WO2016159745A1 - Ethynylxanthines, preparation and use for cancer treatment - Google Patents

Ethynylxanthines, preparation and use for cancer treatment Download PDF

Info

Publication number
WO2016159745A1
WO2016159745A1 PCT/LV2015/000001 LV2015000001W WO2016159745A1 WO 2016159745 A1 WO2016159745 A1 WO 2016159745A1 LV 2015000001 W LV2015000001 W LV 2015000001W WO 2016159745 A1 WO2016159745 A1 WO 2016159745A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
purine
dione
amino
alkoxy
Prior art date
Application number
PCT/LV2015/000001
Other languages
French (fr)
Inventor
Pavels Arsenjans
Jelena VASILJEVA
llona DOMRACHEVA
Irina Shestakova
Anita GULBE
Iveta KANEPE-LAPSA
Valerjans Kauss
Ivars Kalvins
Original Assignee
Latvian Institute Of Organic Synthesis
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 Latvian Institute Of Organic Synthesis filed Critical Latvian Institute Of Organic Synthesis
Priority to GB1714548.3A priority Critical patent/GB2553684B/en
Priority to PCT/LV2015/000001 priority patent/WO2016159745A1/en
Priority to CA2989161A priority patent/CA2989161C/en
Publication of WO2016159745A1 publication Critical patent/WO2016159745A1/en

Links

Classifications

    • 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
    • C07D473/04Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms
    • C07D473/06Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3
    • C07D473/12Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3 with methyl radicals in positions 1, 3, and 7, e.g. caffeine
    • 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
    • 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
    • C07D473/04Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms
    • C07D473/06Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3
    • 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
    • C07D473/04Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms
    • C07D473/06Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3
    • C07D473/10Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3 with methyl radicals in positions 3 and 7, e.g. theobromine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention relates to novel ethynylxanthine derivatives, methods for their synthesis and use for the treatment and/or prevention of cancer.
  • Cancer is one of the main causes of death among economically developed countries (Jermal, CA Cancer J. Clin., 2011, 61 69-90). According to the International Health Organization, more than 7 million people diagnosed with various forms of cancer, die each year (Jermal, CA Cancer J. Clin., 2010, 60, 277-300; Siegel, CA Cancer J. Clin. 2012, 62, 10-29). Unfortunately, the number of cancer diagnosis in past few years has increased almost twice. In clinical practice, the treatment of cancer involves a wide range of chemotherapy drugs. Besides, most of them exhibit various side effects, high toxicity and moderate selectivity. Therefore, a new generation of selective, low toxic anticancer agent development is one of the main tasks in medicinal chemistry and pharmaceutical industries.
  • xanthine derivatives are able to cross through the blood brain barrier (BBB).
  • BBB blood brain barrier
  • anticancer drugs based on the structure of the natural purine analogues were developed (cladribine, fludarabine, mercaptopurine, thioguanine, clofarabine, nelai'abine, etc.). These are the first line therapy agents to cure hematologic malignant diseases.
  • Caffeine derivatives possess CNS expression as calcium agonist or antagonist effect. Recent studies show that caffeine-containing coffee daily use is able to lower mouth and brain cancer formation up to 39% (Michaud, Am. J. Clin. Nutr. 2010, 92, 1145-50; Kang, Cancer. Res. 2010, 70, 1173-83). Also, it reduces the risk of women cervical cancer development. Caffeine reduces a chance of prostate cancer formation in men by 60%. The same effect was observed in ability to prevent breast, colon and hepatic cancer (Hepatology, 2007, 46, 430-435).
  • Caffeine enhances doxorubicin, cisplatin activity in metastatic carcinomas, lymphomas, bone and soft tissue sarcomas (Hayashi, Anticancer Res., 2005, 25, 2399-2406). Also, caffeine effectively inhibits breast cancer resistance protein (BCRP) multidrug resistance (MDR) on MCF-7 and MCF-7/MX100 (mitoxantrone-resistant) cell lines.
  • BCRP breast cancer resistance protein
  • MDR multidrug resistance
  • MCF-7 and MCF-7/MX100 mitoxantrone-resistant
  • Drugs used in neuro-oncology have a limited ability to cross through the BBB and are highly toxic.
  • caffeine analogues such as 8-(3- (dimethylamino)propoxy)caffeine (proxyfeine)
  • proxyfeine 8-(3- (dimethylamino)propoxy)caffeine
  • Proxyfeine (RU 2166948, 20.05.2001) is used in chemotherapy for cancer patients at high risk of brain metastases and the rehabilitation of the metastatic lesions to the brain, as well as the early stages of cancer metastasis prevention in Russia and other countries.
  • PCT Patent application No. WO2008077557 discloses preparation of 8-ethynyl xanthine derivatives as selective A2A receptor antagonists and their use as medicines, for example, in the treatment of dopamine-related movement disorders,
  • Ri and R 2 represent, e.g., hydrogen, Q ⁇ a-kyl, cycloalkyl, heterocycloalkyl, aryl (wherein these groups may be further substituted), etc.; R 3 represents e.g., aryl, hetaryl group.
  • ethynylxanthine derivatives exhibit high antiproliferative activity against various tumor cell lines. Therefore, these substances may be therapeutically beneficial in the treatment of tumors.
  • These substances may be administered in the form of a pharmaceutical composition, wherein they are present together with one or more pharmaceutically acceptable diluents, carriers, or excipients.
  • R 1 represents hydrogen, C ⁇ alkyl, hydroxy-C 2 - 4 alkyl, Ci -3 alkoxy-C 2 - 4 alkyl, Ci. salkylcarbonyl-C ⁇ alkyl or C 1-3 alkyl(C 1 _3alkyl)amino-C 2-4 alkyl;
  • R 2 represents C ⁇ alkyl, hydroxy-C 2 . 4 alkyl, C 1-4 alkylcarbonyl-Ci- 4 alkyl, C]. 3 alko y-C 2 - 4 alkyl, Ci -3 alkyl(C 1 _ 3 alkyl)amino-C 2 - 4 alkyl or halo-C 2 . 4 alkyl;
  • R 3 represents Ci_ 4 alkyl, allyl or Ci- 3 alkoxy-C 2-4 alkyl; with the proviso that if substituent R 3 is at purine N(7) atom the dotted line between N(7) and C(8) represents no bond, and the dotted line between C(8) and N(9) represents chemical bond;
  • R 3 represents Q ⁇ alkyl, hydroxy-C 1-4 alkyl, C i .. 3 alkoxy-C i _ 4 alkyl, amino-C
  • heterocyclyl represents saturated 4-7 membered heterocycle containing one or two heteroatoms selected from oxygen, sulfur and nitrogen, wherein the heterocyclyl may be azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, tetrahydrofuryl, morpholinyl, thiomorpholinyl and piperazinyl;
  • aryl represents phenyl or phenyl substituted by one or more substituents selected independently from halogen, cyano, C 1 _ 4 alkoxycarbonyl, N-Q.
  • the invention also relates to a manufacturing process of a compound selected from those of Formula I as defined above, comprising reaction of a compound of Formula II:
  • R l , R 2 and R 3 are as defined for Formula I above, with a compound of Formula III:
  • R 4 is as defined for Formula I above, optionally in the presence of base in an appropriate solvent (e.g., DIEA in DMF, NMP, DMAC or EtOAc), in the presence of Cul and palladium catalyst generated in situ (e.g., from PdCl 2 or Pd(OAc) 2 and PPh 3 ) or commercially available (Ph 3 P) 4 Pd to yield a compound of Formula I, which may be converted, if desired, into an optical isomer, polymorph, pharmaceutically-acceptable salt, hydrate or solvate.
  • an appropriate solvent e.g., DIEA in DMF, NMP, DMAC or EtOAc
  • Cul and palladium catalyst generated in situ e.g., from PdCl 2 or Pd(OAc) 2 and PPh 3
  • Ph 3 P commercially available
  • Ci. 4 alkyl represents straight or branched chain alkyl groups having 1, 2, 3 or 4 carbon atoms, examples of such alkyl groups include methyl, ethyl, n-propyl, 2-propyl, n-butyl, 2-butyl, iso-butyl and tert-butyl.
  • cyclo-C 3-6 alkyl represents monocyclic alkyl groups having 3, 4, 5 or 6 carbon atoms, including cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • heterocyclyl represents a saturated 4-7 membered heterocycle containing one or two heteroatoms selected from oxygen, sulfur and nitrogen
  • heterocyclyl groups include azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, tetrahydrofuryl, morpholinyl, thiomorpholinyl and piperazinyl.
  • halo or halogen represents fluorine, chlorine, bromine and iodine.
  • analogs and derivatives of the compounds of the invention can be created which have improved therapeutic efficacy, i.e., higher potency and/or selectivity at a specific targeted receptor type, greater ability to penetrate mammalian blood-brain barriers, fewer side effects, etc.
  • the compounds of Formula I can be in the form of a pharmaceutically acceptable salt or a solvate.
  • pharmaceutically acceptable refers here to the therapeutically active non-toxic salt forms, which the compounds of Formula I are able to form.
  • the latter can conveniently be obtained by treating the base form with such appropriate acids as inorganic acids such as hydrochloric acid, hydrobromic acid; sulfuric acid; nitric acid; phosphoric acid and the like; or organic acids such as acetic, propanoic, hydroxyacetic, 2-hydroxypropanoic, oxopropanoic, oxalic, malonic, succinic, maleic, fumaric, malic, tartaric, methanesulfonic, benzenesulfonic, 4-methylbenzenesulfonic, 2-hydroxybenzoic, and like acids.
  • the salt may be converted to the free base by treatment with alkali.
  • Scheme 1 describes the preparation of compounds of Formula I of the present invention. All of the starting materials II are prepared by representative procedures described in Schemes 2 and 3, by procedures well known to one of ordinary skill in organic chemistry or can be obtained commercially. All of the final compounds of the present invention are prepared by procedures described in these charts or by procedures analogous thereto, which procedures would be well known to one of ordinary skill in organic chemistry. All of the variables used in the schemes are as defined below or as in the claims.
  • Method C Pd(OAc) 2 , Ph 3 P, Cul, N-methylpyrrolidine/toluene (1:1), DIEA, 50 °C; Method D. Pd(PPh 3 ) 4 , PdCl 2 , Ph 3 P, Cul, ethyl acetate, DIEA, 40 °C.
  • DMF is defined as ⁇ , ⁇ -dimethylformamide
  • DMAC is defined as ⁇ , ⁇ -dimethylacetamide
  • NMP is defined as N-methylpyrrolidone
  • DMSO dimethyl sulfoxide
  • HCl as hydrochloric acid
  • NH 3 as aqueous ammonia solution
  • MeCN acetonitrile
  • EtOAc diisopropylethylamine
  • rt room temperature
  • Method D A vial charged with Pd(PPh 3 ) 4 (346 mg, 0.3 mmol), PdCl 2 (51 mg, 0.3 mmol), Ph 3 P (157 mg, 0.6 mmol), Cul (58 mg, 0.3 mmol), 8-bromo- 1,3,7- trimethyl-lH-purine-2,6(3H,7H)-dione (4.08 g, 15.0 mmol) and 2-methylbut-3-yn-2- ol (2.05 mL, 21.0 mmol), DIEA (5.0 mL) and ethyl acetate (70 mL) was stirred for 15 min at 40 °C with simultaneous barbotation with argon.
  • Pd(PPh 3 ) 4 346 mg, 0.3 mmol
  • PdCl 2 51 mg, 0.3 mmol
  • Ph 3 P 157 mg, 0.6 mmol
  • Cul 58 mg, 0.3 mmol
  • reaction mixture was stirred for additional 2-4 h. After cooling to rt, the reaction mixture was filtered through a silica gel pad and washed with EtOAc (200 mL). Then solvent was evaporated under reduced pressure. The crude product was purified by flash chromatography on silica gel using mixture of water (containing 0.1% aq.
  • MDA-MB-435s human melanoma
  • H9C2 rat embrio cardiomyoblast
  • MCF-7 human breast adenocarcinoma, estrogen-positive
  • HepG2 human hepatocellular carcinoma
  • SH-SY5Y human neuroblastoma
  • C6 rat glioma
  • U937 human histiocytic leukemia
  • A549 human lung carcinoma
  • Monolayer tumor cell line MDA-MB-435s (human melanoma), H9C2 (rat embrio cardiomyoblast), MCF-7 (human breast adenocarcinoma, estrogen-positive), HepG2 (human hepatocellular carcinoma), SH-SY5Y (human neuroblastoma), C6 (rat glioma), U937 (human histiocytic leukemia), and A549 (human lung carcinoma), and normal cell line NIH 3T3 (mouse fibroblasts) were cultured in standard medium DMEM (Dulbecco's modified Eagle's medium) containing 1% non-essential amino- acids, 2 niM glutamine and supplemented with 10% fetal bovine serum (FBS, Sigma) ("Sigma").
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • NIH 3T3 normal mouse fibroblasts, "ATCC” cell line according the basal toxicity test (INVITOX Protocol No 64, 1992) and non-toxic compounds were selected.
  • NIH 3T3 cells/well were placed into 96-well plates for 24 h and then exposed to the test compound over a range of eight concentration (1- 1000 ⁇ g/mL) for 24 h. Upon that, the cells were incubated with the neutral red dye for 4 h and then OD was determined at 540 nm.
  • LD50 value is the amount of the drug that is taken to kill 50% of the test animals
  • the IC 5 o values were calculated using the program Graph Pad 5 Prism® 3.0.
  • MDA-MB-435s human melanoma
  • H9C2 rat embrio cardiomyoblast
  • MCF-7 human breast adenocarcinoma, estrogen-positive
  • HepG2 human hepatocellular carcinoma
  • SH- SY5Y human neuroblastoma
  • C6 rat glioma
  • U937 human histiocytic leukemia
  • A549 human lung carcinoma

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to novel ethynylxanthine derivatives of formula (I) which exhibit high antiproliferative activity against various tumor cell lines, methods for their synthesis and use for the treatment and/or prevention of cancer.

Description

ETHYNYLXA THINES, PREPARATION AND USE FOR CANCER TREATMENT
FIELD OF THE INVENTION
The present invention relates to novel ethynylxanthine derivatives, methods for their synthesis and use for the treatment and/or prevention of cancer.
BACKGROUND OF THE INVENTION
Cancer is one of the main causes of death among economically developed countries (Jermal, CA Cancer J. Clin., 2011, 61 69-90). According to the International Health Organization, more than 7 million people diagnosed with various forms of cancer, die each year (Jermal, CA Cancer J. Clin., 2010, 60, 277-300; Siegel, CA Cancer J. Clin. 2012, 62, 10-29). Unfortunately, the number of cancer diagnosis in past few years has increased almost twice. In clinical practice, the treatment of cancer involves a wide range of chemotherapy drugs. Besides, most of them exhibit various side effects, high toxicity and moderate selectivity. Therefore, a new generation of selective, low toxic anticancer agent development is one of the main tasks in medicinal chemistry and pharmaceutical industries. Despite the advanced studies in the elaboration of anticancer drugs, the treatment outcome for brain malignant tumors still remains a challenge. The majority of promising antitumor drugs used against brain tumors in clinical trials, unfortunately, had limited impact on human survival, due to inadequacy of efficient drug delivery to the target in the central nervous system (CNS) at a sufficient concentration (Siegal, Neuro Oncol, 2013 10.1093/neuonc/not016; Lampson, Drug Discov. Today, 2009, 14, 185-191; Huse, Nat. Rev., 2010, 10, 319- 331).
Increased interest in xanthines stems from the fact that this heterocyclic system occurs in a number of natural substances; xanthine derivatives are able to cross through the blood brain barrier (BBB). It is an important class of compounds with a wide range of pharmacological effects, including anticancer, anti-HIV, anticoagulant, antispasmodic and antibacterial activity. Nowadays, in clinical practice, a number of anticancer drugs based on the structure of the natural purine analogues were developed (cladribine, fludarabine, mercaptopurine, thioguanine, clofarabine, nelai'abine, etc.). These are the first line therapy agents to cure hematologic malignant diseases. Agents in the therapeutic effect of complete remission constitute 80% after monochemotherapy course. Such compounds act as antimetabolites by replacing the natural nucleoside in DNA and RNA synthesis such as multi-cellular enzyme inhibitors. Unfortunately, these drugs showed a wide range of side effects, and high treatment costs limit the possibilities in clinical practice.
To minimize side effects currently being developed antitumor drugs based on caffeine core. Caffeine derivatives possess CNS expression as calcium agonist or antagonist effect. Recent studies show that caffeine-containing coffee daily use is able to lower mouth and brain cancer formation up to 39% (Michaud, Am. J. Clin. Nutr. 2010, 92, 1145-50; Kang, Cancer. Res. 2010, 70, 1173-83). Also, it reduces the risk of women cervical cancer development. Caffeine reduces a chance of prostate cancer formation in men by 60%. The same effect was observed in ability to prevent breast, colon and hepatic cancer (Hepatology, 2007, 46, 430-435). Caffeine enhances doxorubicin, cisplatin activity in metastatic carcinomas, lymphomas, bone and soft tissue sarcomas (Hayashi, Anticancer Res., 2005, 25, 2399-2406). Also, caffeine effectively inhibits breast cancer resistance protein (BCRP) multidrug resistance (MDR) on MCF-7 and MCF-7/MX100 (mitoxantrone-resistant) cell lines. One of the advantages of caffeine derivatives action is the ability to cross through the BBB. It opens the possibility to cure malignant diseases, such as neuroblastoma and glioblastoma multiforme, in the brain (Vartanyan, Psychopharm. Biol. Narc., 2005, 5, 1093-1095). Drugs used in neuro-oncology (temodar, carmustine (BCNU), lomusthine (CCNU), etc.) have a limited ability to cross through the BBB and are highly toxic. In a series of caffeine analogues, such as 8-(3- (dimethylamino)propoxy)caffeine (proxyfeine), antitumor agents were developed, however introduction in the clinic of the EU and the US interferes, due to high levels of toxicity (LD50=355 mg/kg), as well as a large number of serious side effects. Proxyfeine (RU 2166948, 20.05.2001) is used in chemotherapy for cancer patients at high risk of brain metastases and the rehabilitation of the metastatic lesions to the brain, as well as the early stages of cancer metastasis prevention in Russia and other countries.
Figure imgf000004_0001
Proxyfeine
PCT Patent application No. WO2008077557 discloses preparation of 8-ethynyl xanthine derivatives as selective A2A receptor antagonists and their use as medicines, for example, in the treatment of dopamine-related movement disorders,
Figure imgf000004_0002
8-Ethynyl xanthines wherein Ri and R2 represent, e.g., hydrogen, Q^a-kyl, cycloalkyl, heterocycloalkyl, aryl (wherein these groups may be further substituted), etc.; R3 represents e.g., aryl, hetaryl group.
PCT Patent application No. WO2014/143799 A2, 2014 discloses preparation of N7- benzyl 8-ethynyl xanthine derivatives as agents for treatment of short transient receptor potential channel 5 (TrpC5) disorders A2A receptor antagonists and their use as medicines, for example, in the treatment of dopamine-related movement disorders.
Figure imgf000004_0003
THE PRESENT INVENTION
We have discovered that certain ethynylxanthine derivatives exhibit high antiproliferative activity against various tumor cell lines. Therefore, these substances may be therapeutically beneficial in the treatment of tumors. These substances may be administered in the form of a pharmaceutical composition, wherein they are present together with one or more pharmaceutically acceptable diluents, carriers, or excipients. OBJECTS OF THE INVENTION
It is an object of the present invention to provide novel antiproliferative compounds, methods for their synthesis and use for treatment and/or prevention cancer.
SUMMARY OF THE INVENTION
What we therefore believe to be comprised by our invention may be summarized inter alia in the following words:
A compound selected from those of Formula I
Figure imgf000005_0001
wherein
R1 represents hydrogen, C^alkyl, hydroxy-C2-4alkyl, Ci-3alkoxy-C2-4alkyl, Ci. salkylcarbonyl-C^alkyl or C1-3alkyl(C1_3alkyl)amino-C2-4alkyl;
R2 represents C^alkyl, hydroxy-C2.4alkyl, C1-4alkylcarbonyl-Ci-4alkyl, C].3alko y-C2- 4alkyl, Ci-3alkyl(C1_3alkyl)amino-C2-4alkyl or halo-C2.4alkyl;
R3 represents Ci_4alkyl, allyl or Ci-3alkoxy-C2-4alkyl; with the proviso that if substituent R3 is at purine N(7) atom the dotted line between N(7) and C(8) represents no bond, and the dotted line between C(8) and N(9) represents chemical bond;
and
with the proviso that if substituent R3 is at purine N(9) atom the dotted line between N(7) and C(8) represents chemical bond, and the dotted line between C(8) and N(9) represents no bond; R4 represents Q^alkyl, hydroxy-C1-4alkyl, C i ..3alkoxy-C i _4alkyl, amino-C|,4alkyl, 1- hydroxy-di-(Ci.3alkyl)methyl, 1 -amino-di-f C i . jalkyl )methyl, l-hydiOxy- cyclo-Cj_ 6alkyl, l-aniino~cyclo-C3_6alkyl, 1 -(hydroxy-Ci _3alkyl)-cycloC3-6alkyl, C\. 3alkylamino-Ci-3alkyl.
Figure imgf000006_0001
di-(Ci_3alkoxy~C2-4alkyl)- amino-Ci-3alkyK heterocyclyl-C i .salkyl , aryl or heteroaryl; wherein the term "heterocyclyl" represents saturated 4-7 membered heterocycle containing one or two heteroatoms selected from oxygen, sulfur and nitrogen, wherein the heterocyclyl may be azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, tetrahydrofuryl, morpholinyl, thiomorpholinyl and piperazinyl; the term "aryl" represents phenyl or phenyl substituted by one or more substituents selected independently from halogen, cyano, C1_4alkoxycarbonyl, N-Q. 4alkylaminocarbonyl, N,N-di-(Ci_3aikyl)aminocarbonyl, CH2OH, trifluoromethyl, Q. 4alkyl, allyl, C2-4alkynyl, C1-4alkoxy, difluoromethoxy, trifluoromethoxy, cyclo- C3-6alkoxy, hydroxy-C1-4alkyl, Ci^alkoxy-Ci^alkyl, C1_3alkoxy-C2_4alkoxy, di-(Ci_ 3alkyl)amino, di-CCi.salky amino-Ct-salkyl, di-(C1-3alkyl)amino-C2-4alkoxy, C1-4alkylsulfonylamino and d-^lkyl-aminosulfonyl; the term "heteroaryl" represents an aromatic 5 or 6 membered ring comprising one to three heteroatoms selected from oxygen, sulfur and nitrogen, wherein the heteroaryl may be unsubstituted or optionally substituted by one or more substituents selected independently from halogen, cyano, trifluoromethyl, C1-4alkyl, Q^alkoxy, difluoromethoxy, trifluoromethoxy, cyclo-C3_6alkoxy, Ci-salkoxy-C^alkyl, cyclo-C3. 6alkylamino and di-(Ci_3alkyl)amino; its optical isomers, polymorphs and pharmaceutically acceptable acid addition salts and hydrates and solvates thereof.
Specific compounds of Formula I within the present invention include but are not limited to: 8 (3- Hydroxy 3- methylbu ^^^
8-(( 1 -Hydroxycyclohexyl)ethynyl)- 1 ,3 ,7-trimcthyl- 11 I-purine-2,6(3I I,7H)-dione, 8-(( 1 -Aminocydohexyl)ethynyl)-3 ,7-dimethyl- 1 H-purine-2,6(3H,7H)-dione,
8-(3-(Dimethylamino)prop- 1 -yn- 1 -yl)- 1 ,3 ,7-trimethyl- lH-purine-2,6(3H,7H)-dione, 8-(3-(bis(2-methoxyethyl)amino)prop- 1 -yn- 1 -yl)- 1 ,3 ,7-trimethyl- IH-purine- 2,6(3H,7H)-dione,
l,3,7-Trimemyl-8-(3-(pyrrolidin-l-yl)prop-l-yn-l-yl)-lH-purine-2,6(3H,7H)-dione, 1 ,3 ,7-Trimethyl-8-(3-(piperidin- 1 -yl)prop- 1 -yn- 1 -yl)- lH-purine-2,6(3H,7H)-dione, 8-(3-(Azepan- 1 -yl)prop- 1 -yn- 1 -yl)- 1 ,3 ,7-trimethyl- lH-purine-2,6(3H,7H)-dione, l,3,7-Trimethyl-8-(3-mo holinoprop-l-yn-l-yl)-lH-purine-2,6(3H,7H)-dione, 8-(3-Hydroxy-3-methylbut-l-yn-l-yl)-3,7-dimethyl-l-(5-oxohexyl)-lH-purine- 2,6(3H,7H)-dione,
8-(( 1 -Hydroxycyclohexyl)ethynyl)-3,7-dimethyl- 1 -(5-oxohexyl)- 1 H-purine- 2,6(3H,7H)-dione,
8-(( 1 - Aminocyclohexyl)ethynyl)-3 ,7-dimethyl- 1 -(5-oxohexyl)- lH-purine- 2,6(3H,7H)-dione,
8-(3-(Bis(2-methoxyethyl)amino)prop- 1 -yn- 1 -yl)-3 ,7-dimethyl- 1 -(5-oxohexyl)- 1 H- purine-2,6(3H,7H)-dione,
3,7-Dimethyl-l-(5-oxohexyl)-8-(3-(pyrrolidin-l-yl)prop-l-yn-l-yl)-lH-purine- 2,6(3H,7H)-dione,
3 ,7-Dimethyl- l-(5-oxohexyl)-8-(3-(piperidin- 1 -yl)prop- 1 -yn- 1 -yl)- 1 H-purine- 2,6(3H,7H)-dione,
l,3,7-Trimethyl-8-(phenylethynyl)-lH-purine-2,6(3H,7H)-dione,
1 ,3,9-Trimethyl-8-(3-(pyrrolidin- 1 -yl)prop- 1-yn- 1 -yl)-lH-purine-2,6(3H,9H)-dione, 1 ,3 ,9-Trimethyl-8-(phenylethynyl)- lH-purine-2,6(3H,9H)-dione
and optical isomers, polymorphs, and pharmaceutically- acceptable acid addition salts, hydrates, and solvates thereof. The invention also relates to a manufacturing process of a compound selected from those of Formula I as defined above, comprising reaction of a compound of Formula II:
Figure imgf000008_0001
II
wherein R l, R2 and R3 are as defined for Formula I above, with a compound of Formula III:
≡≡ R4
"I wherein R4 is as defined for Formula I above, optionally in the presence of base in an appropriate solvent (e.g., DIEA in DMF, NMP, DMAC or EtOAc), in the presence of Cul and palladium catalyst generated in situ (e.g., from PdCl2 or Pd(OAc)2 and PPh3) or commercially available (Ph3P)4Pd to yield a compound of Formula I, which may be converted, if desired, into an optical isomer, polymorph, pharmaceutically-acceptable salt, hydrate or solvate.
DETAILED DESCRIPTION OF THE INVENTION
As used herein, the term "Ci.4alkyl" represents straight or branched chain alkyl groups having 1, 2, 3 or 4 carbon atoms, examples of such alkyl groups include methyl, ethyl, n-propyl, 2-propyl, n-butyl, 2-butyl, iso-butyl and tert-butyl.
The term "cyclo-C3-6alkyl" represents monocyclic alkyl groups having 3, 4, 5 or 6 carbon atoms, including cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
The term "heterocyclyl" represents a saturated 4-7 membered heterocycle containing one or two heteroatoms selected from oxygen, sulfur and nitrogen, examples of such heterocyclyl groups include azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, tetrahydrofuryl, morpholinyl, thiomorpholinyl and piperazinyl.
The term "halo" or "halogen" represents fluorine, chlorine, bromine and iodine. In addition, using methods known to those skilled in the art, analogs and derivatives of the compounds of the invention can be created which have improved therapeutic efficacy, i.e., higher potency and/or selectivity at a specific targeted receptor type, greater ability to penetrate mammalian blood-brain barriers, fewer side effects, etc.
It will be appreciated by those skilled in the art that compounds of the invention having a chiral center may exist in and be isolated in optically active and racemic forms. It is to be understood that the present invention encompasses any racemic, optically-active, tautomeric, or stereoisomeric form of a compound of the invention, which possesses the useful properties described herein.
For therapeutic use, the compounds of Formula I can be in the form of a pharmaceutically acceptable salt or a solvate. The term "pharmaceutically acceptable" refers here to the therapeutically active non-toxic salt forms, which the compounds of Formula I are able to form. The latter can conveniently be obtained by treating the base form with such appropriate acids as inorganic acids such as hydrochloric acid, hydrobromic acid; sulfuric acid; nitric acid; phosphoric acid and the like; or organic acids such as acetic, propanoic, hydroxyacetic, 2-hydroxypropanoic, oxopropanoic, oxalic, malonic, succinic, maleic, fumaric, malic, tartaric, methanesulfonic, benzenesulfonic, 4-methylbenzenesulfonic, 2-hydroxybenzoic, and like acids. Conversely, the salt may be converted to the free base by treatment with alkali.
Scheme 1 describes the preparation of compounds of Formula I of the present invention. All of the starting materials II are prepared by representative procedures described in Schemes 2 and 3, by procedures well known to one of ordinary skill in organic chemistry or can be obtained commercially. All of the final compounds of the present invention are prepared by procedures described in these charts or by procedures analogous thereto, which procedures would be well known to one of ordinary skill in organic chemistry. All of the variables used in the schemes are as defined below or as in the claims.
We have found that product yields in palladium catalyzed cross-coupling of terminal acetylenes and 8-bromoxanthines strongly depends on the nature of catalyst and solvent. In accordance with experimental data, reaction of terminal acetylenes III with 8-bromoxanthines under routine experimental conditions (Methods A and B) in general led to the formation of the corresponding 8-ethynylxanthines I in very low yields. Suprisingly, we have found that use of the mixture of N-methylpyrrolidine and toluene (1:1) (Method C) gave the desired products in high yields. Alternative method was elaborated (Method D). Performing the reaction in ethyl acetate and using 2 mol- % of Pd(PPh3)4 and 2 mol-% of PdCl2 decreases the cost of reaction. It should be noted that in the representative example the treatment of 8-bromocaffeine with two equivalents of 2-methylbut-3-yn-2-ol under conditions of Method D led to the formation of desired 8-(3-hydroxy-3-methylbut-l-yn-l-yl)-l,3,7-trimethyl-lH-purine- 2,6(3H,7H)-dione (1-1) in 55% yield and by-product 8-[(£)-5-hydroxy-2-(l-hydroxy- 1 -methylethyl)-5-methylhex- 1 -en-3-ynyl]- 1 ,3 ,7-trimethyl- 1 H-purine-2,6(3H,7H)- dione in 26% yield similar to the method elaborated previously (Arsenyan, Tetrahedron Lett., 2013, 54, 6524-6528).
Scheme 1. General procedure toward compounds of Formula I .
Figure imgf000010_0001
Reaction conditions:
Method A. (Ph3P)2PdCl2, Cul, N-methylpyrrolidine or DMF, DIEA, 50 °C;
Method B. Pd(OAc)2, Ph3P, Cul, N-methylpyrrolidine or DMAC, DIEA, 55 °C;
Method C. Pd(OAc)2, Ph3P, Cul, N-methylpyrrolidine/toluene (1:1), DIEA, 50 °C; Method D. Pd(PPh3)4, PdCl2, Ph3P, Cul, ethyl acetate, DIEA, 40 °C.
Compounds II, wherein R3 is at purine N(7), can be prepared by bromination of position 8 of corresponding 8-unsubstituted 1,3,7-substituted lH-purine-2,6(3H,7H)- diones. Representative method for the synthesis of compound II wherein R1 and R3 are methyl groups shown in Scheme 2. Commercially available caffeine (1) and pentoxifylline (2) are brominated in position 8 by N-bromosuccinimide (NBS) in dichloromethane in analogy to published procedure [Synlett, 2012, 23, 1191-1198]. Both products 3 and 4 were isolated in almost quantitative yields. Scheme 2. General procedure for the preparation of 8-bromo lH-purine-2,6(3H,7H)~ diones II, wherein R3 is at purine N(7) (3, 4).
Figure imgf000011_0001
1 (R1 = Me) Me)
2 (R1 = MeC(0)(CH2)4) MeC(0)(CH2)4)
Compounds II, wherein R3 is at purine N(9), can be prepared by bromination of position 8 of respective 8-unsubstituted 1,3,9-substituted lH-purine-2,6(3H,9H)-
1 2 diones. Representative method for the synthesis of compound II wherein R , R and R3 are methyl groups (compound 11) is shown in Scheme 3. Thus, l,3-dimethyl-6- chlorouracil (6) was prepared by the treatment of 1,3-dimethylbarbituric acid (5) in phosphorous oxychloride (POCI3) with water followed by heating under reflux. Then treatment of compound 6 with a mixture of fuming nitric acid and sulfuric acid resulted in formation of 6-chloro-l,3-dimethyl-5-nitropyrimidine-2,4(lH,3H)-dione (7). Next, choro substituent was substituted by methylamino moiety to give intermediate 8 and nitro group was reducted by hydrogen using palladium on charcoal as a catalyst. Finally, condensation of 5-amino-l,3-dimethyl-6-methylamino- pyrimidine-2,4(lH,3H)-dione (9) with formic acid gave l,3,9-trimethyl-3,9-dihydro- purin-2,6(3H,9H)-dione (10). Necessary 8-bromo- 1,3, -trimethyl-3, 9-dihydropurin- 2,6(3H,9H)-dione (11) was obtained in the reaction of compound 10 with N- bromosuccinimide (NBS) in acetonitrile. The procedures shown in the Scheme 3 are general and can be used to prepare analogous 8-bromo 1,3,9-substituted lH-purine- 2,6(3H,9H)-diones.
Scheme 3. Preparation of 8-bromo lH-purine-2,6(3H,9H)-dione 11 (II, wherein R3 is at purine N(9)).
Figure imgf000012_0001
5 6 7
8
Figure imgf000012_0002
10 11
It will be appreciated that in the above transformations it may be necessary or desirable to protect any sensitive groups in the molecule of the compound in question in order to avoid undesirable side reactions.
EXPERIMENTAL PART
The compounds and their preparation of the present invention will be better understood in connection with the following examples, which are intended as an illustration of and not a limitation upon the scope of the invention.
Hereinafter, "DMF" is defined as Ν,Ν-dimethylformamide, "DMAC" is defined as Ν,Ν-dimethylacetamide, "NMP" is defined as N-methylpyrrolidone, "DMSO" as dimethyl sulfoxide, "HCl" as hydrochloric acid, „aq. NH3" as aqueous ammonia solution,„"MeCN" as acetonitrile, "DIEA" as diisopropylethylamine, "EtOAc" as ethyl acetate, "rt" as room temperature.
Intermediate 3.
8-Bromo-l,3,7-trimethyl-lH-pu e (3)
Figure imgf000012_0003
To a round-botton flask containing freshly distilled CH2C12 (150 mL) was added caffeine (10.0 g, 0.05 mol) and NBS (17.6 g, 0.10 mol). When the solids had dissolved in solvent, water (50 mL) was added and the reaction mixture was stirred for 5 days. Then cold 2 M aq.NaOH (30 mL) was added and the mixture was shaken till decolorization. The organic layer was separated, washed with water (2x200 mL), dried over sodium sulfate, filtered and evaporated to give the title compound (13.5 g, 99%). Ή NMR (CDC13™S, 400 MHz) δ (ppm): 3.41 (s, 3H, CH3), 3.57 (s, 3H, CH3), 3.97 (s, 3H, CH3). Intermediate 4
8-Bromo-3,7-dimethyl-l-(5 -dione (4)
Figure imgf000013_0001
To a round-botton flask containing freshly distilled CH2C12 (150 mL) was added pentoxifylline (13.4 g, 0.05 mol) and NBS (17.6 g, 0.10 mol). When the solids had dissolved, water (50 mL) was added and the reaction mixture stirred for 5 days. Then cold 2 M aq. NaOH (30 mL) was added and the mixture was shaken till decolorization. The organic layer was separated, washed with water (2x200 mL), dried over sodium sulfate, filtered and evaporated to give the title compound (16.95 g, 95%). Ή NMR (CDCI3 TMS, 400 MHz) £ (ppm): 1.61-1.67 (m, 4H), 2.14 (s, 3H), 2.49 (t, 2H), 3.54 (s, 3H), 3.95 (s, 3H), 3.99 (t, 2H).
Intermediate 11.
8-Bromo-l,3,9-trimethyl-lH-purine-2,6(3H,9H)-dione (11).
a) 6-Chloro-l,3-dimethylpyrimidine-2,4(lH,3H)-dione (6).
To a suspension of 1,3-dimethylbarbituric acid (7.8 g, 50 mmol) in POCl3 (60 mL) was slowly added water (2.5 mL, 0.14 mol) and the reaction mixture was refluxed for 1 h under nitrogen atmosphere. An excess of POCI3 was distilled off under reduced presssure and the residue was quenched with 40 mL of ice water. The mixture was extracted with chloroform (2x 100 mL). The organic phase was dried over anhydrous Na2S04, evaporated to dryness, and the residue crystallized with ether to give the title compound (8.0 g) as a yellow crystals. Ή NMR (400 MHz, DMSO- d6) δ (ppm): 3.10 and 3.15 (both s, both 311), 6.07 (s, 1H).
b) 6-Chloro-l^-dimethyl-5-nitropyrimidine-2,4(lH H)-dione (7).
Compound 6 (15 g, 74.5 mmol) was added portionwise to a cooled solution of cone. H2SO4 (40 mL) . The reaction temperature was maintained below 10 °C. Fuming nitric acid (15 mL) was added dropwise to the above reaction mixture and it was stirred for 2 h at the same temperature. The reaction nixture was poured onto the ice cold water (500 mL) and extracted with chloroform (2x260 mL). The combined organic extracts were washed with water (260 mL), dried over anhydrous Na2S04 and concentrated in vacuo to give the title compound (12.0 g) as a yellow solid. 1H NMR (400 MHz, DMSO-de) δ (ppm): 3.07 and 3.26 (both s, both 3H).
c) l,3-Dimethyl-6-methylamino-5-nitropyrimidine-2,4(lH,3H)-dione (8).
To a stirred solution of 6-chloro-l,3-dimethyl-5-nitropyrimidine-2,4(lH,3H)-dione (7) (11.0 g, 50.1 mmol) in chloroform (90 mL) was added dropwise a solution of 40% aq. methylamine (7.76 mL, 100.2 mmol) in dichloromethane (20 mL) under nitrogen atmosphere. After stirring at room temperature for 1 h the reaction mixture was concentrated under reduced pressure. The residue was crystallized from ether to give the title compound (12.5 g) as a yellow solid. lH NMR (400 MHz, DMSO- d6) δ (ppm): 2.49, 2.75 and 3.35 (all s, all 3H), 7.74 (br s, 1H).
d) 5-Amino-l,3-dimethyl-6-methylamino-pyrimidine-2,4(lH,3H)-dione (9).
To a stirred solution of l,3-dimethyl-6-methylamino-5-nitropyrimidine-2,4(lH,3H)- dione (8) (2 g, 10 mmol) in wet methanol (140 mL) was added 10% Pd-C (1 g) under hydrogen balloon atmosphere at room temperature. After overnight stirring, the reaction mixture was filtered and the filtrate was concentrated to give the title compound (1.5 g). 1H NMR (400 MHz, DMSO-d6) δ (ppm): 2.89 (d, 3H), 3.31 (s, 3H), 3.40 (s, 3H), 4.75 (br s, 1H). MS (EI) mlv 185 [M]+.
e) l,3,9-Trimethy]-lH-purine-2,6(3H,9H)-dione (10).
A mixture of 5-amino-l,3-dimethyl-6-methylamino-pyrimidine-2,4(lH,3H)-dione (9) (1.5 g) and formic acid (10 mL) was refluxed for 3 h under nitrogen atmosphere. An excess of formic acid was evaporated under reduce pressure. The residue was extracted with CH2CI2, washed with aq. Na2C03, dried over Na2S04 and evaporated to dryness. The residue was purified by column chromatography (eluent CH2C12 : MeOH, 10: 1 ) to give the title compound (0.5 g) as a white solid. ]H NMR (400 MHz, DMSO-de) δ (ppm): 3.22, 3.68 and 3.93 (all s, all 3H), 7.66 (s, IH). MS (EI) rnlz: 195 [M+2]+.
f) 8-Bromo-l,3,9-trimethyl-l//-purine-2,6(3//,9H)-dione (11).
A mixture of 1.3 ,9-trimethyl- 1 H-purine-2,6(3H,9H)-dione (10) (1.0 g, 5.15 mmol) and NBS (1.2 g, 6.7 mmol) in dry MeCN (40 mL) was stirred for 3 h at room temperature. Water (30 mL) and CH2C12 (150 mL) was added, the organic phase was separated, dried over Na2S04 and evaporated to dryness. The residue was purified by column chromatography (CH2C12 : MeOH, 9:1) to give the title compound (0.57 g) as a white solid with m.p. >200 °C. Ή NMR (400 MHz, DMSO-d6) δ (ppm): 3.22, 3.69 and 3.88 (all s, all 3H). MS (EI) mlz: 375 [M+2]+.
Example 1
8-(3-Hydroxy-3-methylbut-l-yn-l-yl)-l,3,7-trimethyl-lH-purine-2,6(3H,7H)- dione (1-1)
Figure imgf000015_0001
Method A. To a mixture of PdCl2 (113 mg, 0.637 mmol), Cul (242 mg, 1.27 mmol), and triphenylphosphine (333 mg, 1.27 mmol) was added dry NMP or DMF (40 mL). The reaction mixture was allowed to stir at 40 °C for 15 min with simultaneous barbotation with argon. Then a solution of 8-bromo-l,3,7-trimethyl-lH-purine- 2,6(3H,7H)-dione (1.73 g, 6.37 mmol) and 2-methylbut-3-yn-2-ol (0.93 mL, 9.56 mmol) and dry DIEA (3.6 mL) was added and stirring was continued at 50 °C for 24 h. After cooling to rt, the reaction mixture was poured to EtOAc (300 mL), washed with brine (80 mL) containing aq. NH3 (0.5 mL) and stirred at rt for the additional 30 min. The aqueous phase was separated and the organic phase was washed with brine (3 x 80 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude product was purified by flash chromatography on silica gel using a mixture of water (containing 0.1% HC1) - MeCN (5% - 70%) as eluent to give the title compound in 42% yield, mp = 180-182 °C. Ή NMR (CDC13 TMS, 400 MHz) <5> (ppm): 1.63 (s, 6H), 3.36 (s, 3H), 3.51 (s, 3H), 3.52 (br s, 1H), 3.90 (s, 3H).
13C NMR (CDCI3/TMS, 100.6 MHz) ^(ppm): 28.0, 29.8, 30.8, 32.9, 65.1, 69.6, 102.5, 107.5, 135.0, 147.4, 151.5, 154.6. MS (EI) mlz: 277.2 [M+l]+. Method B. To a mixture of Pd(OAc)2 ( 100 mg, 0.446 mmol), Cul (169 mg, 0.868 mmol), and triphenylphosphine (233 mg, 0.890 mmol) dry NMP or DMAC (40 mL) was added. Reaction mixture was allowed to stir for 15 min at 40 °C with simultaneous barbotation with argon. Then a solution of 8-bromo- 1 ,3,7-trimethyl 1 //- purine-2,6(3H,7H)-dione (1.21 g, 4.46 mmol) and 2-methylbut-3-yn-2-ol (0.65 mL, 6.69 mmol) and dry DIEA (4.0 mL) was added and stirring was continued at 55 °C for 24 h. Compound 1-1 (31% yield) was isolated as in Method A. Method C. To a mixture of Pd(OAc)2 (100 mg, 0.446 mmol), Cul (169 mg, 0.868 mmol), and triphenylphosphine (233 mg, 0.890 mmol) dry NMP (10 mL) was added. Reaction mixture was allowed to stir for 15 min at 40 °C with simultaneous barbotation with argon. Then solution of 8-bromo- 1,3, 7-trimethyl-lH-purine- 2,6(3H,7H)-dione (1.21 g, 4.46 mmol) and 2-methylbut-3-yn-2-ol (0.65 mL, 6.69 mmol) and dry DIEA (4.0 mL) in NMP (10 mL) and toluene (20 mL) was added and stirring was continued at 50 °C for 24 h. Compound 1-1 (72% yield) was isolated as in Method A.
Method D. A vial charged with Pd(PPh3)4 (346 mg, 0.3 mmol), PdCl2 (51 mg, 0.3 mmol), Ph3P (157 mg, 0.6 mmol), Cul (58 mg, 0.3 mmol), 8-bromo- 1,3,7- trimethyl-lH-purine-2,6(3H,7H)-dione (4.08 g, 15.0 mmol) and 2-methylbut-3-yn-2- ol (2.05 mL, 21.0 mmol), DIEA (5.0 mL) and ethyl acetate (70 mL) was stirred for 15 min at 40 °C with simultaneous barbotation with argon. Then reaction mixture was stirred for additional 2-4 h. After cooling to rt, the reaction mixture was filtered through a silica gel pad and washed with EtOAc (200 mL). Then solvent was evaporated under reduced pressure. The crude product was purified by flash chromatography on silica gel using mixture of water (containing 0.1% aq. HQ) - MeCN (5% - 70%) as eluent to give compound 1-1 in 55% yield and 8-[(£)-5- hydroxy-2-( 1 -hydroxy- 1 -methylethyl)-5-methylhex- l-en-3-ynyl]- 1 ,3 ,7-trimefhyl- 1 H- purine-2,6(3H,7H)-dione as a by-product in 26% yield. By-product: Ή NMR (CDC13 TMS, 400 MHz) (ppm): 1.32 (s, 6H), 1.52 (s, 6H), 3.35 (s, 3H), 3.49 (s, 3H), 4.03 (s, 3H). 5.13 (br s, 2H), 6.65 (s, 1H). 13C NMR (CDC13/TMS, 100.6 MHz) δ ippm): 28.1 , 29.7, 29.9, 31.0, 33.7, 65.3, 70.7, 78.6, 95.9, 107.9, 109.6, 146.5, 147.3, 151.2, 155.0, 156.1.
Example 2
8-((l-Hydroxyeyclohexyl)ethynyl)-l,3,7-trimethyl-lH-purine-2,6f3//,7//)-dione (1-2)
Figure imgf000017_0001
Yield: 49% (Method C), 54% (Method D), 11% (Method B); mp = 194-196 °C. Ή NMR (CDC13/TMS, 400 MHz) (ppm): 1.24-1.33 (m, 1H), 1.48-1.57 (m, 3H), 1.66- 1.75 (m, 4H), 1.94-2.02 (m, 2H), 3.30 (br s, 1H), 3.34 (s, 3H), 3.49 (s, 3H), 3.89 (s, 3H). 13C NMR (CDC13 TMS, 100.6 MHz) £ (ppm): 22.9, 24.8, 27.9, 29.7, 32.9, 39.2, 68.6, 71.5, 102.0, 107.4, 135.1, 147.3, 151.4, 154.5. MS (EI) m/Z: 317.5 [M+l]+.
Example 3
8-((l-Aminocyclohexyl)ethynyl)-3,7-dimethyl-lH-purine-2,6(3H,7H)-dione (I-3)
Figure imgf000017_0002
Yield: 48% (Method D), 43% (Method C), 16% (Method B), 7 % (Method A); mp = 187-189 °C. 1H NMR (CDC13 TMS, 400 MHz) £(ppm): 1.20-1.29 (m, 1H), 1.48-1.67 (m, 5H), 1.72-1.77 (m, 2H), 1.81 (br s, 2H), 1.93-1.97 (m, 2H), 3.39 (s, 3H), 3.55 (s, 3H), 3.98 (s, 3H). 13C NMR (CDCI3/TMS, 100.6 MHz) £(ppm): 23.2, 25.1, 27.9, 29.7, 33.0, 39.6, 50.4, 70.9, 104.5, 107.6, 135.8, 147.6, 151.5, 154.8. MS (EI) m/z 302.3 [M+l]+.
Example 4
8-(3-(Dimethylamino)prop-l-yn-l-yl)-l,3»7-trimetliyl-lH-purinc-2,6i3H,7H)- diorie (1-4)
Figure imgf000018_0001
Isolated as hydrochloride. Yield: 58% (Method D), 47% (Method C), 4% (Method B); mp = 197- 199 °C (dec. ) Ή NMR (DMSO-d6, 400 MHz) (ppm): 2.88 (s, 611), 3.21 (s, 3H), 3.38 (s, 3H), 3.96 (s, 3H), 4.50 (s, 2H). 13C NMR (DMSO-d6, 100.6 MHz) δ (ppm): 27.6, 29.3, 33.0, 41.6, 45.7, 76.4, 87.1, 107.6, 132.9, 146.8, 150.7, 154.0. MS (EI) m/z: 277.5 [M+l].
Example 5
8-(3-(bis(2-methoxyethyI)amino)prop-l-yn-l-yI)-l,3>7-trimethyI-lH-purine- 2,6(3H,7H)-dione (1-5)
Figure imgf000018_0002
Yield: 33% (Method D), 30% (Method C), 10% (Method B); foam. 1H NMR (CDCI3/TMS, 400 MHz) £(ppm): 2.79 (t, 4H), 3.31 (s, 6H), 3.34 (s, 3H), 3.48 (t, 4H), 3.51 (s, 3H), 3.82 (s, 2H), 3.96 (s, 3H). 13C NMR (CDCI3 TMS, 100.6 MHz) S(pp ): 27.8, 29.6, 33.0, 44.1, 53.5, 58.7, 70.9, 73.2, 94.1, 107.5, 135.4, 147.5, 151.4, 154.7. MS (EI) mlz: 364.5 [M+l]+.
Example 6
l,3,7-Trimethyl-8-(3-(pyrrolidin-l-yl)prop-l-yn-l-yl)-lH-purine-2,6(3H,7H)- dione (1-6)
Figure imgf000018_0003
Yield: 57% (Method D), 52% (Method C), 8% (Method A); mp = 149- 150 °C. Ή NMR (CDCI3 TMS, 400 MHz) δ (ppm): 1.83-1.86 (m, 4H), 2.68-2.72 (m, 4H), 3.39 (s, 3H), 3.56 (s, 3H), 3.75 (s, 2H), 4.00 (s, 3H). C NMR (CDCI3 TMS, 100.6 MHz) S (ppm): 23.8, 27.9, 29.7, 33.1, 43.6, 52.7, 72.6, 94.5, 107.6, 135.4, 147.6, 151.5, 154.8. MS (EI) mlz: 302.3 [M+l]+. Example 7
13 -Trimethyl-8-(3-(piperidin-l-y!)prop-l-yn -yl)-li?-purine-2,6(3H,7H)-dione
(1-7)
Figure imgf000019_0001
Yield: 35% (Method C), 30% (Method D); mp = 137-138 °C. 1H NMR (CDCI3/TMS, 400 MHz) <?(ppm): 1.40-1.48 (m, 2H), 1.60-1.67 (m, 4H), 2.54-2.60 (m, 4H), 3.39 (s, 3H), 3.55 (s, 3H), 3.60 (s, 2H), 4.00 (s, 3H). 13C NMR (CDCI3/TMS, 100.6 MHz) δ (ppm): 23.6, 25.8, 27.9, 28.0, 29.8, 48.3, 53.4, 73.2, 94.2, 107.6, 135.5, 147.6, 151.5, 154.8. MS (EI) w/z: 316.3 [M+l]+. Example 8
8-(3-(Azepan-l-yl)prop-l-yn-l-yl)-l,3,7-trimethyl-lH-purine-2,6(3H,7H)-dione
(1-8)
Figure imgf000019_0002
Yield: 34% (Method C), foam. lR NMR (CDCI3/TMS, 400 MHz) < (ppm): 1.61-1.64 (m, 4H), 1.70-1.76 (m, 4H), 2.81 (t, 4H), 3.39 (s, 3H), 3.55 (s, 3H), 3.74 (s, 2H), 4.01 (s, 3H). 13C NMR (CDCI3/TMS, 100.6 MHz) δ (ppm): 26.6, 27.9, 28.0, 29.7, 33.1, 48.7, 55.3, 72.9, 93.2, 107.6, 135.4, 147.6, 151.5, 154.8. MS (EI) mlz: 330.3 [M+lf.
Example 9 1 J-Trimethyl-8 3-morpholinoprop-l-yn-l-yl)-lH»piirine-256(3 H)-dione
9)
Figure imgf000020_0001
Yield: 38% (Method C), mp =188-190 °C. 1H NMR (CDCI3 TMS, 400 MHz) δ (ppm): 2.64 (t, 4H), 3.40 (s, 3H), 3.56 (s, 3H), 3.64 (s, 2H), 3.76 (t, 4H), 4.01 (s, 3H). 13C NMR (CDCI3 TMS, 100.6 MHz) <?(ppm): 28.0, 29.7, 33.2, 47.8, 52.3, 66.7, 73.7, 93.1, 107.7, 135.2, 147.6, 151.5, 154.8. MS (EI) ml v. 318.3 [M+l]+.
Example 10
8-(3-Hydroxy-3-methyIbut-l-yn-l-yl)-3,7-dimethyl-l-(5-oxohexyl)-lH-purine- 2,6(3H,7H)-dione (1-10
Figure imgf000020_0002
Yield: 47% (Method D), 44% (Method C); foam. 1H NMR (CDCI3/TMS, 400 MHz) δ (ppm): 1.62-1.66 (m, 4H), 1.65 (s, 6H), 2.12 (s, 3H), 2.48 (t, 2H), 2.72 (br s, IH), 3.53 (s, 3H), 3.96 (s, 3H), 3.97 (t, 2H). 13C NMR (CDCVTMS, 100.6 MHz) (ppm): 20.9, 27.3, 29.7, 29.9, 30.9, 33.0, 40.9, 43.1, 65.4, 70.0, 102.2, 107.7, 135.1, 147.6, 151.2, 154.6, 208.7. MS (EI) mlz: 361.1 [M+l]+.
Example 11
8-((l-Hydroxycyclohexyl)ethynyl)-3,7-dimethyl-l-(5-oxohexyl)-lH-purine- 2,6(3H,7H)-dione (1-1
Figure imgf000020_0003
Yield: 42% (Method C), mp = 126-128 °C. lH NMR (CDCI3/TMS, 400 MHz) δ (ppm): 1.27-1.37 (m, I H), 1.54-1.79 (m, 7H), 2.02-2.08 (m, 2H), 2.13 (s, 3H), 2.48 (t, 2H), 3.54 (s, 3H), 3.97 (s, 3H). 3.97-4.01 (2H, m). 13C NMR (CDC13 TMS, 100.6 MHz) <5" (ppm): 20.9, 23.0, 24.9, 27.4, 29.7, 29.9, 33.1 , 39.3, 40.9, 43.1 , 69.0, 72.0, 101.5, 107.8, 135.3, 147.6, 151.2, 154.6, 208.6. MS (EI) m/z: 401.6 | M+1 ]+.
Example 12
8-((l-Aininocyclohexyl)ethyiiyl)-3 -dimethyl-l-(5-ox hexyl)-l//-purine- 2,6(3//,7//)-dione (1-1
Figure imgf000021_0001
Yield: 45% (Method C), mp > 200° C. 1H NMR (CDC13 TMS, 400 MHz) (ppm): 1.24-1.34 (m, 1H), 1.61-1.84 (m, 7H), 1.95-2.01 (m, 2H), 2.13 (s, 3H), 2.34-2.37 (m, 2H), 2.48 (t, 2H), 3.39 (s, 3H), 3.49 (s, 3H, 3.93-3.97 (m, 2H), 4.01 (s, 3H), 9.28 (br s, 2H). 13C NMR (CDCl3 TMS, 100.6 MHz) (ppm): 20.9, 22.5, 24.1, 27.3, 25.1 , 29.7, 29.9, 33.5, 35.9, 41.0, 43.1, 53.6, 75.7, 93.9, 108.0, 133.8, 147.4, 150.9, 154.4, 208.6. MS (EI) m/z: 400.5 [M+l]+. Example 13
8-(3-(Bis(2-methoxyethyl)amino)prop-l-yn-l-yl)-3,7-dimethyl-l-(5-oxohexyl)-lH- purine-2,6(3H,7H)-dione (1-13)
Figure imgf000021_0002
Yield: 35% (Method C), foam. 1H NMR (CDC1 TMS, 400 MHz) (ppm): 1.60-1.65 (m, 4H), 2.10 (s, 3H), 2.47 (t, 2H), 2.34-2.37 (m, 2H), 2.83 (t, 4H), 3.30 (s, 6H), 3.45 (t, 4H), 3.54-3.55 (m, 2H), 3.56 (s, 3H), 3.92 (s, 3H), 3.97 (t, 2H). 13C NMR (CDC13/TMS, 100.6 MHz) (ppm): 20.9, 27.4, 29.8, 31.9, 40.7, 43.1, 54.2, 58.8, 63.0, 66.5, 71.0, 76.9, 107.2, 110.3, 143.1 , 147.3, 148.1, 151.3, 155.1, 208.6. MS (EI) m/z: 448.6 [M+l f.
Example 14 397-Dimethyl-l-(5-oxohexyl)-8-(3 pyrrolidlE-l-y!)prop-l-yn-l-yl)-lH-purine- 2,6(3H,7H)-dione (1-14)
Figure imgf000022_0001
Isolated as hydrochloride. Yield: 41%, foam. 'Η NMR (CDCl TMS, 400 MHz) δ (ppm): 1.61-1.70 (m, 4H), 1.82-1.86 (m, 4H), 2.13 (s, 3H), 2.49 (t, 2H), 2.64-2.68 (m, 4H), 3.48 (d, 2H), 3.58 (s, 3H), 3.93 (s, 3H), 4.01 (t, 2H), 6.78 (br s, 1H). 13C NMR (CDC1 TMS, 100.6 MHz) £(ppm): 20.8, 23.9, 27.3, 29.6, 29.9, 33.6, 40.9, 43.0, 43.3, 52.7, 77.6, 85.1, 108.4, 133.0, 147.4, 151.0, 154.5, 208.5. MS (EI) m/z: 386.3 [M+l]+. Example 15
3,7-Dimethyl-l-(5-oxohexyl)-8-(3-(piperidin-l-yI)prop-l-yn-l-yI)-lH-purine- 2,6(3H,7H>dione (1-1
Figure imgf000022_0002
Yield: 39% (Method C), mp = 182-184 °C. 1H NMR (CDC13/TMS, 400 MHz) δ (ppm): 1.34-1.48 (m, 1H), 1.58-1.66 (m, 4H), 1.90-1.93 (m, 3H), 2.11 (s, 3H), 2.21- 2.29 (m, 2H), 2.47 (t, 2H), 2.94-2.97 (m, 2H), 3.51 (s, 3H), 3.60-3.63 (m, 2H), 3.97 (t, 2H), 4.07 (s, 3H), 4.22 (s, 2H). 13C NMR (CDC13 TMS, 100.6 MHz) <S (ppm): 20.8, 21.5, 22.7, 27.3, 29.6, 29.9, 33.7, 40.9, 43.0, 46.7, 52.6, 78.7, 84.6, 108.5, 133.1, 147.5, 151.0, 154.5, 208.5. MS (EI) m/z: 400.2 [M+l]+.
Example 16
l,3,7-Trimethyl-8-(phenylethynyl)-lH-purine-2,6(3H,7H)-dione (1-16)
Figure imgf000022_0003
Ή NMR (CDCl TMS, 400 MHz) δ (ppm): Yield: 36% (Method C). Ή NMR
(CDCI3/TMS, 400 MHz) £ (ppm): 3.43 (s, 3H), 3.61 (s, 3H), 4.10 (s, 3H), 7.39-7.46 (m, 3H), 7.61 -7.63 (m, 2H). MS (EI) mix;. 295.5 [M]+. Example 17
l,3,9-Trimethyl-8-(3^pyrrolidin-l-yl)prop- l-yn-I-yl)-l//-purine-2,6(3//,9H)- dione (1-17)
Figure imgf000023_0001
Isolated as hydrochloride. 1H NMR (DMSO-d6, 400 MHz) δ (ppm): 1.86-1.99 (m, 4H), 3.07-3.20 (m, 4H), 3.25 (s, 3H), 3.72 (s, 3H), 3.93 (s, 3H), 4.35 (s, 2H). MS (EI) m/z: 302.1 [M]+.
Example 18
l,3,9-Trimethyl-8-(phenyl -dione (I-18)
Figure imgf000023_0002
Yield: 75% (Method C), mp > 200 °C. 1H NMR (CDCI3 TMS, 400 MHz) £ (ppm): 3.42 (s, 3H), 3.80 (s, 3H), 4.06 (s, 3H), 7.35-7.42 (m, 3H), 7.53-7.55 (m, 2H). 13C NMR (DMSO-d6, 100.6 MHz) £(ppm): 28.1, 31.0, 33.8, 78.1, 93.6, 116.6, 120.1, 128.9, 129.9, 131.2, 131.6, 140.0, 150.8, 156.1. MS (El) m/Z: 295.3 [M]+.
ANTIPROLIFERATIVE ACTIVITY
Anticancer activity of 8-ethynylxanthines was tested on monolayer tumor cell lines: MDA-MB-435s (human melanoma), H9C2 (rat embrio cardiomyoblast), MCF-7 (human breast adenocarcinoma, estrogen-positive), HepG2 (human hepatocellular carcinoma), SH-SY5Y (human neuroblastoma), C6 (rat glioma), U937 (human histiocytic leukemia), and A549 (human lung carcinoma). The borderline concentration, relevant to the highest tolerated dose, was determined for each compound using the NIH 3T3 (Mouse Swiss Albino embryo fibroblasts) cell line. The basal cytotoxicity was used to predict starting doses for in vivo acute oral LD50 values in rodent. The results of these experiments are summarized in Table 1. Caffeine, Proxyfeine, Pentoxifylline and Temodar were used as references. Caffeine and Pentoxifylline exhibit no cytotoxic effect on all studied cell lines. Despite the fact that Temodar is widely used drag for brain cancer prevention, in vitro results showed more than a modest activity on studied tumor cell lines (IC5o=298 μΜ on SH-SY5Y neuroblastoma and IC5o=31 μΜ on U937 lymphoma cell lines). Cancer cells are more sensitive to Proxyfeine (IC5o=96-283 μΜ). Surprisingly, 8-ethynylcaffeines have an extended ability to suppress cancer cell growth. Thus, caffeine derivatives 1-6-8 showed high in vitro antiproliferative effect against majority of cancer cells, moreover, these compounds are active against brain tumors (IC5o=8.2÷11.5 μΜ on SH-SY5Y and C6 cell lines). It should be noted that caffeine 1-3 selectively inhibits lymphoma U937 cell growth (IC50=5.1 μΜ), and derivative 1-6 - estrogen-positive human breast adenocarcinoma MCF-7 (IC5o=5.7 μΜ). In a series of pentoxifylline derivatives 1-14- 15 piperidylmethyl derivative 1-15 showed high cytotoxicity against studied cancer cells, especially, on lymphoma U937 (ICso=4.7 μΜ) and glioma C6 (Κ¾ο=7.1 μΜ).
In vitro cytotoxicity assay
Monolayer tumor cell line: MDA-MB-435s (human melanoma), H9C2 (rat embrio cardiomyoblast), MCF-7 (human breast adenocarcinoma, estrogen-positive), HepG2 (human hepatocellular carcinoma), SH-SY5Y (human neuroblastoma), C6 (rat glioma), U937 (human histiocytic leukemia), and A549 (human lung carcinoma), and normal cell line NIH 3T3 (mouse fibroblasts) were cultured in standard medium DMEM (Dulbecco's modified Eagle's medium) containing 1% non-essential amino- acids, 2 niM glutamine and supplemented with 10% fetal bovine serum (FBS, Sigma) ("Sigma"). All cells obtained from the American Type Culture Collection. About 2-10T04 cells/mL (depending on line nature) were placed in 96-well plates immediately after compounds were added to the wells. The control cells without test compounds were cultured on separate plate. The plates were incubated for 72 h, 37 °C, 5% C02. The number of surviving cells was determined using 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolinium bromide (MTT). MTT-test: after incubating with preparations culture medium was removed and 200 ih of fresh medium with 10 mM HEPES was added in each well of the plate, then 20 μΐ . of MTT (2 mg/mL in HBSS) was added. After incubation (3 h., 37 °C, 5%C02) the medium with MTT was removed and 200 μL· of DMSO and 25 of glycine buffer (pl 10.5) were added at once to each sample. The samples were tested at 540 rim on Anthos HT II photometer.
Basal toxicity test
Compounds were tested on NIH 3T3 (normal mouse fibroblasts, "ATCC") cell line according the basal toxicity test (INVITOX Protocol No 64, 1992) and non-toxic compounds were selected. 9,000 NIH 3T3 cells/well were placed into 96-well plates for 24 h and then exposed to the test compound over a range of eight concentration (1- 1000 μg/mL) for 24 h. Upon that, the cells were incubated with the neutral red dye for 4 h and then OD was determined at 540 nm. Alternative LD50 values (LD50 value is the amount of the drug that is taken to kill 50% of the test animals) was calculated according to the formula: log (LD50 [mmol/kg] = 0.435 x log (IC50 [mmol/1]) = 0.625. The IC5o values were calculated using the program Graph Pad 5 Prism® 3.0.
Table 1. In vitro cytotoxicity caused by 8-cthynylxanthines was tested on monolayer tumor cell lines: MDA-MB-435s (human melanoma), H9C2 (rat embrio cardiomyoblast), MCF-7 (human breast adenocarcinoma, estrogen-positive), HepG2 (human hepatocellular carcinoma), SH- SY5Y (human neuroblastoma), C6 (rat glioma), U937 (human histiocytic leukemia), and A549 (human lung carcinoma).
MDA-MB-435s H9C2 MCF-7 HepG2 SH-SY5Y C6 U937 A549 3T3
Compound
ICso ICso ICso ICso ICso ICso ICso ICso LD50, mg/kg
Caffeine * * * * * * * * 1670
Prox feine 283 * 135 * 111 96 97 * 1891
Pentoxifylline * * * * * * * >2000
Teinodar 515 * * * 298 * 31 * >2000
1-1 119 141 * * 83 261 79 239 1 1 16
1-2 * * * * * 187 95 * 1329
1-3 13 41 36 96 41 15 5.1 52 347
1-4 28 57 25 28 25 28 9.2 42 873
1-5 * * * * * * * * 1924
1-6 8.6 75 5.7 17 8.6 11.5 26 8.6 262
1-7 14 68 14 60 14 8.5 5.7 17 383
1-8 8.2 8.2 8.2 13.7 8.2 8.2 5.5 8.2 287
1-9 * * * * * * 124 * 1676
1-10 * 22 169 86 105 277 83 188 2235
I-ll * 259 * 116 183 85 259 1766
1-12 80 46 73 96 64 20 87 78
1-14 7.3 9.8 4.9 12 4.9 17 12 7.3
1-15 19 52 11.8 26 9.5 7.1 4.7 11.8
1-16 122 51 44 34 17 17 13.6 85
1-17 * * * * 193 * 151 *
1-18 54 126 98 105 85 139 133 143 aIC50 - Concentration (μΜ) providing 50% cell killing effect (MTT).

Claims

1. A compound selected from those of Formula I:
Figure imgf000028_0001
wherein
R1 represents hydrogen, C1-4alkyl, hydroxy-C2_4alkyl, C^alkoxy-C^alkyl, Q. 3alkylcarbonyl-C i .4alkyl or C ^alkyKC! .3alkyl)amino-C2-4alkyl ;
R2 represents C1.4alkyl, hydroxy-C2-4alkyl, Q^alkylcarbonyl-Q^alkyl, C1_3alkoxy-C2. 4alkyl, Ci-3alkyl(C1-3alkyl)amino-C2-4alkyl or halo-C2.4alkyl;
R represents C1-4alkyl, allyl or C1-3alkoxy-C2-4alkyl; with the proviso that if substituent R3 is at purine N(7) atom the dotted line between N(7) and C(8) represents no bond, and the dotted line between C(8) and N(9) represents chemical bond;
and
with the proviso that if substituent R3 is at purine N(9) atom the dotted line between N(7) and C(8) represents chemical bond, and the dotted line between C(8) and N(9) represents no bond;
R4 represents C1-4alkyl, hydroxy-Ci.4alkyl, C i .3alkoxy-C i .4alkyl, amino-Ci_4alkyl, 1- hydroxy-di-(C1- alkyl)methyl, l-amino-di-(Ci.3alkyl)methyl, l-hydroxy-cyclo-C3. 6alkyl, l-amino-cyclo-Cs^alkyl, l-(hydroxy-C1-3alkyl)-cycloC3-6alkyl, Ci_ 3alkylamino-Ci-3alkyl, C i .3alkyl(C i .3alkyl)amino-C i -3alkyl, di-(C1-3alkoxy-C2-4alkyl)- aniino-Ci-3alkyl, heterocyclyl-C1-3alkyl, aryl or heteroaryl; wherein the term "heterocyclyl" represents saturated 4-7 membered heterocycle containing one or two heteroatoms selected from oxygen, sulfur and nitrogen, wherein the heterocyclyl may be azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, tetrahydrofuryl, morpholinyl, thiomorpholinyl and piperazinyl; the term "aryl" represents phenyl or phenyl substituted by one or more substituents selected independently from halogen, cyano, Ci_4alkoxycarbonyl, N-Cj. 4alkylaminocarbonyl, N,N-di-(Ci-3alkyl)aminocarbonyl, CH2OH, trifluoromefhyl, Ci_ 4alkyl, allyl, C2-4alkynyl, C1-4alkoxy, difluoromethoxy, trifluoromethoxy, cyclo- C3_6alkoxy, hydroxy-Ci-4alkyl, Ci^alkoxy-Ct^alkyl, C1-3alkoxy-C2-4alkoxy, di-(Ci_ 3alkyl)amino, di-(Ci-3alkyl)amino-C1-3alkyl, di-(C1-3alkyl)amino-C2-4alkoxy, C1_4alkylsulfonylamino and Ct^alkyl-aminosulfonyl; the term "heteroaryl" represents an aromatic 5 or 6 membered ring comprising one to three heteroatoms selected from oxygen, sulfur and nitrogen, wherein the heteroaryl may be unsubstituted or optionally substituted by one or more substituents selected independently from halogen, cyano, trifluoromethyl, C1-4alkyl, C1-4alkoxy, difluoromethoxy, trifluoromethoxy, cyclo-C3.6alkoxy, C1-3alkoxy-Ci.4alkyl, cyclo-C3- 6alkylamino and di-(C1-3alkyl)amino; its optical isomers, polymorphs and pharmaceutically acceptable acid addition salts and hydrates and solvates thereof.
2. The compound as claimed in Claim 1, wherein R3 is at purine N(7) atom.
3. The compound as claimed in Claim 1, wherein R3 is at purine N(9) atom.
4. The compound as claimed in Claims 1, 2 and 3, wherein R4 represents hydroxy- Ci„4alkyl, amino-Ci.4alkyl, l-hydroxy-di-(Ci_3alkyl)methyl, l-amino-di-(Ci_ 3alkyl)mefhyl, l-hydroxy-cyclo-C3_6alkyl, l-amino-cyclo-C3.6alkyl, 1 -(hydroxy- Ci-3alkyl)-cycloC3-6alkyl, Ci 3alkyl(Ci salkyljamino-Ci-aalkyl, di-iCVialkoxy- C2-4alkyl)-amino-Ci.3alkyl, heterocyclyl-Ci-3alkyl or aryl.
5. The compound as claimed in Claim 4, wherein R2 and R3 each independently represent Ci_4alkyl or C1_3alkoxy-C2_4alkyl.
6. The compound as claimed in Claim 5, wherein R and R each represent methyl.
7. The compound as claimed in any of Claims 1 to 6, wherein R1 represents methyl or 5-oxohexyl.
8. The compound as claimed in Claim 1, which is selected from:
8-(3-Hydroxy-3-methylbut- 1-yn- 1-yl)- 1 ,3,7-trimethyl- lH-purine-2,6(3H,7H)- dione,
8-((l-Hydroxycyclohexyl)emynyl)-13 -trimethyl-lH-purine-2,6(3H,7H)-dione, 8-((l-Aminocyclohexyl)emynyl)-3,7-dimethyl-lH-purine-2,6(3H,7H)-dione, 8-(3 -(Dimethylamino)prop- 1 -yn- 1-yl)- 1 ,3 ,7-trimethyl- 1 H-purine-2,6(3H,7H)- dione,
8-(3-(bis(2-methoxyethyl)amino)prop- 1 -yn-1 -yl)- 1 ,3 ,7-trimethyl- lH-purine- 2,6(3H,7H)-dione,
l,3,7-Trimethyl-8-(3-(pyrrolidin-l-yl)prop-l-yn-l-yl)-lH-purine-2,6(3H,7H)- dione,
l,3,7-Trimethyl-8 3-(piperidin-l-yl)prop-l-yn-l-yl)-lH-purine-2,6(3H,7H)- dione,
8-(3-(Azepan-l-yl)prop-l-yn-l-yl)-l,3,7-trimethyl-lH-purine-2,6(3H,7H)-dione, l,3,7-Trimemyl-8-(3-moφholinopro -l-yn-l-yl)-lH-purine-2,6(3H,7H)-dione, 8-(3-Hydroxy-3-methylbut-l-yn-l-yl)-3,7-dimethyl-l-(5-oxohexyl)-lH-purine- 2,6(3H,7H)-dione,
8-(( 1 -Hydroxycyclohexyl)ethynyl)-3 ,7-dimethyl- 1 -(5-oxohexyl)- 1 H-purine- 2,6(3H,7H)-dione,
8-(( 1 - Aminocyclohexyl)ethynyl)-3 ,7-dimethyl- 1 -(5-oxohexyl)- 1 H-purine- 2,6(3H,7H)-dione,
8-(3-(Bis(2-methoxyethyl)amino)prop-l-yn-l-yl)-3,7-dimethyl-l-(5-oxohexyl)- lH-purine-2,6(3H,7H)-dione, 3 ,7-Dimethyl- l-(5-oxohexyl)-8-(3-(pyrrolidin- l-yl)prop- 1 -yn- 1 -yl)-lH-purine-
2,6(3H,7H)-dione,
3 ,7 -Dimethyl- 1 -(5-oxohexyl> 8--(3-(piperidin- 1 -yl)prop- 1 - yn- 1 ~yl)- 11 I-purine- 2,6(3H,7H)-dione,
1 ,3,7 'lYimethyl 8 -(phenylethynyl) l H- purine 2,6(3H,7H) dione,
1 ,3,9-Trimethyl-8-(3-(pyrrolidin- 1 -yl)prop- 1 -yn-l-yl)- 1 H-purine-2,6(3H,9H)- dione,
1 ,3 ,9-Trimethyl-8-(phenylethynyl)- 1 H-purine-2,6(3H,9H)-dione and
optical isomers, polymorphs, and pharmaceutically-acceptable acid addition salts, hydrates, and solvates thereof.
9. The compound as claimed in any of Claims 1 to 8 for use in the manufacture of a medicament for the treatment or prevention of melanoma, breast adenocarcinoma, hepatocellular carcinoma, neuroblastoma, glioma, lymphoma, and lung cancer.
A process for the synthesis of a compound selected from those of Formula I:
Figure imgf000031_0001
wherein
R represents hydrogen, C1-4alkyl, hydroxy-C2-4alkyl, C1-3alkoxy-C2_4alkyl, Q 3alkylcarbonyl-Ci_4alkyl or C1-3alkyl(C1-3alkyl)amino-C2-4alkyl;
R2 represents C1-4alkyl, hydroxy-C2.4alkyl, Ci^alkylcarbonyl-Ci^alkyl, C1.3alkoxy-C2 4alkyl, C1-3alkyl(C1-3alkyl)amino-C2-4alkyl or halo-C2.4alkyl;
R3 represents Q^alkyl, allyl or C i _3alko y-C2-4alkyl ; with the proviso that if substituent R3 is at purine N(7) atom the dotted line between N(7) and C(8) represents no bond, and the dotted line between C(8) and N(9) represents chemical bond;
and
with the proviso that if substituent R3 is at purine N(9) atom the dotted line between N(7) and C(8) represents chemical bond, and the dotted line between C(8) and N(9) represents no bond;
R4 represents Ci-4alkyl, hydroxy-C^alkyl, C]_3alkoxy-C]-4alkyl, amino-Ci^alkyl, 1-
Figure imgf000032_0001
l-amino-di-(Ci.3alkyl)methyl, l-hydroxy-cyclo-C3- 6alkyl, l-amino-cyclo-C3.6alkyl, l- iydroxy-Q^alky -cycloC^alkyl, Ci_ 3alkylamino-C1-3alkyl, Q-salkyliQ.aalky amino-Ci.salkyl, di-(C1-3alkoxy-C2-4alkyl)- amino-Ci-3alkyl, heterocyclyl-Q^alkyl, aryl or heteroaryl; and its optical isomers, polymorphs and pharmaceutically acceptable acid and base addition salts and hydrates and solvates thereof; comprising reaction of a compound of Formula II:
Figure imgf000032_0002
with a compound of Formula III:
≡-R4
III
optionally in the presence of base in an appropriate solvent (e.g., DIEA in DMF, NMP, DMAC or EtOAc), in the presence of Cul and palladium catalyst (e.g. Pd(PPh3)4) or palladium catalyst generated in situ (e.g., from PdCl2 or Pd(OAc)2 and PPh3) to yield a compound of Formula I, which may be converted, if desired, into an optical isomer, polymorph, pharmaceutically-acceptable salt, hydrate or solvate.
PCT/LV2015/000001 2015-03-27 2015-03-27 Ethynylxanthines, preparation and use for cancer treatment WO2016159745A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
GB1714548.3A GB2553684B (en) 2015-03-27 2015-03-27 Ethynylxanthines, preparation and use for cancer treatment
PCT/LV2015/000001 WO2016159745A1 (en) 2015-03-27 2015-03-27 Ethynylxanthines, preparation and use for cancer treatment
CA2989161A CA2989161C (en) 2015-03-27 2015-03-27 Ethynylxanthines, preparation and use for cancer treatment

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/LV2015/000001 WO2016159745A1 (en) 2015-03-27 2015-03-27 Ethynylxanthines, preparation and use for cancer treatment

Publications (1)

Publication Number Publication Date
WO2016159745A1 true WO2016159745A1 (en) 2016-10-06

Family

ID=52815227

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/LV2015/000001 WO2016159745A1 (en) 2015-03-27 2015-03-27 Ethynylxanthines, preparation and use for cancer treatment

Country Status (3)

Country Link
CA (1) CA2989161C (en)
GB (1) GB2553684B (en)
WO (1) WO2016159745A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2166948C1 (en) 2000-05-29 2001-05-20 Центральный научно-исследовательский рентгенорадиологический институт Method for treating gliomae aggravated with epileptic syndrome
WO2008077557A1 (en) 2006-12-22 2008-07-03 Schwarz Pharma Ag 8-ethinylxanthine derivatives as selective a2a receptor antagonists
WO2009024542A2 (en) * 2007-08-17 2009-02-26 Boehringer Ingelheim International Gmbh Purin derivatives for use in the treatment of fab-related diseases
WO2011005871A1 (en) * 2009-07-07 2011-01-13 Pgxhealth, Llc Substituted 8-[6-carbonylamine-3-pyridyl]xanthines as adenosine a2b antagonists
WO2014143799A2 (en) 2013-03-15 2014-09-18 Hydra Biosciences, Inc. Substituted xanthines and methods of use thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2166948C1 (en) 2000-05-29 2001-05-20 Центральный научно-исследовательский рентгенорадиологический институт Method for treating gliomae aggravated with epileptic syndrome
WO2008077557A1 (en) 2006-12-22 2008-07-03 Schwarz Pharma Ag 8-ethinylxanthine derivatives as selective a2a receptor antagonists
WO2009024542A2 (en) * 2007-08-17 2009-02-26 Boehringer Ingelheim International Gmbh Purin derivatives for use in the treatment of fab-related diseases
WO2011005871A1 (en) * 2009-07-07 2011-01-13 Pgxhealth, Llc Substituted 8-[6-carbonylamine-3-pyridyl]xanthines as adenosine a2b antagonists
WO2014143799A2 (en) 2013-03-15 2014-09-18 Hydra Biosciences, Inc. Substituted xanthines and methods of use thereof

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
ARSENYAN, TETRAHEDRON LETT., vol. 54, 2013, pages 6524 - 6528
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 July 2006 (2006-07-12), XP002739192, Database accession no. 892164-23-7 *
HAYASHI, ANTICANCER RES., vol. 25, 2005, pages 2399 - 2406
HEPATOLOGY, vol. 46, 2007, pages 430 - 435
HUSE, NAT. REV., vol. 10, 2010, pages 319 - 331
JERMAL, CA, CANCER J. CLIN., vol. 60, 2010, pages 277 - 300
JERMAL, CA, CANCER J. CLIN., vol. 61, 2011, pages 69 - 90
KANG, CANCER. RES., vol. 70, 2010, pages 1173 - 83
LAMPSON, DRUG DISCOV. TODAY, vol. 14, 2009, pages 185 - 191
MICHAUD, AM. J. CLIN. NUTR., vol. 92, 2010, pages 1145 - 50
SIEGAL, NEURO ONCOL., 2013
SIEGEL, CA, CANCER J. CLIN., vol. 62, 2012, pages 10 - 29
SYNLETT, vol. 23, 2012, pages 1191 - 1198
VARTANYAN, PSYCHOPHARM. BIOL. NARC., vol. 5, 2005, pages 1093 - 1095

Also Published As

Publication number Publication date
CA2989161A1 (en) 2016-10-06
GB2553684A8 (en) 2018-04-18
GB2553684B (en) 2020-06-03
GB2553684A (en) 2018-03-14
CA2989161C (en) 2022-03-01

Similar Documents

Publication Publication Date Title
KR20060120393A (en) Triazolopyrimidine derivatives as glycogen synthase kinase 3 inhibitors
TW201018696A (en) Fused bicyclic and tricyclic pyrimidine compounds as tyrosine kinase inhibitors
CA2870062A1 (en) Pyrrolopyrazone inhibitors of tankyrase
CN110167941A (en) Substituted fused heteroaryl compounds are as kinase inhibitor and its application
JP2016517845A (en) Novel N (2,3-dihydro-1H-pyrrolo [2,3-b] pyridin-5-yl) -4-quinazolinamine derivatives and N- (2,3-dihydro-1H-indoles as PERK inhibitors -5-yl) -4-quinazolineamine derivatives
US9416143B2 (en) Griseofulvin derivatives
JP2024054247A (en) 1-isopropyl-3-methyl-8-(pyridin-3-yl)-1,3-dihydro-2H-imidazo[4,5-c]cinnolin-2-one and its uses as selective modulators of ataxia telangiectasia mutated (atm) kinase - Patents.com
CN117940432A (en) Camptothecins compound, preparation method and application thereof
EP2960241A1 (en) Method for producing tricyclic compound, and tricyclic compound capable of being produced by said production method
JP2023145547A (en) Cd73 inhibitor, preparation method therefor and application thereof
JP2022116171A (en) 6-fluoro-2-methylbenzo[d]thiazol-5-yl compounds
CN113527300B (en) Bruton&#39;s tyrosine protein kinase inhibitor
El-Gamal et al. Diarylureas and diarylamides with pyrrolo [2, 3-d] pyrimidine scaffold as broad-spectrum anticancer agents
CN109081828B (en) Poly (ADP-ribose) polymerase inhibitor, preparation method and application
JP6606806B2 (en) Deuterated quinazolinone compound and drug composition containing the compound
CN110105356B (en) Azaindole compound and preparation method and application thereof
TW201006836A (en) Anticancer derivatives, preparation thereof and therapeutic use thereof
CN114075219B (en) Quinoline condensed ring derivative, preparation method and medical application thereof
CA2989161C (en) Ethynylxanthines, preparation and use for cancer treatment
WO2016159747A1 (en) Ethynylxanthines, preparation and use as calcium ion channel modulators
CN111433201B (en) Benzepine derivative, process for preparing the same and use thereof in medicine
JP2022554385A (en) WDR5 inhibitors and modulators
WO2016159746A1 (en) Novel ethynylxanthines, their preparation and use as inhibitors of matrix metalloproteinases and angiogenesis
EP3098222B1 (en) Fused acridine derivative and pharmaceutical composition, preparation method and use thereof
RU2615986C1 (en) Substituted methyl(2-{4-[3-(3-methanesulfonylamino-2-fluoro-5-chloro-phenyl)-1h-pyrazol-4-yl]pyrimidin-2-ylamino}ethyl) carbamates, process for their preparation and application

Legal Events

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

Ref document number: 15714972

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 201714548

Country of ref document: GB

Kind code of ref document: A

Free format text: PCT FILING DATE = 20150327

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2989161

Country of ref document: CA

122 Ep: pct application non-entry in european phase

Ref document number: 15714972

Country of ref document: EP

Kind code of ref document: A1