WO2018037120A1 - Promédicaments activés par des espèces réactives de l'oxygène destinés à être utilisés dans le traitement de maladies inflammatoires et du cancer - Google Patents

Promédicaments activés par des espèces réactives de l'oxygène destinés à être utilisés dans le traitement de maladies inflammatoires et du cancer Download PDF

Info

Publication number
WO2018037120A1
WO2018037120A1 PCT/EP2017/071457 EP2017071457W WO2018037120A1 WO 2018037120 A1 WO2018037120 A1 WO 2018037120A1 EP 2017071457 W EP2017071457 W EP 2017071457W WO 2018037120 A1 WO2018037120 A1 WO 2018037120A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
amino
compound
group
phenyl
Prior art date
Application number
PCT/EP2017/071457
Other languages
English (en)
Inventor
Jorge PEIRÓ CADAHÍA
Mads Hartvig Clausen
Jon Bondebjerg
Christian Abildgaard HANSEN
Original Assignee
Danmarks Tekniske Universitet
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 Danmarks Tekniske Universitet filed Critical Danmarks Tekniske Universitet
Publication of WO2018037120A1 publication Critical patent/WO2018037120A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic Table
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds

Definitions

  • the present invention relates to prodrugs which are activated predominantly or exclusively in inflammatory tissue. More particularly, the present invention relates to prodrugs of methotrexate and derivatives thereof, which are selectively activated by Reactive Oxygen Species (ROS) in inflammatory tissues associated with cancer and inflammatory diseases such as rheumatoid arthritis (RA), juvenile dermatomyositis, psoriasis, psoriatic arthritis, lupus, sarcoidosis, Crohn's disease, colitis ulcerosa, multiple sclerosis, Amyotropic Lateral Sclerosis (ALS), atopic dermatitis, eczema etc.
  • ROS Reactive Oxygen Species
  • RA rheumatoid arthritis
  • Prodrugs are masked forms of pharmacologically active agents designed to undergo in vivo activation by specific stimuli.
  • prodrug chemical moieties that makes the drug in question inactive in healthy tissue and selectively activated in diseased tissue the side-effect profile and the selectivity may be improved significantly.
  • ROS Reactive Oxygen Species
  • Methotrexate is a well-known anti-cancer drug, a so-called anti-folate acting by inhibiting the metabolism of folic acid via dihydrofolate reductase. Methotrexate is also widely used as a disease- modifying treatment for some autoimmune diseases, including rheumatoid arthritis, juvenile dermatomyositis, psoriasis, psoriatic arthritis, lupus, sarcoidosis, and Crohn's disease. US 2015/0005352 Al discloses ROS-sensitive prodrug compositions and methods of treating ROS- associated diseases by administering the ROS-sensitive prodrug compositions.
  • WO 2012/123076 Al relates to ferrocene-based compounds and their use as ROS-regulating prodrugs.
  • Therapeutic Delivery 2012, 3, 823-833 discloses the use of boronic acids/esters as triggers for developing ROS-activated anticancer prodrugs that target cancer cells.
  • ACHILLI, C. ET AL. "Folic acid-conjugated 4-Amino-Phenylboronate, a Boron-Containing Compound Designed for Boron Neutron Capture Therapy, is an Unexpected Agonist for Human Neutrophils and Platelets", CHEM BIO DRUG DES, vol. 83, 2013, p. 532-540 discloses folic acid-conjugated 4-amino- phenylboronate as a possible compound for the selective delivery of 10 B in Boron Neutron Capture Therapy (BNCT).
  • BNCT Boron Neutron Capture Therapy
  • ROSOWSKY A ET AL. "SYNTHESIS OF BIOLOGICAL ACTIVITY OF METHOTREXATE ANALOGUES WITH TWO ACID GROUPS AND A HYDROPHOBIC AROMATIC RING IN THE SIDE CHAIN", JOURNAL OF MEDICINAL CHEMISTRY AMERICAN CHEMICAL SOCIETY, US, vol. 34, no. 2, 1 January 1991, p. 574-579 discloses Y-(m-carboxyanilide) and Y-(m-boronoanilide) deivatives of methotrexate and ⁇ - (m-carboxyanilide) derivatives of aminopterin.
  • US 2013/045949 Al discloses compounds that may be selectively activated to produce active anci- cancer agents in tumor cells.
  • US 2014/0378673 Al relates to hypoxia selective prodrugs.
  • prodrugs of ROS-sensitive drug compositions in particular prodrugs of methotrexate, which may be used for site-specific treatment, are stable and lend themselves for up-scaling.
  • prodrugs of ROS-sensitive drug compositions in particular prodrugs of methotrexate, which are selectively activated in inflammatory tissues, have a beneficial cytotoxicity in target cells, low (no) cytotoxicity in healthy cells, are stable, and have a satisfactory bioavailability at the intended site of action.
  • methotrexate of formula I are ROS-sensitive and are selectively activated in inflammatory tissues and thus lend themselves for site-specific treatment with methotrexate.
  • the present invention relates to a compound of the formula I
  • Rl and R2 are independently selected from the group consisting of hydrogen and a moiety of the formula II, III, IV, VII, VIII, IX, X, XI, or XII
  • R3 is selected from the group consisting of hydrogen, Ci -6 alkyl, C 2 - 6 alkenyl, C 2 - 4 alkynyl, C 5 -i 2 aryl, C 4 . 11 heteroaryl and a moiety of the formula II, III, IV, VII, VIII, IX, X, XI, or XII above;
  • R4 and R5 are independently selected from the group consisting of OH, 0-Ci- 6 alkyl, 0-C 2 - 6 alkenyl, 0-C 5 -i 2 aryl, 0-C 4 -uheteroaryl and a moiety of the formula V, VI, XIII, XIV, XV, XVI, or XVII;
  • R6, R7 and R12 are independently selected from the group consisting of hydrogen, CF 3 , C 2 - 6 alkenyl, C 5 -i 2 aryl, and C 4 -uheteroaryl ;
  • R8 and R9 are independently hydroxyl groups or R8 and R9 form, together with the intervening B and O atoms, a pinacol, catechol, diethanolamine, N-methyldiethanolamine or N- methyliminodiacetic acid (MIDA) boronate group;
  • MIDA N-methyliminodiacetic acid
  • W and Q are independently C or N; wherein each of X, Y and Z are selected from the group consisting of halogen, amino, nitro, cyano, hydroxyl, CF 3 , Ci -6 alkyl, Ci- 6 alkoxy, C 2 - 6 alkenyl, C 2 - 6 alkenyloxy, C 6 -i 2 aryl, C 4 -uheteroaryl ; wherein each of said Ci- 6 alkyl, C 2 - 6 alkenyl, C 6 -i 2 aryl, C 4 -uheteroaryl may be substituted by one or more substituents selected from the group consisting of halogen, amino, nitro, cyano, hydroxyl, CF 3 , and Ci- 6 alkyl; and each of a, b and c are integers in the range 0-4;
  • X' and Y' are independently S or O, and R', R", R'" and R"" are independently selected from hydrogen, Ci- 5 alkyl, C 2 - 6 alkenyl, C 5 -i 2 aryl, C 4 -uheteroaryl, and Ci- 5 alkyl-C 5 -i 2 aryl ; wherein, if each of Rl, R2 and R3 are different from a moiety selected from a moiety of the formula II, III, IV, VII, VIII, IX, X, XI, or XII then at least one of R4 and R5 is a moiety of the formula V, XIII, XIV, XV, XVI, or XVII; as well as pharmaceutically acceptable salts, solvates, and stereoisomers thereof.
  • the present invention relates to a method for the preparation of a compound according to the invention, comprising the steps: a) Providing methotrexate (MTX) of the formula 12 or aminopterin (AMT) of the formula 31
  • RIO and Rl l is a leaving group LG; and c) Reacting optionally protected MTX (12) or optionally protected aminopterin (31) with a compound of formula Ila, Ilia, IVa, Vila, Villa, IXa, Xa, XIa, Xlla, XVIIIa, XlXa, XXa, XXIa, XXIIa, or XXIIIa to obtain a compound of formula I according to the invention;
  • step c) or d) optionally reacting the compound obtained in step c) or d), as appropriate, with a compound of the formula Ila, Ilia, IVa, Via, Vila, Villa, IXa, Xa, XIa, Xlla, XVIIIa, XlXa, XXa, XXIa, XXIIa, or XXIIIa, followed by an optional deprotection step to obtain a compound of formula I according to the invention.
  • a compound of the formula Ila, Ilia, IVa, Via, Vila, Villa, IXa, Xa, XIa, Xlla, XVIIIa, XlXa, XXa, XXIa, XXIIa, or XXIIIa followed by an optional deprotection step to obtain a compound of formula I according to the invention.
  • a peptide coupling agent e.g . BOP, PyBOP, DCC, EDC, HATU, HOBt, etc. followed by addition of a compound of the formula XVIIIa, XlXa, or XXa, and a final deprotection step when needed, to obtain a compound of the formula I.
  • Suitable coupling agents are known to a person skilled in the art and are disclosed in e.g . Chem. Rev. , 2011, 111 (11), 6557-6602.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to the invention, optionally in combination with one or more excipients.
  • the present invention relates to a compound according to the invention as a prodrug for the treatment of inflammatory diseases or cancer.
  • FIGURE 2 shows MCF-7 in vitro cell viability assay incubated with compounds 12 (methotrexate) and prodrug 16
  • FIGURE 3 shows in vitro cell viability study of MCF-7 cells incubated for 48 h with 0.25, 0.062 and 0.015 ⁇ concentrations of compounds 12 (methotrexate) and 16;
  • FIGURE 4 shows NCI-H460 in vitro cell viability assay incubated with compounds 12 (methotrexate) and prodrug 16
  • FIGURE 6 shows activation of prodrugs under oxidative conditions (H 2 0 2 );
  • FIGURE 7 shows NCI-H460 in vitro cell viability assay incubated with compounds 31 (aminopterin) and prodrug 23;
  • FIGURE 8 shows MCF-7 in vitro cell viability assay incubated with compounds 31 (aminopterin) and prodrug 23;
  • alkyl means a linear, cyclic or branched hydrocarbon group having 1 to 24 carbon atoms, such as methyl, ethyl, propyl, /so-propyl, cyclopropyl, butyl, /so-butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl, hexyl, and cyclohexyl .
  • alkenyl means a linear, cyclic or branched hydrocarbon groups having 2 to 24 carbon atoms, and comprising (at least) one unsaturated bond .
  • alkenyl groups are vinyl, allyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl and decaenyl .
  • Preferred examples of alkenyl are vinyl, allyl, butenyl, especially allyl .
  • alkynyl means a linear, cyclic or branched hydrocarbon groups having 2 to 24 carbon atoms, and comprising (at least) one triple bond. Examples of alkynyl groups are acetylene, propynyl, butynyl, pentynyl, and hexynyl.
  • halogen includes fluoro, chloro, bromo, and iodo.
  • alkoxy refers to a group -OR, wherein R is alkyl as defined above.
  • alkenyloxy refers to a group -OR, wherein R is alkenyl as defined above.
  • aryl refers to an unsaturated cyclic system.
  • Aryl groups may comprise from 4-12 atoms, suitably from 6-8 atoms, most suitably 6 atoms.
  • “Aryl” is preferably phenyl (-C 5 H 5 ).
  • aromatic is intended to mean a carbocyclic ring system, such as phenyl, naphthyl, 1,2,3,4-tetrahydronaphthyl, anthracyl, phenanthracyl, pyrenyl, benzopyrenyl, fluorenyl and xanthenyl.
  • heteroaryl groups are oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, coumaryl, furanyl, thienyl, quinolyl, benzothiazolyl, benzotriazolyl, benzodiazolyl, benzooxozolyl, phthalazinyl, phthalanyl, triazolyl, tetrazolyl, isoquinolyl, acridinyl, carbazolyl, dibenzazepinyl, indolyl, benzopyrazolyl, phenoxazonyl.
  • heteroaryl groups are benzimidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, furyl, thienyl, quinolyl, triazolyl, tetrazolyl, isoquinolyl, indolyl in particular benzimidazolyl, pyrrolyl, imidazolyl, pyridinyl, pyrimidinyl, furyl, thienyl, quinolyl, tetrazolyl, and isoquinolyl.
  • pharmaceutically acceptable salt is intended to indicate salts prepared by reacting a compound of formula I with a suitable inorganic or organic acid, such as hydrochloric, hydrobromic, hydroiodic, sulfuric, nitric, phosphoric, formic, acetic, 2,2-dichloroacetic, choline, adipic, ascorbic, L- aspartic, L-glutamic, galactaric, lactic, maleic, L-malic, phthalic, citric, propionic, benzoic, glutaric, gluconic, D-glucuronic, methanesulfonic, salicylic, succinic, malonic, tartaric, benzenesulfonic, ethane-l,2-disulfonic, 2-hydroxy ethanesulfonic acid, toluenesulfonic, sulfamic or fumaric acid .
  • a suitable inorganic or organic acid such as hydrochloric, hydrobromic, hydroiodic,
  • Pharmaceutically acceptable salts of compounds of formula I may also be prepared by reaction with a suitable base such as sodium hydroxide, potassium hydroxide, magnesium hydroxide, calcium hydroxide, ammonia, or suitable non-toxic amines, such as lower alkylamines, for example triethylamine, hydroxy-lower alkylamines, for example 2-hydroxyethylamine, bis-(2-hydroxyethyl)- amine, cycloalkylamines, for example dicyclohexylamine, or benzylamines, for example ⁇ , ⁇ ' - dibenzylethylenediamine, and dibenzylamine, or L-arginine or L-lysine.
  • a suitable base such as sodium hydroxide, potassium hydroxide, magnesium hydroxide, calcium hydroxide, ammonia, or suitable non-toxic amines, such as lower alkylamines, for example triethylamine, hydroxy-lower alkylamines, for example 2-hydroxyethyl
  • solvate is intended to indicate a species formed by interaction between a compound, e.g . a compound of formula I, and a solvent, e.g. alcohol, glycerol or water, wherein said species is in a solid form .
  • a solvent e.g. alcohol, glycerol or water
  • water is the solvent
  • said species is referred to as a hydrate.
  • the compounds of the formula I according to the invention may be prepared by the following steps: a) Providing methotrexate (MTX) of the formula 12 or aminopterin (AMT) of the formula 31 or any protected version of them. ;
  • R4, R5, R6, R7, R8, R9, R12, R', R", R'", R"", W, Q, X, X', Y, Y' Z, a, b, c are as defined above,
  • RIO and Rll is a leaving group LG; and c) Reacting optionally protected MTX ( 12) or optionally protected aminopterin (31) with a compound of formula Ila, Ilia, IVa, Vila, Villa, IXa, Xa, XIa, Xlla, XVIIIa, XlXa, XXa, XXIa, XXIIa, or XXIIIa to obtain a compound of formula I according to the invention ;
  • step c) or d) optionally reacting the compound obtained in step c) or d), as appropriate, with a compound of the formula Ila, Ilia, IVa, Via, Vila, Villa, IXa, Xa, XIa, Xlla, XVIIIa, XlXa, XXa, XXIa, XXIIa, or XXIIIa, followed by an optional deprotection step to obtain a compound of formula I according to the invention .
  • a compound of the formula Ila, Ilia, IVa, Via, Vila, Villa, IXa, Xa, XIa, Xlla, XVIIIa, XlXa, XXa, XXIa, XXIIa, or XXIIIa followed by an optional deprotection step to obtain a compound of formula I according to the invention .
  • Methotrexate (MTX) of the formula 12 or aminopterin of the formula 31 are prepared with protecting groups at the desired positions when necessary. Suitable protective groups are known to a person skilled in the art and are disclosed in e.g . Wuts, P. G . M . & Greene, T. W. Greene's Protective Groups in Organic Synthesis. (Wiley, 2006) .
  • Step b A non-limiting example of the process step a can be found in Preparation Example 8, 9 and 21 below. Step b
  • a compound of the formula Ila, Ilia, IVa, Via, Vila, Villa, IXa, Xa, XIa, Xlla, XVIIIa, XlXa, XXa, XXIa, XXIIa, or XXIIIa may be provided commercially or may be prepared by a method known per se from commercially available starting compounds.
  • Non-limiting examples of possible leaving groups include CI, Br, I, CDI, p-nitrophenol, etc..
  • a compound of the formula Ila, Ilia or IVa may be prepared from the corresponding alcohol, transforming it into reactive species such a chloroformate or halide.
  • Non-limiting examples are shown in Preparation Examples 1, 2 and 3, wherein the synthesis of compounds 1 , 3 and 5 is illustrated .
  • a compound of the formula XXIa may be prepared from the corresponding amine, transforming it into the reactive isocyanate.
  • Non-limiting examples are shown in Preparation Example 22, wherein the synthesis of compound 34 is illustrated .
  • Step c Optionally protected methotrexate (MTX) of the formula 12 or aminopterin of the formula 31 is reacted with one or more compounds of the formula Ila, Ilia, IVa, Vila, Villa, IXa, Xa, XIa, Xlla, XVIIIa, XlXa, XXa, XXIa, XXIIa, XXIIIa in a manner known per se.
  • Illustrative, non-limiting examples of said reaction can be found in Examples 1, 2, 3, 4, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 below.
  • Step e The compound obtained in step c) or d), as appropriate, may optionally be reacted with a compound of the formula Ila, Ilia, IVa, Via, Vila, Villa, IXa, Xa, XIa, Xlla, XVIIIa, XlXa, XXa, XXIa, XXIIa, or XXIIIa, followed by an optional deprotection step, to obtain a compound of formula I according to the invention.
  • Illustrative, non-limiting examples of said reaction can be found in Examples 19, 20, 21, 22, 23, 24 below.
  • a peptide coupling agent e.g . BOP, PyBOP, DCC, EDC, HATU, HOBt, etc. followed by addition of a compound of the formula XVIIIa, XlXa, or XXa, and a final deprotection step when needed, to obtain a compound of the formula I.
  • Suitable coupling agents are known to a person skilled in the art and are disclosed in e.g . Chem. Rev. , 2011, 111 (11), 6557-6602.
  • An embodiment of the invention is a compound of the formula I, wherein Rl and R2 are independently selected from the group consisting of hydrogen and a moiety of the formula II, III, or IV.
  • An embodiment of the invention is a compound of the formula I, wherein R3 is selected from the group consisting of hydrogen, Ci_ 5 alkyl, C 2 - 5 alkenyl, C 2 . 4 alkynyl, C 5 _i 2 aryl, C 4 .nheteroaryl and a moiety of the formula II, III, or IV.
  • An embodiment of the invention is a compound of the formula I, wherein R4 and R5 are independently selected from the group consisting of OH, 0-Ci- 6 alkyl, 0-C 2 - 6 alkenyl, 0-C 6 -i 2 aryl, O- C 4 -uheteroaryl and a moiety of the formula V, or VI
  • An embodiment of the invention is a compound of the formula I, wherein Rl and R2 are independently selected from the group consisting of hydrogen and a moiety of the formula II, III, or IV;
  • R3 is selected from the group consisting of hydrogen, Ci- 6 alkyl, C 2 - 6 alkenyl, C 2 . 4 alkynyl, C 6 -i 2 aryl, C 4 . 11 heteroaryl and a moiety of the formula II, III, or IV;
  • R4 and R5 are independently selected from the group consisting of OH, 0-Ci- 6 alkyl, 0-C 2 - 6 alkenyl, 0-C 6 -i 2 aryl, 0-C 4 -uheteroaryl and a moiety of the formula V, or VI; and wherein, if each of Rl, R2 and R3 are different from a moiety selected from a moiety of the formula II, III, and IV, then at least one of R4 and R5 is a moiety of the formula V.
  • An embodiment of the invention is a compound of the formula I, wherein W and Q are both C.
  • Another embodiment of the invention is a compound of the formula I, wherein W is C and Q is N.
  • Another embodiment of the invention is a compound of the formula I, wherein W is N and Q is C.
  • Another embodiment of the invention is a compound of the formula I, wherein W and Q are both N.
  • An embodiment of the invention is a compound of formula I, wherein R6 and R7 are independently selected from the group consisting of hydrogen and Ci- 6 alkyl, preferably selected from the group consisting of hydrogen and Ci- 4 alkyl, preferably selected from the group consisting of hydrogen and methyl, preferably wherein R6 and R7 are both hydrogen.
  • An embodiment of the invention is a compound of formula I, wherein R8 and R9 are independently hydroxyl groups or R8 and R9 form, together with the intervening B and O atoms, a pinacol or catechol group, preferably wherein R8 and R9 are independently hydroxyl groups or R8 and R9 form, together with the intervening B and O atoms, a pinacol group, preferably wherein R8 and R9 are both hydroxyl groups.
  • An embodiment of the invention is a compound of formula I, wherein each of X, Y and Z are selected from the group consisting of halogen, cyano, hydroxyl, CF 3 , and Ci- 6 alkyl ; and each of a, b and c are 0, 1 or 2.
  • An embodiment of the invention is a compound of formula I, wherein each of X, Y and Z are selected from the group consisting of halogen, cyano, hydroxyl, and Ci- 6 alkyl ; and each of a, b and c are 0 or 1.
  • An embodiment of the invention is a compound of formula I, wherein each of X, Y and Z are selected from the group consisting of halogen and Ci- 4 alkyl; and each of a, b and c are 0 or 1.
  • An embodiment of the invention is a compound of formula I, wherein each of X, Y and Z are selected from the group consisting of fluoro and methyl; and each of a, b and c are 0 or 1.
  • An embodiment of the invention is a compound of the formula I, wherein R2 is selected from the group consisting of
  • Rl is hydrogen
  • R3 is selected from the group consisting of hydrogen and Ci_ 5 alkyl, preferably selected from the group consisting of hydrogen and Ci- 4 alkyl, preferably wherein said R3 is methyl;
  • R4 and R5 are selected from the group consisting of OH and 0-Ci- 5 alkyl, preferably selected from the group consisting of OH and 0-Ci- 4 alkyl, preferably wherein R4 and R5 are both methoxy or hydroxy.
  • An embodiment of the invention is a compound of the formula I, wherein R3 is selected from the group consisting of
  • Rl and R2 are hydrogen
  • R4 and R5 are selected from the group consisting of OH and 0-Ci_ 5 alkyl, preferably selected from the group consisting of OH and 0-Ci- 4 alkyl, preferably wherein R4 and R5 are both methoxy or hydroxy.
  • An embodiment of the invention is a compound of the formula I, wherein R4 and/or R5 is a moiety of the formula
  • Rl and R2 are hydrogen
  • R3 is selected from the group consisting of hydrogen and Ci- 6 alkyl, preferably selected from the group consisting of hydrogen and Ci- 4 alkyl, preferably wherein said R3 is methyl .
  • An embodiment of the invention is a compound of the formula I, wherein Y' is S.
  • An embodiment of the invention is a compound of the formula I, wherein X' is O.
  • An embodiment of the invention is a compound of the formula I, wherein R' and R" are both hydrogen .
  • Compounds of formula I may comprise asymmetrically substituted (chiral) carbon atoms and carbon-carbon double bonds which may give rise to the existence of stereoisomeric forms, e.g . enantiomers, diastereomers and geometric isomers.
  • the present invention includes all such isomers, either in pure form or as mixtures thereof.
  • the compounds of formula I may be obtained in crystalline form either directly by concentration from an organic solvent or by crystallisation or recrystallisation from an organic solvent or mixture of said solvent and a cosolvent that may be organic or inorganic, such as water.
  • the crystals may be isolated in essentially solvent-free form or as a solvate, such as a hydrate.
  • the invention covers all crystalline modifications and forms and also mixtures thereof.
  • the present invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising as an active ingredient an effective amount of at least one compound of the Formula I, or pharmaceutically acceptable salt thereof and/or stereoisomer thereof, optionally in combination with one or more conventional excipients.
  • the pharmaceutical composition of the present invention usually comprises 0.1-90wt% of the compound of Formula I and/or physiologically acceptable salt thereof.
  • the pharmaceutical composition can be prepared according to methods known in the art. For this purpose, if necessary, the compound of Formula I and/or a stereoisomer thereof is combined with one or more solid or liquid pharmaceutically acceptable excipients and/or adjuvants, to form an application form or dosage form suitable for administration to a human.
  • the compound of Formula I of the present invention or the pharmaceutical composition containing the same can be administered in unit dosage form, and the administration routes can be intestinal or parenteral administration, such as oral, intramuscular, subcutaneous, nasal, oral mucosal, skin, intraperitoneal or rectal administration.
  • the administration dosage form can be, for example, tablets, capsules, drop pills, aerosols, pills, powders, solutions, suspensions, emulsions, granules, liposomes, transdermal agents, buccal tablets, suppositories, lyophilized powder injections, can be normal preparations, sustained-release preparations, controlled-release preparations, and various microparticle administration systems.
  • various carriers well known in the art can be widely used.
  • the examples of the carriers can be, for example, diluents and absorbents, such as starch, dextrin, calcium sulfate, lactose, mannitol, sucrose, sodium chloride, glucose, urea, calcium carbonate, kaolin, microcrystalline cellulose, aluminum silicate; wetting agent and binding agent, such as water, glycerol, polyethylene glycol, ethanol, propanol, starch slurry, dextrin, syrup, honey, glucose solution, acacia mucilage, gelatin mucilage, sodium carboxymethylcellulose, shellac, methylcellulose, potassium phosphate, polyvinylpyrrolidone; disintegrants, such as, dry starch powder, alginate, agar powder, laminarin powder, sodium hydrogen carbonate and citric acid, calcium carbonate, polyoxyethylene sorbitol fatty acid ester, sodium dodecyl sulfate, methyl cellulose, ethyl cellulose; disintegr
  • the tablets can be further processed into coated tablets, for example, sugar coated tablets, thin film coated tablets, enteric-coated tablets, or double-layer tablets or multi-layer tablets.
  • various carriers known in the art can be used.
  • the examples of the carriers can be, for example, diluents and absorbing agents, such as glucose, lactose, starch, cocoa butter, hydrogenated vegetable oil, polyvinylpyrrolidone, Gelucire, kaolin, talc; binding agent, such as acacia gum, tragacanth gum, gelatin, ethanol, honey, liquid sugar, rice paste or panada; disintegrants, such as agar powder, dry starch powder, alginate, sodium dodecyl sulfonate, methyl cellulose, ethyl cellulose.
  • various carriers known in the art can be widely used.
  • the examples of the carriers can be, for example, polyethylene glycol, lecithin, cocoa butter, fatty alcohol, ester of fatty alcohol, gelatin, semi-synthetic ester.
  • the compound of Formula I or stereoisomer thereof as effective component is mixed with the various carriers, and the resultant mixture is placed in hard gelatin capsule shells or soft capsules.
  • the compound of Formula I or stereoisomer thereof as effective component can also be processed into microcapsules, suspended in aqueous medium to form a suspension, or placed in hard capsules or processed into injections.
  • a preparation for injection such as solution, emulsion, lyophilized powder injection and suspension
  • all diluents known in the art for example, water, ethanol, polyethylene glycol, 1,3-propylene glycol, ethoxylated isostearyl alcohol, multi-oxidized isostearyl alcohol, polyoxyethylene sorbitol fatty acid ester, could be used.
  • an isotonic injection solution an suitable amount of sodium chloride, glucose or glycerol can be added to the injection preparation, and conventional co-solvent, buffer agent, and pH regulator can further added.
  • flavouring agents preservatives, flavoring agents, correctants, sweetening agents or other materials can also be added to the pharmaceutical compositions.
  • the administration dose of the compound of Formula I, or a stereoisomer thereof may depend on many factors, for example, the properties and severity of the diseases to be prevented or treated, the gender, age, bodyweight and individual reaction of patient or animal, the specific compound to be used, the administration routes and times, and so on.
  • the dose can be of single dose form or can be divided into several dose forms, such as, two, three or four dose forms.
  • the compounds according to the invention may be used as a prodrug for the treatment of inflammatory diseases or cancer.
  • inflammatory diseases include rheumatoid arthritis (RA), juvenile dermatomyositis, juvenile rheumatoid arthritis, psoriasis, psoriatic arthritis, lupus, sarcoidosis, atopic dermatitis, eczema, Crohn's disease, uveitis associated with juvenile idiopathic arthritis or ulcerative colitis, colitis ulcerosa, multiple sclerosis, Amyotropic Lateral Sclerosis (ALS), non-infectious ocular inflammation, vasculitis, systemic lupus erythematosus, and eosinophilic fasciitis.
  • RA rheumatoid arthritis
  • psoriasis juvenile rheumatoid arthritis
  • psoriatic arthritis psoriatic arthritis
  • lupus sarcoidosis
  • Non-limiting examples of cancer diseases include acute lymphocytic leukemia, meningeal leukemia, myeloproliferative neoplasm, breast cancer, squamous cell carcinoma, lymphosarcoma, osteosarcoma, advanced mycosis fungoides (cutaneous T cell lymphoma), small cell types lung cancer, non-small cell lung cancer, and non-Hodgkin's lymphoma.
  • Analytical TLC was conducted on Merck aluminium sheets covered with silica (C60) .
  • the plates were either visualized under UV-light or stained by dipping in a developing agent followed by heating .
  • KMn0 4 (3 g in water (300 mL) along with K 2 C0 3 (20 g) and 5% aqueous NaOH (5 mL)) or Ninhydrin (3 g in a mixture of n-butanol (200 mL) and AcOH (6 mL)) were used as developing agents.
  • Flash column chromatography was performed using Matrex 60 A, 35-70 ⁇ silicagel.
  • Method A eluents A (0.1% HC0 2 H in mili-Q water) and B (0.1% HC0 2 H in CH 3 CN) were used in a linear gradient (5% B to 100% B) in a total run time of 2.6 min.
  • Method B eluents A (0.1% HC0 2 H in H 2 0) and B (0.1% HC0 2 H in CH 3 CN) were used in a linear gradient (5% B to 100% B) in a total run time of 5.0 min.
  • Method C eluents A (lOmM NH 4 OAc in mili-Q water) and B (0.1% NH 4 OAc in mili-Q water /MeCN, 90/10, v/v) were used in a linear gradient (5% B to 100% B) in a total run time of 2.6 min.
  • Method D eluents A (0.1% NH 4 OAc in H 2 0) and B (0.1% NH 4 OAc in CH 3 CN) were used in a linear gradient (5% B to 100% B) in a total run time of 5.0 min
  • the LC system was coupled to a SQD mass spectrometer operating in both positive and negative electrospray modes. The temperature for all recordings was approximately 20 °C.
  • Analytical LC- HRMS (ESI) analysis was performed on an Agilent 1100 RP-LC system equipped with a diode array detector using a Phenomenex Luna C18 column (d 3 ⁇ , 2.1 x 50 mm ; column temp: 40 °C; flow: 0.4 mL/min) .
  • Eluents A (0.1% HC0 2 H in H 2 0) and B (0.1% HC0 2 H in CH 3 CN) were used in a linear gradient (20% B to 100% B) in a total run time of 15 min.
  • the LC system was coupled to a Micromass LCT orthogonal time-of-flight mass spectrometer equipped with a Lock Mass probe operating in positive or negative electrospray mode.
  • Elution was carried out in a reversed-phase gradient fashion combining Al (0.1% HC0 2 H in mili-Q water) and Bl (0.1% HC0 2 H in CH 3 CN) or A2 (5 m M N H 4 OAc in H 2 0) and B2 (5 m M N H 4 OAc in CH 3 CN) : 5% B to 70 % B in 10 min, hold for 3.5 min, then 70% B to 100% B in 1.5 min, and hold 3 minutes. Total run time : 20 min .
  • 2,4-diamino-6-(hydroxymethyl)pteridine hydrochloride (4.40 g, 19.2 mmol) was dissolved in hot water (150 mL) and after cooling to 21 °C the solution was neutralized with 1M NaOH aq. solution to pH 7 (ca. 20 mL) . The formed precipitates were collected by filtration, washed with water, and dried in vacuo over P 2 0 5 to afford an orange-beige solid corresponding to 2,4-diamino-6- (hydroxymethyl)pteridine. The solid was suspended in dry DMAc (25 mL) and triphenylphosphine dibromide ( 18.1 g, 42.9 mmol) was added to the suspension.
  • the reaction was allowed to warm to 21 °C and stirred for 5 h under a N 2 atmosphere.
  • the mixture was diluted with CH 2 CI 2 (100 mL), washed with 1M HCI (2 x75 mL), sat. NaHC0 3 (2 x 75mL), and brine (75 mL) .
  • the organic phase was dried over Na 2 S0 4 , filtered, and concentrated in vacuo to afford a yellow solid (770 mg) that was purified by preparative HPLC.
  • the CH 3 CN/H 2 0 fractions containing the pinacolate intermediate were poured together into a 250mL round bottom flask and HCI cc. (0.3 mL, ca. pH 2) was added .
  • reaction mixture was stirred for 16 h at 21 °C and quenched with sat. NaHC0 3 (ca. 50 mL) . After removal of the CH 3 CN in vacuo the formed precipitate was filtered, washed with H 2 0 and dried in vacuo to afford the title compound 9 (122 mg, 27%) as a yellow solid.
  • the solid residue was purified by preparative HPLC.
  • the CH 3 CN/H 2 0 fractions containing the pinacolate intermediate were poured together into a 250mL round bottom flask and HCI cc. (0.3 mL, ca. pH 2) was added.
  • the reaction mixture was stirred for 16h at room temperature and quenched with sat. NaHC0 3 (ca. 50 mL) .
  • After removal of the CH 3 CN in vacuo the formed precipitate was filtered, washed with H 2 0 and dried in vacuo to afford the title compound 10 (179 mg, 22%) as a yellow solid.
  • the reaction was allowed to warm to 21 °C and stirred for 5 h under a N 2 atmosphere.
  • the mixture was diluted with CH 2 CI 2 (100 mL), washed with 1M HCI (2 x75 mL), sat. NaHC0 3 (2 x 75mL) and brine (75 mL) .
  • the organic phase was dried over Na 2 S0 4 , filtered and concentrated in vacuo.
  • the solid residue was purified by preparative HPLC.
  • the CH 3 CN/H 2 0 fractions containing the pinacolate intermediate were poured together into a 250mL round bottom flask and HCI cc. (0.3 mL, ca. pH 2) was added .
  • methotrexate (12) 1.0 g, 2.20 mmol
  • anhydrous DMF 50 mL
  • 1, 1,3,3-tetramethylguanidine 0.55 mL, 4.4 mmol
  • 4-methoxybenzyl chloride 0.59 mL, 4.4 mmol
  • reaction mixture was stirred for 16h at 21 °C and quenched with sat. NaHC0 3 (ca. 50 mL) . After removal of CH 3 CN in vacuo, the precipitate was filtered, washed with H 2 0 and dried in vacuo to afford 30 as a yellow solid (131 mg, 39%) .
  • the reaction was allowed to warm to 21 °C and stirred for 5 h under a N 2 atmosphere.
  • the crude mixture was concentrated in vacuo to afford a dark yellow solid .
  • the solid residue was purified by preparative HPLC.
  • the CH 3 CN/H 2 0 fractions containing the pinacolate intermediate were poured together into a 250mL round bottom flask and HCI cc. (0.3 mL, ca. pH 2) was added .
  • the reaction mixture was stirred for 16h at room temperature and quenched with excess of sat. NaHC0 3 (ca. 50 mL) .
  • the reaction was allowed to warm to 21 °C and stirred for 16 h under a N 2 atmosphere.
  • the mixture was diluted with CH 2 CI 2 (100 mL), washed with 1 M aq. HCI (2 x75 ml_), sat. aq. NaHC0 3 (2 x 75mL), and brine (75 mL).
  • the organic phase was dried over Na 2 S0 4 , filtered, and concentrated in vacuo to afford a solid that was purified by preparative HPLC.
  • the CH 3 CN/H 2 0 fractions containing the pinacolate intermediate were poured together and HCI cc. was added until pH ⁇ 2.
  • the reaction mixture was stirred for 16 h at 21 °C and quenched with sat.
  • Chemical Stability pH 7.4 / PBS stability Chemical stability at pH 7.4 for compounds 16 and 23 was assessed using diclofenac as an internal standard.
  • To 490 ⁇ _ of a pre-warmed solution of 20 ⁇ diclofenac in PBS (0.1% DMSO) is added 10 ⁇ ⁇ . of a ImM DMSO solution of compound 16 and 23 (in triplicates) .
  • the mixture was incubated at 1000 rpm at 37 °C in an Eppendorf Thermomixer C ( 1.5 mL) and samples were taken for analysis by RP-UPLC-MS after 30 min, lh, 2h, 4h, 8h, 24h and 48h.
  • Solubility Kinetic solubility at 100 ⁇ with 1% DMSO. Briefly, the kinetic solubility, utilizing test compound from 10 mM DMSO stock solution, is measured at a final compound concentration of 100 ⁇ and 1% DMSO. Test compound is added to 100 mM potassium phosphate buffer, pH 7.4, and incubated at 37 °C for 20 hours in a heater-shaker. After incubation, the samples are centrifuged at 3000xg at 37 °C for 30 min to pellet insoluble material and an aliquot of the supernatant is taken for analysis. After dilution of the sample, the concentration of dissolved compound is quantified by LC-MS/MS analysis. Plasma protein binding assay.
  • the fraction unbound drug (f u ) in plasma from human or other animal species was determined by equilibrium dialysis at 37 °C for 4 hours using a Rapid Equilibrium Dialysis (RED) device.
  • the drug molecule at a concentration of 10 ⁇ is added to 50% plasma and dialyzed against isotonic phosphate buffer (67 mM, pH 7.4). After dialysis, the drug concentration in the buffer and plasma is quantified by LC-MS/MS analysis.
  • the stability of the drug molecule in plasma is determined by incubating drug- spiked plasma (10 ⁇ ) at 37 °C for 4 hours, meanwhile the control plasma sample is kept in the freezer. The concentration of drug in both samples is quantified by LC-MS/MS analysis.
  • the in vitro metabolic stability assay uses liver microsomes. Compound is dissolved in 100 mM KP0 4 buffer pH 7.4 to a ⁇ final concentration. The assay is initiated by addition of NADPH and incubated for up to 40 min (THERMOstar, BMG Lab Technologies) with microsomes. Experiments are terminated at different time points by addition of acetonitrile. The amount of parent compound remaining is analyzed by LC-MS/MS. The natural logarithm of relative amount parent compound remaining is plotted against time and the first-order rate constant of consumption is determined by linear regression. In vitro half life is expressed in minutes and in vitro clearance in ⁇ /img x min), respectively.
  • the human breast cancer MCF-7 (Sigma) and human large cell lung cancer NCIH-460 (ATTC) cell lines were cultured in a humidified, 5% C0 2 atmosphere at 37 °C in Dulbecco ' s Modified Eagle ' s Medium (DMEM) (Sigma) or Roswell Park Memorial Institute medium (RPMI) 1640 (Sigma) supplemented with 10% fetal bovine serum (FBS, heat- inactivated, Fisher Scientific) and 1% penicillin/streptomycin. Both cell lines were subcultured every 2-3 days.
  • DMEM Dulbecco ' s Modified Eagle ' s Medium
  • RPMI 1640 Roswell Park Memorial Institute medium
  • FBS fetal bovine serum
  • Fisher Scientific heat- inactivated, Fisher Scientific
  • MTS 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2- (4-sulfophenyl)-2H-te-trazolium (MTS) assay (Promega Biotech AB, Sweden ) was used to determine the in vitro antiproliferative effect of the compounds. This assay is based on the principle that cells have the ability to reduce MTS tetrazolium, while, when dead, they lose this ability. MCF-7 and NCI-H460 cells were cultured in 96-well plates at an initial density of 10 4 cells/well (MCF-7) or 7xl0 3 cells/well (NCI-H460) in their respective growth medium.
  • the medium was removed and the cells were incubated in the presence or absence of pre-activated compounds at different concentrations.
  • the MTS reagent was added to each well .
  • the cells were further incubated for a period of time between 30-60 min at 37 °C until colorimetric reaction was developed within the linear range and the absorbance of the samples was measured at 490 nm using a 96-well plate spectrophotometer (Victor 3 plate reader with Wallac 1420 Workstation vs 3.0 software).
  • a control was used for each tested compound, where cells were incubated with DMEM or RPMI containing the equivalent concentration of DMSO (maximum of 0.4% v/v). Each concentration of tested compounds was done in triplicates. The final concentration of H202 in each well was always ⁇ 10 ⁇ (non-cytotoxic concentration in MCF-7 and NCI-H460 cell lines, determined with the described assay). The IC 50 values were calculated using GraphPad Prism v6.0 (California, USA) as the concentration of the compounds required to cause 50% response compared to cells exposed to controls using a non-linear dose-response regression.
  • FIGURE 2 shows MCF-7 in vitro cell viability assay incubated with compounds 12 (methotrexate) and prodrug 16.
  • Cells were incubated at increasing concentrations of tested compouns for 48 h before MTS reagent was added .
  • Pre-activation of tested compounds with H 2 0 2 was performed 24 h before experiment started as described in the experimental section.
  • FIGURE 4 shows NCI-H460 in vitro cell viability assay incubated with compounds 12 (methotrexate) and prodrug 16. Cells were incubated at increasing concentrations of tested compouns for 48 h before MTS reagent was added .
  • Pre-activation of tested compounds with H 2 0 2 was performed 24 h before experiment started as described in the experimental section.
  • FIGURE 6 shows activation of prodrugs under oxidative conditions (H 2 0 2 ) . The activation was run at a test compound concentration of 50 ⁇ and H 2 0 2 of 0.5 mM in a mixture of 30% DMSO in PBS. The experiment was run in triplicates. Values are represented as mean and error bars as SD. Error bars not shown are smaller than the symbol .
  • FIGURE 7 shows NCI-H460 in vitro cell viability assay incubated with compounds 31 (aminopterin) and prodrug 23.
  • Cells were incubated at increasing concentrations of tested compounds for 48 h before MTS reagent was added .
  • Pre-activation of tested compounds with H 2 0 2 was performed 24 h before experiment started as described in the experimental section.
  • FIGURE 8 shows MCF-7 in vitro cell viability assay incubated with compounds 31 (aminopterin) and prodrug 23.
  • mice Male, 8-9 weeks were obtained from Janvier, France. The mice were maintained in the animal house at Redoxis, Medicon Village, Lund, Sweden, where they were acclimatized for approximately one week before initiation of the experiment. All animal experiments were approved by the local animal ethic committee Malmo/Lund, Sweden, approved under the license N165-15.
  • CIA collagen induced arthritis
  • CFA Complete Freund's Adjuvant
  • Vehicle and compound 2% DMSO in PBS, Life Technologies, injection volume 370 ⁇
  • mice with score exceeding 45 were removed from the experiment.
  • the general health of mice was evaluated three times per week after disease induction. As an indicator of general health, animal body weight was used.
  • DBA/IJ mice were given the indicated amounts of compound daily and disease progression vas evaluated three times per week starting on day 27.
  • One animal in vehicle group and one animal in 23 group were sacrificed pre-termination due to high score.
  • the AMT group was removed pre- termination due to a decline in health.
  • Data represents mean values of arthritic score ⁇ SEM.
  • * represents a p-value ⁇ 0.05 and ** represents a p-value ⁇ 0.01 for comparison between MTX and vehicle, while ⁇ represents a p-value ⁇ 0.05 for comparison between 16 and vehicle.
  • Rl and R2 are selected from the group consisting of hydrogen and a moiety of the formula II, III or IV
  • R3 is selected from the group consisting of hydrogen, Ci_ 5 alkyl, C 2 - 6 alkenyl, C 2 - 4 alkynyl, C 5 . i 2 aryl, C 4- n heteroaryl and a moiety of the formula II, III or IV above,
  • R4 and R5 are independently selected from the group consisting of OH, 0-Ci- 6 alkyl, 0-C 2 . 5 alkenyl, 0-C 6 -i 2 aryl, 0-C 4 -uheteroaryl and a moiety of the formula V or VI;
  • R6 and R7 are independently selected from the group consisting of hydrogen, CF 3 , Ci- 6 alkyl, C 2 - 6 alkenyl, C 6 -i 2 aryl, and C 4 -uheteroaryl;
  • R8 and R9 are independently hydroxyl groups or R8 and R9 form, together with the intervening B and O atoms, a pinacol, catechol, diethanolamine, N-methyldiethanolamine or MIDA boronate group;
  • W and Q are independently C or N; wherein each of X, Y and Z are selected from the group consisting of halogen, amino, nitro, cyano, hydroxyl, CF 3 , Ci- 6 alkyl, Ci- 6 alkoxy, C 2 - 6 alkenyl, C 2 - 6 alkenyloxy, C 6 -i 2 aryl, C 4 .
  • Ci- 6 alkyl, C 2 - 6 alkenyl, C 6 -i 2 aryl, C 4 -uheteroaryl may be substituted by one or more substituents selected from the group consisting of halogen, amino, nitro, cyano, hydroxyl, CF 3 , and Ci_ 5 alkyl; and each of a, b and c are integers in the range 0-4;
  • X' and Y' are independently S or O, and R' and R" are independently selected from hydrogen, Ci- 6 alkyl, C 2 - 6 alkenyl, aryl and Ci- 6 alkyl-aryl; wherein, if each of Rl, R2 and R3 are different from a moiety selected from a moiety of the formula II, III and IV, then at least one of R4 and R5 is a moiety of the formula V; as well as pharmaceutically acceptable salts, solvates, and stereoisomers thereof.
  • ASPECT 4 The compound according to any one of the preceding aspects, wherein R8 and R9 are independently hydroxyl or R8 and R9 form, together with the intervening B and O atoms, a pinacol or catechol group, preferably wherein R8 and R9 are independently hydroxyl or R8 and R9 form, together with the intervening B and O atoms, a pinacol group, preferably wherein R8 and R9 are both hydroxyl groups.
  • each of X, Y and Z are selected from the group consisting of halogen, cyano, hydroxyl, CF 3 , and Ci- 5 alkyl ; and each of a, b and c are 0, 1 or 2.
  • R2 is selected from the group consisting of
  • Rl is hydrogen
  • R3 is selected from the group consisting of hydrogen and Ci- 6 alkyl, preferably selected from the group consisting of hydrogen and Ci- 4 alkyl, preferably wherein said R3 is methyl; and R4 and R5 are selected from the group consisting of OH and 0-Ci- 5 alkyl, preferably selected from the group consisting of OH and 0-Ci_ 4 alkyl, preferably wherein R4 and R5 are both methoxy or hydroxy.
  • Rl and R2 are hydrogen
  • R4 and R5 are selected from the group consisting of OH and 0-Ci- 5 alkyl, preferably selected from the group consisting of OH and 0-Ci- 4 alkyl, preferably wherein R4 and R5 are both methoxy or hydroxy.
  • Rl and R2 are hydrogen
  • R3 is selected from the group consisting of hydrogen and Ci- 6 alkyl, preferably selected from the group consisting of hydrogen and Ci- 4 alkyl, preferably wherein said R3 is methyl.
  • a method for the preparation of a compound of the formula I comprising the steps: a) Providing methotrexate (MTX) of the formula 12 or aminopterin of the formula 31 or any protected versions of them
  • R4, R5, R6, R7, R8, R9, R', R", W, Q, X, X', Y, Y' Z, a, b, c are as defined above,
  • RIO and Rl l is a leaving group LG; and c) Reacting optionally protected MTX(12) or optionally protected aminopterin (31) with a compound of formula Ila, Ilia, IVa, or VI to obtain a compound of formula I according to the invention;
  • a pharmaceutical composition comprising a compound according to any one of aspects 1-12 optionally in combination with one or more excipients.
  • a compound according to any one of aspects 1-12 as a prodrug for the treatment of inflammatory diseases or cancer such as wherein said inflammatory disease is selected from the group consisting of rheumatoid arthritis (RA), juvenile dermatomyositis, psoriasis, psoriatic arthritis, lupus, sarcoidosis, atopic dermatitis, eczema Crohn's disease, colitis ulcerosa, multiple sclerosis, and Amyotropic Lateral Sclerosis (ALS).
  • RA rheumatoid arthritis
  • juvenile dermatomyositis juvenile dermatomyositis
  • psoriasis psoriatic arthritis
  • lupus lupus
  • sarcoidosis atopic dermatitis
  • eczema Crohn's disease colitis ulcerosa
  • multiple sclerosis multiple sclerosis
  • ALS Amyotropic Lateral Sclerosis

Landscapes

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

Abstract

L'invention concerne des promédicaments activés principalement ou exclusivement dans des tissus inflammatoires, plus particulièrement des promédicaments de méthotrexate et leurs dérivés, qui sont activés sélectivement par des espèces réactives de l'oxygène (ROS) dans des tissus inflammatoires associés au cancer et à des maladies inflammatoires, ainsi qu'un procédé de préparation desdits promédicaments.
PCT/EP2017/071457 2016-08-26 2017-08-25 Promédicaments activés par des espèces réactives de l'oxygène destinés à être utilisés dans le traitement de maladies inflammatoires et du cancer WO2018037120A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP16185890 2016-08-26
EP16185890.7 2016-08-26

Publications (1)

Publication Number Publication Date
WO2018037120A1 true WO2018037120A1 (fr) 2018-03-01

Family

ID=56842670

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/071457 WO2018037120A1 (fr) 2016-08-26 2017-08-25 Promédicaments activés par des espèces réactives de l'oxygène destinés à être utilisés dans le traitement de maladies inflammatoires et du cancer

Country Status (1)

Country Link
WO (1) WO2018037120A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109096319A (zh) * 2018-10-09 2018-12-28 贺州学院 一种检测过氧化氢的新型近红外比率荧光探针及其制备方法和应用
CN110156823A (zh) * 2019-06-14 2019-08-23 淮阴工学院 一种无毒甲氨蝶呤前药及其制备方法
US20200113906A1 (en) * 2017-05-10 2020-04-16 University Of Massachusetts Crosslinked polymer nanoparticles for targeted cellular uptake and therapeutics, and compositions and methods thereof
CN112321625A (zh) * 2020-11-30 2021-02-05 河西学院 一种基于叶酸的mida硼酸酯-萘二甲酰亚胺类化合物及其制备与应用
CN113896657A (zh) * 2021-11-16 2022-01-07 山东盛安贝新能源有限公司南京分公司 一种索马鲁肽保护氨基酸的合成方法及纯化方法
CN114249731A (zh) * 2020-09-24 2022-03-29 南京海润医药有限公司 一种甲氨蝶呤中间体的精制方法

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012123076A1 (fr) 2011-03-11 2012-09-20 Ruprecht-Karls-Universität Heidelberg Composés à base de ferrocène et leur utilisation comme promédicaments régulant les espèces réactives de l'oxygène (ros)
US20130045949A1 (en) 2011-07-01 2013-02-21 Uwm Research Foundation, Inc. Anti-cancer agents
US20140378673A1 (en) 2012-02-13 2014-12-25 University Of Tsukuba Prodrug using nitroimidazole
US20150005352A1 (en) 2013-06-26 2015-01-01 The Regents Of The University Of California Reactive oxygen species-based prodrugs

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012123076A1 (fr) 2011-03-11 2012-09-20 Ruprecht-Karls-Universität Heidelberg Composés à base de ferrocène et leur utilisation comme promédicaments régulant les espèces réactives de l'oxygène (ros)
US20130045949A1 (en) 2011-07-01 2013-02-21 Uwm Research Foundation, Inc. Anti-cancer agents
US20140378673A1 (en) 2012-02-13 2014-12-25 University Of Tsukuba Prodrug using nitroimidazole
US20150005352A1 (en) 2013-06-26 2015-01-01 The Regents Of The University Of California Reactive oxygen species-based prodrugs

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
ACHILLI, C. ET AL.: "Folic acid-conjugated 4-Amino-Phenylboronate, a Boron-Containing Compound Designed for Boron Neutron Capture Therapy, is an Unexpected Agonist for Human Neutrophils and Platelets", CHEM BIO DRUG DES, vol. 83, 2013, pages 532 - 540, XP002762878
CHEM. REV., vol. 111, no. 11, 2011, pages 6557 - 6602
KHAN, Z.A. ET AL: "Methotrexate: a detailed review on drug delivery and clinical aspects", EXPERT OPINION ON DRUG DELIVERY, vol. 9, 2012, pages 151 - 169, XP002762879 *
KHAN; Z.A. ET AL.: "Methotrexate: a detailed review on drug delivery and clinical spects", EXPERT OPINION ON DRUG DELIVERY, vol. 9, 2012, pages 151 - 169, XP002762879
ROSOWSKY A ET AL.: "JOURNAL OF MEDICINAL CHEMISTRY", vol. 34, 1 January 1991, AMERICAN CHEMICAL SOCIETY, article "SYNTHESIS OF BIOLOGICAL ACTIVITY OF METHOTREXATE ANALOGUES WITH TWO ACID GROUPS AND A HYDROPHOBIC AROMATIC RING IN THE SIDE CHAIN", pages: 574 - 579
ROSOWSKY A ET AL: "SYNTHESIS AND BIOLOGICAL ACTIVITY OF METHOTREXATE ANALOGUES WITH TWO ACID GROUPS AND A HYDROPHOBIC AROMATIC RINGIN THE SIDE CHAIN", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 34, no. 2, 1 January 1991 (1991-01-01), pages 574 - 579, XP002927775, ISSN: 0022-2623, DOI: 10.1021/JM00106A016 *
WEI WEN-HAO ET AL.: "ORGANIC & BIOMOLECULAR CHEMISTRY", vol. 3, 21 September 2005, ROYAL SOCIETY OF CHEMISTRY, article "Gadolinium texaphyrin-methotrexate conjugates. Towards improved cancer chemotherapeutic agents", pages: 3290 - 3296
WEI WEN-HAO ET AL: "Gadolinium texaphyrin-methotrexate conjugates. Towards improved cancer chemotherapeutic agents", ORGANIC & BIOMOLECULAR CHEMISTRY, ROYAL SOCIETY OF CHEMISTRY, GB, vol. 3, no. 18, 21 September 2005 (2005-09-21), pages 3290 - 3296, XP002608176, ISSN: 1477-0520 *
WUTS, P. G. M.; GREENE, T. W.: "Greene's Protective Groups in Organic Synthesis", 2006, WILEY
XIAOHUA PENG; VARSHA GANDHI: "ROS-activated anticancer prodrugs: a new strategy for tumor-specific damage", THERAPEUTIC DELIVERY, vol. 3, 2012, pages 823 - 833
YUNYAN KUANG; KUMUDHA BALAKRISHNAN; VARSHA GANDHI; XIAOHUA PENG: "Hydrogen peroxide inducible DNA cross-linking agents: targeted anticancer prodrugs", J. AM. CHEM. SOC., vol. 133, 2011, pages 19278 - 19281

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200113906A1 (en) * 2017-05-10 2020-04-16 University Of Massachusetts Crosslinked polymer nanoparticles for targeted cellular uptake and therapeutics, and compositions and methods thereof
CN109096319A (zh) * 2018-10-09 2018-12-28 贺州学院 一种检测过氧化氢的新型近红外比率荧光探针及其制备方法和应用
CN110156823A (zh) * 2019-06-14 2019-08-23 淮阴工学院 一种无毒甲氨蝶呤前药及其制备方法
CN114249731A (zh) * 2020-09-24 2022-03-29 南京海润医药有限公司 一种甲氨蝶呤中间体的精制方法
CN114249731B (zh) * 2020-09-24 2023-09-29 南京海润医药有限公司 一种甲氨蝶呤中间体的精制方法
CN112321625A (zh) * 2020-11-30 2021-02-05 河西学院 一种基于叶酸的mida硼酸酯-萘二甲酰亚胺类化合物及其制备与应用
CN113896657A (zh) * 2021-11-16 2022-01-07 山东盛安贝新能源有限公司南京分公司 一种索马鲁肽保护氨基酸的合成方法及纯化方法

Similar Documents

Publication Publication Date Title
WO2018037120A1 (fr) Promédicaments activés par des espèces réactives de l'oxygène destinés à être utilisés dans le traitement de maladies inflammatoires et du cancer
EP3039020B1 (fr) Inhibiteurs d'ido
US8865756B2 (en) Inhibitors of HCV NS5A
AU2020214802A1 (en) Compounds and uses thereof
JP6616411B2 (ja) 新規な、nik阻害剤としてのピラゾール誘導体
CA3042731A1 (fr) Inhibiteurs de kinases associees au recepteur de l'interleukine -1 et leurs utilisations
KR102659213B1 (ko) Jak1 선택적 억제제
WO2011150243A1 (fr) Inhibiteurs de ns5a de vhc
EP3917517A1 (fr) Composés et leurs utilisations
EA024853B1 (ru) Ингибиторы ns5a вгс
EP3686199B9 (fr) Dérivé à cycle fusionné utilisé en tant qu'inhibiteur du récepteur a2a
CN115484952A (zh) 被取代的吡啶酮-吡啶基化合物的方法、组合物和结晶形式
CN110759908A (zh) 用于抑制Bcl-2蛋白的N-苯磺酰基苯甲酰胺类化合物及其组合物及应用
CN107987072B (zh) 作为crth2抑制剂的吲哚类化合物
EP3560928A1 (fr) Composé imidazole fusionné ayant une activité inhibitrice de l'indoléamine 2,3-dioxygénase
CN106414468A (zh) 富马酸‑co‑释放分子杂合物、其在治疗炎性或心血管疾病中的用途和其制备方法
CN106232602B (zh) 氘代喹唑啉酮化合物以及包含该化合物的药物组合物
EP3475285B1 (fr) Cible mécaniste d'inhibiteurs de la voie de signalisation de la rapamycine et ses applications thérapeutiques
EP4053108A1 (fr) Composé contenant un noyau benzénique et son application
CN108264500B (zh) 取代的2-氨基吡啶类化合物及制备方法
WO2021175317A1 (fr) Cristal de composé tricyclique agissant sur la protéine crbn et son procédé de préparation
WO2019233366A1 (fr) Antagoniste sélectif du récepteur a2a
KR20220110744A (ko) 암 줄기 세포를 표적화하는 암 치료
RU2428414C2 (ru) Способ получения амидов креатина
WO2018037119A1 (fr) Promédicaments activés par des espèces réactives de l'oxygène destinés à être utilisés dans le traitement de maladies inflammatoires et du cancer

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: 17761225

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17761225

Country of ref document: EP

Kind code of ref document: A1