EP3758707A1 - 7-substituted sulfonimidoylpurinone compounds and derivatives for the treatment and prophylaxis of liver cancer - Google Patents

7-substituted sulfonimidoylpurinone compounds and derivatives for the treatment and prophylaxis of liver cancer

Info

Publication number
EP3758707A1
EP3758707A1 EP19708981.6A EP19708981A EP3758707A1 EP 3758707 A1 EP3758707 A1 EP 3758707A1 EP 19708981 A EP19708981 A EP 19708981A EP 3758707 A1 EP3758707 A1 EP 3758707A1
Authority
EP
European Patent Office
Prior art keywords
amino
methyl
purine
compound
mcthyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP19708981.6A
Other languages
German (de)
English (en)
French (fr)
Inventor
Thomas POESCHINGER
Carola Ries
Hong Shen
Hongying Yun
Sabine HOVES
Carina HAGE
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Hoffmann La Roche AG
Original Assignee
F Hoffmann La Roche AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by F Hoffmann La Roche AG filed Critical F Hoffmann La Roche AG
Publication of EP3758707A1 publication Critical patent/EP3758707A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/24Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one nitrogen and one sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to novel sulfonimidoylpurinones derivatives that have in vivo Toll-like receptor agonism activity, for (use in) the treatment and/or prophylaxis of liver cancer
  • Liver cancer is the fifth most common form of cancer. Each year, approximately 750,000 cases are diagnosed and about 700,000 people die from the disease each year, making it the third most common cause of cancer death in the world (Ferlay et al., Int. J. Cancer 127:2893-2917 (2010)). In the United States, the incidence of primary liver cancer has been rising, and while some progress has been made in detecting and treating localized disease, the five year survival rate for late stage liver cancer is still well below 10% (American-Cancer- Society. 2012. Cancer Facts & Figures 2012. Atlanta: American Cancer Society).
  • liver cancer treatments for liver cancer include surgical removal of the part of the liver containing the tumor (partial hepatectomy), liver transplantation, transcatheter arterial chemoembolization (TACE), in situ tumor destruction by various methods such as radiofirequency ablation (RFA) or cryosurgery and administration of Sorafenib.
  • TACE transcatheter arterial chemoembolization
  • RFA radiofirequency ablation
  • Sorafenib treatments for liver cancer remain a significant unmet medical need.
  • the present invention relates to compounds of formula (I),
  • TLRs Toll-like receptors
  • PAMPs pathogen-associated molecular patterns
  • TLR3, TLR7, TLR8 and TLR9 are located within endosomes.
  • TLR7 can be activated by binding to a specific small molecule ligand (i.e., TLR7 agonist) or its native ligand (i.e., single- stranded RNA, ssRNA). Following binding of ssRNA to TLR7, the receptor in its dimerized form is believed to undergo a structural change leading to the subsequent recruitment of adapter proteins at its cytoplasmic domain, including the myeloid differentiation primary response gene 88 (MyD88). Following the initiation of the receptor signalling cascade via the MyD88 pathway, cytoplasmic transcription factors such as interferon regulatory factor 7 (IRF-7) and nuclear factor kappa B (NF-kB) are activated. These transcription factors then translocate to the nucleus and initiate the transcription of various genes, e.g., IFN-a and other antiviral cytokine genes.
  • IRF-7 interferon regulatory factor 7
  • NF-kB nuclear factor kappa B
  • WO201772662 relates to TLR7 agonist-anti HER2 conjugates for the treatment of HER2 positive cancers.
  • Hotz et al, Oncoimmunology 2012, 227-228 relates to cancer treatment with TLR7 agonists.
  • TLR7 agonists are used systemically for the treatment of cancer.
  • Only topical TLR7 agonist imiquimod is known to induce immune- mediated rejection of skin metastases in patients with breast cancer (Adams S., Kozhaya L., Martiniuk F., Meng T.C., Chiriboga L., Liebes L., Hochman T., Shuman N., Axelrod D., Speyer J., et al. Clin. Cancer Res. 2012;18:6748-6757.
  • the present invention relates to a series of novel 6-amino-2-sulfonimidoyl-9- substituted-7-substituted-purin-8-one compounds with Toll-like receptor agonistic activity and their prodrugs for use in the treatment or prophylaxis (prevention) of liver cancer.
  • the present invention provides a series of novel 6-amino-2-sulfonimidoyl-9- substituted-7-substituted-purin-8-one compounds that have Toll-like receptor agonistic activity and their prodrugs.
  • the invention also provides the bio-activity of such compounds to induce cytokine/chemokine release, SEAP level increase by activating Toll-like receptors, such as TLR7 receptor, the metabolic conversion of prodrugs to parent compounds in the presence of human hepatocytes, and the therapeutic or prophylactic use of such compounds and their pharmaceutical compositions comprising these compounds and their prodrugs to treat or prevent liver cancer.
  • the present invention also provides compounds with superior activity.
  • the compounds of formula (I) also show good solubility and PK profiles.
  • the present invention relates to novel compounds of formula (I),
  • R 1 is Ci- 6 alkyl
  • R 2 is benzyl, said benzyl being unsubstituted or substituted by one, two or three
  • R 3 is -NR 4 R 5 , wherein
  • R 4 is Ci- 6 alkyl or Ci- 6 alkoxyCi- 6 alkyl
  • R 5 is (Ci-6alkyl)2NCOOCi-6alkyl, Ci- 6 alkoxyCi- 6 alkyl, Ci_
  • prodrug compounds are especially useful for the treatment of liver cancer as they are activated (converted into their active form) in the liver. They show valuable anti-tumor efficacy in vivo in liver cancer cell models (either alone or in combination with anti- PD1/PD1 antibodies or with anti- angiogenic agents) and in vitro against liver cancer cells (by activation of peripheral blood cells and/or factors).
  • the invention also relates to their use for the manufacture of a medicament for the treatment or prophylaxis of liver cancer, medicaments based on a compound in accordance with the invention for the treatment or prophylaxis of liver cancer.
  • the compounds of formula (I) are useful for the treatment or prophylaxis of liver cancer, especially for the treatment or prophylaxis of hepatocellular carcinoma, hepatoma, cholangiocarcinoma, hepatoblastoma, hepatic carcinoma, hepatic angiosarcoma, or metastatic liver cancer.
  • Ci- 6 alkyl denotes a saturated, linear or branched chain alkyl group containing 1 to 6, particularly 1 to 4 carbon atoms, for example methyl, ethyl, «-propyl, isopropyl, «-butyl, isobutyl, /c/t-butyl and the like.
  • Particular“Ci- 6 alkyl” groups are methyl, ethyl and «-propyl.
  • Ci- 6 alkoxy denotes a group of the formula Ci- 6 alkyl-0-.
  • Examples of Ci_ 6 alkoxy group include, but not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n- butoxy, isobutoxy and /c/t-butoxy.
  • Particular“Ci- 6 alkoxy” groups are methoxy, ethoxy and isopropoxy.
  • a more particular Ci- 6 alkoxy group is ethoxy.
  • halogen and“halo” are used interchangeably herein and denote fluoro, chloro, bromo, or iodo.
  • heterocyclyl denotes a monovalent saturated or partly unsaturated mono or bicyclic ring system of 3 to 10 ring atoms, comprising 1 to 5 ring heteroatoms selected from N, O and S, the remaining ring atoms being carbon.
  • heterocyclyl is a monovalent saturated monocyclic ring system of 4 to 7 ring atoms, comprising 1, 2, or 3 ring heteroatoms selected from N, O and S, the remaining ring atoms being carbon.
  • Examples for monocyclic saturated heterocyclyl are aziridinyl, oxiranyl, azetidinyl, oxetanyl, pyrrolidinyl, dimethylpyrrolidinyl, ethoxycarbonylpyrrolidinyl, tetrahydrofuranyl, tetrahydro-thienyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperazinyl, morpholinyl, thiomorpholinyl, dioxothiomorpholinyl, azepanyl, diazepanyl, homopiperazinyl, or oxazepanyl.
  • Monocyclic saturated heterocyclyl can be further substituted by one to three substituents independently selected from halogen, Ci- 6 alkyl and Ci- 6 alkoxycarbonyl.
  • substituted monocyclic saturated heterocyclyl are 4- methylpiperazinyl, dimethylpyrrolidinyl, ethoxycarbonylpyrrolidinyl, difluoropyrrolidinyl, fluoro(methyl)pyrrolidinyl.
  • Examples for bicyclic saturated heterocyclyl are
  • azabicyclo[3.2.1]octyl quinuclidinyl, oxaazabicyclo[3.2.1]octyl, azabicyclo[3.3.1]nonyl, oxaazabicyclo[3.3.1]nonyl, thiaazabicyclo[3.3.1]nonyl, azaspiro[3.3]heptanyl and oxaazaspiro[3.3]heptanyl.
  • Examples for partly unsaturated heterocyclyl are dihydro furyl, imidazolinyl, dihydrooxazolyl, tetrahydropyridinyl and dihydropyranyl.
  • carbonyl alone or in combination refers to the group -C(O)-.
  • Ci- 6 alkylcarbonyl refers to a group Ci- 6 alkyl-C(0)-, wherein the“Ci_ 6 alkyl” is as defined above.
  • Particular“Ci- 6 alkylcarbonyl” group is acetyl.
  • enantiomer denotes two stereoisomers of a compound which are non- superimposable mirror images of one another.
  • diastereomer denotes a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities.
  • pharmaceutically acceptable salts denotes salts which are not biologically or otherwise undesirable.
  • Pharmaceutically acceptable salts include both acid and base addition salts.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid, and organic acids selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, / olucncsulfonic acid, and salicyclic acid.
  • inorganic acids such as hydroch
  • salts derived from pharmaceutically acceptable organic nontoxic bases includes salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, trimethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, /V-cthylpipcridinc, and polyamine resins
  • racemates can be separated according to known methods into the enantiomers.
  • diastereomeric salts which can be separated by crystallization are formed from the racemic mixtures by reaction with an optically active acid such as e.g. D- or L-tartaric acid, mandelic acid, malic acid, lactic acid or
  • prodrug denotes a form or derivative of a compound which is metabolized in vivo, e.g., by biological fluids or enzymes by a subject after administration, into a pharmacologically active form of the compound in order to produce the desired pharmacological effect.
  • Prodrugs are described e.g. in“The Organic Chemistry of Drug Design and Drug Action”, by Richard B. Silverman, Academic Press, San Diego, 2004, Chapter 8 Prodrugs and Drug Delivery Systems, pp. 497-558.
  • a pharmaceutically active metabolite is intended to mean a pharmacologically active product produced through metabolism in the body of a specified compound or salt thereof. After entry into the body, most drugs are substrates for chemical reactions that may change their physical properties and biologic effects. These metabolic conversions, which usually affect the polarity of the compounds of the invention, alter the way in which drugs are distributed in and excreted from the body. However, in some cases, metabolism of a drug is required for therapeutic effect.
  • therapeutically effective amount denotes an amount of a compound or molecule of the present invention that, when administered to a subject, (i) treats or prevents the particular disease, condition or disorder, (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described herein.
  • the therapeutically effective amount will vary depending on the compound, the disease state being treated, the severity of the disease treated, the age and relative health of the subject, the route and form of administration, the judgement of the attending medical or veterinary practitioner, and other factors.
  • pharmaceutical composition denotes a mixture or solution comprising a therapeutically effective amount of an active pharmaceutical ingredient together with pharmaceutically acceptable excipients to be administered to a mammal, e.g., a human in need thereof.
  • the present invention relates to a compound of formula (I),
  • R 1 is Ci- 6 alkyl
  • R 2 is benzyl, said benzyl being unsubstituted or substituted by one, two or three
  • R 3 is -NR 4 R 5 , wherein
  • R 4 is Ci- 6 alkyl or Ci- 6 alkoxyCi- 6 alkyl
  • R 5 is (Ci-6alkyl)2NCOOCi-6alkyl, Ci- 6 alkoxyCi- 6 alkyl, Ci_
  • a further embodiment of present invention is (ii) a compound of formula (I), wherein
  • R 1 is Ci-ealkyl
  • R 2 is benzyl, said benzyl being unsubstituted or substituted by halogen or Ci_ 6 alkyl;
  • R 3 is azetidinyl
  • R 4 is Ci-ealkyl or Ci- 6 alkoxyCi- 6 alkyl
  • R 5 is (Ci-6alkyl)2NCOOCi-6alkyl, Ci- 6 alkoxyCi- 6 alkyl, Ci- 6alkoxycarbonyl(Ci- 6 alkyl)aminoCi- 6 alkyl, Ci_
  • a further embodiment of present invention is (iii) a compound of formula (I), wherein
  • R 1 is ethyl or propyl
  • R 2 is benzyl, bromobenzyl, chlorobenzyl, fluorobenzyl or methylbenzyl;
  • R 3 is azetidinyl
  • R 4 is methyl, ethyl, propyl or methoxyethyl
  • R 5 is acetyl(methyl)aminoethyl, butyl, butyl(methyl)carbamoyloxyethyl, diethylcarbamoyloxyethyl, ethoxycarbonyl(methyl)aminoethyl, ethoxycarbonylethyl, ethoxycarbonylisobutyl,
  • ethoxycarbonylisopentyl ethoxycarbonylmethyl, ethoxycarbonyloxyethyl, ethoxycarbonyl(phenyl)ethyl, ethyl, isobutyl, isopropoxycarbonylisopentyl, isopropoxycarbonyl(phenyl)ethyl, isopropyl, methoxycarbonyl(methyl)aminoethyl, methoxyethyl, methoxypropyl, propyl, propyl(methyl)carbamoyloxyethyl, pyrrolidinylcarbamoyloxyethyl, tert- butoxycarbonyl(methyl)aminoethyl, tert-butoxycarbonylethyl, tert- butoxycarbonylisopentyl or tert-butoxycarbonyl(phenyl)ethyl;
  • a further embodiment of present invention is (iii-l) a compound of formula (I), wherein
  • R 1 is ethyl or propyl
  • R 2 is benzyl, chlorobenzyl, fluorobenzyl or methylbenzyl
  • R 3 is azetidinyl
  • R 4 is methyl, ethyl, propyl or methoxyethyl
  • R 5 is acetyl(methyl)aminoethyl, butyl, butyl(methyl)carbamoyloxyethyl, diethylcarbamoyloxyethyl, ethoxycarbonyl(methyl)aminoethyl, ethoxycarbonylethyl, ethoxycarbonylisobutyl,
  • ethoxycarbonylisopentyl ethoxycarbonylmethyl, ethoxycarbonyloxyethyl, ethoxycarbonyl(phenyl)ethyl, ethyl, isobutyl, isopropoxycarbonylisopentyl, isopropoxycarbonyl(phenyl)ethyl, isopropyl, methoxycarbonyl(methyl)aminoethyl, methoxyethyl, methoxypropyl, propyl, propyl(methyl)carbamoyloxyethyl, pyrrolidinylcarbamoyloxyethyl, tert- butoxycarbonyl(methyl)aminoethyl, tert-butoxycarbonylethyl, tert- butoxycarbonylisopentyl or tert-butoxycarbonyl(phenyl)ethyl;
  • a further embodiment of present invention is (iv) a compound of formula (I), wherein R 3 is azetidinyl, 4-methylpiperazinyl, piperidinylpiperidinyl, pyrrolidinyl, acetyl(methyl)aminoethyl(methyl)amino, bis(methoxyethyl)amino, butyl(ethyl)amino, butyl(methyl)amino, butyl(methyl)carbamoyloxyethyl(methyl)amino,
  • a further embodiment of present invention is (v) a compound of formula (I), wherein R 1 is ethyl, for use in the treatment or prophylaxis of liver cancer.
  • a further embodiment of present invention is (vi) a compound of formula (I), wherein R 2 is benzyl substituted by halogen or Ci_ 6 alkyl, for use in the treatment or prophylaxis of liver cancer.
  • a further embodiment of present invention is (vii) a compound of formula (I), wherein R 2 is bromobenzyl, chlorobenzyl, fluorobenzyl or methylbenzyl, for use in the treatment or prophylaxis of liver cancer.
  • a further embodiment of present invention is (vii-l) a compound of formula (I), wherein R 2 is chlorobenzyl, fluorobenzyl or methylbenzyl, for use in the treatment or prophylaxis of liver cancer.
  • a further embodiment of present invention is (viii) a compound of formula (I), wherein R 2 is bromobenzyl, chlorobenzyl or fluorobenzyl, for use in the treatment or prophylaxis of liver cancer.
  • a further embodiment of present invention is (viii-l) a compound of formula (I), wherein R 2 is chlorobenzyl or fluorobenzyl, for use in the treatment or prophylaxis of liver cancer.
  • a further embodiment of present invention is (ix) a compound of formula (I), wherein R 3 is -NR 4 R 5 , wherein R 4 is Ci- 6 alkyl, R 5 is Ci- 6 alkyl, for use in the treatment or prophylaxis of liver cancer.
  • a further embodiment of present invention is (x) a compound of formula (I), wherein R 3 is propyl(methyl)amino or ethyl(methyl)amino, for use in the treatment or prophylaxis of liver cancer.
  • a further embodiment of present invention is (xi) a compound of formula (I), wherein
  • R 1 is Ci-ealkyl
  • R 2 is benzyl, said benzyl being substituted by halogen or Ci- 6 alkyl;
  • R 3 is -NR 4 R 5 , wherein R 4 is Ci- 6 alkyl, R 5 is Ci- 6 alkyl;
  • a further embodiment of present invention is (xii) a compound of formula (I), wherein
  • R 1 is ethyl
  • R 2 is methylbenzyl, bromobenzyl, chlorobenzyl or fluorobenzyl
  • R 3 is propyl(methyl)amino or ethyl(methyl)amino
  • a further embodiment of present invention is (xii- 1 ) a compound of formula (I), wherein
  • R 1 is ethyl
  • R 2 is methylbenzyl, chlorobenzyl or fluorobenzyl
  • R 3 is propyl(methyl)amino or ethyl(methyl)amino
  • the compounds of the present invention can be prepared by any conventional means. Suitable processes for synthesizing these compounds as well as their starting materials are provided in the schemes below and in the examples. All substituents, in particular, R 1 to R 14 are as defined above unless otherwise indicated. Furthermore, and unless explicitly otherwise stated, all reactions, reaction conditions, abbreviations and symbols have the meanings well known to a person of ordinary skill in organic chemistry.
  • a compound of formula VI is prepared by cyclization of isocyanate VII with aminomalononitrile /3-tolucncsulfonatc. Then bicycle V is synthesized by reaction of compound of formula VI with benzoyl isothiocyanate in the presence of inorganic base, such as NaOH or KOH. Alkylation of bicycle V with alkylhalide in the presence of base, such as K 2 CO 3 , NaH or CS 2 CO 3 , gives compound of formula IV.
  • Compound of formula III is prepared by oxidation of compound of formula IV with an oxidant, such as meta- chloroperoxybenzoic acid, urea-hydrogen peroxide adduct and HIO 4 .
  • Compound of formula II is obtained by imination of compound of formula III with imination reagent, such as sodium azide in acid, said acid is, for example, Eaton’s reagent or PPA.
  • imination reagent such as sodium azide in acid
  • said acid is, for example, Eaton’s reagent or PPA.
  • Compound of formula I is obtained by reaction of compound of formula II with carbamoyl chloride in the presence of a mixed base such as pyridine and triethylamine, pyridine and DIPEA, DMAP and triethylamine, or DMAP and DIPEA.
  • a compound of formula X is prepared by reaction of compound of formula XI with R 2 NH 2 . Reduction of compound X with reducing reagent, such as Zinc or Iron powder in AcOH, gives the compound of formula IX. Cyclization of compound of formula IX with cyclization reagents, such as phosgene, carbonyl diimidazole, diethyl carbonate and triphosgene, affords compound of formula VIII.
  • a compound of formula IVa is prepared by treating the compound of formula VIII with PMBNEb.
  • a compound of formula III is prepared by deprotection of compound of formula IVa with acid, such as CF 3 COOH, followed by oxidation with an oxidant, such as meto-chloroperoxybenzoic acid, urea- hydrogen peroxide adduct and HIO4.
  • acid such as CF 3 COOH
  • an oxidant such as meto-chloroperoxybenzoic acid, urea- hydrogen peroxide adduct and HIO4.
  • Compound of formula II is obtained by the imination of compound of formula III with imination reagent, such as sodium azide in acid, said acid is for example Eaton’s reagent or PPA.
  • R 1 and R 2 are defined above.
  • the mixed base can be, for example, pyridine and triethylamine, pyridine and DIPEA, DMAP and triethylamine, or DMAP and DIPEA.
  • a compound of formula (I) when manufactured according to the above process, for use in the treatment or prophylaxis of liver cancer is also an object of the invention.
  • compositions or medicaments for use in the treatment or prophylaxis of liver cancer containing the compounds of the invention and a therapeutically inert carrier, diluent or excipient, as well as methods of using the compounds of the invention to prepare such compositions and medicaments.
  • compounds of formula (I) may be formulated by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a galenical administration form.
  • physiologically acceptable carriers i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a galenical administration form.
  • the pH of the formulation depends mainly on the particular use and the concentration of compound, but preferably ranges anywhere from about 3 to about 8.
  • a compound of formula (I) are formulated in an acetate buffer, at pH 5.
  • the compounds of formula (I) are sterile.
  • the compound may be stored, for example, as a solid or amorphous composition, as a lyophilized formulation or as an aqueous solution.
  • compositions are formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • The“effective amount” of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to activate TLR7 receptor and lead to produce INF-a and other cytokines, which can be used, but not limited, for the treatment or prevention of hepatitis B and/or C viral infected patients.
  • the pharmaceutically effective amount of the compound of the invention administered parenterally per dose will be in the range of about 0.1 to 50 mg/kg, alternatively about 0.1 to 30 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day.
  • oral unit dosage forms such as tablets and capsules, preferably contain from about 20 to about 1000 mg of the compound of the invention.
  • the compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal and epidural and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the compounds of the present invention may be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches, etc.
  • Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents.
  • a typical formulation is prepared by mixing a compound of the present invention and a carrier or excipient.
  • Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C., et ah, Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005.
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • buffers stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing
  • An example of a suitable oral dosage form is a tablet containing about 20 to 1000 mg of the compound of the invention compounded with about 30 to 90 mg anhydrous lactose, about 5 to 40 mg sodium croscarmellose, about 5 to 30 mg polyvinylpyrrolidone (PVP) K30, and about 1 to 10 mg magnesium stearate.
  • the powdered ingredients are first mixed together and then mixed with a solution of the PVP.
  • the resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment.
  • An example of an aerosol formulation can be prepared by dissolving the compound, for example 20 to 1000 mg, of the invention in a suitable buffer solution, e.g. a phosphate buffer, adding a tonicifier, e.g. a salt such sodium chloride, if desired.
  • the solution may be filtered, e.g., using a 0.2 micron filter, to remove impurities and contaminants.
  • An embodiment therefore, includes a pharmaceutical composition comprising a compound of formula (I) or pharmaceutically acceptable salts or enantiomers or diastereomers thereof.
  • composition comprising a compound of formula (I) or pharmaceutically acceptable salts or enantiomers or diastereomers thereof, together with a pharmaceutically acceptable carrier or excipient.
  • Another embodiment includes a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or pharmaceutically acceptable salts or enantiomers or diastereomers thereof for use in the treatment of hepatitis B virus infection.
  • the present invention provides methods for treating or preventing liver cancer in a patient in need thereof.
  • the liver cancer is hepatocellular carcinoma, hepatoma, cholangiocarcinoma, hepatoblastoma, hepatic carcinoma, hepatic angiosarcoma, or metastatic liver cancer.
  • the liver cancer is a refractory cancer.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation.
  • liver cancer examples include, but are not limited to, hepatocellular carcinoma, hepatoma, hepatoblastoma, cholangiocarcinoma, hepatoblastoma, hepatic carcinoma, sarcoma, lymphoma and hepatic angiosarcoma.
  • the liver cancer e.g., HCC
  • the liver cancer e.g., HCC
  • the liver cancer e.g., HCC
  • the liver cancer can be metastatic or non-metastatic.
  • the liver cancer e.g., HCC
  • the liver cancer can comprise a single tumor, multiple tumors, or a poorly defined tumor with an infiltrative growth pattern (into portal veins or hepatic veins).
  • the liver cancer e.g., HCC
  • the liver cancer can comprise a fibrolamellar, pseudoglandular (adenoid), pleomorphic (giant cell), or clear cell pattern.
  • the liver cancer e.g., HCC
  • the liver cancer (e.g., HCC) can comprise a poorly differentiated form, and malignant epithelial cells are discohesive, pleomorphic, anaplastic, and/or giant.
  • the liver cancer (e.g., HCC) is associated with hepatits B, hepatitis C, cirhhosis, or type 2 diabetes.
  • the terms "cell proliferative disorder” and "proliferative disorder” refer to disorders that are associated with some degree of abnormal cell proliferation.
  • the cell proliferative disorder is cancer.
  • liver cancer prophylaxis/prevention of liver cancer in patients which have a high risk of developing liver cancer.
  • the compounds described herein are especially useful as prodrugs which are converted into the active drug predominantly in the liver.
  • One embodiment of the invention embodiment are the prodrug compounds described herein for use in the treatment of liver cancer wherein the compounds are prodrugs of the formula (I),
  • R 1 is Ci- 6 alkyl
  • R 2 is benzyl, said benzyl being unsubstituted or substituted by one, two or three
  • R 3 is -NR 4 R 5 , wherein
  • R 4 is Ci_ 6 alkyl or Ci_ 6 alkoxyCi_ 6 alkyl
  • R 5 is (Ci-6alkyl)2NCOOCi_6alkyl, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_
  • prodrug compounds of formula I are converted in the human liver into the active drug of the formula II
  • example 61 demonstrates the liver as the primary site of conversion of the prodrug into its active form.
  • One preferred embodiment of the invention are the (prodrug) compounds described herein wherein the compounds are susceptible for the conversion into its active form by the liver enzymes CYP2C9 and CYP2C19.
  • One preferred embodiment of the invention are the (prodrug) compounds described herein wherein the compounds show a conversion rate into the active compound of >10 nmol/min/mg protein in human hepatocytes and of ⁇ 2 nmol/min/mg protein in human enterocytes (as measured in an appropriate assay using human hepatocytes and human enterocytes.
  • One aspect of the invention is the combined treatment (combination treatment) of a patient suffering from liver cancer with the compound of formula I with Surprisingly, we found that that a combination therapy of the compounds of formula I and an anti-PD-Ll/PDl axis treatment is highly effective for liver tumors
  • one aspect of the invention is a compound of the formula (I) (or a medicament or a pharmaceutical composition comprising such compound),
  • R 1 is Ci- 6 alkyl
  • R 2 is benzyl, said benzyl being unsubstituted or substituted by one, two or three substituents independently selected from halogen and Ci- 6 alkyl;
  • R 3 is -NR 4 R 5 , wherein
  • R 4 is Ci- 6 alkyl or Ci- 6 alkoxyCi- 6 alkyl
  • R 5 is (Ci-6alkyl)2NCOOCi-6alkyl, Ci- 6 alkoxyCi- 6 alkyl, Ci_
  • One embodiment of the invention is a compound of the formula (I) (or a medicament or a pharmaceutical composition comprising such compound),
  • R 1 is Ci_ 6 alkyl
  • R 2 is benzyl, said benzyl being unsubstituted or substituted by one, two or three substituents independently selected from halogen and Ci_ 6 alkyl;
  • R 3 is -NR 4 R 5 , wherein
  • R 4 is Ci_ 6 alkyl or Ci_ 6 alkoxyCi_ 6 alkyl
  • R 5 is (Ci-6alkyl)2NCOOCi-6alkyl, Ci- 6 alkoxyCi- 6 alkyl, Ci_
  • an antagonistic PD1 or antagonistic PD-L1 antibody is co-administered (wherein the treatment is in combination with an antagonistic PD 1 or antagonistic PD-L1 antibody).
  • One embodiment of the invention is the use of a compound of the formula (I),
  • R 1 is Ci- 6 alkyl
  • R 2 is benzyl, said benzyl being unsubstituted or substituted by one, two or three substituents independently selected from halogen and Ci- 6 alkyl;
  • R 3 is -NR 4 R 5 , wherein
  • R 4 is Ci_ 6 alkyl or Ci_ 6 alkoxyCi_ 6 alkyl
  • R 5 is (Ci-6alkyl)2NCOOCi_6alkyl, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_
  • the particular compounds of formula (I) which are used in the combination therapy with the antagonistic PD 1 or antagonistic PD- Ll antibody are selected from the following:
  • the particular compounds of formula (I) which are used in the combination therapy with the antagonistic PD 1 or antagonistic PD- Ll antibody are selected from the following:
  • the particular compound of formula (I) which is used in the combination therapy with the antagonistic PD1 or antagonistic PD-L1 antibody is: 6- Amino-9- [(4-chlorophcny l)mcthy l]-/V-cthy 1-2 [S(S)-cthy lsulfonimidoy 1]-/V- methyl-8-oxo-purine-7-carboxamide
  • the co-administration (or combination therapy or treatment in combination with or combination treatment) of the compound of formula I and the antagonistic PD 1 or antagonistic PD-L1 antibody is simultaneously. In one embodiment the co-administration (or combination therapy or treatment in combination with or combination treatment) of the compound of formula I and the antagonistic PD 1 or antagonistic PD-L1 antibody is sequentially.
  • the co-administration can be simultaneous or sequential in either order, wherein there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • the co-administration is either simultaneously or sequentially (e.g. intravenous (i.v.) through a continuous infusion.
  • the co-administration is simultaneously.
  • the co-administration is sequentially.
  • the co-administration is either simultaneously or sequentially (e.g. intravenous (i.v.) through a continuous infusion.
  • a“therapeutically effective amount” or simply“effective amount” which is the amount of the respective compound or combination that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • the amount of co-administration and the timing of co-administration will depend on the type (species, gender, age, weight, etc.) and condition of the patient being treated and the severity of the disease or condition being treated.
  • Said compounds of formula I and said antibodies are suitably co-administered to the patient at one time or over a series of treatments e.g. on the same day or on the day after.
  • PD-l programmed death - 1 receptor
  • PD-L2 B7-DC, CD273
  • the negative regulatory role of PD-l was revealed by PD-l knock outs (Pdcdl-/-), which are prone to
  • PD-l is related to CD28 and CTLA-4, but lacks the membrane proximal cysteine that allows homodimerization.
  • the cytoplasmic domain of PD-l contains an immunoreceptor tyrosine-based inhibition motif (ITIM, V/IxYxxL/V).
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • PD-l only binds to PD-L1 and PD-L2.
  • PD-l can be expressed on T cells, B cells, natural killer T cells, activated monocytes and dendritic cells (DCs). PD-l is expressed by activated, but not by unstimulated human CD4+ and CD8+ T cells, B cells and myeloid cells. This stands in contrast to the more restricted expression of CD28 and CTLA-4. Nishimura et al., Int. Immunol. 8: 773-80 (1996); Boettler et al., J. Virol. 80: 3532-40 (2006).
  • PD-l There are at least 4 variants of PD-l that have been cloned from activated human T cells, including transcripts lacking (i) exon 2, (ii) exon 3, (iii) exons 2 and 3 or (iv) exons 2 through 4. Nielsen et al., Cell. Immunol. 235: 109-16 (2005). With the exception of PD-ldeltaex3, all variants are expressed at similar levels as full length PD-l in resting peripheral blood mononuclear cells (PBMCs). Expression of all variants is significantly induced upon activation of human T cells with anti-CD3 and anti-CD28.
  • PBMCs peripheral blood mononuclear cells
  • the PD-ldeltaex3 variants lacks a transmembrane domain, and resembles soluble CTLA-4, which plays an important role in autoimmunity. Ueda et al., Nature 423: 506-11 (2003). This variant is enriched in the synovial fluid and sera of patients with rheumatoid arthritis. Wan et al., J. Immunol. 177: 8844-50 (2006).
  • PD-L1 is constitutively expressed on mouse T and B cells, CDs, macrophages, mesenchymal stem cells and bone marrow- derived mast cells. Yamazaki et al., J. Immunol. 169: 5538-45 (2002). PD-L1 is expressed on a wide range of nonhematopoietic cells (e.g., cornea, lung, vascular epithelium, liver nonparenchymal cells, mesenchymal stem cells, pancreatic islets, placental
  • JAK2 has also been implicated in PD-L1 induction.
  • PI3K phosphatidylinositol 3-kinase
  • Akt phosphatidylinositol 3-kinase
  • PD-L2 expression is more restricted than PD-L1.
  • PD-L2 is inducibly expressed on DCs, macrophages, and bone marrow- derived mast cells.
  • PD-L2 is also expressed on about half to two-thirds of resting peritoneal Bl cells, but not on conventional B2 B cells.
  • Zhong et al. Eur. J. Immunol. 37: 2405-10 (2007).
  • PD-L2+ Bl cells bind phosphatidylcholine and may be important for innate immune responses against bacterial antigens.
  • Induction of PD- L2 by IFN-gamma is partially dependent upon NF-kappaB. Liang et al., Eur. J. Immunol.
  • PD-L2 can also be induced on monocytes and macrophages by GM- CF, IL-4 and IFN-gamma. Yamazaki et al., J. Immunol. 169: 5538-45 (2002); Loke et al., PNAS 100:5336-41 (2003).
  • PD-l signaling typically has a greater effect on cytokine production than on cellular proliferation, with significant effects on IFN-gamma, TNF-alpha and IL-2 production.
  • PD- 1 mediated inhibitory signaling also depends on the strength of the TCR signaling, with greater inhibition delivered at low levels of TCR stimulation. This reduction can be overcome by costimulation through CD28 [Freeman et al., J. Exp. Med. 192: 1027-34 (2000)] or the presence of IL-2 [Carter et al., Eur. J. Immunol. 32: 634-43 (2002)].
  • Evidence is mounting that signaling through PD-L1 and PD-L2 may be bidirectional.
  • signaling may also be delivered back to the cells expressing PD-L1 and PD-L2.
  • dendritic cells with a naturally human anti-PD-L2 antibody isolated from a patient with Waldenstrom’s macroglobulinemia was not found to upregulate MHC II or B7 costimulatory molecules, such cells did produce greater amount of proinflammatory cytokines, particularly TNF- alpha and IL-6, and stimulated T cell proliferation. Nguyen et ah, J. Exp. Med. 196: 1393- 98 (2002). Treatment of mice with this antibody also (1) enhanced resistance to transplanted b 16 melanoma and rapidly induced tumor-specific CTL.
  • Radhakrishnan et ah J. Immunol. 170: 1830-38 (2003); Radhakrishnan et ah, Cancer Res. 64: 4965-72 (2004); Heckman et ah, Eur. J. Immunol. 37: 1827-35 (2007); (2) blocked development of airway inflammatory disease in a mouse model of allergic asthma. Radhakrishnan et al., J.
  • DC dendritic cells
  • B7.1 has already been identified as a binding partner for PD-L1. Butte et al., Immunity 27: 111-22 (2007). Chemical crosslinking studies suggest that PD-L1 and B7.1 can interact through their IgV-like domains. B7.1 :PD-Ll interactions can induce an inhibitory signal into T cells. Ligation of PD-L 1 on CD4+ T cells by B7.1 or ligation of B7.1 on CD4+ T cells by PD-L1 delivers an inhibitory signal. T cells lacking CD28 and CTLA-4 show decreased proliferation and cytokine production when stimulated by anti- CD3 plus B7.1 coated beads.
  • T cells lacking all the receptors for B7.1 i.e., CD28, CTLA-4 and PD-L1
  • B7.1 acts specifically through PD- Ll on the T-cell in the absence of CD28 and CTLA-4.
  • T cells lacking PD-l showed decreased proliferation and cytokine production when stimulated in the presence of anti-CD3 plus PD-L1 coated beads, demonstrating the inhibitory effect of PD-L 1 ligation on B7.1 on T cells.
  • T cells lacking all known receptors for PD-L1 i.e., no PD-l and B7.1
  • T cell proliferation was no longer impaired by anti-CD3 plus PD-L1 coated beads.
  • PD-L1 can exert an inhibitory effect on T cells either through B7.1 or PD-l.
  • B7.1 and PD-L1 The direct interaction between B7.1 and PD-L1 suggests that the current understanding of costimulation is incomplete, and underscores the significance to the expression of these molecules on T cells.
  • Studies of PD-L1-/- T cells indicate that PD-L1 on T cells can downregulate T cell cytokine production. Latchman et al., Proc. Natl. Acad. Sci. USA 101 : 10691-96 (2004). Because both PD-L1 and B7.1 are expressed on T cells, B cells, DCs and macrophages, there is the potential for directional interactions between B7.1 and PD-L1 on these cells types.
  • PD-L1 on non-hematopoietic cells may interact with B7.1 as well as PD-l on T cells, raising the question of whether PD-L1 is involved in their regulation.
  • B7.1 PD-L 1 interaction
  • T cell PD-L1 may trap or segregate away APC B7.1 from interaction with CD28.
  • the antagonism of signaling through PD-L1, including blocking PD-L1 from interacting with either PD-l, B7.1 or both, thereby preventing PD-L1 from sending a negative co-stimulatory signal to T-cells and other antigen presenting cells is likely to enhance immunity in response to infection (e.g., acute and chronic) and tumor immunity.
  • An exemplary PD-L1 antagonist is the anti-PD-Ll antibody atezolizumab.
  • Other antagonistic PD-L1 antibodies are durvalumab and avelumab.
  • the anti-PD-Ll /PD 1 interaction can blocked by antagonist anti- PD-l antibodies like the antagonistic PD1 antibodies pembrolizumab or nivolumab or an anti-PD 1 antibody comprising the variable heavy chain and light chain domainss of PD 1 - 0103-0312.
  • human PD-L1 refers to the human protein PD-L1 (SEQ ID NO: 13, PD-l signaling typically).
  • binding to human PD-L1 or “specifically binding to human PD-L1” or“which binds to human PD-L1” or“anti- PD-L1 antibody” or “antagonistic PD-L1” refers to an antibody specifically binding to the human PD-L1 antigen with a binding affinity of KD-value of 1.0 x 10-8 mol/1 or lower, in one embodiment of a KD-value of 1.0 xlO-9 mol/1 or lower.
  • an“antibody binding to human PD-L1” as used herein refers to an antibody specifically binding to the human PD-L1 antigen with a binding affinity of KD 1.0 x 10-8 mol/1 or lower (in one embodiment 1.0 x 10-8 mol/1 - 1.0 x 10-13 mol/1), in on embodiment of a KD 1.0 xlO-9 mol/1 or lower (in one embodiment 1.0 x 10-9 mol/1 - 1.0 x 10-13 mol/1).
  • human PD1 refers to the human protein PD1 (SEQ ID NO: 14, PD-l signaling typically).
  • binding to human PD1 or “specifically binding to human PD1” or“which binds to human PD1” or“anti-PDl antibody” or“antagonistic PD1” refers to an antibody specifically binding to the human PD 1 antigen with a binding affinity of KD-value of 1.0 x 10-8 mol/1 or lower, in one embodiment of a KD-value of 1.0 xlO-9 mol/1 or lower.
  • the binding affinity is determined with a standard binding assay, such as surface plasmon resonance technique (BIAcore®, GE-Healthcare Uppsala, Sweden).
  • an“antibody binding to human PD1” as used herein refers to an antibody specifically binding to the human PD1 antigen with a binding affinity of KD 1.0 x 10-8 mol/1 or lower (in one embodiment 1.0 x 10-8 mo 1/1 - 1.0 x 10-13 mol/1), in on embodiment of a KD 1.0 xlO-9 mol/l or lower (in one embodiment 1.0 x 10-9 mol/1 - 1.0 x 10-13 mol/1).
  • variable domain denotes each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen.
  • the domains of variable human light and heavy chains have the same general structure and each domain comprises four framework (FR) regions whose sequences are widely conserved, connected by three "hypervariable regions” (or complementarity determining regions, CDRs).
  • the framework regions adopt a b-sheet conformation and the CDRs may form loops connecting the b- sheet structure.
  • the CDRs in each chain are held in their three-dimensional structure by the framework regions and form together with the CDRs from the other chain the antigen binding site.
  • the antibody heavy and light chain CDR3 regions play a particularly important role in the binding specificity/affinity of the antibodies according to the invention and therefore provide a further object of the invention.
  • constant region denotes the sum of the domains of an antibody other than the variable region.
  • the constant region is not involved directly in binding of an antigen, but exhibits various effector functions.
  • antibodies are divided in the classes: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses, such as IgGl, IgG2, IgG3, and IgG4, IgAl and IgA2.
  • the heavy chain constant regions that correspond to the different classes of antibodies are called a, d, e, g, and m, respectively.
  • the light chain constant regions which can be found in all five antibody classes are called k (kappa) and l (lambda).
  • constant region derived from human origin or“human constant region” as used in the current application denotes a constant heavy chain region of a human antibody of the subclass IgGl, IgG2, IgG3, or IgG4 and/or a constant light chain kappa or lambda region.
  • constant regions are well known in the state of the art and e.g. described by Kabat, E.A., et ah, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991) (see also e.g. Johnson, G., and Wu, T.T., Nucleic Acids Res.
  • the antagonistic anti-PD 1 antibody which binds to human PD 1 used in the combination therapy described herein is nivolumab or pembrolizumab and is characterized in comprising the following VH and VL sequences as described herein:
  • the antagonistic anti-PD 1 antibody which binds to human PD 1 used in the combination therapy described herein is either a mono- or multispecific antagonistic PD 1 antibody and comprises the following heavy chain variable domain VH and light chain variable domain VL sequences as described herein:
  • such anti-PD 1 antibody based on the heavy chain variable domain VH and light chain variable domain VL sequences of PD 1-0103 -0312 comprises a heavy chain constant region of IgGl subtype (e.g. SEQ ID NO: 16 or SEQ ID NO: 17, eventually also comprising further mutations, see below the bispecific embodiment) and a human kappa light chain constant region (e.g. SEQ ID NO: 15).
  • a heavy chain constant region of IgGl subtype e.g. SEQ ID NO: 16 or SEQ ID NO: 17, eventually also comprising further mutations, see below the bispecific embodiment
  • a human kappa light chain constant region e.g. SEQ ID NO: 15
  • such anti-PD 1 antibody based on the heavy chain variable domain VH and light chain variable domain VL sequences of PD1-0103-0312 is e.g. bispecific and i) the bispecific antibody comprises a constant heavy chain region of human IgGl subclass comprising the mutations L234A, L235A and P329G (numberings according to EU Index of Kabat); and wherein ii)) in the constant heavy chain region a S354C and T366W mutations are comprised in one CH3 domain and Y349C, T366S, L368A and Y407V mutations are comprised the other CH3 domain (numberings according to EU Index of Kabat).
  • the compound of formula I used in the combination therapy described herein is selected from the following:
  • the antagonistic PD 1 antibody used in the combination therapy comprises a heavy chain variable domain VH with an amino acid sequence of SEQ ID NO: 5 and a light chain variable domain VL with an amino acid sequence of SEQ ID NO: 6.
  • the antibody which binds to human PD-L1 used in the combination therapy described herein is atezolizumab or durvalumab or avelumab and is characterized in comprising the following VH and VL sequences as described herein:
  • the antagonistic PD-L1 antibody used in the combination therapy is atezolizumab or durvalumab or avelumab (in one preferred embodiment atezolizumab).
  • Another aspect of the invention is the combined treatment (combination treatment) of a patient suffering from liver cancer with the compound of formula I as described above in combination with an anti-angiogenic agent.
  • the anti- angiogenic agent can be co administered either with compounds of formula I alone or in addition to the combination therapy of the compounds of formula I with an anti-PD-Ll/PDl axis treatment.
  • Antiangiogenic agents as used herein include (but are not limited to) small molecule tyrosine kinase inhibitors (TKIs) that bind competitively to the intracellular receptor domains for VEGF, PDGF, and other angiogenic growth factors, like e.g.
  • TKIs small molecule tyrosine kinase inhibitors
  • sorafenib (4- ⁇ 4- [3-(4-Chlor-3-trifluormethylphenyl)ureido]phenoxy ⁇ pyridin-2-carbonsauremethylamid; NexavarTM), regorafenib (4-[4-( ⁇ [4-Chlor-3-(trifluormethyl)phenyl]carbamoyl ⁇ amino)-3- fluorphenoxy]-N-methylpyridin-2-carboxamid-Hydrat; StivargaTM), and sunitinib (N-[2- (Diethylamino)ethyl]-5-[(Z)-(5-fluor-l,2-dihydro-2-oxo-3H-indol-3-yliden)-methyl]-2,4- dimethyl-lH-pyrrol-3-carboxamid; SutentTM), but include also anti-VEGF or anti-VEGF receptor antibodies like e.g. bevacizumab (AvastinTM).
  • the anti-angiogenic agent used in the combination therapy is sorafenib, regorafenib, sunitinib or bevacizumab (preferably sorafenib or bevacizumab).
  • the compound of formula I used in the combination therapy with an antagonistic PD1 or antagonistic PD-L1 antibody and an anti- angiogenic agent described herein is selected from the following:
  • the antagonistic PD 1 antibody is nivolumab or pembrolizumab or comprises a heavy chain variable domain VH of SEQ ID NO:5 and a light chain variable domain VL of SEQ ID NO: 6;
  • the antagonistic PD-L1 antibody is atezolizumab or durvalumab or avelumab (in one preferred embodiment atezolizumab)
  • the anti-angiogenic agent used in the combination therapy is sorafenib, regorafenib, sunitinib or bevacizumab (preferably sorafenib or bevacizumab).
  • the compound of formula I used in the combination therapy with an antagonistic PD1 or antagonistic PD-L1 antibody and an anti- angiogenic agent described herein is selected from the following:
  • the antagonistic PD 1 antibody is nivolumab or pembrolizumab or comprises a heavy chain variable domain VH of SEQ ID NO:5 and a light chain variable domain VL of SEQ ID NO: 6;
  • the antagonistic PD-L1 antibody is atezolizumab or durvalumab or avelumab (in one preferred embodiment atezolizumab)
  • anti-angiogenic agent used in the combination therapy is sorafenib, regorafenib, sunitinib or bevacizumab (preferably sorafenib or bevacizumab).
  • R 1 is Ci- 6 alkyl
  • R 2 is benzyl, said benzyl being unsubstituted or substituted by one, two or three
  • R 3 is -NR 4 R 5 , wherein
  • R 4 is Ci- 6 alkyl or Ci- 6 alkoxyCi- 6 alkyl
  • R 5 is (Ci-6alkyl)2NCOOCi-6alkyl, Ci- 6 alkoxyCi- 6 alkyl, Ci_
  • compositions or medicament thereof for use in the treatment or prophylaxis of liver cancer; with the proviso that
  • R 1 is Ci- 6 alkyl
  • R 2 is benzyl, said benzyl being unsubstituted or substituted by halogen or Ci- 6 alkyl;
  • R 3 is azetidinyl
  • R 4 is Ci- 6 alkyl or Ci- 6 alkoxyCi- 6 alkyl
  • R 5 is (Ci-6alkyl)2NCOOCi-6alkyl, Ci- 6 alkoxyCi- 6 alkyl, Ci- 6alkoxycarbonyl(Ci- 6 alkyl)aminoCi- 6 alkyl, Ci- 6alkoxycarbonyl(phenyl)Ci- 6 alkyl, Ci- 6 alkoxycarbonylCi- 6 alkyl, Ci- ealkoxycarbonyloxyCi-ealkyl, Ci-ealkyl, Ci_ 6 alkylcarbonyl(Ci_ 6alkyl)aminoCi_ 6 alkyl or pyrrolidinylcarbamoyloxyCi_ 6 alkyl.
  • R 1 is ethyl or propyl
  • R 2 is benzyl, bromobenzyl, chlorobenzyl, fluorobenzyl or methylbenzyl;
  • R 3 is azetidinyl
  • R 4 is methyl, ethyl, propyl or methoxyethyl
  • R 5 is acetyl(methyl)aminoethyl, butyl, butyl(methyl)carbamoyloxyethyl, diethylcarbamoyloxyethyl, ethoxycarbonyl(methyl)aminoethyl, ethoxycarbonylethyl, ethoxycarbonylisobutyl,
  • ethoxycarbonylisopentyl ethoxycarbonylmethyl, ethoxycarbonyloxyethyl, ethoxycarbonyl(phenyl)ethyl, ethyl, isobutyl, isopropoxycarbonylisopentyl, isopropoxycarbonyl(phenyl)ethyl, isopropyl, methoxycarbonyl(methyl)aminoethyl, methoxyethyl, methoxypropyl, propyl, propyl(methyl)carbamoyloxyethyl, pyrrolidinylcarbamoyloxyethyl, tert- butoxycarbonyl(methyl)aminoethyl, tert-butoxycarbonylethyl, tert- butoxycarbonylisopentyl or tert-butoxycarbonyl(phenyl)ethyl. 4. The compound for use according to embodiment 3, wherein R 3 is
  • R 2 is bromobenzyl, chlorobenzyl or fluorobenzyl.
  • R 2 is benzyl, said benzyl being substituted by halogen or Ci- 6 alkyl;
  • R 3 is -NR 4 R 5 , wherein R 4 is Ci- 6 alkyl, R 5 is Ci- 6 alkyl.
  • R 1 is ethyl
  • R 2 is methylbenzyl, bromobenzyl, chlorobenzyl or fluorobenzyl
  • R 3 is propyl(methyl)amino or ethyl(methyl)amino.
  • a compound for use in the treatment or prophylaxis of liver cancer selected from: 6-Amino-9-benzyl-N-methyl-8-oxo-N-propyl-2-(propylsulfonimidoyl)purine-7- carboxamide;
  • the compound is 6-Amino-9-[(4-chloiOphcnyl)mcthyl]-/V-cthyl-2[S(S)- ethylsulfonimidoyl] - /V- m c t hy 1 - 8 - v - p u ri n c - 7 - c a r b o x a m i d c .
  • liver cancer is hepatocellular carcinoma, hepatoma, cholangiocarcinoma, hepatoblastoma, hepatic carcinoma, hepatic angiosarcoma, or metastatic liver cancer.
  • a pharmaceutical composition or medicament comprising a compound according to any one of embodiments 1 to 15 and a therapeutically inert carrier, for use in the treatment or prophylaxis of liver cancer.
  • a method for the treatment or prophylaxis of liver cancer comprises administering a therapeutically effective amount of a compound as defined in any one of embodiments 1 to 15.
  • the treatment is in combination with an antagonistic PD 1 antibody or antagonistic PD-L1 antibody.
  • an anti-angiogenic agent selected from is sorafenib, regorafenib, sunitinib or bevacizumab (in one preferred embodiment the anti- angiogenic agent is sorafenib; in one preferred embodiment the anti- angiogenic agent is bevacizumab) is used in the combination therapy.
  • 35. A compound as defined in any one of embodiments 1 to 15, or a pharmaceutical composition or a medicament comprising such compound for use in the treatment or prophylaxis of liver cancer wherein the treatment is in combination with an anti-angiogenic agent.
  • treatment is in combination with an anti-angiogenic agent.
  • anti-angiogenic agent selected from is sorafenib, regorafenib, sunitinib or bevacizumab (in one preferred embodiment the anti- angiogenic agent is sorafenib; in one preferred embodiment the anti-angiogenic agent is bevacizumab).
  • SEQ ID NO: 14 exemplary human PD 1
  • SEQ ID NO: 16 human heavy chain constant region derived from IgGl
  • SEQ ID NO: 17 human heavy chain constant region derived from IgGl mutated on
  • FIG. 41 (compound 41 -A) induces PD-L1 expression on tumor cells in the iAST mouse model of hepatocellular carcinoma.
  • Figure 2A CD45+ total immune cell infiltrate
  • Figure 2B PD-L1 on CD45-
  • Figure 2C CD1 lb- lymphoid cells
  • Figure 2D CD1 lb+ myeloid cells
  • Figure 6 Treatment with an active form of the compounds of the present invention does not induce enhanced tumor cell proliferation in cell lines originating from hepatocellular carcinoma and cholangiocarcinoma.
  • Figure 6A compound 41 c-B
  • Figure 6B compound 41 c-A
  • Figure 7 7A and 7B: Factors released in peripheral blood upon treatment with an active form of the compounds of the present invention (compound 41c-B) result in inhibition of proliferation in tumor cell lines.
  • Figure 7A Cell lines Hep3B, SNU449, HLF, JHH2, Huh7, OZ, JHH1, HepG2
  • Figure 7B Cell lines JHH4, HLE, JHH6, JHH5, SkHepl, EGI1.
  • 7C Factors released in peripheral blood upon treatment with an active form of the compounds of the present invention (compound 41c-A) result in inhibition of proliferation in tumor cell lines.
  • Figure 8 Single crystal X-ray diffraction of Example 41-B.
  • Figure 9 Single crystal X-ray diffraction of Example 42-A.
  • Figure 10 Single crystal X-ray diffraction of Example 43 -B.
  • CDI /V,/V’-carbonyl diimidazole
  • EC 50 the molar concentration of an agonist, which produces
  • EDC Al-((ethylimino)methylene)-A3,A3-dimethylpropane- 1,3- diamine
  • HOBt TV-hydroxybenzotri azole
  • PE petroleum ether
  • PPA polyphosphoric acid
  • Acidic condition A: 0.1% formic acid and 1% acetonitrile in FLO; B: 0.1% formic acid in acetonitrile;
  • Mass spectra generally only ions which indicate the parent mass are reported, and unless otherwise stated the mass ion quoted is the positive mass ion (M+H) + .
  • NMR Spectra were obtained using Bruker Avance 400MHz. All reactions involving air-sensitive reagents were performed under an argon atmosphere. Reagents were used as received from commercial suppliers without further purification unless otherwise noted.
  • Intermediate AC was prepared in analogy to Intermediate AA by using A- ethylpropan- 1 -amine instead of A-methylpropan- 1 -amine.
  • A-Ethyl-A-propyl-carbamoyl chloride (12.6 g, Intermediate AC) was obtained as a yellow oil and used for next step without further purification.
  • Intermediate AD was prepared in analogy to Intermediate AA by using A-ethyl-2- methoxyethanamine instead of A-methylpropan- 1 -amine.
  • the crude /V-cthyl-/V-(2- methoxyethyl)carbamoyl chloride (2.5 g, Intermediate AD) was obtained as a light yellow oil and used for next step without further purification.
  • Intermediate AE was prepared in analogy to Intermediate AA by using N- ethylbutan- 1 -amine (5 g) instead of A-methylpropan- 1 -amine.
  • the crude /V-butyl-/V-cthyl- carbamoyl chloride (6.3 g, Intermediate AE) was obtained as a light yellow oil and used for next step without further purification.
  • Intermediate AG was prepared in analogy to Intermediate AA by using of bis(2- methoxyethyl)amine (2 g, 15 mmol) instead of /V-mcthylpropan- 1 -amine.
  • the crude product /V, /V- b i s ( 2 - m c t h o x y c t h y I ) c a rb a m o y I chloride (2.6 g, Intermediate AG) was obtained as a light yellow oil and used for next step without further purification.
  • Intermediate AH was prepared in analogy to Intermediate AA by using azetidine hydrochloride (10.7 g, 107 mmol) and sodium bicarbonate (3 equiv.) instead of N- methylpropan- 1 -amine and sodium bicarbonate (2 equiv.).
  • the crude azetidine-l-carbonyl chloride (1.5 g, Intermediate AH) was obtained as a light yellow oil and used for next step without further purification.
  • Step 2 Preparation of tert- butyl 3-[chlorocarbonyl(methyl)amino]propanoate (Intermediate AR)
  • Step 1 Preparation of ethyl (25)-2-(methylamino)propanoate hydrochloride (Compound AS-1)
  • Step 1 Preparation of tert- butyl (25)-4-methyl-2-(methylamino)pentanoate
  • Step 2 Preparation of tert- butyl (25)-2-[chlorocarbonyl(methyl)amino]-4-methyl- pentanoate (Intermediate AT)
  • Step 1 Preparation of isopropyl (25)-4-methyl-2-(methylamino)pentanoate hydrochloride (Compound AU-1)
  • Intermediate AU was prepared in analogy to Intermediate AP by using isopropyl (25 ' )-4-mcthyl-2-(mcthylamino)pcntanoatc hydrochloride (500 mg, Compound AU-1) instead of ethyl 2-(methylamino)acetate hydrochloride.
  • the crude isopropyl (2S)-2- [chlorocarbonyl(methyl)amino]-4-methyl-pentanoate (650 mg, Intermediate AU) was obtained as a light yellow oil and used for the next step without further purification.
  • Step 1 Preparation of ethyl (25)-3-methyl-2-(methylamino)butanoate hydrochloride (Compound AV-1)
  • Step 2 Preparation of ethyl (25)-2-[chlorocarbonyl(methyl)amino]-3-methyl- butanoate (Intermediate AV)
  • Step 2 Preparation of ethyl (25)-2-[chlorocarbonyl(methyl)amino]-4-methyl- pentanoate (Intermediate AW)
  • Intermediate AX was prepared in analogy to Intermediate AP by using (S)-cthyl- 2-(methylamino)-3-phenylpropanoate instead of ethyl 2-(methylamino)acetate hydrochloride.
  • the crude ethyl (25)-2-[chlorocarbonyl(methyl)amino] -3-phenyl- propanoate (200 mg, Intermediate AX) was obtained as a light yellow oil and used for the next step without further purification
  • Intermediate AY was prepared in analogy to Intermediate AP by using isopropyl (25)-2-(methylamino)-3-phenyl-propanoate (190 mg) instead of ethyl 2- (methylamino) acetate hydrochloride.
  • the crude isopropyl (2S)-2- [chlorocarbonyl(methyl)amino] -3-phenyl-propanoate (220 mg, Intermediate AY) was obtained as light brown oil and used for the next step without further purification.
  • Step 1 Preparation of tert- butyl (25)-2-(methylamino)-3-phenyl-propanoate
  • AX-1 2-Methylpropene (25 g, 446 mmol) was bubbled into DCM (50 mL) at -78 °C. Then the 2-methylpropene solution was added to a solution of (5 ' )-2-(mcthylamino)-3- phenylpropanoic acid (500 mg) and H 2 SO 4 (3.68 g, 2 mL) in dioxane (20 mL) at 0 °C. The reaction mixture was stirred at room temperature for 18 hrs in a sealed tube. The reaction mixture was poured into an ice cold aqueous KOH solution (8.4 g in water (30 mL)) and the resulting mixture was extracted with DCM (50 mL) twice.
  • Step 2 Preparation of (5) -tert- butyl 2-((chlorocarbonyl)(methyl)amino)-3- phenylpropanoate (Intermediate AZ)
  • Intermediate AZ was prepared in analogy to intermediate AP by using /er/-butyl (2S)-2-(mcthylamino)-3-phcnyl-propanoatc (Compound AZ-1) instead of ethyl 2- (methylamino) acetate hydrochloride.
  • the crude /er/-butyl (2S)-2- [chlorocarbonyl(methyl)amino]-3-phenyl-propanoate 360 mg, Intermediate AZ
  • Step 1 Preparation of tert-butyl /V-[2-[acetyl(methyl)amino]ethyl]-/V-methyl- carbamate (Compound BA-1)
  • Step 3 Preparation of A-[2-[acetyl(methyl)amino]ethyl]-A-methyl-carbamoyl chloride (Intermediate BA)

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
EP19708981.6A 2018-02-28 2019-02-26 7-substituted sulfonimidoylpurinone compounds and derivatives for the treatment and prophylaxis of liver cancer Pending EP3758707A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2018077501 2018-02-28
PCT/EP2019/054729 WO2019166432A1 (en) 2018-02-28 2019-02-26 7-substituted sulfonimidoylpurinone compounds and derivatives for the treatment and prophylaxis of liver cancer

Publications (1)

Publication Number Publication Date
EP3758707A1 true EP3758707A1 (en) 2021-01-06

Family

ID=65685310

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19708981.6A Pending EP3758707A1 (en) 2018-02-28 2019-02-26 7-substituted sulfonimidoylpurinone compounds and derivatives for the treatment and prophylaxis of liver cancer

Country Status (19)

Country Link
EP (1) EP3758707A1 (zh)
JP (1) JP7089596B2 (zh)
KR (1) KR20200128414A (zh)
CN (1) CN111801100B (zh)
AR (1) AR114419A1 (zh)
AU (1) AU2019228654A1 (zh)
BR (1) BR112020016509A2 (zh)
CA (1) CA3091950A1 (zh)
CL (1) CL2020002139A1 (zh)
CO (1) CO2020010306A2 (zh)
IL (1) IL276817A (zh)
MA (1) MA52412A (zh)
MX (1) MX2020008746A (zh)
PE (1) PE20211456A1 (zh)
PH (1) PH12020551343A1 (zh)
RU (1) RU2020131012A (zh)
SG (1) SG11202008291XA (zh)
TW (1) TW202003518A (zh)
WO (1) WO2019166432A1 (zh)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JOP20170169A1 (ar) 2016-08-29 2019-01-30 Novartis Ag مركبات بيريدازين ثلاثية الحلقة مندمجة تفيد في علاج العدوى بفيروس أورثوميكسو
JP7065989B2 (ja) 2018-02-28 2022-05-12 ノバルティス アーゲー インフルエンザを処置するためのオルソミクソウイルス複製の阻害剤としての10-(ジ(フェニル)メチル)-4-ヒドロキシ-8,9,9a,10-テトラヒドロ-7H-ピロロ[1’,2’:4,5]ピラジノ[1,2-b]ピリダジン-3,5-ジオン誘導体および関連化合物
KR102409595B1 (ko) 2020-06-29 2022-06-17 한국과학기술연구원 단백질 카이네이즈 csf-1r 억제제로서의 신규 퓨리논 유도체
CN112420196A (zh) * 2020-11-20 2021-02-26 长沙市弘源心血管健康研究院 急性心肌梗死患者5年内生存率的预测方法和系统
WO2024013205A1 (en) * 2022-07-14 2024-01-18 F. Hoffmann-La Roche Ag Phosphorylpurinone compounds for the treatment of cancer

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4189048B2 (ja) * 1997-12-26 2008-12-03 大日本住友製薬株式会社 複素環化合物
TW201639852A (zh) 2008-12-09 2016-11-16 吉李德科學股份有限公司 用於製備可用作類鐸受體調節劑之化合物的中間體化合物
CA2760766A1 (en) * 2009-05-21 2010-11-25 Nicholas James Bennett Novel pyrimidine derivatives and their use in the treatment of cancer and further diseases
US8962652B2 (en) * 2009-10-22 2015-02-24 Gilead Sciences, Inc. Derivatives of purine or deazapurine useful for the treatment of (inter alia) viral infections
US20110150836A1 (en) * 2009-12-22 2011-06-23 Gilead Sciences, Inc. Methods of treating hbv and hcv infection
ES2691671T3 (es) 2010-06-24 2018-11-28 Alkermes Pharma Ireland Limited Profármacos de compuestos NH-acídicos: derivados de éster, carbonato, carbamato y fosfonato
US20120039916A1 (en) * 2010-08-13 2012-02-16 Baylor Research Institute Novel vaccine adjuvants based on targeting adjuvants to antibodies directly to antigen-presenting cells
SI3294740T1 (sl) * 2015-05-08 2019-12-31 F. Hoffmann-La Roche Ag Nove sulfonimidoilpurinonske spojine in derivati za zdravljenje virusne okužbe in zaščito pred njo
MA44334A (fr) 2015-10-29 2018-09-05 Novartis Ag Conjugués d'anticorps comprenant un agoniste du récepteur de type toll
WO2018041763A1 (en) 2016-08-29 2018-03-08 F. Hoffmann-La Roche Ag 7-substituted sulfonimidoylpurinone compounds for the treatment and prophylaxis of virus infection
JP7101663B2 (ja) * 2016-09-13 2022-07-15 エフ.ホフマン-ラ ロシュ アーゲー Tlr7アゴニストとhbvキャプシドアセンブリ阻害剤の併用療法

Also Published As

Publication number Publication date
IL276817A (en) 2020-10-29
AR114419A1 (es) 2020-09-02
MA52412A (fr) 2021-06-02
BR112020016509A2 (pt) 2020-12-15
CO2020010306A2 (es) 2020-08-31
PH12020551343A1 (en) 2021-06-21
PE20211456A1 (es) 2021-08-05
KR20200128414A (ko) 2020-11-12
WO2019166432A1 (en) 2019-09-06
CN111801100A (zh) 2020-10-20
JP7089596B2 (ja) 2022-06-22
AU2019228654A1 (en) 2020-09-03
MX2020008746A (es) 2020-09-28
RU2020131012A (ru) 2022-03-28
SG11202008291XA (en) 2020-09-29
CN111801100B (zh) 2023-10-24
CA3091950A1 (en) 2019-09-06
TW202003518A (zh) 2020-01-16
JP2021514972A (ja) 2021-06-17
CL2020002139A1 (es) 2021-01-04

Similar Documents

Publication Publication Date Title
EP3758707A1 (en) 7-substituted sulfonimidoylpurinone compounds and derivatives for the treatment and prophylaxis of liver cancer
JP6688352B2 (ja) 補体媒介障害の治療のためのアリール化合物、ヘテロアリール化合物及び複素環式化合物
CA2774145C (en) Hcv protease inhibitors
KR102459155B1 (ko) 바이러스 감염의 치료 및 예방을 위한 7-치환된 설폰이미도일푸린온 화합물
TWI476201B (zh) 抗病毒治療用的吡唑並[1,5-a]嘧啶類
WO2018129287A1 (en) Heteroaryldiazepine derivatives as rsv inhibitors
JP2019513129A (ja) B型肝炎抗ウイルス剤
WO2019097479A1 (en) Novel dihydroisoxazole compounds and their use for the treatment of hepatitis b
US20200140383A1 (en) 2-(4-chlorophenoxy)-n-((1 -(2-(4-chlorophenoxy)ethynazetidin-3-yl)methyl)acetamide derivatives and related compounds as atf4 inhibitors for treating cancer and other diseases
WO2019008506A1 (en) N- (3- (2- (4-CHLOROPHENOXY) ACETAMIDO) BICYCLO [1.1.1] PENTAN-1-YL) -2-CYCLOBUTANE-1-CARBOXAMIDE DERIVATIVES AND RELATED COMPOUNDS AS ATF4 INHIBITORS FOR THE TREATMENT OF CANCER AND OTHER DISEASES
JP7139568B2 (ja) ジヒドロピリミジン化合物並びにその調製方法及び使用
CA3133300A1 (en) Benzodiazepine derivatives as rsv inhibitors
US20200268762A1 (en) 7-substituted sulfonimidoylpurinone compounds and derivatives for the treatment and prophylaxis of liver cancer
WO2022166923A1 (zh) 苯基二氢嘧啶类化合物及其应用
EP3433257A1 (en) Alkynyl nucleoside analogs as inhibitors of human rhinovirus
WO2015193229A1 (de) Bet-proteininhibitorische 1,4-dihydropyrido[3,4-b]pyrazinone mit meta-substituierter aromatischer amino- oder ethergruppe
IL296182A (en) Inhibitors of human immunodeficiency virus replication
TW202409011A (zh) 人類呼吸道融合病毒及間質肺病毒抑制劑
CN118574834A (zh) 稠合的四环化合物、其组合物和用途

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20200928

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAX Request for extension of the european patent (deleted)
RAV Requested validation state of the european patent: fee paid

Extension state: MA

Effective date: 20200928

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20230629