WO2007054725A2 - Produit compose - Google Patents

Produit compose Download PDF

Info

Publication number
WO2007054725A2
WO2007054725A2 PCT/GB2006/004224 GB2006004224W WO2007054725A2 WO 2007054725 A2 WO2007054725 A2 WO 2007054725A2 GB 2006004224 W GB2006004224 W GB 2006004224W WO 2007054725 A2 WO2007054725 A2 WO 2007054725A2
Authority
WO
WIPO (PCT)
Prior art keywords
thiazol
pyrimidin
dimethyl
methyl
phenylamino
Prior art date
Application number
PCT/GB2006/004224
Other languages
English (en)
Other versions
WO2007054725A3 (fr
Inventor
Simon Green
Sheelagh Frame
Ian Fleming
Original Assignee
Cyclacel Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB0523040.4A external-priority patent/GB0523040D0/en
Priority claimed from GB0523160A external-priority patent/GB0523160D0/en
Application filed by Cyclacel Limited filed Critical Cyclacel Limited
Priority to EP06808516A priority Critical patent/EP1951307A2/fr
Publication of WO2007054725A2 publication Critical patent/WO2007054725A2/fr
Publication of WO2007054725A3 publication Critical patent/WO2007054725A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a pharmaceutical combination suitable for the treatment of proliferative disorders.
  • the present invention relates to combinations for the treatment of cancer, preferably non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • CDKs Cyclin-dependent kinases
  • CDKs are serine/threonine kinases that play a crucial regulatory role in the cell cycle. CDKs regulate cell cycle progression by phosphorylation of various proteins involved in DNA replication and cell division, including transcription factors and tumour suppressor proteins (Senderowicz, AM. Small-molecule cyclin-dependent kinase modulators, Oncogene, 2003; 22: 6609- 6620). Certain CDKs also play a role in the regulation of RNA synthesis by their involvement in the phosphorylation of the carboxy terminal domain (CTD) of the largest subunit of RNA polymerase II (pol II). It is not surprising, therefore, that CDKs have become attractive therapeutic targets.
  • CCD carboxy terminal domain
  • Cdc2 (also known as cdkl) is a catalytic sub-unit of a family of cyclin dependent kinases that are involved in cell cycle regulation.
  • kinases comprise at least two sub-units, namely a catalytic sub-unit (of which cdc2 is the prototype) and a regulatory sub-unit (cyclin).
  • the cdks are regulated by transitory association with a member of the cyclin family: cyclin A (cdc2, CDK2), ⁇ : cyclin B1-B3 (cdc2), cyclin C (CDK8), cyclin D1-D3 (CDK2-CDK4- CDK5-CDK6), cyclin E (CDK2), cyclin H (CDK7).
  • CDK activity is regulated by post-translatory modification, by transitory associations with other proteins and by modifications of their intra-cellular localization.
  • the CDK regulators comprise activators (cyclins, CDK7/cyclin H, cdc25 phosphateses), the p9.sup.CKS and pi 5. sup. CDK-BP sub-units, and the inhibiting proteins (pl6.sup.INK4A, pl5.sup.INK4B, p21.sup.Cipl, pl8, p27.sup.Kipl).
  • Roscovitine has been demonstrated to be a potent inhibitor of cyclin dependent kinase enzymes, particularly CDK2.
  • CDK inhibitors are understood to block passage of cells from the Gl /S and the G2/M phase of the cell cycle.
  • the pure R-enantiomer of roscovitine, seliciclib (R-Roscovitine; CYC202) has recently emerged as a potent inducer of apoptosis in a variety of tumour cells (McClue SJ, Blake D, Clarke R, et al,
  • Roscovitine has also been shown to be an inhibitor of retinoblastoma phosphorylation and therefore implicated as acting more potently on Rb positive tumors.
  • active pharmaceutical agents can often be administered in combination in order to optimise the treatment regime.
  • a CDK inhibitor in combination with a second chemotherapeutic agent is described in WO 03/077999, WO 03/082337, WO 2004/041262, WO 2004/041267, WO 2004/041268, WO 2004/041308, WO 2004/110455 and WO 2005/053699 (all to Cyclacel Limited).
  • the present invention seeks to provide a new combination of known pharmaceutical agents that is particularly suitable for the treatment of proliferative disorders, especially cancer. More specifically, a preferred aspect of the invention centres on combinations useful in the treatment of non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • a first aspect of the present invention relates to a combination comprising a histone deacetylase inhibitor and a compound of formula I or II, or a pharmaceutically acceptable salt thereof,
  • R 1 and R 2 are methyl, ethyl or isopropyl, and the other is H;
  • R 3 and R 4 are each independently H, branched or unbranched C 1 -C 6 alkyl, or aryl, and wherein at least one of R 3 and R 4 is other than H;
  • R 5 is a branched or unbranched Cj -C 5 alkyl group or a Ci-C 6 cycloalkyl group, each of which may be optionally substituted with one or more OH groups;
  • R 6 , R 7 , R 8 and R 9 are each independently H, halogen, NO 2 , OH, OMe, CN,
  • R 10 and R 14 are each independently H, C(OR' ) or a hydrocarbyl group optionally substituted by one or more R 15 groups;
  • R 11 , R 12 , and R 13 are each independently H, alkyl or alkenyl, each of which may be optionally substituted with one or more R 1 groups;
  • R 15 and R 16 are each independently halogen, NO 2 , CN, (CH 2 ) m OR a ,
  • R 8 , R h , R 1 and R j are each independently selected from alkyl, aryl, aralkyl and heteroaryl, each of which may be optionally substituted with one or more substituents selected from halogen, OH, NO 2 , NH 2 CF 3 and COOH; m, p, q and r are each independently O, 1, 2 or 3; n and s are each independently 1, 2, or 3; and
  • R a'n and R a "F are each independently H or alkyl.
  • a second aspect relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a combination according to the invention and a pharmaceutically acceptable carrier, diluent or excipient.
  • a third aspect relates to the use of a combination according to the invention in the preparation of a medicament for treating a proliferative disorder.
  • a fourth aspect relates to a pharmaceutical product comprising a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, and a HDAC inhibitor, as a combined preparation for simultaneous, sequential or separate use in therapy.
  • a fifth aspect relates to a method of treating a proliferative disorder, said method comprising simultaneously, sequentially or separately administering a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, and a HDAC inhibitor.
  • a sixth aspect relates to the use of a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for the treatment of a proliferative disorder, wherein said treatment comprises simultaneously, sequentially or separately administering a HDAC inhibitor.
  • a seventh aspect relates to the use of a HDAC inhibitor in the preparation of a medicament for the treatment of a proliferative disorder, wherein said treatment comprises simultaneously, sequentially or separately administering a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof.
  • An eighth aspect relates to the use of (i) a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, and (ii) a HDAC inhibitor, in the preparation of a medicament for treating a proliferative disorder.
  • a ninth aspect relates to the use of a HDAC inhibitor, in the preparation of a medicament for treating a proliferative disorder, wherein said medicament is for use in combination therapy with a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof.
  • a tenth aspect relates to the use of a HDAC inhibitor, in the preparation of a medicament for treating a proliferative disorder, wherein said medicament is for use in pretreatment therapy with a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof.
  • An eleventh aspect relates to the use of a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating a proliferative disorder, wherein said medicament is for use in combination therapy with a HDAC inhibitor.
  • a twelfth aspect relates to the use of a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating a proliferative disorder, wherein said medicament is for use in pretreatment therapy with a HDAC inhibitor.
  • a thirteenth aspect relates to the use of a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, and a HDAC inhibitor, in the preparation of a medicament for the treatment of non small cell lung cancer (NSCLC).
  • NSCLC non small cell lung cancer
  • the invention relates to a combination which comprises a histone deacetylase (HDAC) inhibitor and a compound of formula I, or a pharmaceutically acceptable salt thereof.
  • HDAC histone deacetylase
  • the invention provides a combination comprising a 2,6,9-substituted purine derivative of formula I, which exhibits improved resistance to metabolic deactivation, and a HDAC inhibitor.
  • one of R 1 and R 2 is ethyl or isopropyl, and the other is H.
  • R s is isopropyl or cyclopentyl.
  • R 6 , R 7 , R 8 and R 9 are all H.
  • R 1 or R 2 is ethyl and the other is H.
  • R 3 and R 4 are each independently H 5 methyl, ethyl, propyl, butyl or phenyl.
  • R 3 and R 4 are each independently H, methyl, ethyl, n-propyl, isopropyl, n-butyl, s-butyl, t-butyl or phenyl.
  • R 3 and R 4 are each independently H, methyl, ethyl, propyl or butyl.
  • R 3 and R 4 are each independently H, methyl, ethyl, n-propyl, isopropyl, n-butyl, s-butyl or t-butyl.
  • R 3 and R 4 are each independently H, methyl, ethyl, isopropyl or t-butyl.
  • said compound of formula I is selected from the following:
  • said compound of formula I is selected from the following: (26'3J?)-3- ⁇ 9-Isopropyl-6-[(pyridin-3-ylmethyl)-amino]-9H-purin-2-ylamino ⁇ -pentan-2- ol; (2i?3jS)-3- ⁇ 9-Isopropyl-6-[(pyridin-3-ylmethyl)-amino]-9H-purm-2-ylamino ⁇ -pentan-2- ol;
  • said compound of formula I is selected from the following: (3R)-3- ⁇ 9-isopropyl-6-[(pyridin-3-ylmethyl)-amino]-9H-purin-2-ylamino ⁇ -2-methyl- pentan-2-ol; (3 S)-3 - ⁇ 9-isopropy l-6-[(pyridin-3 -ylmethy l)-amino] -9H-purin-2-ylamino ⁇ -2-methyl- pentan-2-ol;
  • said compound of formula I is selected from the following:
  • the present invention provides a combination comprising a compound of formula II and a HDAC inhibitor.
  • One embodiment of the present invention relates to a combination comprising a compound of formula Ha, or a pharmaceutically acceptable salt thereof,
  • R 10 and R 14 are each independently H or a hydrocarbyl group optionally substituted by one or more R 15 groups;
  • R 11 , R 12 , and R 13 are each independently H, alkyl or alkenyl, each of which may be optionally substituted with one or more R groups;
  • R 15 and R 16 are each independently halogen, NO 2 , CN, (CH 2 ) m OR a , where m is 0, 1, 2 or 3, O(CH 2 ) n OR b , where n is 1, 2, or 3, NR c R d , CF 3 , COOR e , CONR f R g , COR h , SO 3 H 5 SO 2 R 1 , SO 2 NR j R k , heterocycloalkyl or heteroaryl, wherein said heterocycloalkyl and heteroaryl may be optionally substituted by one or more substituents selected from R m and COR n ; and
  • R a"n are each independently H or alkyl.
  • hydrocarbyl refers to a group comprising at least C and H. If the hydrocarbyl group comprises more than one C then those carbons need not necessarily be linked to each other. For example, at least two of the carbons may be linked via a suitable element or group. Thus, the hydrocarbyl group may contain heteroatoms. Suitable heteroatoms will be apparent to those skilled in the art and include, for instance, sulphur, nitrogen, oxygen, phosphorus and silicon.
  • the hydrocarbyl group is an aryl, heteroaryl, alkyl, cycloalkyl, aralkyl or alkenyl group.
  • alkyl includes both saturated straight chain and branched alkyl groups which may be substituted (mono- or poly-) or unsubstituted.
  • the alkyl group is a Ci -20 alkyl group, more preferably a Ci. 15 , more preferably still a Ci -J2 alkyl group, more preferably still, a Cj -6 alkyl group, more preferably a C1. 3 alkyl group.
  • Particularly preferred alkyl groups include, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl and hexyl.
  • Suitable substituents include, for example, one or more R 5 groups.
  • cycloalkyl refers to a cyclic alkyl group which may be substituted (mono- or poly-) or unsubstituted.
  • the cycloalkyl group is a C3-12 cycloalkyl group.
  • Suitable substituents include, for example, one or more R 15 groups.
  • heterocycloalkyl refers to a cycloalkyl group containing one or more heteroatoms selected from O, N and S.
  • heterocycloalkyl include 1- (1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, pyrrolidinyl, dihydrofuranyl, tetrahydropyranyl, pyranyl, thiopyranyl, aziridinyl, oxiranyl, methylenedioxyl, chromenyl, isoxazolidinyl, l,3-oxazolidin-3-yl, isothiazo
  • heterocycloalkyl a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule.
  • connection of said heterocycloalkyl rings is through a carbon or a sp 3 hybridized nitrogen heteroatom.
  • Preferred heterocycloalkyl groups include piperazine, morpholine, piperidine and pyrrolidine.
  • alkenyl refers to a group containing one or more carbon- carbon double bonds, which may be branched or unbranched, substituted (mono- or poly-) or unsubstituted.
  • the alkenyl group is a C 2-20 alkenyl group, more preferably a C 2-15 alkenyl group, more preferably still a C 2-I2 alkenyl group, or preferably a C 2-6 alkenyl group, more preferably a C 2 - 3 alkenyl group.
  • Suitable substituents include, for example, one or more R 15 groups as defined above.
  • aryl refers to a Q ⁇ -n aromatic group which may be substituted (mono- or poly-) or unsubstituted. Typical examples include phenyl and naphthyl etc. Suitable substituents include, for example, one or more R 15 groups.
  • heteroaryl refers to a C 4-12 aromatic, substituted (mono- or poly-) or unsubstituted group, which comprises one or more heteroatoms.
  • Preferred heteroaryl groups include pyrrole, pyrazole, pyrimidine, pyrazine, pyridine, quinoline, triazole, tetrazole, thiophene and furan.
  • suitable substituents include, for example, one or more R 15 groups.
  • R 8 , R h , R 1 and R* are each independently selected from alkyl, phenyl, benzyl and pyridyl, each of which may be optionally substituted with one or more substituents selected from halogen, OH, NO 2 , NH 2 CF 3 and COOH;
  • R a"n and R a "f are each independently H, methyl, ethyl or isopropyl.
  • R 10 and R 14 are each independently H or a C 1-20 hydrocarbyl group optionally comprising up to six heteroatoms selected from from N, O, and S, and which is optionally substituted by one, two or three R 15 groups;
  • R 14 is aryl or heteroaryl, each of which may be optionally substituted by one or more R 15 groups.
  • R 14 is H, CO(R J ), aryl or heteroaryl, wherein said aryl or heteroaryl groups may be optionally substituted by one or more R 15 groups.
  • R 14 is H, COMe, phenyl or pyridyl, wherein said phenyl or pyridyl groups may be optionally substituted by one or more R 15 groups.
  • R 14 is phenyl or pyridinyl, each of which may be optionally substituted by one or more R 15 groups.
  • R 10 is H or alkyl. More preferably, R 10 is H, methyl, ethyl or 3-methylbutyl.
  • R 11 , R 12 , and R 13 are each independently H, C 1 -C 6 alkyl or C 2 -C 6 alkenyl, each of which may be optionally substituted with one, two or three R 16 groups.
  • R is C 1-6 alkyl. More preferably still, R 11 is methyl.
  • R 12 and R 13 are both H.
  • R 15 and R 16 are each independently F, Cl, Br, I, NO 2 , CN 3 OH, OMe, OEt, CH 2 OH, O(CH 2 ) 2 OMe, NH 2 , NHMe, NMe 2 , CF 3 , COOH, CONH 2 , CONHMe, CONMe 2 , COMe, SO 3 H, SO 2 Me, SO 2 NH 2 , SO 2 NHMe, SO 2 NMe 2 , morpholine, piperidine, piperazine, N-acetylpiperazine, N-methylpiperazine, triazole, or tetrazole.
  • R 12 and R 13 are both H and R 11 is Me.
  • the compound of the invention is of formula III, or a pharmaceutically acceptable salt thereof,
  • R 10 is as defined above in claim 1 or claim 12;
  • X is C; or X is N and R 17 is absent;
  • R , 1 1 7 ', ⁇ R> 1 l 8 iS ⁇ R» 1 i 9 y and R >2 z 0 u are each independently H or as defined for R 15 and R , 16
  • R 10 is H or alkyl
  • R 17 is H, NO 2 , OR", halogen, CF 3 , CN, COR q , alkyl, NR r R s , 0(CHa) 1 OR 1 ;
  • R 18 is H, OR U , halogen, alkyl, NR V R W , or a heterocycloalkyl optionally substituted with one or more substituents selected from R m and C0R n ;
  • t is O, 1, 2 or 3;
  • R 19 is H, alkyl or NR x R y ; and R p"y are each independently H or alkyl.
  • R 10 is H, Me, Et or 3-methylbutyl.
  • R 17 is H, NO 2 , OH, Me, I, CF 3 , CN, CH 2 OH, CO 2 H, CO 2 Me OrNH 2 ;
  • R 18 is H, F, OH, I, Cl, Br, OMe, NMe 2 , morpholine, Me, N-methylpiperazine, N- acetylpiperazine or piperazine; and R 19 is H, Me or NMe 2 .
  • R 17 is selected from H, NO 2 , halogen, CN, CF 3 , SO 3 H, (CH 2 ) m OR a , COOR e , (CH 2 ) p NR c R d , (CH 2 ) r NR b' SO 2 R h' , (CH 2 ) q NR a' C0R gl , SO 2 NR j R k , C0NR f R g , SO 2 NR e' (CH 2 ) s OR c> , S0 2 NR d R' and heterocycloalkyl optionally substituted by one or more COR" or sulfonyl groups.
  • R 17 is selected from H, NO 2 , OH, Me, I, CN, CH 2 OH, CF 3 , CO 2 H, CO 2 Me, NH 2 , Cl, 4-acetylpiperazin-l-yl, OMe, SO 3 H, CH 2 NHSO 2 Me, CH 2 NHCOPh, CH 2 NHSO 2 CF 3 , SO 2 NH 2 , CONH 1 Pr, SO 2 NHEt, SO 2 NH(CH 2 ) 2 OMe, SO 2 NH 1 Pr, SO 2 NH(CH 2 ) 2 OH, NHMe, SO 2 NH-benzyl and morpholin-4-sulfonyl.
  • R 18 is selected from H, NO 2 , SO 3 H, halogen, (CH 2 ) m OR a , (CH 2 ) p NR c R d , (CH 2 ) q NR a' COR s> , SO 2 NR e' (CH 2 ) s OR c> , SO 2 NR d> R' ' and heterocycloalkyl optionally substituted by one or more COR", R m or aralkyl groups.
  • R 18 is selected from H, F, OH, Cl, Br, OMe, NMe 2 , morpholin-4-yl, 4-methylpiperazin-l-yl, Me, 4-acetyl-piperazin-l-yl, I, CH 2 NHCOMe, NO 2 , SO 3 H, SO 2 NH(CH 2 ) 2 OMe, 4-benzylpiperazin-l-yl, SO 2 NH(CH 2 ) 2 OH, SO 2 NH-benzyl, CH 2 NH 2 , CH 2 NHCO-(pyrid-2-yl) and piperazin-1-yl.
  • R 19 is selected from H, R f and (CH 2 ) p NR 0 R d .
  • R 19 is selected from H, Me and NMe 2 .
  • R 20 is selected from H 5 R f , CF 3 , halogen and (CH 2 ) q NR a COR g> .
  • R 20 is selected from H, NHCOMe, CF 3 , Br and Me.
  • X is N and R 1 is absent.
  • X is C.
  • the compound of formula II is selected from the following:
  • the compound of formula II is selected from the following:
  • said compound of formula II is 3,4-dimethyl-5 ⁇ [2-(4-piperazin-l-yl-phenylamino)pyrimidin-4-yl]-3H-thiazol-2-one.
  • the presently claimed combinations comprise a compound of formula I or formula II and a histone deacetylase (HDAC) inhibitor.
  • HDAC histone deacetylase
  • Histones are small positively charged proteins that are rich in basic amino acids (positively charged at physiological pH). There are five main types of histones namely, Hl, H2A, H2B, H3, and H4 which exhibit a high degree of structural similarity. Histones are not found in eubacteria (e.g., E. coli), although the DNA of these bacteria is associated with other proteins that presumably function like histones to package the DNA within the bacterial cell. Archaebacteria, however, do contain histones that package their DNAs in structures similar to eukaryotic chromatin (G. M. Cooper, "The Cell - A Molecular Approach", 2 nd Edition, Chapter II). The majority of histones are synthesized during the S phase of the cell cycle, and newly synthesized histones quickly enter the nucleus to become associated with DNA. Within minutes of its synthesis, new DNA becomes associated with histones in nucleosomal structures.
  • eubacteria e.g., E. coli
  • the amino-terminal tail domains of histones may be enzymatically modified by post- translational addition of methyl (to lysine and arginine groups), acetyl (to lysine groups), or phosphate groups (to serine groups) (Spencer et al, Gene, 1999, 240(1), 1). This results in a reduction of the net positive charge of the histone which, consequently, may weaken the binding of the histone to DNA.
  • HDACs histone deacetylators
  • HDACs therefore, are believed to be associated with a number of different diseases which include proliferative disorders such as leukemia (Lin et al, Nature, 1998, 391, 811), melanomas/squamous cell carcinomas (Gillenwater et al, Int. J. Cancer, 1998, 75217; Saunders et al, Cancer Res., 1999, 59, 399), breast cancer, prostrate cancer, bladder cancer (Gelmetti et al, MoI. Cell Biol., 1998, 18, 7185; Wang et al, PNAS, 1998, 951, 10860) and colon cancer (C. A. Hassig, et al, 1997, Chem. Biol., 4, 783; S. Y. Archer et al, PNAS, 1998, 95(12), 6791).
  • proliferative disorders such as leukemia (Lin et al, Nature, 1998, 391, 811), melanomas/squamous cell carcinomas (Gillenwater e
  • US 2005/0004007 discloses a method for promoting apoptosis in cancer cells which involves administering a cyclin dependent kinase inhibitor and an agent which induces cellular differentiation.
  • agent which induces cellular differentiation namely, histone deacetylase inhibitors, protein kinase C, retinoids and vitamin D3.
  • combinations comprising a compound of formula I or formula II and a HDAC inhibitor are not specifically disclosed, nor is the use of this combination in the treatment of solid tumours, such as NSCLC.
  • the exemplification of US 2005/0004007 is limited to combinations of flavopiridol with selected HDAC inhibitors tested on leukemia cell lines. Accordingly, to date, there has been no disclosure of the specific combinations claimed in the present application, let alone any suggestion that they would be therapeutically useful in the treatment of lung cancers such as NSCLC.
  • the HDAC inhibitor is sodium butyrate.
  • the HDAC inhibitor is a prodrug of sodium butyrate.
  • the prodrug is pivaloyloxymethyl butyrate.
  • Pivaloyloxymethyl butyrate (Pivanex ® ) is an acyloxyalkyl ester prodrug of butyric acid and has been shown to induce the instrinsic pathway of apoptosis in leukemia and neuroblastoma cells (S. Mei et al, International Journal of Oncology, 2004, 25, 1509).
  • the HDAC inhibitor is trichostatin A (TSA).
  • Trichostatin A is a specific and reversible inhibitor of HDAC. At nanomolar concentrations, TSA causes a marked accumulation of highly acetylated histones in vivo and strongly inhibits the activity of the partially purified histone deacetylase in vitro (M. Yoshida et al, J. Biol. Chem., 1990, 265(28), 17174).
  • TSA arrests cell cycle progression in Gl and inhibits the activity of the HDl deacetylase with an IC 50 of 70 nM (Y. Hoshikawa et al, Exp. Cell Res., 1994, 214, 189).
  • TSA can also concomitantly modify the expression of genes.
  • Mishra et al demonstrated that TSA significantly downregulated CDl 54 and IL-IO and up- regulated IFN- ⁇ gene expression in systemic lupus erythematosis (SLE) T cells.
  • SLE is an autoimmune disease characterised by dysregulated production of antibodies which leads to irreversible, immune complex-mediated end-organ failure ( N. Mishra et al, PNAS, 2001, 98(5), 2628).
  • the HDAC inhibitor is suberoylanilide hyroxamic acid (SAHA).
  • SAHA Suberoylanilide hyroxamic acid
  • WO 2005/097747 discloses the use of prodrugs of hydroxamic based HDAC inhibitors, such as SAHA, in the treatment of neoplasms, thioredoxin (TRX)-mediated diseases and in the prevention and/or treatment of CNS diseases.
  • WO 2005/039498 and WO 2005/018578 disclose further methods for the treatment of neoplasms, wherein WO2005/039498 relates to leukemia and WO 2005/018578 relates to mesothelioma or lymphoma.
  • the HDAC inhibitor is sodium valproate (otherwise known as sodium 2-propylpentanoate).
  • Sodium valproate is the sodium salt of valproic acid and is a NICE-approved anticonvulsant drug used in the treatment of epilepsy. More recently, studies have investigated the use of sodium valproate for the treatment of advanced solid tumour malignancies and cancer-related neuropathic pain. Combination studies involving valproic acid and UCN-Ol have also been undertaken. In this regard, although valproic acid itself has only a weak anticancer effect, studies have shown that it becomes highly effective against cancer cells when used in combination.
  • Another aspect of the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, and a HDAC inhibitor.
  • Another aspect relates to a pharmaceutical product comprising the combination of the present invention for use in the treatment of a proliferative disorder, wherein the disorder is preferably cancer, and more preferably, NSCLC.
  • a further aspect of the present invention relates to a pharmaceutical product comprising a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, and a HDAC inhibitor as a combined preparation for simultaneous, sequential or separate use in therapy.
  • the HDAC inhibitor is selected from sodium butyrate, or a prodrug thereof, suberoylanilide hydroxamic acid (SAHA), sodium valproate and trichostatin (TSA).
  • SAHA suberoylanilide hydroxamic acid
  • TSA sodium valproate
  • TSA trichostatin
  • Yet another aspect relates to a method of treating a proliferative disorder, said method comprising simultaneously, sequentially or separately administering a combination of the present invention.
  • “simultaneously” is used to mean that the two agents are administered concurrently, whereas the term “in combination” is used to mean that they are administered, if not simultaneously, then “sequentially” with a timeframe that they are able to act therapeutically within the same time frame.
  • administration “sequentially” may permit one agent to be administered within 5 minutes, 10 minutes or a matter of hours after the other provided that they are both concurrently present in therapeutic amounts.
  • the time delay between administration of the components will vary depending on the exact nature of the components, the interaction therebetween and their respective half-lives.
  • the HDAC inhibitor is administered sequentially or separately prior to the compound of formula I or formula II, or a pharmaceutically acceptable salt thereof.
  • the compound of formula I or formula II, or a pharmaceutically acceptable salt thereof is administered sequentially or separately prior to the HDAC inhibitor.
  • the compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, and the HDAC inhibitor are administered simultaneously.
  • the HDAC inhibitor is sodium butyrate, or a prodrug thereof
  • the compound of formula I or formula II is administered simultaneously with, or separately or sequentially with, the HDAC inhibitor, irrespective of the order of administration. More preferably, the sodium butyrate and compound of formula I or formula II are administered separately or sequentially.
  • the HDAC inhibitor is sodium butyrate and the compound of formula I is compound [I].
  • the sodium butyrate and compound [1] are administered separately or sequentially, irrespective of the order of administration.
  • the HDAC inhibitor is sodium butyrate and the compound of formula II is compound [2].
  • the sodium butyrate and compound [2] are administered separately or sequentially, irrespective of the order of administration, or simultaneously.
  • the HDAC inhibitor is sodium valproate
  • the compound of formula I or formula II is administered simultaneously with, or separately or sequentially with, the HDAC inhibitor, irrespective of the order of administration. More preferably, the sodium valproate and compound of formula I or formula II are administered separately or sequentially. Even more preferably, the sodium valproate is administered separately or sequentially prior to the compound of formula I or II.
  • the HDAC inhibitor is sodium valproate
  • the compound of formula I is compound [I].
  • sodium valproate and compound [1] are administered separately or sequentially, irrespective of the order of administration.
  • the HDAC inhibitor is sodium valproate
  • the compound of formula II is compound [2].
  • sodium valproate and compound [2] are administered separately or sequentially, irrespective of the order of administration.
  • the HDAC inhibitor is sodium valproate
  • the compound of formula I is compound [3].
  • the sodium valproate is administered separately or sequentially prior to compound [3], i.e. the subject is pretreated with sodium valproate.
  • the HDAC inhibitor is sodium valproate
  • the compound of formula I is compound [4].
  • the sodium valproate is administered separately or sequentially prior to compound [4], i.e. the subject is pretreated with sodium valproate.
  • the compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, and the HDAC inhibitor are each administered in a therapeutically effective amount with respect to the individual components. In another preferred embodiment, the compound of formula I or formula II, or pharmaceutically acceptable salt thereof, and the HDAC inhibitor are each administered in a sub-therapeutic amount with respect to the individual components.
  • sub-therapeutic amount means an amount that is lower than that typically required to produce a therapeutic effect with respect to treatment with the compound of formula I or formula II alone or the HDAC inhibitor alone.
  • a further aspect relates to the use of the combination of the present invention in the preparation of a medicament for treating a proliferative disorder.
  • preparation of a medicament includes the use of one or more of the above described components directly as the medicament or in any stage of the manufacture of such a medicament.
  • Another aspect relates to the use of a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for the treatment of a proliferative disorder, wherein said treatment comprises simultaneously, sequentially or separately administering a HDAC inhibitor to a subject.
  • Yet another aspect relates to the use of a HDAC inhibitor, in the preparation of a medicament for the treatment of a proliferative disorder, wherein said medicament is for use in combination therapy with the compound of formula I or formula II, or a pharmaceutically acceptable salt thereof.
  • the therapy can be pretreatment therapy.
  • a further aspect relates to the use of the compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for the treatment of a proliferative disorder, wherein said medicament is for use in combination therapy with a HDAC inhibitor.
  • the therapy can be pretreatment therapy.
  • the term “combination therapy” refers to therapy in which the HDAC inhibitor and a compound of formula I or formula II are administered, if not simultaneously, then sequentially within a time frame that they both are available to act therapeutically within the same time frame.
  • pretreatment therapy means a regimen in which one agent is administered prior to, either separately or sequentially, the second agent.
  • the second agent is administered at least 2 hours after the administration of the first agent. More preferably, the second agent is administered at least 4 hours, or more preferably at least 6 or 8 hours, after the administration of the first agent. Even more preferably, the second agent is administered at least 12 hours, or more preferably at least 18 or 24 hours, after the administration of the first agent.
  • the compound of formula I or formula II and the HDAC inhibitor interact in a synergistic manner.
  • the term “synergistic” means that the compound of formula I or formula II and the HDAC inhibitor produce a greater effect when used in combination than would be expected from adding the individual effects of the two components.
  • a synergistic interaction may allow for lower doses of each component to be administered to a patient, thereby decreasing the toxicity of chemotherapy, whilst producing and/or maintaining the same therapeutic effect.
  • each component can be administered in a sub-therapeutic amount.
  • the CDK inhibitor and the HDAC inhibitor interact in a manner so as to alleviate or eliminate adverse side effects associated with use of the individual components in monotherapy, or associated with their use in known combinations.
  • the HDAC inhibitor is selected from sodium butyrate, or a prodrug thereof, suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA).
  • SAHA suberoylanilide hydroxamic acid
  • TSA trichostatin A
  • Another aspect of the invention relates to a combination comprising:
  • a HDAC inhibitor (i) a HDAC inhibitor; and (ii) (a) a 2,6,9-substituted purine derivative or a pharmaceutically acceptable salt thereof; or (b) a 2-substituted-4-heteroaryl-pyrimidine derivative, or a pharmaceutically acceptable salt thereof.
  • proliferative disorder is used herein in a broad sense to include any disorder that requires control of the cell cycle, for example cardiovascular disorders such as restenosis and cardiomyopathy, auto-immune disorders such as glomerulonephritis and rheumatoid arthritis, dermatological disorders such as psoriasis, anti-inflammatory, anti-fungal, antiparasitic disorders such as malaria, emphysema and alopecia.
  • cardiovascular disorders such as restenosis and cardiomyopathy
  • auto-immune disorders such as glomerulonephritis and rheumatoid arthritis
  • dermatological disorders such as psoriasis, anti-inflammatory, anti-fungal, antiparasitic disorders such as malaria, emphysema and alopecia.
  • the compounds of the present invention may induce apoptosis or maintain stasis within the desired cells as required.
  • the proliferative disorder is cancer.
  • the proliferative disorder is lung cancer, more preferably, non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • Lung cancers may be divided into two broad categories namely, small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). The distinction between these two types of cancer is based on the appearance of the tumour cells when viewed under a microscope.
  • SCLC small cell lung cancer
  • NSCLC non-small cell lung cancer
  • SCLC accounts for 20% of lung cancers diagnosed and it is characterised by small cells which are mostly filled with the nucleus (hence the name). It is sometimes also referred to as "oat cell” cancer. SCLC is the most aggressive type of cancer, which metastasizes rapidly to other parts of the body. Diagnosis with SCLC often occurs only after the cancer has spread throughout the body. In general, SCLC is almost always caused as a result of smoking. NSCLC can be subdivided into a group of related lung cancers which include epidermoid or squamous cell carcinoma, adenocarcinoma and large cell carcinoma.
  • Squamous cell lung cancer accounts for approximately 30% of all lung cancer cases and develops from reserve cells (which have the role of replacing damaged epithelium cells) in the lining of the lungs and bronchi. As a result, the cancer often initially develops in the centre of the chest. Squamous cell lung cancers are frequently slow growing and can take several years to progress from a confined tumour into invasive cancer. In 10-20% of cases, the cancer cavitates within the lungs. On metastasis, it often spreads to the bone, liver, adrenal glands, small intestine and brain.
  • Adenocarcinoma is the most common form of lung cancer making up 30-40% of all lung cancer cases. Adenocarcinoma develops in the outer part of the lung and develops from mucus-producing cells. The course of this cancer varies widely but often progresses slowly and the patient will present with few or no symptoms. In some cases, however, it can be extremely aggressive and rapidly fatal. In 50% of cases when it metastasises, it spreads only to the brain. Other locations to which adrenocarcinoma spreads include the liver, the adrenal glands, and bone.
  • the incidence of large cell carcinoma occurs less frequently than that of either adenocarcinoma or squamous cell carcinoma and accounts for 10-20% of lung cancer cases.
  • the cancer is composed of large-sized cells that are anaplastic in nature and often arise in the bronchi. Large cell carcinoma develops on the periphery of the lungs and can spread to the plura.
  • NSCLC drugs and regimens include camptosar (irinotecan; CPT-I l), camptothecin, carboplatin (paraplatin), cisplatin (platinol), epirubicin, gemcitabine, navelbine (vinorelbine), oxaliplatin, taxol (paclitaxel) and taxotere (docetaxol) (NSCLC Treatment - Chemotherapy, Lung Cancer Online).
  • Cisplatin is acknowledged to have certain disadvantages in that significant non-hematological toxicity (ototoxicity and nephroxicity) occurs in patients, along with emesis (P. Zatloukal et al, Lung Cancer, 2002, 38, S33).
  • the pharmaceutical product of the invention is in the form of a pharmaceutical composition comprising a pharmaceutically acceptable carrier, diluent or excipient.
  • the compounds of the present invention can be administered alone, they will generally be administered in admixture with a pharmaceutical carrier, excipient or diluent, particularly for human therapy.
  • a pharmaceutical carrier excipient or diluent
  • the pharmaceutical compositions may be for human or animal usage in human and veterinary medicine.
  • suitable carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like.
  • suitable diluents include ethanol, glycerol and water.
  • compositions may comprise as, or in addition to, the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s).
  • Suitable binders include starch, gelatin, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol.
  • Suitable lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition.
  • preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid.
  • Antioxidants and suspending agents may be also used.
  • the agents of the present invention can be present as salts or esters, in particular pharmaceutically acceptable salts or esters.
  • compositions of the agents of the invention include suitable acid addition or base salts thereof.
  • suitable pharmaceutical salts may be found in Berge et al, J Pharm Sci, 66, 1-19 (1977). Salts are formed, for example with strong inorganic acids such as mineral acids, e.g.
  • sulphuric acid, phosphoric acid or hydrohalic acids with strong organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (Cl-C4)-alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-toluene sulfonic acid.
  • Esters are formed either using organic acids or alcohols/hydroxides, depending on the functional group being esterified.
  • Organic acids include carboxylic acids, such as alkanecarboxylic acids of 1 to 12 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acid, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (Cl-C4)-alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-toluen
  • Suitable hydroxides include inorganic hydroxides, such as sodium hydroxide, potassium hydroxide, calcium hydroxide, aluminium hydroxide.
  • Alcohols include alkanealcohols of 1-12 carbon atoms which may be unsubstituted or substituted, e.g. by a halogen).
  • the invention also includes where appropriate all enantiomers and tautomers of the agents.
  • the man skilled in the art will recognise compounds that possess optical properties (one or more chiral carbon atoms) or tautomeric characteristics.
  • the corresponding enantiomers and/or tautomers may be isolated/prepared by methods known in the art. STEREO AND GEOMETRIC ISOMERS
  • agents of the invention may exist as stereoisomers and/or geometric isomers — e.g. they may possess one or more asymmetric and/or geometric centres and so may exist in two or more stereoisomeric and/or geometric forms.
  • the present invention contemplates the use of ail the individual stereoisomers and geometric isomers of those inhibitor agents, and mixtures thereof.
  • the terms used in the claims encompass these forms, provided said forms retain the appropriate functional activity (though not necessarily to the same degree).
  • the present invention also includes all suitable isotopic variations of the agent or pharmaceutically acceptable salts thereof.
  • An isotopic variation of an agent of the present invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature.
  • isotopes that can be incorporated into the agent and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine and chlorine such as 2H, 3H, 13C, 14C, 15N, 17O, 180, 31P, 32P, 35S, 18F and 36Cl, respectively.
  • isotopic variations of the agent and pharmaceutically acceptable salts thereof are useful in drug and/or substrate tissue distribution studies. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium, i.e., 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances. Isotopic variations of the agent of the present invention and pharmaceutically acceptable salts thereof of this invention can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents.
  • the invention furthermore relates to agents of the present invention in their various crystalline forms, polymorphic forms and (an)hydrous forms. It is well established within the pharmaceutical industry that chemical compounds may be isolated in any of such forms by slightly varying the method of purification and or isolation form the solvents used in the synthetic preparation of such compounds.
  • the invention further includes agents of the present invention in prodrug form.
  • prodrugs are generally compounds wherein one or more appropriate groups have been modified such that the modification may be reversed upon administration to a human or mammalian subject.
  • Such reversion is usually performed by an enzyme naturally present in such subject, though it is possible for a second agent to be administered together with such a prodrug in order to perform the reversion in vivo.
  • Examples of such modifications include ester (for example, any of those described above), wherein the reversion may be carried out be an esterase etc.
  • Other such systems will be well known to those skilled in the art.
  • compositions of the present invention may be adapted for oral, rectal, vaginal, parenteral, intramuscular, intraperitoneal, intraarterial, intrathecal, intrabronchial, subcutaneous, intradermal, intravenous, nasal, buccal or sublingual routes of administration.
  • compositions For oral administration, particular use is made of compressed tablets, pills, tablets, gellules, drops, and capsules. Preferably, these compositions contain from 1 to 2000 mg and more preferably from 50-1000 mg, of active ingredient per dose.
  • compositions of the present invention may also be in form of suppositories, pessaries, suspensions, emulsions, lotions, ointments, creams, gels, sprays, solutions or dusting powders.
  • the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin.
  • the active ingredient can also be incorporated, at a concentration of between 1 and 10% by weight, into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
  • Injectable forms may contain between 10 - 1000 mg, preferably between 10 - 500 mg, of active ingredient per dose.
  • compositions may be formulated in unit dosage, form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose.
  • the combination or pharmaceutical composition of the invention is administered intravenously.
  • a person of ordinary skill in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation.
  • a physician will determine the actual dosage which will be most suitable for an individual patient and it will depend on a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy.
  • the dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • the compound of formula I may be administered at a dose of from 0.01 to 30 mg/kg body weight, such as from 0.1 to 10 mg/kg, more preferably from 0.1 to 1 mg/kg body weight.
  • the compound of formula II may be administered at a dose of from 0.01 to 30 mg/kg body weight, such as from 0.1 to 10 mg/kg, more preferably from 0.1 to 1 mg/kg body weight.
  • one or more doses of 10 to 150 mg/day of the compounds of formula I or formula II will be administered to the patient for the treatment of malignancy.
  • the HDAC inhibitor is typically administered in accordance with a physician's direction at dosages described in the relevant references discussed above.
  • Pivanex is typically administered at about 2.34g/m 2 per day. Pivanex is preferably administered intravenously.
  • Suberoylanilide hydroxamic acid (SAHA) is typically administered from about 100-600mg per day.
  • SAHA is preferably administered orally.
  • the total daily dose of HDAC inhibitor can be administered as a single dose or divided into separate dosages preferably administered two, three or four time a day.
  • the HDAC inhibitor is administered at least 2 hours before the administration of the compound of formula I or formula II. More preferably, the HDAC inhibitor is administered at least 4 hours, or more preferably at least 6 or 8 hours, before the administration of the compound of formula I or formula II. Even more preferably, the HDAC inhibitor is administered at least 12 hours, or more preferably at least 18 or 24 hours, before the administration of the compound of formula I or formula II.
  • the HDAC inhibitor is administered at least 2 hours after the administration of the compound of formula I or formula II. More preferably, the HDAC inhibitor is administered at least 4 hours, or more preferably at least 6 or 8 hours, after the administration of the compound of formula I or formula II. Even more preferably, the HDAC inhibitor is administered at least 12 hours, or more preferably at least 18 or 24 hours, after the administration of the compound of formula I or formula II. KIT OF PARTS
  • a further aspect of the invention relates to a kit of parts comprising:
  • a HDAC inhibitor optionally admixed with a pharmaceutically acceptable diluent, excipient or carrier, optionally admixed with a pharmaceutically acceptable diluent, excipient or carrier.
  • the HDAC inhibitor is selected from sodium butyrate, or a prodrug thereof, suberoylanilide hydroxamic acid (SAHA), sodium valproate and trichostatin A (TSA).
  • SAHA suberoylanilide hydroxamic acid
  • TSA trichostatin A
  • the HDAC inhibitor is selected from sodium butyrate, or a prodrug thereof, suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA).
  • SAHA suberoylanilide hydroxamic acid
  • TSA trichostatin A
  • the compound of formula I or formula II and the HDAC inhibitor are each in unit dosage form.
  • the kit of parts contains a plurality of unit dosage forms of each component, i.e. of components (i) and (ii) above.
  • the kit of parts may further comprise a means for facilitating compliance with a particular dosing regimen, for example, instructions indicating when, how, and how frequently the unit dosage forms of each component should be taken.
  • Figure 1 shows that concomitant treatment of seliciclib and sodium butyrate leads to a synergistic increase in apoptosis in A549 cells, as determined by an increase in sub- Gl cell fragments, following 72 hours treatment.
  • Figure 2 shows that concomitant treatment of seliciclib and sodium butyrate in H460 cells leads to a synergistic increase in apoptosis following 72 hours treatment, as determined by annexin V staining.
  • Figure 3 shows that in respect of the seliciclib/sodium butyrate combination there is an increase in apoptosis at the IC50 concentration, as determined by caspase cleavage of cytokeratin 18 (M30 ELISA) and PARP cleavage.
  • Figure 3 also shows a synergistic decrease in McIl levels which probably relates to the loss of this anti- apoptotic protein, pushing the cells into apoptosis.
  • Figure 4 shows the molecular pathways of apoptosis with regard to the seliciclib/sodium butyrate combination in more detail.
  • Loss of McIl and Bcl2 both anti-apoptotic proteins pushes the cells towards apoptosis.
  • Both XIAP and survivin are inhibitors of the apoptotic process therefore the loss of these proteins again push the cells towards apoptosis.
  • This effect may explain the synergistic induction of apoptosis as shown by the appearance of PARP.
  • the Histone Western blot shows that the HDAC inhibitor increases the amount of acetylated histone (since deacetylation is inhibited).
  • Figure 5 shows the time course of cellular events at the IC50 in respect of the seliciclib/sodium butyrate combination. At later time points, the synergistic activation of caspases 3 and 9 (indicating apoptosis is induced) can now be seen.
  • Figure 6 shows the cell cycle distribution after treatment with DMSO (control), sodium valproate, seliciclib and sodium valproate/seliciclib in combination.
  • H460 cells were treated with the indicated drug(s) for the times shown prior to PI analysis on the flow cytometer. The results are the average of two duplicate samples.
  • Figure 7 shows the cell cycle distribution after treatment with DMSO (control), sodium valproate, the indicated CDK inhibitor and sodium valproate/CDK inhibitor in combination.
  • H460 cells were treated with the indicated drug(s) for the times shown prior to PI analysis on the flow cytometer. The results are the average of two duplicate samples.
  • Compound [1] referred to herein is (2R,3S)-3-( ⁇ 9-isopropyl-6-[(pyridin-3- ylmethyl)amino]-9H-purin-2-yl ⁇ amino)pentan-2-ol.
  • Sodium butyrate and sodium valproate were obtained from Sigma; TSA was obtained from AG Scientific, Inc.; SAHA was obtained from Toronto Research Chemicals, Inc.
  • the concomitant treatment 1.5-fold serial dilutions of the CDK inhibitor (seliciclib, compounds [l]-[4]), HDAC inhibitor, or both drugs simultaneously were added to cells 24h after plating, and left for 72h at 37 0 C.
  • the drug concentrations selected were chosen to span the IC 50 values for the drugs tested.
  • the first drug was added 2h after cells were plated, and left for 24h.
  • Medium was aspirated and replaced with fresh medium containing the second drug, and incubated for 72h.
  • the two controls for each sequential treatment involved substituting one of the drug treatments with medium.
  • Protein lysates were generated from 10cm plates that were seeded at approximately 5 x 10 5 cells/well, in medium containing 10% FCS. Cells were incubated with HDAC inhibitor and/or seliciclib at the indicated concentrations and times prior to harvest. After incubation, the supernatants were removed and centrifuged at 2000rpm for 5 min to pellet any floating cells.
  • Flow Cytometry H460 cells were seeded in 10cm plates at approximately 3x10 5 cells/plate and left to settle overnight. Next day, seliciclib, sodium butyrate or both drugs were added at the indicated concentrations. After either 24h or 72h treatment, cells were harvested by trypsinisation. Cell cycle analysis by propidium iodide (PI) staining involved fixing the cells overnight in 70% (v/v) ethanol at -2O 0 C prior to analysis on the flow cytometer. Annexin V staining was performed as indicated in manufacturers instructions, on live, non-fixed cells.
  • PI propidium iodide
  • H460 cells were seeded onto 10cm plates at approximately 0.5 x 10 6 cells/plate and allowed to settle for 24h.
  • Cells were treated with sodium valproate for 24 hours followed by the CDK inhibitor (seliciclib, compounds [l]-[4]) for a further 24 hours.
  • the concentrations of compound used were equivalent to 1 x IC 5 O.
  • Single agent control treatments were also carried out. These involved treating the cells with sodium valproate for 24 hours followed by drug-free medium for a further 24 hours, or drug- free medium for 24 hours followed by the CDK inhibitor for a further 24 hours. All cells were harvested by collecting the media prior to media changes, as well as at 48 hours.
  • Adherent cells were harvested by trypsinisation, pooled with the cells in suspension, washed twice in PBS and fixed by resuspending in ImI ice-cold 70% ethanol. Standard cell cycle analysis by propidium iodide staining was carried out on the flow cytometer. Results are the average of duplicate samples.
  • M30/TPS analysis of time-gap experiment A549 cells were seeded in 96 well plates and left to settle overnight. Cells were treated with seliciclib, sodium butyrate or the combination at the indicated concentrations. After 72h treatment, medium was harvested, retained and stored at - 2O 0 C. Samples were analysed in the M30 ELISA as described in the manufacturers instructions.
  • Seliciclib was tested in combination with the indicated HDAC inhibitors in H460 and A549 cell lines, using three different treatment regimes.
  • the Combination Index values from each drug treatment are shown for ED50, ED75 and ED90 values (the point on the curve where 50%, 75% and 90% of the cells have been killed).
  • Data are the average of at least three independent experiments (Table 1).
  • Table 1 Data for the effect of seliciclib and the HDAC inhibitors on the A549 cell lines are shown in parentheses.
  • seliciclib is synergistic when used in combination with all three HDAC inhibitors tested, demonstrating that combining seliciclib with a HDAC inhibitor is a good concept for treating NSCLC cell lines.
  • butyrate was tested in combination with the indicated CDK inhibitors in H460 cells, using three different treatment regimes.
  • the Combination Index values from each drug treatment are shown for ED50, ED75 and ED90 values (the point on the curve where 50%, 75% and 90% of the cells have been killed).
  • Data are the average of three independent experiments (Table 2).
  • Table 2 Data for the effect of compounds [l]-[4] in combination with sodium butyrate.
  • Seliciclib and Butyrate induce a synergistic increase in sub-Gl A549 cells.
  • A549 cells were incubated with IC50 butyrate, 0.25 - 1.5 X IC50 seliciclib, or 0.25 - 1.5 X IC50 seliciclib in the presence of IC50 butyrate for 72h. Cells were then harvested, stained with propidium iodide and their DNA content analysed by flow cytometry. Data are representative of two independent experiments ( Figure 1).
  • Seliciclib and Butyrate induce a synergistic increase in apoptotic H460 cells.
  • H460 cells were incubated with 0.25 - 1.5X IC50 butyrate, 0.25 - 1.5 X IC50 seliciclib, or 0.25 - 1.5 X IC50 seliciclib and butyrate for 72h. Cells were then harvested, stained with annexin V and analysed on the flow cytometer. Data are representative of two independent experiments ( Figure 2).
  • Annexin V labels live cells that are undergoing apoptosis.
  • butyrate and seliciclib induced a much larger annexin V signal than the two single drug treatments combined, indicating a synergistic increase in apoptotic cells.
  • the highest concentration of butyrate and seliciclib (1.5 X IC50) appears to contain fewer cells undergoing apoptosis than those treated with 0.67 X or 1 X IC50, the reason for this is not clear at present.
  • A549 cells were treated with DMSO (control) or with IC50 concentrations of seliciclib, sodium butyrate, or seliciclib and butyrate for 72h, as indicated.
  • Cell culture supernatants were harvested and tested in the M30 apoptosense ELISA, and the cells harvested and analysed for cleaved PARP and McI-I by western blotting. Data are representative of two independent experiments ( Figure 3).
  • H460 cells were treated with butyrate, seliciclib or seliciclib and butyrate at 1 X or 1.5 X IC50 concentrations for 24h. Cells were harvested and the resulting cell lysates analysed by western blotting with the indicated antibodies. Data are representative of two independent experiments ( Figure 4).
  • H460 cells were treated with IX IC50 butyrate, seliciclib or seliciclib and butyrate for the indicated times. Cells were harvested and the resulting cell lysates analysed by western blotting with the indicated antibodies. Data are representative of two independent experiments ( Figure 5).

Landscapes

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

Abstract

La présente invention concerne, selon un premier aspect, un produit composé comprenant un inhibiteur de HDAC et un composé représenté par la formule (I) ou (II), ou un sel pharmaceutiquement acceptable de celui-ci, et un inhibiteur de HDAC. Cette invention concerne, selon un deuxième aspect, un produit pharmaceutique comprenant un inhibiteur de HDAC et un composé représenté par la formule (I) ou (II), ou un sel pharmaceutiquement acceptable de celui-ci, comme préparation combinée destinée à être utilisée de manière simultanée, séquentielle ou séparée en thérapie. Cette invention concerne, selon un troisième aspect, une méthode de traitement d'un trouble proliférant, laquelle méthode consiste à administrer simultanément, séquentiellement ou séparément un composé représenté par la formule (I) ou (II), ou un sel pharmaceutiquement acceptable de celui-ci, et un inhibiteur de HDAC à un sujet.
PCT/GB2006/004224 2005-11-11 2006-11-13 Produit compose WO2007054725A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP06808516A EP1951307A2 (fr) 2005-11-11 2006-11-13 Combinaison d'un inhibiteur cdk avec un inhibiteur d'hdac

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GBGB0523040.4A GB0523040D0 (en) 2005-11-11 2005-11-11 Combination
GB0523040.4 2005-11-11
GB0523160A GB0523160D0 (en) 2005-11-14 2005-11-14 Combination
GB0523160.0 2005-11-14

Publications (2)

Publication Number Publication Date
WO2007054725A2 true WO2007054725A2 (fr) 2007-05-18
WO2007054725A3 WO2007054725A3 (fr) 2007-08-09

Family

ID=37603117

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2006/004224 WO2007054725A2 (fr) 2005-11-11 2006-11-13 Produit compose

Country Status (2)

Country Link
EP (1) EP1951307A2 (fr)
WO (1) WO2007054725A2 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150203517A1 (en) * 2014-01-17 2015-07-23 Methylgene Inc. Prodrugs of Compounds that Enhance Antifungal Activity and Compositions of Said Prodrugs
WO2017020065A1 (fr) * 2015-08-04 2017-02-09 University Of South Australia Dérivés de n-(pyridin-2-yl)-4-(thiazol-5-yl) pyrimidin-2-amine utilisés comme composés thérapeutiques
WO2021175200A1 (fr) * 2020-03-02 2021-09-10 Sironax Ltd Inhibiteurs de ferroptose-diarylamine para-acétamides

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004016612A2 (fr) * 2002-08-15 2004-02-26 Cyclacel Limited Nouveaux derives puriques
US20050004007A1 (en) * 2000-09-12 2005-01-06 Steven Grant Promotion of adoptosis in cancer cells by co-administration of cyclin dependent kinase inhibitiors and cellular differentiation agents
WO2005012298A1 (fr) * 2003-07-30 2005-02-10 Cyclacel Limited Derives de pyridinylamino-pyrimidine utiles comme inhibiteurs de la proteine kinase
WO2005042525A1 (fr) * 2003-10-21 2005-05-12 Cyclacel Limited Composes de pyrimidin-4-yl-3, 4-thione et leur utilisation a des fins therapeutiques

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050004007A1 (en) * 2000-09-12 2005-01-06 Steven Grant Promotion of adoptosis in cancer cells by co-administration of cyclin dependent kinase inhibitiors and cellular differentiation agents
WO2004016612A2 (fr) * 2002-08-15 2004-02-26 Cyclacel Limited Nouveaux derives puriques
WO2005012298A1 (fr) * 2003-07-30 2005-02-10 Cyclacel Limited Derives de pyridinylamino-pyrimidine utiles comme inhibiteurs de la proteine kinase
WO2005042525A1 (fr) * 2003-10-21 2005-05-12 Cyclacel Limited Composes de pyrimidin-4-yl-3, 4-thione et leur utilisation a des fins therapeutiques

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BIBLE K C ET AL: "FLAVOPIRIDOL: a CYTOTOXIC FLAVONE THAT INDUCES CELL DEATH IN NONCYCLING A549 HUMAN LUNG CARCINOMA CELLS" CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, BALTIMORE, MD, US, vol. 56, 1 November 1996 (1996-11-01), pages 4856-4861, XP002125982 ISSN: 0008-5472 *
REID TONY ET AL: "Phase II trial of the histone deacetylase inhibitor pivaloyloxymethyl butyrate (Pivanex, AN-9) in advanced non-small cell lung cancer." LUNG CANCER (AMSTERDAM, NETHERLANDS) SEP 2004, vol. 45, no. 3, September 2004 (2004-09), pages 381-386, XP002415089 ISSN: 0169-5002 *
ROSATO ROBERTO R ET AL: "Evidence of a functional role for p21WAF1/CIP1 down-regulation in synergistic antileukemic interactions between the histone deacetylase inhibitor sodium butyrate and flavopiridol." MOLECULAR PHARMACOLOGY, vol. 65, no. 3, March 2004 (2004-03), pages 571-581, XP002415088 ISSN: 0026-895X *
SHABBEER S ET AL: "FOCUS ON DEACETYLATION FOR THERAPEUTIC BENEFIT" IDRUGS, CURRENT DRUGS LTD, GB, vol. 8, no. 2, 2 February 2005 (2005-02-02), pages 144-154, XP009066252 ISSN: 1369-7056 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150203517A1 (en) * 2014-01-17 2015-07-23 Methylgene Inc. Prodrugs of Compounds that Enhance Antifungal Activity and Compositions of Said Prodrugs
US9636298B2 (en) * 2014-01-17 2017-05-02 Methylgene Inc. Prodrugs of compounds that enhance antifungal activity and compositions of said prodrugs
WO2017020065A1 (fr) * 2015-08-04 2017-02-09 University Of South Australia Dérivés de n-(pyridin-2-yl)-4-(thiazol-5-yl) pyrimidin-2-amine utilisés comme composés thérapeutiques
CN108349964A (zh) * 2015-08-04 2018-07-31 常州千红生化制药股份有限公司 N-(吡啶-2-基)-4-(噻唑-5-基)嘧啶-2-胺类化合物作为治疗性化合物
JP2018527334A (ja) * 2015-08-04 2018-09-20 ユニバーシティー オブ サウス オーストラリアUniversity Of South Australia 治療化合物としてのn−(ピリジン−2−イル)−4−(チアゾール−5−イル)ピリミジン−2−アミン誘導体
US10479785B2 (en) 2015-08-04 2019-11-19 Aucentra Therapeutics Pty Ltd N-(pyridin-2-yl)-4-(thiazol-5-yl)pyrimidin-2-amine derivatives as therapeutic compounds
CN108349964B (zh) * 2015-08-04 2021-06-01 常州千红生化制药股份有限公司 N-(吡啶-2-基)-4-(噻唑-5-基)嘧啶-2-胺类化合物作为治疗性化合物
USRE49850E1 (en) 2015-08-04 2024-02-27 Aucentra Therapeutics Pty Ltd N-(pyridin-2-yl)-4-(thiazol-5-yl)pyrimidin-2-amine derivatives as therapeutic compounds
US11970491B2 (en) 2015-08-04 2024-04-30 Aucentra Therapeutics Pty Ltd N-(pyridin-2-yl)-4-(thiazol-5-yl)pyrimidin-2-amine derivatives as therapeutic compounds
WO2021175200A1 (fr) * 2020-03-02 2021-09-10 Sironax Ltd Inhibiteurs de ferroptose-diarylamine para-acétamides
CN115151252A (zh) * 2020-03-02 2022-10-04 维泰瑞隆有限公司 铁死亡抑制剂-二芳基胺对乙酰胺类
EP4114375A4 (fr) * 2020-03-02 2024-05-29 Sironax Ltd Inhibiteurs de ferroptose-diarylamine para-acétamides

Also Published As

Publication number Publication date
EP1951307A2 (fr) 2008-08-06
WO2007054725A3 (fr) 2007-08-09

Similar Documents

Publication Publication Date Title
JP7058345B2 (ja) Bcl-2阻害剤及びMDM2阻害剤の組合せ製品並びに疾患の予防及び/又治療におけるその使用
JP5478262B2 (ja) 抗癌剤
US20170157134A1 (en) Combination therapy
JP7114478B2 (ja) 癌の治療のための配合剤
WO2020103921A1 (fr) Composition pharmaceutique et utilisation associée
JP6076498B2 (ja) ナンセンス突然変異抑制剤としてのピリミド[4,5−b]キノリン−4,5(3h,10h)−ジオン
JP2005200419A (ja) 癌治療法
WO2012104823A2 (fr) Composés pyridopyrimidinones dans le traitement de maladies neurodégénératives
US20130196938A1 (en) Combination comprising cndac (2'-cyano-2'-deoxy-n4-palmitoyl-1-beta-d-arabinofuranosyl-cytosine) and a cytotoxic agent
WO2014170873A1 (fr) Composés présentant une activité d'inhibition de la sirtuine
ES2385850T3 (es) Potenciador de radioterapia
US20090306098A1 (en) Combination of roscovitine and a hdca inhibitor to treat proliferative diseases
WO2020103922A1 (fr) Nouvelle composition pharmaceutique et son utilisation
EP1951307A2 (fr) Combinaison d'un inhibiteur cdk avec un inhibiteur d'hdac
US9872874B2 (en) Dosage regimen for sapacitabine and seliciclib
JP2015510945A (ja) オーロラキナーゼ阻害薬を使用する癌の治療方法
KR20080044277A (ko) 유기 화합물의 조합물
WO2018186366A1 (fr) Nouvel inhibiteur de kinase dépendante de la cycline 8 et/ou 19
AU2015324072B2 (en) A compound for anti-cancer therapy that acts by targeting GOF mutant P53 and stimulates P73
US20050164976A1 (en) Combination of a cdk inhibitor and 5-fu for the tratment of cancer
CN113004207B (zh) Plk4小分子抑制剂的化合物及其应用
US20230404973A1 (en) New carbazole derivatives sensitizing cells to anti-cancer agents
US20180303797A1 (en) Chalcone Compounds
JP2013166703A (ja) インドール化合物を含有する抗腫瘍剤

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2006808516

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

WWP Wipo information: published in national office

Ref document number: 2006808516

Country of ref document: EP