WO2022034587A1 - Traf2- AND Nck-INTERACTING KINASE (TNIK) INHIBITORS AND USES THEREOF - Google Patents

Traf2- AND Nck-INTERACTING KINASE (TNIK) INHIBITORS AND USES THEREOF Download PDF

Info

Publication number
WO2022034587A1
WO2022034587A1 PCT/IL2021/050976 IL2021050976W WO2022034587A1 WO 2022034587 A1 WO2022034587 A1 WO 2022034587A1 IL 2021050976 W IL2021050976 W IL 2021050976W WO 2022034587 A1 WO2022034587 A1 WO 2022034587A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
substituted
tnk
group
Prior art date
Application number
PCT/IL2021/050976
Other languages
French (fr)
Inventor
Joel Moshe VAN GELDER
Original Assignee
Tnik Therapeutics Ltd
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 Tnik Therapeutics Ltd filed Critical Tnik Therapeutics Ltd
Publication of WO2022034587A1 publication Critical patent/WO2022034587A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • 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
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings

Definitions

  • the present invention in some embodiments thereof, relates to therapy and, more particularly, but not exclusively, to novel compounds which act as inhibitors of Traf2- and Nck- interacting kinase (TNIK), to pharmaceutical compositions comprising same and to uses thereof in the treatment of medical conditions associated with Traf2- and Nck-interacting kinase (TNIK), such as, for example, cancer.
  • TPIK Traf2- and Nck-interacting kinase
  • TNIK Traf2- and Nck-interacting kinase
  • TNIK plays an important role in the growth of colorectal cancer, and was reported to be a target for controlling aberrant Wnt signaling in colorectal cancer (Cancer Res 2010;70:5024-5033). TNIK gene was found to be over-expressed in 7 % of gastric cancer patients’ tissue samples, thus being a target for treating gastric cancer (Oncogenesis 2014, 3, e89). TNIK was also reported to be associated with proliferation and differentiation of leukemia stem cells in chronic myelogenous leukemia (Journal of Clinical Investigation, 2012, 122, 624).
  • cancers or tumors such as, for example, hepatocellular carcinoma, desmoid tumor, medulloblastoma (pediatric brain tumor), Wilms tumor (pediatric kidney cancer), and thyroid tumor, which are associated with aberrant Wnt signaling, or which are otherwise associated with one or more other TNIK signaling pathways, can benefit from TNIK inhibition.
  • TNIK inhibition was also reported to be useful in treating other medical conditions, including, for example, chronic obstructive pulmonary disease (COPD), lupus nephritis, diabetic nephropathy, focal segmental glomerulosclerosis, renal fibrosis, pulmonary fibrosis, and scleroderma, and other inflammatory diseases and disorders.
  • COPD chronic obstructive pulmonary disease
  • lupus nephritis diabetic nephropathy
  • focal segmental glomerulosclerosis renal fibrosis
  • pulmonary fibrosis pulmonary fibrosis
  • scleroderma scleroderma
  • WO 2019/156439 and WO 2019/156438 disclose compounds featuring a lH-pyrazol-3- amine skeleton, substituted by aryls and/or heteroaryls, which act as TNIK inhibitors. Additional background art includes WO 2009/030890 and WO 2010/100431, which disclose pyrimidine and pyrrolopyrimidine compounds, respectively, that are capable of inhibiting one or more kinases; Karthikeyan et al., Bioorg. Biomed. Chem Lett., 2017, Vol.
  • Y is N or CRs; Z is N or CIU;
  • Ri, and R2 are each independently hydrogen, alkyl or cycloalkyl;
  • R3 and R4 are each independently hydrogen, alkyl, haloalkyl, or halo;
  • R5 and Re are each independently hydrogen, alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, or aryloxy;
  • B is hydrogen, alkyl, haloalkyl, halo, alkoxy, thioalkoxy, or cycloalkyl;
  • A is a substituted or unsubstituted aryl or heteroaryl, or, alternatively, forms with Ri a substituted or unsubstituted heteroalicyclic ring or forms with R3 a substituted or unsubstituted cyclic ring;
  • X is -O-C
  • Ri, and R2 are each hydrogen.
  • Y is CR5. According to some of any of the embodiments described herein, R5 is hydrogen.
  • Z is CRe
  • Rs is hydrogen
  • R4 is hydrogen
  • B is alkyl
  • A is a substituted or unsubstituted aryl.
  • A is a substituted aryl.
  • the aryl is substituted by a heteroaryl.
  • the aryl is substituted by pyrazole.
  • W is or comprises a nitrogen-containing heteroalicyclic ring.
  • W is a nitrogen-containing heteroalicyclic ring attached to the X via a nitrogen atom of the nitrogen-containing heteroalicyclic ring.
  • the nitrogen-containing heteroalicyclic ring is substituted by an alkyl, cycloalkyl or a heteroalicyclic.
  • W is a substituted piperidinyl.
  • W is 1,4-bipiperidinyl.
  • the piperidinyl is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom.
  • W is a substituted 1,4- piperazinyl.
  • the piperazinyl is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom.
  • W is an aminoalkyl, attached to the X via the amine.
  • the aminoalkyl is substituted by the nitrogen-containing heteroalicyclic.
  • A is phenyl substituted by pyrazole.
  • a pharmaceutical composition comprising the compound of Formula I as described herein in any of the respective embodiments and any combination thereof and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is formulated for oral administration.
  • the medical condition is cancer
  • the treating comprises orally administering the compound or the composition to a subject in need thereof.
  • FIGs. 1A-B present the chemical structures of exemplary compounds according to some embodiments of the present invention
  • FIGs. 2A-B present schemes showing exemplary unsuccessful synthetic pathways for providing TNK-108
  • FIGs. 3A-B present schemes showing additional exemplary unsuccessful synthetic pathways for providing TNK-108
  • FIG. 4 presents an exemplary synthetic pathway for providing TNK-108
  • FIGs. 5A-B present another exemplary synthetic pathway for providing TNK-108
  • FIG. 6 presents an exemplary synthetic pathway for providing TNK-127
  • FIG. 7 presents an exemplary synthetic pathway for providing TNK-135
  • FIG. 8 presents an exemplary synthetic pathway for providing TNK-128
  • FIG. 9 presents comparative plots showing the mean plasma concentration following a single IV or PO administration of TNK-108;
  • FIG. 10 presents comparative plots showing the mean plasma concentration of TNK-002 (KY-08405) following a single IV or PO administration of mol equivalent amounts of TNK-108 and TNK-002 (KY-08405);
  • FIG. 11 presents comparative plots showing the mean plasma concentration of a respective glucuronide metabolite following a single IV or PO of mol equivalent amounts of TNK-108 and TNK-002 (KY-08405).
  • FIGs. 14A-B are bar graphs showing the effect of treatment with TNK-128 (FIG. 14A) and TNK-108 (FIG. 14B) on the tumor volume of mice inoculated with SW620 human colorectal cancer, at day 25 of the study.
  • the present invention in some embodiments thereof, relates to therapy and, more particularly, but not exclusively, to novel compounds which act as Traf2- and Nck-interacting kinase (TNIK), to pharmaceutical compositions comprising same and to uses thereof in the treatment of medical conditions associated with Traf2- and Nck-interacting kinase (TNIK), such as, for example, cancer.
  • TPIK Traf2- and Nck-interacting kinase
  • the present inventors While exploring the activity of TNIK inhibitors such as disclosed in, for example, WO 2019/156439, the present inventors have uncovered that (i) the oral bioavailability of such compounds is very low (e.g., lower than 10 % and even lower than 5 %); and (ii) the blood exposure (AUC) of such compounds is low.
  • the present inventors have uncovered that compounds such as disclosed in WO 2019/156439, which possess a phenol moiety, undergo substantial Phase 2 metabolism, for example, glucuronidation and possibly also sulfation, which adversely affects their activity; causes low blood exposure after IV and especially PO administration; and results in a low oral bioavailability.
  • the present inventors have first selected 1,4’ -bipiperidine as an exemplary protecting group, based on a successful performance of such a group in the drug known as CPT-11 (Irinotecan®) [See, for example, Kunimoto T et al., Cancer Res., 1987, 47, 5944-5947],
  • TNK-002 an exemplary TNIK inhibitor
  • TNK-108 a compound referred to herein as TNK-108 (see, FIG. 1A)
  • the present inventors have recognized that a more sophisticated synthetic route should be employed, which is not based on coupling the 1,4’ -bipiperidine to a phenol moiety, but rather involves “building” the final molecule differently.
  • the present inventors have uncovered a synthetic pathway which was successfully employed for preparing compounds in which a piperidine- or piperazine-containing moiety “masks” a phenol or aniline moiety in a TNIK compound, and have practiced this synthetic pathway for preparing compounds such as shown in FIGs. 4-8.
  • TNK-108 an exemplary such compound, which is referred to herein as TNK-108, exhibits high bioavailability (e.g., higher than 50 % and even higher than 60 %, when tested in mice), high plasma concentration following oral administration, as shown in FIGs. 9 and 10, which remains even 24 hours following administration, as shown in FIG. 10, and which remains around the EC50 of the compound almost 8 hours following oral administration, as shown in FIG. 9.
  • This compound also exhibits Tl/2 which is higher by about 30 % compared to its parent compound TNK-002.
  • TNK-128 presents the plasma concentration of the glucuronide metabolite and show much lower amounts of this metabolite compared to the parent compound TNK-002. Similar observations were obtained for another exemplary compound, referred to herein as TNK-128, as shown, for example, in FIGs. 12A-B and in Table 1 in the Examples section that follows. The anti-cancer activity of TNK-128 and TNK-108 is demonstrated in FIGs. 14A-B.
  • Non-cleavable compounds, referred to herein as TNK-127 and TNK-135 also presented improved performance (e.g., improved solubility and plasma stability), as shown for example, in FIG. 13 and Table 1.
  • TNIK inhibitors that have structural features similar to those described in WO 2019/156439, yet exhibit improved performance at least in terms of their pharmacokinetic properties, paves the way to a myriad of compounds that can be beneficially utilized for inhibiting TNIK and as therapeutically active agents for treating medical conditions in which downregulation of TNIK is beneficial.
  • Embodiments of the present invention relate to novel TNIK inhibitors, to processes of preparing same and to uses thereof in treating medical conditions that are associated with TNIK (e.g., mediated by TNIK activity).
  • Y is N or CR 5 ;
  • Z is N or CRs
  • Ri and R2 are each independently hydrogen, alkyl or cycloalkyl
  • R3 and R4 are each independently hydrogen, alkyl, haloalkyl, or halo;
  • R5 and Re are each independently hydrogen, alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, or aryloxy;
  • B is hydrogen, alkyl, haloalkyl, halo, alkoxy, thioalkoxy, or cycloalkyl;
  • A is a substituted or unsubstituted aryl or heteroaryl, or, alternatively, forms with Ri a substituted or unsubstituted heteroalicyclic ring or forms with R3 a substituted or unsubstituted cyclic ring;
  • W is a nitrogen-containing moiety attached to said X via said nitrogen atom.
  • Ri is hydrogen.
  • Ri is alkyl.
  • R2 is hydrogen.
  • R2 is alkyl.
  • Ri and R2 are each hydrogen.
  • R2 is hydrogen
  • the compound is incapable of undergoing a simple coupling to the moiety W, due to the reactivity of the respective bridging amine.
  • Ri is hydrogen
  • the compound is incapable of undergoing a simple coupling to the moiety W, due to the high reactivity of the respective pyrazole nitrogen, as shown, for example , in FIG. 2B.
  • R4 is hydrogen.
  • R4 is halo, alkyl (e.g., methyl or ethyl) or haloalkyl (e.g., trihaloalkyl such as CF3).
  • Y is CR5.
  • R5 is hydrogen.
  • R5 is halo or alkyl (e.g., methyl or ethyl) or haloalkyl (e.g., trihaloalkyl such as CF3).
  • Z is CRe.
  • Rs is hydrogen.
  • Re is halo or alkyl (e.g., methyl or ethyl) or haloalkyl (e.g., trihaloalkyl such as CF3).
  • R5 is hydrogen.
  • R5 is halo or alkyl (e.g., methyl or ethyl) or haloalkyl (e.g., trihaloalkyl such as CF3).
  • Y is CR5 and Z is CRs, such that the respective ring is a phenyl ring, which can be substituted, in case one or more of R4, R5, Re or B is other than hydrogen.
  • B is hydrogen, halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3).
  • one R4, R5, Rs or B is halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3).
  • the other substituents on the phenyl are all hydrogen.
  • B is alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3). According to some of any of the embodiments described herein, B is alkyl (e.g., methyl or ethyl).
  • B is alkyl (e.g., methyl or ethyl)
  • Y is CR5
  • Z is CRs
  • R4, R5, and Rs are each hydrogen.
  • R3 is hydrogen.
  • R3 is alkyl (e.g., methyl or ethyl).
  • A is a substituted or unsubstituted aryl (e.g., substituted or unsubstituted phenyl).
  • A is a substituted aryl (e.g., substituted phenyl).
  • the aryl e.g., phenyl
  • the aryl is substituted at least at the para position with respect to the pyrazole to which the phenyl is linked.
  • the aryl e.g., phenyl
  • A is a substituted or substituted heteroaryl, which, when substituted, include at least a substituent at the para position with respect to the pyrazole to which the heteroaryl is linked.
  • the heteroaryl is substituted at the meta position with respect to the pyrazole ring to which the heteroaryl is linked.
  • exemplary A moieties include, but are not limited to:
  • exemplary A moieties include, but are not limited to:
  • Y is CR5
  • Z is CRe
  • Rs, Rs and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
  • X, W, R3, R4, Rs, Rs and B are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
  • Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • n is 1 or 2 and at least one Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
  • n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl.
  • one of the Ra substituents is pyrazole, which can be substituted or unsubstituted.
  • Ra, X, W, R3, R4, Rs, Re and B are as defined herein in any of the respective embodiments;
  • m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1);
  • k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
  • Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • m is 0 or 1, and in some embodiments, m is 0.
  • k is 0.
  • W is a nitrogen-containing moiety which is attached to X via the nitrogen atom, and which is or comprises a nitrogen-containing heteroalicyclic ring.
  • W is a substituted nitrogen-containing heteroalicyclic ring, which is attached to the X via the nitrogen atom of the heteroalicyclic ring.
  • W is a substituted nitrogen-containing heteroalicyclic ring.
  • W is a substituted nitrogen-containing heteroalicyclic ring, which is attached to the X via the nitrogen atom.
  • the nitrogen-containing heteroalicyclic ring is substituted by an alkyl, cycloalkyl or a heteroalicyclic.
  • W is or comprises a piperidine, a piperazine, or a morpholino group, which can be substituted, and which can be linked to X via the nitrogen atom.
  • W is or comprises a piperidine or a piperazine, which can be substituted, and which can be linked to X via the nitrogen atom.
  • W is a piperidine or a piperazine, which is linked to X via the nitrogen atom. According to some of these embodiments, the piperazine or piperidine is substituted.
  • the compounds can be collectively represented by Formula II:
  • A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof;
  • V is N (such that W is or comprises a piperazine) or CRn (such that W is or comprises a piperidine);
  • R11 is hydrogen, halo, alkyl, cycloalkyl, and optionally other substituents as described herein;
  • Rio is selected from the group consisting of hydrogen, a substituted or unsubstituted alkyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heteroalicyclic.
  • Rio can alternatively be a hydrocarbon chain, which can be linear or branched, saturated or unsaturated (preferably saturated), optionally interrupted by one or more heteroatoms (e.g., oxygen or an amine as defined herein).
  • Rio is or comprises an alkylene glycol as defined herein.
  • Y is CR5
  • Z is CRs, R4, Rs, Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3))
  • A is substituted or unsubstituted phenyl.
  • X, V, R3, R4, Rs, Rs, Rio and B are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
  • Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • n is 1 or 2 and at least one Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
  • n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl.
  • one of the Ra substituents is pyrazole, which can be substituted or unsubstituted.
  • Ra, X, V, R3, R4, Rs, Rs, Rio and B are as defined herein in any of the respective embodiments;
  • m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1);
  • k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
  • Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • m is 0 or 1, and in some embodiments, m is 0.
  • k is 0.
  • V is CRn, and Rio is other than hydrogen.
  • Rn is hydrogen.
  • W in Formula I, la or lb is a substituted 1,4-bi-piperidinyl.
  • V is CRn, and Rio is a heteroalicyclic.
  • Rn is hydrogen.
  • Rio is a nitrogen-containing heteroalicyclic and in some embodiments, Rio is piperidinyl, such that W in Formula I, la or lb is 1,4-bi-piperidinyl.
  • the bi-piperidinyl can be substituted or unsubstituted and is preferably unsubstituted. Exemplary such compounds are referred to herein as TNK-108 and TNK-127 (see, for example, FIGs. 1 A and IB, respectively).
  • Rio is a hydrocarbon chain, which can be linear or branched, saturated or unsaturated (preferably saturated), optionally interrupted by one or more heteroatoms (e.g., oxygen or an amine as defined herein).
  • Rio is a hydrocarbon chain, which can be linear or branched, saturated or unsaturated (preferably saturated), optionally interrupted by one or more heteroatoms (e.g., oxygen or an amine as defined herein).
  • heteroatoms e.g., oxygen or an amine as defined herein.
  • Rio is a branched, saturated hydrocarbon chain, interrupted by one or more heteroatoms. In some of these embodiments, Rio is a branched, saturated hydrocarbon chain, interrupted by one or more (e.g., two) oxygen atoms.
  • V is CRn
  • Rn is hydrogen
  • V is N.
  • Moieties that are or comprise a branched saturated hydrocarbon chain interrupted by one or more heteroatoms, and which can be included as Rio in any of the respective Formulae can be collectively represented by Formula V:
  • L is an alkylene chain of, for example, 1 to 6, or 1 to 4, carbon atoms, which is linked to V;
  • R21 is hydrogen or an alkyl, which can be substituted or unsubstituted, and is preferably an unsubstituted alkyl; the two moieties [(CH 2 )j(CH2-O)q(R22)] can be the same or different, and each of these moieties, j is independently an integer of from 0 to 6, or from 0 to 4, or from 1 to 4; q is independently an integer of from 0 to 6, or from 0 to 4, or from 0 to 2; and R22 is an alkyl (e.g., a branched alkyl), or cycloalkyl.
  • the two moieties [(CH2)j(CH2-O)q(R22)] are the same.
  • R22 is a branched alkyl, for example, t-butyl.
  • Exemplary branched saturated hydrocarbon chains of Formula V are shown in the compounds referred to as TNK-124 and TNK-128 (see, for example, FIG. 1 A).
  • W is 1,4-bipiperidinyl.
  • such compounds are collectively represented by Formula III:
  • A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof.
  • Y is CR5
  • Z is CRs, R4, R5,
  • Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
  • X, R3, R4, Rs, Rs, and B are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
  • Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • n is 1 or 2 and at least one
  • Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
  • n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl.
  • one of the Ra substituents is pyrazole, which can be substituted or unsubstituted.
  • Ra, X, R3, R4, Rs, Rs, and B are as defined herein in any of the respective embodiments; m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1); k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
  • Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • m is 0 or 1, and in some embodiments, m is 0.
  • k is 0.
  • W is a moiety of Formula V as described herein in any of the respective embodiments.
  • A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof;
  • V is as defined herein for Formula II, Ila or lib, and
  • q, j, L, R21 and R22 are as defined herein for Formula V in any of the respective embodiments and any combination thereof.
  • Y is CR5
  • Z is CRe
  • Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
  • X, V, B, R3, R4, Rs, Re, q, j, L, R21 and R22 are as defined herein in any of the respective embodiments;
  • n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2);
  • Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • n is 1 or 2 and at least one
  • Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
  • n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl.
  • one of the Ra substituents is pyrazole, which can be substituted or unsubstituted.
  • Ra, X, B, V, R3, R4, Rs, Re, q, j, L, R21 and R22 are as defined herein in any of the respective embodiments;
  • m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1);
  • k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
  • Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • n is 0 or 1, and in some embodiments, m is 0.
  • k is 0.
  • W is an aminoalkyl, which is attached to X via said amino.
  • the aminoalkyl is substituted, e.g., the alkyl is substituted by one or more substituents.
  • the one or more substituents comprise one or more heteroatoms such as N and/or O and/or S.
  • the aminoalkyl can be substituted by one or more of amine, alkoxy, thioalkoxy, heteroalicyclic, heteroaryl, carboxy, and other heteroatom-containing substituents.
  • Such compounds can be collectively represented by Formula VI:
  • A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof; and R13 and R14 are each independently selected from hydrogen and alkyl, wherein preferably at least one of R13 and R14 is an alkyl, and further preferably, at least one such alkyl is substituted as described herein. When one or both of R13 and R14 is an alkyl, the alkyl is independently of 1 to 10, or from
  • Y is CR5
  • Z is CRe
  • Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
  • X, V, B, R3, R4, Rs, Rs, R12, RB and R14 are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
  • Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • n is 1 or 2 and at least one
  • Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
  • n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl.
  • one of the Ra substituents is pyrazole, which can be substituted or unsubstituted.
  • Ra, X, B, V, R3, R4, Rs, Re, R12, R13 and R14 are as defined herein in any of the respective embodiments;
  • m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1);
  • k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
  • Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • n is 0 or 1, and in some embodiments, m is 0.
  • k is 0.
  • R12 is hydrogen
  • W is an aminoalkyl which is substituted by a nitrogen-containing heteroalicyclic (e.g., a piperidinyl or piperazinyl group).
  • a nitrogen-containing heteroalicyclic e.g., a piperidinyl or piperazinyl group.
  • A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof;
  • V is as defined herein for Formula II, Ila or lib,
  • R12 is as defined herein for R’ and R” and K is an alkyl, as defined herein, e.g., an alkylene chain, preferably of from 1 to 10, or from 1 to 6, or from 1 to 4, carbon atoms in length.
  • Y is CR5
  • Z is CRe
  • Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
  • X, V, B, R3, R4, R5, Re, R12 and K are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
  • Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • n is 1 or 2 and at least one Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
  • n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl.
  • one of the Ra substituents is pyrazole, which can be substituted or unsubstituted.
  • Ra, X, B, V, R3, R4, Rs, Re, R12 and K are as defined herein in any of the respective embodiments;
  • m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1);
  • k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
  • Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
  • m is 0 or 1, and in some embodiments, m is 0.
  • R12 is hydrogen
  • K is an unsubstituted alkyl.
  • V is CRn.
  • Rn is hydrogen.
  • W is or comprises a nitrogen-containing heteroalicyclic, for example, a piperidine or a piperazine.
  • W is a piperidinyl which is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom, such as described, for example, by Formula V or in Formula IV, IVa or IVb.
  • W is or comprises a substituted 1,4-piperazinyl.
  • the piperazinyl is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom, such as described, for example, by Formula V or in Formula IV, IVa or IVb.
  • physiological conditions include presence of esterases and/or acidic environment.
  • Such compounds are also referred to herein as biocleavable compounds or biocleavable prodrugs.
  • compounds of such embodiments are such that the substituent on the respective phenyl is other than hydroxy (e.g., A in Formula I, II, III, IV, VI or VII is a phenyl that if substituted, is not substituted by hydroxy; or Ra in Formula la, Ila, Illa, Iva, Via or Vila is other than hydroxy).
  • Such compounds are also referred to herein as non-biocleavable compounds or non-biocleavable prodrugs.
  • an amine or an amide group within a substituent on the phenol group that is attached to the pyrazole via an amine group is replaced by a -NH-
  • compounds of such embodiments are such that the substituent on the respective phenyl is other than hydroxy (e.g., A in Formula I, II, III, IV, VI or VII is a phenyl that if substituted, is not substituted by hydroxy; or Ra in Formula la, Ila, Illa, Iva, Via or Vila is other than hydroxy).
  • the process as described herein is a large scale process or is at least a scalable process, that is, can be scaled up to be a large scale process.
  • large scale in the context of the process as described herein, it is meant a production process in which at least one of the compounds, whether a starting material, an intermediate and/or the final product, is used or prepared in an amount of at least one mole, or at least 2 moles, or at least 3 moles, or at least 5 moles, for example, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50 moles, or even more, including any intermediate values therebetween.
  • a “large scale” process is defined herein, alternatively or in addition, by the weight of the one or more of the starting materials, the intermediates and/or the final product, such that one or more these materials is/are used or prepared in an amount of at least 1 Kg, or at least 2 Kg, or at least 3 Kg, or at least 5 Kg, for example, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 Kg, or even more, including any intermediate values therebetween.
  • a “large scale” process is defined, alternatively or in addition, by the volumes of the reactors used in the process, as comprising at least one production step that is performed in a reactor having a volume of at least 10 liters, or at least 15 liters, or at least 20 liters, or at least 30 liters, or at least 40, 50, 60, 70, 80, 90, 100 liters or even more, including any intermediate values therebetween.
  • the process as described herein is a gram-scale process.
  • gram-scale in the context of the process as described herein, it is meant a production process in which at least one of the compounds, whether a starting material, an intermediate and/or the final product, is used or prepared in an amount of at least one gram.
  • reactive group it is meant a chemical group that is capable of undergoing a chemical reaction that typically leads to a bond formation.
  • the bond is preferably a covalent bond.
  • Chemical reactions that lead to a bond formation include, for example, nucleophilic and electrophilic substitutions, nucleophilic and electrophilic addition reactions, addition-elimination reactions, cycloaddition reactions, rearrangement reactions and any other known organic reactions that involve a bond formation.
  • the Q and L2 reactive groups are such that can undergo a chemical reaction that leads to formation of an X moiety, as described herein.
  • W-L2 can be such that comprises a respective reactive group that is attached to W and forms, when coupled with Q, the moiety X.
  • L2 can be hydrogen, and the second reactive group is an amine (that forms a part of W, such that W-L2 comprise the second reactive group).
  • the first and second reactive groups can be hydroxy and amine, respectively, while the amine forms a part of the W group, and the conditions under which the reaction is performed are such that promote a formation of the respective X moiety (as a part of a carbamate group) upon reacting said hydroxy and said amine.
  • Conditions for reacting an amine and a hydroxy group to provide a carbamate group include, for example, suitable coupling agents, optionally combined with oxidizing agents and/or other agents that promote formation of carbamate by reacting an amine (reactive group of W-L2) and phenol (which comprises the hydroxy group).
  • suitable coupling agents optionally combined with oxidizing agents and/or other agents that promote formation of carbamate by reacting an amine (reactive group of W-L2) and phenol (which comprises the hydroxy group).
  • CDI carbonylimidazolide
  • BTC bis(trichloromethyl) carbonate
  • the first and second reactive groups can be each amine, while one amine forms a part of the W group and the other forms a part of a respective aniline, and the conditions under which the reaction is performed are such that promote a formation of the respective X moiety (as a part of an ureido group), upon reacting said amines.
  • Conditions for reacting amines to provide an ureido/urea group include, for example, suitable coupling agents, optionally combined with other agents or conditions that promote formation of ureido moiety by reacting amine (reactive group of W-L2) and aniline (which comprises the other amine).
  • suitable coupling agents optionally combined with other agents or conditions that promote formation of ureido moiety by reacting amine (reactive group of W-L2) and aniline (which comprises the other amine).
  • CDI carbonylimidazolide
  • the first reactive group Li and the third reactive group L3 are selected such that upon reacting these groups an amine group is generated.
  • the first and third reactive groups are such that participate in a nucleophilic reaction, for example, the third reactive group is amine and the first reactive group is a leaving group, and the reaction conditions are such that promote a nucleophilic reaction. Exemplary such agents are described in the Examples section that follows.
  • the process further comprises preparing a compound of Formula Xd.
  • preparing a compound of Formula Xd comprises reacting a compound of Formula Xf with hydrazine.
  • the compounds disclosed herein are usable as modulators of TNIK activity, typically as inhibitors of TNIK activity, and/or as capable of generating, preferably under physiological conditions, compounds that act as modulators of TNIK activity, typically as inhibitors of TNIK activity.
  • the compounds disclosed herein are usable as therapeutically active agents for treating medical conditions that are associated with abnormal TNIK activity and/or in which modulating (e.g., inhibiting) TNIK activity is beneficial.
  • a method of modulating e.g., inhibiting a TNIK activity, which comprises contacting cells that exhibit abnormal (e.g., hyperactive) TNIK activity with a compound as described herein in any of the respective embodiments and any combination thereof.
  • such compounds exhibit modulation (e.g., inhibition) of TNIK activity when used per se.
  • the contacting can be effected in vitro, ex-vivo, or in vivo.
  • the cells are tumor cells or cells that are characterized by abnormal proliferation.
  • a method of modulating e.g., inhibiting a TNIK activity in a subject in need thereof, which comprises administering to the subject a compound as described herein in any of the respective embodiments and any combination thereof.
  • a compound as described herein in any of the respective embodiments and any combination thereof for use in modulating (e.g., inhibiting) a TNIK activity in a subject in need thereof.
  • a compound as described herein in any of the respective embodiments and any combination thereof for use in the preparation of medicament for modulating (e.g., inhibiting) a TNIK activity in a subject in need thereof.
  • the methods and uses described herein are usable for modulating a TNIK activity is cells that exhibit an abnormal TNIK activity, and in subjects hosting such cells.
  • the methods and uses described herein are usable in treating medical conditions that are associated with abnormal TNIK activity.
  • the medical conditions are associated with TNIK hyperactivity.
  • the medical conditions are mediated by TNIK.
  • the medical conditions are associated with signaling pathways mediated by TNIK.
  • Medical conditions that are treatable according to some embodiments of the present invention include proliferative diseases and disorders, such as cancer.
  • Exemplary cancer types include, but are not limited to, colorectal cancer, breast cancer, brain tumor, gastric cancer, liver cancer, ovarian cancer, lung cancer, gastrointestinal cancer, leukemia, and melanoma.
  • a method of treating a medical condition associated with an abnormal TNIK activity comprising administering to a subject in need thereof a therapeutically effective amount of a compound as described herein in any of the respective embodiments and any combination thereof.
  • a use of compound as described herein in any of the respective embodiments and any combination thereof in the treatment of a medical condition associated with an abnormal TNIK activity there is provided a use of compound as described herein in any of the respective embodiments and any combination thereof in the manufacture of a medicament for the treatment of a medical condition associated with an abnormal TNIK activity.
  • a method of treating a cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound as described herein in any of the respective embodiments and any combination thereof.
  • a method of treating a proliferative disease or disorder in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound as described herein in any of the respective embodiments and any combination thereof.
  • cancer and “tumor” are interchangeably used.
  • the terms refer to a malignant growth and/or tumor caused by abnormal and uncontrolled cell proliferation (cell division).
  • cancer encompasses tumor metastases.
  • cancer cell describes the cells forming the malignant growth or tumor.
  • Non-limiting examples of cancers and/or tumor metastases which can be treated according to some embodiments of any of the embodiments described herein relating to cancer include any solid or non-solid cancer and/or tumor metastasis, including, but not limiting to, tumors of the gastrointestinal tract (e.g., colon carcinoma, rectal carcinoma, colorectal carcinoma, colorectal cancer, colorectal adenoma, hereditary nonpolyposis type 1, hereditary nonpolyposis type 2, hereditary nonpolyposis type 3, hereditary nonpolyposis type 6; colorectal cancer, hereditary nonpolyposis type 7, small and/or large bowel carcinoma, esophageal carcinoma, tylosis with esophageal cancer, stomach carcinoma, pancreatic carcinoma, pancreatic endocrine tumors), endometrial carcinoma, dermatofibrosarcoma protuberans, gallbladder carcinoma, biliary tract tumors, prostate cancer,
  • tumors of the gastrointestinal tract
  • the term “subject” refers to a mammal (e.g., human), for example, one who has been diagnosed with a condition described herein (e.g., cancer). Subjects may be of either gender and at any stage of development.
  • the compounds of the present embodiments are characterized by enhanced solubility, high bioavailability, plasma stability, and high AUC values. These features render these compounds suitable for oral administration, and further allow administering the compounds with relatively high intervals.
  • the compounds are administered once, twice or trice daily. According to some embodiments of any of the methods and uses described herein the compounds are administered once, twice or trice weekly, or, for example, once in two days or once in three days. Compounds that feature high Tl/2 values are suitable for use in such a regimen.
  • the compounds of the present disclosure can otherwise be administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • An effective dosage is typically in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 0.01 to about 50 mg/kg/day, in single or divided doses. Depending on age, species and disease or condition being treated, dosage levels below the lower limit of this range may be suitable.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intracardiac, e.g., into the right or left ventricular cavity, into the common coronary artery, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • the compounds are administered orally (PO).
  • the compounds are administered intravenously (IV).
  • the compounds described herein according to any of the aspects of embodiments of the invention described herein can be utilized (e.g., administered to a subject) per se or in a pharmaceutical composition where the compound is mixed with suitable carriers or excipients.
  • a "pharmaceutical composition” refers to a preparation of one or a compound according to any of the embodiments described herein with other chemical components such as physiologically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • physiologically acceptable carrier and “pharmaceutically acceptable carrier” which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • An adjuvant is included under these phrases.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • the compound per se When utilized per se or in a pharmaceutically acceptable composition, the compound per se (that is, not including, weight of carriers or excipients co-formulated with the compound, as described herein) is optionally at least 80 % pure (by dry weight), optionally at least 90 % pure (by dry weight), at least 95 % pure (by dry weight), at least 98 % pure (by dry weight), and optionally at least 99 % pure (by dry weight). Purity may be enhanced, e.g., by removing impurities associated with synthesis of the compound or isolation of the compound from a natural source, by any suitable technique known in the art. Techniques for formulation and administration of drugs may be found in “Remington’s Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference.
  • tissue refers to part of an organism consisting of cells designed to perform a function or functions. Examples include, but are not limited to, brain tissue, retina, skin tissue, hepatic tissue, pancreatic tissue, breast tissue, bone, cartilage, connective tissue, blood tissue, muscle tissue, cardiac tissue brain tissue, vascular tissue, renal tissue, pulmonary tissue, gonadal tissue, hematopoietic tissue.
  • compositions of some embodiments of the invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with some embodiments of the invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the active ingredients of the pharmaceutical composition may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank’s solution, Ringer’s solution, or physiological salt buffer.
  • physiologically compatible buffers such as Hank’s solution, Ringer’s solution, or physiological salt buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the pharmaceutical composition can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the pharmaceutical composition to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient.
  • Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
  • the pharmaceutical composition is formulated for oral administration.
  • the carrier is an aqueous carrier which comprises a surfactant, preferably a surfactant, and one or both of a solubilizer (e.g., cyclodextrin or Transcutol®) and a water-soluble or water-miscible polymer (e.g., polyalkylene glycol).
  • a solubilizer e.g., cyclodextrin or Transcutol®
  • a water-soluble or water-miscible polymer e.g., polyalkylene glycol
  • the surfactant is a non-ionic surfactant, for example a Vitamin E-based surfactant such as, for example, VE-TPGS or a surfactant such as Gelucire® 44/14 (lauroyl polyoxyl-32 glycerides).
  • the surfactant is of the Tween® family, for example, Tween®80.
  • An exemplary pharmaceutical composition which is suitable for oral administration of compounds such as the exemplified TNK-108, comprises 20 % PEG400, 10 % VE-TPGS and 70 % of a 5 % aqueous solution of HP-P-CD.
  • An exemplary pharmaceutical composition which is suitable for oral administration of compounds such as the exemplified TNK-108, comprises 10 % Transcutol®, 5 % Tween®80 and 85 % a solution of 10 % Gelucire® 44/14 in Water.
  • An exemplary pharmaceutical composition which is suitable for oral administration of compounds such as the exemplified TNK-108, comprises 20 % PEG400, 5 % Tween®80 and 75 % a solution of 5 % HP-P-CD in Water.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the active ingredients for use according to some embodiments of the invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in a dispenser may be formulated containing a powder mix of the active compound and a suitable powder base such as lactose or starch.
  • compositions described herein may be formulated for parenteral administration, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative.
  • the compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the active ingredients to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water based solution, before use.
  • a suitable vehicle e.g., sterile, pyrogen-free water based solution
  • compositions of some embodiments of the invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
  • the pharmaceutical composition is formulated for oral administration.
  • compositions suitable for use in context of some embodiments of the invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of the active ingredient(s) effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., cancer or metastatic cancer) or prolong the survival of the subject being treated.
  • a disorder e.g., cancer or metastatic cancer
  • the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays.
  • a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals.
  • the data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage may vary depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see, e.g., Fingl et al. (1975), in "The Pharmacological Basis of Therapeutics", Ch. 1 p. l).
  • Dosage amount and interval may be adjusted individually to provide protein (e.g., TNIK) inhibitory levels of the active ingredient are sufficient to induce or suppress the biological effect (minimal effective concentration, MEC).
  • MEC minimum effective concentration
  • the MEC will vary for each preparation, but can be estimated from in vitro data, e.g., based on results on TNIK inhibition assay described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.
  • an effective amount is at least 100 % of the IC50 of the compound towards a TNIK inhibition. In some embodiments, an effective amount is at least 200 % of the IC50 of the compound towards TNIK inhibition. In some embodiments, an effective amount is at least 300 % of the IC50 of the compound towards TNIK inhibition. In some embodiments, an effective amount is at least 500 % of the IC50 of the compound towards TNIK inhibition. In some embodiments, an effective amount is at least 1000 % of the IC50 of the compound towards TNIK inhibition.
  • dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
  • compositions to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • compositions of some embodiments of the invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert.
  • Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as is further detailed herein.
  • the compound may be administered with an additional anti-cancer agent, as described herein in any of the respective embodiments, either together in a co-formulation (e.g., in the same pharmaceutical composition) or in separate formulations.
  • the treatment of cancer is effected in combination with an additional anti-cancer as described herein in any of the respective embodiments.
  • a pharmaceutical composition as described herein can further comprise any of the additional agents as described herein, or alternatively, be identified for use in combination with any of the additional agents as described herein.
  • kits for the treatment of a condition e.g., treatment of cancer
  • the kit comprising a packaging material packaging the compound described herein.
  • the kit further comprises an additional agent as described herein in any of the respective embodiments, and the two agents are packaged individually within the kit.
  • the kit further comprises instructions to use the compound in combination with an additional agent (e.g., an additional anti-cancer cancer) as described herein in any of the respective embodiments.
  • an additional agent e.g., an additional anti-cancer cancer
  • the one or more additional agent(s) is an anti-cancer drug.
  • the anti-cancer drug is EGFR kinase inhibitors, MEK inhibitors, VEGFR inhibitors, anti-VEGFR2 antibodies, KDR antibodies, AKT inhibitors, PDK-1 inhibitors, PI3K inhibitors, c-kit/Kdr tyrosine kinase inhibitors, Bcr-Abl tyrosine kinase inhibitors, VEGFR2 inhibitors, PDGFR-beta inhibitors, KIT inhibitors, Flt3 tyrosine kinase inhibitors, PDGF receptor family inhibitors, Flt3 tyrosine kinase inhibitors, RET tyrosine kinase receptor family inhibitor, VEGF-3 receptor antagonists, Raf protein kinase family inhibitor, angiogenesis inhibitors, Erb2 inhibitors, mTOR inhibitors, IGF-1R antibodies,
  • agents include, but are not limited to, alkylating agents such as nitrogen mustard, chlorambucil, cytoxan, ifosfamide, melphalan, thiptepa and busulfan; antimetabolites such as methotrexate, 5-fluorouracil, cytoxine arabinoside (ara-C), 5 -azacitidine, 6- mercaptopurine, 6-thioguanine, and fludarabine phosphat; antitumor antibiotics such as doxorubicin, adriamycin, daunorubicin, dactinomycin, bleomycin, mitomycin C, plicamycin, idarubicin, and mitoxantrone; vinca alkaloids and epipodophyllotoxins such as vincristine, vinblastine, vindesine, etoposide, and teniposide; nitrosoureas such as carmustine, lomustine, semustine and streptozotocin; synthetic
  • the additional agent is a drug for colorectal cancer.
  • the drug for colorectal cancer is based on regimens FOLFOX or FOLFIRI including 5-FU, leucovorin, oxaliplatin, irinotecan or their combinations.
  • the combination therapy is used together with cetuximab and/or bevacizumab.
  • the additional agent e.g., anti-cancer drug or therapy
  • compositions, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
  • a compound or “at least one compound” may include a plurality of compounds, including mixtures thereof.
  • range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
  • a numerical range is indicated herein, it is meant to include any cited numeral (fractional or integral) within the indicated range.
  • the phrases “ranging/ranges between” a first indicate number and a second indicate number and “ranging/ranges from” a first indicate number “to” a second indicate number are used herein interchangeably and are meant to include the first and second indicated numbers and all the fractional and integral numerals therebetween.
  • method refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
  • the term “treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical or aesthetical symptoms of a condition.
  • the term "subject” means an animal, preferably a mammal such as a nonprimate (e.g., cow, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit or guinea pig) or a primate (e.g. monkey and human), preferably a human being.
  • linking group describes a group (a substituent) that is attached to another moiety in the compound via two or more atoms thereof.
  • end group a group (a substituent) that is attached to another moiety in the compound via one atom thereof.
  • amine describes both a -NR’R” end group and a -NR'- linking group, wherein R’ and R" are each independently hydrogen, alkyl, cycloalkyl, aryl, as these terms are defined hereinbelow.
  • the amine group can therefore be a primary amine, where both R’ and R” are hydrogen, a secondary amine, where R’ is hydrogen and R” is alkyl, cycloalkyl or aryl, or a tertiary amine, where each of R’ and R” is independently alkyl, cycloalkyl or aryl.
  • R' and R" can each independently be hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, cyano, nitro, azo, sulfonamide, carbonyl, C-carboxylate, O-carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, N-carbamate, O-carbamate, C-amide, N-amide, guanyl, guanidine and hydrazine.
  • amine is used herein to describe a -NR'R" group in cases where the amine is an end group, as defined hereinunder, and is used herein to describe a -NR'- group in cases where the amine is or forms a part of a linking group.
  • alkyl describes a saturated aliphatic hydrocarbon including straight chain and branched chain groups.
  • the alkyl group has 1 to 20 carbon atoms.
  • the alkyl is a medium size alkyl having 1 to 10 carbon atoms. Unless otherwise indicated, the alkyl is a lower alkyl having 1 to 4 carbon atoms.
  • the alkyl has at least 4 carbon atoms, for example, the alkyl is having 4 to 12 or 4 to 10 or 4 to 8 carbon atoms.
  • the alkyl group may be substituted or unsubstituted.
  • Substituted alkyl may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, oxo, carbonyl, cyano, nitro, azo, sulfonamide, C- carboxylate, O-carboxylate, N-thiocarbamate, O-thiocarba
  • the alkyl group can be an end group, as this phrase is defined hereinabove, wherein it is attached to a single adjacent atom, or a linking group, as this phrase is defined hereinabove, which connects two or more moieties via at least two carbons in its chain.
  • a linking group it is also referred to herein as “alkylene”, e.g., methylene, ethylene, propylene, etc.
  • alkenyl describes an alkyl, as defined herein, in which at least one pair of carbon atoms are linked to one another via a double bond.
  • alkynyl or “alkyne” describes an alkyl, as defined herein, in which at least one pair of carbon atoms are linked to one another via a triple bond.
  • cycloalkyl describes an all-carbon monocyclic or fused ring (/. ⁇ ., rings which share an adjacent pair of carbon atoms) group where one or more of the rings does not have a completely conjugated pi-electron system.
  • the cycloalkyl group may be substituted or unsubstituted.
  • Substituted cycloalkyl may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, oxo, carbonyl, cyano, nitro, azo, sulfonamide, C-carboxylate, O-carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, N-carbamate, O-carbamate, C-amide, N-amide, guanyl, guanidine and hydrazine.
  • substituent group can independently
  • the cycloalkyl group can be an end group, as this phrase is defined hereinabove, wherein it is attached to a single adjacent atom, or a linking group, as this phrase is defined hereinabove, connecting two or more moieties at two or more positions thereof.
  • heteroalicyclic describes a monocyclic or fused ring group having in the ring(s) one or more atoms such as nitrogen, oxygen and sulfur.
  • the rings may also have one or more double bonds. However, the rings do not have a completely conjugated pi-electron system.
  • the heteroalicyclic may be substituted or unsubstituted.
  • Substituted heteroalicyclic may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, oxo, carbonyl, cyano, nitro, azo, sulfonamide, C- carboxylate, O-carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, O-carbamate, N- carbamate, C-amide, N-amide, guanyl, guanidine and hydrazine.
  • substituent group can independently be, for
  • the heteroalicyclic group can be an end group, as this phrase is defined hereinabove, where it is attached to a single adjacent atom, or a linking group, as this phrase is defined hereinabove, connecting two or more moieties at two or more positions thereof.
  • Representative examples are piperidine, piperazine, tetrahydrofurane, tetrahydropyrane, morpholino and the like.
  • nitrogen-containing heteroalicyclic groups or moieties include, but are not limited to, Representative examples of nitrogen-containing heteroalicyclic include, but are not limited to, morpholine, thiomorpholine, piperidine, piperazine, hexahydroazepine and tetrahydropyrane. Other moieties are also contemplated.
  • piperidine refers to a group or a group, with R’ as defined herein.
  • pyrrolidine refers to a group or a group, with R’ as defined herein. group, with R’ as defined herein.
  • morpholine refers to a ⁇ - / group, and encompasses also thiomorpholine.
  • thiomorpholine refers t group.
  • hexahydroazepine refers t group.
  • aryl describes an all-carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi-electron system. The aryl group may be substituted or unsubstituted.
  • Substituted aryl may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, cyano, nitro, azo, sulfonamide, C-carboxylate, O- carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, N-carbamate, O-carbamate, C- amide, N-amide, guanyl, guanidine and hydrazine.
  • substituent group can independently be, for example, hydroxyalkyl, tri
  • the aryl group can be an end group, as this term is defined hereinabove, wherein it is attached to a single adjacent atom, or a linking group, as this term is defined hereinabove, connecting two or more moieties at two or more positions thereof.
  • the aryl is phenyl.
  • the aryl is naphthalenyl.
  • heteroaryl describes a monocyclic or fused ring (/. ⁇ ., rings which share an adjacent pair of atoms) group having in the ring(s) one or more atoms, such as, for example, nitrogen, oxygen and sulfur and, in addition, having a completely conjugated pi-electron system.
  • heteroaryl groups include pyrrole, furane, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrimidine, triazine, tetrazine, quinoline, isoquinoline and purine.
  • the heteroaryl group may be substituted or unsubstituted.
  • Substituted heteroaryl may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, cyano, nitro, azo, sulfonamide, C-carboxylate, O- carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, O-carbamate, N-carbamate, C- amide, N-amide, guanyl, guanidine and hydrazine.
  • the heteroaryl group can be an end group, as this phrase is defined here
  • alkaryl describes an alkyl, as defined herein, which is substituted by one or more aryl or heteroaryl groups.
  • An example of alkaryl is benzyl.
  • halide and “halo” describes fluorine, chlorine, bromine or iodine.
  • haloalkyl describes an alkyl group as defined above, further substituted by one or more halide.
  • dithiosulfide refers to a -S-SR’ end group or a -S-S- linking group, as these phrases are defined hereinabove, where R’ is as defined herein.
  • hydroxyl or “hydroxy” describes a -OH group.
  • alkoxy describes both an -O-alkyl and an -O-cycloalkyl group, as defined herein.
  • aryloxy describes both an -O-aryl and an -O-heteroaryl group, as defined herein.
  • thiohydroxy or “thio” describes a -SH group.
  • thioalkoxy describes both a -S-alkyl group, and a -S-cycloalkyl group, as defined herein.
  • thioaryloxy describes both a -S-aryl and a -S-heteroaryl group, as defined herein.
  • nitro describes an -NO2 group.
  • carboxylate as used herein encompasses C-carboxylate and O-carboxylate.
  • thiocarboxylate encompasses “C-thiocarboxylate and O- thiocarb oxy late.
  • carboxylate as used herein encompasses N-carbamate and O-carbamate.
  • thiocarbamate as used herein encompasses N-thiocarbamate and O- thiocarbamate.
  • dithiocarbamate encompasses N-dithiocarbamate and S- dithiocarbamate.
  • amide as used herein encompasses C-amide and N-amide.
  • hydrozine describes a -NR’-NR”R”’ end group or a -NR’ -NR”- linking group, as these phrases are defined hereinabove, with R’, R”, and R'" as defined herein.
  • alkylene glycol describes a -O-[(CR’R”) Z -O]y-R’” end group or a -O-[(CR’R”) Z -O]y- linking group, with R’, R” and R’” being as defined herein, and with z being an integer of from 1 to 10, preferably, from 2 to 6, more preferably 2 or 3, and y being an integer of 1 or more.
  • R’ and R are both hydrogen.
  • z is 2 and y is 1, this group is ethylene glycol.
  • z is 3 and y is 1, this group is propylene glycol.
  • y 2-4, the alkylene glycol is referred to herein as oligo(alkylene glycol).
  • the compound described herein may be in a form of a salt thereof, for example, a pharmaceutically acceptable salt thereof, and/or in a form of a prodrug thereof.
  • the phrase “pharmaceutically acceptable salt” refers to a charged species of the parent compound and its counter-ion, which is typically used to modify the solubility characteristics of the parent compound and/or to reduce any significant irritation to an organism by the parent compound, while not abrogating the biological activity and properties of the administered compound.
  • a pharmaceutically acceptable salt of the compounds described herein may optionally be a base addition salt comprising at least one acidic (e.g., phenol and/or carboxylic acid) group of the compound which is in a negatively charged form (e.g., wherein the acidic group is deprotonated), in combination with at least one counter-ion, derived from the selected base, that forms a pharmaceutically acceptable salt.
  • a base addition salt comprising at least one acidic (e.g., phenol and/or carboxylic acid) group of the compound which is in a negatively charged form (e.g., wherein the acidic group is deprotonated), in combination with at least one counter-ion, derived from the selected base, that forms a pharmaceutically acceptable salt.
  • the base addition salts of the compounds described herein may therefore be complexes formed between one or more acidic groups of the drug and one or more equivalents of a base.
  • the base addition salts may include a variety of organic and inorganic counter-ions and bases, such as, but not limited to, sodium (e.g., by addition of NaOH), potassium (e.g., by addition of KOH), calcium (e.g., by addition of Ca(OH)2, magnesium (e.g., by addition of Mg(OH)2), aluminum (e.g., by addition of Al(0H)3 and ammonium (e.g., by addition of ammonia).
  • bases such as, but not limited to, sodium (e.g., by addition of NaOH), potassium (e.g., by addition of KOH), calcium (e.g., by addition of Ca(OH)2, magnesium (e.g., by addition of Mg(OH)2), aluminum (e.g., by addition of Al(0H)3 and ammonium (e.g., by addition of ammonia).
  • bases such as, but not limited to, sodium (e.g., by addition of NaOH), potassium (e.g.,
  • a pharmaceutically acceptable salt of the compounds described herein may optionally be an acid addition salt comprising at least one base group (e.g., amine or amide group) of the compound which is in a positively charged form (e.g., wherein an -NH- group is protonated), in combination with at least one counter-ion, derived from the selected acid, that forms a pharmaceutically acceptable salt.
  • base group e.g., amine or amide group
  • a positively charged form e.g., wherein an -NH- group is protonated
  • the acid addition salts of the compounds described herein may therefore be complexes formed between one or more basic groups of the drug and one or more equivalents of an acid.
  • the acid addition salts may include a variety of organic and inorganic acids, such as, but not limited to, hydrochloric acid which affords a hydrochloric acid addition salt, hydrobromic acid which affords a hydrobromic acid addition salt, acetic acid which affords an acetic acid addition salt, ascorbic acid which affords an ascorbic acid addition salt, benzenesulfonic acid which affords a besylate addition salt, camphorsulfonic acid which affords a camphorsulfonic acid addition salt, citric acid which affords a citric acid addition salt, maleic acid which affords a maleic acid addition salt, malic acid which affords a malic acid addition salt, methanesulfonic acid which affords a methanesulfonic acid (mesylate) addition salt, naphthalenesulfonic acid which affords a naphthalenesulfonic acid addition salt, oxalic acid which affords an oxalic acid addition salt,
  • the acid or base additions salts can be either monoaddition salts or poly-addition salts.
  • addition salt refers to a salt in which the stoichiometric ratio between the counter-ion and charged form of the compound is 1 : 1, such that the addition salt includes one molar equivalent of the counter-ion per one molar equivalent of the compound.
  • poly-addition salt refers to a salt in which the stoichiometric ratio between the counter-ion and the charged form of the compound is greater than 1 : 1 and is, for example, 2: 1, 3 : 1, 4: 1 and so on, such that the addition salt includes two or more molar equivalents of the counter-ion per one molar equivalent of the compound.
  • prodrug refers to a compound which is converted in the body to an active compound (e.g., the compound of the formula described hereinabove).
  • a prodrug is typically designed to facilitate administration, e.g., by enhancing absorption.
  • a prodrug may comprise, for example, the active compound modified with ester groups, for example, wherein any one or more of the hydroxyl groups of a compound is modified by an acyl group, optionally (Ci- 4)acyl (e.g., acetyl) group to form an ester group, and/or any one or more of the carboxylic acid groups of the compound is modified by an alkoxy or aryloxy group, optionally (Ci-4)alkoxy (e.g., methyl, ethyl) group to form an ester group.
  • an acyl group optionally (Ci- 4)acyl (e.g., acetyl) group to form an ester group
  • any one or more of the carboxylic acid groups of the compound is modified by an alkoxy or aryloxy group, optionally (Ci-4)alkoxy (e.g., methyl, ethyl) group to form an ester group.
  • each of the compounds described herein, including the salts thereof, can be in a form of a solvate or a hydrate thereof.
  • solvate refers to a complex of variable stoichiometry (e.g., di-, tri-, tetra-, penta- , hexa-, and so on), which is formed by a solute (the heterocyclic compounds described herein) and a solvent, whereby the solvent does not interfere with the biological activity of the solute.
  • hydrate refers to a solvate, as defined hereinabove, where the solvent is water.
  • the compounds described herein can be used as polymorphs and the present embodiments further encompass any isomorph of the compounds and any combination thereof.
  • the present embodiments further encompass any enantiomers and diastereomers of the compounds described herein.
  • enantiomer refers to a stereoisomer of a compound that is superposable with respect to its counterpart only by a complete inversion/reflection (mirror image) of each other. Enantiomers are said to have “handedness” since they refer to each other like the right and left hand. Enantiomers have identical chemical and physical properties except when present in an environment which by itself has handedness, such as all living systems.
  • a compound may exhibit one or more chiral centers, each of which exhibiting an R- or an ⁇ '-configuration and any combination, and compounds according to some embodiments of the present invention, can have any their chiral centers exhibit an R- or an S- configuration.
  • diastereomers refers to stereoisomers that are not enantiomers to one another. Diastereomerism occurs when two or more stereoisomers of a compound have different configurations at one or more, but not all of the equivalent (related) stereocenters and are not mirror images of each other. When two diastereoisomers differ from each other at only one stereocenter they are epimers. Each stereo-center (chiral center) gives rise to two different configurations and thus to two different stereoisomers.
  • embodiments of the present invention encompass compounds with multiple chiral centers that occur in any combination of stereo-configuration, namely any diastereomer.
  • the present inventors have first selected 1,4’ -bipiperidine as an exemplary protecting group, based on a successful performance of such a group in the drug known as CPT-11 (Irinotecan®) [See, for example, Kunimoto T et al., Cancer Res., 1987, 47, 5944-5947],
  • TNK-002 an exemplary TNIK inhibitor
  • TNK-108 a compound referred to herein as TNK-108 (see, FIG. 1 A)
  • 1,4'-Bipiperidine (99 %; Compound 4A) was obtained from BIDE;
  • reaction mixture was concentrated give a residue, which was purified by reversed-phase HPLC (0.1 % FA condition), followed by further purification by preparative HPLC (HC1 condition; column: Phenomenex Synergi C18 150x25x10pm; mobile phase : [water(0.05%HCl)-ACN];B% : 20%-40%,10 minutes) to give TNK-108 (57.95 mg, 2.68 % yield, 97.6 % purity, HC1 salt) as a yellow solid.
  • FIGs. 5A-B A general synthetic pathway for preparing TNK-108 on a gram scale is shown in FIGs. 5A-B.
  • 1,4'-Bipiperidine (95 %; Compound 13) was obtained from BIDE;
  • TNK-127 was prepared, upon using respective starting materials.
  • FIG. 6 A general synthetic pathway for preparing TNK-127 on a gram scale is shown in FIG. 6.
  • 1,4'-Bipiperidine (95 %; Compound 13) was obtained from BIDE;
  • N-bromosuccinimide (NBS; 99 %) was obtained from TaiTan; N,N’ -Carbonyldiimidazole (CDI; 99 %) was obtained from Aladdin;
  • reaction mixture was diluted with ethyl acetate (50 mL), washed with brine (40 mL x 3), dried over Na2SO4, filtered and concentrated under vacuum to give compound 22 (1.60 gram, crude) as brown liquid, which was confirmed by 1 H-NMR.
  • TNK-135 was prepared, upon using respective starting materials.
  • FIG. 7 A general synthetic pathway for preparing TNK-135 is shown in FIG. 7.
  • N-bromosuccinimide (NBS; 99 %) was obtained from TaiTan;
  • N,N’ -Carbonyldiimidazole (CDI; 99 %) was obtained from Aladdin;
  • Exemplary such compounds are TNK-124 and TNK-128 (see, FIG. 1A), prepared as follows.
  • FIG. 8 A general synthetic pathway for preparing TNK-128 is shown in FIG. 8.
  • TNK-108 The properties of TNK-108 were tested and compared to those of the parent compound TNK-002 (also referred to herein as KY-08405), according to the following protocols.
  • TNIK Kinase Assay is a luminescent kinase assay that measures ADP formed from TNIK kinase reaction.
  • the assay uses ADP-GloTM Kinase Assay (Promega Cat# V910) and TNIK Kinase Enzyme System (Promega cat# V4158).
  • TNIK kinase domain
  • MBP maltose binding protein
  • ATP phosphatidylcholine
  • IC50 for compound inhibition is obtained from a 4-parameter logistic curve using GraphPad Prism software.
  • the CellTiter-Glo® Luminescent Cell Viability Assay (Promega Cat# G7570) is used to determine the number of viable cells in culture based on quantitation of the ATP present, which signals the presence of metabolically active cells.
  • SW620 cells/well are seeded in a 96 well plate. 24 hours later, tested compounds in DMSO are added at a nine-point dilution curve in medium with 0.1% FBS and 1% DMSO. Compounds are tested in an independent triplicate. After 48 hours incubation at 37 °C, CellTiter Gio reagent is added and luminescence is recorded. A direct relationship exists between luminescence measured with the CellTiter-Glo® Assay and the number of cells in culture. IC50 for compound inhibition is obtained from a 4-parameter logistic curve using GraphPad Prism software.
  • the tested compound (10 mM) in DMSO 10 pL was added into the lower chamber of Whatman miniuniprep vials. 490 pL of a selected phosphate buffer (pH 3.5, 4.5 or 7.4) and 0.1N HC1 were added thereto and the samples were vortexed for 2 minutes. The vials were then shaked on a Barnstead shaker for 24 hours at room temperature, at a speed of 800 rpm, and were thereafter centrifuged for 20 minutes (at about 4000 rpm). The miniunipreps were then compressed to prepare the filtrate for injection into HPLC, and concentrations were calculated. HPLC analyses were performed using HPLC-AW(19#139), equipped with Waters XBridge C18 4.6*100 mm column.
  • Detector was operated at 282 nm. Flow rate: 1.4 mL/minute. Mobile phase A: 0.1% TFA in water. Mobile phase B: 0.1 % TFA in CAN. Gradient: from 70:30 to 30:70 in 5 minutes.
  • An intermediate solution was prepared by diluting 5 pL of a stock solution containing 10 mM of the tested compound in dimethyl sulfoxide (DMSO)) with 495 pL of methanol (MeOH). The intermediate solution concentration was 100 pM in 99 % MeOH).
  • DMSO dimethyl sulfoxide
  • MeOH methanol
  • NADPH solution P-Nicotinamide adenine dinucleotide phosphate reduced form, tetrasodium salt
  • NADPH 4Na Vendor: Chem-Impex International, Cat. No. 00616 powder was weighed and diluted into a 10 mM MgCb solution, to thereby obtained an NADPH working solution at a concentration of 10 unit/mL.
  • the final concentration of NADPH in in the reaction system is 1 unit/mL.
  • Microsome solutions Human liver microsomes (HLM) were obtained from Corning (Cat No. 452117; Lot No. 38292). CD-I mouse microsomes (MLM) were obtained from Biopredic (CatNo. BQM1000; LotNo. MIC255036). The appropriate concentrations of microsome working solutions were prepared in 100 mM potassium phosphate buffer.
  • Stop Solution Cold (4 °C) acetonitrile (ACN) containing 100 ng/mL tolbutamide and 100 ng/mL labetalol as internal standards (IS) was used as the stop solution.
  • an Apricot automation workstation was used to add 10 pL/well of NADPH regenerating system to every reaction plate except NCF60 (Blank, TO, T5, T10, T20, T30, and T60) to start the reaction.
  • NCF60 Binary, TO, T5, T10, T20, T30, and T60.
  • the reaction plates were incubated at 37 °C, and timer 2 was started.
  • the final concentration of microsomes in the incubation medium was 0.5 mg protein/mL.
  • the final concentration of a tested or control compound in the incubation medium was 1 pM.
  • the final concentration of MeOH in the incubation medium was 0.99 %.
  • the final concentration of DMSO in the incubation medium was 0.01 %.
  • An Apricot automation workstation was used to add 300 pL/well of stop solution to each reaction plate at its appropriate end time point (5, 10, 20, 30 or 60 minutes) to terminate the reaction.
  • stop solution was added prior to the addition of microsomes and NADPH.
  • Each plate was sealed and shaken for 10 minutes. After shaking, each plate was centrifuged at 4000 rpm and 4 °C for 20 minutes.
  • an Apricot automation workstation was used to add 300 pL/well of HPLC grade water to eight new 96-well plates. After centrifugation, an Apricot automation workstation was used to transfer 100 pL of supernatant from each reaction plate to its corresponding bioanaylsis plate. Each bioanalysis plate was sealed and shaken for 10 minutes prior to LC-MS/MS analysis.
  • TNK-108 TNK-108
  • KY-08405 TNK-128, TNK-127
  • Propantheline bromide positive control
  • 1 mM intermediate solution was prepared by diluting 10 pL of a stock solution (10 mM in DMSO) with 90 pL DMSO
  • Propantheline was prepared by diluting 10 pL of a stock solution (10 mM in DMSO) with 90 pL ultrapure water.
  • CD-I mouse and human plasma were tested. The pooled frozen plasma was thawed in a water bath at 37 °C prior to experiment. Plasma was centrifuged at 4000 rpm for 5 minutes and the clots, if present, were removed. The pH was adjusted to 7.4 ⁇ 0.1 when required.
  • 100 pM dosing solution was prepared by diluting 20 pL of the intermediate solution (1 mM) with 180 pL 45 % MeOH/H 2 O.
  • a stop solution 200 ng/mL tolbutamide and 200 ng/mL Labetalol in 50 % ACN/MeOH was added to precipitate protein, and mixed thoroughly.
  • Sample plates were thereafter centrifuged at 4,000 rpm for 10 minutes. An aliquot of supernatant (50 pL) was transferred from each well and mixed with 100 pL ultrapure water. The samples were shaked at 800 rpm for about 10 minutes before being subjected to LC-MS/MS analysis.
  • LC-MS was performed as described hereinabove, equipped with ACQUITY UPLC BEH C18, 1.7 pm, 2.1x50 mm, while using as a mobile phase A: 0.1% formic acid in water; B: 0.1% formic acid in acetonitrile, at a flow rate of 700 pL/minute.
  • TNK-128, and of TNK-127 and TNK-135 and their counterpart TNK- 007 were also tested using the above protocols.
  • the biocleavable compounds TNK-108 and TNK-128 are inactive in the in vitro kinase assay, probably due to steric hindrance within the TNIK pocket.
  • the biocleavable compounds become active in the low micromolar range in the cell-based assay after generating TNK-002 upon contacting intracellular esterases.
  • TNK-108 exhibits improved solubility compared to the parent TNK-002 and TNK-128.
  • TNK-108 is quite stable in mouse and human microsomes. It generates minute amounts of TNK-002 in mice plasma.
  • TNK-128 generates substantial amount of TNK-002 in mice plasma.
  • TNK-127 and TNK-135 are less active in the kinase assay than TNK-007 while the activity of these 3 compounds in the CellTiter Gio assay is moderate. Both TNK-127 and TNK- 135 are significantly more soluble and more stable in microsomes than TNK-007 and are very stable in mouse and human plasma. Table 1
  • IV: KY-08405, 10 mg/kg (5 mL/kg) via tail vein injection (N 3/time point)
  • the plasma samples were analyzed by LC-MS, as described hereinabove. Concentrations of TNK-002, TNK-108 and glucuronidation metabolites thereof were determined.
  • FIG. 9 presents comparative plots showing the mean plasma concentration following a single IV or PO administration of TNK-108, and clearly show that the bioavailability of TNK- 108 is high, about 77%; and its plasma concentration after oral administration remains around the EC50 (determined in a cell-based assay) for many hours, indicating low clearance.
  • the area under the curve (AUC) is high.
  • FIG. 10 presents comparative plots showing the mean plasma concentration following a single IV or PO administration of mol equivalent amounts of TNK-108 and TNK-002.
  • TNK-002 The measured oral bioavailability of TNK-002 was 1.3 %, much lower than that of TNK- 108.
  • the AUC values are 4-5-folds lower for TNK-002 compared to TNK-108.
  • the half-life in blood (Tl/2) measured for TNK-108 is 5.7 hours, and for TNK-002 4.2 hours.
  • FIG. 11 presents comparative plots showing the mean plasma concentration of a respective glucuronide metabolite following a single IV or PO of mol equivalent amounts of TNK-108 and TNK-002. As can be seen, much lower amounts of the glucuronide metabolite were observed following administration of TNK-108, compared to TNK-002. As can be further seen, following PO administration, the amount of the glucuronide metabolite of TNK-002 is higher by several orders of magnitude compared to that of TNK-108.
  • TNK-128 Similar assays were conducted with TNK-128. The obtained data is presented in FIGs. 12A-B. As can be seen, plasma concentrations of TNK-128 are higher than those of TNK-002, both when administered orally and IV.
  • TNK-127 exhibits higher plasma concentrations upon IV administration, and for a longer time period.
  • mice Female BALB/c nude mice (Mus miiscuhis). aged 6-8 weeks, and weighing 18-22 grams were kept in individual ventilation cages at constant temperature (20-26 °C) and humidity (40- 70 %) with 5 animals in each cage. Animals had free access to irradiation sterilized dry granule food and to sterile drinking water during the entire study period.
  • SW620 tumor cells were maintained in vitro as a monolayer culture in Leibovitz's L-15 medium supplemented with 10 % heat inactivated fetal bovine serum, 100 U/mL Penicillin and 100 pg/mL Streptomycin at 37 °C in an atmosphere of 0 % CO2 in air.
  • the tumor cells were routinely subcultured twice weekly by trypsin-EDTA treatment.
  • the cells growing in an exponential growth phase were harvested and counted for tumor inoculation.
  • Each mouse was inoculated subcutaneously at the right flank with SW620 cancer cells (5 x 10 6 ) in 0.1 mL of PBS for tumor development. Treatments were started on day 14 after tumor inoculation when the average tumor size reached approximately 162 mm 3 .
  • the animals were assigned into groups according to a sorting SOP based upon their tumor volumes. Each group consisted of 10 tumor-bearing mice.
  • Vehicle 1 includes 10 % propylene glycol (PG), 10 % Cremophor and 80 % or a 5 %HP-P- CD aqueous solution.
  • Vehicle 2 includes 20 % PEG400, 5 % Tween®80 and 75 % or a 5 %HP-P-CD aqueous solution.
  • TNK-128 preparations at a concentration of 3.75, 7.5, or 15, mg/ml were prepared by dissolving TNK-128 in PG, adding Cremophor EL to the solution and stirring until a homogeneous mixture is obtained, and thereafter adding 5 % HP-P-CD in water and vortexing for 1 minute, for obtaining a clear solution.
  • TNK-108 preparations at a concentration of 3.5 or 5, mg/ml were prepared by dissolving TNK-108 in PEG400, adding Tween®80 to the solution and stirring until a homogeneous mixture is obtained, and thereafter adding 5 % HP-P-CD in water and vortexing for 1 minute, for obtaining a clear solution.
  • the animals were daily checked for any effects of tumor growth and treatments on normal behavior such as mobility, food and water consumption (by looking only), body weight gain/loss (body weights were measured twice per week), eye/hair matting and any other abnormal effect as stated in the protocol. Death and observed clinical signs were recorded on the basis of the numbers of animals within each subset.
  • the tumor size was then used for calculation of RTV and T/C values.
  • Tumor weight was measured at the study termination.
  • FIGs. 14A-B are bar graphs showing the average tumor volume at day 25 of animals treated with a TNK-128 preparation at the indicated dose or with the respective vehicle (FIG. 14 A) and of animals treated with a TNK-108 preparation, at the indicated dose, and of the respective vehicle (FIG. 14B), and show that treatment with both compounds resulted in reduction in tumor volume.

Landscapes

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

Abstract

Compounds represented by Formula (I): wherein W is a nitrogen-containing moiety attached to said X via said nitrogen atom, and all other variables are as defined in the specification, processes of preparing same and uses thereof for treating medical conditions associated with upregulated activity of TNIK are provided.

Description

Traf2- AND Nck-INTERACTING KINASE (TNIK) INHIBITORS AND USES THEREOF
RELATED APPLICATION/S
This application claims the benefit of priority under 35 USC §119(e) of U.S. Provisional Patent Application No. 63/063,491 filed on August 10, 2020, the contents of which are incorporated herein by reference in their entirety.
FIELD AND BACKGROUND OF THE INVENTION
The present invention, in some embodiments thereof, relates to therapy and, more particularly, but not exclusively, to novel compounds which act as inhibitors of Traf2- and Nck- interacting kinase (TNIK), to pharmaceutical compositions comprising same and to uses thereof in the treatment of medical conditions associated with Traf2- and Nck-interacting kinase (TNIK), such as, for example, cancer.
Inhibition of Traf2- and Nck-interacting kinase (TNIK) has been described in the art as useful for treating cancer (see, e.g., U.S. Patent Application Publication no. 2010/0216795). It has been shown that TNIK is hyperactive in various types of cancers, including, for example, colorectal cancer, a cancer, brain tumor, gastric cancer, liver cancer, and ovarian cancer [J.S. Boehm et al., Cell 129, 1065-1079, 2007],
For example, TNIK plays an important role in the growth of colorectal cancer, and was reported to be a target for controlling aberrant Wnt signaling in colorectal cancer (Cancer Res 2010;70:5024-5033). TNIK gene was found to be over-expressed in 7 % of gastric cancer patients’ tissue samples, thus being a target for treating gastric cancer (Oncogenesis 2014, 3, e89). TNIK was also reported to be associated with proliferation and differentiation of leukemia stem cells in chronic myelogenous leukemia (Journal of Clinical Investigation, 2012, 122, 624).
In addition, other cancers or tumors, such as, for example, hepatocellular carcinoma, desmoid tumor, medulloblastoma (pediatric brain tumor), Wilms tumor (pediatric kidney cancer), and thyroid tumor, which are associated with aberrant Wnt signaling, or which are otherwise associated with one or more other TNIK signaling pathways, can benefit from TNIK inhibition.
TNIK inhibition was also reported to be useful in treating other medical conditions, including, for example, chronic obstructive pulmonary disease (COPD), lupus nephritis, diabetic nephropathy, focal segmental glomerulosclerosis, renal fibrosis, pulmonary fibrosis, and scleroderma, and other inflammatory diseases and disorders.
WO 2019/156439 and WO 2019/156438 disclose compounds featuring a lH-pyrazol-3- amine skeleton, substituted by aryls and/or heteroaryls, which act as TNIK inhibitors. Additional background art includes WO 2009/030890 and WO 2010/100431, which disclose pyrimidine and pyrrolopyrimidine compounds, respectively, that are capable of inhibiting one or more kinases; Karthikeyan et al., Bioorg. Biomed. Chem Lett., 2017, Vol. 27, 99.2663-7, which discloses N-(lH-pyrazole-3-yl)-quinazoline-4-amines as kinase inhibitors usable in treating neurodegenerative diseases and cancer; and WO 01/79198, WO 2005/009435; WO 2015/120390; Bahaj et al., Chem. Pharm. Bull., 2005, Vol. 53, No. 6, pp. 611-615; and Singh et al., J. Chem. Res., 2007, Vol. 4, pp. 229-232, which disclose amino-pyrazole compounds. Additional background art includes Fandrick D.R., Org. Lett. 2015, 17, 12, 2964-2967. Additional background art includes U.S. Patent No. 8,530,444; U.S. Patent No. 4,604,463 and Kunimoto T et al., Cancer Res., 1987, 47, 5944-5947.
SUMMARY OF THE INVENTION
According to an aspect of some embodiments of the present invention there is provided a compound represented by Formula I: a
Figure imgf000004_0001
Formula I wherein: Y is N or CRs; Z is N or CIU; Ri, and R2 are each independently hydrogen, alkyl or cycloalkyl; R3 and R4 are each independently hydrogen, alkyl, haloalkyl, or halo; R5 and Re are each independently hydrogen, alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, or aryloxy; B is hydrogen, alkyl, haloalkyl, halo, alkoxy, thioalkoxy, or cycloalkyl; A is a substituted or unsubstituted aryl or heteroaryl, or, alternatively, forms with Ri a substituted or unsubstituted heteroalicyclic ring or forms with R3 a substituted or unsubstituted cyclic ring; X is -O-C(=O)- or -NH-C(=O)-; and W is a nitrogen-containing moiety attached to the X via the nitrogen atom.
According to some of any of the embodiments described herein, Ri, and R2 are each hydrogen.
According to some of any of the embodiments described herein, Y is CR5. According to some of any of the embodiments described herein, R5 is hydrogen.
According to some of any of the embodiments described herein, Z is CRe.
According to some of any of the embodiments described herein, Rs is hydrogen.
According to some of any of the embodiments described herein, R4 is hydrogen.
According to some of any of the embodiments described herein, B is alkyl.
According to some of any of the embodiments described herein, X is -O-C(=O).
According to some of any of the embodiments described herein, A is a substituted or unsubstituted aryl.
According to some of any of the embodiments described herein, A is a substituted aryl.
According to some of any of the embodiments described herein, the aryl is substituted by a heteroaryl.
According to some of any of the embodiments described herein, the aryl is substituted by pyrazole.
According to some of any of the embodiments described herein, W is or comprises a nitrogen-containing heteroalicyclic ring.
According to some of any of the embodiments described herein, W is a nitrogen-containing heteroalicyclic ring attached to the X via a nitrogen atom of the nitrogen-containing heteroalicyclic ring.
According to some of any of the embodiments described herein, the nitrogen-containing heteroalicyclic ring is substituted by an alkyl, cycloalkyl or a heteroalicyclic.
According to some of any of the embodiments described herein, W is a substituted piperidinyl.
According to some of any of the embodiments described herein, W is 1,4-bipiperidinyl.
According to some of any of the embodiments described herein, the piperidinyl is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom.
According to some of any of the embodiments described herein, W is a substituted 1,4- piperazinyl.
According to some of any of the embodiments described herein, the piperazinyl is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom.
According to some of any of the embodiments described herein, W is an aminoalkyl, attached to the X via the amine.
According to some of any of the embodiments described herein, the aminoalkyl is substituted by the nitrogen-containing heteroalicyclic. According to some of any of the embodiments described herein, A is phenyl substituted by pyrazole.
According to some of any of the embodiments described herein, X is -O-C(=O)- and W is 1,4-bipiperidinyl.
According to some of any of the embodiments described herein, X is -NH-C(=O)- and W is 1,4-bipiperidinyl.
According to some of any of the embodiments described herein, X is -O-C(=O)- and W is 1,4-piperazinyl substituted by a linear hydrocarbon chain interrupted by at least one O atom.
According to some of any of the embodiments described herein, X is -NH-C(=O)- and W is an aminoalkyl, attached to the X via the amine, and wherein the aminoalkyl is substituted by the nitrogen-containing heteroalicyclic.
According to an aspect of some embodiments of the present invention there is provided a pharmaceutical composition comprising the compound of Formula I as described herein in any of the respective embodiments and any combination thereof and a pharmaceutically acceptable carrier.
According to some of any of the embodiments described herein, the pharmaceutical composition is formulated for oral administration.
According to an aspect of some embodiments of the present invention there is provided a compound of Formula I as described herein in any of the respective embodiments and any combination thereof, or the pharmaceutical composition as described herein in any of the respective embodiments and any combination thereof, for use in treating a medical condition associated with upregulated activity of TNIK.
According to some of any of the embodiments described herein, the medical condition is cancer.
According to some of any of the embodiments described herein, the treating comprises orally administering the compound or the composition to a subject in need thereof.
According to an aspect of some embodiments of the present invention there is provided a process of preparing the compound of Formula I according to any of the respective embodiments and any combination thereof, the process comprising: reacting a compound of Formula Xa:
Figure imgf000007_0001
wherein Q is hydroxy, in case X is -O-C(=O)-, or amine, in case X is -NH-C(=O)-, and Li is a first reactive group; with a compound of Formula Xb:
W-L2
Formula Xb, wherein W-L2 is or comprises a second reactive group, under conditions that generate the X upon coupling the W-L2 with the Q, to thereby generate a compound of Formula Xc:
Figure imgf000007_0002
and reacting the compound of Formula Xc with a compound of Formula Xd:
Figure imgf000007_0003
Formula Xd wherein L3 is a third reactive group that generates, upon reacting with Li, the -N(R2)-, under conditions that generate the -N(R.2)- upon coupling the Li and L3, , to thereby generate the compound of Formula I.
Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWING(S)
Some embodiments of the invention are herein described, by way of example only, with reference to the accompanying drawings. With specific reference now to the drawings in detail, it is stressed that the particulars shown are by way of example and for purposes of illustrative discussion of embodiments of the invention. In this regard, the description taken with the drawings makes apparent to those skilled in the art how embodiments of the invention may be practiced.
In the drawings:
FIGs. 1A-B present the chemical structures of exemplary compounds according to some embodiments of the present invention;
FIGs. 2A-B present schemes showing exemplary unsuccessful synthetic pathways for providing TNK-108;
FIGs. 3A-B present schemes showing additional exemplary unsuccessful synthetic pathways for providing TNK-108;
FIG. 4 presents an exemplary synthetic pathway for providing TNK-108;
FIGs. 5A-B present another exemplary synthetic pathway for providing TNK-108;
FIG. 6 presents an exemplary synthetic pathway for providing TNK-127;
FIG. 7 presents an exemplary synthetic pathway for providing TNK-135;
FIG. 8 presents an exemplary synthetic pathway for providing TNK-128;
FIG. 9 presents comparative plots showing the mean plasma concentration following a single IV or PO administration of TNK-108;
FIG. 10 presents comparative plots showing the mean plasma concentration of TNK-002 (KY-08405) following a single IV or PO administration of mol equivalent amounts of TNK-108 and TNK-002 (KY-08405); FIG. 11 presents comparative plots showing the mean plasma concentration of a respective glucuronide metabolite following a single IV or PO of mol equivalent amounts of TNK-108 and TNK-002 (KY-08405).
FIGs. 12A-B present comparative plots showing the mean plasma concentration profiles of TNK-128, TNK-002 (KY-08405) and its glucuronide metabolite following a single IV (FIG. 12A) or PO (FIG. 12B) administration of TNK-128 at 10 mg/kg in male C57BL/6 mice (N=3/time point).
FIG. 13 presents comparative plots showing the mean plasma concentration profiles of TNK-127 after single IV and PO administrations in male C57BL/6 mice (N=3/time point).
FIGs. 14A-B are bar graphs showing the effect of treatment with TNK-128 (FIG. 14A) and TNK-108 (FIG. 14B) on the tumor volume of mice inoculated with SW620 human colorectal cancer, at day 25 of the study.
DESCRIPTION OF SPECIFIC EMBODIMENTS OF THE INVENTION
The present invention, in some embodiments thereof, relates to therapy and, more particularly, but not exclusively, to novel compounds which act as Traf2- and Nck-interacting kinase (TNIK), to pharmaceutical compositions comprising same and to uses thereof in the treatment of medical conditions associated with Traf2- and Nck-interacting kinase (TNIK), such as, for example, cancer.
Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not necessarily limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other embodiments or of being practiced or carried out in various ways.
While exploring the activity of TNIK inhibitors such as disclosed in, for example, WO 2019/156439, the present inventors have uncovered that (i) the oral bioavailability of such compounds is very low (e.g., lower than 10 % and even lower than 5 %); and (ii) the blood exposure (AUC) of such compounds is low. The present inventors have uncovered that compounds such as disclosed in WO 2019/156439, which possess a phenol moiety, undergo substantial Phase 2 metabolism, for example, glucuronidation and possibly also sulfation, which adversely affects their activity; causes low blood exposure after IV and especially PO administration; and results in a low oral bioavailability.
In a search for a modification of such compounds that would overcome the above limitations, and would result, for example, in enhanced bioavailability, improved pharmacokinetic properties (e.g. enhanced blood exposure (AUC)), and reduced or nullified glucuronidation for compounds that possess a phenol moiety, while maintaining the compounds’ TNIK inhibition activity, the present inventors have conceived a methodology for modifying the chemical structure of such compounds.
As a first step, while compounds that feature a phenol moiety were identified as effective TNIK inhibitor, the present inventors have search for a methodology for protecting the phenol moiety to thereby reduce or nullify Phase 2 metabolism that results in low oral bioavailability and low blood exposure.
The present inventors have first selected 1,4’ -bipiperidine as an exemplary protecting group, based on a successful performance of such a group in the drug known as CPT-11 (Irinotecan®) [See, for example, Kunimoto T et al., Cancer Res., 1987, 47, 5944-5947],
During the laborious studies conducted for practicing such a modification, several strategies for coupling 1,4’ -bipiperidine to the phenol moiety in an exemplary TNIK inhibitor, referred to herein as TNK-002, so as to provide a compound referred to herein as TNK-108 (see, FIG. 1A), have been tested. As described in further detail in the Examples section that follows, and shown in FIGs. 2A-B and 3A-B, a simple coupling of 1,4’ -bipiperidine to the phenol moiety, using common coupling methodologies, did not provide the desired compound.
The present inventors have recognized that a more sophisticated synthetic route should be employed, which is not based on coupling the 1,4’ -bipiperidine to a phenol moiety, but rather involves “building” the final molecule differently. During laborious experimentation, the present inventors have uncovered a synthetic pathway which was successfully employed for preparing compounds in which a piperidine- or piperazine-containing moiety “masks” a phenol or aniline moiety in a TNIK compound, and have practiced this synthetic pathway for preparing compounds such as shown in FIGs. 4-8.
In studies conducted to explore the activity and ADME (absorption, distribution, metabolism, excretion) and pharmacokinetic properties of these compounds, it has been shown that an exemplary such compound, which is referred to herein as TNK-108, exhibits high bioavailability (e.g., higher than 50 % and even higher than 60 %, when tested in mice), high plasma concentration following oral administration, as shown in FIGs. 9 and 10, which remains even 24 hours following administration, as shown in FIG. 10, and which remains around the EC50 of the compound almost 8 hours following oral administration, as shown in FIG. 9. This compound also exhibits Tl/2 which is higher by about 30 % compared to its parent compound TNK-002. FIG. 11 presents the plasma concentration of the glucuronide metabolite and show much lower amounts of this metabolite compared to the parent compound TNK-002. Similar observations were obtained for another exemplary compound, referred to herein as TNK-128, as shown, for example, in FIGs. 12A-B and in Table 1 in the Examples section that follows. The anti-cancer activity of TNK-128 and TNK-108 is demonstrated in FIGs. 14A-B. Non-cleavable compounds, referred to herein as TNK-127 and TNK-135, also presented improved performance (e.g., improved solubility and plasma stability), as shown for example, in FIG. 13 and Table 1.
The novel methodology for preparing TNIK inhibitors that have structural features similar to those described in WO 2019/156439, yet exhibit improved performance at least in terms of their pharmacokinetic properties, paves the way to a myriad of compounds that can be beneficially utilized for inhibiting TNIK and as therapeutically active agents for treating medical conditions in which downregulation of TNIK is beneficial.
Embodiments of the present invention relate to novel TNIK inhibitors, to processes of preparing same and to uses thereof in treating medical conditions that are associated with TNIK (e.g., mediated by TNIK activity).
Compounds:
The compounds of the present embodiments can be collectively represented by Formula I:
Figure imgf000011_0001
Formula I wherein:
Y is N or CR5;
Z is N or CRs;
Ri and R2 are each independently hydrogen, alkyl or cycloalkyl;
R3 and R4 are each independently hydrogen, alkyl, haloalkyl, or halo;
R5 and Re are each independently hydrogen, alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, or aryloxy;
B is hydrogen, alkyl, haloalkyl, halo, alkoxy, thioalkoxy, or cycloalkyl;
A is a substituted or unsubstituted aryl or heteroaryl, or, alternatively, forms with Ri a substituted or unsubstituted heteroalicyclic ring or forms with R3 a substituted or unsubstituted cyclic ring; X is -O-C(=O)- or -NH-C(=O)-; and
W is a nitrogen-containing moiety attached to said X via said nitrogen atom.
According to some of any of the embodiments described herein, Ri is hydrogen. Alternatively, Ri is alkyl.
According to some of any of the embodiments described herein, R2 is hydrogen. Alternatively, R2 is alkyl.
According to some of any of the embodiments described herein, Ri and R2 are each hydrogen.
It is to be noted that when R2 is hydrogen, the compound is incapable of undergoing a simple coupling to the moiety W, due to the reactivity of the respective bridging amine. It is to be moreover noted that when Ri is hydrogen, the compound is incapable of undergoing a simple coupling to the moiety W, due to the high reactivity of the respective pyrazole nitrogen, as shown, for example , in FIG. 2B.
According to some of any of the embodiments described herein, R4 is hydrogen. Alternatively, R4 is halo, alkyl (e.g., methyl or ethyl) or haloalkyl (e.g., trihaloalkyl such as CF3).
According to some of any of the embodiments described herein, Y is CR5. According to exemplary embodiments, R5 is hydrogen. Alternatively, R5 is halo or alkyl (e.g., methyl or ethyl) or haloalkyl (e.g., trihaloalkyl such as CF3).
According to some of any of the embodiments described herein, Z is CRe. According to exemplary embodiments, Rs is hydrogen. Alternatively, Re is halo or alkyl (e.g., methyl or ethyl) or haloalkyl (e.g., trihaloalkyl such as CF3).
According to exemplary embodiments, R5 is hydrogen. Alternatively, R5 is halo or alkyl (e.g., methyl or ethyl) or haloalkyl (e.g., trihaloalkyl such as CF3).
According to some of any of the embodiments described herein, Y is CR5 and Z is CRs, such that the respective ring is a phenyl ring, which can be substituted, in case one or more of R4, R5, Re or B is other than hydrogen.
According to some of any of the embodiments described herein, B is hydrogen, halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3).
According to some of any of the embodiments described herein, one R4, R5, Rs or B is halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3). According to some of these embodiments, the other substituents on the phenyl are all hydrogen.
According to some of any of the embodiments described herein, B is alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3). According to some of any of the embodiments described herein, B is alkyl (e.g., methyl or ethyl).
According to some of any of the embodiments described herein, B is alkyl (e.g., methyl or ethyl), Y is CR5, Z is CRs, and R4, R5, and Rs are each hydrogen.
According to exemplary embodiments, R3 is hydrogen. Alternatively, R3 is alkyl (e.g., methyl or ethyl).
According to some of any of the embodiments described herein, A is a substituted or unsubstituted aryl (e.g., substituted or unsubstituted phenyl).
According to some of any of the embodiments described herein, A is a substituted aryl (e.g., substituted phenyl).
According to some of these embodiments, the aryl (e.g., phenyl) is substituted at least at the para position with respect to the pyrazole to which the phenyl is linked. Alternatively, or in addition, the aryl (e.g., phenyl) is substituted at the meta position with respect to the pyrazole ring to which the phenyl is linked.
Alternatively, A is a substituted or substituted heteroaryl, which, when substituted, include at least a substituent at the para position with respect to the pyrazole to which the heteroaryl is linked. Alternatively, or in addition, the heteroaryl is substituted at the meta position with respect to the pyrazole ring to which the heteroaryl is linked.
According to some of any of the embodiments described herein, exemplary A moieties include, but are not limited to:
Figure imgf000014_0001
Figure imgf000015_0001
According to some of any of the embodiments described herein, exemplary A moieties include, but are not limited to:
Figure imgf000016_0001
According to some of any of the embodiments described herein, Y is CR5, Z is CRe, R4, Rs, Rs and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
Compounds according to these embodiments can be collectively represented by Formula la:
Figure imgf000017_0001
Formula la wherein:
X, W, R3, R4, Rs, Rs and B are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
According to some of any of the embodiments of Formula la, n is 1 or 2 and at least one Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
According to exemplary embodiments of Formula la, n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl. According to some of these embodiments, one of the Ra substituents is pyrazole, which can be substituted or unsubstituted.
Compounds according to these embodiments can be collectively represented by Formula lb:
Figure imgf000018_0001
Formula lb wherein:
Ra, X, W, R3, R4, Rs, Re and B are as defined herein in any of the respective embodiments; m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1); k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
According to some of any of the embodiments of Formula lb, m is 0 or 1, and in some embodiments, m is 0.
According to some of any of the embodiments of Formula lb, k is 0.
According to some of any of the embodiments described herein for Formula I, la or lb, W is a nitrogen-containing moiety which is attached to X via the nitrogen atom, and which is or comprises a nitrogen-containing heteroalicyclic ring.
According to some of any of the embodiments described herein for Formula I, la or lb, W is a substituted nitrogen-containing heteroalicyclic ring, which is attached to the X via the nitrogen atom of the heteroalicyclic ring.
According to some of any of the embodiments described herein for Formula I, la or lb, W is a substituted nitrogen-containing heteroalicyclic ring. According to some of any of the embodiments described herein for Formula I, la or lb, W is a substituted nitrogen-containing heteroalicyclic ring, which is attached to the X via the nitrogen atom. In some of any of these embodiments, the nitrogen-containing heteroalicyclic ring is substituted by an alkyl, cycloalkyl or a heteroalicyclic. According to some of any of the embodiments described herein for Formula I, la or lb, W is or comprises a piperidine, a piperazine, or a morpholino group, which can be substituted, and which can be linked to X via the nitrogen atom.
According to some of any of the embodiments described herein for Formula I, la or lb, W is or comprises a piperidine or a piperazine, which can be substituted, and which can be linked to X via the nitrogen atom.
According to some of any of the embodiments described herein for Formula I, la or lb, W is a piperidine or a piperazine, which is linked to X via the nitrogen atom. According to some of these embodiments, the piperazine or piperidine is substituted.
According to some of any of these embodiments, the compounds can be collectively represented by Formula II:
Figure imgf000019_0001
Formula II wherein:
A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof;
V is N (such that W is or comprises a piperazine) or CRn (such that W is or comprises a piperidine);
R11 is hydrogen, halo, alkyl, cycloalkyl, and optionally other substituents as described herein; and
Rio is selected from the group consisting of hydrogen, a substituted or unsubstituted alkyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heteroalicyclic. Rio can alternatively be a hydrocarbon chain, which can be linear or branched, saturated or unsaturated (preferably saturated), optionally interrupted by one or more heteroatoms (e.g., oxygen or an amine as defined herein). In exemplary embodiments, Rio is or comprises an alkylene glycol as defined herein.
According to some of any of the embodiments described herein for Formula II, Y is CR5, Z is CRs, R4, Rs, Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
Compounds according to these embodiments can be collectively represented by Formula Ila:
Figure imgf000020_0001
wherein:
X, V, R3, R4, Rs, Rs, Rio and B are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
According to some of any of the embodiments of Formula Ila, n is 1 or 2 and at least one Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
According to exemplary embodiments of Formula Ila, n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl. According to some of these embodiments, one of the Ra substituents is pyrazole, which can be substituted or unsubstituted.
Compounds according to these embodiments can be collectively represented by Formula lib:
Figure imgf000021_0001
wherein:
Ra, X, V, R3, R4, Rs, Rs, Rio and B are as defined herein in any of the respective embodiments; m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1); k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
According to some of any of the embodiments of Formula lib, m is 0 or 1, and in some embodiments, m is 0.
According to some of any of the embodiments of Formula lib, k is 0.
According to some of any of the embodiments described herein for Formula II, Ila or lib,
V is CRn, and Rio is other than hydrogen. In some of these embodiments, Rn is hydrogen. According to these embodiments, W in Formula I, la or lb is a substituted 1,4-bi-piperidinyl.
According to some of any of the embodiments described herein for Formula II, Ila or lib,
V is CRn, and Rio is a heteroalicyclic. In some of these embodiments, Rn is hydrogen. In some of these embodiments, Rio is a nitrogen-containing heteroalicyclic and in some embodiments, Rio is piperidinyl, such that W in Formula I, la or lb is 1,4-bi-piperidinyl. The bi-piperidinyl can be substituted or unsubstituted and is preferably unsubstituted. Exemplary such compounds are referred to herein as TNK-108 and TNK-127 (see, for example, FIGs. 1 A and IB, respectively).
According to some of any of the embodiments described herein for Formula II, Ila or lib, Rio is a hydrocarbon chain, which can be linear or branched, saturated or unsaturated (preferably saturated), optionally interrupted by one or more heteroatoms (e.g., oxygen or an amine as defined herein).
According to some of these embodiments, Rio is a hydrocarbon chain, which can be linear or branched, saturated or unsaturated (preferably saturated), optionally interrupted by one or more heteroatoms (e.g., oxygen or an amine as defined herein).
In some of these embodiments, Rio is a branched, saturated hydrocarbon chain, interrupted by one or more heteroatoms. In some of these embodiments, Rio is a branched, saturated hydrocarbon chain, interrupted by one or more (e.g., two) oxygen atoms.
In some of any of the embodiments in which Rio is a hydrocarbon chain as described herein, V is CRn, and in some of these embodiments, Rn is hydrogen.
In some of any of the embodiments in which Rio is a hydrocarbon chain as described herein, V is N.
Moieties that are or comprise a branched saturated hydrocarbon chain interrupted by one or more heteroatoms, and which can be included as Rio in any of the respective Formulae can be collectively represented by Formula V:
-(L)C(R2i)[(CH2)j(CH2-O)q(R22)]2
Formula V wherein:
L is an alkylene chain of, for example, 1 to 6, or 1 to 4, carbon atoms, which is linked to V;
R21 is hydrogen or an alkyl, which can be substituted or unsubstituted, and is preferably an unsubstituted alkyl; the two moieties [(CH2)j(CH2-O)q(R22)] can be the same or different, and each of these moieties, j is independently an integer of from 0 to 6, or from 0 to 4, or from 1 to 4; q is independently an integer of from 0 to 6, or from 0 to 4, or from 0 to 2; and R22 is an alkyl (e.g., a branched alkyl), or cycloalkyl.
In some embodiments of Formula V, the two moieties [(CH2)j(CH2-O)q(R22)] are the same.
In some embodiments of Formula V, in at least one of the [(CH2)j(CH2-O)q(R22)] moieties, or in both, R22 is a branched alkyl, for example, t-butyl.
In some embodiments of Formula V, in at least one of the [(CH2)j(CH2-O)q(R22)] moieties, or in both, j is 0.
In some embodiments of Formula V, in at least one of the [(CH2)j(CH2-O)q(R22)] moieties, or in both, q is 1. In some embodiments of Formula V, R21 is methyl.
Exemplary branched saturated hydrocarbon chains of Formula V are shown in the compounds referred to as TNK-124 and TNK-128 (see, for example, FIG. 1 A).
According to some of any of the embodiments described herein for Formula I, la and lb, W is 1,4-bipiperidinyl.
In some embodiments, such compounds are collectively represented by Formula III:
Figure imgf000023_0001
Formula III wherein:
A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof.
According to some of any of the embodiments described herein for Formula III, Y is CR5, Z is CRs, R4, R5, Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
Compounds according to these embodiments can be collectively represented by Formula Illa:
Figure imgf000024_0001
wherein:
X, R3, R4, Rs, Rs, and B are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide. According to some of any of the embodiments of Formula Illa, n is 1 or 2 and at least one
Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
According to exemplary embodiments of Formula Illa, n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl. According to some of these embodiments, one of the Ra substituents is pyrazole, which can be substituted or unsubstituted. Compounds according to these embodiments can be collectively represented by Formula
Illb:
Figure imgf000025_0001
wherein:
Ra, X, R3, R4, Rs, Rs, and B are as defined herein in any of the respective embodiments; m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1); k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
According to some of any of the embodiments of Formula Illb, m is 0 or 1, and in some embodiments, m is 0.
According to some of any of the embodiments of Formula Illb, k is 0.
According to some of any of the embodiments described herein for Formula I, la and lb, W is a moiety of Formula V as described herein in any of the respective embodiments.
In some of these embodiments, the compounds are collectively represented by Formula
IV:
Figure imgf000026_0001
Formula IV wherein:
A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof; V is as defined herein for Formula II, Ila or lib, and q, j, L, R21 and R22 are as defined herein for Formula V in any of the respective embodiments and any combination thereof.
According to some of any of the embodiments described herein for Formula IV, Y is CR5, Z is CRe, R4, R5, Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
Compounds according to these embodiments can be collectively represented by Formula
IVa:
Figure imgf000027_0001
wherein:
X, V, B, R3, R4, Rs, Re, q, j, L, R21 and R22 are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide. According to some of any of the embodiments of Formula IVa, n is 1 or 2 and at least one
Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
According to exemplary embodiments of Formula IVa, n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl. According to some of these embodiments, one of the Ra substituents is pyrazole, which can be substituted or unsubstituted. Compounds according to these embodiments can be collectively represented by Formula
IVb:
Figure imgf000028_0001
wherein:
Ra, X, B, V, R3, R4, Rs, Re, q, j, L, R21 and R22 are as defined herein in any of the respective embodiments; m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1); k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
According to some of any of the embodiments of Formula IVb, m is 0 or 1, and in some embodiments, m is 0.
According to some of any of the embodiments of Formula IVb, k is 0.
According to some of any of the embodiments of Formula IV, IVa or IVb, V is N.
According to some of any of the embodiments described herein for Formula I, la or lb, W is an aminoalkyl, which is attached to X via said amino. In some of these embodiments, the aminoalkyl is substituted, e.g., the alkyl is substituted by one or more substituents. In some of these embodiments, the one or more substituents comprise one or more heteroatoms such as N and/or O and/or S. For example, the aminoalkyl can be substituted by one or more of amine, alkoxy, thioalkoxy, heteroalicyclic, heteroaryl, carboxy, and other heteroatom-containing substituents. Such compounds can be collectively represented by Formula VI:
Figure imgf000029_0001
Formula VI wherein:
A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof; and R13 and R14 are each independently selected from hydrogen and alkyl, wherein preferably at least one of R13 and R14 is an alkyl, and further preferably, at least one such alkyl is substituted as described herein. When one or both of R13 and R14 is an alkyl, the alkyl is independently of 1 to 10, or from
1 to 8, or from 1 to 6, or from 2 to 6, carbon atoms in length, including any intermediate values and subranges therebetween.
According to some of any of the embodiments described herein for Formula VI, Y is CR5, Z is CRe, R4, R5, Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
Compounds according to these embodiments can be collectively represented by Formula Via:
Figure imgf000030_0001
Formula Via wherein:
X, V, B, R3, R4, Rs, Rs, R12, RB and R14 are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide. According to some of any of the embodiments of Formula Via, n is 1 or 2 and at least one
Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
According to exemplary embodiments of Formula Via, n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl. According to some of these embodiments, one of the Ra substituents is pyrazole, which can be substituted or unsubstituted. Compounds according to these embodiments can be collectively represented by Formula
Vlb:
Figure imgf000031_0001
wherein:
Ra, X, B, V, R3, R4, Rs, Re, R12, R13 and R14 are as defined herein in any of the respective embodiments; m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1); k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
According to some of any of the embodiments of Formula VIb, m is 0 or 1, and in some embodiments, m is 0.
According to some of any of the embodiments of Formula VIb, k is 0.
According to some of any of the embodiments of Formula VI, Via or VIb, R12 is hydrogen.
According to exemplary embodiments of Formula VI, Via or VIb, W is an aminoalkyl which is substituted by a nitrogen-containing heteroalicyclic (e.g., a piperidinyl or piperazinyl group).
In some of these embodiments, the compounds are collectively represented by Formula
VII:
Figure imgf000032_0001
wherein:
A, B, X, Y, Z, Ri, R2, R3, R4, R5 and Re are as defined herein for Formula I, la or lb, in any of the respective embodiments and any combination thereof; V is as defined herein for Formula II, Ila or lib, R12 is as defined herein for R’ and R” and K is an alkyl, as defined herein, e.g., an alkylene chain, preferably of from 1 to 10, or from 1 to 6, or from 1 to 4, carbon atoms in length.
According to some of any of the embodiments described herein for Formula VII, Y is CR5, Z is CRe, R4, R5, Re and B are as defined herein in any of the respective embodiments (e.g., each independently selected from halo, alkyl (e.g., methyl or ethyl), or haloalkyl (e.g., trihaloalkyl such as CF3)), and A is substituted or unsubstituted phenyl.
Compounds according to these embodiments can be collectively represented by Formula
Vila:
Figure imgf000033_0001
Formula Vila wherein:
X, V, B, R3, R4, R5, Re, R12 and K are as defined herein in any of the respective embodiments; n represents the number of substituents on the phenyl which are other than hydrogen and can be 0, 1, 2, 3, 4 or 5 (preferably 0, 1 or 2); and
Ra is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
According to some of any of the embodiments of Formula Vila, n is 1 or 2 and at least one Ra substituent is at the para position with respect to the pyrazole to which the phenyl is linked.
According to exemplary embodiments of Formula Vila, n is 1 or 2 and one of the Ra substituents is a (substituted or unsubstituted) heteroaryl. According to some of these embodiments, one of the Ra substituents is pyrazole, which can be substituted or unsubstituted.
Compounds according to these embodiments can be collectively represented by Formula Vllb:
Figure imgf000034_0001
Formula Vllb wherein:
Ra, X, B, V, R3, R4, Rs, Re, R12 and K are as defined herein in any of the respective embodiments; m represents the number of substituents on the phenyl (Ra) which are other than hydrogen and can be 0, 1, 2, 3 or 4 (preferably 0 or 1); k represents the number of substituents on the terminal pyrazole ring (Rb) which are other than hydrogen, and can be 0, 1, 2 or 3 (preferably 0 or 1); and
Rb is selected from alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, aryloxy, cyano, aryl, heteroaryl, heteroalicyclic, cycloalkyl, sulfonyl, sulfinyl, and sulfonamide.
According to some of any of the embodiments of Formula Vllb, m is 0 or 1, and in some embodiments, m is 0.
According to some of any of the embodiments of Formula Vllb, k is 0.
According to some of any of the embodiments of Formula VII, Vila or Vllb, R12 is hydrogen.
According to some of any of the embodiments of Formula VII, Vila or Vllb, K is an unsubstituted alkyl.
According to some of any of the embodiments of Formula VII, Vila or Vllb, V is CRn. In some of these embodiments Rn is hydrogen.
According to some of any of the embodiments described herein for Formula I, la or lb, W is or comprises a nitrogen-containing heteroalicyclic, for example, a piperidine or a piperazine. According to some of any of the embodiments described herein for Formula I, la or lb, W is a piperidinyl which is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom, such as described, for example, by Formula V or in Formula IV, IVa or IVb.
According to some of any of the embodiments described herein for Formula I, la or lb, W is or comprises a substituted 1,4-piperazinyl.
In some of these embodiments, the piperazinyl is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom, such as described, for example, by Formula V or in Formula IV, IVa or IVb.
According to some of any of the embodiments described herein for Formula I, la, lb, II, Ila, lib, III, Illa, IIIB, IV, IVa, IVb, VI, Via, VIb, VII, Vila and Vllb, X is -O-C(=O). In some embodiments, such compounds can act as prodrugs under physiological conditions that promote cleavage of the -O-C(=O)- so as to generate a -OH group at the respective position. Exemplary physiological conditions include presence of esterases and/or acidic environment. Such compounds are also referred to herein as biocleavable compounds or biocleavable prodrugs.
Table A below presents exemplary compounds as disclosed and referenced in WO 2019/156439 for which a respective compound represented by Formula I, II, III, IV, VI or VII wherein X is a -O-C(=O)- can be generated as a biocleavable prodrug according to the present embodiments. For each of these compounds, the hydroxy on the phenol group that is attached to the pyrazole via an amine group is replaced by a -0-C(=0)-W group as described herein in any of the respective embodiments, as shown below, with the added circles showing the modification made to the compounds disclosed in WO 2019/156439, according to the present embodiments.
It is to be noted that, as explained herein, these modifications are not merely of converting the respective hydroxy group to a respective carbamate, since such a conversion is not feasible. Rather, in order to practice such a modification, a dedicated synthetic pathway had to be designed.
Figure imgf000036_0001
Compounds as disclosed in WO Compounds of Formula I 2019/156439 wherein X is -O-C(=O)-
Table A
Figure imgf000036_0002
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
(Table A; Cont.)
It is to be noted that whenever compounds of Formula I, II, III, IV or VI feature a phenyl group as variable A, as shown for example in Formula la, Ila, Illa, Iva, Via or Vila, respectively, and the phenyl is substituted by hydroxy, as shown for example, for compounds 39, 40, 49, 61, 62, 81, 82, 85, 87, 89, 92, 94, 108, 124, 126, 150, 160, 162, 168 and 275 in Table A, this hydroxy group can either also be modified to a -0-C(=0)-W group or remain intact. Preferably, however, compounds of such embodiments are such that the substituent on the respective phenyl is other than hydroxy (e.g., A in Formula I, II, III, IV, VI or VII is a phenyl that if substituted, is not substituted by hydroxy; or Ra in Formula la, Ila, Illa, Iva, Via or Vila is other than hydroxy).
In some of any of the embodiments described herein for Formula I, la, lb, II, Ila or lib, X is -O-C(=O)- and W is 1,4-bipiperidinyl.
In some of any of the embodiments described herein for Formula I, la, lb, IV, IVa, or IVb, X is -O-C(=O)- and W is 1,4-piperazinyl substituted by a linear hydrocarbon chain interrupted by at least one O atom such as described in Formula V.
According to some of any of the embodiments described herein for Formula I, la, lb, II, Ila, lib, III, Illa, IIIB, IV, IVa, IVb, VI, Via, VIb, VII, Vila and Vllb, X is -NH-C(=O)-. In some embodiments, such compounds can be considered as non-cleavable modifications, since under most prevalence physiological conditions cleavage of the -NH-C(=O)- does not occur. Such compounds are also referred to herein as non-biocleavable compounds or non-biocleavable prodrugs.
Table B below presents exemplary compounds as disclosed and referenced in WO 2019/156439 for which a respective compound represented by Formula I, II, III, IV, VI or VII wherein X is a -NH-C(=O)- can be generated as a non-biocleavable compound (therapeutically active agent) according to the present embodiments. For each of these compounds, an amine or an amide group within a substituent on the phenol group that is attached to the pyrazole via an amine group is replaced by a -NH-C(=0)-W group as described herein in any of the respective embodiments, as shown below, with the added circles showing the modification made to the compounds disclosed in WO 2019/156439, according to the present embodiments.
Figure imgf000052_0001
Compounds as disclosed in WO Compounds of Formula I 2019/156439 wherein X is -NR’-C(=O)- wherein R’ and R” are as defined herein.
It is to be noted that, as explained herein, these modifications are not merely of converting the respective amine or amide group to a respective ureido group, since such a conversion is not feasible. Rather, in order to practice such a modification, a dedicated synthetic pathway had to be designed.
Table B
Figure imgf000052_0002
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
It is to be noted that whenever compounds of Formula I, II, III, IV, VI or VII feature a phenyl group as variable A, as shown for example in Formula la, Ila, Illa, Iva, Via or Vila, respectively, and the phenyl is substituted by hydroxy, as shown for example, for compounds 152, 169, 266, 270, 142, 143, 145, 147, 149, 151, 154, 166 and 179 in Table B, this hydroxy group can either also be modified to a -0-C(=0)-W group or remain intact. Preferably, however, compounds of such embodiments are such that the substituent on the respective phenyl is other than hydroxy (e.g., A in Formula I, II, III, IV, VI or VII is a phenyl that if substituted, is not substituted by hydroxy; or Ra in Formula la, Ila, Illa, Iva, Via or Vila is other than hydroxy).
In some of any of the embodiments described herein for Formula I, la, lb, II, Ila or lib, X is -NH-C(=O)- and W is 1,4-bipiperidinyl.
In some of any of the embodiments described herein for Formula I, la, lb, IV, IVa or IVb, X is -NH-C(=O)- and W is 1,4-piperazinyl substituted by a linear hydrocarbon chain interrupted by at least one O atom such as described in Formula V.
In some of any of the embodiments described herein for Formula I, la, and lb, X is -NH- C(=O)- and W is an aminoalkyl, optionally substituted by piperidinyl or piperazinyl. According to some of any of the embodiments described herein for Formula I, la or lb, X is -NH-C(=O)- and W is 1,4-bipiperidinyl.
According to some of any of the embodiments described herein for Formula I, la or lb, X is -NH-C(=O)- and W is an aminoalkyl (e.g., aminopropyl) substituted by a piperidinyl.
Process:
According to an aspect of some embodiments of the present invention there is provided a process of preparing compounds of Formula I, as described herein in any of the respective embodiments.
According to some of these embodiments, the process as described herein is a large scale process or is at least a scalable process, that is, can be scaled up to be a large scale process.
Herein, by “large scale” in the context of the process as described herein, it is meant a production process in which at least one of the compounds, whether a starting material, an intermediate and/or the final product, is used or prepared in an amount of at least one mole, or at least 2 moles, or at least 3 moles, or at least 5 moles, for example, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50 moles, or even more, including any intermediate values therebetween.
A “large scale” process is defined herein, alternatively or in addition, by the weight of the one or more of the starting materials, the intermediates and/or the final product, such that one or more these materials is/are used or prepared in an amount of at least 1 Kg, or at least 2 Kg, or at least 3 Kg, or at least 5 Kg, for example, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 Kg, or even more, including any intermediate values therebetween.
A “large scale” process is defined, alternatively or in addition, by the volumes of the reactors used in the process, as comprising at least one production step that is performed in a reactor having a volume of at least 10 liters, or at least 15 liters, or at least 20 liters, or at least 30 liters, or at least 40, 50, 60, 70, 80, 90, 100 liters or even more, including any intermediate values therebetween.
According to some of these embodiments, the process as described herein is a gram-scale process.
Herein, by “gram-scale” in the context of the process as described herein, it is meant a production process in which at least one of the compounds, whether a starting material, an intermediate and/or the final product, is used or prepared in an amount of at least one gram.
The process, according to the present embodiments, comprises reacting a compound of Formula Xa:
Figure imgf000058_0001
wherein Q is hydroxy, in case X is -O-C(=O)-, or amine, in case X is -NH-C(=O)-, and Li is a first reactive group; with a compound of Formula Xb:
W-L2
Formula Xb, wherein W is as described herein in any of the respective embodiments and W-L2 comprise a second reactive group, under conditions that generate said X upon coupling said W-L2 with said Q, to thereby generate a compound of Formula Xc:
Figure imgf000059_0001
and reacting the compound of Formula Xc with a compound of Formula Xd:
Figure imgf000059_0002
Formula Xd wherein L3 is a third reactive group that generates, upon reacting with Li, said -N(R.2)-, under conditions that generate said -N(R.2)- upon coupling said Li and L3, to thereby generate the compound of Formula I.
Herein, by “reactive group” it is meant a chemical group that is capable of undergoing a chemical reaction that typically leads to a bond formation. The bond, according to the present invention, is preferably a covalent bond. Chemical reactions that lead to a bond formation include, for example, nucleophilic and electrophilic substitutions, nucleophilic and electrophilic addition reactions, addition-elimination reactions, cycloaddition reactions, rearrangement reactions and any other known organic reactions that involve a bond formation.
According to the present embodiments, the Q and L2 reactive groups are such that can undergo a chemical reaction that leads to formation of an X moiety, as described herein. W-L2 can be such that comprises a respective reactive group that is attached to W and forms, when coupled with Q, the moiety X.
Alternatively, when W is a substituted amine or a nitrogen-containing heterocyclic, L2 can be hydrogen, and the second reactive group is an amine (that forms a part of W, such that W-L2 comprise the second reactive group).
For example, when X is -O-C(=O)- and W is a substituted amine or a nitrogen-containing heteroalicyclic, the first and second reactive groups can be hydroxy and amine, respectively, while the amine forms a part of the W group, and the conditions under which the reaction is performed are such that promote a formation of the respective X moiety (as a part of a carbamate group) upon reacting said hydroxy and said amine.
Conditions for reacting an amine and a hydroxy group to provide a carbamate group (when X is -O-C(=O)- and W is a nitrogen-containing moiety) include, for example, suitable coupling agents, optionally combined with oxidizing agents and/or other agents that promote formation of carbamate by reacting an amine (reactive group of W-L2) and phenol (which comprises the hydroxy group). These include, for example, using carbonylimidazolide (CDI) in water, as described, for example, in Padiya et al., Org. Lett., 2012, 14, 2814-2817. Exemplary such agents and conditions are described in the Examples section that follows.
Other exemplary conditions include, for example, using bis(trichloromethyl) carbonate (BTC), under conditions such as described, for example, in Varjosaari et al., Synthesis, 2016, 48, 43-47.
For example, when X is -NH-C(=O)- and W is a substituted amine or a nitrogen-containing heteroalicyclic, the first and second reactive groups can be each amine, while one amine forms a part of the W group and the other forms a part of a respective aniline, and the conditions under which the reaction is performed are such that promote a formation of the respective X moiety (as a part of an ureido group), upon reacting said amines.
Conditions for reacting amines to provide an ureido/urea group (when X is -NH-C(=O)- and W is a nitrogen-containing moiety) include, for example, suitable coupling agents, optionally combined with other agents or conditions that promote formation of ureido moiety by reacting amine (reactive group of W-L2) and aniline (which comprises the other amine). These include, for example, using carbonylimidazolide (CDI) in water, as described, for example, in Padiya et al., Org. Lett., 2012, 14, 2814-2817.
Exemplary such agents and conditions are described in the Examples section that follows.
The first reactive group Li and the third reactive group L3 are selected such that upon reacting these groups an amine group is generated. In some embodiments, the first and third reactive groups are such that participate in a nucleophilic reaction, for example, the third reactive group is amine and the first reactive group is a leaving group, and the reaction conditions are such that promote a nucleophilic reaction. Exemplary such agents are described in the Examples section that follows.
According to some of any of these embodiments, the process further comprises preparing a compound of Formula Xd. In some embodiments, preparing a compound of Formula Xd comprises reacting a compound of Formula Xf with hydrazine.
A-C(=O)-CH2CN
Formula Xf wherein A is as described herein in any of the respective embodiments and any combination thereof.
In any of the reactions performed in the described process, functional groups of the reacting compounds, which do not participate in a respective reaction, can be protected and then deprotected as required or desired, and the process as described herein can further comprises such protection and de-protection reactions.
Uses/ Applications:
The compounds disclosed herein are usable as modulators of TNIK activity, typically as inhibitors of TNIK activity, and/or as capable of generating, preferably under physiological conditions, compounds that act as modulators of TNIK activity, typically as inhibitors of TNIK activity. The compounds disclosed herein are usable as therapeutically active agents for treating medical conditions that are associated with abnormal TNIK activity and/or in which modulating (e.g., inhibiting) TNIK activity is beneficial.
According to an aspect of some embodiments of the present invention there is provided a method of modulating (e.g., inhibiting) a TNIK activity, which comprises contacting cells that exhibit abnormal (e.g., hyperactive) TNIK activity with a compound as described herein in any of the respective embodiments and any combination thereof.
According to some embodiments of this aspect of the present invention, the compound has Formula I, la, lb, II, Ila, lib, III, Illa, Illb, IV, IVa, IVb, VI, Via, VIb, VII, Vila or Vllb, wherein X is -NH-C(=O)-.
As exemplified herein (see, for example, Table 1 in the Examples section that follows), such compounds exhibit modulation (e.g., inhibition) of TNIK activity when used per se.
The contacting, according to these embodiments, can be effected in vitro, ex-vivo, or in vivo. According to some embodiments of this aspect of the present invention, the compound has Formula I, la, lb, II, Ila, lib, III, Illa, Illb, IV, IVa, IVb, VI, Via, VIb, VII, Vila or Vllb, wherein X is -O-C(=O)-, and the contacting is effected in vivo. In some of these embodiments, the cells are tumor cells or cells that are characterized by abnormal proliferation.
According to an aspect of some embodiments of the present invention there is provided a method of modulating (e.g., inhibiting) a TNIK activity in a subject in need thereof, which comprises administering to the subject a compound as described herein in any of the respective embodiments and any combination thereof.
According to an aspect of some embodiments of the present invention there is provided a compound as described herein in any of the respective embodiments and any combination thereof for use in modulating (e.g., inhibiting) a TNIK activity in a subject in need thereof.
According to an aspect of some embodiments of the present invention there is provided a compound as described herein in any of the respective embodiments and any combination thereof for use in the preparation of medicament for modulating (e.g., inhibiting) a TNIK activity in a subject in need thereof.
The methods and uses described herein are usable for modulating a TNIK activity is cells that exhibit an abnormal TNIK activity, and in subjects hosting such cells.
The methods and uses described herein are usable in treating medical conditions that are associated with abnormal TNIK activity. In some embodiments, the medical conditions are associated with TNIK hyperactivity. In some embodiments, the medical conditions are mediated by TNIK. In some of these embodiments, the medical conditions are associated with signaling pathways mediated by TNIK.
Medical conditions that are treatable according to some embodiments of the present invention include proliferative diseases and disorders, such as cancer.
Exemplary cancer types include, but are not limited to, colorectal cancer, breast cancer, brain tumor, gastric cancer, liver cancer, ovarian cancer, lung cancer, gastrointestinal cancer, leukemia, and melanoma.
According to an aspect of some embodiments of the present invention there is provided a method of treating a medical condition associated with an abnormal TNIK activity, the method comprising administering to a subject in need thereof a therapeutically effective amount of a compound as described herein in any of the respective embodiments and any combination thereof.
According to an aspect of some embodiments of the present invention there is provided a use of compound as described herein in any of the respective embodiments and any combination thereof in the treatment of a medical condition associated with an abnormal TNIK activity. According to an aspect of some embodiments of the present invention there is provided a use of compound as described herein in any of the respective embodiments and any combination thereof in the manufacture of a medicament for the treatment of a medical condition associated with an abnormal TNIK activity.
According to an aspect of some embodiments of the present invention there is provided a method of treating a cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound as described herein in any of the respective embodiments and any combination thereof.
According to an aspect of some embodiments of the present invention there is provided a use of compound as described herein in any of the respective embodiments and any combination thereof in the treatment of cancer.
According to an aspect of some embodiments of the present invention there is provided a use of compound as described herein in any of the respective embodiments and any combination thereof in the manufacture of a medicament for the treatment of cancer.
According to an aspect of some embodiments of the present invention there is provided a method of treating a proliferative disease or disorder in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound as described herein in any of the respective embodiments and any combination thereof.
According to an aspect of some embodiments of the present invention there is provided a use of compound as described herein in any of the respective embodiments and any combination thereof in the treatment of a proliferative disease or disorder.
According to an aspect of some embodiments of the present invention there is provided a use of compound as described herein in any of the respective embodiments and any combination thereof in the manufacture of a medicament for the treatment of a proliferative disease or disorder.
As used herein, the terms “cancer” and “tumor” are interchangeably used. The terms refer to a malignant growth and/or tumor caused by abnormal and uncontrolled cell proliferation (cell division). The term “cancer” encompasses tumor metastases.
The term “cancer cell” describes the cells forming the malignant growth or tumor.
Non-limiting examples of cancers and/or tumor metastases which can be treated according to some embodiments of any of the embodiments described herein relating to cancer (including any of the aspects described herein) include any solid or non-solid cancer and/or tumor metastasis, including, but not limiting to, tumors of the gastrointestinal tract (e.g., colon carcinoma, rectal carcinoma, colorectal carcinoma, colorectal cancer, colorectal adenoma, hereditary nonpolyposis type 1, hereditary nonpolyposis type 2, hereditary nonpolyposis type 3, hereditary nonpolyposis type 6; colorectal cancer, hereditary nonpolyposis type 7, small and/or large bowel carcinoma, esophageal carcinoma, tylosis with esophageal cancer, stomach carcinoma, pancreatic carcinoma, pancreatic endocrine tumors), endometrial carcinoma, dermatofibrosarcoma protuberans, gallbladder carcinoma, biliary tract tumors, prostate cancer, prostate adenocarcinoma, renal cancer (e.g., Wilms’ tumor type 2 or type 1), liver cancer (e.g., hepatoblastoma, hepatocellular carcinoma, hepatocellular cancer), bladder cancer, embryonal rhabdomyosarcoma, germ cell tumor, trophoblastic tumor, testicular germ cells tumor, immature teratoma of ovary, uterine, epithelial ovarian, sacrococcygeal tumor, choriocarcinoma, placental site trophoblastic tumor, epithelial adult tumor, ovarian carcinoma, serous ovarian cancer, ovarian sex cord tumors, cervical carcinoma, uterine cervix carcinoma, small-cell and non-small cell lung carcinoma, nasopharyngeal, breast carcinoma (e.g., ductal breast cancer, invasive intraductal breast cancer, sporadic breast cancer, susceptibility to breast cancer, type 4 breast cancer, breast cancer- 1, breast cancer-3, breast-ovarian cancer), squamous cell carcinoma (e.g., in head and neck), neurogenic tumor, astrocytoma, ganglioblastoma, neuroblastoma, lymphomas (e.g., Hodgkin's disease, nonHodgkin's lymphoma, B-cell lymphoma, Diffuse large B-cell lymphoma (DLBCL), Burkitt lymphoma, cutaneous T-cell lymphoma, histiocytic lymphoma, lymphoblastic lymphoma, T-cell lymphoma, thymic lymphoma), gliomas, adenocarcinoma, adrenal tumor, hereditary adrenocortical carcinoma, brain malignancy (tumor), various other carcinomas (e.g., bronchogenic large cell, ductal, Ehrlich-Lettre ascites, epidermoid, large cell, Lewis lung, medullary, mucoepidermoid, oat cell, small cell, spindle cell, spinocellular, transitional cell, undifferentiated, carcinosarcoma, choriocarcinoma, cystadenocarcinoma), ependimoblastoma, epithelioma, erythroleukemia (e.g., Friend, lymphoblast), fibrosarcoma, giant cell tumor, glial tumor, glioblastoma (e.g., multiforme, astrocytoma), glioma hepatoma, heterohybridoma, heteromyeloma, histiocytoma, hybridoma (e.g., B-cell), hypernephroma, insulinoma, islet tumor, keratoma, leiomyoblastoma, leiomyosarcoma, leukemia (e.g., acute lymphatic leukemia, acute lymphoblastic leukemia, acute lymphoblastic pre-B cell leukemia, acute lymphoblastic T cell leukemia, acute megakaryoblastic leukemia, monocytic leukemia, acute myelogenous leukemia, acute myeloid leukemia, acute myeloid leukemia with eosinophilia, B-cell leukemia, basophilic leukemia, chronic myeloid leukemia, chronic B-cell leukemia, eosinophilic leukemia, Friend leukemia, granulocytic or myelocytic leukemia, hairy cell leukemia, lymphocytic leukemia, megakaryoblastic leukemia, monocytic leukemia, monocytic-macrophage leukemia, myeloblastic leukemia, myeloid leukemia, myelomonocytic leukemia, plasma cell leukemia, pre-B cell leukemia, promyelocytic leukemia, subacute leukemia, T-cell leukemia, lymphoid neoplasm, predisposition to myeloid malignancy, acute nonlymphocytic leukemia), lymphosarcoma, melanoma, mammary tumor, mastocytoma, medulloblastoma, mesothelioma, metastatic tumor, monocyte tumor, multiple myeloma, myelodysplastic syndrome, myeloma, nephroblastoma, nervous tissue glial tumor, nervous tissue neuronal tumor, neurinoma, neuroblastoma, oligodendroglioma, osteochondroma, osteomyeloma, osteosarcoma (e.g., Ewing's), papilloma, transitional cell, pheochromocytoma, pituitary tumor (invasive), plasmacytoma, retinoblastoma, rhabdomyosarcoma, sarcoma (e.g., Ewing's, histiocytic cell, Jensen, osteogenic, reticulum cell), schwannoma, subcutaneous tumor, teratocarcinoma (e.g., pluripotent), teratoma, testicular tumor, thymoma and trichoepithelioma, gastric cancer, fibrosarcoma, glioblastoma multiforme, multiple glomus tumors, Li-Fraumeni syndrome, liposarcoma, lynch cancer family syndrome II, male germ cell tumor, mast cell leukemia, medullary thyroid, multiple meningioma, endocrine neoplasia myxosarcoma, paraganglioma, familial nonchromaffin, pilomatricoma, papillary, familial and sporadic, rhabdoid predisposition syndrome, familial, rhabdoid tumors, soft tissue sarcoma, and Turcot syndrome with glioblastoma.
According to some of any of the embodiments described herein the term “subject” refers to a mammal (e.g., human), for example, one who has been diagnosed with a condition described herein (e.g., cancer). Subjects may be of either gender and at any stage of development.
As described and exemplified herein, the compounds of the present embodiments are characterized by enhanced solubility, high bioavailability, plasma stability, and high AUC values. These features render these compounds suitable for oral administration, and further allow administering the compounds with relatively high intervals.
According to some embodiments of any of the methods and uses described herein the compounds are administered once, twice or trice daily. According to some embodiments of any of the methods and uses described herein the compounds are administered once, twice or trice weekly, or, for example, once in two days or once in three days. Compounds that feature high Tl/2 values are suitable for use in such a regimen.
The compounds of the present disclosure can otherwise be administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. An effective dosage is typically in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 0.01 to about 50 mg/kg/day, in single or divided doses. Depending on age, species and disease or condition being treated, dosage levels below the lower limit of this range may be suitable.
Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intracardiac, e.g., into the right or left ventricular cavity, into the common coronary artery, intravenous, intraperitoneal, intranasal, or intraocular injections.
According to some embodiments of any of the methods and uses described herein, the compounds are administered orally (PO).
According to some embodiments of any of the methods and uses described herein, the compounds are administered intravenously (IV).
Pharmaceutical compositions:
The compounds described herein according to any of the aspects of embodiments of the invention described herein can be utilized (e.g., administered to a subject) per se or in a pharmaceutical composition where the compound is mixed with suitable carriers or excipients.
As used herein a "pharmaceutical composition" refers to a preparation of one or a compound according to any of the embodiments described herein with other chemical components such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
Hereinafter, the phrases "physiologically acceptable carrier" and "pharmaceutically acceptable carrier" which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound. An adjuvant is included under these phrases.
Herein the term "excipient" refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
When utilized per se or in a pharmaceutically acceptable composition, the compound per se (that is, not including, weight of carriers or excipients co-formulated with the compound, as described herein) is optionally at least 80 % pure (by dry weight), optionally at least 90 % pure (by dry weight), at least 95 % pure (by dry weight), at least 98 % pure (by dry weight), and optionally at least 99 % pure (by dry weight). Purity may be enhanced, e.g., by removing impurities associated with synthesis of the compound or isolation of the compound from a natural source, by any suitable technique known in the art. Techniques for formulation and administration of drugs may be found in “Remington’s Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference.
Alternatively, one may administer the pharmaceutical composition in a local rather than systemic manner, for example, via injection of the pharmaceutical composition directly into a tissue region of a patient. The term “tissue” refers to part of an organism consisting of cells designed to perform a function or functions. Examples include, but are not limited to, brain tissue, retina, skin tissue, hepatic tissue, pancreatic tissue, breast tissue, bone, cartilage, connective tissue, blood tissue, muscle tissue, cardiac tissue brain tissue, vascular tissue, renal tissue, pulmonary tissue, gonadal tissue, hematopoietic tissue.
Pharmaceutical compositions of some embodiments of the invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
Pharmaceutical compositions for use in accordance with some embodiments of the invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
For injection, the active ingredients of the pharmaceutical composition may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank’s solution, Ringer’s solution, or physiological salt buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
For oral administration, the pharmaceutical composition can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the pharmaceutical composition to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient. Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
Pharmaceutical compositions which can be used orally include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
In some embodiments, the pharmaceutical composition is formulated for oral administration.
In exemplary such embodiments, the carrier is an aqueous carrier which comprises a surfactant, preferably a surfactant, and one or both of a solubilizer (e.g., cyclodextrin or Transcutol®) and a water-soluble or water-miscible polymer (e.g., polyalkylene glycol). In exemplary embodiments, the surfactant is a non-ionic surfactant, for example a Vitamin E-based surfactant such as, for example, VE-TPGS or a surfactant such as Gelucire® 44/14 (lauroyl polyoxyl-32 glycerides). In exemplary embodiments, the surfactant is of the Tween® family, for example, Tween®80.
An exemplary pharmaceutical composition, which is suitable for oral administration of compounds such as the exemplified TNK-108, comprises 20 % PEG400, 10 % VE-TPGS and 70 % of a 5 % aqueous solution of HP-P-CD.
An exemplary pharmaceutical composition, which is suitable for oral administration of compounds such as the exemplified TNK-108, comprises 10 % Transcutol®, 5 % Tween®80 and 85 % a solution of 10 % Gelucire® 44/14 in Water.
An exemplary pharmaceutical composition, which is suitable for oral administration of compounds such as the exemplified TNK-108, comprises 20 % PEG400, 5 % Tween®80 and 75 % a solution of 5 % HP-P-CD in Water.
For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
For administration by nasal inhalation, the active ingredients for use according to some embodiments of the invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in a dispenser may be formulated containing a powder mix of the active compound and a suitable powder base such as lactose or starch.
The pharmaceutical composition described herein may be formulated for parenteral administration, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative. The compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
Pharmaceutical compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the active ingredients to allow for the preparation of highly concentrated solutions.
Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water based solution, before use.
The pharmaceutical composition of some embodiments of the invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
According to some of any of the embodiments described herein, the pharmaceutical composition is formulated for oral administration.
Pharmaceutical compositions suitable for use in context of some embodiments of the invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of the active ingredient(s) effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., cancer or metastatic cancer) or prolong the survival of the subject being treated.
Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. For any preparation used in the methods of the invention, the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays. For example, a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.
Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals. The data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see, e.g., Fingl et al. (1975), in "The Pharmacological Basis of Therapeutics", Ch. 1 p. l).
Dosage amount and interval may be adjusted individually to provide protein (e.g., TNIK) inhibitory levels of the active ingredient are sufficient to induce or suppress the biological effect (minimal effective concentration, MEC). The MEC will vary for each preparation, but can be estimated from in vitro data, e.g., based on results on TNIK inhibition assay described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.
In some embodiments of any of the embodiments described herein, an effective amount is at least 100 % of the IC50 of the compound towards a TNIK inhibition. In some embodiments, an effective amount is at least 200 % of the IC50 of the compound towards TNIK inhibition. In some embodiments, an effective amount is at least 300 % of the IC50 of the compound towards TNIK inhibition. In some embodiments, an effective amount is at least 500 % of the IC50 of the compound towards TNIK inhibition. In some embodiments, an effective amount is at least 1000 % of the IC50 of the compound towards TNIK inhibition.
Depending on the severity and responsiveness of the condition to be treated, dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
The amount of a composition to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
Compositions of some embodiments of the invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert. Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as is further detailed herein.
It will be appreciated that the compounds described herein can be provided alone or in combination with other active ingredients, which are well known in the art for alleviating the medical condition.
The compound may be administered with an additional anti-cancer agent, as described herein in any of the respective embodiments, either together in a co-formulation (e.g., in the same pharmaceutical composition) or in separate formulations.
According to a specific embodiment, the treatment of cancer (and other hyperproliferative disorders) is effected in combination with an additional anti-cancer as described herein in any of the respective embodiments.
A pharmaceutical composition as described herein can further comprise any of the additional agents as described herein, or alternatively, be identified for use in combination with any of the additional agents as described herein.
According to another aspect described herein, there is provided a kit for the treatment of a condition (e.g., treatment of cancer) described herein, the kit comprising a packaging material packaging the compound described herein.
In some of these embodiments, the kit further comprises an additional agent as described herein in any of the respective embodiments, and the two agents are packaged individually within the kit.
In some of these embodiments, the kit further comprises instructions to use the compound in combination with an additional agent (e.g., an additional anti-cancer cancer) as described herein in any of the respective embodiments.
In some of any of the respective embodiments described herein, the one or more additional agent(s) is an anti-cancer drug. For example, the anti-cancer drug is EGFR kinase inhibitors, MEK inhibitors, VEGFR inhibitors, anti-VEGFR2 antibodies, KDR antibodies, AKT inhibitors, PDK-1 inhibitors, PI3K inhibitors, c-kit/Kdr tyrosine kinase inhibitors, Bcr-Abl tyrosine kinase inhibitors, VEGFR2 inhibitors, PDGFR-beta inhibitors, KIT inhibitors, Flt3 tyrosine kinase inhibitors, PDGF receptor family inhibitors, Flt3 tyrosine kinase inhibitors, RET tyrosine kinase receptor family inhibitor, VEGF-3 receptor antagonists, Raf protein kinase family inhibitor, angiogenesis inhibitors, Erb2 inhibitors, mTOR inhibitors, IGF-1R antibodies, NFkB inhibitors, proteasome inhibitors, chemotherapy agents, or glucose reduction agents.
Exemplary agents include, but are not limited to, alkylating agents such as nitrogen mustard, chlorambucil, cytoxan, ifosfamide, melphalan, thiptepa and busulfan; antimetabolites such as methotrexate, 5-fluorouracil, cytoxine arabinoside (ara-C), 5 -azacitidine, 6- mercaptopurine, 6-thioguanine, and fludarabine phosphat; antitumor antibiotics such as doxorubicin, adriamycin, daunorubicin, dactinomycin, bleomycin, mitomycin C, plicamycin, idarubicin, and mitoxantrone; vinca alkaloids and epipodophyllotoxins such as vincristine, vinblastine, vindesine, etoposide, and teniposide; nitrosoureas such as carmustine, lomustine, semustine and streptozotocin; synthetic agents such as dacrabazine, hexamethyl melamine, hydroxyurea, mitotane procabazine, cisplatin, cisplatinum and carboplatin; corticosteroids (cortisone acetate, hydrocortisone, prednisone, prednisolone, methyl prednisolone and dexamethasone), estrogens (diethylstilbestrol, estradiol, esterified estrogens, conjugated estrogens, chi orotriani sene), progesterones (medroxyprogesterone acetate, hydroxy progesterone caproate, megestrol acetate), anti-estrogens (tamoxifen), aromastase inhibitors (aminoglutethimide), androgens (testosterone propionate), methyl testosterone, fluoxymesterone, testolactone), anti-androgens (flutamide), LHRH analogues (leuprolide acetate), and endocrines for prostate cancer (ketoconazole).
In some embodiments, the additional agent is a drug for colorectal cancer. In some embodiments, the drug for colorectal cancer is based on regimens FOLFOX or FOLFIRI including 5-FU, leucovorin, oxaliplatin, irinotecan or their combinations. In a conventional standard method of treatment, the combination therapy is used together with cetuximab and/or bevacizumab.
The additional agent (e.g., anti-cancer drug or therapy) can be administered to the subject simultaneously or sequentially with the compound of the present embodiments, or can be formulated with the compounds of the present embodiments in the same pharmaceutical composition.
It is expected that during the life of a patent maturing from this application many relevant medical conditions associated with abnormal activity of TNTK will be developed and the scope of this phrase is intended to include all such new technologies a priori.
As used herein the term “about” refers to ± 10 % or ± 5 %. The terms "comprises", "comprising", "includes", "including", “having” and their conjugates mean "including but not limited to".
The term “consisting of’ means “including and limited to”.
The term "consisting essentially of means that the composition, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
As used herein, the singular form "a", "an" and "the" include plural references unless the context clearly dictates otherwise. For example, the term "a compound" or "at least one compound" may include a plurality of compounds, including mixtures thereof.
Throughout this application, various embodiments of this invention may be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
Whenever a numerical range is indicated herein, it is meant to include any cited numeral (fractional or integral) within the indicated range. The phrases “ranging/ranges between” a first indicate number and a second indicate number and “ranging/ranges from” a first indicate number “to” a second indicate number are used herein interchangeably and are meant to include the first and second indicated numbers and all the fractional and integral numerals therebetween.
As used herein the term "method" refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
As used herein, the term “treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical or aesthetical symptoms of a condition. As used herein, the term "subject" means an animal, preferably a mammal such as a nonprimate (e.g., cow, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit or guinea pig) or a primate (e.g. monkey and human), preferably a human being. Herein throughout, the phrase “linking group” describes a group (a substituent) that is attached to another moiety in the compound via two or more atoms thereof. In order to differentiate a linking group from a substituent that is attached to another moiety in the compound via one atom thereof, the latter will be referred to herein and throughout as an “end group”.
As used herein, the term “amine” describes both a -NR’R” end group and a -NR'- linking group, wherein R’ and R" are each independently hydrogen, alkyl, cycloalkyl, aryl, as these terms are defined hereinbelow.
The amine group can therefore be a primary amine, where both R’ and R” are hydrogen, a secondary amine, where R’ is hydrogen and R” is alkyl, cycloalkyl or aryl, or a tertiary amine, where each of R’ and R” is independently alkyl, cycloalkyl or aryl.
Alternatively, R' and R" can each independently be hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, cyano, nitro, azo, sulfonamide, carbonyl, C-carboxylate, O-carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, N-carbamate, O-carbamate, C-amide, N-amide, guanyl, guanidine and hydrazine.
The term “amine” is used herein to describe a -NR'R" group in cases where the amine is an end group, as defined hereinunder, and is used herein to describe a -NR'- group in cases where the amine is or forms a part of a linking group.
The term "alkyl" describes a saturated aliphatic hydrocarbon including straight chain and branched chain groups. Preferably, the alkyl group has 1 to 20 carbon atoms. Whenever a numerical range; e.g., " 1-20", is stated herein, it implies that the group, in this case the alkyl group, may contain 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms. In some embodiments, the alkyl is a medium size alkyl having 1 to 10 carbon atoms. Unless otherwise indicated, the alkyl is a lower alkyl having 1 to 4 carbon atoms. In some embodiments, the alkyl has at least 4 carbon atoms, for example, the alkyl is having 4 to 12 or 4 to 10 or 4 to 8 carbon atoms. The alkyl group may be substituted or unsubstituted. Substituted alkyl may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, oxo, carbonyl, cyano, nitro, azo, sulfonamide, C- carboxylate, O-carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, N-carbamate, O-carbamate, C-amide, N-amide, guanyl, guanidine and hydrazine.
The alkyl group can be an end group, as this phrase is defined hereinabove, wherein it is attached to a single adjacent atom, or a linking group, as this phrase is defined hereinabove, which connects two or more moieties via at least two carbons in its chain. When an alkyl is a linking group, it is also referred to herein as “alkylene”, e.g., methylene, ethylene, propylene, etc.
The term “alkenyl” describes an alkyl, as defined herein, in which at least one pair of carbon atoms are linked to one another via a double bond.
The term “alkynyl” or “alkyne” describes an alkyl, as defined herein, in which at least one pair of carbon atoms are linked to one another via a triple bond.
The term "cycloalkyl" describes an all-carbon monocyclic or fused ring (/.< ., rings which share an adjacent pair of carbon atoms) group where one or more of the rings does not have a completely conjugated pi-electron system. The cycloalkyl group may be substituted or unsubstituted. Substituted cycloalkyl may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, oxo, carbonyl, cyano, nitro, azo, sulfonamide, C-carboxylate, O-carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, N-carbamate, O-carbamate, C-amide, N-amide, guanyl, guanidine and hydrazine. The cycloalkyl group can be an end group, as this phrase is defined hereinabove, wherein it is attached to a single adjacent atom, or a linking group, as this phrase is defined hereinabove, connecting two or more moieties at two or more positions thereof.
The term "heteroalicyclic" describes a monocyclic or fused ring group having in the ring(s) one or more atoms such as nitrogen, oxygen and sulfur. The rings may also have one or more double bonds. However, the rings do not have a completely conjugated pi-electron system. The heteroalicyclic may be substituted or unsubstituted. Substituted heteroalicyclic may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, oxo, carbonyl, cyano, nitro, azo, sulfonamide, C- carboxylate, O-carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, O-carbamate, N- carbamate, C-amide, N-amide, guanyl, guanidine and hydrazine. The heteroalicyclic group can be an end group, as this phrase is defined hereinabove, where it is attached to a single adjacent atom, or a linking group, as this phrase is defined hereinabove, connecting two or more moieties at two or more positions thereof. Representative examples are piperidine, piperazine, tetrahydrofurane, tetrahydropyrane, morpholino and the like.
Exemplary nitrogen-containing heteroalicyclic groups or moieties include, but are not limited to, Representative examples of nitrogen-containing heteroalicyclic include, but are not limited to, morpholine, thiomorpholine, piperidine, piperazine, hexahydroazepine and tetrahydropyrane. Other moieties are also contemplated.
The term “piperazine" refers
Figure imgf000076_0001
group
Figure imgf000076_0002
Figure imgf000076_0003
group, where R’ and R’ ’ are as defined hereinabove.
The term “piperidine” refers to a
Figure imgf000076_0005
group or a
Figure imgf000076_0004
group, with R’ as defined herein.
- N,
The term “pyrrolidine” refers to a
Figure imgf000076_0011
group or a
Figure imgf000076_0006
group, with R’ as defined herein.
Figure imgf000076_0010
group, with R’ as defined herein.
Figure imgf000076_0007
- N O
The term “morpholine” refers to a \ - / group, and encompasses also thiomorpholine.
The term “thiomorpholine” refers t
Figure imgf000076_0008
group.
The term “hexahydroazepine” refers t
Figure imgf000076_0009
group. The term "aryl" describes an all-carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi-electron system. The aryl group may be substituted or unsubstituted. Substituted aryl may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, cyano, nitro, azo, sulfonamide, C-carboxylate, O- carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, N-carbamate, O-carbamate, C- amide, N-amide, guanyl, guanidine and hydrazine. The aryl group can be an end group, as this term is defined hereinabove, wherein it is attached to a single adjacent atom, or a linking group, as this term is defined hereinabove, connecting two or more moieties at two or more positions thereof. Preferably, the aryl is phenyl. Optionally, the aryl is naphthalenyl.
The term "heteroaryl" describes a monocyclic or fused ring (/.< ., rings which share an adjacent pair of atoms) group having in the ring(s) one or more atoms, such as, for example, nitrogen, oxygen and sulfur and, in addition, having a completely conjugated pi-electron system. Examples, without limitation, of heteroaryl groups include pyrrole, furane, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrimidine, triazine, tetrazine, quinoline, isoquinoline and purine. The heteroaryl group may be substituted or unsubstituted. Substituted heteroaryl may have one or more substituents, whereby each substituent group can independently be, for example, hydroxyalkyl, trihaloalkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroalicyclic, amine, halide, sulfinate, sulfate, sulfonate, sulfoxide, phosphonate, hydroxy, alkoxy, aryloxy, thiohydroxy, thioalkoxy, thioaryloxy, cyano, nitro, azo, sulfonamide, C-carboxylate, O- carboxylate, N-thiocarbamate, O-thiocarbamate, urea, thiourea, O-carbamate, N-carbamate, C- amide, N-amide, guanyl, guanidine and hydrazine. The heteroaryl group can be an end group, as this phrase is defined hereinabove, where it is attached to a single adjacent atom, or a linking group, as this phrase is defined hereinabove, connecting two or more moieties at two or more positions thereof.
The term “alkaryl” describes an alkyl, as defined herein, which is substituted by one or more aryl or heteroaryl groups. An example of alkaryl is benzyl.
The term "halide" and “halo” describes fluorine, chlorine, bromine or iodine.
The term “haloalkyl” describes an alkyl group as defined above, further substituted by one or more halide. The term “sulfate” describes a -0-S(=0)2-0R’ end group, as this term is defined hereinabove, or an -O-S(=O)2-O- linking group, as these phrases are defined hereinabove, where R’ is as defined hereinabove.
The term “thiosulfate” describes a -O-S(=S)(=O)-OR’ end group or a -O-S(=S)(=O)-O- linking group, as these phrases are defined hereinabove, where R’ is as defined hereinabove.
The term “sulfite” describes an -O-S(=O)-O-R’ end group or a -O-S(=O)-O- group linking group, as these phrases are defined hereinabove, where R’ is as defined hereinabove.
The term “thiosulfite” describes a -O-S(=S)-O-R’ end group or an -O-S(=S)-O- group linking group, as these phrases are defined hereinabove, where R’ is as defined hereinabove.
The term “sulfinate” or “sulfinyl” describes a -S(=O)-OR’ end group or an -S(=O)-O- group linking group, as these phrases are defined hereinabove, where R’ is as defined hereinabove.
The term “sulfoxide” describes a -S(=O)R’ end group or an -S(=O)- linking group, as these phrases are defined hereinabove, where R’ is as defined hereinabove.
The term "sulfonate” or “sulfonyl” describes a -S(=O)2-OR’ end group (also referred to herein as -SO3R’ or -SO3H) or an — O-S(=O)2- linking group, as these phrases are defined hereinabove, where R’ is as defined herein.
The term “S-sulfonamide” describes a -S(=O)2-NR’R” end group or a -S(=O)2-NR’- linking group, as these phrases are defined hereinabove, with R’ and R” as defined herein.
The term "N-sulfonamide" describes an R’ S(=O)2-NR”- end group or a -S(=O)2-NR’- linking group, as these phrases are defined hereinabove, where R’ and R” are as defined herein.
The term “disulfide” refers to a -S-SR’ end group or a -S-S- linking group, as these phrases are defined hereinabove, where R’ is as defined herein.
The term “phosphonate” describes a -P(=O)(OR’)(OR”) end group or a -P(=O)(OR’)(O)- linking group, as these phrases are defined hereinabove, with R’ and R” as defined herein.
The term “thiophosphonate” describes a -P(=S)(OR’)(OR”) end group or a -P(=S)(OR’)(O)- linking group, as these phrases are defined hereinabove, with R’ and R” as defined herein.
The term "carbonyl" or "carbonate" or “ketone” as used herein, describes a -C(=O)-R’ end group or a -C(=O)- linking group, as these phrases are defined hereinabove, with R’ as defined herein.
The term "thiocarbonyl" as used herein, describes a -C(=S)-R’ end group or a -C(=S)- linking group, as these phrases are defined hereinabove, with R’ as defined herein.
The term “oxo” as used herein, described a =0 end group.
The term “thiooxo” as used herein, described a =S end group. The term “oxime” describes a =N-OH end group or a =N-O- linking group, as these phrases are defined hereinabove.
The term “hydroxyl” or “hydroxy” describes a -OH group.
The term "alkoxy" describes both an -O-alkyl and an -O-cycloalkyl group, as defined herein.
The term "aryloxy" describes both an -O-aryl and an -O-heteroaryl group, as defined herein.
The term "thiohydroxy" or “thio” describes a -SH group.
The term "thioalkoxy" describes both a -S-alkyl group, and a -S-cycloalkyl group, as defined herein.
The term "thioaryloxy" describes both a -S-aryl and a -S-heteroaryl group, as defined herein.
The term "cyano" or “nitrile” describes a -C=N group.
The term “isocyanate” describes an -N=C=O group.
The term "nitro" describes an -NO2 group.
The term “carboxylate” as used herein encompasses C-carboxylate and O-carboxylate.
The term “C-carboxylate” describes a -C(=O)-OR’ end group or a -C(=O)-O- linking group, as these phrases are defined hereinabove, where R’ is as defined herein.
The term “O-carboxylate” describes a -OC(=O)R’ end group or a -OC(=O)- linking group, as these phrases are defined hereinabove, where R’ is as defined herein.
The term “thiocarboxylate” as used herein encompasses “C-thiocarboxylate and O- thiocarb oxy late.
The term “C-thiocarboxylate” describes a -C(=S)-OR’ end group or a -C(=S)-O- linking group, as these phrases are defined hereinabove, where R’ is as defined herein.
The term “O -thiocarb oxy late” describes a -OC(=S)R’ end group or a -OC(=S)- linking group, as these phrases are defined hereinabove, where R’ is as defined herein.
The term “carbamate” as used herein encompasses N-carbamate and O-carbamate.
The term “N-carbamate” describes an R”OC(=O)-NR’- end group or a -OC(=O)-NR’- linking group, as these phrases are defined hereinabove, with R’ and R” as defined herein.
The term “O-carbamate” describes an -OC(=O)-NR’R” end group or an -OC(=O)- NR’- linking group, as these phrases are defined hereinabove, with R’ and R” as defined herein.
The term “thiocarbamate” as used herein encompasses N-thiocarbamate and O- thiocarbamate. The term “O-thiocarbamate” describes a -OC(=S)-NR’R” end group or a -OC(=S)-NR’- linking group, as these phrases are defined hereinabove, with R’ and R” as defined herein.
The term “N-thiocarbamate” describes an R”OC(=S)NR’- end group or a -OC(=S)NR’- linking group, as these phrases are defined hereinabove, with R’ and R” as defined herein.
The term “dithiocarbamate” as used herein encompasses N-dithiocarbamate and S- dithiocarbamate.
The term “S-dithiocarbamate” describes a -SC(=S)-NR’R” end group or a -SC(=S)NR’- linking group, as these phrases are defined hereinabove, with R’ and R” as defined herein.
The term “N-dithiocarbamate” describes an R”SC(=S)NR’- end group or a -SC(=S)NR’- linking group, as these phrases are defined hereinabove, with R’ and R” as defined herein.
The term "urea", which is also referred to herein as “ureido”, describes a -NR’C(=O)- NR”R’ ’ ’ end group or a -NR’C(=O)-NR”- linking group, as these phrases are defined hereinabove, where R’ and R” are as defined herein and R'" is as defined herein for R' and R".
The term “thiourea”, which is also referred to herein as “thioureido”, describes a -NR’- C(=S)-NR”R”’ end group or a -NR’-C(=S)-NR”- linking group, with R’, R” and R’” as defined herein.
The term “amide” as used herein encompasses C-amide and N-amide.
The term “C-amide” describes a -C(=O)-NR’R” end group or a -C(=O)-NR’- linking group, as these phrases are defined hereinabove, where R’ and R” are as defined herein.
The term “N-amide” describes a R’C(=O)-NR”- end group or a R’C(=O)-N- linking group, as these phrases are defined hereinabove, where R’ and R” are as defined herein.
The term “guanyl” describes a R’R”NC(=N)- end group or a -R’NC(=N)- linking group, as these phrases are defined hereinabove, where R’ and R” are as defined herein.
The term “guanidine” describes a -R’NC(=N)-NR”R”’ end group or a - R’NC(=N)- NR”- linking group, as these phrases are defined hereinabove, where R’, R" and R'" are as defined herein.
The term “hydrazine” describes a -NR’-NR”R”’ end group or a -NR’ -NR”- linking group, as these phrases are defined hereinabove, with R’, R”, and R'" as defined herein.
As used herein, the term “hydrazide” describes a -C(=O)-NR’-NR”R”’ end group or a - C(=O)-NR’-NR”- linking group, as these phrases are defined hereinabove, where R’, R” and R’” are as defined herein. As used herein, the term “thiohydrazide” describes a -C(=S)-NR’-NR”R”’ end group or a -C(=S)-NR’-NR”- linking group, as these phrases are defined hereinabove, where R’, R” and R’” are as defined herein.
As used herein, the term “alkylene glycol” describes a -O-[(CR’R”)Z-O]y-R’” end group or a -O-[(CR’R”)Z-O]y- linking group, with R’, R” and R’” being as defined herein, and with z being an integer of from 1 to 10, preferably, from 2 to 6, more preferably 2 or 3, and y being an integer of 1 or more. Preferably R’ and R” are both hydrogen. When z is 2 and y is 1, this group is ethylene glycol. When z is 3 and y is 1, this group is propylene glycol. When y is 2-4, the alkylene glycol is referred to herein as oligo(alkylene glycol).
For any of the embodiments described herein, the compound described herein may be in a form of a salt thereof, for example, a pharmaceutically acceptable salt thereof, and/or in a form of a prodrug thereof.
As used herein, the phrase “pharmaceutically acceptable salt” refers to a charged species of the parent compound and its counter-ion, which is typically used to modify the solubility characteristics of the parent compound and/or to reduce any significant irritation to an organism by the parent compound, while not abrogating the biological activity and properties of the administered compound.
In the context of some of the present embodiments, a pharmaceutically acceptable salt of the compounds described herein may optionally be a base addition salt comprising at least one acidic (e.g., phenol and/or carboxylic acid) group of the compound which is in a negatively charged form (e.g., wherein the acidic group is deprotonated), in combination with at least one counter-ion, derived from the selected base, that forms a pharmaceutically acceptable salt.
The base addition salts of the compounds described herein may therefore be complexes formed between one or more acidic groups of the drug and one or more equivalents of a base.
The base addition salts may include a variety of organic and inorganic counter-ions and bases, such as, but not limited to, sodium (e.g., by addition of NaOH), potassium (e.g., by addition of KOH), calcium (e.g., by addition of Ca(OH)2, magnesium (e.g., by addition of Mg(OH)2), aluminum (e.g., by addition of Al(0H)3 and ammonium (e.g., by addition of ammonia). Each of these acid addition salts can be either a mono-addition salt or a poly-addition salt, as these terms are defined herein.
In the context of some of the present embodiments, a pharmaceutically acceptable salt of the compounds described herein may optionally be an acid addition salt comprising at least one base group (e.g., amine or amide group) of the compound which is in a positively charged form (e.g., wherein an -NH- group is protonated), in combination with at least one counter-ion, derived from the selected acid, that forms a pharmaceutically acceptable salt.
The acid addition salts of the compounds described herein may therefore be complexes formed between one or more basic groups of the drug and one or more equivalents of an acid.
The acid addition salts may include a variety of organic and inorganic acids, such as, but not limited to, hydrochloric acid which affords a hydrochloric acid addition salt, hydrobromic acid which affords a hydrobromic acid addition salt, acetic acid which affords an acetic acid addition salt, ascorbic acid which affords an ascorbic acid addition salt, benzenesulfonic acid which affords a besylate addition salt, camphorsulfonic acid which affords a camphorsulfonic acid addition salt, citric acid which affords a citric acid addition salt, maleic acid which affords a maleic acid addition salt, malic acid which affords a malic acid addition salt, methanesulfonic acid which affords a methanesulfonic acid (mesylate) addition salt, naphthalenesulfonic acid which affords a naphthalenesulfonic acid addition salt, oxalic acid which affords an oxalic acid addition salt, phosphoric acid which affords a phosphoric acid addition salt, toluenesulfonic acid which affords a p-toluenesulfonic acid addition salt, succinic acid which affords a succinic acid addition salt, sulfuric acid which affords a sulfuric acid addition salt, tartaric acid which affords a tartaric acid addition salt and trifluoroacetic acid which affords a trifluoroacetic acid addition salt. Each of these acid addition salts can be either a mono-addition salt or a poly-addition salt, as these terms are defined herein.
Depending on the stoichiometric proportions between the charged group(s) in the compound and the counter-ion in the salt, the acid or base additions salts can be either monoaddition salts or poly-addition salts.
The phrase “mono-addition salt”, as used herein, refers to a salt in which the stoichiometric ratio between the counter-ion and charged form of the compound is 1 : 1, such that the addition salt includes one molar equivalent of the counter-ion per one molar equivalent of the compound.
The phrase “poly-addition salt”, as used herein, refers to a salt in which the stoichiometric ratio between the counter-ion and the charged form of the compound is greater than 1 : 1 and is, for example, 2: 1, 3 : 1, 4: 1 and so on, such that the addition salt includes two or more molar equivalents of the counter-ion per one molar equivalent of the compound.
As used herein, the term “prodrug” refers to a compound which is converted in the body to an active compound (e.g., the compound of the formula described hereinabove). A prodrug is typically designed to facilitate administration, e.g., by enhancing absorption. A prodrug may comprise, for example, the active compound modified with ester groups, for example, wherein any one or more of the hydroxyl groups of a compound is modified by an acyl group, optionally (Ci- 4)acyl (e.g., acetyl) group to form an ester group, and/or any one or more of the carboxylic acid groups of the compound is modified by an alkoxy or aryloxy group, optionally (Ci-4)alkoxy (e.g., methyl, ethyl) group to form an ester group.
Further, each of the compounds described herein, including the salts thereof, can be in a form of a solvate or a hydrate thereof.
The term “solvate” refers to a complex of variable stoichiometry (e.g., di-, tri-, tetra-, penta- , hexa-, and so on), which is formed by a solute (the heterocyclic compounds described herein) and a solvent, whereby the solvent does not interfere with the biological activity of the solute.
The term “hydrate” refers to a solvate, as defined hereinabove, where the solvent is water.
The compounds described herein can be used as polymorphs and the present embodiments further encompass any isomorph of the compounds and any combination thereof.
The present embodiments further encompass any enantiomers and diastereomers of the compounds described herein.
As used herein, the term "enantiomer" refers to a stereoisomer of a compound that is superposable with respect to its counterpart only by a complete inversion/reflection (mirror image) of each other. Enantiomers are said to have “handedness” since they refer to each other like the right and left hand. Enantiomers have identical chemical and physical properties except when present in an environment which by itself has handedness, such as all living systems. In the context of the present embodiments, a compound may exhibit one or more chiral centers, each of which exhibiting an R- or an ^'-configuration and any combination, and compounds according to some embodiments of the present invention, can have any their chiral centers exhibit an R- or an S- configuration.
The term "diastereomers", as used herein, refers to stereoisomers that are not enantiomers to one another. Diastereomerism occurs when two or more stereoisomers of a compound have different configurations at one or more, but not all of the equivalent (related) stereocenters and are not mirror images of each other. When two diastereoisomers differ from each other at only one stereocenter they are epimers. Each stereo-center (chiral center) gives rise to two different configurations and thus to two different stereoisomers. In the context of the present invention, embodiments of the present invention encompass compounds with multiple chiral centers that occur in any combination of stereo-configuration, namely any diastereomer.
Herein throughout, the term “about” describes ± 10 % or ± 5 %.
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements.
Various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below find experimental support in the following examples.
EXAMPLES
Reference is now made to the following examples, which together with the above descriptions illustrate some embodiments of the invention in a non limiting fashion.
EXAMPLE 1
Design and synthesis of bipiperidine-containing compounds
While practicing the TNIK inhibitors disclosed in, for example, WO 2019/156439, it has been uncovered that (i) the oral bioavailability of such compounds is very low (e.g., lower than 10 % and even lower than 5 %); and (ii) the blood exposure (AUC) of such compounds is low. The present inventors have uncovered that compounds such as disclosed in WO 2019/156439, which possess a phenol moiety, undergo substantial Phase 2 metabolism, for example, glucuronidation and possibly also sulfation, which adversely affects their activity; causes low blood exposure after IV and especially PO administration; and results in a low oral bioavailability.
In a search for a modification of such compounds that would overcome the above limitations, and would result, for example, in enhanced bioavailability, improved pharmacokinetic properties (e.g. enhanced blood exposure (AUC)), and reduced or nullified glucuronidation for compounds that possess a phenol moiety, while maintaining the compounds’ TNIK inhibition activity, the present inventors have conceived a methodology for protecting the phenol moiety.
The present inventors have first selected 1,4’ -bipiperidine as an exemplary protecting group, based on a successful performance of such a group in the drug known as CPT-11 (Irinotecan®) [See, for example, Kunimoto T et al., Cancer Res., 1987, 47, 5944-5947],
During the laborious studies conducted in a search for such a modification, several strategies for coupling l,4’-bipiperidine to an exemplary TNIK inhibitor, referred to herein as TNK-002 so as to provide a compound referred to herein as TNK-108 (see, FIG. 1 A), have been tested.
The present inventors have uncovered that a simple coupling of l,4’-bipiperidine to TNK- 002, using common coupling agents such as, for example, triphosgene, p-nitrophenyl chloroformate or carbonyldiimidazole (CDI), as exemplified in FIG. 2A, did not result in the desired coupling (for the first two) or resulted in a mixture of products that are inseparable by common techniques such as preparative HPLC. Attempts to perform such reactions while protecting nucleophilic amines on the compound, as exemplified in FIG. 2B, also failed.
The present inventors have therefore recognized that a more sophisticated synthetic route should be employed, which is not based on coupling the 1,4’ -bipiperidine to TNK-002, but rather involves “building” the final molecule differently. To this end, several possible building blocks were synthesized, as shown in FIGs. 3 A and 3B, yet these synthetic pathways also did not result in the desired compound.
After further laborious experimentation, the present inventors have uncovered a synthetic pathway in which TNK-108 was successfully prepared, as shown schematically in FIG. 4, and is detailed in the following.
Preparation of TNK-108 (Procedure I)
Materials and Methods:
2-Bromo-4-methoxyaniline (98 %; Compound 1) was obtained from BIDE;
Ethylboronic acid (95 %; Compound 2A) was obtained from TCI;
1,4'-Bipiperidine (99 %; Compound 4A) was obtained from BIDE;
Di(lH-imidazol-l-yl)methanethione (99 %; Compound 6B) was obtained from TCI; and l-(4-(lH-Pyrazol-l-yl)-phenyl)ethanone (100 %; Compound 7A) was prepared by Wuxi.
All other reagents and solvents were obtained from known vendors.
LC-MS analyses were performed using SHIMADZU LCMS-2020; Column: Kinetex EVO C18 30*2.1mm, 5pm
NMR measurements were performed using Bruker AVANCE III HD 400 MHz.
Preparation of compound 2 (see, FIG. 4):
Figure imgf000085_0001
To a solution of Compound 1 (5.00 grams, 24.8 mmol, 1.00 mol equiv.) in dichloromethane (DCM; 50.0 mL) was added TEA (5.01 grams, 49.5 mmol, 6.89 mL, 2.00 mol equiv.), (Boc)2O (5.67 grams, 26.0 mmol, 5.97 mL, 1.05 mol equiv.) and DMAP (908 mg, 7.42 mmol, 0.300 mol equiv.). The resulting mixture was stirred at 20 °C for 12 hours. TLC (petroleum ether: ethyl acetate = 5: 1) indicated that Compound 1 (Rf = 0.45) was consumed completely. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by column chromatography (SiCh, petroleum etherethyl acetate = 50: 1 to 20: 1, Rf = 0.55) to give Compound 2 (2.50 grams, 31.4 % yield, 93.8 % purity) as a yellow oil.
The compound’s structure was confirmed by 'H-NMR and LC-MS (not shown).
XH NMR (400 MHz, CDCh): 8 = 7.96 (d, J= 8.80 Hz, 1H), 7.08 (d, J= 2.90 Hz, 1H), 6.86 (dd, J= 2.90, 9.1 Hz, 1H), 6.73 (br s, 1H), 3.78 (s, 3H), 1.53 (s, 9H).
Preparation of Compound 3 (See FIG. 4):
Figure imgf000086_0001
2 3
A mixture of Compound 2 (2.20 grams, 7.28 mmol, 1.00 mol equiv.), Compound 2A (646 mg, 8.74 mmol, 1.20 mol equiv.), K3PO4 (3.09 grams, 14.6 mmol, 2.00 mol equiv.) and Pd(dppf)C12 (266 mg, 364 pmol, 0.05 mol equiv.) in dioxane (25.0 mL) was degassed and purged with N2 for 3 times and stirred at 110 °C for 12 hours under N2 atmosphere. LC-MS showed that Compound 2 was consumed completely and one main peak (RT = 0.879 minutes) with desired mass was detected. The reaction mixture was filtered and the filtrate was concentrated to give a residue, which was purified by column chromatography (SiCh, petroleum ether: ethyl acetate = 50: 1 to 5:1, TLC: petroleum etherethyl acetate = 5:1, Rf = 0.51) to give Compound 3 (1.60 gram, 87.4 % yield) as a white solid. The compound’s structure was confirmed by 'H-NMR.
XH NMR (400 MHz, CDCh): 6 = 7.62-7.42 (m, 1H), 6.81-6.65 (m, 2H), 6.06 (br s, 1H), 3.86-3.74 (m, 3H), 2.58 (q, J= 7.60 Hz, 2H), 1.53-1.48 (m, 9H), 1.23 (t, J= 7.60 Hz, 3H)
Figure imgf000086_0002
3 3 1
To a solution of Compound 3 (1.10 gram, 4.38 mmol, 1.00 mol equiv.) in DCM (5.00 mL) was added BBn (2.19 grams, 8.75 mmol, 843 pL, 2.00 mol equiv.) at 0 °C and the mixture was stirred at 20 °C for 2 hours. LC-MS showed that Compound 3 was consumed completely (not shown). The reaction mixture was quenched by saturated aqueous NaHCOs (30.0 mL), and extracted with ethyl acetate (30.0 mL x 3). The combined organic layers were dried over Na2SO4, filtered and concentrated to give Compound 3_1 (700 mg, crude) as an off-white solid.
Figure imgf000087_0001
3 1 4
To a solution of Compound 3_1 (700 mg, 5.10 mmol, 1.00 mol equiv.) in DCM (5.00 mL) and THF (5.00 mL) was added DIEA (1.32 gram, 10.2 mmol, 1.78 mL, 2.00 mol equiv.) and (Boc)2O (1.23 gram, 5.61 mmol, 1.29 mL, 1.10 mol equiv.). The resulting mixture was stirred at 20 °C for 2 hours. TLC (petroleum etherethyl acetate = 1 : 1) indicated that Compound 3_1 was consumed completely. The reaction mixture was diluted with ethyl acetate (20.0 mL) and washed with HC1 (1 M, 20.0 mL x 2). The combined organic layers were washed with brine (30.0 mL), dried over Na2SO4, filtered and concentrated to give a residue, which was purified by prep-TLC (SiO2, petroleum etherethyl acetate = 1 : 1, Rf = 0.32) to give Compound 4 (800 mg, 66.1 % yield) as a yellow solid. The compound’s structure was confirmed by 'H-NMR
'H-NMR (400 MHz, CDCh): 8 = 7.44-7.31 (m, 1H), 6.64 (d, J= 2.90 Hz, 1H), 6.59 (dd, J = 2.90, 8.50 Hz, 1H), 6.05 (br s, 1H), 5.16 (br s, 1H), 2.55 (q, J= 7.60 Hz, 2H), 1.55-1.47 (m, 9H), 1.23-1.13 (m, 3H).
Preparation of Compound 5 (see, FIG. 4):
Figure imgf000087_0002
To a solution of triphosgene (6.31 grams, 21.3 mmol, 2.52 mol equiv.) in DCM (60.0 mL) was added a solution of Compound 4A (6.38 grams, 37.9 mmol, 4.50 mol equiv.) and TEA (11.9 grams, 118 mmol, 16.4 mL, 14.0 mol equiv.) in DCM (40.0 mL) at 0 °C drop-wise and the mixture was stirred at 20 °C for 30 minutes. Then the reaction mixture was concentrated and dissolved with toluene (60.0 mL). Compound 4 (2.00 grams, 8.43 mmol, 1.00 mol equiv.) and pyridine (Py; 10.0 mL) was added to the mixture. The resulting mixture was stirred at 120 °C for 12 hours. LC- MS showed that the desired mass (RT = 0.722 minutes) was detected. The reaction mixture was filtered and the filtrate was concentrated to give a residue, which was purified by column chromatography (SiCL, petroleum ether: ethyl acetate = 3: 1 to about 0: 1, TLC: di chloromethane methanol NH ’JLO = 10: 1 :0.01, Rf = 0.14) to give Compound 5 (2.60 grams, 71.5 % yield) as a yellow solid. The compound’s structure was confirmed by 'H-NMR.
'H-NMR (400 MHz, DMSO): 5 = 8.51 (s, 1H), 7.21 (d, J = 8.60 Hz, 1H), 6.95-6.85 (m, 2H), 4.17 (br s, 1H), 4.10-3.97 (m, 1H), 3.64-3.50 (m, 1H), 2.99 (br s, 1H), 2.84 (br s, 1H), 2.56 (d, J = 7.60 Hz, 2H), 1.80 (d, J = 8.60 Hz, 2H), 1.58-1.48 (m, 6H), 1.45 (s, 9H), 1.41 (br s, 4H), 1.06-1.01 (m, 3H).
Figure imgf000088_0001
To a solution of Compound 5 (2.60 grams, 6.02 mmol, 1.00 mol equiv.) in ethyl acetate (10.0 mL) was added HC1-EA (4.00 M, 28.9 mL, 19.2 mol equiv.) and the mixture was stirred at 25 °C for 12 hours. TLC (dichloromethane:methanol = 10: 1) indicated that Compound 5 (Rf = 0.25) was consumed completely and one new spot (Rf = 0.15) formed. The reaction mixture was concentrated to give a residue, which was basified with basic resin to give Compound 6 (2.00 grams, 99.0 % yield, 98.8 % purity) as a yellow oil, as confirmed by LC-MS (not shown). Preparation of Compound 7 (see, FIG. 4):
Figure imgf000089_0001
6 7
To a solution of Compound 6 (2.00 grams, 6.03 mmol, 1.00 mol equiv.) in THF (60.0 mL) was added Compound 6B (1.13 gram, 6.34 mmol, 1.05 mol equiv.) and the mixture was stirred at 25 °C for 12 hours. Then NH2NH2*H2O (610 mg, 12.2 mmol, 592 pL, 2.02 mol equiv.) was added to the mixture and stirred at 25 °C for 16 hours. Then NH NH ’H O (2.22 grams, 44.3 mmol, 2.16 mL, 7.35 mol equiv.) was added again and the mixture was stirred at 25 °C for another 6 hours. LCMS showed that Compound 6 was consumed and one main peak (RT = 0.946 minutes) with desired mass was detected (not shown). The reaction mixture was quenched with water (40.0 mL), and extracted with ethyl acetate (30.0 mL x 3). The combined organic layers were washed with brine (80.0 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give Compound 7 (1.75 gram, crude) as a yellow solid.
Preparation of Compound 7 A:
Figure imgf000089_0002
7A_1 7A
To a solution of Compound 7A_1 (2.00 grams, 10.7 mmol, 1.00 mol equiv.) in AcOH (20.0 mL) and DCM (10.0 mL) was added HBr (2.63 grams, 10.7 mmol, 1.77 mL, 33 % purity in AcOH, 1.00 mol equiv.) and the mixture was stirred at 25 °C for 1 hour, then Bn (1.89 gram, 11.8 mmol, 609 pL, 1.10 mol equiv.) was added at 0 °C. The resulting mixture was stirred at 70 °C for 4 hours. LCMS showed that about 19 % of Compound 7A_1 (RT = 0.787 minutes) was remained and about 71% of desired mass (RT = 0.853 minutes) was detected (not shown). The reaction mixture was diluted with ethyl acetate (20.0 mL) and washed with saturated aqueous NaHCCL (30.0 mL x 3). The combined organic layers were washed with brine (90.0 mL), dried over Na2SO4, filtered and concentrated to give a residue, which was triturated with ethyl acetate (5.00 mL) and filtered to give the compound 7A (1.80 gram, 63.2 % yield) as a yellow solid.
Preparation of Compound 8 (see, FIG. 4):
Figure imgf000090_0001
To a solution of Compound 7 (1.40 gram, 3.45 mmol, 1.00 mol equiv.) in EtOH (60.0 mL) and THF (10.0 mL) was added a solution of compound 7A (915 mg, 3.45 mmol, 1.00 mol equiv.) in EtOH (20.0 mL) and THF (10.0 mL) at 0 °C and the mixture was stirred at 25 °C for 22 hours. LCMS showed that the reaction was completed (not shown). The reaction mixture was concentrated to give Compound 8 (2.10 grams, crude) as a yellow solid.
Preparation of TNK-108 (see, FIG. 4):
A mixture of Compound 8 (2.10 grams, 3.66 mmol, 1.00 mol equiv.), NaHCOs (924 mg, 11.0 mmol, 428 pL, 3.01 mol equiv.) in EtOH (600 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 80 °C for 48 hours under N2 atmosphere. LC-MS showed about 13.7 % of Compound 8 (RT = 0.786 minutes) was remained and 23.7 % of desired mass (RT = 0.824 minutes) was detected. The reaction mixture was concentrated give a residue, which was purified by reversed-phase HPLC (0.1 % FA condition), followed by further purification by preparative HPLC (HC1 condition; column: Phenomenex Synergi C18 150x25x10pm; mobile phase : [water(0.05%HCl)-ACN];B% : 20%-40%,10 minutes) to give TNK-108 (57.95 mg, 2.68 % yield, 97.6 % purity, HC1 salt) as a yellow solid.
The compound’s structure was confirmed by LC-MS (not shown) and 'H-NMR.
E-NMR (400 MHz, DMSO): 8 = 10.29 (br s, 1H), 8.58 (d, J= 2.50 Hz, 1H), 7.98-7.86 (m, 4H), 7.79 (d, J= 1.60 Hz, 1H), 7.50 (d, J= 8.70 Hz, 1H), 6.94-6.83 (m, 2H), 6.62-6.55 (m, 1H), 6.37 (s, 1H), 4.36-4.25 (m, 1H), 4.21-4.13 (m, 1H), 3.41 (d, J= 11.3 Hz, 4H), 3.12-2.87 (m, 4H), 2.68-2.61 (m, 2H), 2.15 (d, J= 11.5 Hz, 2H), 1.91-1.78 (m, 4H), 1.72 (d, J= 12.8 Hz, 2H), 1.48-1.34 (m, 1H), 1.18 (t, J= 7.50 Hz, 3H). Preparation of TNK-108 (Procedure II; gram scale):
A general synthetic pathway for preparing TNK-108 on a gram scale is shown in FIGs. 5A-B.
Materials and Methods:
3 -Ethylphenol (98 %; Compound 11) was obtained from TCI;
1,4'-Bipiperidine (95 %; Compound 13) was obtained from BIDE;
Ethyl 4-fluorobenzoate (99 %; Compound 15-1) was obtained from BIDE;
IH-Pyrazole (98 %; Compound 15-4) was obtained from TCI;
BrettPhos Pd G3 (CAS Number 1470372-59-8) (100 %) was obtained from BIDE.
All other reagents and solvents were obtained from known vendors.
LC-MS analyses were performed using SHIMADZU LCMS-2020; Column: Kinetex EVO C18 30*2.1mm, 5pm
NMR measurements were performed using Bruker AVANCE III HD 400 MHz.
Preparation of Compound 15-2 (see, FIG. 5B):
Figure imgf000091_0001
A solution of Compound 15-1 (5.00 grams, 29.7 mmol, 4.39 mL, 1.00 mol equivalent), K2CO3 (8.22 grams, 59.4 mmol, 2.00 mol equivalent) and Compound 15-4 (2.13 grams, 31.2 mmol, 1.05 mol equivalent) in DMSO (50.0 mL) was stirred at 110 °C for 18 hours under N2 atmosphere. LCMS showed that Compound 15-1 was consumed completely and a desired mass (RT = 0.88) was detected. The reaction mixture was diluted with H2O (350 mL) and thereafter extracted with ethyl acetate (250 mL). The organic phase was washed with brine (60.0 mL x 3), dried over Na2SO4, filtered and concentrated under reduced pressure to give Compound 15-2 (6.40 grams, 29.6 mmol, 99.5 % yield) as a white solid.
Preparation of Compound 15-3 (see, FIG. 5B):
Figure imgf000091_0002
To a solution of Compound 15-2 (5.00 grams, 23.1 mmol, 1.00 mol equivalent) in THF (15.0 mL) was added t-BuOK (5.19 grams, 46.2 mmol, 2.00 mol equivalents). The mixture was stirred at 25 °C for 10 minutes. Then, MeCN (1.90 gram, 46.2 mmol, 2.43 mL, 2.00 mol equivalents) was added and the resulting mixture was stirred at 25 °C for 2 hours. LCMS showed that Compound 15-2 was consumed and a desired mass (RT = 0.580) was detected. The reaction mixture was quenched with ice water (80.0 mL), acidified with IM HC1 to pH of about 5, and thereafter extracted with ethyl acetate (80.0 mL x 3). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give Compound 15-3 as a yellow solid.
Preparation of Compound 15-5 (see, FIG. 5B):
Figure imgf000092_0001
To a solution of Compound 15-3 (4.80 grams, 22.7 mmol, 1.00 mol equivalent) in toluene (48.0 mL) was added NH NH ’H O (11.3 grams, 227 mmol, 11.0 mL, 10.0 mol equivalents). The resulting mixture was stirred at 110 °C for 18 hours. TLC (petroleum ether/ethyl acetate = 1/1) indicated that Compound 15-3 (Rf = 0.4) was consumed completely and one new spot (Rf = 0.3) formed. The reaction mixture was filtered and the filter cake was concentrated under reduced pressure to give a residue, which was triturated with petroleum ether/ethyl acetate (1/1, 20.0 mL) at 30 °C for 15.0 minutes, filtered and the filter cake was collected and concentrated to give Compound 15-5 (5.00 grams, 22.2 mmol, 97.6 % yield) as a yellow solid.
Preparation of Compound 15 (see, FIG. 5B):
Figure imgf000092_0002
15-5 15
To a solution of Compound 15-5 (5.00 grams, 22.2 mmol, 1.00 mol equivalent) in THF (50.0 mL) was added KOH (4.50 M, 39.46 mL, 8.00 mol equivalents) and BOC2O (5.09 grams, 23.3 mmol, 5.35 mL, 1.05 mol equivalent). The mixture was stirred at 30 °C for 18 hours. TLC (petroleum ether/ethyl acetate = 1/1) indicated that Compound 15-5 (Rf = 0.2) was consumed completely and one new spot (Rf= 0.5) formed. The reaction mixture was filtered and the filter cake was concentrated under reduced pressure to give a residue, which was triturated with ethyl acetate (20.0 mL) at 30 °C for 15 minutes, filtered and the filter cake was collected and concentrated under pressure to give Compound 15 (6.00 grams, 17.7 mmol, 79.7 % yield, 96.0 % purity) as a light yellow solid.
'H-NMR (400 MHz, DMSO): 3 = 8.54 (d, J= 2.3 Hz, 1H), 7.88 (d, J= 2.1 Hz, 4H), 7.77 (d, J= 1.5 Hz, 1H), 6.58-6.55 (m, 1H), 6.43 (s, 2H), 5.82 (s, 1H), 1.60 (s, 9H).
Preparation of Compound 12 (see, FIG. 5 A):
Figure imgf000093_0001
To a solution of Compound 11 (5.00 grams, 40.9 mmol, 4.95 mL, 1.00 mol equivalent) in DCM (15 mL) and MeOH (10 mL) was added TBATB (20.5 grams, 42.5 mmol, 1.04 mol equivalent). The mixture was stirred at 20 °C for 1 hour. LCMS showed that Compound 11 was consumed. The reaction mixture was washed with 1 M HC1 (100 mL), water (100 mL), and brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give Compound 12 (8.00 grams, 39.7 mmol, 97.2 % yield), as a yellow liquid.
Preparation of Compound 14 (see, FIG. 5 A):
Figure imgf000093_0002
To a solution of triphosgene (29.1 grams, 98.17 mmol, 2.00 mol equivalents) in DCM (90 mL) was added a solution of Compound 13 (8.26 grams, 49.0 mmol, 1.00 mol equivalent) and TEA (14.9 grams, 147 mmol, 20.5 mL, 3.00 mol equivalent) in DCM (60 mL) at 0 °C drop-wise. The mixture was stirred at 20 °C for 30 minutes. The reaction mixture was then concentrated and diluted with toluene (100 mL). Then Compound 2* (4.93 grams, 24.5 mmol, 0.50 mol equivalent) and Py (15.5 grams, 196 mmol, 15.85 mL, 4.00 mol equivalents) were added. The resulting mixture was stirred at 120 °C for 12 hours. LCMS showed that a desired mass (RT = 0.842 minutes) was detected. The reaction mixture was washed with brine (200 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography (ISCO®; 16.0 g SepaFlash® Silica Flash Column, Eluent of 0-10% dichloromethane/methanol, TLC: dichloromethane/methanol = 10/1, Rf = 0.40) to give Compound 14 (2.00 grams, 5.06 mmol, 10.3 % yield) as a yellow solid.
Preparation of Compound 16 (see, FIG. 5 A):
Figure imgf000094_0001
14 16
A mixture of Compound 14 (1.50 gram, 2.85 mmol, 1.00 mol equivalent), Compound 15 (925 mg, 2.85 mmol, 1.00 mol equivalent), CS2CO3 (2.78 grams, 8.54 mmol, 3.00 mol equivalents), BrettPhos Pd G3 (515 mg, 569 pmol, 0.20 mol equivalents) in dioxane (50 mL) was degassed and purged with N2 3 times, and then the mixture was stirred at 100 °C for 15 hours under N2 atmosphere. LCMS showed that Compound 14 was consumed completely and a desired mass (RT = 0.941 minutes) was detected. The reaction mixture was diluted with H2O (250 mL) and extracted with EA (200 mL x 3). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give Compound 16 (3.50 grams, crude) as a yellow oil. Preparation of TNK-108 (see, FIG. 5 A):
Figure imgf000095_0001
To a solution of Compound 16 (3.50 grams, 5.47 mmol, 1.00 mol equivalent) in DCM (25 mL) was added TFA (38.5 grams, 337 mmol, 25 mL, 61.7 mol equivalents). The mixture was stirred at 25 °C for 1 hour. LCMS showed that Compound 6* was consumed completely and a desired mass (RT = 0.827 minutes) was detected. The reaction mixture was basified with saturated aqueous Na2CCf to pH of about 8, and thereafter extracted with ethyl acetate (100 mL x 3). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by reversed-phase HPLC (0.1% TFA condition) to give TNK-108 (829 mg, 28.0 % yield) as a light solid, as confirmed by LCMS (not shown) and 1 H-NMR.
XH NMR (400 MHz, DMSO): 3 = 9.10 (d, J= 7.2 Hz, 1H), 8.55 (d, J= 2.4 Hz, 1H), 8.04- 7.69 (m, 5H), 7.65-7.30 (m, 2H), 6.92-6.76 (m, 2H), 6.63-6.51 (m, 1H), 6.35 (s, 1H), 4.33-4.17 (m, 2H), 3.43 (br d, J= 11.2 Hz, 3H), 3.06-2.89 (m, 4H), 2.65 (q, J = 7.3 Hz, 2H), 2.07 (d, J = 11.6 Hz, 2H), 1.86 (br d, J= 13.4 Hz, 2H), 1.75-1.63 (m, 5H), 1.48-1.38 (m, 1H), 1.18 (t, J= 7.5 Hz, 3H).
Preparation of TNK-127 (see, FIG. IB)
Using a similar synthetic pathway, TNK-127 was prepared, upon using respective starting materials.
A general synthetic pathway for preparing TNK-127 on a gram scale is shown in FIG. 6.
Materials and Methods:
3 -Ethylaniline (98 %; Compound 21) was obtained from TCI;
1,4'-Bipiperidine (95 %; Compound 13) was obtained from BIDE;
N-bromosuccinimide (NBS; 99 %) was obtained from TaiTan; N,N’ -Carbonyldiimidazole (CDI; 99 %) was obtained from Aladdin;
BrettPhos Pd G3 (100 %) was obtained from BIDE.
All other reagents and solvents were obtained from known vendors.
LC-MS analyses were performed using SHIMADZU LCMS-2020; Column: Kinetex EVO C18 30*2.1mm, 5pm
NMR measurements were performed using Bruker AVANCE III HD 400 MHz.
Figure imgf000096_0001
To a solution of Compound 21 (1.00 gram, 8.25 mmol, 1.03 mL, 1.00 mol equivalent) in DMF (10 mL) was added NBS (1.47 gram, 8.25 mmol, 1.00 mol equivalent) at 0 °C. The mixture was stirred at 25 °C for 1 hour. TLC (petroleum ether/ethyl acetate = 10/1) showed that Compound 21 (Rf = 0.32) was consumed and a new spot (Rf = 0.29) was formed. The reaction mixture was diluted with ethyl acetate (50 mL), washed with brine (40 mL x 3), dried over Na2SO4, filtered and concentrated under vacuum to give compound 22 (1.60 gram, crude) as brown liquid, which was confirmed by 1 H-NMR.
XHNMR (400 MHz, DMSO): <5 = 7.13 (d, J= 8.63 Hz, 1H), 6.54 (d, J= 2.75 Hz, 1H), 6.36 (dd, J= 8.50, 2.88 Hz, 1H), 5.18 (br s, 2H), 2.51-2.56 (m, 2H), 1.12 (t, J= 7.57 Hz, 3H)
Preparation of Compound 23 (see, FIG. 6):
Figure imgf000096_0002
To a solution of Compound 22 (300 mg, 1.50 mmol, 1.00 mol equivalent), TEA (151 mg, 1.50 mmol, 208 pL, 1.00 mol equivalent) in DMF (2 mL) was added CDI (243 mg, 1.50 mmol, 1.00 mol equivalent) at 0 °C. The mixture was stirred at 25 °C for 1 hour. Then a solution of Compound 13 (252 mg, 1.50 mmol, 1.00 mol equivalent) in DMF (1 mL) was added and the resulting mixture was stirred at 25 °C for 8 hours. LCMS showed that Compound 22 was consumed and the desired mass (RT = 0.829) was detected. The reaction mixture was diluted with ethyl acetate (10 mL), acidified to pH of about 6 with 1 M HC1, filtered and the filtrate cake was collected to give Compound 23 (500 mg, 1.27 mmol, 84.5 % yield) as a white solid.
Preparation of TNK-127 (see, FIG. 6):
Figure imgf000097_0001
23 TNK-127
A suspension of Compound 15 (209 mg, 553 pmol, 1.00 mol equivalent), prepared as described hereinabove and shown in FIG. 5B, Compound 23 (218 mg, 553 pmol, 1.00 mol equivalent) and CS2CO3 (360 mg, 1.11 mmol, 2.00 mol equivalents) in dioxane (4 mL) was degassed and purged with N2 for 3 times, and thereafter charged with BrettPhos Pd G3 (25.0 mg,
27.6 pmol, 0.05 mol equivalent) and the resulting mixture was stirred at 90 °C for 1.5 hours under nitrogen. TLC (plate 1, petroleum ether/ethyl acetate = 1/2) showed that Compound 23 (Rf = 0.41) was consumed and a major spot (Rf = 0.20) was formed. The mixture was filtered through a pad of Celite®, the filtrate was concentrated under vacuum to give a residue, which was triturated with petroleum ether/ethyl acetate (1/1, 5.00 mL) at 25 °C for 1 hour, filtered and the filtrate cake was collected. TLC (plate 2, petroleum ether/ethyl acetate = 1/2) of the cake showed the spot of Rf = 0.2 was disappeared, and LCMS showed the desired mass of TNK-127. The cake was purified by preparative HPLC (column: Phenomenex luna C18 150x40mmxl5pm;mobile phase: [water(0.1%TFA)-ACN];B%: 10%-40%, 11 minutes) to give TNK-127 (74.6 mg, 136 pmol,
24.6 % yield, 98.5 % purity) as a yellow gum, as confirmed by 'H-NMR and LCMS (not shown).
XH NMR (400 MHz, DMSO): 3 = 9.08 (br dd, J= 4.16, 2.32 Hz, 1H), 8.55 (d, J= 2.45 Hz, 1H), 8.41 (s, 1H), 7.88-7.95 (m, 2H), 7.80-7.87 (m, 2H), 7.77 (d, J= 1.47 Hz, 1H), 7.37 (br d, J= 8.19 Hz, 2H), 7.11-7.25 (m, 2H), 6.45-6.64 (m, 1H), 6.25 (s, 1H), 4.27 (br d, J= 13.45 Hz, 2H), 3.33-3.48 (m, 3H), 2.85-3.04 (m, 2H), 2.69-2.84 (m, 2H), 2.61 (q, J= 7.38 Hz, 2H), 2.03 (br d, J = 10.76 Hz, 2H), 1.85 (br d, J= 13.45 Hz, 2H), 1.65-1.72 (m, 2H), 1.36-1.64 (m, 4H), 1.16 (t, J = 7.46 Hz, 3H). Preparation of TNK-135 (See, FIG. IB)
Using a similar synthetic pathway, TNK-135 was prepared, upon using respective starting materials.
A general synthetic pathway for preparing TNK-135 is shown in FIG. 7.
Materials and Methods:
3 -Ethylaniline (98 %; Compound 21) was obtained from TCI;
3-(piperidin-l-yl)propan-l-amine (97 %; Compound 25) was obtained from Sigma;
N-bromosuccinimide (NBS; 99 %) was obtained from TaiTan;
N,N’ -Carbonyldiimidazole (CDI; 99 %) was obtained from Aladdin;
BrettPhos Pd G3 (100 %) was obtained from BIDE.
All other reagents and solvents were obtained from known vendors.
LC-MS analyses were performed using SHIMADZU LCMS-2020; Column: Kinetex EVO C18 30*2.1mm, 5pm
NMR measurements were performed using Bruker AVANCE III HD 400 MHz.
Preparation of Compound 22 (see, FIG. 7):
Figure imgf000098_0001
21 22
To a solution of Compound 21 (1.00 gram, 8.25 mmol, 1.03 mL, 1.00 mol equivalent) in DMF (10 mL) was added NBS (1.47 gram, 8.25 mmol, 1.00 mol equivalent) at 0 °C. The reaction mixture was stirred at 25 °C for 1 hour. TLC (petroleum ether/ethyl acetate = 10/1) showed that Compound 21 (Rf = 0.32) was consumed and a new spot (Rf = 0.27) was formed. The reaction mixture was diluted with ethyl acetate (50 mL), washed with brine (50 mL x 3). The organic layer was dried over Na2SC>4, filtered and concentrated under vacuum to give a residue, which was purified by flash silica gel chromatography (ISCO®; 2.3 g SepaFlash® Silica Flash Column, Eluent of 0-10% ethyl acetate/petroleum ether, TLC: petroleum ether/ethyl acetate = 10/1, Rf = 0.27) to give Compound 22 (1.50 gram, 7.42 mmol, 89.9 % yield, 99.0 % purity) as a brown liquid, as confirmed by LCMS (not shown). Preparation of Compound 24 (see, FIG. 7):
Figure imgf000099_0001
22 24
To a solution of Compound 22 (400 mg, 2.00 mmol, 1.00 mol equivalent), TEA (202 mg, 2.00 mmol, 278 pL, 1.00 mol equivalent) in DMF (4 mL) was added CDI (324 mg, 2.00 mmol, 1.00 mol equivalent) at 0 °C. The mixture was stirred at 20 °C for 1 hour. Then a solution of Compound 25 (283 mg, 2.00 mmol, 1.00 mol equivalent) in DMF (1 mL) was added and the resulting mixture was stirred at 20 °C for 8 hours. LCMS (not shown) showed one main peak with desired mass (RT = 1.837 minutes). The reaction mixture was diluted with H2O (20.0 mL), and extracted with ethyl acetate (40 mL x 3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was triturated with ethyl acetate (10.0 mL) at 15 °C for 10 minutes, filtered and the cake was collected to give Compound 24 (300 mg, 814 pmol, 40.7 % yield) as a white solid as confirmed by LCMS (not shown) and 1 H-NMR.
XHNMR (400 MHz, DMSO): 3 = 9.61 (s, 1H), 8.92 (s, 1H), 7.38 (s, 2H), 7.21 (dd, J = 8.8, 2.8, 1H), 6.54 (t, J= 5.8, 1H), 3.40 (d, = 11.6, 2H), 3.25 (q, J = 6.4, 2H), 3.02-3.03 (m, 2H), 3.81-3.84 (m, 2H), 2.61 (q, J= 7.6, 2H), 1.68-1.86 (m, 6H), 1.37-1.39 (m, 1H), 1.14 (t, J=
7.4, 3H).
Preparation of TNK-135 (see, FIG. 7):
Figure imgf000100_0001
TNK-135
A mixture of Compound 24 (291 mg, 790 pmol, 1.47 mol equivalent), Compound 15 (175 mg, 537 pmol, 1.00 mol equivalent), prepared as described hereinabove and shown in FIG. 5B, BrettPhos Pd G3 (292 mg, 322 pmol, 0.60 mol equivalent), CS2CO3 (613 mg, 1.88 mmol, 3.50 mol equivalent) in dioxane (30.0 mL) was degassed and purged with N2 for 3 times, and thereafter was stirred at 115 °C for 2 hours under N2 atmosphere. LCMS (not shown) showed that Compound 24 was consumed and a desired mass was detected. The reaction mixture was filtered and the filtrate was concentrated to give a residue. Part of the residue (about 150 mg crude) was purified by prep-HPLC (column: Waters Xbridge 150x25mmx5pm; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 20%-50%, 9 minutes) to give TNK-135 (26.73 mg, 52.1 pmol, 9.69 % yield), as confirmed by LCMS (not shown) and NMR.
XH NMR (400 MHz, DMSO): 3 = 12.64-11.83 (m, 1H), 8.54 (d, J= 2.4 Hz, 1H), 8.14 (br s, 1H), 7.98-7.72 (m, 5H), 7.56-7.24 (m, 1H), 7.22-7.04 (m, 3H), 6.62-6.52 (m, 1H), 6.19 (br s, 1H), 5.98 (t, J= 5.6 Hz, 1H), 3.07 (q, J= 6.5 Hz, 2H), 2.60 (q, J= 7.5 Hz, 2H), 2.37-2.21 (m, 6H), 1.60-1.53 (m, 2H), 1.49 (quin, J= 5.5 Hz, 4H), 1.37 (br d, J= 5.0 Hz, 2H), 1.15 (t, J= 7.5 Hz, 3H).
The remaining part of the residue (about 100 mg) was purified by preparative HPLC (FA condition; column: Phenomenex Synergi C18 150x25mmxl0pm; mobile phase: [water(0.225%FA)-ACN];B%:40%-70%,10 minutes) to give desired a compound (about 60 mg, crude) as a white solid, which was further separated on preparative HPLC (neutral conditiomcolumn: Waters Xbridge 150x25mmx5pm; mobile phase: [water(10 mM NH4HCO3)- ACN];B%: 20%-50%, 9 minutes) to give TNK-135 (13.91 mg, 27.1 pmol, 5.04 % yield), as confirmed by LCMS (not shown) and NMR. XH NMR (400 MHz, DMSO): 3 = 12.67-12.09 (m, 1H), 8.54 (d, J= 1.8 Hz, 1H), 8.13 (br s, 1H), 7.95-7.72 (m, 5H), 7.64-7.26 (m, 1H), 7.24-7.01 (m, 3H), 6.61-6.52 (m, 1H), 6.20 (br s, 1H), 5.98 (br t, J= 5.6 Hz, 1H), 3.07 (q, J= 6.6 Hz, 2H), 2.60 (q, J= 7.4 Hz, 2H), 2.36-2.21 (m, 6H), 1.60-1.53 (m, 2H), 1.48 (quin, J= 5.5 Hz, 4H), 1.41-1.33 (m, 2H), 1.15 (t, J= 7.5 Hz, 3H).
Additional designs:
Based on the synthetic pathways described herein, compounds corresponding to, for example, TNK-002 or TNK-007 and featuring other piperazine- or piperidine-containing moieties can be prepared.
Exemplary such compounds are TNK-124 and TNK-128 (see, FIG. 1A), prepared as follows.
Preparation of TNK-128:
A general synthetic pathway for preparing TNK-128 is shown in FIG. 8.
Preparation of compound b (see, FIG. 8):
Figure imgf000101_0001
To a solution ofNaH (1.15 gram, 28.7 mmol, 60% purity, 1.00 mol equivalent) in THF (50 mL) was added Compound b_l (5.00 grams, 28.7 mmol, 4.90 mL, 1.00 mol equivalent) at 0 °C. The mixture was stirred at 0 °C for 30 minutes, and Compound b_0 (10.7 grams, 57.4 mmol, 4.33 mL, 2.00 mol equivalent) was thereafter added. The resulting mixture was heated to 80 °C and stirred at that temperature for 3 hours. TLC (petroleum ether/ethyl acetate = 10/1, L) showed that Compound b_l (Rf = 0.46) was consumed and a major new spot (Rf = 0.48) was formed. The reaction mixture was quenched by water (30 mL), and extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed by brine (50 mL) and dried over Na2SO4, filtered and concentrated under vacuum to get a residue. The residue was purified by column chromatography (SiC>2, petroleum ether/ethyl acetate = 1/0-50/1, Rf = 0.48) to give Compound b (5.80 grams, 20.6 mmol, 71.8 % yield) as light yellow liquid, as confirmed by 'H-N R.
E-NMR (400 MHz, CDCh): 3 4= .19 (q, J= 7.15 Hz, 4H), 3.38 (t, J= 8.2 Hz, 2 H), 2.44 (t, J= 8.4 Hz, 2H), 1.44 (s, 3H), 1.25 (t, J= 7 Hz, 6H). Preparation of Compound 12 (see, FIG. 8):
Figure imgf000102_0001
11 12
To a solution of compound 11 (3.00 grams, 24.5 mmol, 2.97 mL, 1.00 mol equivalent) in DCM (9 mL) and MeOH (6 mL) was added tetrabutylammonium tribromide (11.8 grams, 24.5 mmol, 1.00 mol equivalent). The resulting mixture was stirred at 20 °C for 1 hour. The reaction mixture was then washed with HC1 (1 M, 50 mL), water (50 mL), and brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give compound 12 (4.00 grams, 19.8 mmol, 81.0 % yield) as a yellow liquid.
Preparation of Compound 33 (see, FIG. 8):
Figure imgf000102_0002
Boc
12 33
To a solution of Compound a (2.50 grams, 11.2 mmol, 1.00 mol equivalent) in DMF (25 mL) were added TEA (1.70 gram, 16.8 mmol, 2.34 mL, 1.50 mol equivalent) and CDI (1.82 gram, 11.2 mmol, 1.00 mol equivalent) at 0 °C and the obtained mixture was stirred at 20 °C for 1 hour. Then compound 12 (2.26 grams, 11.2 mmol, 1.00 mol equivalent) and DBU (1.71 gram, 11.2 mmol, 1.69 mL, 1.00 mol equivalent) were added, and the resulting mixture was stirred at 70 °C for 2 hours. LC-MS showed a desired mass (RT = 1.074). The reaction mixture was diluted with ethyl acetate (150 mL), washed with water (100 mL) and brine (100 mL x 2), dried over Na2SO4, filtered and concentrated under vacuum to give compound 33 (4.50 grams, crude) as a yellow oil. Preparation of compound 34 (see, FIG. 8):
Figure imgf000103_0001
To a solution of Compound 33 (4.50 grams, 10.8 mmol, 1.00 mol equivalent) in DCM (20 mL) was added TFA (15.4 grams, 135 mmol, 10.0 mL, 12.4 mol equivalents) and the reaction mixture was stirred at 20 °C for 2 hours. TLC (dichloromethane/methanol = 10/1, 12) showed that Compound 33 (Rf = 0.89) was consumed and a major new spot (Rf = 0.18) was formed. The mixture was basified with saturated aqueous NaHCOs to pH of about 8, and extracted with dichloromethane (30 mL x 3). The combined organic phase was washed by brine (50 mL) and dried over Na2SO4, filtered and concentrated under vacuum to get a residue. The residue was purified by column chromatography (SiCL, dichloromethane/methanol = 1/0-5/1, Rf = 0.18) to give Compound 34 (1.60 gram, 4.78 mmol, 43.9 % yield, 93.6 % purity) as a yellow oil.
Preparation of Compound 36 (see, FIG. 8):
Figure imgf000103_0002
To a solution of Compound 34 (668 mg, 2.13 mmol, 1.00 mol equivalent) in THF (1.8 mL) were added TEA (1.08 gram, 10.6 mmol, 1.49 mL, 5.00 mol equivalent) and compound b (1.80 gram, 6.40 mmol, 3.00 mol equivalent). The mixture was stirred at 70 °C for 4 hours. TLC (ethyl acetate/petroleum ether = 2/1) indicated residual amount of Compound 34 (Rf = 0.01) and appearance of a new spot (Rf = 0.60). The mixture was diluted with ethyl acetate (50 mL), washed with water (50 mL), and brine (50 mL x 2), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (SiCh, petroleum ether/ethyl acetate = 30/1-1/1, Rf = 0.60) to get Compound 36 (500 mg, 869 pmol, 40.7 % yield, 89.3 % purity) as a yellow oil, as confirmed by LCMS (not shown).
Preparation of Compound 37 (see, FIG. 8):
Figure imgf000104_0001
To a mixture of Compound 36 (1.70 gram, 3.05 mmol, 92.1 % purity, 1.00 mol equivalent) in t-BuOH (15 mL) and MeOH (3 mL) were added NaBFL (520 mg, 13.7 mmol, 4.51 mol equivalents) and CaCL (50.0 mg, 450 pmol, 1.48xl0-1 mol equivalent). The reaction mixture was stirred at 80 °C for 18 hours. LCMS (not shown) showed residual amount of Compound 36 (RT = 0.817) and the appearance of the desired mass (RT = 0.705). The reaction mixture was quenched by saturated aqueous NH4CI (80 mL), and extracted with ethyl acetate (50 mL x 3). The combined organic phase washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under vacuum to give a residue, which was purified by column chromatography (SiCL, dichloromethane/methanol = 1/0-10/1, TLC: dichloromethane/methanol = 10/1, Rf = 0.42, 12) to give Compound 37 (450 mg, 1.05 mmol, 34.3 % yield) as yellow oil.
Preparation of Compound 38 (see, FIG. 8):
Figure imgf000104_0002
To a solution of Compound 37 (450 mg, 1.05 mmol, 1.00 mol equivalent) in DCM (2 mL) were added BOC2O (1.14 gram, 5.24 mmol, 1.20 mL, 5.00 mol equivalent) and Mg(C104)2 (23.3 mg, 104 pmol, 0.10 mol equivalent). The mixture was stirred at 40 °C for 72 hours. LCMS (not shown) indicated appearance of the desired mass (RT = 0.874). 8 mL Water was added and the mixture was extracted with dichloromethane (5 mL x 3). The combined organic phase was washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under vacuum to give Compound
38 (100 mg, 184 pmol, 17.6 % yield) as yellow oil.
Figure imgf000105_0001
A mixture of Compound 38 (100 mg, 184 pmol, 1.00 mol equivalent), Compound 15 (60.0 mg, 184 pmol, 1.00 mol equivalent), BrettPhos Pd G3 (8.37 mg, 9.23 pmol, 0.05 mol equivalent) and CS2CO3 (120 mg, 369 pmol, 2.00 mol equivalents) in dioxane (1 mL) was stirred at 90 °C for 1 hour under N2. TLC (petroleum ether/ethyl acetate = 2/1, I2) showed that Compound 38 (Rf = 0.66) was consumed and a major spot (Rf = 0.44) was formed. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL x 3). The combined organic phase was washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under vacuum to give a residue. The residue was purified by column chromatography (SiCL, petroleum ether/ethyl acetate = 10/1-0/1, Rf = 0.44) to give Compound 39 (60.0 mg, 64.8 pmol, 35.1 % yield, 85 % purity) as yellow oil, as confirmed by LCMS (not shown). Preparation of TNK-128 (see, FIG. 8):
Figure imgf000106_0001
To a solution of Compound 39 (60.0 mg, 64.8 pmol, 85 % purity, 1.00 mol equivalent) in THF (2 mL) was added TBAF (1 M, 648.85 pL, 10.0 mol equivalent) and the mixture was stirred at 78 °C for 1 hour. LCMS (not shown) showed that Compound 39 was consumed and the desired mass (RT = 0.872) was detected. The mixture was diluted with water (8 mL) and extracted with ethyl acetate (5 mL x 3). The combined organic phase was washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under vacuum to get a residue. The residue was purified by reversed-phase HPLC (Eluent of 0-50% MeCN/0.1% FA aq. gradient) to give TNK-128 (22.12 mg, 30.5 pmol, 47.0 % yield, 94.7 % purity) as an off-white solid, as confirmed by LCMS (not shown) and 1 H-NMR.
XH NMR (400 MHz, DMSO): 3 = 8.55 (
Figure imgf000106_0002
2.50 Hz, 1H), 7.87-7.95 (m, 2H), 7.80-7.87
(m, 2H), 7.77 (d, J = 1.50 Hz, 1H), 7.38 (s, 1H), 6.76-6.88 (m, 2H), 6.52-6.61 (m, 1H), 6.34 (s, 1H), 3.52-3.58 (m, 2H), 3.27 (br s, 4H), 3.05 (s, 4H), 2.61-2.65 (m, 2H), 2.39 (br s, 4H), 1.36-1.42 (m, 2H), 1.17 (t, J= 7.50 Hz, 3H), 1.11 (s, 18H), 0.80 (s, 3H).
EXAMPLE 2
In Vitro ADME characterization
The properties of TNK-108 were tested and compared to those of the parent compound TNK-002 (also referred to herein as KY-08405), according to the following protocols.
Inhibition of TNIK kinase activity:
TNIK Kinase Assay is a luminescent kinase assay that measures ADP formed from TNIK kinase reaction. The assay uses ADP-Glo™ Kinase Assay (Promega Cat# V910) and TNIK Kinase Enzyme System (Promega cat# V4158).
Briefly, 20 ng TNIK (kinase domain) are incubated with 30 pM MBP (maltose binding protein) and 100 pM ATP. Tested compounds in DMSO are added at an eleven-point dilution curve in an independent triplicate. Final DMSO concentration is 1 %. ADP formed in the reaction during 1 hour incubation at 37 °C, is converted into ATP, which in turn is converted into light by Ultra-Gio™ Luciferase. The luminescent signal obtained positively correlates with ADP amount and TNIK kinase activity. IC50 for compound inhibition is obtained from a 4-parameter logistic curve using GraphPad Prism software.
CellTiter Gio assay:
The CellTiter-Glo® Luminescent Cell Viability Assay (Promega Cat# G7570) is used to determine the number of viable cells in culture based on quantitation of the ATP present, which signals the presence of metabolically active cells.
Briefly, 5000 SW620 cells/well are seeded in a 96 well plate. 24 hours later, tested compounds in DMSO are added at a nine-point dilution curve in medium with 0.1% FBS and 1% DMSO. Compounds are tested in an independent triplicate. After 48 hours incubation at 37 °C, CellTiter Gio reagent is added and luminescence is recorded. A direct relationship exists between luminescence measured with the CellTiter-Glo® Assay and the number of cells in culture. IC50 for compound inhibition is obtained from a 4-parameter logistic curve using GraphPad Prism software.
Kinetic Solubility:
The tested compound (10 mM) in DMSO 10 pL, was added into the lower chamber of Whatman miniuniprep vials. 490 pL of a selected phosphate buffer (pH 3.5, 4.5 or 7.4) and 0.1N HC1 were added thereto and the samples were vortexed for 2 minutes. The vials were then shaked on a Barnstead shaker for 24 hours at room temperature, at a speed of 800 rpm, and were thereafter centrifuged for 20 minutes (at about 4000 rpm). The miniunipreps were then compressed to prepare the filtrate for injection into HPLC, and concentrations were calculated. HPLC analyses were performed using HPLC-AW(19#139), equipped with Waters XBridge C18 4.6*100 mm column. Detector was operated at 282 nm. Flow rate: 1.4 mL/minute. Mobile phase A: 0.1% TFA in water. Mobile phase B: 0.1 % TFA in CAN. Gradient: from 70:30 to 30:70 in 5 minutes.
Microsomal stability:
Preparation of working solutions:
Compound solutions. An intermediate solution was prepared by diluting 5 pL of a stock solution containing 10 mM of the tested compound in dimethyl sulfoxide (DMSO)) with 495 pL of methanol (MeOH). The intermediate solution concentration was 100 pM in 99 % MeOH).
50 pL of the intermediate solution (100 pM) were diluted with 450 pL of 100 mM potassium phosphate buffer, to thereby obtain a working solution in which compound’s concentration is 10 pM, 9.9% MeOH.
NADPH solution: P-Nicotinamide adenine dinucleotide phosphate reduced form, tetrasodium salt; NADPH 4Na (Vendor: Chem-Impex International, Cat. No. 00616) powder was weighed and diluted into a 10 mM MgCb solution, to thereby obtained an NADPH working solution at a concentration of 10 unit/mL. The final concentration of NADPH in in the reaction system is 1 unit/mL.
Microsome solutions: Human liver microsomes (HLM) were obtained from Corning (Cat No. 452117; Lot No. 38292). CD-I mouse microsomes (MLM) were obtained from Biopredic (CatNo. BQM1000; LotNo. MIC255036). The appropriate concentrations of microsome working solutions were prepared in 100 mM potassium phosphate buffer.
Stop Solution: Cold (4 °C) acetonitrile (ACN) containing 100 ng/mL tolbutamide and 100 ng/mL labetalol as internal standards (IS) was used as the stop solution.
Assay Protocol:
Using an Apricot automation workstation, 10 pL/well of the tested compound working solution were added to all 96-well reaction plates except the blank (TO, T5, T10, T20, T30, T60, and NCF60). An Apricot automation workstation was used to add 80 pL/well of microsome solution to all reaction plates (Blank, TO, T5, T10, T20, T30, T60, and NCF60). All reaction plates containing mixtures of compound and microsomes were pre-incubated at 37 °C for 10 minutes. An Apricot automation workstation was used to add 10 pL/well of 100 mM potassium phosphate buffer to reaction plate NCF60. Reaction plate NCF60 was incubated at 37 °C, and timer 1 was started.
After pre-incubation, an Apricot automation workstation was used to add 10 pL/well of NADPH regenerating system to every reaction plate except NCF60 (Blank, TO, T5, T10, T20, T30, and T60) to start the reaction. The reaction plates were incubated at 37 °C, and timer 2 was started.
The final concentration of microsomes in the incubation medium was 0.5 mg protein/mL. The final concentration of a tested or control compound in the incubation medium was 1 pM. The final concentration of MeOH in the incubation medium was 0.99 %. The final concentration of DMSO in the incubation medium was 0.01 %.
An Apricot automation workstation was used to add 300 pL/well of stop solution to each reaction plate at its appropriate end time point (5, 10, 20, 30 or 60 minutes) to terminate the reaction. For TO, stop solution was added prior to the addition of microsomes and NADPH. Each plate was sealed and shaken for 10 minutes. After shaking, each plate was centrifuged at 4000 rpm and 4 °C for 20 minutes. During centrifugation, an Apricot automation workstation was used to add 300 pL/well of HPLC grade water to eight new 96-well plates. After centrifugation, an Apricot automation workstation was used to transfer 100 pL of supernatant from each reaction plate to its corresponding bioanaylsis plate. Each bioanalysis plate was sealed and shaken for 10 minutes prior to LC-MS/MS analysis.
Data Analyses:
The following equation of first order kinetics was used to calculate T1/2 and CLint(mic) (pL/min/mg):
Figure imgf000109_0001
g g y g
Plasma stability:
The following compounds were tested: TNK-108; KY-08405 (TNK-002), TNK-128, TNK-127, and Propantheline bromide (positive control). 1 mM intermediate solution was prepared by diluting 10 pL of a stock solution (10 mM in DMSO) with 90 pL DMSO; 1 mM intermediate solution of positive control Propantheline was prepared by diluting 10 pL of a stock solution (10 mM in DMSO) with 90 pL ultrapure water. CD-I mouse and human plasma were tested. The pooled frozen plasma was thawed in a water bath at 37 °C prior to experiment. Plasma was centrifuged at 4000 rpm for 5 minutes and the clots, if present, were removed. The pH was adjusted to 7.4 ± 0.1 when required.
100 pM dosing solution was prepared by diluting 20 pL of the intermediate solution (1 mM) with 180 pL 45 % MeOH/H2O.
At each time point (0, 10, 30, 60 and 120 minutes), 400 pL of a stop solution (200 ng/mL tolbutamide and 200 ng/mL Labetalol in 50 % ACN/MeOH) was added to precipitate protein, and mixed thoroughly.
Sample plates were thereafter centrifuged at 4,000 rpm for 10 minutes. An aliquot of supernatant (50 pL) was transferred from each well and mixed with 100 pL ultrapure water. The samples were shaked at 800 rpm for about 10 minutes before being subjected to LC-MS/MS analysis.
The % remaining of test compound after incubation in plasma was calculated using following equation:
% Remaining= 100 x (PAR at appointed incubation time / PAR at To time) where PAR is the peak area ratio of analyte versus internal standard (IS), and the appointed incubation time points are To (0 minute), Tn (n=0, 10, 30, 60, 120 minutes).
LC-MS was performed as described hereinabove, equipped with ACQUITY UPLC BEH C18, 1.7 pm, 2.1x50 mm, while using as a mobile phase A: 0.1% formic acid in water; B: 0.1% formic acid in acetonitrile, at a flow rate of 700 pL/minute.
The properties of TNK-128, and of TNK-127 and TNK-135 and their counterpart TNK- 007 were also tested using the above protocols.
The obtained data is presented in Table 1 below.
As can be seen in Table 1, the biocleavable compounds TNK-108 and TNK-128 are inactive in the in vitro kinase assay, probably due to steric hindrance within the TNIK pocket. The biocleavable compounds become active in the low micromolar range in the cell-based assay after generating TNK-002 upon contacting intracellular esterases. TNK-108 exhibits improved solubility compared to the parent TNK-002 and TNK-128. TNK-108 is quite stable in mouse and human microsomes. It generates minute amounts of TNK-002 in mice plasma. TNK-128 generates substantial amount of TNK-002 in mice plasma.
TNK-127 and TNK-135 are less active in the kinase assay than TNK-007 while the activity of these 3 compounds in the CellTiter Gio assay is moderate. Both TNK-127 and TNK- 135 are significantly more soluble and more stable in microsomes than TNK-007 and are very stable in mouse and human plasma. Table 1
Figure imgf000111_0001
EXAMPLES
In Vivo pharmacokinetic studies The pharmacokinetic parameters of TNK-108 were tested and compared to those of TNK-
002 (KY-08405) and those of TNK-002 which was subjected to glucuronidation (KY-08405G).
C57BL/6 mice, male, N=18, 6-8 week-old, weighing about 20-21 grams, were purchased from Jihui Laboratory Animal Co. LTD, and were given free access to food and water.
Mice were divided into 4 treatment groups: IV: TNK-108, 15.6 mg/kg (5 mL/kg) via tail vein injection (N=3/time point)
PO: TNK-108, 31.2 mg/kg (10 mL/kg) via oral gavage (N=3/time point) IV: KY-08405, 10 mg/kg (5 mL/kg) via tail vein injection (N=3/time point) PO: KY-08405, 20 mg/kg (10 mL/kg) via oral gavage (N=3/time point)
The animals were restrained manually, and approximately 115 pL of blood/time point was collected into pre-cooled EDTA-K2 tubes via facial vein. 3 pL of dichlorvos (1 mg/mL) was added to 110 pL of blood immediately, and the sample was thereafter centrifuged at 2000 g for 5 minutes (4 °C) to obtain plasma within 15 minutes after sample collection. The following Table presents the design of the in vivo study conducted.
Figure imgf000112_0001
The plasma samples were analyzed by LC-MS, as described hereinabove. Concentrations of TNK-002, TNK-108 and glucuronidation metabolites thereof were determined.
FIG. 9 presents comparative plots showing the mean plasma concentration following a single IV or PO administration of TNK-108, and clearly show that the bioavailability of TNK- 108 is high, about 77%; and its plasma concentration after oral administration remains around the EC50 (determined in a cell-based assay) for many hours, indicating low clearance. The area under the curve (AUC) is high.
FIG. 10 presents comparative plots showing the mean plasma concentration following a single IV or PO administration of mol equivalent amounts of TNK-108 and TNK-002.
The measured oral bioavailability of TNK-002 was 1.3 %, much lower than that of TNK- 108. The AUC values are 4-5-folds lower for TNK-002 compared to TNK-108. The half-life in blood (Tl/2) measured for TNK-108 is 5.7 hours, and for TNK-002 4.2 hours.
FIG. 11 presents comparative plots showing the mean plasma concentration of a respective glucuronide metabolite following a single IV or PO of mol equivalent amounts of TNK-108 and TNK-002. As can be seen, much lower amounts of the glucuronide metabolite were observed following administration of TNK-108, compared to TNK-002. As can be further seen, following PO administration, the amount of the glucuronide metabolite of TNK-002 is higher by several orders of magnitude compared to that of TNK-108.
Similar assays were conducted with TNK-128. The obtained data is presented in FIGs. 12A-B. As can be seen, plasma concentrations of TNK-128 are higher than those of TNK-002, both when administered orally and IV.
Similar assays were conducted with TNK-127, and the obtained data is presented in FIG. 13. As can be seen, TNK-127 exhibits higher plasma concentrations upon IV administration, and for a longer time period. EXAMPLE 4
In vivo activity studies
In vivo studies were conducted for evaluating the anti -turn or efficacy of TNK-108, and TNK- 128 in the subcutaneous SW620 human colorectal cancer xenograft model in female BALB/c Nude mice.
Animals:
Female BALB/c nude mice (Mus miiscuhis). aged 6-8 weeks, and weighing 18-22 grams were kept in individual ventilation cages at constant temperature (20-26 °C) and humidity (40- 70 %) with 5 animals in each cage. Animals had free access to irradiation sterilized dry granule food and to sterile drinking water during the entire study period.
Cell Culture’.
SW620 tumor cells were maintained in vitro as a monolayer culture in Leibovitz's L-15 medium supplemented with 10 % heat inactivated fetal bovine serum, 100 U/mL Penicillin and 100 pg/mL Streptomycin at 37 °C in an atmosphere of 0 % CO2 in air. The tumor cells were routinely subcultured twice weekly by trypsin-EDTA treatment. The cells growing in an exponential growth phase were harvested and counted for tumor inoculation.
Tumor Inoculation and Animal Grouping:
Each mouse was inoculated subcutaneously at the right flank with SW620 cancer cells (5 x 106) in 0.1 mL of PBS for tumor development. Treatments were started on day 14 after tumor inoculation when the average tumor size reached approximately 162 mm3. The animals were assigned into groups according to a sorting SOP based upon their tumor volumes. Each group consisted of 10 tumor-bearing mice.
Tested Preparations:
Vehicle 1 includes 10 % propylene glycol (PG), 10 % Cremophor and 80 % or a 5 %HP-P- CD aqueous solution.
Vehicle 2 includes 20 % PEG400, 5 % Tween®80 and 75 % or a 5 %HP-P-CD aqueous solution.
TNK-128 preparations at a concentration of 3.75, 7.5, or 15, mg/ml, were prepared by dissolving TNK-128 in PG, adding Cremophor EL to the solution and stirring until a homogeneous mixture is obtained, and thereafter adding 5 % HP-P-CD in water and vortexing for 1 minute, for obtaining a clear solution.
TNK-108 preparations at a concentration of 3.5 or 5, mg/ml, were prepared by dissolving TNK-108 in PEG400, adding Tween®80 to the solution and stirring until a homogeneous mixture is obtained, and thereafter adding 5 % HP-P-CD in water and vortexing for 1 minute, for obtaining a clear solution.
Observations:
At the time of routine monitoring, the animals were daily checked for any effects of tumor growth and treatments on normal behavior such as mobility, food and water consumption (by looking only), body weight gain/loss (body weights were measured twice per week), eye/hair matting and any other abnormal effect as stated in the protocol. Death and observed clinical signs were recorded on the basis of the numbers of animals within each subset.
Tumor Measurements and Endpoints:
The major endpoint was to see if the tumor growth could be delayed or mice could be cured. Tumor size was measured twice weekly in two dimensions using a caliper, and the volume was expressed in mm3 using the formula: V = 0.5 a x b2 where a and b are the long and short diameters of the tumor, respectively. The tumor size was then used for calculation of RTV and T/C values. T/C was calculated for each group using the formula: T/C(%)=TRTV / CRTV x 100 %; TRTV was the average RTV of treatment group, while CRTV is the average RTV of control group on the same day. RTV for each tumor was calculated as RTV = Vt / Vi, Vtis the tumor volume on a given day, Vi is the tumor volume on the day of treatment start for the same tumor.
TGI was calculated for each group using the formula: TGI (%) = [l-(Ti-TO)/ (Vi-V0)] x 100; Ti is the average tumor volume of a treatment group on a given day, TO is the average tumor volume of the treatment group on the day of treatment start, Vi is the average tumor volume of the vehicle control group on the same day with Ti, and V0 is the average tumor volume of the vehicle group on the day of treatment start.
Tumor weight was measured at the study termination.
Statistical Analysis:
Statistical analysis of difference in the tumor volume among the groups were conducted on the data obtained at day 25 after the start of treatment before animals with tumor volumes larger than 3000 mm3 started to be sacrificed. A one-way ANOVA analysis was performed for all 10 groups, and when a significant F-statistics (a ratio of treatment variance to the error variance) was obtained, comparisons between groups were carried out with Games-Howell test. Two-tailed unpaired T-test (day 25), and two-way ANOVA with repeated measures (multiple time points) were performed for pairwise comparisons to complement the statistical analysis.
Results:
FIGs. 14A-B are bar graphs showing the average tumor volume at day 25 of animals treated with a TNK-128 preparation at the indicated dose or with the respective vehicle (FIG. 14 A) and of animals treated with a TNK-108 preparation, at the indicated dose, and of the respective vehicle (FIG. 14B), and show that treatment with both compounds resulted in reduction in tumor volume.
Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims.
All publications, patents and patent applications mentioned in this specification are herein incorporated in their entirety by reference into the specification, to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention. To the extent that section headings are used, they should not be construed as necessarily limiting. In addition, any priority document(s) of this application is/are hereby incorporated herein by reference in its/their entirety.

Claims

WHAT IS CLAIMED IS:
1. A compound represented by Formula I:
Figure imgf000116_0001
wherein:
Y is N or CR5;
Z is N or C s;
Ri, and R2 are each independently hydrogen, alkyl or cycloalkyl;
R3 and R4 are each independently hydrogen, alkyl, haloalkyl, or halo;
R5 and Re are each independently hydrogen, alkyl, halo, hydroxy, thiol, hydroxyalkyl, thioalkyl, alkoxy, thioalkoxy, carboxylate, amine, amide, or aryloxy;
B is hydrogen, alkyl, haloalkyl, halo, alkoxy, thioalkoxy, or cycloalkyl;
A is a substituted or unsubstituted aryl or heteroaryl, or, alternatively, forms with Ri a substituted or unsubstituted heteroalicyclic ring or forms with R3 a substituted or unsubstituted cyclic ring;
X is -O-C(=O)- or -NH-C(=O)-; and
W is a nitrogen-containing moiety attached to said X via said nitrogen atom.
2. The compound of claim 1, wherein Ri, and R2 are each hydrogen.
3. The compound of claim 1 or 2, wherein Y is CR5.
4. The compound of claim 3, wherein R5 is hydrogen.
5. The compound of any one of claims 1-4, wherein Z is CRs.
6. The compound of claim 5, wherein Rs is hydrogen.
7. The compound of any one of claims 1-6, wherein R4 is hydrogen.
8. The compound of any one of claims 1-7, wherein B is alkyl.
9. The compound of any one of claims 1-8, wherein X is -O-C(=O)-.
10. The compound of any one of claims 1-9, wherein A is a substituted or unsubstituted aryl.
11. The compound of claim 10, wherein A is a substituted aryl.
12. The compound of claim 11, wherein said aryl is substituted by a heteroaryl.
13. The compound of claim 12, wherein said aryl is substituted by pyrazole.
14. The compound of any one of claims 1-13, wherein W is or comprises a nitrogencontaining heteroalicyclic ring.
15. The compound of any one of claims 1-14, wherein W is a nitrogen-containing heteroalicyclic ring attached to said X via a nitrogen atom of said nitrogen-containing heteroalicyclic ring.
16. The compound of claim 15, wherein said nitrogen-containing heteroalicyclic ring is substituted by an alkyl, cycloalkyl or a heteroalicyclic.
17. The compound of any one of claims 1-12, wherein W is a substituted piperidinyl.
18. The compound of claim 17, wherein W is 1,4-bipiperidinyl.
19. The compound of claim 17, wherein said piperidinyl is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom.
20. The compound of any one of claims 1-12, wherein W is a substituted 1,4- piperazinyl. 116
21. The compound of claim 20, wherein said piperazinyl is substituted by a linear or branched hydrocarbon chain interrupted by at least one O atom.
22. The compound of any one of claims 1-13, wherein W is an aminoalkyl, attached to said X via said amine.
23. The compound of claim 22, wherein said aminoalkyl is substituted by said nitrogencontaining heteroalicyclic.
24. The compound of any one of claims 1-23, wherein A is phenyl substituted by pyrazole.
25. The compound of claim 24, wherein X is -O-C(=O)- and W is 1,4-bipiperidinyl.
26. The compound of claim 24, wherein X is -NH-C(=O)- and W is 1,4-bipiperidinyl.
27. The compound of claim 24, wherein X is -O-C(=O)- and W is 1,4-piperazinyl substituted by a linear hydrocarbon chain interrupted by at least one O atom.
28. The compound of claim 24, wherein X is -NH-C(=O)- and W is an aminoalkyl, attached to said X via said amine, and wherein said aminoalkyl is substituted by said nitrogencontaining heteroalicyclic.
29. A pharmaceutical composition comprising the compound of any one of claims 1- 28 and a pharmaceutically acceptable carrier.
30. The pharmaceutical composition of claim 29, being formulated for oral administration.
31. The compound of any one of claims 1-28, or the composition of claim 29 or 30, for use in treating a medical condition associated with upregulated activity of TNIK.
32. The compound or composition of claim 31, wherein said medical condition is cancer. 117
33. The compound or composition of claim 31 or 32, wherein said treating comprises orally administering the compound or the composition to a subject in need thereof.
34. A process of preparing the compound of Formula I according to any one of claims
1-28, the process comprising: reacting a compound of Formula Xa:
Figure imgf000119_0001
wherein Q is hydroxy, in case X is -O-C(=O)-, or amine, in case X is -NH-C(=O)-, and Li is a first reactive group; with a compound of Formula Xb:
W-L2
Formula Xb, wherein W-L2 is or comprises a second reactive group, under conditions that generate said X upon coupling said W-L2 with said Q, to thereby generate a compound of Formula Xc:
Figure imgf000119_0002
Formula Xc, and reacting the compound of Formula Xc with a compound of Formula Xd: 118
Figure imgf000120_0001
Formula Xd wherein L3 is a third reactive group that generates, upon reacting with Li, said -N(R.2)-, under conditions that generate said -N(R.2)- upon coupling said Li and L3, to thereby generate the compound of Formula I.
PCT/IL2021/050976 2020-08-10 2021-08-10 Traf2- AND Nck-INTERACTING KINASE (TNIK) INHIBITORS AND USES THEREOF WO2022034587A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063063491P 2020-08-10 2020-08-10
US63/063,491 2020-08-10

Publications (1)

Publication Number Publication Date
WO2022034587A1 true WO2022034587A1 (en) 2022-02-17

Family

ID=80247853

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2021/050976 WO2022034587A1 (en) 2020-08-10 2021-08-10 Traf2- AND Nck-INTERACTING KINASE (TNIK) INHIBITORS AND USES THEREOF

Country Status (1)

Country Link
WO (1) WO2022034587A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105294680A (en) * 2014-06-25 2016-02-03 中国药科大学 VEGFR-2 irreversible inhibitor and use thereof
WO2019156439A1 (en) * 2018-02-07 2019-08-15 Korea Research Institute Of Chemical Technology Compounds for inhibiting tnik and medical uses thereof
WO2019224790A2 (en) * 2018-05-24 2019-11-28 Kashiv Biosciences, Llc Prodrugs of fulvestrant

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105294680A (en) * 2014-06-25 2016-02-03 中国药科大学 VEGFR-2 irreversible inhibitor and use thereof
WO2019156439A1 (en) * 2018-02-07 2019-08-15 Korea Research Institute Of Chemical Technology Compounds for inhibiting tnik and medical uses thereof
WO2019224790A2 (en) * 2018-05-24 2019-11-28 Kashiv Biosciences, Llc Prodrugs of fulvestrant

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MATHIJSSEN RON, ALPHEN ROBBERT, VERWEIJ JAAP, LOOS WALTER, NOOTER KEES, STOTER GERRIT, SPARREBOOM ALEX: "Clinical Pharmacokinetics and Metabolism of Irinotecan (CPT-11)", CLINICAL CANCER RESEARCH, ASSOCIATION FOR CANCER RESEARCH, US, vol. 7, no. 8, 1 August 2001 (2001-08-01), US, pages 2182 - 2194, XP055908247, ISSN: 1078-0432 *
SATO H ET AL.: "Prediction of multiple binding modes of the CDK2 inhibitors, anilinopyrazoles, using the automated docking programs GOLD, FlexX, and LigandFit: an evaluation of performance", JOURNAL OF CHEMICAL INFORMATION AND MODELING, vol. 46, no. 6, 27 November 2006 (2006-11-27), pages 2552 - 62, XP055143558, DOI: 10.1021/ci600186b *

Similar Documents

Publication Publication Date Title
US10596174B2 (en) Pyrrolopyrimidine compounds as inhibitors of protein kinases
DK2880035T3 (en) Hitherto UNKNOWN PYRROLOPYRIMIDINE COMPOUNDS AS PROTEIN CHINAS INHIBITORS
AU2009227003B2 (en) Crystalline forms and two solvated forms of 4-amino-5-fluoro-3-[5-(4-methylpiperazin-1-yl)-1H-benzimidazol-2-yl]quinolin-2(1H)-one lactic acid salts
US9604935B2 (en) Deuterated phenyl amino pyrimidine compound and pharmaceutical composition containing the same
JP2020502065A (en) 8,9-dihydroimidazole [1,2-a] pyrimido [5,4-e] pyrimidine-5 (6H) -ketone compound
ES2716165T3 (en) (5,6-Dihydro) pyrimido [4,5-e] indolizines
US9586965B2 (en) Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases
AU2019297889A1 (en) Biaryl ether-type quinazoline derivatives
JP2023109924A (en) Dihydroimidazopyrazinone compound, composition including the compound, and use thereof
EP3101020B1 (en) Deuterated quinazolinone compound and pharmaceutical composition comprising same
CN102725288A (en) Method for manufacturing a 6-substituted-1-methyl-1H-benzimidazole derivative, and manufacturing intermediate from said method
WO2022034587A1 (en) Traf2- AND Nck-INTERACTING KINASE (TNIK) INHIBITORS AND USES THEREOF
TW202333667A (en) Pyrimidine or pyridine derivative and pharmaceutical use thereof
JP2023036957A (en) Diarylpyrazole compound, composition comprising the compound, and use thereof
CN108137614B (en) Fused pyrimidine compound, composition containing compound and application of compound
CN111303024B (en) Quinoline-structured pan-KIT kinase inhibitor and application thereof
JP7110335B2 (en) Pyridoquinazoline derivatives useful as protein kinase inhibitors
EP3766883A1 (en) Imidaxopyrolone compound and application thereof
CN116903627B (en) Imidazo triazine thiobenzamide derivative and preparation method and application thereof
CN108383849B (en) Imidazoquinazoline derivative and application thereof in anti-tumor and anti-inflammation
CA3228228A1 (en) Urea compound containing 2-heteroaromatic ring substitution, preparation method therefor and use thereof

Legal Events

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

Ref document number: 21855754

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: 21855754

Country of ref document: EP

Kind code of ref document: A1