Li et al., 2015, Nat Chern Biol, 1 1 , 235), To test if cGAMP analogues have improved stability, 5 uL of of synthetic cGAMP analogues (100 μΜ stock) were incubated with 45 uL of FBS in a final volume of 50 uL at 37 °C for 1, 2 and 4 hours. At the indicated time, 10 uL of the reaction mixture was taken out and mixed with 10 ,uL phosphate buffered saline (PBS), then heated at 95 °C to denature proteins, which were removed by centrifugation at 13000 g for 5 minutes. The supernatants were delivered to THPl-ISG-luciferase cell line in the presence of PFO to measui'e the activity of remaining cGAMP analogues, as described above. Category A indicates less than 10% decrease of activity after 4-hour incubation, B indicates 10-75% decrease of activity after 4-hour incubation, and C indicates more than 75% loss of activity after 4-hour incubation.
Table 7. Stability of cyclic dinucleotides and analogues in fetal bovine serum
Figure imgf000259_0001
Figure imgf000259_0002
Figure imgf000259_0003
Figure imgf000259_0004
C >75% C >75%
[0340] The following series of prophetic examples are also compounds of the present invention:
Figure imgf000261_0001
260
Figure imgf000262_0001
261
Figure imgf000263_0001
262
Figure imgf000264_0001
263
Figure imgf000265_0001
264
Figure imgf000266_0001
265
Figure imgf000267_0001
266
Figure imgf000268_0001
267

Claims

CLAIMS:
1 , A compound of Formula la or a pharmaceutically acceptable salt thereof,
Figure imgf000269_0001
Formula la wherein: a and b are independently 0 or 1 and a + b = 1, when a is 1, b is 0 and R5 is not present; and when a is 0, b is 1 and R4 is not present;
X* and X2 are independently 0, S or Se in a five-membered ring;
L1, starting from the carbon alpha to X1, and L2, starting from the carbon alpha to X , are independently -CH20-P(0)R6-0-,-CH20-P(S)R6-0-, -C(Y1)(Y2)0-P(0)R6-C(Y )(Y4)-, - CH2NHSO2NH-, -CH2NHC(0)NH-, -CH2NHC(S)NH-5 -CH2NHC(NH)NH-? - (Ή-\ί ΙΠί)}ί i -C H AHSiK I K -( I H I K'l O i'M I-. -O W ί SO XI i~. ~CH2NH 3,4-
di oxocy clobuien- 1 ,2-diy 1)NH-,
Figure imgf000269_0002
c is 0, 1. or 2; d, e, and f are independently 0 or 1 ;
Y1, Y2, Y3, and Y4 are independently H or F;
R6 is hydroxy!, thiol, Ci-eaikyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyioxyl, Ci-6alkoxy, Ci- ehydroxyalkoxy, amino, Ci-salkylamino, di(Ci-6alkyl)aniino, or azido groups, Ci- ealkoxy, Cj-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyioxyl, Ci-bhydroxyalkoxy, amino, Ci- ealky amiRO, di(Ci-6alkyl)ammo, or azido groups, C3-salkenyl-0-, Ci-salkynyl-O-, oligo(ethylene glycol), poly(ethyiene glycol), borano (-BH3"), or -NR7R8; 7 and R8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively iunctionalized with one or more halogen, thiol, hvdroxyi, carbonyl, caiboxy 1, carbonyloxyl, Ci -ealkoxy, Ci-6hydroxyalkoxy, amino, Ci-ealkylaniino, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Ct-ealkyl)- selectively iunctionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cj.-6alkoxy, C.^hydroxyalkoxy, amino, O- ealkylamino, or di(Ci-6alk}d)amino groups, cyclic -(Cwoxaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively iunctionalized with one or more halogen, thiol, hydroxy], carbonyl, caiboxy], carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- 6alkylamino, or di(Ci-6alkyl)animo groups;
R1 and l 2 are independently aromatic rings or heteroaromatic rings with the following general structure including its tautomeric forms:
Figure imgf000270_0001
g and h are independently 0 or 1;
W1 and W2 are independently hydrogen, halogen, hvdroxyi, Ci-ealk l, Ci- ealkyl selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci-ealkylamino, di(Ci- 6aikyl)amino, or azido groups, Ci-ealkoxy, Cwalkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci- &hydroxyalkoxy, amino, Ci-6alkylammo, di(Ci-6alkyi)amino, or azido groups, C:,- saikenyl-O-, Cs-salkynyl-O-, oligo(ethylene glycol), polyethylene glycol), azido, or - \ R"RS:
Z Z2, Z3, Z4, Z5, and Z6 are independently CH or ; if present, Z ', Z8, Z9, Z10, and Z11 are independently CH or N, and then W1 is CH or N: RA R4, and R5 are independently hydrogen, halogen, hydroxyl, amino, Ci-ealk l, Ci- 6alkyi selectively function ah zed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonvloxyl, Ci-6aIkoxy, Ci-ehyciroxyalkoxy, amino,
Figure imgf000271_0001
di(Ci- 6alkyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonvloxyl, Ci-ehydroxyalkoxy, amino, C.-ealkyl amino, di(Ci.6alkyl)amino, or azido groups, Cj-salkenyl-O-, C3-5alkynyl-0-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7R8; and the compound is not 2'3'-cGAMP.
2, The compound of claim 1 wherein the compound of Formula lb, or a pharmaceutically acceptable salt thereof,
Figure imgf000271_0002
Formula lb wherein:
X"! and X2 are independently 0, S or Se;
Z12, Z13, Z14, Z15, Z36, and Z17 are independently CH or N;
L1, starting from the carbon alpha to X1, and U, starting from the carbon alpha to X2, are independently -CH20-P(0)R6-0-,-CH20-P(S)R6-0-. -C(Y1)(Y2)0-P(0)R6-C(Y3)(Y4)-, - CH2NHSO2NH-, -CH2NHC(0)NH-, -CH_NHC(S)NH-, -CH2NHC(NH)NH-, - CH2NHC 0)CH2-, -CH2NHSO2CH2-, -CH2CH2C(0)NH-, -CH2CH2SO2NH-, -CI !-M 1(3.-1-
Figure imgf000271_0003
c i i), L or 2; d, e, and f are independently 0 or 1 ;
Y3, Y% Y3, and Y4 are independently H or F;
R" is hydroxy 1, thiol, Ci-ealkyl, Ci ealkyl selectively functionalized with one or more halogen, thiol, hvdroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci- ehydroxyalkoxy, amino,
Figure imgf000272_0001
di(Ci-r,alkyi)amino, or azido groups, Ci- 6alkoxy, Ci-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-6hydroxyalkoxy, amino, Ci- 6alkylamino, di(Ci-6alkyl)amino, or azido groups, Cs-salkenyl-O-, Cs-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), borano (-BH3"), or -NR7R8;
R7 and R8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hvdroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-ohydroxyalkoxy, amino,
Figure imgf000272_0002
di(Ci- 6a3kyl)amino, or azido groups, cyclic -(d^alkyl)-, cyclic -(Ci-ealkyl)- selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl carboxyl, carbonyloxyl, C]-6alkoxy. Ci-fthydroxyalkoxy, amino, Ci- ea!kylamino, or di(Ci-6alkyl)amino groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-6hydroxyalkoxy, amino, Ci- 6alkylamino, or di(Ci-6alkyl)aniino groups;
R3 and R4 are independently hydrogen, halogen, hydroxyl, ammo, Ci-6alkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cwalkoxy, Ci-ehydroxyaikoxy, amino, Ci-ealkylarnino, di(Ci-6alkyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl. carbonyloxyl, Ci-6hydroxyalkoxy, amino, Ci- 6alkylamino, di(Ci-6alkyl)amino, or azido groups, Cs-salkenyl-O-, C3-5alkynyl-0-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7R8; and the compound is not a previously disclosed compound, including 2'3'-cGAMP.
3. The compound of claim 1, wherein a compound of Formula Ic, or a pharmaceutically acceptable salt thereof, Formula Ic
Figure imgf000273_0001
Z12, Z , Z14, /■"'. Z16, and Z17 are independently CH or N;
R3 and R4 are independently hydrogen, halogen, hydroxyl, ammo, Ci-6alkyl, C i -ealkyl selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxy], Cwalkoxy, Ci-ehydroxyalkoxy, amino, Ci^alkylamino, di(Ci-6alkyl)amino, or azido groups, Ci-ealkoxy, Ci-^alkoxy selectively funciionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl carbonyloxyl, Cwhydroxyalkoxy, amino, Ci- 6alkylamino, di(Ci-6ailiyi)amino, or azido groups, C saikenyl-O, Cs-salkynyl-O-, oligo(ethyiene glycol), polyiethylene glycol), azido, or -NR7RS;
R7 and R8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-6hydroxyalkoxy, amino, Ci-6alkylamino, di(Ci- 6aikyl)amino, or azido groups, cyclic -(Ci-ealkyi)-, cyclic -(Ci-eaikyl)- selectively funciionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, C whydroxy alkoxy, amino, Ci-6alkyiammo, or di(Ci- 6aIkyl)amino groups, cyclic -(Ci-60xaalkyl)-, or cyclic -(Ci-60xaalkyl)- selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-bhydroxyalkoxy, amino, Ci-eaikyiarmno, or di(Ci-6aik}'1)amino groups;
R9 and R!0 are independently hydroxyl, thiol, Ci-ealkyl, Ci-ealkyl selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-6alkoxy, Ci-bhydroxyalkoxy, amino, Ci-6aIkyiamino, di(Ci-6alkyl)ainino, or azido groups, G-ealkoxy, Ci-6alkoxy selectively iunctionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl,
Figure imgf000274_0001
amino, Ci-ealkylamino, di(Cr- 6alkyi)ammo, or azido groups, Cs-salkenyl-O-, Ci-salkynyl-O-, oligo(ethyleRe glycol), poly(ethylene glycol), borano {-RH . ). or -NR7RS; the oxygen atom in one or both of the tetrahydrofuranyl rings of Formula Ic can be optionally replaced by a sulfur or a selenium atom; and the compound is not 2'3'-cGAMP.
4, The compound of claim 1, wherein a compound of Formula id, or a pharmaceutically acceptable salt thereof
Figure imgf000274_0002
Formula Id wherein:
X1 and X2 are independently 0, S or Se;
L!, starting from the carbon alpha to X1, and L2, starting from the carbon alpha to X2, are independently -CH20-P(0)R6-0-,-CH20-P(S)R6-0-, -C(Y1)(Y2)0-P(0)R6-C(Y3)(Y4)-, - C! I M iSOM i-. -( l b\! l( i )NH-. ~CH2NHC(S)NH~, -CH2NHC(NH)NH-, - CH2NHC(0)CH2-, (l l 'M ISO'CH '-. -CH2CH2C 0)NH-, -Π 1 H bSi) Xi k -CH2NH(3,4-
dioxocy clobuten- 1 ,2-diyl)NH-,
Figure imgf000274_0003
c is 0, 1, or 2; d, e, and f are independently 0 or 1 ;
Y1, Y2, Y3, and Y4 are independently H or F; R6 is hydroxy!, thiol, Ci-ealk l, Ci-ealkyl selectively functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl Ci-ealkoxy, Ci- ehydroxyalkoxy, amino, Ci-ealkylarnino, di(Cwalk>'l)amino, or azido groups, Ci- ealkoxy, Cwalkoxy selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- ealkw!amino, di(C :-6alkyl)amino, or azido groups, Cs-salkenyl-O-, C3.5alkyn.yl-O-, oligo(ethylene glycol), poly(ethylene glycol), borano (-BH3"), or -NR7R8;
R? and R8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Cwhydroxyalkoxy, amino, Ci-ealkylamino, di(G- 6alkyl)amino, or azido groups, cyclic -(Ci-salkyi)-, cyclic -(Ci-ealkyl)- seiective!y functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-e.hydroxyalkoxy, amino, Ci- ea!kylammo, or di(Ci-6alkyl)amino groups, cyclic -(Ci-60xaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionahzed with one or more halogen, thio!, hydroxy], carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxya!koxy, amino, Ci- 6alkylarnino, or di(Ci-6alkyl)amino groups;
R3 and R4 are independently hydrogen, halogen, hydroxyl, amino, Cwalkyl, C 3 -ealkyl selectively functionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cwalkoxy, Ci-ehydroxyalkoxy, amino, Ci-6aikyiamino, di(Ci-6a3kyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionahzed with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cwhydroxyalkoxy, amino, Ci- 6alkylamino, di(Ci-6aiky!)amino, or azido groups, C saikenyl-Q-, Cs-salkynyl-O-, oligo(ethy!ene glycol), poly(ethy!ene glycol), azido, or -NR7R8;
R11 and R12 are independently selected from the group consisting of:
Figure imgf000276_0001
212 ZH Z15 Z16 Z17 Z18 Z19 Z20 Z2i Z22 Z23 Z24 Z25 Z26 Z27 Z28 Z29, Z30, Z31, Z32, Z33, Z34, Z35, Z36, and Z37 are each independently CH or N; and
W3, W4, W5, W6, W7, Ws, and W9 are independently hydrogen, halogen, hydroxyl, Ci-ealkyl, Cwalkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-6alkoxy, Ci-ehydroxyalkoxy, amino, Ci-ealkylamino, di(Ci-6alkyl)amino, or azido groups, Cwalkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, C why droxyalkoxy, amino, Ci-ealkylamino, di(Ci- 6aikyl)amino, or azido groups, Cs-salkenyl-O-, C - alk> m ! ·()·. oligoiethyiene glycol), poly(ethylene glycol), azido, or -\R~R\ 5, The compound of claim 1, wherein a compound of Formula ie, or a pharmaceutically acceptable salt thereof,
Figure imgf000277_0001
wherein:
X3, X4, X5, and X6 are independently 0, NH, CFfc, CHF, or CF; ;
R33 and R34 are independently selected from the group consisting of:
Figure imgf000277_0002
7.} ZP, Zu, 7) Z16, 7) Zm Z39, Z20, 7.} TP; 7. , 7? 7.P, Z26, ZP, and Z2i are each independently CH or N; and
W3, W4, W\ W6, and W7 are independently hydrogen, halogen, hydroxyl, Ct~ salkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, d-ealkoxy, Ci-ehydroxyalkoxy, amino, Ci- eaikylarnino, di(Ci-6alkyl)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-6hydroxyalkoxy, amino, Ci-eaikylamino, di(Ci-6alkyl)amino, or azido groups, Cs-salkenyl-O-, C salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7R8; 7 and R8 are independently hydrogen, Ci-ealkyl, Ci-eaikyl selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl, carbonyloxyl, d-ealkoxy, Ci-fthydroxyalkoxy, amino, d-ealkyl amino, di(Ci- 6alkyl)amino, or azido groups, cy clic -(Ci-ealkyl)-, cyclic -(Ci-ealkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl carbonyloxyl, Cj.-6alkoxy, d-ehydroxyalkoxy, amino, d- ealkylamino, or di(Ci-6alk>d)amino groups, cyclic -(Cwoxaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonyloxyl, d-ehydroxyalkoxy, amino, Ci- 6alkylamino, or di(d-6alkyl)amino groups;
R3 and R4 are independently hydrogen, halogen, hydroxyl, amino, d^alkyl, C 3 -ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ealkoxy, d-ehydroxyalkoxy, amino, d^alkylamino, di(d-6alkyl)amino, or azido groups, Ci-6alkoxy, Ci-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl carbonyloxyl, d-ohydroxyaikoxy, amino, Ci- ealkyiammo, di(d-6alkyl)amino, or azido groups, d-salkenyl-O-, d-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7R8;
R15 and R16 are independently hydroxyl, thiol, methoxy, ethoxy, amino, N- methylamino, AyV-dimethylamino, N-ethylamino, N,N-diethylammo, N-morpholino, or borano (~Bi h }; the oxygen atom in one or both of the tetrahydrofuranyl rings of Formula le can be optionally replaced by a sulfur or a selenium atom; and the compound is not 2'3'-cGAMP.
6, The compound of claim 1, wherein a compound of Formula If, or a pharmaceutically acceptable salt thereof,
Figure imgf000279_0001
Formula If wherein:
X1 and X2 are independently 0, S or Se;
Z12, Z!3, Z14, Z15, Z16, and Z17 are independently CH or N;
L1, starting from the carbon alpha to X1, and L2, starting from the carbon alpha to X2, are independently -CFi20-P(0)R6~0~ CH20-P(S)R6~Ck -C(Y])(Y2)0-P(0)R6-C(Y )(Y4)-, - CH2NHSO2NH-, -CH2NHC(0)NH-, -CH2NHC(S)NH-, -CH2NHC (NH)NH-, - CH2NHC(0)CH2-, -CH2 HSO2CH2- -CH2CH2C(0)NH-, -CH2CH2SO2NH-, -CH?.NH(3,4-
dioxocy clobuten- 1 ,2-diy 1)NH-,
Figure imgf000279_0002
c is 0, 1 , or 2; d, e, and f are independently 0 or 1;
Y3, Y , Y3, and Y4 are independently H or F;
R:' is hydroxyi, thiol, d-6alkyl, (Ί i-al'ky! selectively functi onalized with one or more halogen, thiol, hydroxyi, carbonyl, carboxyl, carbonyioxyl, Ci-6alkoxy, Ci- bh droxyalkoxy, amino, Ci-salkylaimno, di(d-6alkyl)arnino, or azido groups, Ci- ealkoxy, d-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxyi, carbonyl, carboxyl, carbonyioxyl, d-fthydroxyalkoxy, amino, d- eaikylamino, di(Ci-6alkyl)amino, or azido groups, d-salkenyl-O-, d-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), borano (-Bilr), or -NR7RS; 7 and R8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl, carbonyloxyl, C i-ealkoxy, Ci-fthydroxyalkoxy, amino, d-ealkyl amino, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic -(Ci-ealkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl carboxyl carbonyloxyl, d.-6alkoxy, d-ehydroxyalkoxy, amino, d- ealkylamino, or di(Ci-6alkyd)amino groups, cyclic -(Cwoxaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxy], carbonyl, carboxyl, carbonyloxyl, d-ehydroxyalkoxy, amino, Ci- 6alkylamino, or di(Ci-6alkyl)animo groups;
R3 and R5 are independently hydrogen, halogen, hydroxyl, amino, Cwalkyl, C 3 -ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl Cwalkoxy, d-ehydroxyalkoxy, amino, d^alkylamino, di(d-6alkyl)amino, or azido groups, Ci-6alkoxy, Ci-6alkoxy selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl carbonyloxyl, d-ohydroxyaikoxy, amino, Ci- ealkylammo, di(d-6alkyl)amino, or azido groups, d-salkenyl-O-, d-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7R8; and the compound is not 2'2'-cGAMP.
7. The compound of claim 1, wherein a compound of Formula Ig, or a pharmaceutically acceptable salt thereof,
Figure imgf000280_0001
Formula Ig wherein:
Zn, ZB, Z14, 7.)K Z16, find Zi7 are independently CH or N;
R5 and R5 are independently hydrogen, halogen, hydroxyl, amino, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxvl, carbonyloxyl, Ci-eaikoxy, Ci-ehydroxyalkoxy, amino, Cwalkylamino, di(C walky l)amino, or azido groups, Ci-ealkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxvl carbonyloxyl Ci-shydroxyalkoxy, amino, Ci- 6alkylamino, di(Cj-6alkyl)amino, or azido groups, Ci-salkenyl-O-, Cs-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7R8;
R7 and Rs are independently hydrogen, d-eal l, Ci-6alkyi selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxvl, carbonyloxyl, Cwalkoxy, C whydroxy alkoxy, amino, Cwalkylamino, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Ci-ea!kyf)-, cyclic -(Ci-ealkyl)- selectively functionalized with one or more halogen, thiol hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cj-ealkoxy, C J -(-hydroxy alkoxy, amino, Ci-ealkylamino, or di(G- 6alkyl)amino groups, cyclic -(Cwoxaalkyl)-, or cyclic -(Cwoxaalkyl)- selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cwhydrox alkoxy, amino,
Figure imgf000281_0001
or di(Ci-6alkyl)amino groups;
R9 and R1C are independently hydroxyl, thiol, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-ehydroxyalkoxy, amino,
Figure imgf000281_0002
di(Ci-6alky!)amino, or azido groups, Ci-6alkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Cwhydroxy alkoxy, amino, Ci-ealkylamino, di(Ci- 6a!kyl)amino, or azido groups, Cs-salkenyl-O-, Cs-salkynyl-O-, o!igoiethy!ene glycol), poly(ethylene glycol), borano i-BR ; ). or -NR7RS; the oxygen atom in one or both of the tetrahydrofuranyl rings of Formula Ig can be optionally replaced by a sulfur or a selenium atom; and the compound is not 2'2'-cGAMP.
8. The compound of claim 1, wherein a compound of Formula Ih, or a pharmaceutically acceptable salt thereof,
Figure imgf000282_0001
Formula Ih wherein:
X1 and X2 are independently 0, S or Se;
L1, starting from the carbon alpha to X1, and L2, siartmg from the carbon alpha to X2, are independently -CH20-P(0)R6-0-,-C¾0-P(S)R6-0-, -C(Y3)(Y2)0-P(0)R6-C(Y3)(Y4)-, - CH2NHSO2NH-, -i l l 'NHCiOiM k -CH2NHC(S)NH-, -CH2NHC ( H)NH-, - CH2NHC(0)CH2-5 -CH2 HSO2CH2- -CH2CH2C(0)NH-, -CH2CH2SO2NH-, -CH2NH(3,4-
dioxocy clobuten- 1 ,2-diyl)NH-,
Figure imgf000282_0002
c is 0, 1, or 2; d, e, and f are independently 0 or 1 ;
Y\ Y2, Y and V 1 are independently H or F;
R6 is hydroxyl thiol, Ci-salkyl, Ci-ealkyl selectively fimctionahzed with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl Ci-6alkoxy, Ci- eh droxyalkoxy, amino, Ci-6alkylarmno, di(Ci-6aIkyi)amino, or azido groups, Ci- 6alkoxy, Cr.6alkoxy selectively functionalized with one or more halogen, thiol, hydroxyl carbonyl, carboxyl, carbonyloxyl, Ci-ohydroxyalkoxy, amino, Ci- ealkylamino, di(Ci-6alkyl)amino, or azido groups, Cs-salkenyl-O-, C3-5alkynyl-0-, oligo(ethylene glycol), poly (ethylene glycol), borano (-BH3"), or -NR.7R.8;
R7 and R8 are independently hydrogen, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cwalkoxy, Ci-&hydroxyalkoxy, amino, Ci-6alkylammo, di(Ci- 6alkyl)amino, or azido groups, cyclic -(Ci-ealkyl)-, cyclic ~(Ci-6alkyl)~ selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl carboxyl, carbonyloxyl, Ci-ealkoxy, Ci-6hydroxyalkoxy, amino, Ci- 6alkylamino, or
Figure imgf000283_0001
groups, cyclic ~(Ci-60xaalkyl)~, or cyclic - (Ci-60xaalkyl)- selectively functional ized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, amino, Ci- ealkyl amino, or di(Ci-6alkyl)amino groups;
R3 and R5 are independently hydrogen, halogen, hydroxvl, amino, Ci-ealkyl, Ci-ealkyl selectively functionalized with one or more halogen, thiol, hvdroxyl, carbonyl, carboxyl, carbonyloxyl, Ci-ea!koxy, Ci-bhydroxyalkoxy, amino, Ci-ealkyf amino, di(Ci-6alkyi)amino, or azido groups, Ci-6alkoxy, Ci-ealkoxy selectively functionalized with one or more halogen, thiol, hydroxvl, carbonyl, carboxyl, carbonyloxyl, Ci-ehydroxyalkoxy, ammo, Ci - 6alkyiamino, di(Ci-6alkyi)amino, or azido groups, (\.--aiken> ! -()·. Cs-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7R8;
R11 and R.'2 are independently selected from the group consisting of
Figure imgf000283_0002
Figure imgf000284_0001
. with at least one of R11 and R12 being
Figure imgf000284_0002
Zi2, Z13, Z14, Z15, 7} Z17, Z1S, Z19, Z20,
Figure imgf000284_0003
z22, z23, z24, z25, z26, z27, z28, Z29, Z30, Z31, Z32, Z33, Z34, Z35, Z36, and Z37 are each independently CH or N; and
W3, W4, W5, W6, W7, Ws, and W9 are independently hydrogen, halogen, hydroxy!,
Figure imgf000284_0004
Ci-eaikyl selectively functionalized with one or more halogen, thiol, hydroxy., carbonyl, carbox l, carbonyloxyl, Cwalkoxy, Ci-eliydroxyalkoxy, amino, Ci-ealkylamino, di(Ci-6alkyl)arnino, or azido groups, Ci-6alkoxy, Ci-eaikoxy selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyl, C l-ehy droxyaJkoxy, amino, Ci-ealkylamino, di(Ci- 6alkyl)amino, or azido groups, C3-5alkenyl-0-, C?-5alkynyl-0-, oligo(ethylene glycol), po!y(ethylene glycol), azido, or -NR7R8.
9, The compound of claim 1, wherein a compound of Formula Ii, or a pharmaceutically acceptable salt thereof,
Figure imgf000284_0005
Formula !i wherein: X3, X5, X6, and X7 are independently 0, NH, CFfc, CHF, or CF2;
R and R14 are independently selected from the roup consisting of:
Figure imgf000285_0001
Zn, y} /} >. 7) X' \ 7) 7 7 Z20, 7? Z22, TP, 7: Z25, Z26, Z27, and Z28 are each independently CH or N; and
W3, W4, W3, W&, and W" are independently hydrogen, halogen, hydroxyl, Ci- eaikyl, Ci-6alkyl selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxvi, Cwalkoxy, C why droxy aikoxy, amino, Ci- 6alkylaniino, di(Ci-6alkyl)aniino, or azido groups, Ci-6alkoxy, Ci-c,alkoxy selectively functionalized with one or more halogen, thiol, hydroxyl, carbonyl, carboxyl, carbonyloxvi, Cwhydroxyalkoxy, amino, Ci-f.alkylamino, di(Ci-6alkyl)amino, or azido groups, d-saikenyl-O, d-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7R8;
R? and R8 are independently hydrogen, Ci-ealkyl, Cwalkyl selectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxvi, C alkoxy, Ci-ebydroxyalkoxy, amino, d-ealkyl amino, di(Ci- 6aikyl)amino, or azido groups, cyclic -(Cwalkyi)-, cyclic -(Cwalkyl)- seiectively functionalized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbon vioyd. Cwalkoxy, Cwhydroxyalkoxy, amino, Ci- ealkylanimo, or di(Cwalkyl)amino groups, cyclic -(Cwoxaalkyl)-, or cyclic - (Ci-60xaalkyl)- selectively functionalized with one or more halogen, thio!, hydroxyl, carbonyl, carboxyl, carbonyloxyl, Cwhydroxyalkoxy, amino, Ci- 6a!ky!amino, or di(d-6aIky!)ammo groups; R3 and R3 are independently hydrogen, halogen, hydroxy 1, amino, Ci-ealkyl, Ci-eall selectively fundi on alized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyi,
Figure imgf000286_0001
Ci -ehydroxyalkoxy, amino, Cwdkylamino, di(Ci-6al3<yl)amino, azido groups, Ci-6alkoxy, Ci-6alkoxy selectively functionaiized with one or more halogen, thiol, hydroxy!, carbonyl, carboxyl, carbonyloxyi, Ci-ehydroxy alkoxy, amino, Ci- 6alkylamino, di(C.-6alkyl)arriino, or azido groups, C3-salkerryl-Q~, Cs-salkynyl-O-, oligo(ethylene glycol), poly(ethylene glycol), azido, or -NR7R8;
Ri 5 and R16 are independently hydroxy!, thiol, methoxy, ethoxy, amino, ΛΓ- methylamino, N,N-dimethylamino, N-ethylamino, AyV-diethylamino, N-morpholino, or borano (-Pil l ; ): the oxygen atom in one or both of the tetrahydrofuranyl rings of Formula li can be optionally replaced by a sulfur or a selenium atom; and the compound is not 2'2'-cGAMP.
A compound selected from the group consisting of:
Figure imgf000286_0002
Figure imgf000287_0001
286
Figure imgf000288_0001
Figure imgf000289_0001
288
Figure imgf000290_0001
289
Figure imgf000291_0001
290
Figure imgf000292_0001
291
Figure imgf000293_0001
292
Figure imgf000294_0001
293
Figure imgf000295_0001
294
Figure imgf000296_0001
295
Figure imgf000297_0001
296
Figure imgf000298_0001
297
Figure imgf000299_0001
Figure imgf000299_0002
298
Figure imgf000300_0001
299
Figure imgf000301_0001
300
Figure imgf000302_0001
301
Figure imgf000303_0001
302
Figure imgf000304_0001
303
Figure imgf000305_0001
304
Figure imgf000306_0001
305
Figure imgf000307_0001
306
Figure imgf000308_0001
307
Figure imgf000309_0001
308
Figure imgf000310_0001
Figure imgf000310_0002
309
Figure imgf000311_0001
Figure imgf000311_0002
310
Figure imgf000312_0001
Figure imgf000312_0002
311
Figure imgf000313_0001
39. A compound according to the formula:
Figure imgf000313_0002
Figure imgf000314_0001
Figure imgf000314_0002
313
Figure imgf000315_0001
Figure imgf000315_0002
314
Figure imgf000316_0001
Figure imgf000316_0002
315
Figure imgf000317_0001
Figure imgf000317_0002
316
Figure imgf000318_0001
A compound according to the formula:
Figure imgf000318_0002
Figure imgf000319_0001
Figure imgf000319_0002
318
Figure imgf000320_0001
Figure imgf000320_0002
319
Figure imgf000321_0001
Figure imgf000321_0002
320
36. A compound according to the formula:
Figure imgf000322_0001
37. A compound according to the formula:
Figure imgf000322_0002
A compound selected from the group consisting of:
Figure imgf000323_0001
322
Figure imgf000324_0001
323
Figure imgf000325_0001
Figure imgf000325_0002
324
Figure imgf000326_0001
39. A compoimd selected from the group consisting of
Figure imgf000327_0001
326
Figure imgf000328_0001
327
Figure imgf000329_0001
328
Figure imgf000330_0001
329
Figure imgf000331_0001
Figure imgf000331_0002
330
Figure imgf000332_0001
Figure imgf000332_0002
331 A compound according to the formula:
Figure imgf000333_0001
43. A compound according to the form la:
Figure imgf000333_0002
A compound according to the formula:
Figure imgf000334_0001
Figure imgf000334_0002
A compound according to the formula:
Figure imgf000335_0001
A compound selected from the group consisting of {¾>)- and (5p)~ boranophosphates of the formula:
Figure imgf000335_0002
48. A compound selected from the group consisting of (!¾>)- and (<Sp)-
boranophosphates of the formul a:
Figure imgf000336_0001
A compound according to the formula:
Figure imgf000336_0002
A compound according to the formula:
Figure imgf000337_0001
51. A compound according to the formula:
Figure imgf000337_0002
Figure imgf000338_0001
Figure imgf000338_0002
337 A compound according to the formula:
Figure imgf000339_0001
55. A pharmaceutical composition comprising a compound of formula L or a pharmaceutically acceptable salt thereof,
56. A pharmaceutical composition wherein the composition is a compound of formula I in a pharmaceutical formulation including a nanoparticle or another delivery vehicle,
57. A pharmaceutical composition wherein the composition is a compound of formula I in combination with at least one further therapeutic agent.
58. A pharmaceutical composition of claim 57 wherein the addi tonal therapeutic agent is an immune checkpoint inhibitor including but is not limited to antibodies against PD- ! . P -l. s or CTLA-4.
59. A pharmaceutical composition of claim 57 wherein the additional therapeutic agent is radiation including stereotactic body radiation therapy (SBRT).
60. A pharmaceutical composition of claim 57 wherein the additional therapeutic agent is an anu-neoplastic agent, including cheniotherapeutic agent and therapeutic antibodies.
61. A pharmaceutical composition wherein the composition is a compound of formula I in a pharmaceutical formulation with an immunogen of a cancer cell (tumor antigen) or of an infectious pathogen.
62. A pharmaceutical composition of claim 57 wherein the composition further comprises one or more of pharmaceutically acceptable carriers, diluents and excipients.
63. A method of inducing or promoting an immune response comprising administering to a patient in need thereof, an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.
64. A method of inducing or promoting an immune response comprising administering to a patient in need thereof, an effective amount of a pharmaceutical composition according to claim 63.
65. A method of inducing or promoting a STING-dependent immune response comprising administering to a patient in need thereof, an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.
66. A method of inducing or promoting a STING-dependent immune response comprising administering to a patient in need thereof, an effective amount of a
pharmaceutical composition according to claim 63.
67. A method of treating cancer comprising administering to a patient in need thereof, a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.
68. A method of treating cancer comprising administering to a patient in need thereof, a therapeutically effective amount of a pharmaceutical composition according to claim 67.
69. A method of treating cancer comprising administering to a patient in need thereof, a therapeutically effective amount of a pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, in combination with immune checkpoint antibodies, including but is not limited to, antibodies against PD-1, PD- LI or CTLA-4.
70. A method of treating cancer comprising administering to a patient in need thereof, a therapeutically effective amount of a pharmaceutical composition comprising a compound of formula I, or a phai'maceuticaily acceptable salt thereof, in combination with radiation including stereotactic body radiation therapy (SBRT).
71. A method of treating cancer comprising administering to a patient in need thereof, a therapeutically effective amount of a pharmaceutical composition comprising a compound of formula Ϊ, or a pharmaceutically acceptable salt thereof, in combination with an anti-neoplastic agent, including chemotherapeutic agent and therapeutic antibodies,
72. A method of treating infectious diseases comprising administering to a patient in need thereof, a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.
73. A method of treating infectious diseases comprising administering to a patient in need thereof, a therapeutically effective amount of a pharmaceutical composition according to claims 72,
74. A method of treating immune disorders comprising administering to a patient in need thereof, a therapeuticaliy effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.
75. A method of treating immune disorders comprising administering to a patient in need thereof, a therapeutically effective amount of a pharmaceutical composition according to claims 74.
76. A vaccine composition comprising an antigen or antigen composition and a compound of Formula i, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a disease,
77. The use of a compound of Formula I. or a pharmaceutically acceptable salt thereof, for the manufacture of a vaccine composition comprising an antigen or antigen composition for the treatment or prevention of a disease,
78. A method of treating or preventing a disease comprising the administration to a patient suffering from or susceptible to the disease, a vaccine composition comprising an antigen or antigen composition and a compound of Formula I, or a pharmaceutically acceptable salt thereof.
79. A method of treating or preventing a disease comprising administering to a patient in need thereof, a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof, wherein the patient is a mammal.
80. A method of treating or preventing a disease comprising administering to a patient in need thereof, a therapeutically effective amount of a compound of formula I. or a pharmaceutically acceptable salt thereof, wherein the patient is a human.
PCT/US2017/023093 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use WO2017161349A1 (en)

Priority Applications (24)

Application Number Priority Date Filing Date Title
BR112018068748-0A BR112018068748B1 (en) 2016-03-18 2017-03-17 CYCLIC DINUCLEOTIDE COMPOUND, PHARMACEUTICAL COMPOSITION, AND, USE OF A COMPOUND
ES17767683T ES2929628T3 (en) 2016-03-18 2017-03-17 Cyclic Dinucleotide Compounds and Methods of Use
IL280430A IL280430B2 (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
LTEPPCT/US2017/023093T LT3429596T (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
CN202210558244.6A CN114751950A (en) 2016-03-18 2017-03-17 Cyclic dinucleotide compounds and methods of use
CN202111504995.1A CN114230625A (en) 2016-03-18 2017-03-17 Cyclic dinucleotide compounds and methods of use
DK17767683.0T DK3429596T3 (en) 2016-03-18 2017-03-17 CYCLIC DINUCLEOTIDE COMPOUNDS AND METHODS OF USE
EA201892095A EA037513B1 (en) 2016-09-17 2017-03-17 Cyclic dinucleotide compounds and methods of use thereof
AU2017233068A AU2017233068C1 (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
CN201780030376.9A CN109475570B (en) 2016-03-18 2017-03-17 Cyclic dinucleotide compounds and methods of use
EP19218894.4A EP3692996A1 (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
NZ746112A NZ746112A (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
SG11201807660QA SG11201807660QA (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
CA3017524A CA3017524A1 (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
KR1020187029283A KR102530488B1 (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
JP2018568181A JP6980198B2 (en) 2016-03-18 2017-03-17 Cyclic dinucleotide compound and usage
EP17767683.0A EP3429596B1 (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
HRP20221263TT HRP20221263T1 (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
PL17767683.0T PL3429596T3 (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use
US15/953,492 US10519188B2 (en) 2016-03-18 2018-04-15 Cyclic di-nucleotide compounds and methods of use
ZA2018/06074A ZA201806074B (en) 2016-03-18 2018-09-11 Cyclic di-nucleotide compounds and methods of use
IL261827A IL261827B (en) 2016-03-18 2018-09-17 Cyclic di-nucleotide compounds and methods of use
US16/438,153 US11299512B2 (en) 2016-03-18 2019-06-11 Cyclic di-nucleotide compounds and methods of use
US17/697,247 US20220340613A1 (en) 2016-03-18 2022-03-17 Cyclic di-nucleotide compounds and methods of use

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201662310364P 2016-03-18 2016-03-18
US62/310,364 2016-03-18
US201662355382P 2016-06-28 2016-06-28
US62/355,382 2016-06-28
US201662396140P 2016-09-17 2016-09-17
US62/396,140 2016-09-17

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/953,492 Continuation US10519188B2 (en) 2016-03-18 2018-04-15 Cyclic di-nucleotide compounds and methods of use

Publications (1)

Publication Number Publication Date
WO2017161349A1 true WO2017161349A1 (en) 2017-09-21

Family

ID=59851227

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/023093 WO2017161349A1 (en) 2016-03-18 2017-03-17 Cyclic di-nucleotide compounds and methods of use

Country Status (22)

Country Link
US (3) US10519188B2 (en)
EP (2) EP3429596B1 (en)
JP (3) JP6980198B2 (en)
KR (1) KR102530488B1 (en)
CN (3) CN114230625A (en)
AU (1) AU2017233068C1 (en)
BR (1) BR112018068748B1 (en)
CA (1) CA3017524A1 (en)
DK (1) DK3429596T3 (en)
ES (1) ES2929628T3 (en)
HR (1) HRP20221263T1 (en)
HU (1) HUE060396T2 (en)
IL (2) IL280430B2 (en)
LT (1) LT3429596T (en)
MA (1) MA43827A (en)
MX (1) MX2022001755A (en)
NZ (1) NZ746112A (en)
PL (1) PL3429596T3 (en)
PT (1) PT3429596T (en)
SG (2) SG11201807660QA (en)
WO (1) WO2017161349A1 (en)
ZA (1) ZA201806074B (en)

Cited By (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9994607B2 (en) 2015-12-03 2018-06-12 Glaxosmithkline Intellectual Property Development Limited Compounds
WO2018100558A3 (en) * 2016-12-01 2018-07-19 Takeda Pharmaceutical Company Limited Cyclic purine dinucleotides as sting (stimulator of inteferon genes) agonists
WO2018138685A3 (en) * 2017-01-27 2018-10-04 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
US10106574B2 (en) 2015-08-13 2018-10-23 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
WO2018198084A1 (en) * 2017-04-27 2018-11-01 Lupin Limited Cyclic di-nucleotide compounds with tricyclic nucleobases
WO2019046511A1 (en) * 2017-08-31 2019-03-07 Sperovie Biosciences, Inc. Compounds, compositions, and methods for the treatment of disease
US10246480B2 (en) 2017-02-17 2019-04-02 Eisai R&D Management Co., Ltd. Compounds for the treatment of cancer
WO2019074887A1 (en) * 2017-10-10 2019-04-18 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
WO2019079261A1 (en) * 2017-10-16 2019-04-25 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
WO2019084060A1 (en) 2017-10-24 2019-05-02 Silverback Therapeutics, Inc. Conjugates and methods of use thereof for selective delivery of immune-modulatory agents
WO2019092660A1 (en) 2017-11-10 2019-05-16 Takeda Pharmaceutical Company Limited Sting modulator compounds, and methods of making and using
WO2019118839A1 (en) * 2017-12-15 2019-06-20 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
WO2019125974A1 (en) * 2017-12-20 2019-06-27 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
WO2019123338A1 (en) * 2017-12-20 2019-06-27 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2019123340A1 (en) * 2017-12-20 2019-06-27 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2019123339A1 (en) * 2017-12-20 2019-06-27 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2019165374A1 (en) 2018-02-26 2019-08-29 Gilead Sciences, Inc. Substituted pyrrolizine compounds as hbv replication inhibitors
WO2019173587A1 (en) * 2018-03-08 2019-09-12 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
US10414747B2 (en) 2016-10-04 2019-09-17 Merck Sharp & Dohme Corp. Benzo[b]thiophene compounds as sting agonists
WO2019180683A1 (en) 2018-03-23 2019-09-26 Takeda Pharmaceutical Company Limited Sting modulator compounds with sulfamate linkages, and methods of making and using
CN110291096A (en) * 2016-11-25 2019-09-27 詹森生物科技公司 Cyclic annular dinucleotides is as STING agonist
WO2019193543A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
WO2019193542A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides
WO2019195181A1 (en) 2018-04-05 2019-10-10 Gilead Sciences, Inc. Antibodies and fragments thereof that bind hepatitis b virus protein x
WO2019193533A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2'-cyclic dinucleotides
WO2019200247A1 (en) 2018-04-12 2019-10-17 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
WO2019205180A1 (en) * 2018-04-28 2019-10-31 杭州星鳌生物科技有限公司 Chemical composition of txs-wx compounds, preparation method and use of same against tumors
WO2019211799A1 (en) 2018-05-03 2019-11-07 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide
US10519188B2 (en) 2016-03-18 2019-12-31 Immunesensor Therapeutics, Inc. Cyclic di-nucleotide compounds and methods of use
WO2020016782A1 (en) * 2018-07-17 2020-01-23 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
WO2020028097A1 (en) 2018-08-01 2020-02-06 Gilead Sciences, Inc. Solid forms of (r)-11-(methoxymethyl)-12-(3-methoxypropoxy)-3,3-dimethyl-8-0x0-2,3,8,13b-tetrahydro-1h-pyrido[2,1-a]pyrrolo[1,2-c] phthalazine-7-c arboxylic acid
WO2020041720A1 (en) 2018-08-24 2020-02-27 Codiak Biosciences, Inc. Extracellular vesicles targeting dendritic cells and uses thereof
WO2020050406A1 (en) 2018-09-06 2020-03-12 第一三共株式会社 Novel cyclic dinucleotide derivative and antibody-drug conjugate thereof
WO2020056008A1 (en) 2018-09-12 2020-03-19 Silverback Therapeutics, Inc. Compositions for the treatment of disease with immune stimulatory conjugates
JP2020508310A (en) * 2017-02-21 2020-03-19 ボード オブ レジェンツ, ザ ユニバーシティ オブ テキサス システムBoard Of Regents, The University Of Texas System Cyclic dinucleotides as agonists of interferon gene stimulator-dependent signaling
WO2020057546A1 (en) 2018-09-21 2020-03-26 上海迪诺医药科技有限公司 Cyclic dinucleotide analogue, pharmaceutical composition thereof, and application
US10604542B2 (en) 2016-01-11 2020-03-31 Innate Tumor Immunity, Inc. Cyclic dinucleotides for treating conditions associated with sting activity such as cancer
WO2020074004A1 (en) * 2018-10-12 2020-04-16 上海济煜医药科技有限公司 Cyclic dinucleotide compound and uses thereof
WO2020092528A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
WO2020092621A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
US10662416B2 (en) 2016-10-14 2020-05-26 Precision Biosciences, Inc. Engineered meganucleases specific for recognition sequences in the hepatitis B virus genome
CN111263767A (en) * 2017-08-30 2020-06-09 北京轩义医药科技有限公司 Cyclic dinucleotides as stimulators of interferon gene modulators
CN111349132A (en) * 2018-12-21 2020-06-30 上海济煜医药科技有限公司 Tumor immunity compound and application thereof
EP3558324A4 (en) * 2016-12-20 2020-08-05 Merck Sharp & Dohme Corp. Cyclic dinucleotide sting agonists for cancer treatment
WO2020178768A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
WO2020178770A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides and prodrugs thereof
WO2020178769A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides and prodrugs thereof
WO2020191361A2 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Extracellular vesicles for vaccine delivery
WO2020191369A1 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
US10793557B2 (en) 2018-04-03 2020-10-06 Merck Sharp & Dohme Corp. Sting agonist compounds
WO2020214663A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020211668A1 (en) * 2019-04-15 2020-10-22 Bioardis Llc Cd73 inhibitors
WO2020214652A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020227421A1 (en) * 2019-05-09 2020-11-12 Aligos Therapeutics, Inc. Modified cyclic dinucleoside compounds as sting modulators
WO2020237025A1 (en) 2019-05-23 2020-11-26 Gilead Sciences, Inc. Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors
US10875872B2 (en) 2018-07-31 2020-12-29 Incyte Corporation Heteroaryl amide compounds as sting activators
WO2020263830A1 (en) 2019-06-25 2020-12-30 Gilead Sciences, Inc. Flt3l-fc fusion proteins and methods of use
WO2021003445A1 (en) 2019-07-03 2021-01-07 Codiak Biosciences, Inc. Extracellular vesicles targeting t cells and uses thereof
WO2021016204A1 (en) * 2019-07-19 2021-01-28 Immunesensor Therapeutics, Inc. Antibody-sting agonist conjugates and their use in immunotherapy
WO2021034804A1 (en) 2019-08-19 2021-02-25 Gilead Sciences, Inc. Pharmaceutical formulations of tenofovir alafenamide
US10947227B2 (en) 2018-05-25 2021-03-16 Incyte Corporation Tricyclic heterocyclic compounds as sting activators
WO2021062317A1 (en) 2019-09-25 2021-04-01 Codiak Biosciences, Inc. Extracellular vesicle compositions
WO2021067181A1 (en) 2019-09-30 2021-04-08 Gilead Sciences, Inc. Hbv vaccines and methods treating hbv
WO2021067644A1 (en) 2019-10-01 2021-04-08 Silverback Therapeutics, Inc. Combination therapy with immune stimulatory conjugates
WO2021076666A1 (en) * 2019-10-14 2021-04-22 Immunesensor Therapeutics, Inc. Methods of treating cancer with a sting agonist
US11008344B2 (en) 2018-07-31 2021-05-18 Incyte Corporation Tricyclic heteroaryl compounds as STING activators
US11021511B2 (en) 2017-01-27 2021-06-01 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
WO2021113765A1 (en) 2019-12-06 2021-06-10 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
WO2021168274A1 (en) 2020-02-21 2021-08-26 Silverback Therapeutics, Inc. Nectin-4 antibody conjugates and uses thereof
US11110106B2 (en) 2018-10-29 2021-09-07 Venenum Biodesign, LLC Sting agonists for treating bladder cancer and solid tumors
WO2021177438A1 (en) 2020-03-06 2021-09-10 第一三共株式会社 Antibody-drug conjugate including novel cyclic dinucleotide derivative
WO2021184017A1 (en) 2020-03-13 2021-09-16 Codiak Biosciences, Inc. Extracellular vesicles for treating neurological disorders
WO2021188959A1 (en) 2020-03-20 2021-09-23 Gilead Sciences, Inc. Prodrugs of 4'-c-substituted-2-halo-2'-deoxyadenosine nucleosides and methods of making and using the same
WO2021189047A2 (en) 2020-03-20 2021-09-23 Codiak Biosciences, Inc. Extracellular vesicles for therapy
WO2021206158A1 (en) 2020-04-10 2021-10-14 小野薬品工業株式会社 Method of cancer therapy
EA038805B1 (en) * 2017-11-21 2021-10-21 Такеда Фармасьютикал Компани Лимитед Cyclic dinucleotides as sting (stimulator of interferon genes) agonists
US11161864B2 (en) 2018-10-29 2021-11-02 Venenum Biodesign, LLC Sting agonists
WO2021232019A1 (en) 2020-05-15 2021-11-18 Immunesensor Therapeutics, Inc. Sting agonist combination treatments with immune checkpoint inhibitors
WO2022006327A1 (en) 2020-07-01 2022-01-06 Silverback Therapeutics, Inc. Anti-asgr1 antibody conjugates and uses thereof
WO2022031894A1 (en) 2020-08-07 2022-02-10 Gilead Sciences, Inc. Prodrugs of phosphonamide nucleotide analogues and their pharmaceutical use
WO2022050300A1 (en) 2020-09-02 2022-03-10 第一三共株式会社 NOVEL ENDO-β-N-ACETYLGLUCOSAMINIDASE
US11285131B2 (en) 2017-08-04 2022-03-29 Merck Sharp & Dohme Corp. Benzo[b]thiophene STING agonists for cancer treatment
WO2022066883A1 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Extracellular vesicles comprising kras antigens and uses thereof
WO2022066934A2 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
WO2022066898A2 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Methods of producing extracellular vesicles
WO2022066928A2 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
US11312772B2 (en) 2017-08-04 2022-04-26 Merck Sharp & Dohme Corp. Combinations of PD-1 antagonists and benzo [b] thiophene STING agonists for cancer treatment
TWI769195B (en) * 2016-12-01 2022-07-01 日商武田藥品工業股份有限公司 Cyclic dinucleotide
RU2776060C2 (en) * 2017-12-15 2022-07-13 Янссен Байотек, Инк. Cyclic dinucleotides as sting agonists
US11453697B1 (en) 2015-08-13 2022-09-27 Merck Sharp & Dohme Llc Cyclic di-nucleotide compounds as sting agonists
US11466047B2 (en) 2017-05-12 2022-10-11 Merck Sharp & Dohme Llc Cyclic di-nucleotide compounds as sting agonists
WO2022241134A1 (en) 2021-05-13 2022-11-17 Gilead Sciences, Inc. COMBINATION OF A TLR8 MODULATING COMPOUND AND ANTI-HBV siRNA THERAPEUTICS
WO2022245671A1 (en) 2021-05-18 2022-11-24 Gilead Sciences, Inc. Methods of using flt3l-fc fusion proteins
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2023004440A2 (en) 2021-07-23 2023-01-26 Immunesensor Therapeutics, Inc. Sting agonist combination treatments with cytokines
EP4137499A1 (en) 2021-08-17 2023-02-22 Ustav organicke chemie a biochemie AV CR, v.v.i. 7-substituted 7-deazaadenine-containing 2,3 -cyclic dinucleotides
US11596692B1 (en) 2018-11-21 2023-03-07 Incyte Corporation PD-L1/STING conjugates and methods of use
US11691990B2 (en) 2018-08-16 2023-07-04 Eisai R&D Management Co., Ltd Salts of compounds and crystals thereof
US11702430B2 (en) 2018-04-03 2023-07-18 Merck Sharp & Dohme Llc Aza-benzothiophene compounds as STING agonists
US11725024B2 (en) 2020-11-09 2023-08-15 Takeda Pharmaceutical Company Limited Antibody drug conjugates
TWI827720B (en) * 2018-11-02 2024-01-01 大陸商上海濟煜醫藥科技有限公司 Cyclic dinucleotide compounds and uses thereof
JP7440504B2 (en) 2018-07-10 2024-02-28 エフ-スター・セラピューティクス・インコーポレイテッド Compounds, compositions and methods for treating diseases

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019195658A1 (en) 2018-04-05 2019-10-10 Dana-Farber Cancer Institute, Inc. Sting levels as a biomarker for cancer immunotherapy
CN109929894B (en) * 2019-04-17 2021-06-01 中国农业科学院兰州兽医研究所 Preparation and activity identification method of porcine second messenger molecule 2 '3' -cGAMP
WO2021041532A1 (en) 2019-08-26 2021-03-04 Dana-Farber Cancer Institute, Inc. Use of heparin to promote type 1 interferon signaling
CN115417906B (en) * 2022-09-14 2024-02-27 杭州星鳌生物科技有限公司 Cyclic dinucleotide metal compound and preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014144666A2 (en) * 2013-03-15 2014-09-18 The University Of Chicago Methods and compositions related to t-cell activity
US20140329889A1 (en) * 2013-05-03 2014-11-06 The Regents Of The University Of California Cyclic di-nucleotide induction of type i interferon
US20150010613A1 (en) * 2012-06-08 2015-01-08 The John Hopkins University Compositions and methods for cancer immunotherapy

Family Cites Families (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9716557D0 (en) 1997-08-06 1997-10-08 Glaxo Group Ltd Benzylidene-1,3-dihydro-indol-2-one derivatives having anti-cancer activity
US8367716B2 (en) 2003-07-28 2013-02-05 Karaolis David K R Method for attentuating virulence of microbial pathogens and for inhibiting microbial biofilm formation
EP1729781B1 (en) 2004-03-15 2012-10-24 Karaolis, David K. R. A method for inhibiting cancer cell proliferation or increasing cancer cell apoptosis
EP1782826A1 (en) 2005-11-08 2007-05-09 GBF Gesellschaft für Biotechnologische Forschung mbH PQS and c-diGMP and its conjugates as adjuvants and their uses in pharmaceutical compositions
US20110262485A1 (en) 2008-08-04 2011-10-27 University Of Miami Sting (stimulator of interferon genes), a regulator of innate immune responses
US20120178710A1 (en) 2009-07-01 2012-07-12 Rutgers, The State University Of New Jersey Synthesis of cyclic diguanosine monophosphate and thiophosphate analogs thereof
US9061048B2 (en) 2010-12-15 2015-06-23 The Regents Of The University Of California Cyclic di-AMP induction of type I interferon
JP5650780B2 (en) 2012-04-04 2015-01-07 日東電工株式会社 Vaccine composition
CN104540945A (en) 2012-04-30 2015-04-22 格伦·N·巴伯 Modulating immune responses
EP2931738B1 (en) 2012-12-13 2019-02-06 Aduro Biotech, Inc. Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use
AU2013363087B2 (en) 2012-12-19 2018-07-19 Board Of Regents, The University Of Texas System Pharmaceutical targeting of a mammalian cyclic di-nucleotide signaling pathway
JP2016503655A (en) * 2012-12-27 2016-02-08 アデュロ バイオテック,インコーポレイテッド Signal peptide fusion partner facilitating expression of the antigenic sequence of Listeria, its preparation method and its use
JP6153116B2 (en) 2013-01-09 2017-06-28 国立大学法人東北大学 Triazole-linked cyclic dinucleotide analogs
SG11201508165VA (en) 2013-04-29 2015-11-27 Sloan Kettering Inst Cancer Compositions and methods for altering second messenger signaling
US9549944B2 (en) 2013-05-18 2017-01-24 Aduro Biotech, Inc. Compositions and methods for inhibiting “stimulator of interferon gene”—dependent signalling
MX354057B (en) * 2013-05-18 2018-02-09 The Regents Of The Univ Of California Star Compositions and methods for activating "stimulator of interferon gene"-dependent signalling.
CN105188373B (en) * 2013-05-18 2017-09-22 艾杜罗生物科技公司 Suppress the composition and method of " interferon gene stimulates the protein " dependent signals conduction
WO2015017652A1 (en) 2013-07-31 2015-02-05 Memorial Sloan-Kettering Cancer Center Sting crystals and modulators
CA2927009C (en) 2013-10-21 2019-04-23 Drexel University Use of sting agonists to treat chronic hepatitis b virus infection
US20160287623A1 (en) 2013-11-19 2016-10-06 The University Of Chicago Use of sting agonist as cancer treatment
US10421971B2 (en) * 2014-01-15 2019-09-24 The University Of Chicago Anti-tumor therapy
CN103908468B (en) 2014-04-21 2017-02-08 上海捌加壹医药科技有限公司 Application of cyclic dinucleotide cGAMP in preparing anti-tumor medicaments
US10450341B2 (en) * 2014-06-04 2019-10-22 Glaxosmithkline Intellectual Property Development Limited Cyclic di-nucleotides as modulators of STING
US11058758B2 (en) 2014-11-20 2021-07-13 National Institutes Of Biomedical Innovation, Health And Nutrition TH1-inducing adjuvant comprising combination of different nucleic acid adjuvants, and use of same
EP3233882B1 (en) 2014-12-16 2019-10-30 Kayla Therapeutics Fluorinated cyclic dinucleotides for cytokine induction
US20170340658A1 (en) 2014-12-16 2017-11-30 Invivogen Combined use of a chemotherapeutic agent and a cyclic dinucleotide for cancer treatment
EP3233089A4 (en) 2014-12-17 2018-11-14 Lipogen LLC Method of treating cancer with cgamp or cgasmp
GB201501462D0 (en) 2015-01-29 2015-03-18 Glaxosmithkline Ip Dev Ltd Novel compounds
MY190404A (en) 2015-03-10 2022-04-21 Aduro Biotech Inc Compositions and methods for activating "stimulator of interferon gene"-dependent signalling
US10329258B2 (en) 2015-04-30 2019-06-25 University Of Washington CGAS in systemic lupus erythematosus (SLE)
US20180169159A1 (en) 2015-06-11 2018-06-21 University Of Miami Cancer treatment and diagnosis
CN104962561A (en) * 2015-06-25 2015-10-07 河南农业大学 RNA aptamer and detection method for enzymatic cGAMP generation amount detection
CN106318997A (en) 2015-07-03 2017-01-11 聊城市奥润生物医药科技有限公司 Efficient preparation and purification method of thio- (seleno-) phosphoric acid cyclic di-nucleotide cGAMP
WO2017011444A1 (en) 2015-07-13 2017-01-19 The Wistar Institute Of Anatomy And Biology Methods and compositions for treating b cell cancers
TW201717968A (en) 2015-07-14 2017-06-01 春季銀行製藥公司 Compounds and compositions that induce RIG-I and other pattern recognition receptors
EP3334745B1 (en) * 2015-08-13 2024-05-15 Merck Sharp & Dohme LLC Cyclic di-nucleotide compounds as sting agonists
EP3368072A4 (en) 2015-10-28 2019-07-03 Aduro BioTech, Inc. Compositions and methods for activating "stimulator of interferon gene"-dependent signalling
US20170146519A1 (en) 2015-11-20 2017-05-25 Oregon Health & Science University Sting agonists and methods of selecting sting agonists
CN107849084B (en) 2015-12-03 2021-09-14 葛兰素史密斯克莱知识产权发展有限公司 Cyclic purine dinucleotides as STING modulators
WO2017100305A2 (en) 2015-12-07 2017-06-15 Opi Vi - Ip Holdco Llc Composition of antibody construct-agonist conjugates and methods of use thereof
US20170158772A1 (en) 2015-12-07 2017-06-08 Opi Vi - Ip Holdco Llc Compositions of antibody construct - agonist conjugates and methods of use thereof
CN106540260A (en) 2015-12-09 2017-03-29 聊城市奥润生物医药科技有限公司 Interferon gene stimulatory protein(SP)(STING)Application of the agonist in anti-alzheimer's disease
CN106554416B (en) 2015-12-09 2019-03-15 聊城市奥润生物医药科技有限公司 A kind of application of anti-PD-L1 Humanized monoclonal antibodies joint interferon gene stimulates the protein (STING) agonist in antitumor
US20180369268A1 (en) 2015-12-16 2018-12-27 Aduro Biotech, Inc. Methods for identifying inhibitors of "stimulator of interferon gene"- dependent interferon production
KR20180097751A (en) 2016-01-11 2018-08-31 인네이트 튜머 이뮤니티, 인코포레이티드 RTI ID = 0.0 &gt; STING &lt; / RTI &gt; activity,
US10723756B2 (en) 2016-01-11 2020-07-28 Innate Tumor Immunity Inc. Cyclic dinucleotides for treating conditions associated with STING activity such as cancer
US20170239283A1 (en) 2016-02-23 2017-08-24 Providence Health & Services - Oregon Use of sting agonists to treat virally-induced and pre-malignant growths
AU2017225769B2 (en) 2016-03-02 2023-01-05 The Board Of Regents Of The University Of Texas System Sting activating nanovaccine for immunotherapy
US20190070212A1 (en) 2016-03-11 2019-03-07 Spring Bank Pharmaceuticals, Inc. Compounds and compositions for the treatment of infections
CN108779473A (en) 2016-03-16 2018-11-09 法国居里学院 It is used to prepare the purposes of the method and the particle of the virion with ring dinucleotides for treating cancer
KR102530488B1 (en) * 2016-03-18 2023-05-08 이뮨 센서, 엘엘씨 Cyclic di-nucleotide compounds and methods of use
CN106539757A (en) 2016-03-20 2017-03-29 聊城市奥润生物医药科技有限公司 Application of the ring dinucleotide cGAMP- liposomees in antitumor
KR102527784B1 (en) 2016-04-07 2023-04-28 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Heterocyclic amides useful as protein modulators
KR20180132783A (en) 2016-04-07 2018-12-12 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Heterocyclic amides useful as protein modulators
WO2017186711A1 (en) 2016-04-25 2017-11-02 Invivogen Novel complexes of immunostimulatory compounds, and uses thereof
CN107412260B (en) 2016-05-23 2022-07-19 北京大学 cGAS-STING pathway activators and uses thereof
WO2017218358A1 (en) 2016-06-13 2017-12-21 The Regents Of The University Of California FLUORESCENT BIOSENSOR FOR 2', 3'-cGAMP
CN106552265A (en) 2016-06-21 2017-04-05 聊城市奥润生物医药科技有限公司 STING agonist and application of the IDO1 inhibitor drug combinations in antitumor
KR20190033526A (en) 2016-06-24 2019-03-29 인피니티 파마슈티칼스, 인코포레이티드 Combination therapy
WO2018009466A1 (en) 2016-07-05 2018-01-11 Aduro Biotech, Inc. Locked nucleic acid cyclic dinucleotide compounds and uses thereof
MX2019000216A (en) 2016-07-06 2019-11-12 Sperovie Biosciences Inc Compounds, compositions, and methods for the treatment of disease.
WO2018009652A1 (en) 2016-07-06 2018-01-11 Sperovie Biosciences, Inc. Compounds, compositions, and methods for the treatment of disease
WO2018013887A1 (en) 2016-07-15 2018-01-18 Sperovie Biosciences, Inc. Compounds, compositions, and methods for the treatment of disease
WO2018029256A1 (en) 2016-08-09 2018-02-15 Aarhus Universitet Modulation of ifi16 and sting activity
EP3922279A1 (en) 2016-08-30 2021-12-15 Dana Farber Cancer Institute, Inc. Drug delivery compositions and uses thereof
WO2018045204A1 (en) 2016-08-31 2018-03-08 Ifm Therapeutics, Inc Cyclic dinucleotide analogs for treating conditions associated with sting (stimulator of interferon genes) activity
WO2018053508A1 (en) 2016-09-19 2018-03-22 The University Of North Carolina At Chapel Hill Methods and compositions for inducing an immune response
US10537590B2 (en) 2016-09-30 2020-01-21 Boehringer Ingelheim International Gmbh Cyclic dinucleotide compounds
CR20190168A (en) 2016-10-04 2019-05-17 Merck Sharp & Dohme BENZO[b]THIOPHENE COMPOUNDS AS STING AGONISTS
WO2018065360A1 (en) 2016-10-07 2018-04-12 Biolog Life Science Institute Forschungslabor Und Biochemica-Vertrieb Gmbh Cyclic dinucleotides containing benzimidazole, method for the production of same, and use of same to activate stimulator of interferon genes (sting)-dependent signaling pathways
AU2017341735B2 (en) 2016-10-11 2022-01-13 Helmholtz Center for Infection Research Hepatitis C virus immunogenic compositions comprising as an adjuvant a cyclic dinucleotide or an archaeosome and methods of use thereof
JOP20170188A1 (en) 2016-11-25 2019-01-30 Janssen Biotech Inc Cyclic dinucleotides as sting agonists
JOP20170192A1 (en) 2016-12-01 2019-01-30 Takeda Pharmaceuticals Co Cyclic dinucleotide
WO2018118664A1 (en) 2016-12-20 2018-06-28 Merck Sharp & Dohme Corp. Combinations of pd-1 antagonists and cyclic dinucleotide sting agonists for cancer treatment
RU2019122598A (en) 2016-12-20 2021-01-22 Мерк Шарп И Доум Корп. CYCLIC DINUCLEOTIDE STING AGONISTS FOR CANCER TREATMENT
KR20190126761A (en) 2016-12-22 2019-11-12 마브파마, 인크. Compositions and Methods of Enhancing or Increasing Type I IFN Production
CA3047589A1 (en) 2016-12-22 2018-06-28 Mavupharma, Inc. Phosphodiesterase inhibitors and methods of microbial treatment
WO2018119117A1 (en) 2016-12-22 2018-06-28 The Regents Of The University Of California Methods of producing cyclic dinucleotides
ES2891326T3 (en) 2017-01-27 2022-01-27 Janssen Biotech Inc Cyclic dinucleotides as STING agonists
WO2018140831A2 (en) 2017-01-27 2018-08-02 Silverback Therapeutics, Inc. Tumor targeting conjugates and methods of use thereof
AU2018212788A1 (en) 2017-01-27 2019-07-25 Janssen Biotech, Inc. Cyclic dinucleotides as STING agonists
AU2017397458A1 (en) 2017-02-01 2019-08-15 Modernatx, Inc. RNA cancer vaccines
JP2018131427A (en) 2017-02-17 2018-08-23 国立研究開発法人理化学研究所 Technology for controlling immune cells
US10933078B2 (en) 2017-02-21 2021-03-02 Board Of Regents, The University Of Texas System Cyclic dinucleotides as agonists of stimulator of interferon gene dependent signalling
JOP20190218A1 (en) 2017-03-22 2019-09-22 Boehringer Ingelheim Int Modified cyclic dinucleotide compounds
WO2018198084A1 (en) 2017-04-27 2018-11-01 Lupin Limited Cyclic di-nucleotide compounds with tricyclic nucleobases
EP3621624B1 (en) 2017-05-12 2023-08-30 Merck Sharp & Dohme LLC Cyclic di-nucleotide compounds as sting agonists
EP3431484A1 (en) 2017-07-21 2019-01-23 Ludwig-Maximilians-Universität München A fluorescent cyclic dinucleotide and its use in methods of identifying substances having an ability to modulate the cgas/sting pathway
CN107335049B (en) 2017-08-18 2019-10-18 中国药科大学 Application of the composite family type cyclic peptide compounds as cGAS-STING signal pathway inhibitor
JP7311514B2 (en) 2017-08-30 2023-07-19 ベイジン シュエンイー ファーマサイエンシズ カンパニー, リミテッド Cyclic Dinucleotides as Interferon Gene Stimulator Modulators
US11638716B2 (en) 2017-08-31 2023-05-02 F-star Therapeutics, Inc. Compounds, compositions, and methods for the treatment of disease
EP4242212A3 (en) 2018-03-23 2023-11-15 Takeda Pharmaceutical Company Limited Sting modulator compounds with sulfamate linkages, and methods of making and using
CN108498529A (en) 2018-06-20 2018-09-07 福建师范大学 Dnmt rna inhibitor and cGAMP pharmaceutical compositions for tumor prevention treatment

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150010613A1 (en) * 2012-06-08 2015-01-08 The John Hopkins University Compositions and methods for cancer immunotherapy
WO2014144666A2 (en) * 2013-03-15 2014-09-18 The University Of Chicago Methods and compositions related to t-cell activity
US20140329889A1 (en) * 2013-05-03 2014-11-06 The Regents Of The University Of California Cyclic di-nucleotide induction of type i interferon

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
SHANAHAN ET AL.: "Identification of c-di-GMP Derivatives Resistant to an EAL Domain Phosphordiesterase", BIOCHEMISTRY, vol. 52, 2013, pages 365 - 377, XP055172476 *
ZHANG ET AL.: "Cyclic GMP-AMP Containing Mixed Phosphodiester Linkages is an Endogenous High-Affinity Ligand for STING", MOLECULAR CELL, vol. 51, 2013, pages 226 - 235, XP055300246 *

Cited By (222)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10766919B2 (en) 2015-08-13 2020-09-08 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
US11453697B1 (en) 2015-08-13 2022-09-27 Merck Sharp & Dohme Llc Cyclic di-nucleotide compounds as sting agonists
US10106574B2 (en) 2015-08-13 2018-10-23 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
US10759825B2 (en) 2015-08-13 2020-09-01 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as STING agonists
US10738074B2 (en) 2015-08-13 2020-08-11 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as STING agonists
US10730907B2 (en) 2015-12-03 2020-08-04 Glaxosmithkline Intellectual Property Development Limited Compounds
US10364266B2 (en) 2015-12-03 2019-07-30 Glaxosmithkline Intellectual Property Development Limited Compounds
US9994607B2 (en) 2015-12-03 2018-06-12 Glaxosmithkline Intellectual Property Development Limited Compounds
US11505571B2 (en) 2016-01-11 2022-11-22 Innate Tumor Immunity, Inc. Compounds and compositions for treating conditions associated with sting activity
US10961270B2 (en) 2016-01-11 2021-03-30 Innate Tumor Immunity, Inc. Compounds and compositions for treating conditions associated with sting activity
US10604542B2 (en) 2016-01-11 2020-03-31 Innate Tumor Immunity, Inc. Cyclic dinucleotides for treating conditions associated with sting activity such as cancer
US10519188B2 (en) 2016-03-18 2019-12-31 Immunesensor Therapeutics, Inc. Cyclic di-nucleotide compounds and methods of use
US11299512B2 (en) 2016-03-18 2022-04-12 Immunesensor Therapeutics, Inc. Cyclic di-nucleotide compounds and methods of use
US10414747B2 (en) 2016-10-04 2019-09-17 Merck Sharp & Dohme Corp. Benzo[b]thiophene compounds as sting agonists
US10703738B2 (en) 2016-10-04 2020-07-07 Merck Sharp & Dohme Corp. Benzo[b]thiophene compounds as STING agonists
US10730849B2 (en) 2016-10-04 2020-08-04 Merck Sharp & Dohme Corp. Benzo[b]thiophene compounds as STING agonists
US11274285B2 (en) 2016-10-14 2022-03-15 Precision Biosciences, Inc. Engineered meganucleases specific for recognition sequences in the Hepatitis B virus genome
US10662416B2 (en) 2016-10-14 2020-05-26 Precision Biosciences, Inc. Engineered meganucleases specific for recognition sequences in the hepatitis B virus genome
CN110291096A (en) * 2016-11-25 2019-09-27 詹森生物科技公司 Cyclic annular dinucleotides is as STING agonist
JP2020500862A (en) * 2016-11-25 2020-01-16 ヤンセン バイオテツク,インコーポレーテツド Cyclic dinucleotides as STING agonists
JP7316936B2 (en) 2016-11-25 2023-07-28 ヤンセン バイオテツク,インコーポレーテツド Cyclic dinucleotides as STING agonists
JP2020055860A (en) * 2016-12-01 2020-04-09 武田薬品工業株式会社 Cyclic dinucleotide as sting (stimulator of interferon genes) agonist
US11666594B2 (en) 2016-12-01 2023-06-06 Takeda Pharmaceutical Company Limited Antibody-drug conjugates comprising a cyclic dinucleotide
JP7229907B2 (en) 2016-12-01 2023-02-28 武田薬品工業株式会社 Cyclic dinucleotides as STING (stimulator of interferon genes) agonists
KR20190113768A (en) * 2016-12-01 2019-10-08 다케다 야쿠힌 고교 가부시키가이샤 Cyclic dinucleotides as STING (stimulators of the interferon gene) agonists
WO2018100558A3 (en) * 2016-12-01 2018-07-19 Takeda Pharmaceutical Company Limited Cyclic purine dinucleotides as sting (stimulator of inteferon genes) agonists
US10980825B2 (en) 2016-12-01 2021-04-20 Takeda Pharmaceutical Company Limited Cyclic dinucleotide
TWI769195B (en) * 2016-12-01 2022-07-01 日商武田藥品工業股份有限公司 Cyclic dinucleotide
KR102640802B1 (en) 2016-12-01 2024-02-26 다케다 야쿠힌 고교 가부시키가이샤 Cyclic dinucleotides as STING (stimulator of interferon genes) agonists
EP3868384A1 (en) * 2016-12-01 2021-08-25 Takeda Pharmaceutical Company Limited Cyclic dinucleotides as sting (stimulator of interferon genes) agonists
EP3558324A4 (en) * 2016-12-20 2020-08-05 Merck Sharp & Dohme Corp. Cyclic dinucleotide sting agonists for cancer treatment
WO2018138685A3 (en) * 2017-01-27 2018-10-04 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
US11021511B2 (en) 2017-01-27 2021-06-01 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
JP2020505405A (en) * 2017-01-27 2020-02-20 ヤンセン バイオテツク,インコーポレーテツド Cyclic dinucleotides as STING agonists
US11492367B2 (en) 2017-01-27 2022-11-08 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
CN110234403A (en) * 2017-01-27 2019-09-13 詹森生物科技公司 Cyclic annular dinucleotides as STING agonist
JP7275031B2 (en) 2017-01-27 2023-05-17 ヤンセン バイオテツク,インコーポレーテツド Cyclic dinucleotides as STING agonists
US11339188B2 (en) 2017-02-17 2022-05-24 Eisai R&D Management Co., Ltd. Compounds for the treatment of cancer
US10246480B2 (en) 2017-02-17 2019-04-02 Eisai R&D Management Co., Ltd. Compounds for the treatment of cancer
US10618930B2 (en) 2017-02-17 2020-04-14 Eisai R&D Management Co., Ltd. Compounds for the treatment of cancer
US11400108B2 (en) 2017-02-21 2022-08-02 Board Of Regents, The University Of Texas System Cyclic dinucleotides as agonists of stimulator of interferon gene dependent signalling
JP2020508310A (en) * 2017-02-21 2020-03-19 ボード オブ レジェンツ, ザ ユニバーシティ オブ テキサス システムBoard Of Regents, The University Of Texas System Cyclic dinucleotides as agonists of interferon gene stimulator-dependent signaling
WO2018198084A1 (en) * 2017-04-27 2018-11-01 Lupin Limited Cyclic di-nucleotide compounds with tricyclic nucleobases
US11466047B2 (en) 2017-05-12 2022-10-11 Merck Sharp & Dohme Llc Cyclic di-nucleotide compounds as sting agonists
US11285131B2 (en) 2017-08-04 2022-03-29 Merck Sharp & Dohme Corp. Benzo[b]thiophene STING agonists for cancer treatment
US11312772B2 (en) 2017-08-04 2022-04-26 Merck Sharp & Dohme Corp. Combinations of PD-1 antagonists and benzo [b] thiophene STING agonists for cancer treatment
US11773132B2 (en) 2017-08-30 2023-10-03 Beijing Xuanyi Pharmasciences Co., Ltd. Cyclic di-nucleotides as stimulator of interferon genes modulators
CN111263767A (en) * 2017-08-30 2020-06-09 北京轩义医药科技有限公司 Cyclic dinucleotides as stimulators of interferon gene modulators
CN111263767B (en) * 2017-08-30 2023-07-18 北京轩义医药科技有限公司 Cyclic dinucleotides as stimulators of interferon gene modulators
WO2019046511A1 (en) * 2017-08-31 2019-03-07 Sperovie Biosciences, Inc. Compounds, compositions, and methods for the treatment of disease
US11638716B2 (en) 2017-08-31 2023-05-02 F-star Therapeutics, Inc. Compounds, compositions, and methods for the treatment of disease
JP2020536897A (en) * 2017-10-10 2020-12-17 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Cyclic dinucleotide as an anticancer drug
JP7212683B2 (en) 2017-10-10 2023-01-25 ブリストル-マイヤーズ スクイブ カンパニー Cyclic dinucleotides as anticancer agents
WO2019074887A1 (en) * 2017-10-10 2019-04-18 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
US11660311B2 (en) 2017-10-10 2023-05-30 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
JP2020536971A (en) * 2017-10-16 2020-12-17 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Cyclic dinucleotide as an anticancer drug
WO2019079261A1 (en) * 2017-10-16 2019-04-25 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
CN111406063A (en) * 2017-10-16 2020-07-10 百时美施贵宝公司 Cyclic dinucleotides as anticancer agents
CN111406063B (en) * 2017-10-16 2023-09-15 百时美施贵宝公司 Cyclic dinucleotides as anticancer agents
JP7254821B2 (en) 2017-10-16 2023-04-10 ブリストル-マイヤーズ スクイブ カンパニー Cyclic dinucleotides as anticancer agents
US11427610B2 (en) 2017-10-16 2022-08-30 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
WO2019084060A1 (en) 2017-10-24 2019-05-02 Silverback Therapeutics, Inc. Conjugates and methods of use thereof for selective delivery of immune-modulatory agents
CN111566119A (en) * 2017-11-10 2020-08-21 武田药品工业有限公司 STING modulator compounds and methods of making and using
WO2019092660A1 (en) 2017-11-10 2019-05-16 Takeda Pharmaceutical Company Limited Sting modulator compounds, and methods of making and using
JP7195317B2 (en) 2017-11-10 2022-12-23 武田薬品工業株式会社 STING MODULATOR COMPOUNDS AND METHODS OF MAKING AND USING
JP2021502383A (en) * 2017-11-10 2021-01-28 武田薬品工業株式会社 STING modifier compounds, and methods of manufacture and use
US11542293B2 (en) 2017-11-10 2023-01-03 Takeda Pharmaceutical Company Limited Sting modulator compounds, and methods of making and using
EA038805B1 (en) * 2017-11-21 2021-10-21 Такеда Фармасьютикал Компани Лимитед Cyclic dinucleotides as sting (stimulator of interferon genes) agonists
RU2776060C2 (en) * 2017-12-15 2022-07-13 Янссен Байотек, Инк. Cyclic dinucleotides as sting agonists
JP7317014B2 (en) 2017-12-15 2023-07-28 ヤンセン バイオテツク,インコーポレーテツド Cyclic dinucleotides as STING agonists
WO2019118839A1 (en) * 2017-12-15 2019-06-20 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
AU2018386222B2 (en) * 2017-12-15 2023-04-20 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
JP2021506803A (en) * 2017-12-15 2021-02-22 ヤンセン バイオテツク,インコーポレーテツド Cyclic dinucleotide as a STING agonist
US10966999B2 (en) 2017-12-20 2021-04-06 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
CN111566120B (en) * 2017-12-20 2023-09-29 捷克共和国有机化学与生物化学研究所 3' cyclic dinucleotides with phosphonate bonds of activated STING adaptor protein
JP7098748B2 (en) 2017-12-20 2022-07-11 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 2'3'cyclic dinucleotide with phosphonate binding that activates the STING adapter protein
WO2019123339A1 (en) * 2017-12-20 2019-06-27 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2019123338A1 (en) * 2017-12-20 2019-06-27 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
JP7037667B2 (en) 2017-12-20 2022-03-16 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 3'3'cyclic dinucleotide with phosphonate binding that activates the STING adapter protein
KR20200100136A (en) * 2017-12-20 2020-08-25 인스티튜트 오브 오가닉 케미스트리 앤드 바이오케미스트리 에이에스 씨알 브이.브이.아이. 2'3' cyclic dinucleotide with a phosphonate bond that activates the STING adapter protein
WO2019125974A1 (en) * 2017-12-20 2019-06-27 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
US11203610B2 (en) 2017-12-20 2021-12-21 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
US11685761B2 (en) 2017-12-20 2023-06-27 Merck Sharp & Dohme Llc Cyclic di-nucleotide compounds as sting agonists
CN111511754A (en) * 2017-12-20 2020-08-07 捷克共和国有机化学与生物化学研究所 2'3' cyclic dinucleotides with phosphonate linkages that activate STING transducins
JP2021507943A (en) * 2017-12-20 2021-02-25 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 2'3'cyclic dinucleotide with phosphonic acid binding that activates the STING adapter protein
KR102492115B1 (en) * 2017-12-20 2023-01-27 인스티튜트 오브 오가닉 케미스트리 앤드 바이오케미스트리 에이에스 씨알 브이.브이.아이. 2'3' cyclic dinucleotides with phosphonate linkages that activate STING adapter proteins
TWI786242B (en) * 2017-12-20 2022-12-11 捷克科學院有機化學與生物化學研究所 3'3' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
JP2021508340A (en) * 2017-12-20 2021-03-04 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 3'3'cyclic dinucleotide with phosphonic acid binding that activates the STING adapter protein
CN111511754B (en) * 2017-12-20 2023-09-12 捷克共和国有机化学与生物化学研究所 2'3' cyclic dinucleotides with phosphonate bonds of activated STING adaptor protein
CN111566120A (en) * 2017-12-20 2020-08-21 捷克共和国有机化学与生物化学研究所 3'3' cyclic dinucleotides with phosphonate linkages that activate STING transducins
WO2019123340A1 (en) * 2017-12-20 2019-06-27 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2019165374A1 (en) 2018-02-26 2019-08-29 Gilead Sciences, Inc. Substituted pyrrolizine compounds as hbv replication inhibitors
JP2021515016A (en) * 2018-03-08 2021-06-17 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Cyclic dinucleotide as an anticancer drug
WO2019173587A1 (en) * 2018-03-08 2019-09-12 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
JP7250808B2 (en) 2018-03-08 2023-04-03 ブリストル-マイヤーズ スクイブ カンパニー Cyclic dinucleotides as anticancer agents
CN111801341A (en) * 2018-03-08 2020-10-20 百时美施贵宝公司 Cyclic dinucleotides as anticancer agents
CN111801341B (en) * 2018-03-08 2023-10-24 百时美施贵宝公司 Cyclic dinucleotides as anticancer agents
US11945834B2 (en) 2018-03-08 2024-04-02 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
JP7337086B2 (en) 2018-03-23 2023-09-01 武田薬品工業株式会社 STING MODULATOR COMPOUNDS HAVING SULFAMATE LINKS AND METHODS OF MAKING AND USING
WO2019180683A1 (en) 2018-03-23 2019-09-26 Takeda Pharmaceutical Company Limited Sting modulator compounds with sulfamate linkages, and methods of making and using
EP4242212A2 (en) 2018-03-23 2023-09-13 Takeda Pharmaceutical Company Limited Sting modulator compounds with sulfamate linkages, and methods of making and using
US20210015915A1 (en) * 2018-03-23 2021-01-21 Takeda Pharmaceutical Company Limited Sting modulator compounds with sulfamate linkages, and methods of making and using
JP2021518400A (en) * 2018-03-23 2021-08-02 武田薬品工業株式会社 STING modulator compounds with sulfamate bonds, and methods of manufacture and use
EP4242212A3 (en) * 2018-03-23 2023-11-15 Takeda Pharmaceutical Company Limited Sting modulator compounds with sulfamate linkages, and methods of making and using
US11702430B2 (en) 2018-04-03 2023-07-18 Merck Sharp & Dohme Llc Aza-benzothiophene compounds as STING agonists
US10793557B2 (en) 2018-04-03 2020-10-06 Merck Sharp & Dohme Corp. Sting agonist compounds
WO2019195181A1 (en) 2018-04-05 2019-10-10 Gilead Sciences, Inc. Antibodies and fragments thereof that bind hepatitis b virus protein x
WO2019193542A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides
JP2021521102A (en) * 2018-04-06 2021-08-26 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 3'3'-Cyclic dinucleotide
JP2021520389A (en) * 2018-04-06 2021-08-19 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 2'3'-Cyclic dinucleotide
WO2019193543A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
JP7326321B2 (en) 2018-04-06 2023-08-15 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 2'3'-cyclic dinucleotide
JP7296398B2 (en) 2018-04-06 2023-06-22 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 3'3'-cyclic dinucleotide
WO2019193533A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2'-cyclic dinucleotides
AU2019247905B2 (en) * 2018-04-06 2023-10-05 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
US11292812B2 (en) 2018-04-06 2022-04-05 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′-cyclic dinucleotides
TWI818007B (en) * 2018-04-06 2023-10-11 捷克科學院有機化學與生物化學研究所 2'3'-cyclic dinucleotides
US11149052B2 (en) 2018-04-06 2021-10-19 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′-cyclic dinucleotides
TWI833744B (en) 2018-04-06 2024-03-01 捷克科學院有機化學與生物化學研究所 3'3'-cyclic dinucleotides
AU2019247904B2 (en) * 2018-04-06 2023-11-23 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides
US11788077B2 (en) 2018-04-12 2023-10-17 Precision Biosciences, Inc. Polynucleotides encoding optimized engineered meganucleases having specificity for a recognition sequence in the Hepatitis B virus genome
US11142750B2 (en) 2018-04-12 2021-10-12 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the Hepatitis B virus genome
WO2019200247A1 (en) 2018-04-12 2019-10-17 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
WO2019205180A1 (en) * 2018-04-28 2019-10-31 杭州星鳌生物科技有限公司 Chemical composition of txs-wx compounds, preparation method and use of same against tumors
CN110407879A (en) * 2018-04-28 2019-11-05 杭州星鳌生物科技有限公司 The chemical composition of TXS-WX class compound, preparation method and its application in antitumor
WO2019211799A1 (en) 2018-05-03 2019-11-07 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide
US11713317B2 (en) 2018-05-25 2023-08-01 Incyte Corporation Tricyclic heterocyclic compounds as sting activators
US10947227B2 (en) 2018-05-25 2021-03-16 Incyte Corporation Tricyclic heterocyclic compounds as sting activators
JP7440504B2 (en) 2018-07-10 2024-02-28 エフ-スター・セラピューティクス・インコーポレイテッド Compounds, compositions and methods for treating diseases
CN112424212A (en) * 2018-07-17 2021-02-26 詹森生物科技公司 Cyclic dinucleotides as STING agonists
WO2020016782A1 (en) * 2018-07-17 2020-01-23 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
US11597746B2 (en) 2018-07-17 2023-03-07 Janssen Biotech, Inc. Cyclic dinucleotides as sting agonists
US11912722B2 (en) 2018-07-31 2024-02-27 Incyte Corporation Tricyclic heteroaryl compounds as sting activators
US11008344B2 (en) 2018-07-31 2021-05-18 Incyte Corporation Tricyclic heteroaryl compounds as STING activators
US10875872B2 (en) 2018-07-31 2020-12-29 Incyte Corporation Heteroaryl amide compounds as sting activators
US11427597B2 (en) 2018-07-31 2022-08-30 Incyte Corporation Heteroaryl amide compounds as sting activators
WO2020028097A1 (en) 2018-08-01 2020-02-06 Gilead Sciences, Inc. Solid forms of (r)-11-(methoxymethyl)-12-(3-methoxypropoxy)-3,3-dimethyl-8-0x0-2,3,8,13b-tetrahydro-1h-pyrido[2,1-a]pyrrolo[1,2-c] phthalazine-7-c arboxylic acid
US11691990B2 (en) 2018-08-16 2023-07-04 Eisai R&D Management Co., Ltd Salts of compounds and crystals thereof
WO2020041720A1 (en) 2018-08-24 2020-02-27 Codiak Biosciences, Inc. Extracellular vesicles targeting dendritic cells and uses thereof
RU2809547C2 (en) * 2018-09-06 2023-12-12 Даити Санкио Компани, Лимитед New cyclic dinucleotide derivative and its antibody-drug conjugate
KR20230122685A (en) 2018-09-06 2023-08-22 다이이찌 산쿄 가부시키가이샤 Novel cyclic dinucleotide derivative and antibody-drug conjugate thereof
WO2020050406A1 (en) 2018-09-06 2020-03-12 第一三共株式会社 Novel cyclic dinucleotide derivative and antibody-drug conjugate thereof
KR20210057066A (en) 2018-09-06 2021-05-20 다이이찌 산쿄 가부시키가이샤 Novel Cyclic Dinucleotide Derivatives and Antibody Drug Conjugates
KR102567591B1 (en) 2018-09-06 2023-08-16 다이이찌 산쿄 가부시키가이샤 Novel cyclic dinucleotide derivative and antibody-drug conjugate thereof
KR20230028589A (en) 2018-09-06 2023-02-28 다이이찌 산쿄 가부시키가이샤 Novel cyclic dinucleotide derivative and antibody-drug conjugate thereof
JPWO2020050406A1 (en) * 2018-09-06 2021-08-30 第一三共株式会社 New cyclic dinucleotide derivative and its antibody drug conjugate
JP7483975B2 (en) 2018-09-06 2024-05-15 第一三共株式会社 Novel cyclic dinucleotide derivatives and antibody-drug conjugates thereof
KR102567590B1 (en) 2018-09-06 2023-08-17 다이이찌 산쿄 가부시키가이샤 Novel cyclic dinucleotide derivatives and their antibody drug conjugates
JP7254818B2 (en) 2018-09-06 2023-04-10 第一三共株式会社 Novel cyclic dinucleotide derivatives and their antibody-drug conjugates
AU2019337051B2 (en) * 2018-09-06 2023-11-23 Daiichi Sankyo Company, Limited Novel cyclic dinucleotide derivative and antibody-drug conjugate thereof
WO2020056008A1 (en) 2018-09-12 2020-03-19 Silverback Therapeutics, Inc. Compositions for the treatment of disease with immune stimulatory conjugates
WO2020057546A1 (en) 2018-09-21 2020-03-26 上海迪诺医药科技有限公司 Cyclic dinucleotide analogue, pharmaceutical composition thereof, and application
WO2020074004A1 (en) * 2018-10-12 2020-04-16 上海济煜医药科技有限公司 Cyclic dinucleotide compound and uses thereof
CN112867727A (en) * 2018-10-12 2021-05-28 上海济煜医药科技有限公司 Cyclic dinucleoside compound and application thereof
CN112867727B (en) * 2018-10-12 2024-05-17 上海济煜医药科技有限公司 Cyclic dinucleotide compounds and uses thereof
US11401295B2 (en) 2018-10-12 2022-08-02 Shanghai Jemincare Pharmaceuticals Co., Ltd. Cyclic dinucleotide compound and uses thereof
JP7492523B2 (en) 2018-10-12 2024-05-29 シャンハイ ジェミンケア ファーマシューティカル カンパニー,リミティド Cyclic dinucleotide compounds and uses thereof
US11110106B2 (en) 2018-10-29 2021-09-07 Venenum Biodesign, LLC Sting agonists for treating bladder cancer and solid tumors
US11883420B2 (en) 2018-10-29 2024-01-30 Venenum Biodesign, LLC Sting agonists for treating bladder cancer and solid tumors
US11161864B2 (en) 2018-10-29 2021-11-02 Venenum Biodesign, LLC Sting agonists
WO2020092528A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
EP4371987A1 (en) 2018-10-31 2024-05-22 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
WO2020092621A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
TWI827720B (en) * 2018-11-02 2024-01-01 大陸商上海濟煜醫藥科技有限公司 Cyclic dinucleotide compounds and uses thereof
US11596692B1 (en) 2018-11-21 2023-03-07 Incyte Corporation PD-L1/STING conjugates and methods of use
CN111349132A (en) * 2018-12-21 2020-06-30 上海济煜医药科技有限公司 Tumor immunity compound and application thereof
CN111349132B (en) * 2018-12-21 2021-06-04 上海济煜医药科技有限公司 Tumor immunity compound and application thereof
JP2022523423A (en) * 2019-03-07 2022-04-22 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 2'3'-Cyclic dinucleotide and prodrugs thereof
JP2022523571A (en) * 2019-03-07 2022-04-25 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 3'3'-Cyclic dinucleotide and prodrugs thereof
WO2020178769A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides and prodrugs thereof
WO2020178770A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides and prodrugs thereof
WO2020178768A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
JP7350872B2 (en) 2019-03-07 2023-09-26 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 3'3'-cyclic dinucleotide and its prodrug
US11766447B2 (en) 2019-03-07 2023-09-26 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
JP7350871B2 (en) 2019-03-07 2023-09-26 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 2'3'-cyclic dinucleotide and its prodrug
WO2020191369A1 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
WO2020191361A2 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Extracellular vesicles for vaccine delivery
CN114206841A (en) * 2019-04-15 2022-03-18 博奥阿迪斯生物科技公司 CD73 inhibitor
WO2020211668A1 (en) * 2019-04-15 2020-10-22 Bioardis Llc Cd73 inhibitors
WO2020214663A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020214652A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
US11787833B2 (en) 2019-05-09 2023-10-17 Aligos Therapeutics, Inc. Modified cyclic dinucleoside compounds as sting modulators
WO2020227421A1 (en) * 2019-05-09 2020-11-12 Aligos Therapeutics, Inc. Modified cyclic dinucleoside compounds as sting modulators
WO2020237025A1 (en) 2019-05-23 2020-11-26 Gilead Sciences, Inc. Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors
WO2020263830A1 (en) 2019-06-25 2020-12-30 Gilead Sciences, Inc. Flt3l-fc fusion proteins and methods of use
WO2021003445A1 (en) 2019-07-03 2021-01-07 Codiak Biosciences, Inc. Extracellular vesicles targeting t cells and uses thereof
US11033635B2 (en) 2019-07-19 2021-06-15 Immunesensor Therapeutics, Inc. Antibody-STING agonist conjugates and their use in immunotherapy
US11213592B2 (en) 2019-07-19 2022-01-04 Immunesensor Therapeutics, Inc. Antibody-sting agonist conjugates and their use in immunotherapy
WO2021016204A1 (en) * 2019-07-19 2021-01-28 Immunesensor Therapeutics, Inc. Antibody-sting agonist conjugates and their use in immunotherapy
WO2021034804A1 (en) 2019-08-19 2021-02-25 Gilead Sciences, Inc. Pharmaceutical formulations of tenofovir alafenamide
WO2021062317A1 (en) 2019-09-25 2021-04-01 Codiak Biosciences, Inc. Extracellular vesicle compositions
WO2021067181A1 (en) 2019-09-30 2021-04-08 Gilead Sciences, Inc. Hbv vaccines and methods treating hbv
WO2021067644A1 (en) 2019-10-01 2021-04-08 Silverback Therapeutics, Inc. Combination therapy with immune stimulatory conjugates
WO2021076666A1 (en) * 2019-10-14 2021-04-22 Immunesensor Therapeutics, Inc. Methods of treating cancer with a sting agonist
WO2021113765A1 (en) 2019-12-06 2021-06-10 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
WO2021168274A1 (en) 2020-02-21 2021-08-26 Silverback Therapeutics, Inc. Nectin-4 antibody conjugates and uses thereof
US11179473B2 (en) 2020-02-21 2021-11-23 Silverback Therapeutics, Inc. Nectin-4 antibody conjugates and uses thereof
KR20220151630A (en) 2020-03-06 2022-11-15 다이이찌 산쿄 가부시키가이샤 Antibody Drug Conjugates Containing Novel Cyclic Dinucleotide Derivatives
WO2021177438A1 (en) 2020-03-06 2021-09-10 第一三共株式会社 Antibody-drug conjugate including novel cyclic dinucleotide derivative
WO2021184017A1 (en) 2020-03-13 2021-09-16 Codiak Biosciences, Inc. Extracellular vesicles for treating neurological disorders
WO2021188959A1 (en) 2020-03-20 2021-09-23 Gilead Sciences, Inc. Prodrugs of 4'-c-substituted-2-halo-2'-deoxyadenosine nucleosides and methods of making and using the same
WO2021189047A2 (en) 2020-03-20 2021-09-23 Codiak Biosciences, Inc. Extracellular vesicles for therapy
WO2021206158A1 (en) 2020-04-10 2021-10-14 小野薬品工業株式会社 Method of cancer therapy
WO2021232019A1 (en) 2020-05-15 2021-11-18 Immunesensor Therapeutics, Inc. Sting agonist combination treatments with immune checkpoint inhibitors
US11541126B1 (en) 2020-07-01 2023-01-03 Silverback Therapeutics, Inc. Anti-ASGR1 antibody TLR8 agonist comprising conjugates and uses thereof
WO2022006327A1 (en) 2020-07-01 2022-01-06 Silverback Therapeutics, Inc. Anti-asgr1 antibody conjugates and uses thereof
WO2022031894A1 (en) 2020-08-07 2022-02-10 Gilead Sciences, Inc. Prodrugs of phosphonamide nucleotide analogues and their pharmaceutical use
WO2022050300A1 (en) 2020-09-02 2022-03-10 第一三共株式会社 NOVEL ENDO-β-N-ACETYLGLUCOSAMINIDASE
WO2022066883A1 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Extracellular vesicles comprising kras antigens and uses thereof
WO2022066898A2 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Methods of producing extracellular vesicles
WO2022066928A2 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
WO2022066934A2 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
US11725024B2 (en) 2020-11-09 2023-08-15 Takeda Pharmaceutical Company Limited Antibody drug conjugates
WO2022241134A1 (en) 2021-05-13 2022-11-17 Gilead Sciences, Inc. COMBINATION OF A TLR8 MODULATING COMPOUND AND ANTI-HBV siRNA THERAPEUTICS
WO2022245671A1 (en) 2021-05-18 2022-11-24 Gilead Sciences, Inc. Methods of using flt3l-fc fusion proteins
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2023004440A2 (en) 2021-07-23 2023-01-26 Immunesensor Therapeutics, Inc. Sting agonist combination treatments with cytokines
EP4137499A1 (en) 2021-08-17 2023-02-22 Ustav organicke chemie a biochemie AV CR, v.v.i. 7-substituted 7-deazaadenine-containing 2,3 -cyclic dinucleotides
WO2023020640A1 (en) 2021-08-17 2023-02-23 Ustav Organicke Chemie A Biochemie Av Cr, V. V. I. 7-substituted 7-deazaadenine-containing 2',3'-cyclic dinucleotides

Also Published As

Publication number Publication date
MA43827A (en) 2018-11-28
JP2020100647A (en) 2020-07-02
CN114230625A (en) 2022-03-25
ZA201806074B (en) 2020-02-26
DK3429596T3 (en) 2022-10-24
AU2017233068B2 (en) 2022-07-28
IL261827A (en) 2018-10-31
AU2017233068C1 (en) 2023-05-25
CN109475570A (en) 2019-03-15
PL3429596T3 (en) 2022-12-19
PT3429596T (en) 2022-11-25
MX2022001755A (en) 2022-03-11
JP6980200B2 (en) 2021-12-15
HRP20221263T1 (en) 2023-03-03
HUE060396T2 (en) 2023-02-28
JP2022017500A (en) 2022-01-25
ES2929628T3 (en) 2022-11-30
EP3429596A4 (en) 2020-04-08
NZ746112A (en) 2023-01-27
JP6980198B2 (en) 2021-12-15
JP2019509339A (en) 2019-04-04
EP3429596A1 (en) 2019-01-23
SG11201807660QA (en) 2018-10-30
CN109475570B (en) 2022-04-01
BR112018068748B1 (en) 2024-01-16
IL280430B2 (en) 2023-11-01
US20200010501A1 (en) 2020-01-09
CN114751950A (en) 2022-07-15
KR102530488B1 (en) 2023-05-08
KR20190018408A (en) 2019-02-22
IL280430B1 (en) 2023-07-01
US10519188B2 (en) 2019-12-31
BR112018068748A2 (en) 2019-01-22
EP3429596B1 (en) 2022-08-31
SG10201912074PA (en) 2020-02-27
EP3692996A1 (en) 2020-08-12
IL280430A (en) 2021-03-01
US20220340613A1 (en) 2022-10-27
US20180230177A1 (en) 2018-08-16
LT3429596T (en) 2022-12-12
US11299512B2 (en) 2022-04-12
AU2017233068A1 (en) 2018-10-04
IL261827B (en) 2021-03-25
CA3017524A1 (en) 2017-09-21

Similar Documents

Publication Publication Date Title
AU2017233068B2 (en) Cyclic di-nucleotide compounds and methods of use
CN106459131B (en) Cyclic annular dinucleotides as STING regulator
US10730907B2 (en) Compounds
JP7282778B2 (en) Substituted bicyclic heterocyclic compounds as PRMT5 inhibitors
KR102130745B1 (en) Benzothiophene derivatives as estrogen receptor inhibitors
CA2973806A1 (en) Cyclic dinucleotides useful for the treatment of inter alia cancer
KR20020083532A (en) 4-amino-5-cyano-2-anilino-pyrimidine derivatives and their use as inhibitors of cell-cycle kinases
EA037513B1 (en) Cyclic dinucleotide compounds and methods of use thereof
BR112018011302B1 (en) COMPOUNDS, PHARMACEUTICALLY ACCEPTABLE SALTS THEREOF, PHARMACEUTICAL COMPOSITION AND IMMUNOGENIC COMPOSITION COMPRISING SAID COMPOUNDS AND THERAPEUTIC USES THEREOF

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 11201807660Q

Country of ref document: SG

ENP Entry into the national phase

Ref document number: 3017524

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2018568181

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2018/011224

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2017233068

Country of ref document: AU

Date of ref document: 20170317

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20187029283

Country of ref document: KR

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018068748

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 201892095

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 2017767683

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2017767683

Country of ref document: EP

Effective date: 20181018

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

Ref document number: 17767683

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 112018068748

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20180914