WO2002020015A1 - Inhibitors of prenyl-protein transferase - Google Patents

Inhibitors of prenyl-protein transferase Download PDF

Info

Publication number
WO2002020015A1
WO2002020015A1 PCT/US2001/027013 US0127013W WO0220015A1 WO 2002020015 A1 WO2002020015 A1 WO 2002020015A1 US 0127013 W US0127013 W US 0127013W WO 0220015 A1 WO0220015 A1 WO 0220015A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
heterocycle
aryl
rlo
Prior art date
Application number
PCT/US2001/027013
Other languages
French (fr)
Inventor
S. Jane Desolms
Anthony W. Shaw
Original Assignee
Merck & Co., Inc.
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 Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to AU2001290588A priority Critical patent/AU2001290588A1/en
Publication of WO2002020015A1 publication Critical patent/WO2002020015A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D515/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D515/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D515/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • 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
    • A61K31/4151,2-Diazoles
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0002General or multifunctional contrast agents, e.g. chelated agents

Definitions

  • Ras proteins are part of a signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation.
  • Biological and biochemical studies of Ras action indicate that Ras functions like a G-regulatory protein.
  • Ras In the inactive state, Ras is bound to GDP.
  • Ras Upon growth factor receptor activation Ras is induced to exchange GDP for GTP and undergoes a conformational change.
  • the GTP-bound form of Ras propagates the growth stimulatory signal until the signal is terminated by the intrinsic GTPase activity of Ras, which returns the protein to its inactive GDP bound form (D.R. Lowy and D.M. Willumsen, Ann. Rev. Biochem.
  • Mutated ras genes are found in many human cancers including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias.
  • the protein products of these genes are defective in their GTPase activity and constitutively transmit a growth stimulatory signal.
  • Ras must be localized to the plasma membrane for both normal and oncogenic functions. At least 3 post-translational modifications are involved with Ras membrane localization, and all 3 modifications occur at the C-terminus of Ras.
  • the Ras C-terminus contains a sequence motif termed a "CAAX” or "Cys- Aaal-Aaa2-Xaa” box (Cys is cysteine, Aaa is an aliphatic amino acid, the Xaa is any amino acid) (Willumsen et al., Nature 310:583-586 (1984)).
  • this motif serves as a signal sequence for the enzymes farnesyl- protein transferase or geranylgeranyl-protein transferase type I, which catalyze the alkylation of the cysteine residue of the CAAX motif with a C15 or C20 isoprenoid, respectively.
  • the term prenyl-protein transferase may be used to refer generally to farnesyl-protein transferase and geranylgeranyl- protein transferase type I.
  • the Ras protein is one of several proteins that are known to undergo post-translational farnesylation.
  • Other farnesylated proteins include the Ras-related GTP-binding proteins such as Rho, fungal mating factors, the nuclear lamins, and the gamma subunit of transducin.
  • James, et al., /. Biol. Chem. 269, 14182 (1994) have identified a peroxisome associated protein Pxf which is also farnesylated.
  • James, et al. have also suggested that there are farnesylated proteins of unknown structure and function in addition to those listed above.
  • Farnesyl-protein transferase utilizes farnesyl pyrophosphate to covalently modify the Cys thiol group of the Ras CAAX box with a farnesyl group (Reiss et al, Cell, 62:81-88 (1990); Schaber et al., J. Biol. Chem., 265:14701-14704 (1990); Schafer et al, Science, 249:1133-1139 (1990); Manne et al., Proc. Natl. Acad. Sci USA, 57:7541-7545 (1990)).
  • Inhibition of farnesyl pyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane localization in cultured cells.
  • FPTase farnesyl-protein transferase
  • FPP farnesyl diphosphate
  • Ras protein substrates
  • Such inhibitors may inhibit protein prenylation while serving as alternate substrates for the farnesyl-protein transferase enzyme, or may be purely competitive inhibitors (U.S. Patent No. 5,141,851, University of Texas; N.E. Kohl et al., Science, 260:1934-1937 (1993); Graham, et al., J. Med. Chem., 37, 725 (1994)).
  • deletion of the thiol from a CAAX derivative has been shown to dramatically reduce the inhibitory potency of the compound.
  • thiol group potentially places limitations on the therapeutic application of FPTase inhibitors with respect to pharmacokinetics, pharmacodynamics and toxicity. Therefore, a functional replacement for the thiol is desirable. It has recently been reported that farnesyl-protein transferase inhibitors are inhibitors of proliferation of vascular smooth muscle cells and are therefore useful in the prevention and therapy of arteriosclerosis and diabetic disturbance of blood vessels (JP H7-112930).
  • the present invention comprises peptidomimetic piperidine- and pyrrolidine-containing macrocyclic compounds which inhibit prenyl-protein transferase. Further contained in this invention are chemotherapeutic compositions containing these prenyl-protein transferase inhibitors and methods for their production.
  • the compounds of this invention are useful in the inhibition of prenyl-protein transferase and the prenylation of the oncogene protein Ras.
  • the inhibitors of prenyl-protein transferase are illustrated by the formula A:
  • Rla, Rib, R lc and Rid are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) RlOO-, f) RllS(O) m -, g) Rl0C(O)NRl0-, fa) (RlO) 2 NC(O)-, i) CN, j) NO2, k) RlOC(O)-,
  • substituent on the substituted group is selected from unsubstituted or substituted Ci-C ⁇ alkyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C3-C10 cycloalkyl, unsubstituted or unsubstituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, RlOO-, Rl lS(O) m -, RlOC(O)NRlO-, (RlO) 2 NC(O)-, CN,
  • R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-8 alkyl, c) unsubstituted or substituted C2-8 alkenyl, d) unsubstituted or substituted C2-8 alkynyl, e) unsubstituted or substituted aryl, f) unsubstituted or substituted heterocycle, wherein the substituted group is substituted with one or more of:
  • R4 is selected from Ci-6 alkyl, C ⁇ - ⁇ cycloalkyl, heterocycle, aryl, unsubstituted or substituted with:
  • R5, R6 and R7 are independently selected from: a) hydrogen, b) RlOC(O)-, c) Rl ⁇ C(O)-, or d) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3_6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
  • R6 and R "7 may be joined in a ring; and independently,
  • R5 and R may be joined in a ring;
  • R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) C1-C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O) m -, k) Rl0C(O)NRl0-, 1) (RlO) 2 NC(O)-, m) CN, n) NO2, o) RlOC(O)-, p) Rl ⁇ C(O)-, q) -N(RlO) 2 , r) Rl lOC
  • R9 is selected from: a) hydrogen, b) unsubstituted or substituted C2-C6 alkenyl, c) unsubstituted or substituted C2-C6 alkynyl, d) C1-C6 perfluoroalkyl, e) halo, f) RlOO-, g) RllS(O) m -, h) Rl0C(O)NRl0-, i) (R10) 2 NC(O)-, j) CN, k) NO2,
  • RlO is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-C ⁇ alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
  • Rl 1 is independently selected from a) unsubstituted or substituted C 1 -C alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
  • Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRlO-, d) -NRl C(O)-, e) O, f) -N(Rl )-, g) -S(O) 2 N(R10)-, i) S(O) m ;
  • G is selected from H2 or O;
  • V is selecte. I from: a) heterocycle, or b) aryl;
  • W is a heterocycle
  • X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, f) -O-C(O)NRl0-, g) -NRl0C(O)NRl0-, h) O, i) -N(Rl0)-, j) -S(O) 2 N(RlO)-,
  • Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of: 1) Ci-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) C ⁇ _4 alkoxy, b) NR6R7, c) C3_6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O) m R4, g) -C(O)NR6R7, or h) Ci-4 perfluoroalkyl;
  • Z2 is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
  • inhibitors of prenyl- protein transferase are illustrated by the formula A:
  • Rla, Rib, Rlc and Rid are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3 -C 10 cycloalkyl , e) RlOO-, f) RllS(O) m -, g) Rl0C(O)NRl0-, h) (RlO) 2 NC(O)-, i) CN, j) NO 2 , k) RlOC(O)-, 1) RlO ⁇ C(O)-, m) -N(RlO) 2 , n) RllOC(O)NRl0-, o) unsubstituted or substituted C1-C6 alkyl, p) unsubstituted or substituted C2-C6 alkenyl, or q) unsubstituted or substituted C2-C6 alkyn
  • C3-C10 cycloalkyl unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, RlOO-, RllS(O) m -, RlOC(O)NRlO-, (RlO) 2 NC(O)-, CN, RlOC(O)-, RlO ⁇ C(O)-, -N(RlO)2, and RllOC(O)NRl -,
  • R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-8 alkyl, c) unsubstituted or substituted C2-8 alkenyl, d) unsubstituted or substituted C2-8 alkynyl, e) unsubstituted or substituted aryl, f) unsubstituted or substituted heterocycle, g)
  • substituted group is substituted with one or more of:
  • R4 is selected from C ⁇ _4 alkyl, C3.6 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with: 1) C1-.4 alkoxy,
  • R5, R6 and R7 are independently selected from: a) hydrogen, b) RIOC(O)-, c) Rl°OC(O)-, or d) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
  • R and R7 may be joined in a ring; and independently,
  • R5 and R7 may be joined in a ring
  • R9 is selected from: a) hydrogen, b) unsubstituted or substituted C2-C6 alkenyl, c) unsubstituted or substituted C2-C6 alkynyl, d) C1-C6 perfluoroalkyl, e) halo, f) RlOO-, g) RllS(O) m -, h) Rl0C(O)NRl0-, i) (RlO) 2 NC(O)-, j) CN, k) NO2,
  • RlO is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-C ⁇ alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
  • Rll is independently selected from a) unsubstituted or substituted C1-C6 alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
  • Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(Rl0)-, g) -S(O) 2 N(R10)-, i) S(O) m ;
  • A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(Rl0)-, g) -S(O) 2 N(Rl0)-, i) -NRl0C(O)NRl0-, or j) S(O) m;
  • G is selected from H2 or O;
  • V is selected from pyridyl or phenyl
  • W is a heterocycle
  • X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, f) -O-C(O)NRl0-, g) -NRl0C(O)NRl0-, h) O, i) -N(Rl0)-,
  • Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
  • Z2 is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
  • inhibitors of prenyl- protein transferase are illustrated by the formula B:
  • Rla, Rib and Rlc are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, f) unsubstituted or substituted C2-C6 alkenyl g) -ORlO, or h) unsubstituted or substituted Ci-C ⁇ alkyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, RlOO-, and -N(RlO) 2 ,
  • Rid is selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) RlOO-, ) R n S(O) m -, g) RlOC(O)NRlO-, h) (RlO) 2 NC(O)-, i) CN, j) NO 2 , k) Rl0C(O)-,
  • substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, RlOO-, RllS(O) m -, RlOC(O)NRlO-, (RlO) 2 NC(O)-, CN, RlOC(O)-, RlO ⁇ C(O)-, -N(RlO) 2 , and RHOC(O)NRlO-;
  • R2 is independently selected from a) hydrogen, b) unsubstituted or substituted C ⁇ _8 alkyl, c)
  • substituted group is substituted with one or more of:
  • R4 is selected from C ⁇ _4 alkyl, or C3-6 cycloalkyl, unsubstituted or substituted with: 1) Ci-4 alkoxy, 2) aryl or heterocycle,
  • R and R7 are independently selected from: a) hydrogen, b) Rl0C(O)-, c) RlO ⁇ C(O)-, or d) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3.6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
  • R6 and R7 may be joined in a ring
  • R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) C1-C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O) m -, k) Rl0C(O)NRl0-,
  • R9 is selected from: a) hydrogen, b) Ci-C ⁇ perfluoroalkyl, c) RlOO-, d) Rl0C(O)NRl0-, e) (RlO) 2 NC(O)-, or f) unsubstituted or substituted C1-C6 alkyl optionally substituted by C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O) m -, RlOC(O) NRlO-, (RlO) 2 NC(O)-, CN, RlOC(O)-, RlO ⁇ C(O)-, -N(RlO) 2 , or
  • RlO is independently selected from a) hydrogen, b) unsubstituted or substituted C1-C6 alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
  • Rll is independently selected from a) unsubstituted or substituted C 1 -C alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
  • Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, ) -N(R10)-, g) -S(O) 2 N(R10)-, i) S(O) m ;
  • A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(RlO)-, g) -S(O) 2 N(RlO)-, i) -NRl0C(O)NRl0-, or j) S(O) m;
  • G is selected from H2 or O;
  • W is a heterocycle selected from imidazolyl, pyridyl, or triazolyl;
  • Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
  • kis 0,1,2, 3 or 4 m is 0, 1 or 2; nis 0,1,2, 3, 4 or 5; p is independently 0, 1, 2, 3, 4 or 5; qis 1,2 or 3; ris 0,1,2, 3, 4 or 5; s is independently 0, 1, 2, 3, 4 or 5; tis 0orl;and u is independently 2, 3, 4, 5 or 6;
  • inhibitors of prenyl-protein transferase are illustrated by the formula C:
  • Rla, Rib and Rlc are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C 10 cycloalkyl, f) unsubstituted or substituted C2-C6 alkenyl g) -ORl0, or h) unsubstituted or substituted C1-C6 alkyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, RlOO-, and -N(RlO) 2 ,
  • Rid is selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) RlOO-, f) -N(RlO) , or g) unsubstituted or substituted C1-C6 alkyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, RlOo-, Rl0c(O)NRl0-, (RlO) 2 NC(O)-, Rl0C(O)-, RlO ⁇ C(O)-, -N(RlO) 2 , and RllOC(O)-NRl0-;
  • R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-8 alkyl, c)
  • R4 is selected from C ⁇ _4 alkyl, or C3-6 cycloalkyl, unsubstituted or substituted with: 1) C1-4 alkoxy,
  • R6 and R7 are independently selected from: a) hydrogen, or b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C ⁇ _6 perfluoroalkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
  • R and R7 may be joined in a ring
  • R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) C1-C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O) m -, k) RlOC(O)NRlO-, 1) (RlO) 2 NC(O)-, m) CN, n) NO2, o) Rl0C(O)-, p) RlO ⁇ C(O)-, r) RllOC(O)NRl0-, and s) unsubstituted or substituted C ⁇ -C6 alkyl, optional
  • R9 is selected from: a) hydrogen, b) C1-C6 perfluoroalkyl, or c) unsubstituted or substituted C1-C6 alkyl optionally substituted by C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O) m -, Rl0C(O)NRl0-, (RlO) 2 NC(O)-, CN, RlOC(O)-, RlO ⁇ C(O)-, -N(RlO) 2 , or RllOC(O)NRl0- ;
  • RlO is independently selected from a) hydrogen, b) unsubstituted or substituted C 1 -C6 alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
  • Rl 1 is independently selected from a) unsubstituted or substituted C1-C6 alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
  • Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O,
  • A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(RlO)-, g) -S(O) 2 N(R10)-, h) -N(RlO)S(O) 2 -, i) -NRl0C(O)NRl0-, or j) S(O) m;
  • G is selected from H2 or O;
  • X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, f) -O-C(O)NRl0-, g) -NRl0C(O)NRl0-, h) O, i) -N(Rl0)-, j) -S(O) 2 N(R10)-,
  • Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of: 1) Ci-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7, c ) C3_6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O) m R4, g) -C(O)NR6R7, or h) C 1 _4 perfluoroalkyl ;
  • the compounds of the present invention may have asymmetric centers, chiral axes and chiral planes, and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention.
  • any variable e.g. aryl, heterocycle, Rla, R6 etc.
  • its definition on each occurrence is independent at every other occurrence.
  • combinations of substituents/or variables are permissible only if such combinations result in stable compounds.
  • alkyl is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having from 1 to 20 carbon atoms, unless otherwise specified; "alkoxy” represents an alkyl having from 1 to 20 carbon atoms, unless otherwise indicated, attached through an oxygen bridge.
  • Hydrogen or “halo” as used herein means fluoro, chloro, bromo and iodo.
  • cycloalkyl is intended to include non-aromatic hydrocarbon groups having from 3 to 10 carbon atoms, unless otherwise specified.
  • examples of such cycloalkyl groups includes, but are not limited to, cyclopropyl, cyclobutyl, cyclohexyl, cycloheptyl, cyclooctyl, admantyl and the like.
  • alkenyl refers to a non-aromatic hydrocarbon, straight, branched or cyclic, containing from 2 to 10 carbon atoms, unless otherwise indicated, and at least one carbon to carbon double bond. Preferably one carbon to carbon double bond is present, and up to four non- aromatic carbon-carbon double bonds may be present.
  • C2-C8 alkenyl means an alkenyl radical having from 2 to 8 carbon atoms. Examples of such alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted if a substituted alkenyl group is indicated.
  • alkynyl refers to a hydrocarbon radical straight, branched or cyclic, containing from 2 to 10 carbon atoms, unless otherwise indicated, and at least one carbon to carbon triple bond. Up to three carbon-carbon triple bonds may be present.
  • C2-C8 alkynyl means an alkynyl radical having from 2 to 8 carbon atoms. Examples of such alkynyl groups include, but are not limited to, ethynyl, propynyl and butynyl.
  • the straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated.
  • aryl is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic.
  • aryl elements include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl, acenaphthyl and the like.
  • aralkyl is intended to mean an aryl moiety, as defined above, attached through a C ⁇ -C6 alkyl linker, where alkyl is defined above. Examples of aralkyls include, but are not limited to, benzyl, naphthylmethyl, phenylbutyl and the like.
  • heterocycle or heterocyclic represents a stable 5- to 7-membered monocyclic or stable 8- to 11-membered bicyclic heterocyclic ring which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocycle or heterocyclic includes heteroaryl moieties.
  • heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzo- thiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, 1,3-dioxolanyl, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphth
  • heteroaryl is intended to mean any stable mono- cyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic and wherein from one to four carbon atoms are replaced by heteroatoms selected from the group consisting of N, O, and S.
  • heterocyclic elements include, but are not limited to, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxadiazolyl, pyridyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolyl, quinazolin
  • heterocyclylalkyl is intended to mean a heterocyclic moiety, as defined above, attached through a C ⁇ -C6 alkyl linker, where alkyl is defined above.
  • heterocyclylalkyls include, but are not limited to, 2-pyridylmethyl, 2-imidazolylethyl, 2-quinolinylmethyl, 2-imidazolylmethyl, and the like.
  • heterocycloalkyl is intended to mean a heterocyclic moiety, as defined above, attached through a C ⁇ -C6 alkyl linker, where alkyl is defined above.
  • heterocycloalkyls include, but are not limited to, 2-morpholinylethyl, 1-piperazinylmethyl, 3-pyrrolidinylrriethyl and the like.
  • substituted alkyl As used herein, the terms "substituted alkyl”, “substituted alkenyl”, “substituted alkynyl” and “substituted alkoxy”, unless otherwise defined, are intended to include the branch or straight-chain alkyl group of the specified number of carbon atoms, wherein the carbon atoms may be substituted with F, Cl, Br, I, CF3, OCF3,
  • CN N3, NO2, NH2, N(C ⁇ -C6 alkylfc, oxo, OH, -O(C ⁇ -C6 alkyl), S(O) ⁇ -2, (C ⁇ -C6 alkyl)S(O)0-2-, C2-C6 alkenyl, C2-C6 alkynyl, -(C ⁇ -C6 alkyl)S(O) ⁇ -2(Cl-C6 alkyl), C3-C20 cycloalkyl, -C(O)NH2, HC(O)NH-(C ⁇ -C6 alkyl)C(O)NH-, H2NC(O)NH- (C1-C6 alkyl)C(O)-, -O(C ⁇ -C6 alkyl)CF 3 , (C -C6 alkyl)OC(O)-, (C ⁇ -C6 alkyl) O(C ⁇ -C6 alkyl)-, (C ⁇ -C6 alkyl)C(O)2(
  • substituted aryl As used herein, the terms “substituted aryl”, “substituted heterocycle”, “substituted heteroaryl”, “substituted cycloalkyl”, “substituted benzyl”, “substituted aralkyl” and “substituted heterocyclylalkyl”, “substituted heterocycloalkyl” unless otherwise defined, are intended to include the cyclic group containing from 1 to 3 substitutents in addition to the point of attachment to the rest of the compound.
  • Such substitutents are preferably selected from the group which includes but is not limited to F, Cl, Br, I, CF3, OCF3, NH2, N(C ⁇ -C6 alkyl)2, NO2, CN, N3, C1-C20 alkyl,
  • the substituted Ci-6 alkyl, substituted C2-6 alkenyl, substituted C2-6 alkynyl, substituted C3-6 cycloalkyl, substituted aroyl, substituted aryl, substituted heteroaroyl, substituted arylsulfonyl, substituted heteroarylsulfonyl and substituted heterocycle include moieties containing from 1 to 3 substituents in addition to the point of attachment to the rest of the compound.
  • R6 and R7 or R5 and R7 are joined to form a ring, is illustrated by, but not limited to, the following:
  • Rla and Ri are independently selected from: hydrogen,
  • Rlc is independently selected from: hydrogen, or unsubstituted or substituted C ⁇ -C6 alkyl.
  • Rid is selected from -N(RlO)2, OR 10, unsubstituted or substituted C ⁇ -C6 alkyl, unsubstituted or substituted aryl, or unsubstituted or substituted heterocycle.
  • R2 is selected from: hydrogen and unsubstituted or substituted C ⁇ -C6 alkyl.
  • R and R7 are selected from: hydrogen, unsubstituted or substituted C ⁇ -C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted cycloalkyl.
  • R8 is selected from hydrogen, halo, unsubstituted or substituted C ⁇ -6 alkyl, unsubstituted or substituted Cl-6 alkoxy, unsubstituted or substituted aryl, CN, NO2, Rl0C(O)NRl0-, -ORlOand (RlO) 2 NC(O)-.
  • r is 1 to 3 and at least one R8 is CN.
  • R9 is hydrogen or unsubstituted or substituted C ⁇ -C6 alkyl. Most preferably, R is methyl.
  • Al and A2 are independently selected from: a bond,
  • V is selected from heteroaryl and aryl. More preferably, V is phenyl or pyridyl.
  • X and Y are bonds.
  • Zl and Z2 are independently selected from unsubstituted or substituted phenyl, unsubstituted or substituted naphthyl, unsubstituted or substituted pyridyl, unsubstituted or substituted furanyl and unsubstituted or substituted thienyl. More preferably, Zl is selected from unsubstituted or substituted phenyl and unsubstituted or substituted naphthyl. More preferably, Z2 is selected from a bond and unsubstituted or substituted phenyl.
  • W is selected from imidazolyl, oxazolyl, pyridyl, and triazolyl. More preferably, W is selected from imidazolyl and pyridyl. Most preferably, W is imidazolyl.
  • n 0, 1, or 2.
  • r is 1 or 2.
  • p is 0, 1, 2 or 3.
  • s is 0 or 1.
  • any substituent or variable e.g., Rla, R9, n, etc.
  • -N(RlO)2 represents -NHH, -NHCH3, -NHC2H5, etc. It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials.
  • the pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed, e.g., from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like: and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.
  • the pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a basic moiety by conventional chemical methods. Generally, the salts are prepared either by ion exchange chromatography or by reacting the free base with stoichio- metric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents.
  • Pd2(dba)2 Tiis(dibenzylideneacetone)dipalladinm (0)
  • PYBOP Benzotriazole-1-yl-oxy-trispyrrolidinophosphonium hexafluorophosphate
  • TFA Trifluoroacetic acid
  • THF Tetrahydrofuran
  • Reactions used to generate the compounds of this invention are prepared by employing reactions as shown in the Schemes 1-11, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures. These reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the reactions described in the Schemes.
  • the procedures discussed and illustrated in the following schemes and synopsis may be used in the preparation of the compounds of the instant invention, for either (R) or (S) stereochemistry.
  • variable n is defined as 0 to 6
  • variable p is defined as 0 to 4
  • variable t is defined as 0 or 1.
  • R8 in the schemes represents (R8)r in formula I.
  • substituent Z represents Zl or Z2 of formula I
  • substitutent R represents the optional substitutions provided in the defintion of Zl or Z of formula I.
  • Schemes 1-4 outline the syntheses of various aminopyrrolidine or aminopiperidine intermediates used in the syntheses of the macrocyclic compounds of the instant invention.
  • Boc-protected amino heterocycles 1 and 6 available commerically or by procedures known to those skilled in the art, can be sulfonylated as in Schemes 1 and 2 to give compounds 2 and 7.
  • Compounds 2 and 7 can then deprotected to give intermediates 5 and 8, respectively, which are used in Scheme 5, for example, to form the macrocyclic compounds of the instant invention.
  • Scheme 5 outlines a generic synthesis of the macrocyclic compounds where Rid is OH.
  • the aldehyde 13 reacts with an organolithium or Grignard reagent to give the addition product 14 followed by oxidation to the ketone 15.
  • Addition of the requisite organolithium or Grignard to ketone 15 provides compound 16 which then is oxidized to aldehyde 17.
  • Reductive alkylation of intermediate 5 with compound 17 provides compound 18. Cyclization of 18 with cesium carbonate yields the macrocyclic compound 19 of the instant invention.
  • Scheme 6 illustrates the preparation of the macrocyclic compounds of the instant invention wherein W is imidazole and Rid is NH2- Imidazole 20 is masked at the 2-position with TMSCl, then metallated a second time and reacted with aldehyde 13 to give compound 21.
  • oxidation and treatment with t-butylsulfinamide provides compound 23.
  • Treatment of 23 with silyl-protected oxyalkyl Grignard reagent followed by removal of the silyl group gives alcohol 24.
  • Oxidation of the alcohol gives aldehyde 25 which reductively alkylates amine 5, for example, to give intermediate 26.
  • Cyclization with cesium carbonate provides the macrocycle compound 28 after deprotection.
  • Scheme 7 shows an alternative method for the preparation of aldehyde 25.
  • Compound 23 is reacted with an alkenyl Grignard to give compound 29.
  • Subsequent oxidative cleavage of 29 provides aldehyde 25 which can be carried on as illustrated in Scheme 6.
  • Scheme 8 describes the preparation of the macrocyclic compounds of the instant invention wherein the macrocycle is linked by an amide bond.
  • Aldehyde 25 is oxidized to carboxylic acid 30 which is further coupled to compound 4. Heating of aldehyde 25 with cesium carbonate removes the mesylate and forms a macrocyclic compound of the instant invention 32, after deprotection.
  • Scheme 9 illustrates macrocyclic compounds of the instant invention where the constraining heterocycle is an aminopyrrolidinone.
  • the pyrrolidinone 36 was reductively alkylated with aldehyde 25 to give compound 37. Cyclization of 37 with cesium carbonate followed by deprotection leads to the macrocyclic compound 39 of the instant invention.
  • R is alkyl
  • R a H or CH 3
  • the compounds of the invention are selective inhibitors of farnesyl-protein transferase.
  • a compound is considered a selective inhibitor of farnesyl-protein transferase, for example, when its in vitro farnesyl-protein transferase inhibitory activity, as assessed by the assay described in Example 5, is at least 100 times greater than the in vitro activity of the same compound against geranylgeranyl-protein transferase-type I in the assay described in Example 6.
  • a selective compound exhibits at least 1000 times greater activity against one of the enzymatic activities when comparing geranylgeranyl-protein transferase-type I inhibition and farnesyl-protein transferase inhibition.
  • the selective inhibitor of farnesyl-protein transferase is further characterized by: a) an IC50 (a measure of in vitro inhibitory activity) for inhibition of the prenylation of newly synthesized K-Ras protein more than about 100-fold higher than the EC50 for the inhibition of the farnesylation of hDJ protein.
  • Example 10 When measuring such IC50S and EC50S the assays described in Example 10 may be utilized.
  • the selective inhibitor of farnesyl-protein transferase is further characterized by: b) an IC50 (a measurement of in vitro inhibitory activity) for inhibition of
  • K4B-Ras dependent activation of MAP kinases in cells at least 100-fold greater than the EC50 for inhibition of the farnesylation of the protein hDJ in cells.
  • the selective inhibitor of farnesyl-protein transferase is further characterized by: c) an IC50 (a measurement of in vitro inhibitory activity) against H-Ras dependent activation of MAP kinases in cells at least 1000 fold lower than the inhibitory activity (IC50) against H-r ⁇ -.-CNLL (SEQ.ID. ⁇ O.: 1) dependent activation of MAP kinases in cells.
  • IC50 a measurement of in vitro inhibitory activity
  • H-r ⁇ -.-CNLL SEQ.ID. ⁇ O.: 1
  • the compounds of the invention are dual inhibitors of farnesyl-protein transferase and geranylgeranyl-protein transferase type I.
  • a dual inhibitor may be termed a Class II prenyl-protein transferase inhibitor and will exhibit certain characteristics when assessed in in vitro assays, which are dependent on the type of assay employed.
  • the dual inhibitor compound has an in vitro inhibitory activity (IC50) that is less than about 12 ⁇ M against K4B-Ras dependent activation of MAP kinases in cells.
  • IC50 in vitro inhibitory activity
  • the Class II prenyl-protein transferase inhibitor may also be characterized by: a) an IC50 (a measurement of in vitro inhibitory activity) for inhibiting
  • the Class II prenyl-protein transferase inhibitor may also be characterized by: a) an IC50 (a measurement of in vitro inhibitory activity) against H-Ras dependent activation of MAP kinases in cells greater than 2 fold lower but less than 20,000 fold lower than the inhibitory activity (IC50) against
  • H-r -.-CNLL SEQ.ID. ⁇ O.: 1 dependent activation of MAP kinases in cells; and b) an IC50 (a measurement of in vitro inhibitory activity) against H-ras-
  • the Class II prenyl-protein transferase inhibitor may also be characterized by: a) an IC50 (a measurement of in vitro inhibitory activity) against H-Ras dependent activation of MAP kinases in cells greater than 10-fold lower but less than 2,500 fold lower than the inhibitory activity (IC50) against H-ras-
  • CNLL (SEQ.ID. ⁇ O.: 1) dependent activation of MAP kinases in cells; and b) an IC50 (a measurement of in vitro inhibitory activity) against H-ras- CNLL dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with the pCMN-SEAP plasmid that constitutively expresses the SEAP protein.
  • IC50 inhibitory activity
  • a compound of the instant invention may be a more potent inhibitor of geranylgeranyl-protein transferase-type I than it is an inhibitor of farnesyl-protein transferase.
  • the instant compounds are useful as pharmaceutical agents for mammals, especially for humans. These compounds may be administered to patients for use in the treatment of cancer.
  • Examples of the type of cancer which may be treated with the compounds of this invention include, but are not limited to, colorectal carcinoma, exocrine pancreatic carcinoma, myeloid leukemias and neurological tumors. Such tumors may arise by mutations in the ras genes themselves, mutations in the proteins that can regulate Ras activity (i.e., neurofibromin ( ⁇ F-1), neu, src, abl, lck, fyn) or by other mechanisms.
  • Ras activity i.e., neurofibromin ( ⁇ F-1), neu, src, abl, lck, fyn
  • the compounds of the instant invention inhibit farnesyl-protein transferase and the farnesylation of the oncogene protein Ras.
  • the instant compounds may also inhibit tumor angiogenesis, thereby affecting the growth of tumors (J. Rak et al. Cancer Research, 55:4575-4580 (1995)).
  • Such anti-angiogenesis properties of the instant compounds may also be useful in the treatment of certain forms of vision deficit related to retinal vascularization.
  • the compounds of this invention are also useful for inhibiting other proliferative diseases, both benign and malignant, wherein Ras proteins are aberrantly activated as a result of oncogenic mutation in other genes (i.e., the Ras gene itself is not activated by mutation to an oncogenic form) with said inhibition being accomplished by the administration of an effective amount of the compounds of the invention to a mammal in need of such treatment.
  • the composition is useful in the treatment of neurofibromatosis, which is a benign proliferative disorder.
  • the instant compounds may also be useful in the treatment of certain viral infections, in particular in the treatment of hepatitis delta and related viruses (J.S. Glenn et al. Science, 256:1331-1333 (1992).
  • the compounds of the instant invention are also useful in the prevention of restenosis after percutaneous transluminal coronary angioplasty by inhibiting neointimal formation (C. Indolfi et al. Nature medicine, 1:541-545(1995).
  • the instant compounds may also be useful in the treatment and prevention of polycystic kidney disease (D.L. Schaffner et al. American Journal of Pathology, 142:1051-1060 (1993) and B. Cowley, Jr. et al., FASEB Journal, 2: A3160 (1988)).
  • the instant compounds may also be useful for the treatment of fungal infections.
  • the instant compounds may also be useful as inhibitors of proliferation of vascular smooth muscle cells and therefore useful in the prevention and therapy of arteriosclerosis and diabetic vascular pathologies.
  • the compounds of the instant invention may also be useful in the prevention and treatment of endometriosis, uterine fibroids, dysfunctional uterine bleeding and endometrial hyperplasia.
  • the prenyl-protein transferase inhibitors of the instant invention may also be co- administered with other well known therapeutic agents that are selected for their particular usefulness against the condition that is being treated.
  • the prenyl-protein transferase inhibitor may be useful in further combination with drugs known to supress the activity of the ovaries and slow the growth of the endometrial tissue.
  • drugs include but are not limited to oral contraceptives, progestins, danazol and GnRH (gonadotropin-releasing hormone) agonists.
  • Administration of the prenyl-protein transferase inhibitor may also be combined with surgical treatment of endometriosis (such as surgical removal of misplaced endometrial tissue) where appropriate.
  • the instant compounds may also be useful as inhibitors of corneal inflammation. These compounds may improve the treatment of corneal opacity which results from cauterization-induced corneal inflammation. The instant compounds may also be useful in reducing corneal edema and neovascularization. (K. Sonoda et al., Invest. Ophthalmol. Vis. Sci., 1998, vol. 39, p 2245-2251).
  • the compounds of this invention may be administered to mammals, preferably humans, either alone or, preferably, in combination with pharmaceutically acceptable carriers, excipients or diluents, in a pharmaceutical composition, according to standard pharmaceutical practice.
  • the compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
  • the compounds of the instant invention may be administered to a mammal in need thereof using a gel extrusion mechanism (GEM) device, such as that described in US Serial No. 60/144,643, filed on July 20, 1999, which is hereby incorporated by reference.
  • GEM gel extrusion mechanism
  • composition is intended to encompass a product comprising the specified ingredients in the specific amounts, as well as any product which results, directly or indirectly, from combination of the specific ingredients in the specified amounts.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, or alginic acid; binding agents, for example starch, gelatin, polyvinyl-pyrrolidone or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to mask the unpleasant taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a water soluble taste masking material such as hydroxypropyl-methylcellulose or hydroxypropyl- cellulose, or a time delay material such as ethyl cellulose, cellulose acetate buryrate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water soluble carrier such as polyethyl- eneglycol or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water soluble carrier such as polyethyl- eneglycol or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellu- lose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethylene- oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavoring agents such as sucrose, saccharin or aspartame.
  • sweetening agents such as sucrose, saccharin or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation.
  • These compositions may be preserved by the addition of an anti-oxidant such as butylated hydroxyanisol or alpha-tocopherol.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • the pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring phospha- tides, for example soy bean lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening, flavoring agents, preservatives and antioxidants.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.
  • compositions may be in the form of a sterile injectable aqueous solutions.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • the sterile injectable preparation may also be a sterile injectable oil- in-water microemulsion where the active ingredient is dissolved in the oily phase.
  • the active ingredient may be first dissolved in a mixture of soybean oil and lecithin. The oil solution then introduced into a water and glycerol mixture and processed to form a microemulation.
  • the injectable solutions or microemulsions may be introduced into a patient's blood-stream by local bolus injection.
  • a continuous intravenous delivery device may be utilized.
  • An example of such a device is the Deltec CADD-PLUSTM model 5400 intravenous pump.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension for intramuscular and subcutaneous administration.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Compounds of Formula A may also be administered in the form of a suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
  • compositions, ointments, jellies, solutions or suspensions, etc., containing the compound of Formula A are employed.
  • topical application shall include mouth washes and gargles.
  • the compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles and delivery devices, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • Compounds of the present invention may also be delivered as a suppository employing bases such as cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
  • bases such as cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
  • the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, sex and response of the individual patient, as well as the severity of the patient's symptoms.
  • a suitable amount of compound is administered to a mammal undergoing treatment for cancer.
  • Administration occurs in an amount between about 0.1 mg/kg of body weight to about 60 mg/kg of body weight per day, preferably of between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day.
  • the compounds of the instant invention may also be co-administered with other well known therapeutic agents that are selected for their particular useful- ness against the condition that is being treated.
  • the compounds of the instant invention may also be co-administered with other well known cancer therapeutic agents that are selected for their particular usefulness against the condition that is being treated. Included in such combinations of therapeutic agents are combinations of the instant prenyl-protein transferase inhibitors and an antineoplastic agent. It is also understood that such a combination of antineoplastic agent and inhibitor of prenyl-protein transferase may be used in conjunction with other methods of treating cancer and/or tumors, including radiation therapy and surgery.
  • any of the therapeutic agents described herein may also be used in combination with a compound of the instant invention and an antineoplastic agent.
  • an antineoplastic agent include, in general, microtubule- stabilizing agents such as paclitaxel (also known as Taxol®), docetaxel (also known as Taxotere®), epothilone A, epothilone B, desoxyepothilone A, desoxyepothilone B or their derivatives); microtubule-disruptor agents; alkylating agents, for example, nitrogen mustards, ethyleneimine compounds, alkyl sulfonates and other compounds with an alkylating action such as nitrosoureas, cisplatin, and dacarbazine; anti-metabolites, for example, folic acid, purine or pyrimidine antagonists; epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxan
  • Example classes of antineoplastic agents include, for example, the anthracycline family of drugs, the vinca drugs, the mitomycins, the bleomycins, the cytotoxic nucleosides, the taxanes, the epothilones, discodermolide, the pteridine family of drugs, diynenes and the podophyllotoxins.
  • Particularly useful members of those classes include, for example, doxorubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloro-methotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine arabinoside, podophyllotoxin or podo-phyllotoxin derivatives such as etoposide, etoposide phosphate or teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine, leurosine, paclitaxel and the like.
  • antineoplastic agents include estramustine, cisplatin, carboplatin, cyclophosphamide, bleomycin, tamoxifen, ifosamide, melphalan, hexamethyl melamine, thiotepa, cytarabin, idatrexate, trimetrexate, dacarbazine, L-asparaginase, dactinomycin, mechlorethamine (nitrogen mustard), streptozocin, cyclophosphamide, carmustine (BCNU), lomustine (CCNU), procarbazine, mitomycin, cytarabine, etoposide, methotrexate, bleomycin, chlorambucil, camptothecin, CPT-11, topotecan, ara-C, bicalutamide, flutamide, leuprolide, pyridobenzoindole derivatives, interferons and interleukins
  • antineoplastic, or chemotherapeutic, agents are described, for example, by D. J. Stewart in “Nausea and Vomiting: Recent Research and Clinical Advances", Eds. J. Kucharczyk, et al., CRC Press Inc., Boca Raton, Florida, USA (1991), pages 177-203, especially page 188. See also, R. J. Gralla, et al., Cancer Treatment Reports, 68(1), 163-172 (1984).
  • the preferred class of antineoplastic agents is the taxanes and the preferred antineoplastic agent is paclitaxel.
  • the compounds of the instant invention may also be co-administered with antisense oligonucleotides which are specifically hybridizable with RNA or DNA deriving from human ras gene.
  • antisense oligonucleotides are described in U.S. Patent. No. 5,576,208 and PCT Publication No. WO 99/22772.
  • the instant compounds are particularly useful when co-administered with the antisense oligonucleotide comprising the amino acid sequence of SEQ.ID.NO: 2 of U.S. Patent No. 5,576,208.
  • Certain compounds of the instant invention may exhibit very low plasma concentrations and significant inter-individual variation in the plasma levels of the compound.
  • prenyl-protein transferase inhibitors may be due to extensive metabolism by cytochrome P450 enzymes prior to entry of drug into the systemic circulation.
  • Prenyl-protein transferase inhibitors may be metabolized by cytochrome P450 enzyme systems, such as CYP3A4, CYP2D6, CYP2C9, CYP2C19 or other cytochrome P450 isoform. If a compound of the instant invention demonstrates an affinity for one or more of the cytochrome P450 enzyme systems, another compound with a higher affinity for the P450 enzyme(s) involved in metabolism should be administered concomitantly.
  • Examples of compounds that have a comparatively very high affinity for CYP3 A4, CYP2D6, CYP2C9, CYP2C19 or other P450' isoform include, but are not limited to, piperonyl butoxide, troleandomycin, erythromycin, proadifen, isoniazid, allyliso- propylacetamide, ethinylestradiol, chloramphenicol, 2-ethynylnaphthalene and the like.
  • Such a high affinity compound when employed in combination with a compound of formula A, may reduce the inter-individual variation and increase the plasma concentration of a compound of formula A to a level having substantial therapeutic activity by inhibiting the metabolism of the compound of formula A. Additionally, inhibiting the metabolism of a compound of the instant invention prolongs the pharmacokinetic half-life, and thus the pharmacodynamic effect, of the compound.
  • a compound of the present invention may be employed in conjunction with antiemetic agents to treat nausea or emesis, including acute, delayed, late-phase, and anticipatory emesis, which may result from the use of a compound of the present invention, alone or with radiation therapy.
  • a compound of the present invention may be used in conjunction with other anti-emetic agents, especially neurokinin-1 receptor antagonists, 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, or a corticosteroid such as Decadron (dexa- methasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U.S.Patent Nos.
  • neurokinin-1 receptor antagonists especially 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, or a corticosteroid such as Decadron (dexa- methasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U.S.Pa
  • a particularly preferred neurokinin-1 receptor antagonist for use in conjunction with the compounds of the present invention is 2-(R)-(l-(R)-(3,5-bis (trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-4-(3-(5-oxo-lH,4H-l,2,4- triazolo)methyl)morpholine, or a pharmaceutically acceptable salt thereof, which is described in U.S. Patent No. 5,719,147.
  • a compound of the present invention for the treatment of cancer, it may be desirable to employ a compound of the present invention in conjunction with another pharmacologically active agent(s).
  • a compound of the present invention and the other pharmacologically active agent(s) may be administered to a patient simultaneously, sequentially or in combination.
  • the present compound may employed directly in combination with the other active agent(s), or it may be administered prior, concurrent or subsequent to the administration of the other active agent(s).
  • the currently available dosage forms of the known therapeutic agents for use in such combinations will be suitable.
  • a compound of the present invention may be presented together with another therapeutic agent in a combined preparation, such as with an antiemetic agent for simultaneous, separate, or sequential use in the relief of emesis associated with employing a compound of the present invention and radiation therapy.
  • a combined preparation may be, for example, in the form of a twin pack.
  • a preferred combination comprises a compound of the present invention with antiemetic agents, as described above.
  • Radiation therapy including x-rays or gamma rays which are delivered from either an externally applied beam or by implantation of tiny radioactive sources, may also be used in combination with the instant inhibitor of prenyl-protein transferase alone to treat cancer.
  • compounds of the instant invention may also be useful as radiation sensitizers, as described in WO 97/38697, published on October 23, 1997, and herein incorporated by reference.
  • the instant compounds may also be useful in combination with other inhibitors of parts of the signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation.
  • the instant compounds may be utilized in combination with farnesyl pyrophosphate competitive inhibitors of the activity of farnesyl-protein transferase or in combination with a compound which has Raf antagonist activity.
  • the instant compounds may also be co-administered with compounds that are selective inhibitors of geranylgeranyl protein transferase.
  • the compound of the instant invention is a selective inhibitor of farnesyl-protein transferase
  • co-administration with a compound(s) that is a selective inhibitor of geranylgeranyl protein transferase may provide an improved therapeutic effect.
  • such administration can be orally or parenterally, including intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration. It is preferred that such administration be orally. It is more preferred that such administration be orally and simultaneously.
  • the protein substrate-competitive inhibitor and farnesyl pyrophosphate-competitive inhibitor are administered sequentially, the administration of each can be by the same method or by different methods.
  • an integrin antagonist refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to an integrin(s) that is involved in the regulation of angiogenisis, or in the growth and invasiveness of tumor cells.
  • the term refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the v ⁇ 3 integrin, which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ⁇ v ⁇ 5 integrin, which antagonize, inhibit or counteract binding of a physiological ligand to both the ⁇ v ⁇ 3 integrin and the ⁇ v ⁇ 5 integrin, or which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells.
  • the term also refers to antagonists of the ⁇ l ⁇ l, 2 ⁇ l, ⁇ 5 ⁇ l, 6 ⁇ l and ⁇ 6 ⁇ 4 integrins.
  • the term also refers to antagonists of any combination of v ⁇ 3 integrin, ocv ⁇ 5 integrin, l ⁇ l, 2 ⁇ l, cc5 ⁇ l, ⁇ 6 ⁇ l and ⁇ 6 ⁇ 4 integrins.
  • the instant compounds may also be useful with other agents that inhibit angiogenisis and thereby inhibit the growth and invasiveness of tumor cells, including, but not limited to angiostatin and endostatin.
  • HMG-CoA reductase 3-hydroxy-3-methyIglutaryl-CoA reductase
  • Compounds which have inhibitory activity for HMG-CoA reductase can be readily identified by using assays well-known in the art. For example, see the assays described or cited in U.S. Patent 4,231,938 at col. 6, and WO 84/02131 at pp. 30-33.
  • the terms "HMG-CoA reductase inhibitor” and "inhibitor of HMG-CoA reductase” have the same meaning when used herein.
  • HMG-CoA reductase inhibitors examples include but are not limited to lovastatin (MENACOR®; see US Patent No. 4,231,938; 4,294,926; 4,319,039), simvastatin (ZOCOR®; see US Patent No. 4,444,784; 4,820,850; 4,916,239), pravastatin (PRAVACHOL®; see US Patent Nos. 4,346,227; 4,537,859; 4,410,629; 5,030,447 and 5,180,589), fluvastatin (LESCOL®; see US Patent Nos.
  • HMG-CoA reductase inhibitor as used herein includes all pharmaceutically acceptable lactone and open-acid forms (i.e., where the lactone ring is opened to form the free acid) as well as salt and ester forms of compounds which have HMG-CoA reductase inhibitory activity, and therefor the use of such salts, esters, open-acid and lactone forms is included within the scope of this invention.
  • An illustration of the lactone portion and its corresponding open-acid form is shown below as structures I and II.
  • HMG-CoA reductase inhibitors where an open-acid form can exist
  • salt and ester forms may preferably be formed from the open-acid, and all such forms are included within the meaning of the term "HMG-CoA reductase inhibitor" as used herein.
  • the HMG-CoA reductase inhibitor is selected from lovastatin and simvastatin, and most preferably simvastatin.
  • the term "pharmaceutically acceptable salts" with respect to the HMG-CoA reductase inhibitor shall mean non- toxic salts of the compounds employed in this invention which are generally prepared by reacting the free acid with a suitable organic or inorganic base, particularly those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc and tetramethylammonium, as well as those salts formed from amines such as ammonia, ethylenediamine, N-methylglucamine, lysine, arginine, ornithine, choline, N,N'-dibenzylethylenediamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, l-p-chlorobenzyl-2-pyrrolidine-l'-yl-methyl- benzimidazole, diethylamine, piperazine, and tris(hydroxymethyl) aminomethane.
  • a suitable organic or inorganic base particularly those formed from
  • salt forms of HMG-CoA reductase inhibitors may include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynapthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylsulfate, mucate, napsylate, nitrate, oleate, oxalate, pamao
  • Ester derivatives of the described HMG-CoA reductase inhibitor compounds may act as prodrugs which, when absorbed into the bloodstream of a warm-blooded animal, may cleave in such a manner as to release the drug form and permit the drug to afford improved therapeutic efficacy.
  • the instant compounds may be useful in combination with agents that are effective in the treatment and prevention of NF-1, restenosis, polycystic kidney disease, infections of hepatitis delta and related viruses and fungal infections.
  • combination products employ the combinations of this invention within the dosage range described above and the other pharmaceutically active agent(s) within its approved dosage range.
  • Combinations of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a multiple combination formulation is inappropriate.
  • the instant compounds may also be useful in combination with prodrugs of antineoplastic agents.
  • the instant compounds may be co-administered either concurrently or sequentially with a conjugate (termed a "PSA conjugate") which comprises an oligopeptide, that is selectively cleaved by enzymatically active prostate specific antigen (PSA), and an antineoplastic agent.
  • a conjugate termed a "PSA conjugate”
  • PSA conjugate which comprises an oligopeptide, that is selectively cleaved by enzymatically active prostate specific antigen (PSA), and an antineoplastic agent.
  • PSA conjugate conjugate
  • PSA prostate specific antigen
  • the compounds of the instant invention are also useful as a component in an assay to rapidly determine the presence and quantity of farnesyl-protein transferase (FPTase) in a composition.
  • FPTase farnesyl-protein transferase
  • the composition to be tested may be divided and the two portions contacted with mixtures which comprise a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl pyrophosphate and, in one of the mixtures, a compound of the instant invention.
  • the chemical content of the assay mixtures may be determined by well known immuno-logical, radiochemical or chromatographic techniques. Because the compounds of the instant invention are selective inhibitors of FPTase, absence or quantitative reduction of the amount of substrate in the assay mixture without the compound of the instant invention relative to the presence of the unchanged substrate in the assay containing the instant compound is indicative of the presence of FPTase in the composition to be tested.
  • potent inhibitor compounds of the instant invention may be used in an active site titration assay to determine the quantity of enzyme in the sample.
  • a series of samples composed of aliquots of a tissue extract containing an unknown amount of farnesyl- protein transferase, an excess amount of a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl pyrophosphate are incubated for an appropriate period of time in the presence of varying concentrations of a compound of the instant invention.
  • concentration of a sufficiently potent inhibitor i.e., one that has a Ki substantially smaller than the concentration of enzyme in the assay vessel
  • concentration of a sufficiently potent inhibitor i.e., one that has a Ki substantially smaller than the concentration of enzyme in the assay vessel
  • Step B Preparation of 4-bromo-3-fluorobenzyl alcohol 4-Bromo-3-fluorobenzoic acid (40.8 g, 0.187 mol) was dissolved in THF (250 ml) with magnetic stirring under Ar in an ice-H2 ⁇ bath. The cloudy solution was treated dropwise with borane-THF complex (1 M) (374 mL, 0.374 mol) over a 1 hour period maintaining the internal temperature at ⁇ 10°C. The reaction mixture was left to warm to ambient temperature overnight, then cooled in an ice-H2 ⁇ bath and treated dropwise with H2O (150 mL). The THF was removed on a rotary evaporator, and the residue partitioned between EtOAc and H2O.
  • Step E Preparation of 2-Fluoro-4-[hydroxy-(3-methyl-3H-imidazol-4-yl)- methyll -benzonitrile
  • Step F Preparation of 2-Fluoro-4-(3-methyl-3H-imidazole-4-carbonyl)- benzonitrile 2-Fluoro-4-[hydroxy-(3-methyl-3H-imidazol-4-yl)-methyl]- benzonitrile (0.655 g, 2.83 mmol) and Mn ⁇ 2 (1.23 g, 14.2 mmol) were stirred in C ⁇ 2CI2 (50 mL) and CH3CN (5 mL) for 3 hours. The solution was filtered and concentrated to yield the title compound.
  • Step H Preparation of N-[l-(4-cyano-3-fluoro-phenyl)-l-(3-methyl-
  • Step I Preparation of 4-[(lS)-amino-l-(3-methyl-3H-imidazol-4-yl)- ethyll -2-fluoro-benzoni trile
  • Step J Preparation of l-(3-methoxy-benzenesulfonyl)-piperidine-3-carboxylic acid ethyl ester
  • Step K Preparation of l-(3-methoxy-benzenesulfonyl)-piperidine-3-carboxylic acid
  • l-(3-methoxy-benzenesulfonyl)-piperidine-3R- carboxylic acid ethyl ester 2.02 g, 6.16 mmol
  • absolute ethanol 20 mL
  • 50% w/w NaOH 0.26 mL, 6.47 mmol
  • Step L Preparation of 3-N-[l-(4-cyano-3-fluorophenyl)-l-(l-methyl-lH- imidazol-5-yl)ethyl]-l-[(3-methoxyphenyl)sulfonyl] piperidine-3- carboxamide
  • Step I 4-[(lR)-amino-l-(3-methyl-3H-imidazol-4-yl)-ethyl]-2-fluoro- benzonitrile (Step I) (0.192 g, 0.78 mmol), l-(3-methoxy-benzenesulfonyl)- piperidine-3-carboxylic acid, (0.235 g, 0.78 mmol), EDC (0.284 g, 1.5 mmol), ⁇ OBt (0.230 g, 1.5 mmol) and ⁇ -methylmorpholine (0.172 mL, 1.5 mmol) were stirred in DMF (4 mL) at ambient temperature overnight under ⁇ 2.
  • Step M Preparation of 3-N-[l-(4-cyano-3-fluorophenyl)-l-(l-methyl-lH- imidazol-5-yl)ethyl]-l-[(3-hydroxyphenyl)sulfonyl] piperidine-3- carboxamide
  • Step N Preparation of 5-[(17i?)-10-cyano-14R-methyl-2,2-dioxido-16- oxo-8-oxa-2-thia-l,15-diazatetracyclo[15.3.1.1 3 ' 7 .l 9 ' 13 ]tricosa- 3(23),4,6,9(22),10,12-hexaen-14-yl]-l-methyl-lH-imidazol-l- ium trifluoroacetate
  • the mixture was stirred at 60°C for 17 hours, taken up in water, concentrated to a small volume, and purified on a Waters Delta PrepPak ⁇ PLC column using a 0.1% trifluoroacetic acid water: 0.1% trifluoracetic acid/acetonitrile, 95:5 to 5:95 gradient over 1 hour.
  • Step B Preparation of Magnesium 3-benzyloxyphenylsulfinate bromide
  • benzyl 3-bromophenyl ether as described above in Step A, (5.0 g, 19 mmol) in dry THF (50 mL) dropwise, at a rate that maintained reflux with the heat source removed.
  • the resulting mixture was heated to reflux for 30 min, then cooled to -78°C and SO2 was bubbled into the solution slowly for 1 hr.
  • the reaction mixture was allowed to warm slowly to ambient temperature and stirred for 18 hours. Argon was bubbled into the mixture for 1 hour, then the solution was concentrated in vacuo to give the desired product as a solid.
  • Step C Preparation of 3-Benzyloxyphenylsulfonyl chloride
  • 2-benzyloxyphenylsulfonyl chloride 4.5 g, 14 mmol
  • Sulfuryl chloride (19 mL of a 1.0 M solution in CH2CI2, 19 mmol) was added dropwise, and the mixture was stirred for 18 hrs at ambient temperature, during which time a precipitate formed.
  • the precipitate was removed by filtration and the filtrate was concentrated under reduced pressure to give the product as a yellow oil.
  • Step D Preparation of (R)- 1 -(3-Benzyloxyphenylsulf onyl)-3-(tert- butoxycarbonylamino ' )pyrrolidine
  • Step E Preparation of (R)-3-(tert-Butoxycarbonylamino)-l-(3- hydroxyphenylsulfonyPpyrrolidine
  • Step F Preparation of (R)-3-Amino-l-(3-hydroxyphenyl-sulfonyl)pyrrolidine
  • Step G Preparation of N-[4- ⁇ [tert-butyl(dimethyl)silyl]oxy ⁇ -l-(4-cyano-3- fluorophenyl)- 1 -( 1 -methyl- lH-imidazol-5-yl)butyl] -2-methylpropane-
  • Step H Preparation of N-[l-(4-cyano-3-fluorophenyl)-4-hydroxy-l(S and R)-
  • Step I Preparation of N-[l-(4-cyano-3-fluorophenyl)-l-(l-methyl-lH- imidazol-5-ylV4-oxobutyll-2-methylpropane-2-sulfinamide
  • Step J Preparation of N-[l-(4-cyano-3-fluorophenyl)-4-( ⁇ (3S)-l-[(3- hydroxyphenyl)sulfonyl]pyrrolidin-3-yl ⁇ amino)- 1 -( 1 -methyl- 1H- imidazol-5-yl butyll-2-methylpropane-2-sulfinamide N-[l-(4-Cyano-3-fluorophenyl)-l-(l-methyl-lH-imidazol-5-yl)-4- oxobutyl]-2-methylpropane-2-sulfinamide (0.028 g, 0.0717 mmol) and (S)-3-Amino- l-(3-hydroxyphenylsulfonyl) pyrrolidine hydrochloride (0.021 g, 0.0753 mmol) were dissolved in CH3OH (5 mL) with stirring at ambient temperature, the pH of the solution adjusted to 5 with
  • Step K Preparation of N-[(6S)-15-cyano-l 1-(1 -methyl- lH-imidazol-5-yl)-
  • reaction mixture was concentrated in vacuo, partitioned between aqueous saturated NaHCO3 solution and CH2CI2.
  • the aqueous layer was washed with CH2CI2 (2x), organics combined, washed with brine, dried (MgSO ), filtered and concentrated to dryness to give the title compound.
  • Isoprenyl-protein transferase activity assays are carried out at 30°C unless noted otherwise.
  • a typical reaction contains (in a final volume of 50 ⁇ L): [ 3 H]farnesyl diphosphate, Ras protein, 50 mM HEPES, pH 7.5, 5 mM MgCl2, 5 mM dithiothreitol, 10 ⁇ M ZnCl2, 0.1% polyethyleneglycol (PEG) (15,000-20,000 mw) and isoprenyl-protein transferase.
  • the FPTase employed in the assay is prepared by recombinant expression as described in Omer, C.A., Krai, A.M., Diehl, R.E., Prendergast, G.C., Powers, S., Allen, CM., Gibbs, J.B. and Kohl, N.E. (1993) Biochemistry 32:5167-5176. After thermally pre-equilibrating the assay mixture in the absence of enzyme, reactions are initiated by the addition of isoprenyl- protein transferase and stopped at timed intervals (typically 15 min) by the addition of 1 M HCI in ethanol (1 mL). The quenched reactions are allowed to stand for 15 minutes (to complete the precipitation process).
  • inhibitors are prepared as concentrated solutions in 100% dimethyl sulfoxide and then diluted 20-fold into the enzyme assay mixture.
  • Substrate concentrations for inhibitor IC50 determinations are as follows: FTase, 650 nM Ras-CNLS (SEQ.ID. ⁇ O.: 2), 100 nM farnesyl diphosphate.
  • the compounds of the instant invention are tested for inhibitory activity against human FPTase by the assay described above.
  • the modified geranylgeranyl-protein transferase inhibition assay is carried out at room temperature.
  • a typical reaction contains (in a final volume of
  • the GGTase-type I enzyme employed in the assay is prepared as described in U.S. Patent No. 5,470,832, incorporated by reference.
  • the Ras peptide is derived from the K4B-Ras protein and has the following sequence: biotinyl-G-O-XKKKSKTKCNIM (single amino acid code) (SEQ.-D. ⁇ O.: 3). Reactions are initiated by the addition of GGTase and stopped at timed intervals (typically 15 min) by the addition of 200 ⁇ L of a 3 mg mL suspension of streptavidin SPA beads (Scintillation Proximity Assay beads, Amersham) in 0.2 M sodium phosphate, pH 4, containing 50 mM EDTA, and 0.5% BSA. The quenched reactions are allowed to stand for 2 hours before analysis on a Packard TopCount scintillation counter.
  • biotinyl-G-O-XKKKSKTKCNIM single amino acid code
  • IC50 values are determined with Ras peptide near KM concentrations. Enzyme and substrate concentrations for inhibitor IC50 determinations are as follows: 75 pM GGTase-I, 1.6 ⁇ M Ras peptide, 100 nM geranylgeranyl diphosphate.
  • the compounds of the instant invention are tested for inhibitory activity against human GGTase-type I by the assay described above.
  • the cell line used in this assay is a v-ras line derived from either Ratl or ⁇ IH3T3 cells, which expressed viral Ha-ras p21.
  • the assay is performed essentially as described in DeClue, J.E. et al., Cancer Research 51:712-717, (1991). Cells in 10 cm dishes at 50-75% confluency are treated with the test compound (final concentration of solvent, methanol or dimethyl sulfoxide, is 0.1%).
  • the cells are labeled in 3 ml methionine-free DMEM supple-mented with 10% regular DMEM, 2% fetal bovine serum and 400 ⁇ Ci[35s]methionine (1000 Ci/mmol).
  • the cells are lysed in 1 ml lysis buffer (1% NP40/20 mM HEPES, pH 7.5/5 mM MgCl2/lmM DTT/10 mg/ml aprotinen/2 mg/ml leupeptin/2 mg/ml antipain/0.5 mM PMSF) and the lysates cleared by centrifugation at 100,000 x g for 45 minutes.
  • the immuno-precipitates are washed four times with IP buffer (20 nM HEPES, pH 7.5/1 mM EDTA/1% Triton X-100.0.5% deoxycholate/ 0.1%/SDS/O.l M NaCl) boiled in SDS-PAGE sample buffer and loaded on 13% acrylamide gels. When the dye front reached the bottom, the gel is fixed, soaked in Enlightening, dried and autoradiographed. The intensities of the bands corresponding to farnesylated and nonfarnesylated ras proteins are compared to determine the percent inhibition of farnesyl transfer to protein.
  • IP buffer 20 nM HEPES, pH 7.5/1 mM EDTA/1% Triton X-100.0.5% deoxycholate/ 0.1%/SDS/O.l M NaCl
  • Rat 1 cells transformed with either v-ras, v-raf, or v-mos are seeded at a density of 1 x 10 ⁇ cells per plate (35 mm in diameter) in a 0.3% top agarose layer in medium A (Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum) over a bottom agarose layer (0.6%). Both layers contain 0.1% methanol or an appropriate concentration of the instant compound (dissolved in methanol at 1000 times the final concentration used in the assay).
  • the cells are fed twice weekly with 0.5 ml of medium A containing 0.1% methanol or the concentra- tion of the instant compound. Photomicrographs are taken 16 days after the cultures are seeded and comparisons are made.
  • the SEAP reporter plasmid, pDSElOO was constructed by ligating a restriction fragment containing the SEAP coding sequence into the plasmid pCMV- RE-AKI.
  • the SEAP gene is derived from the plasmid pSEAP2-Basic (Clontech, Palo Alto, CA).
  • the plasmid pCMN-RE-AKI was constructed by Deborah Jones (Merck) and contains 5 sequential copies of the 'dyad symmetry response element' cloned upstream of a 'CAT-TATA' sequence derived from the cytomegalovirus immediate early promoter.
  • the plasmid also contains a bovine growth hormone poly-A sequence.
  • the plasmid, pDSElOO was constructed as follows.
  • a restriction fragment encoding the SEAP coding sequence was cut out of the plasmid pSEAP2- Basic using the restriction enzymes EcoRl and Hpal. The ends of the linear DNA fragments were filled in with the Klenow fragment of E. coli DNA Polymerase I. The 'blunt ended' DNA containing the SEAP gene was isolated by electrophoresing the digest in an agarose gel and cutting out the 1694 base pair fragment.
  • the vector plasmid pCMV-RE-AKI was linearized with the restriction enzyme Bgl-II and the ends filled in with Klenow DNA Polymerase I.
  • the SEAP DNA fragment was blunt end ligated into the pCMN-RE-AKI vector and the ligation products were transformed into DH5-alpha E.
  • coli cells (Gibco-BRL). Transformants were screened for the proper insert and then mapped for restriction fragment orientation. Properly oriented recombinant constructs were sequenced across the cloning junctions to verify the correct sequence. The resulting plasmid contains the SEAP coding sequence downstream of the DSE and CAT-TATA promoter elements and upstream of the BGH poly-A sequence.
  • the SEAP repotrer plasmid, pDSElOl is also constructed by ligating a restriction fragment containing the SEAP coding sequence into the plasmid pCMN- RE-AKI.
  • the SEAP gene is derived from plasmid pGEM7zf (-)/SEAP.
  • the plasmid pDSElOl was constructed as follows: A restriction fragment containing part of the SEAP gene coding sequence was cut out of the plasmid pGEM7zf(-)/SEAP using the restriction enzymes Apa I and Kpnl. The ends of the linear D ⁇ A fragments were chewed back with the Klenow fragment of E. coli D ⁇ A Polymerase I.
  • the "blunt ended" D ⁇ A containing the truncated SEAP gene was isolated by electrophoresing the digest in an agarose gel and cutting out the 1910 base pair fragment. This 1910 base pair fragment was ligated into the plasmid pCMN-RE-AKI which had been cut with Bgl-II and filled in with E. coli Klenow fragment D ⁇ A polymerase. Recombinant plasmids were screened for insert orientation and sequenced through the ligated junctions.
  • the plasmid pCMN-RE-AKI is derived from plasmid pCMNIE-AKI-DHFR (Whang , Y., Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E. (1987) J. Virol., 61, 1796- 1807) by removing an EcoRI fragment containing the DHFR and Neomycin markers.
  • the plasmid pGEM7zf(-)/SEAP was constructed as follows.
  • the SEAP gene was PCRed, in two segments from a human placenta cD ⁇ A library (Clontech) using the following oligos.
  • Sense strand ⁇ -terminal SEAP 5' GAGAGGGAATTCGGGCCCTTCCTGCAT GCTGCTGCTGCTGCTGCTGCTGGGC 3' (SEQ.ID. ⁇ O. :4)
  • Antisense strand ⁇ -terminal SEAP 5' GAGAGAGCTCGAGGTTAACCCGGGT GCGCGGCGTCGGTGGT 3' (SEQ.ID. ⁇ O.:5)
  • Sense strand C-terminal SEAP 5' GAGAGAGTCTAGAGTTAACCCGTGGTCC CCGCGTTGCTTCCT 3' (SEQ.ID.NO.: 6)
  • Antisense strand C-terminal SEAP 5' GAAGAGGAAGCTTGGTACCGCCACTG GGCTGTAGGTGGTGGCT 3' (SEQ.ID.NO.:7)
  • the N-terminal oligos (SEQ.ID.NO.: 4 and SEQ.ID.NO.: 5) were used to generate a 1560 bp N-terminal PCR product that contained EcoRI and Hpal restriction sites at the ends.
  • the Antisense N-terminal oligo (SEQ.ID.NO.: 5) introduces an internal translation STOP codon within the SEAP gene along with the Hpal site.
  • the C-terminal oligos (SEQ.ID.NO.: 6 and SEQ.ID.NO.: 7) were used to amplify a 412 bp C-terminal PCR product containing Hpal and Hind-XT restriction sites.
  • the sense strand C-terminal oligo introduces the internal STOP codon as well as the Hpal site.
  • the N-terminal amplicon was digested with EcoRI and Hpal while the C-terminal amplicon was digested with Hpal and Hindlll.
  • the two fragments comprising each end of the SEAP gene were isolated by electro-phoresing the digest in an agarose gel and isolating the 1560 and 412 base pair fragments. These two fragments were then co-ligated into the vector pGEM7zf (-) (Promega) which had been restriction digested with EcoRI and Hind-XT and isolated on an agarose gel.
  • the resulting clone, pGEM7zf(-)/SEAP contains the coding sequence for the SEAP gene from amino acids.
  • An expression plasmid constitutively expressing the SEAP protein was created by placing the sequence encoding a truncated SEAP gene downstream of the cytomegalovirus (CMV) IE-1 promoter.
  • the expression plasmid also includes the CMV intron A region 5' to the SEAP gene as well as the 3' untranslated region of the bovine growth hormone gene 3' to the SEAP gene.
  • the plasmid pCMVIE-AKI-DHFR (Whang , Y., Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E. (1987) J. Virol., 61:1796-1807) containing the CMV immediate early promoter was cut with EcoRI generating two fragments. The vector fragment was isolated by agarose electro- phoresis and reli gated. The resulting plasmid is named pCMV-AKI. Next, the cytomegalovirus intron A nucleotide sequence was inserted downstream of the CMV IEl promter in pCMV-AKI.
  • the intron A sequence was isolated from a genomic clone bank and subcloned into pBR322 to generate plasmid pl6T-286.
  • the intron A sequence was mutated at nucleotide 1856 (nucleotide numbering as in Chapman, B.S., Thayer, R.M., Vincent, K.A. and Haigwood, N.L., Nuc.Acids
  • Sense strand 5' GGCAGAGCTCGTTTAGTGAACCGTCAG 3' (SEQ.ID.NO.: 8)
  • Antisense strand 5' GAGAGATCTCAAGGACGGTGACTGCAG 3' (SEQ.ID.NO.: 9)
  • oligos generate a 991 base pair fragment with a Sad site incorporated by the sense oligo and a Bgl-II fragment incorporated by the antisense oligo.
  • the PCR fragment is trimmed with Sad and Bgl-II and isolated on an agarose gel.
  • the vector pCMV-AKI is cut with Sad and Bgl-II and the larger vector fragment isolated by agarose gel electrophoresis.
  • the two gel isolated fragments are ligated at their respective Sad and Bgl-II sites to create plasmid pCMV-AKI-InA.
  • the DNA sequence encoding the truncated SEAP gene is inserted into the pCMV-AKI-InA plasmid at the Bgl-II site of the vector.
  • the SEAP gene is cut out of plasmid ⁇ GEM7zf(-)/SEAP (described above) using EcoRI and HindllL
  • the fragment is filled in with Klenow DNA polymerase and the 1970 base pair fragment isolated from the vector fragment by agarose gel electrophoresis.
  • the pCMV-AKI- InA vector is prepared by digesting with Bgl-II and filling in the ends with Klenow DNA polymerase.
  • the final construct is generated by blunt end ligating the SEAP fragment into the pCMV-AKI-InA vector.
  • Transformants were screened for the proper insert and then mapped for restriction fragment orientation. Properly oriented recombinant constructs were sequenced across the cloning junctions to verify the correct sequence.
  • the resulting plasmid named pCMV-SEAP-A (deposited in the ATCC under Budapest Treaty on August 27, 1998, and designated ATCC), contains a modified SEAP sequence downstream of the cytomegalovirus immediately early promoter IE-1 and intron A sequence and upstream of the bovine growth hormone poly-A sequence.
  • the plasmid expresses SEAP in a constitutive manner when transfected into mammalian cells.
  • An expression plasmid constitutively expressing the SEAP protein can be created by placing the sequence encoding a truncated SEAP gene downstream of the cytomegalovirus (CMV) IE-1 promoter and upstream of the 3' unstranslated region of the bovine growth hormone gene.
  • CMV cytomegalovirus
  • the plasmid pCMVIE-AKI-DHFR (Whang , Y., Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E. (1987) J. Virol., 61:1796-1807) containing the CMV immediate early promoter and bovine growth hormone poly-A sequence can be cut with EcoRI generating two fragments. The vector fragment can be isolated by agarose electrophoresis and religated. The resulting plasmid is named pCMV-AKI.
  • the DNA sequence encoding the truncated SEAP gene can be inserted into the pCMV-AKI plasmid at a unique Bgl-II in the vector.
  • the SEAP gene is cut out of plasmid pGEMzf(-)/SEAP (described above) using EcoRI and Hindlll. The fragments are filled in with Klenow DNA polymerase and the 1970 base pair fragment is isolated from the vector fragment by agarose gel electrophoresis.
  • the pCMV-AKI vector is prepared by digesting with Bgl-II and filling in the ends with Klenow DNA polymerase. The final construct is generated by blunt end ligating the SEAP fragment into the vector and transforming the ligation reaction into E. coli DH5 cells.
  • Transformants can then be screened for the proper insert and mapped for restriction fragment orientation. Properly oriented recombinant constructs would be sequenced across the cloning junctions to verify the correct sequence.
  • the resulting plasmid named pCMN-SEAP-B contains a modified SEAP sequence downstream of the cytomegalovirus immediate early promoter, IE1, and upstream of a bovine growth hormone poly-A sequence. The plasmid would express SEAP in a constitutive nammer when transfected into mammalian cells.
  • a D ⁇ A fragment containing viral-H-ras can be PCRed from plasmid "HB-11 (deposited in the ATCC under Budapest Treaty on August 27, 1997, and designated ATCC 209,218) using the following oligos.
  • the sense strand oligo also optimizes the 'Kozak' translation initiation sequence immediately 5' to the ATG start site.
  • cysteine 186 would be mutated to a serine by substituting a G residue for a C residue in the C-terminal antisense oligo.
  • the PCR primer oligos introduce an Xhol site at the 5' end and a Xbal site at the 3 'end.
  • the Xhol-Xbal fragment can be ligated into the mammalian expression plasmid pCI (Promega) cut with Xhol and Xbal. This results in a plasmid, pSMS600, in which the recombinant myr-viral-H-ras gene is constitutively transcribed from the CMV promoter of the pCI vector.
  • a viral-H-ras-CNLL expression plasmid pSMS ⁇ Ol A viral-H-r-w clone with a C-terminal sequence encoding the amino acids CVLL can be cloned from the plasmid "HB-11" by PCR using the following oligos.
  • Antisense strand 5'CACTCTAGACTGGTGTCAGAGCAGCACACACTTGCAGC-3' (SEQ.ID.NO.:
  • the sense strand oligo optimizes the 'Kozak' sequence and adds an Xhol site.
  • the antisense strand mutates serine 189 to leucine and adds an Xbal site.
  • the PCR fragment can be trimmed with Xhol and Xbal and ligated into the Xhol- Xbal cut vector pCI (Promega). This results in a plasmid, pSMS601, in which the mutated viral-H-r ⁇ s-CVLL gene is constitutively transcribed from the CMV promoter of the pCI vector.
  • the human cellular-H-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligonucleotide primers.
  • Sense strand 5'-GAGAGAATTCGCCACCATGACGGAATATAAGCTGGTGG-3' (SEQ.ID.NO.: 14)
  • Antisense strand
  • the primers will amplify a c-H-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, an EcoRI site at the N-terminus and a Sal I site at the C-terminal end.
  • the c-H-ras fragment can be ligated ligated into an EcoRI -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of glutamine-61 to a leucine can be accomplished using the manufacturer's protocols and the following oligonucleotide:
  • the mutated c-H-ras-Leu ⁇ l can be excised from the pAlter-1 vector, using EcoRI and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with EcoRI and Sal I.
  • the new recombinant plasmid, pSMS620 will constitutively transcribe c-H-ras-Leu ⁇ l from the CMV promoter of the pCI vector.
  • the human c-N-ras gene can be PCRed from a human cerebral cortex cD ⁇ A library (Clontech) using the following oligonucleotide primers.
  • Antisense strand
  • the primers will amplify a c-N-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, an EcoRI site at the N-terminus and a Sal I site at the C-terminal end.
  • the c-N-ras fragment can be ligated into an EcoRI -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of glycine-12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide:
  • the mutated c-N-r ⁇ .s-Nal-12 can be excised from the pAlter-1 vector, using EcoRI and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with EcoRI and Sal I.
  • the new recombinant plasmid, pSMS630 will constitutively transcribe c-N-r--_>-Val-12 from the CMV promoter of the pCI vector.
  • c-K4B-ra,f-Val-12 expression plasmid pSMS640 Cloning of a c-K4B-ra,f-Val-12 expression plasmid pSMS640
  • the human c-K4B-r ⁇ .s gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligo-nucleotide primers.
  • Antisense strand
  • the primers will amplify a c-K4B-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, a Kpnl site at the N-terminus and a Sal I site at the C-terminal end.
  • the c-K4B-r ⁇ -? fragment can be ligated into a Kpnl -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of cysteine-12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide:
  • the mutated c-K4B-r ⁇ - ⁇ -Val-12 can be excised from the pAlter-1 vector, using Kpnl and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with Kpnl and Sal I.
  • the new recombinant plasmid will constitutively transcribe c-K4B-ra-?-Val-12 from the CMV promoter of the pCI vector.
  • the human c-K4A-r ⁇ -? gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligo-nucleotide primers.
  • Sense strand 5'-GAGAGGTACCGCCACCATGACTGAATATAAACTTGTGG-3' (SEQ.ID.NO.: 23)
  • Antisense strand 5'-CTCTGTCGACAGATTACATTATAATGCATTTTTTAATTTTCACAC-3' (SEQ.ID.NO.: 24)
  • the primers will amplify a c-K4A-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, a Kpnl site at the N-terminus and a Sal I stite at the C-terminal end.
  • the c-K-ras4A fragment can be ligated into a Kpnl -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of cysteine-12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide:
  • the mutated c-K4A-r ⁇ _?-Val-12 can be excised from the pAlter-1 vector, using Kpnl and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with Kpnl and Sal I.
  • the new recombinant plasmid, pSMS650 will constitutively transcribe c-K4A-ra-?-Val-12 from the CMV promoter of the pCI vector.
  • Human C33A cells (human epitheial carcenoma - ATTC collection) are seeded in 10cm tissue culture plates in DMEM + 10% fetal calf serum + IX Pen/Strep + IX glutamine + IX NEAA. Cells are grown at 37°C in a 5% CO2 atmosphere until they reach 50-80% of confluency. The transient transfection is performed by the CaPO4 method
  • the cells are washed with PBS and trypsinized with 1ml of 0.05% trypsin.
  • the 1 ml of trypsinized cells is diluted into 10 ml of phenol red free DMEM + 0.2% charcoal stripped calf serum + IX (Pen/Strep, Glutamine and NEAA).
  • Transfected cells are plated in a 96 well microtiter plate (100 ⁇ l/well) to which drug, diluted in media, has already been added in a volume of 100 ⁇ l. The final volume per well is 200 ⁇ l with each drug concentration repeated in triplicate over a range of half-log steps.
  • Incubation of cells and drugs is for 36 hours at 37°C under CO2. At the end of the incubation period, cells are examined micro-scopically for evidence of cell distress.
  • 100 ⁇ l of media containing the secreted alkaline phosphatase is removed from each well and transferred to a microtube array for heat treatment at 65 °C for 1 hour to inactivate endogenous alkaline phosphatases (but not the heat stable secreted phosphatase).
  • the heat treated media is assayed for alkaline phosphatase by a luminescence assay using the luminescence reagent CSPD® (Tropix, Bedford, Mass.).
  • Luminescence reflects the level of activation of the fos reporter construct stimulated by the transiently expressed protein.
  • Ratl cells are used for analysis of protein processing. Subconfluent cells in 100 mm dishes are fed with 3.5 ml of media (methionine-free RPMI supplemented with 2% fetal bovine serum or cysteine-free/methionine-free DMEM supplemented with 0.035 ml of 200 mM glutamine (Gibco), 2% fetal bovine serum, respectively) containing the desired concentration of test compound, lovastatin or solvent alone. Cells treated with lovastatin (5-10 ⁇ M), a compound that blocks Ras processing in cells by inhibiting a rate-limiting step in the isoprenoid biosynthetic pathway, serve as a positive control.
  • media methionine-free RPMI supplemented with 2% fetal bovine serum or cysteine-free/methionine-free DMEM supplemented with 0.035 ml of 200 mM glutamine (Gibco), 2% fetal bovine serum, respectively.
  • Test compounds are prepared as lOOOx concentrated solutions in DMSO to yield a final solvent concentration of 0.1%. Following incubation at 37°C for two hours 204 ⁇ Ci/ml [35s]Pro-Mix (Amersham, cell labeling grade) is added.
  • the cells are incubated at 37°C for an additional period of time (typically 6 to 24 hours). The media is then removed and the cells are washed once with cold PBS. The cells are scraped into 1 ml of cold PBS, collected by centrifugation (10,000 x g for 10 sec at room temperature), and lysed by vortexing in 1 ml of lysis buffer (1% Nonidet P-40, 20 mM HEPES, pH 7.5, 150 mM NaCl, 1 mM EDTA, 0.5% deoxycholate, 0.1% SDS, 1 mM DTT, 10 ⁇ g/ml AEBSF, 10 .g/ml aprotinin, 2 ⁇ g/ml leupeptin and 2 ⁇ g/ml antipain). The lysate is then centrifuged at 15,000 x g for 10 min at 4°C and the supernatant saved.
  • lysis buffer 1% Nonidet P-40, 20 mM HEPES, pH 7.5
  • Ki4B-Ras For immunoprecipitation of Ki4B-Ras, samples of lysate supernatant containing equal amounts of protein are utilized. Protein concentration is determined by the bradford method utilizing bovine serum albumin as a standard. The appropri- ate volume of lysate is brought to 1 ml with lysis buffer lacking DTT and 8 ⁇ g of the pan Ras monoclonal antibody, Y13-259, added. The protein/antibody mixture is incubated on ice at 4°C for 24 hours. The immune complex is collected on pansorbin (Calbiochem) coated with rabbit antiserum to rat IgG (Cappel) by tumbling at 4°C for 45 minutes.
  • pansorbin Calbiochem
  • the pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in 100 ⁇ l elution buffer (10 mM Tris pH 7.4, 1% SDS).
  • the Ras is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation (15,000 x g for 30 sec. at room temperature).
  • the supernatant is added to 1 ml of Dilution Buffer 0.1% Triton X- 100, 5 mM EDTA, 50 mM NaCl, 10 mM Tris pH 7.4) with 2 ⁇ g Kirsten-ras specific monoclonal antibody, c-K-ras Ab-1 (Calbiochem).
  • the second protein/antibody mixture is incubated on ice at 4°C for 1-2 hours.
  • the immune complex is collected on pansorbin (Calbiochem) coated with rabbit antiserum to rat IgG (Cappel) by tumbling at 4°C for 45 minutes.
  • the pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in Laemmli sample buffer.
  • the Ras is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation.
  • the supernatant is subjected to SDS-PAGE on a 12% acrylamide gel (bis-acrylamide: acrylamide, 1:100), and the Ras visualized by fluorography.
  • PSN-1 cells are seeded in 24- well assay plates. For each compound to be tested, the cells are treated with a minimum of seven concentrations in half-log steps. The final solvent (DMSO) concentration is 0.1%. A vehicle-only control is included on each assay plate. The cells are treated for 24 hours at 37°C / 5% CO2-
  • the growth media is then aspirated and the samples are washed with PBS.
  • the cells are lysed with SDS-PAGE sample buffer containing 5% 2-mercaptoethanol and heated to 95°C for 5 minutes. After cooling on ice for 10 minutes, a mixture of nucleases is added to reduce viscosity of the samples.
  • the plates are incubated on ice for another 10 minutes.
  • the samples are loaded onto pre-cast 8% acrylamide gels and electrophoresed at 15 mA/gel for 3-4 hours.
  • the samples are then transferred from the gels to PNDF membranes by Western blotting.
  • the membranes are blocked for at least 1 hour in buffer containing
  • hDJ-2 ⁇ eomarkers Cat. # MS-225
  • an alkaline phosphatase-conjugated secondary antibody The membranes are then treated with a fluorescent detection reagent and scanned on a phosphorimager. For each sample, the percent of total signal corresponding to the unprenylated species of hDJ (the slower-migrating species) is calculated by densitometry. Dose-response curves and EC50 values are generated using 4- parameter curve fits in SigmaPlot software.
  • Protocol A Cells are labeled, incubated and lysed as described in Example 10.
  • Rapl For immunoprecipitation of Rapl, samples of lysate supernatant containing equal amounts of protein are utilized. Protein concentration is determined by the bradford method utilizing bovine serum albumin as a standard. The appropriate volume of lysate is brought to 1 ml with lysis buffer lacking DTT and 2 ⁇ g of the Rapl antibody, Rapl/Krevl (121) (Santa Cruz Biotech), is added. The protein/ antibody mixture is incubated on ice at 4°C for 1 hour. The immune complex is collected on pansorbin (Calbiochem) by tumbling at 4°C for 45 minutes.
  • the pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in 100 ⁇ l elution buffer (10 mM Tris pH 7.4, 1% SDS).
  • the Rapl is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation (15,000 x g for 30 sec. at room temperature).
  • the supernatant is added to 1 ml of Dilution Buffer (0.1% Triton X-100, 5 mM EDTA, 50 mM NaCl, 10 mM Tris pH 7.4) with 2 ⁇ g Rapl antibody, Rapl/Krevl (121) (Santa Cruz Biotech).
  • the second protein/antibody mixture is incubated on ice at 4°C for 1-2 hours.
  • the immune complex is collected on pansorbin (Calbiochem) by tumbling at 4°C for 45 minutes.
  • the pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in Laemmli sample buffer.
  • Rapl is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation. The supernatant is subjected to SDS-PAGE on a 12% acrylamide gel (bis-acrylamide: acrylamide, 1:100), and the Rapl visualized by fluorography.
  • Protocol B PSN-1 cells are passaged every 3-4 days in 10cm plates, splitting near-confluent plates 1:20 and 1:40. The day before the assay is set up, 5x 10 cells are plated on 15cm plates to ensure the same stage of confluency in each assay. The media for these cells is RPM1 1640 (Gibco), with 15% fetal bovine serum and lx Pen/Strep antibiotic mix. The day of the assay, cells are collected from the 15 cm plates by trypsinization and diluted to 400,000 cells/ml in media. 0.5ml of these diluted cells are added to each well of 24-well plates, for a final cell number of 200,000 per well. The cells are then grown at 37°C overnight.
  • the compounds to be assayed are diluted in DMSO in 1/2-log dilutions.
  • the range of final concentrations to be assayed is generally 0.1-100 ⁇ M. Four concentrations per compound is typical.
  • the compounds are diluted so that each concentration is lOOOx of the final concentration (i.e., for a 10 ⁇ M data point, a 10 mM stock of the compound is needed).
  • 2 ⁇ L of each lOOOx compound stock is diluted into 1 ml media to produce a 2X stock of compound.
  • a vehicle control solution (2 ⁇ L DMSO to 1ml media), is utilized. 0.5 ml of the 2X stocks of compound are added to the cells.
  • RNAse/DNase mix is added per well. This mix is lmg/ml DNasel (Worthington Enzymes), 0.25 mg/ml Rnase A (Worthington Enzymes), 0.5 M Tris-HCl pH 8.0 and 50 mM MgCl2.
  • the plate is left on ice for 10 minutes. Samples are then either loaded on the gel, or stored at -70°C until use.
  • Each assay plate (usually 3 compounds, each in 4-point titrations, plus controls) requires one 15-well 14% Novex gel. 25 ⁇ l of each sample is loaded onto the gel. The gel is run at 15 mA for about 3.5 hours. It is important to run the gel far enough so that there will be adequate separation between 21 kd (Rapl) and 29kd (Rab6).
  • the blocking solution is discarded and 20ml fresh blocking solution containing the anti Rap la antibody (Santa Cruz Biochemical SC1482) at 1:1000 (diluted in Western blocking buffer) and the anti Rab6 antibody (Santa Cruz Biochemical SC310) at 1:5000 (diluted in Western blocking buffer) are added.
  • the membranes are incubated at room temperature for 1 hour with mild rocking. The blocking solution is then discarded and the membrane is washed 3 times with Western wash buffer for 15 minutes per wash.
  • ECF overhead transparency
  • the developed transparency sheet is scanned on a phosphorimager and the Rapla Minimum Inhibitory Concentration is determined from the lowest concentration of compound that produces a detectable Rapla Western signal.
  • the Rapla antibody used recognizes only unprenylated/unprocessed Rapla, so that the precence of a detectable Rapla Western signal is indicative of inhibition of Rapla prenylation.
  • This protocol allows the determination of an EC50 for inhibition of processing of Rapla.
  • the assay is run as described in Protocol B with the following modifications. 20 ⁇ l of sample is run on pre-cast 10-20% gradient acrylamide mini gels (Novex Inc.) at 15 mA/gel for 2.5-3 hours. Prenylated and unprenylated forms of Rapla are detected by blotting with a polyclonal antibody (Rapl/Krev-1 Ab#121 ; Santa Cruz Research Products #sc-65), followed by an alkaline phosphatase- conjugated anti-rabbit IgG antibody. The percentage of unprenylated Rapla relative to the total amount of Rapla is determined by peak integration using ImagequantTM software (Molecular Dynamics).
  • Unprenylated Rapla is distinguished from prenyl- ated protein by virtue of the greater apparent molecular weight of the prenylated protein. Dose-response curves and EC50 values are generated using 4-parameter curve fits in SigmaPlot software.
  • mice in each oncogene group are randomly assigned to a vehicle or compound treatment group. Animals are dosed subcutaneously starting on day 1 and daily for the duration of the experiment. Alternatively, the farnesyl-protein transferase inhibitor may be administered by a continuous infusion pump. Compound or vehicle is delivered in a total volume of 0.1 ml. Tumors are excised and weighed when all of the vehicle-treated animals exhibited lesions of 0.5-1.0 cm in diameter, typically 11-15 days after the cells were injected. The average weight of the tumors in each treatment group for each cell line is calculated.

Abstract

The present invention is directed to piperidine- and pyrrolidine-containing macrocyclic compounds which inhibit prenyl-protein transferase and the prenylation of the oncogene protein Ras. The invention is further directed to chemotherapeutic compositions containing the compounds of this invention and methods for inhibiting prenyl-protein transferase and the prenylation of the oncogene protein Ras.

Description

TITLE OF THE INVENTION
INHIBITORS OF PRENYL-PROTEIN TRANSFERASE
BACKGROUND OF THE INVENTION
The Ras proteins (Ha-Ras, Ki4a-Ras, Ki4b-Ras and N-Ras) are part of a signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation. Biological and biochemical studies of Ras action indicate that Ras functions like a G-regulatory protein. In the inactive state, Ras is bound to GDP. Upon growth factor receptor activation Ras is induced to exchange GDP for GTP and undergoes a conformational change. The GTP-bound form of Ras propagates the growth stimulatory signal until the signal is terminated by the intrinsic GTPase activity of Ras, which returns the protein to its inactive GDP bound form (D.R. Lowy and D.M. Willumsen, Ann. Rev. Biochem. (52:851-891 (1993)). Mutated ras genes (Ha-ras, Ki4a-r-j_?, Ki4b-rα.s and N-ras) are found in many human cancers including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias. The protein products of these genes are defective in their GTPase activity and constitutively transmit a growth stimulatory signal.
Ras must be localized to the plasma membrane for both normal and oncogenic functions. At least 3 post-translational modifications are involved with Ras membrane localization, and all 3 modifications occur at the C-terminus of Ras. The Ras C-terminus contains a sequence motif termed a "CAAX" or "Cys- Aaal-Aaa2-Xaa" box (Cys is cysteine, Aaa is an aliphatic amino acid, the Xaa is any amino acid) (Willumsen et al., Nature 310:583-586 (1984)). Depending on the specific sequence, this motif serves as a signal sequence for the enzymes farnesyl- protein transferase or geranylgeranyl-protein transferase type I, which catalyze the alkylation of the cysteine residue of the CAAX motif with a C15 or C20 isoprenoid, respectively. (S. Clarke., Ann. Rev. Biochem. 6.7:355-386 (1992); W.R. Schafer and J. Rine, Ann. Rev. Genetics 50:209-237 (1992)). The term prenyl-protein transferase may be used to refer generally to farnesyl-protein transferase and geranylgeranyl- protein transferase type I. The Ras protein is one of several proteins that are known to undergo post-translational farnesylation. Other farnesylated proteins include the Ras-related GTP-binding proteins such as Rho, fungal mating factors, the nuclear lamins, and the gamma subunit of transducin. James, et al., /. Biol. Chem. 269, 14182 (1994) have identified a peroxisome associated protein Pxf which is also farnesylated. James, et al., have also suggested that there are farnesylated proteins of unknown structure and function in addition to those listed above.
Inhibition of farnesyl-protein transferase has been shown to block the growth of Ras-transformed cells in soft agar and to modify other aspects of their transformed phenotype. It has also been demonstrated that certain inhibitors of farnesyl-protein transferase selectively block the processing of the Ras oncoprotein intracellularly (N.E. Kohl et ah, Science, 260:1934-1937 (1993) and G.L. James et al., Science, 260:1937-1942 (1993). Recently, it has been shown that an inhibitor of farnesyl-protein transferase blocks the growth of rαs-dependent tumors in nude mice (N.E. Kohl et al., Proc. Natl. Acad. Sci U.S.A., 97:9141-9145 (1994) and induces regression of mammary and salivary carcinomas in ras transgenic mice (N.E. Kohl et al., Nature Medicine, 1:792-797 (1995).
Indirect inhibition of farnesyl-protein transferase in vivo has been demonstrated with lovastatin (Merck & Co., Rahway, NJ) and compactin (Hancock et al., ibid; Casey et al., ibid; Schafer et al., Science 245:379 (1989)). These drugs inhibit HMG-CoA reductase, the rate limiting enzyme for the production of poly- isoprenoids including farnesyl pyrophosphate. Farnesyl-protein transferase utilizes farnesyl pyrophosphate to covalently modify the Cys thiol group of the Ras CAAX box with a farnesyl group (Reiss et al, Cell, 62:81-88 (1990); Schaber et al., J. Biol. Chem., 265:14701-14704 (1990); Schafer et al, Science, 249:1133-1139 (1990); Manne et al., Proc. Natl. Acad. Sci USA, 57:7541-7545 (1990)). Inhibition of farnesyl pyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane localization in cultured cells. However, direct inhibition of farnesyl- protein transferase would be more specific and attended by fewer side effects than would occur with the required dose of a general inhibitor of isoprene biosynthesis. Inhibitors of farnesyl-protein transferase (FPTase) have been described in two general classes. The first are analogs of farnesyl diphosphate (FPP), while the second class of inhibitors is related to the protein substrates (e.g., Ras) for the enzyme. The peptide derived inhibitors that have been described are generally cysteine containing molecules that are related to the CAAX motif that is the signal for protein prenylation. (Schaber et al., ibid; Reiss et. al., ibid; Reiss et al., PNAS, S<_>:732-736 (1991)). Such inhibitors may inhibit protein prenylation while serving as alternate substrates for the farnesyl-protein transferase enzyme, or may be purely competitive inhibitors (U.S. Patent No. 5,141,851, University of Texas; N.E. Kohl et al., Science, 260:1934-1937 (1993); Graham, et al., J. Med. Chem., 37, 725 (1994)). In general, deletion of the thiol from a CAAX derivative has been shown to dramatically reduce the inhibitory potency of the compound. However, the thiol group potentially places limitations on the therapeutic application of FPTase inhibitors with respect to pharmacokinetics, pharmacodynamics and toxicity. Therefore, a functional replacement for the thiol is desirable. It has recently been reported that farnesyl-protein transferase inhibitors are inhibitors of proliferation of vascular smooth muscle cells and are therefore useful in the prevention and therapy of arteriosclerosis and diabetic disturbance of blood vessels (JP H7-112930).
It has recently been disclosed that certain tricyclic compounds which optionally incorporate a piperidine moiety are inhibitors of FPTase (WO 95/10514, WO 95/10515 and WO 95/10516). Imidazole-containing inhibitors of farnesyl protein transferase have also been disclosed (WO 95/09001 and EP 0 675 112 Al). It is, therefore, an object of this invention to develop peptidomimetic compounds that do not have a thiol moiety, and that will inhibit prenyl-protein transferase and thus, the post-translational prenylation of proteins. It is a further object of this invention to develop chemotherapeutic compositions containing the compounds of this invention and methods for producing the compounds of this invention.
SUMMARY OF THE INVENTION
The present invention comprises peptidomimetic piperidine- and pyrrolidine-containing macrocyclic compounds which inhibit prenyl-protein transferase. Further contained in this invention are chemotherapeutic compositions containing these prenyl-protein transferase inhibitors and methods for their production.
The compounds of this invention are illustrated by the formula A:
Figure imgf000005_0001
DETAILED DESCRIPTION OF THE INVENTION
The compounds of this invention are useful in the inhibition of prenyl-protein transferase and the prenylation of the oncogene protein Ras. In a first embodiment of this invention, the inhibitors of prenyl-protein transferase are illustrated by the formula A:
Figure imgf000005_0002
wherein: Rla, Rib, Rlc and Rid are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) RlOO-, f) RllS(O)m-, g) Rl0C(O)NRl0-, fa) (RlO)2NC(O)-, i) CN, j) NO2, k) RlOC(O)-,
1) RlOθC(O)-, m) -N(R10)2, n) RHOC(O)NR10-, o) unsubstituted or substituted C1-C6 alkyl,
P) unsubstituted or substituted C2-C6 alkenyl, or q) unsubstituted or substituted C2-C6 alkynyl,
wherein the substituent on the substituted group is selected from unsubstituted or substituted Ci-Cβ alkyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C3-C10 cycloalkyl, unsubstituted or unsubstituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, RlOO-, Rl lS(O)m-, RlOC(O)NRlO-, (RlO)2NC(O)-, CN,
RlOC(O)-, RlOθC(O)-, -N(RlO)2, and RllOC(O)-NRlO-,
or two Rlas, two Ribs or two Rlcs, on the same carbon atom may be combined to form -(CH2)u-;
R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-8 alkyl, c) unsubstituted or substituted C2-8 alkenyl, d) unsubstituted or substituted C2-8 alkynyl, e) unsubstituted or substituted aryl, f) unsubstituted or substituted heterocycle,
Figure imgf000007_0001
wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) Ci-4 alkyl, b) (CH2)POR6, c) (CH2)PNR6R75 d) halogen, e) CN,
2) C3-6 < cycloalkyl,
3) OR6,
Figure imgf000007_0002
5) — NR6R7
Figure imgf000007_0003
Figure imgf000008_0001
10) \^ NR6R7 0
11) — S02-NR6R7
Figure imgf000008_0002
15) N3, or
16) F; or
R4 is selected from Ci-6 alkyl, C^-β cycloalkyl, heterocycle, aryl, unsubstituted or substituted with:
1) Ci-6 alkoxy,
2) aryl or heterocycle,
3) halogen, 4) HO,
5)
-11
O 6) SO2Rll,
7) N(RlO)2, or
8) Cι_4 perfluoroalkyl;
R5, R6 and R7 are independently selected from: a) hydrogen, b) RlOC(O)-, c) Rl θC(O)-, or d) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3_6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
1) unsubstituted or substituted C1-C6 alkyl,
2) RlOO-, 3) unsubstituted or substituted aryl or heterocycle,
4) halogen,
5) Rl0C(O)NRl0-,
Figure imgf000009_0001
7) SO2RH,
8) N(R10)2,
9) C3-6 cycloalkyl,
10) C1-C6 perfluoroalkyl.
11) RlOθC(O)-,
12) RllOC(O)NRl0-,
13) CN, and
14) NO2; or
R6 and R"7 may be joined in a ring; and independently,
R5 and R may be joined in a ring; R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) C1-C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O)m-, k) Rl0C(O)NRl0-, 1) (RlO)2NC(O)-, m) CN, n) NO2, o) RlOC(O)-, p) Rl θC(O)-, q) -N(RlO)2, r) Rl lOC(O)NRl0-, and s) unsubstituted or substituted Ci-Cβ alkyl, optionally substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C3-C10 cycloalkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, C1.-C6 perfluoroalkyl, F, Cl, Br, RlOO-, Rl lS(O)m-, Rl0C(O)NRl0-, (RlO)2NC(O)-, CN, Rl0C(O)-,
RlOθC(O)-, -N(R10)2, or R10OC(O)NR10
R9 is selected from: a) hydrogen, b) unsubstituted or substituted C2-C6 alkenyl, c) unsubstituted or substituted C2-C6 alkynyl, d) C1-C6 perfluoroalkyl, e) halo, f) RlOO-, g) RllS(O)m-, h) Rl0C(O)NRl0-, i) (R10)2NC(O)-, j) CN, k) NO2,
1) RlOC(O)-, m) RlOθC(O)-,
Figure imgf000011_0001
o) RllOC(O)NRl0-, or p) unsubstituted or substituted Cχ-C6 alkyl optionally substituted by aryl,
C3-C10 cycloalkyl, C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, RlOC(O)NRlO-, (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, orRllOC(O)NRlO-;
RlO is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-Cβ alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
Rl 1 is independently selected from a) unsubstituted or substituted C 1 -C alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRlO-, d) -NRl C(O)-, e) O, f) -N(Rl )-, g) -S(O)2N(R10)-,
Figure imgf000012_0001
i) S(O)m;
is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(R10)-, g) -S(O)2N(RlO)-,
Figure imgf000012_0002
i) -NRl0C(O)NRl0-, or j) S(O)m;
G is selected from H2 or O;
V is selecte. I from: a) heterocycle, or b) aryl;
W is a heterocycle;
X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, f) -O-C(O)NRl0-, g) -NRl0C(O)NRl0-, h) O, i) -N(Rl0)-, j) -S(O)2N(RlO)-,
Figure imgf000013_0001
1) S(O)m;
Y is selected from a) a bond, b) -C(=O)-, c) -C(O)NRl0-, d) -C(O)O-, or e) -S(=O)m;
Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of: 1) Ci-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Cι_4 alkoxy, b) NR6R7, c) C3_6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or h) Ci-4 perfluoroalkyl;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen,
4) OR6>
5) NR6R7,
6) CN,
7) NO2,
8) CF3;
9) -S(O)mR4
Figure imgf000013_0002
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
Z2 is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
1) Cι_8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Cι_4 alkoxy, b) NR6R7 c) C3_6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or h) Ci_4 perfluoroalkyl;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen,
4) OR6.
5) NR6R7,
6) CN,
7) NO2,
8) CF3,
9) -S(O)mR4,
Figure imgf000014_0001
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
k is 0, 1, 2, 3 or 4; m is 0, 1 or 2; n is 0, 1, 2, 3, 4 or 5; p is independently 0, 1, 2, 3, 4 or 5; q is 1, 2 or 3; r is 0, 1, 2, 3, 4 or 5; s is independently 0, 1, 2, 3, 4 or 5; t is 0 or l; and u is independently 2, 3, 4, 5 or 6;
or a pharmaceutically acceptable salt or stereoisomer thereof.
In another embodiment of this invention, the inhibitors of prenyl- protein transferase are illustrated by the formula A:
Figure imgf000015_0001
wherein:
Rla, Rib, Rlc and Rid are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3 -C 10 cycloalkyl , e) RlOO-, f) RllS(O)m-, g) Rl0C(O)NRl0-, h) (RlO)2NC(O)-, i) CN, j) NO2, k) RlOC(O)-, 1) RlOθC(O)-, m) -N(RlO)2, n) RllOC(O)NRl0-, o) unsubstituted or substituted C1-C6 alkyl, p) unsubstituted or substituted C2-C6 alkenyl, or q) unsubstituted or substituted C2-C6 alkynyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted
C3-C10 cycloalkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, RlOO-, RllS(O)m-, RlOC(O)NRlO-, (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, and RllOC(O)NRl -,
or two Rlas, two Ribs or two Rlcs, on the same carbon atom may be combined to form -(CH2)u-;
R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-8 alkyl, c) unsubstituted or substituted C2-8 alkenyl, d) unsubstituted or substituted C2-8 alkynyl, e) unsubstituted or substituted aryl, f) unsubstituted or substituted heterocycle, g)
or h)
wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) Ci-4 alkyl, b) (CH2)POR6 c) (CH2)PNR6R7, d) halogen, e) CN,
2) C3-.6 cycloalkyl,
Figure imgf000017_0001
5) — NR6R7
Figure imgf000017_0002
11) — S02-NR6R7
Figure imgf000017_0003
Figure imgf000018_0001
14)
^ORβ 0
15) N3, or
16) F; or
R4 is selected from Cι_4 alkyl, C3.6 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with: 1) C1-.4 alkoxy,
2) aryl or heterocycle,
3) halogen,
4) HO,
Figure imgf000018_0002
6) SO2Rll,
7) N(RlO)2, or
8) C1-.4 perfluoroalkyl;
R5, R6 and R7 are independently selected from: a) hydrogen, b) RIOC(O)-, c) Rl°OC(O)-, or d) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
1) RlOO-, 2) aryl or heterocycle, 3) halogen,
4) Rl0C(O)NRl0-,
Figure imgf000019_0001
7) N(R10)2,
8) C3-6 cycloalkyl,
9) C1-C6 perfluoroalkyl,
10) RlOθC(O)-,
11) RllOC(O)NRl0-,
12) CN, and
13) NO2; or
R and R7 may be joined in a ring; and independently,
R5 and R7 may be joined in a ring;
is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) Cl-Cg perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O)m-, k) Rl0C(O)NRl0-,
1) (RlO)2NC(O)-, m) CN, n) NO2, o) RlOC(O)-, p) RlOθC(O)-, q) -N(RlO)2, r) RllOC(O)NRl0-, and s) unsubstituted or substituted C1-C6 alkyl, optionally substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, RlOC(O) NRlO., (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, or Rl0θC(O)NRl0-;
R9 is selected from: a) hydrogen, b) unsubstituted or substituted C2-C6 alkenyl, c) unsubstituted or substituted C2-C6 alkynyl, d) C1-C6 perfluoroalkyl, e) halo, f) RlOO-, g) RllS(O)m-, h) Rl0C(O)NRl0-, i) (RlO)2NC(O)-, j) CN, k) NO2,
1) Rl0C(O)-, m) RlOθC(O)-, n) -N(RlO)2,
0) RllOC(O)NRl0-, or
P) unsubstituted or substituted C1-C6 alkyl optionally substituted by
C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, Rl lS(O)m-, Rl0C(O)
NR10-, (RlO)2NC(O)-, CN, RlOC(O)-, RIOOC(O)-, -N(RlO)2, or RllOC(O)NRl0-;
RlO is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-Cβ alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
Rll is independently selected from a) unsubstituted or substituted C1-C6 alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(Rl0)-, g) -S(O)2N(R10)-,
Figure imgf000021_0001
i) S(O)m;
A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(Rl0)-, g) -S(O)2N(Rl0)-,
Figure imgf000021_0002
i) -NRl0C(O)NRl0-, or j) S(O)m; G is selected from H2 or O;
V is selected from pyridyl or phenyl;
W is a heterocycle;
X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, f) -O-C(O)NRl0-, g) -NRl0C(O)NRl0-, h) O, i) -N(Rl0)-,
Figure imgf000022_0001
1) S(O)m;
Y is selected from a) a bond, b) -C(=O)-, c) -C(O)NRl0-, d) -C(O)O-, or e) -S(=O)m;
Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
1) Ci-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4 g) -C(O)NR6R7, or h) Ci-4 perfluoroalkyl;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen, 4) OR6>
5) NR6R7,
6) CN,
7) NO2,
8) CF3; 9) -S(O)mR4
10) -OS(O)2R4,
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
Z2 is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
1) Cι_8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Cι_4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or h) C 1.4 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen,
4) OR6,
5) NR6R7,
6) CN,
7) NO2,
8) CF3,
9) -S(O)mR4,
Figure imgf000024_0001
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
k is 0, 1, 2, 3 or 4; m is 0, 1 or 2; n is 0, 1, 2, 3, 4 or 5; p is independently 0, 1, 2, 3, 4 or 5; q is 1, 2 or 3; r is 0, 1, 2, 3, 4 or 5; s is independently 0, 1, 2, 3, 4 or 5; t is O or 1; and u is independi sntly 2, 3, 4, 5 or 6;
or a pharmaceutically acceptable salt or stereoisomer thereof.
In another embodiment of this invention, the inhibitors of prenyl- protein transferase are illustrated by the formula B:
Figure imgf000025_0001
wherein:
Rla, Rib and Rlc are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl,
Figure imgf000025_0002
f) unsubstituted or substituted C2-C6 alkenyl g) -ORlO, or h) unsubstituted or substituted Ci-Cβ alkyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, RlOO-, and -N(RlO)2,
or two R as, two Ribs or two Rlcs, on the same carbon atom may be combined to form -(CH2)u-;
Rid is selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) RlOO-, ) RnS(O)m-, g) RlOC(O)NRlO-, h) (RlO)2NC(O)-, i) CN, j) NO2, k) Rl0C(O)-,
1) Rl0θC(O)-, m) -N(RlO)2, n) RllOC(O)NRlO-, o) unsubstituted or substituted Ci-Cβ alkyl,
P) unsubstituted or substituted C2-Cg alkenyl, or q) unsubstituted or substituted C2-C6 alkynyl,
wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, RlOO-, RllS(O)m-, RlOC(O)NRlO-, (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, and RHOC(O)NRlO-;
R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Cι_8 alkyl, c)
R6R7
ϋ or
Figure imgf000026_0001
wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) C1-4 alkyl,
Figure imgf000026_0002
Figure imgf000027_0001
d) halogen, e) CN, 2) OR6,
Figure imgf000027_0002
4) -NR6R7;
R4 is selected from Cι_4 alkyl, or C3-6 cycloalkyl, unsubstituted or substituted with: 1) Ci-4 alkoxy, 2) aryl or heterocycle,
3) halogen,
4) HO,
6) SO2RH,
7) N(RlO)2, or
8) Ci-4 perfluoroalkyl;
R and R7 are independently selected from: a) hydrogen, b) Rl0C(O)-, c) RlOθC(O)-, or d) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3.6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
1) RlOO-,
2) aryl or heterocycle, 3) halogen,
4) Rl0C(O)NRl0-, 5)
0
Figure imgf000028_0001
7) N(R10)2,
8) C3-6 cycloalkyl,
9) C1-C6 perfluoroalkyl,
10) RlOθC(O)-,
11) RllOC(O)NRl0-,
12) CN, and
13) NO2; or
R6 and R7 may be joined in a ring;
R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) C1-C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O)m-, k) Rl0C(O)NRl0-,
1) (RlO)2NC(O)-, m) CN, n) NO2, o) Rl0C(O)-, p) RlOθC(O)-, q) -N(RlO)2, r) RllOC(O)NRl0-, or s) unsubstituted or substituted Ci-Cβ alkyl, optionally substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, Rl0C(O) NRlO-, (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, or RlOθC(O) NRlO-;
R9 is selected from: a) hydrogen, b) Ci-Cβ perfluoroalkyl, c) RlOO-, d) Rl0C(O)NRl0-, e) (RlO)2NC(O)-, or f) unsubstituted or substituted C1-C6 alkyl optionally substituted by C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, RlOC(O) NRlO-, (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, or
RllOC(O)NRl0-;
RlO is independently selected from a) hydrogen, b) unsubstituted or substituted C1-C6 alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
Rll is independently selected from a) unsubstituted or substituted C 1 -C alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, ) -N(R10)-, g) -S(O)2N(R10)-,
Figure imgf000030_0001
i) S(O)m;
A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(RlO)-, g) -S(O)2N(RlO)-,
Figure imgf000030_0002
i) -NRl0C(O)NRl0-, or j) S(O)m;
G is selected from H2 or O;
W is a heterocycle selected from imidazolyl, pyridyl, or triazolyl;
is select© d from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, f) -O-C(O)NRl0-, g) -NRl0C(O)NRl0-, h) O, i) -N(Rl0)-,
J) -S(O) N(RlO)-,
Figure imgf000030_0003
1) S(O)m;
Y is selected from a) a bond, b) -C(=O)-, c) -C(O)NRl0-, d) -C(O)O-, or e) -S(=O)m;
Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
1) Ci-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or h) C 1 -4 perfluoroalkyl ;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen,
4) OR6,
5) NR6R7,
6) CN,
7) NO2,
8) CF3;
9) -S(O)mR4,
Figure imgf000031_0001
11) -C(O)NR6R7,
12) -C(O)OR6, or 13) C3-C6 cycloalkyl;
kis 0,1,2, 3 or 4; m is 0, 1 or 2; nis 0,1,2, 3, 4 or 5; p is independently 0, 1, 2, 3, 4 or 5; qis 1,2 or 3; ris 0,1,2, 3, 4 or 5; s is independently 0, 1, 2, 3, 4 or 5; tis 0orl;and u is independently 2, 3, 4, 5 or 6;
or a pharmaceutically acceptable salt or stereoisomer thereof.
In a further preferred embodiment of this invention, the inhibitors of prenyl-protein transferase are illustrated by the formula C:
Figure imgf000032_0001
wherein:
Rla, Rib and Rlc are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C 10 cycloalkyl,
Figure imgf000033_0001
f) unsubstituted or substituted C2-C6 alkenyl g) -ORl0, or h) unsubstituted or substituted C1-C6 alkyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, RlOO-, and -N(RlO)2,
or two Rlas, two Ribs or two Rlcs, on the same carbon atom may be combined to form -(CH2)u-;
Rid is selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) RlOO-, f) -N(RlO) , or g) unsubstituted or substituted C1-C6 alkyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, RlOo-, Rl0c(O)NRl0-, (RlO)2NC(O)-, Rl0C(O)-, RlOθC(O)-, -N(RlO)2, and RllOC(O)-NRl0-;
R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-8 alkyl, c)
R6R7
U or
Figure imgf000034_0001
wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) Ci-4 alkyl,
Figure imgf000034_0002
d) halogen, e) CN, 2) OR6,
Figure imgf000034_0003
4) -NR6R7;
R4 is selected from Cι_4 alkyl, or C3-6 cycloalkyl, unsubstituted or substituted with: 1) C1-4 alkoxy,
2) aryl or heterocycle,
3) halogen, or
4) -OH;
R6 and R7 are independently selected from: a) hydrogen, or b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cι_6 perfluoroalkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
1) RlOO-,
2) aryl or heterocycle,
3) halogen,
4) Rl0C(O)NRl0-,
5)
Figure imgf000034_0004
6) SO2R11,
7) N(RlO)2, or
8) C3-6 cycloalkyl, or
R and R7 may be joined in a ring;
R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) C1-C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O)m-, k) RlOC(O)NRlO-, 1) (RlO)2NC(O)-, m) CN, n) NO2, o) Rl0C(O)-, p) RlOθC(O)-,
Figure imgf000035_0001
r) RllOC(O)NRl0-, and s) unsubstituted or substituted Cχ-C6 alkyl, optionally substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, Rl0C(O)NRl0-, (RlO)2NC(O)-, CN, Rl0C(O)-, RlOθC(O)-, -N(RlO)2, or
Rl0θC(O)NRl0-;
R9 is selected from: a) hydrogen, b) C1-C6 perfluoroalkyl, or c) unsubstituted or substituted C1-C6 alkyl optionally substituted by C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, Rl0C(O)NRl0-, (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, or RllOC(O)NRl0-;
RlO is independently selected from a) hydrogen, b) unsubstituted or substituted C 1 -C6 alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
Rl 1 is independently selected from a) unsubstituted or substituted C1-C6 alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O,
0 -N(Rl0)-, g) -S(O) N(R10)-,
Figure imgf000036_0001
i) S(O)m;
A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(RlO)-, g) -S(O)2N(R10)-, h) -N(RlO)S(O)2-, i) -NRl0C(O)NRl0-, or j) S(O)m;
G is selected from H2 or O;
X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, f) -O-C(O)NRl0-, g) -NRl0C(O)NRl0-, h) O, i) -N(Rl0)-, j) -S(O)2N(R10)-,
Figure imgf000037_0001
1) S(O)m;
Y is selected from a) a bond, b) -C(=O)-, c) -C(O)NRl0-, d) -C(O)O-, or e) -S(=O)m;
Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of: 1) Ci-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7, c) C3_6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or h) C 1 _4 perfluoroalkyl ;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen,
4) OR6.
5) NR6R7,
6) CN,
7) NO2)
8) CF3;
9) -S(O)mR4,
Figure imgf000038_0001
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
kis 0,1, 2, 3 or 4; mis 0, 1 or 2; nis 0,1, 2, 3, 4 or 5; p is independently 0,1,2, 3, 4 or 5; qis 1,2 or 3; ris 0, 1,2, 3, 4 or 5; s is independently 0,1, 2, 3, 4 or 5; tis Oor l;and u is independently 2, 3, 4, 5 or 6; or a pharmaceutically acceptable salt or stereoisomer thereof.
A specific example of the compounds of the instant invention is illustrated by:
Figure imgf000039_0001
6(R or S)-11(R or S)-amino-ll-(l-methyl-lH-imidazol-5-yl)-17-oxa-2-thia-3,7- diazatetracyclo [16.3.1.13,6.ll2,16]tetracosa-l(22),12(23),13,15,18,20-hexaene-15- carbonitrile 2,2-dioxide,
Figure imgf000039_0002
(14R or S,19S or R)-14-methyl-14-(l-methyl-lH-imidazol-5-yl)-16-oxo-8-oxa-2- thia-l,15-diazatetracyclo[17.3.1.13,7.l9,13]ρentacosa-3(25),4,6,9(24),10,12-hexaene- 10-carbonitrile 2,2-dioxide
Figure imgf000040_0001
(14R or S, 18R or S) -14-methyl-14-(l-methyl-lH-imidazol-5.-yl)-16-oxo-8-oxa-2- thia-l,15-diazatetracyclo[16.3.1.13,7.l9,13]tetracosa-3(24),4,6,9(23),10,12-hexaene-10- carbonitrile 2,2-dioxide
Figure imgf000040_0002
(14R or S, 17R or S)-14-methyl-14-(l-methyl-lH-imidazol-5-yl)-16-oxo-8-oxa-2-thia- l,15-diazatetracyclo[15.3.1.13,7.l9,13]tricosa-3(23),4,6,9(22),10,12-hexaene-10- carbonitrile 2,2-dioxide
or a pharmaceutically acceptable salt or stereoisomer thereof.
The compounds of the present invention may have asymmetric centers, chiral axes and chiral planes, and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention. (See E . Eliel and S.Η. Wilen Stereochemistry of Carbon Compounds (John Wiley and Sons, New York 1994), in particular pages 1119-1190) When any variable (e.g. aryl, heterocycle, Rla, R6 etc.) occurs more than one time in any constituent, its definition on each occurrence is independent at every other occurrence. Also, combinations of substituents/or variables are permissible only if such combinations result in stable compounds.
As used herein, "alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having from 1 to 20 carbon atoms, unless otherwise specified; "alkoxy" represents an alkyl having from 1 to 20 carbon atoms, unless otherwise indicated, attached through an oxygen bridge. "Halogen" or "halo" as used herein means fluoro, chloro, bromo and iodo.
As used herein, "cycloalkyl" is intended to include non-aromatic hydrocarbon groups having from 3 to 10 carbon atoms, unless otherwise specified. Examples of such cycloalkyl groups includes, but are not limited to, cyclopropyl, cyclobutyl, cyclohexyl, cycloheptyl, cyclooctyl, admantyl and the like.
If no number of carbon atoms is specified, the term "alkenyl" refers to a non-aromatic hydrocarbon, straight, branched or cyclic, containing from 2 to 10 carbon atoms, unless otherwise indicated, and at least one carbon to carbon double bond. Preferably one carbon to carbon double bond is present, and up to four non- aromatic carbon-carbon double bonds may be present. Thus, "C2-C8 alkenyl" means an alkenyl radical having from 2 to 8 carbon atoms. Examples of such alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted if a substituted alkenyl group is indicated.
The term "alkynyl" refers to a hydrocarbon radical straight, branched or cyclic, containing from 2 to 10 carbon atoms, unless otherwise indicated, and at least one carbon to carbon triple bond. Up to three carbon-carbon triple bonds may be present. Thus, "C2-C8 alkynyl" means an alkynyl radical having from 2 to 8 carbon atoms. Examples of such alkynyl groups include, but are not limited to, ethynyl, propynyl and butynyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated. As used herein, "aryl" is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic. Examples of such aryl elements include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl, acenaphthyl and the like. As used herein, "aralkyl" is intended to mean an aryl moiety, as defined above, attached through a Cχ-C6 alkyl linker, where alkyl is defined above. Examples of aralkyls include, but are not limited to, benzyl, naphthylmethyl, phenylbutyl and the like.
The term heterocycle or heterocyclic, as used herein, represents a stable 5- to 7-membered monocyclic or stable 8- to 11-membered bicyclic heterocyclic ring which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. The term heterocycle or heterocyclic includes heteroaryl moieties. Examples of such heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzo- thiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, 1,3-dioxolanyl, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, 2-oxopiperazinyl, 2-oxopiperdinyl, 2-oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, 2-pyridinonyl pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, thienyl and triazolyl. As used herein, "heteroaryl" is intended to mean any stable mono- cyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic and wherein from one to four carbon atoms are replaced by heteroatoms selected from the group consisting of N, O, and S. Examples of such heterocyclic elements include, but are not limited to, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxadiazolyl, pyridyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiazolyl, thienofuryl, thienothienyl, thienyl and triazolyl.
As used herein, "heterocyclylalkyl" is intended to mean a heterocyclic moiety, as defined above, attached through a Cχ-C6 alkyl linker, where alkyl is defined above. Examples of heterocyclylalkyls include, but are not limited to, 2-pyridylmethyl, 2-imidazolylethyl, 2-quinolinylmethyl, 2-imidazolylmethyl, and the like.
As used herein, "heterocycloalkyl" is intended to mean a heterocyclic moiety, as defined above, attached through a Cχ-C6 alkyl linker, where alkyl is defined above. Examples of heterocycloalkyls include, but are not limited to, 2-morpholinylethyl, 1-piperazinylmethyl, 3-pyrrolidinylrriethyl and the like.
As used herein, the terms "substituted alkyl", "substituted alkenyl", "substituted alkynyl" and "substituted alkoxy", unless otherwise defined, are intended to include the branch or straight-chain alkyl group of the specified number of carbon atoms, wherein the carbon atoms may be substituted with F, Cl, Br, I, CF3, OCF3,
CN, N3, NO2, NH2, N(Cχ-C6 alkylfc, oxo, OH, -O(Cχ-C6 alkyl), S(O)θ-2, (Cχ-C6 alkyl)S(O)0-2-, C2-C6 alkenyl, C2-C6 alkynyl, -(Cχ-C6 alkyl)S(O)θ-2(Cl-C6 alkyl), C3-C20 cycloalkyl, -C(O)NH2, HC(O)NH-(Cι-C6 alkyl)C(O)NH-, H2NC(O)NH- (C1-C6 alkyl)C(O)-, -O(Cχ-C6 alkyl)CF3, (C -C6 alkyl)OC(O)-, (Cχ-C6 alkyl) O(Cχ-C6 alkyl)-, (Cχ-C6 alkyl)C(O)2(Cχ-C6 alkyl)-, (Cχ-C6 alkyl)OC(O)NH-, aryl, heterocycle, aralkyl, heterocyclylalkyl, heterocycloalkyl, halo-aryl, halo-aralkyl, halo- heterocycle, halo-heterocyclylalkyl, halo-heterocycloalkyl, cyano-aryl, cyano-aralkyl, cyano-heterocycle, cyano-heterocyclylalkyl, and cyano-heterocycloalkyl.
As used herein, the terms "substituted aryl", "substituted heterocycle", "substituted heteroaryl", "substituted cycloalkyl", "substituted benzyl", "substituted aralkyl" and "substituted heterocyclylalkyl", "substituted heterocycloalkyl" unless otherwise defined, are intended to include the cyclic group containing from 1 to 3 substitutents in addition to the point of attachment to the rest of the compound. Such substitutents are preferably selected from the group which includes but is not limited to F, Cl, Br, I, CF3, OCF3, NH2, N(Cχ-C6 alkyl)2, NO2, CN, N3, C1-C20 alkyl,
C3-C2O cycloalkyl, -OH, -O(Cχ-C6 alkyl), S(O)θ-2. (C1-C6 alkyl)S(O)θ-2-, (C1-C6 alkyl)S(O)0-2(Cl-C6 alkyl)-, -C(O)NH2, HC(O)NH-, (Cχ-C6 alkyl)C(O)NH-, H2NC(O)NH-, (Cχ-C6 alkyl)C(O)-, (Cχ-C6 alkyl)OC(O)-, (C1-C6 alkyl)O(Cχ-C6 alkyl)-, (Cχ-C6)C(O)2(Cl-C6 alkyl)-, (Cχ-C6 alkyl)OC(O)NH-, aryl, heterocycle, aralkyl, heterocyclylalkyl, heterocycloalkyl, halo-aryl, halo-aralkyl, halo-heterocycle, halo-heterocyclylalkyl, halo-heterocycloalkyl, cyano-aryl, cyano-aralkyl, cyano- heterocycle, cyano-heterocyclylalkyl, and cyano-heterocycloalkyl.
Preferably, as used herein in the definition of R6 and R7, the substituted Ci-6 alkyl, substituted C2-6 alkenyl, substituted C2-6 alkynyl, substituted C3-6 cycloalkyl, substituted aroyl, substituted aryl, substituted heteroaroyl, substituted arylsulfonyl, substituted heteroarylsulfonyl and substituted heterocycle, include moieties containing from 1 to 3 substituents in addition to the point of attachment to the rest of the compound.
The moiety formed when, in the definition of Rla, Rib, and Rlc, two Rl s, two Ribs, or two Rlcs, on the same carbon atom are combined to form -(CH2)u- is illustrated by the following:
Figure imgf000044_0001
The moiety formed when (in the definition of R5, R6 and R7). R6 and R7 or R5 and R7 are joined to form a ring, is illustrated by, but not limited to, the following:
Figure imgf000044_0002
Lines drawn into the ring systems from substituents (such as from R2, R8, R9 etc.) indicate that the indicated bond may be attached to any of the substitut- able ring carbon or nitrogen atoms. Preferably, Rla and Ri are independently selected from: hydrogen,
-N(RlO)2, Rl0C(O)NRl0- or unsubstituted or substituted C1-C6 alkyl.
Preferably, Rlc is independently selected from: hydrogen, or unsubstituted or substituted Cχ-C6 alkyl. Preferably, Rid is selected from -N(RlO)2, OR 10, unsubstituted or substituted Cχ-C6 alkyl, unsubstituted or substituted aryl, or unsubstituted or substituted heterocycle.
Preferably, R2 is selected from: hydrogen and unsubstituted or substituted Cχ-C6 alkyl.
Preferably, R and R7 are selected from: hydrogen, unsubstituted or substituted Cχ-C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted cycloalkyl.
Preferably, R8 is selected from hydrogen, halo, unsubstituted or substituted Cχ-6 alkyl, unsubstituted or substituted Cl-6 alkoxy, unsubstituted or substituted aryl, CN, NO2, Rl0C(O)NRl0-, -ORlOand (RlO)2NC(O)-. Most preferably, r is 1 to 3 and at least one R8 is CN.
Preferably, R9 is hydrogen or unsubstituted or substituted Cχ-C6 alkyl. Most preferably, R is methyl. Preferably, Al and A2 are independently selected from: a bond,
C(=O), -C(O)NRlO-, -NRlOC(O)-, O, -N(Rl0)-, -S(O)m, -S(O)2N(RlO)-, and -N(RlO)S(O)2". Most preferably, Al is O and A2 is selected from a bond, -S(O)m, or C(=O)-.
Preferably, V is selected from heteroaryl and aryl. More preferably, V is phenyl or pyridyl.
Preferably, X and Y are independently selected from: a bond, -S(=O)m, -C(O)NRl0-, -NRlOC(O)-, -S(O)2N(RlO)-, -N(RlO)S(O)2-.and -C(=O)-.
Most preferably, X and Y are bonds.
Preferably, Zl and Z2 are independently selected from unsubstituted or substituted phenyl, unsubstituted or substituted naphthyl, unsubstituted or substituted pyridyl, unsubstituted or substituted furanyl and unsubstituted or substituted thienyl. More preferably, Zl is selected from unsubstituted or substituted phenyl and unsubstituted or substituted naphthyl. More preferably, Z2 is selected from a bond and unsubstituted or substituted phenyl. Preferably, W is selected from imidazolyl, oxazolyl, pyridyl, and triazolyl. More preferably, W is selected from imidazolyl and pyridyl. Most preferably, W is imidazolyl.
Preferably, n is 0, 1, or 2.
Preferably, r is 1 or 2. Preferably p is 0, 1, 2 or 3. Preferably s is 0 or 1. Preferably, the moiety
Figure imgf000046_0001
is selected from:
Figure imgf000046_0002
It is intended that the definition of any substituent or variable (e.g., Rla, R9, n, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. Thus, -N(RlO)2 represents -NHH, -NHCH3, -NHC2H5, etc. It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials.
The pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed, e.g., from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like: and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.
The pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a basic moiety by conventional chemical methods. Generally, the salts are prepared either by ion exchange chromatography or by reacting the free base with stoichio- metric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents.
Abbreviations which may be used in the description of the chemistry and in the Examples that follow include:
Ac2θ Acetic anhydride;
AIBN 2,2 -Azobisisobutyronitrile;
BINAP 2,2'-Bis(diphenylphosphino)-l,l' binaphthyl; Bn Benzyl;
BOC Boc t-Butoxycarbonyl;
CBz Carbobenzyloxy;
DBAD Di-tert-butyl azodicarboxylate;
DBU 1 ,8-Diazabicyclo[5.4.0]undec-7-ene; DCE 1,2-Dichloroethane;
DIEA N,N-Diisopropylethylamine;
DMAP 4-Dimethylaminopyridine;
DME 1,2-Dimethoxyethane;
DMF N,N-Dimethylformamide; DMSO Methyl sulfoxide; DPPA Diphenylphosphoryl azide;
DTT Dithiothreitol;
EDC l-(3-Dimethylaminopropyl)-3-ethyl-carbodiimide-hydrochloride;
EDTA Ethylenediaminetetraacetic acid;
Et2 Diethyl ether;
Et3N Triethylamine;
EtOAc Ethyl acetate;
EtOH Ethanol;
FAB Fast atom bombardment;
HEPES 4-(2-Hydroxyethyl)- 1 -piperazineethanesulfonic acid;
HOBT 1-Hydroxybenzotriazole hydrate;
HOOBT 3-Hydroxy- 1 ,2,2-benzotriazin-4(3H)-one;
HPLC High-performance liquid chromatography;
LAH Lithium aluminum hydride;
MCPBA -Chloroperoxybenzoic acid;
Me Methyl;
MeOH Methanol;
Ms Methanesulf onyl ;
MsCl Methanesulfonyl chloride; n-Bu rc-butyl; n-Bu3P Tri-n-butylphosphine;
NaHMDS Sodium bis(trimethylsilyl)amide;
NBS N-Bromosuccinimide;
Pd2(dba)2 Tiis(dibenzylideneacetone)dipalladinm (0)
Ph phenyl;
PMSF a-Toluenesulfonyl chloride;
Py or pyr Pyridine;
PYBOP Benzotriazole-1-yl-oxy-trispyrrolidinophosphonium hexafluorophosphate;
RT room temperature; t-Bu tert-Butyl;
TBAF Tetrabutylammoniumfluoride;
RPLC Reverse Phase Liquid Chromatography;
TBSC1 tert-Butyldimethylsilyl chloride;
TFA Trifluoroacetic acid; THF Tetrahydrofuran;
TIPS Triisopropylsilyl;
TMS Tetramethylsilane; and
Tr Trityl.
Reactions used to generate the compounds of this invention are prepared by employing reactions as shown in the Schemes 1-11, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures. These reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the reactions described in the Schemes. The procedures discussed and illustrated in the following schemes and synopsis may be used in the preparation of the compounds of the instant invention, for either (R) or (S) stereochemistry.
Synopsis of Schemes
Unless otherwise indicated, variable n is defined as 0 to 6, variable p is defined as 0 to 4, and variable t is defined as 0 or 1. For simplicity, the number of substitutions possible on a ring has not been included. For example, R8 in the schemes represents (R8)r in formula I. In the following schemes, substituent Z represents Zl or Z2 of formula I and substitutent R represents the optional substitutions provided in the defintion of Zl or Z of formula I. Additionally,
Figure imgf000049_0001
represents substituent W designated in formula I.
The requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures. Schemes 1-4 outline the syntheses of various aminopyrrolidine or aminopiperidine intermediates used in the syntheses of the macrocyclic compounds of the instant invention. Boc-protected amino heterocycles 1 and 6, available commerically or by procedures known to those skilled in the art, can be sulfonylated as in Schemes 1 and 2 to give compounds 2 and 7. Compounds 2 and 7 can then deprotected to give intermediates 5 and 8, respectively, which are used in Scheme 5, for example, to form the macrocyclic compounds of the instant invention. In Scheme 3, the boc-protected amino heterocycle 6 is reductively alkylated with a suitably substituted aldehyde B to give intermediate 9 which is then deprotected to provide compound 10. In Scheme 4, heterocycle 6 is coupled with a suitably substituted carboxylic acid to provide compound 11 which is deprotected as shown in Scheme 3. Compound 1 can be substituted for compound 6 in both Schemes 3 and 4.
Scheme 5 outlines a generic synthesis of the macrocyclic compounds where Rid is OH. The aldehyde 13 reacts with an organolithium or Grignard reagent to give the addition product 14 followed by oxidation to the ketone 15. Addition of the requisite organolithium or Grignard to ketone 15 provides compound 16 which then is oxidized to aldehyde 17. Reductive alkylation of intermediate 5 with compound 17 provides compound 18. Cyclization of 18 with cesium carbonate yields the macrocyclic compound 19 of the instant invention.
Scheme 6 illustrates the preparation of the macrocyclic compounds of the instant invention wherein W is imidazole and Rid is NH2- Imidazole 20 is masked at the 2-position with TMSCl, then metallated a second time and reacted with aldehyde 13 to give compound 21. Next, oxidation and treatment with t-butylsulfinamide provides compound 23. Treatment of 23 with silyl-protected oxyalkyl Grignard reagent followed by removal of the silyl group gives alcohol 24. Oxidation of the alcohol gives aldehyde 25 which reductively alkylates amine 5, for example, to give intermediate 26. Cyclization with cesium carbonate provides the macrocycle compound 28 after deprotection.
Scheme 7 shows an alternative method for the preparation of aldehyde 25. Compound 23 is reacted with an alkenyl Grignard to give compound 29. Subsequent oxidative cleavage of 29 provides aldehyde 25 which can be carried on as illustrated in Scheme 6.
Scheme 8 describes the preparation of the macrocyclic compounds of the instant invention wherein the macrocycle is linked by an amide bond. Aldehyde 25 is oxidized to carboxylic acid 30 which is further coupled to compound 4. Heating of aldehyde 25 with cesium carbonate removes the mesylate and forms a macrocyclic compound of the instant invention 32, after deprotection. Scheme 9 illustrates macrocyclic compounds of the instant invention where the constraining heterocycle is an aminopyrrolidinone. The pyrrolidinone 36 was reductively alkylated with aldehyde 25 to give compound 37. Cyclization of 37 with cesium carbonate followed by deprotection leads to the macrocyclic compound 39 of the instant invention.
In Scheme 10, heterocyclic carboxylic acids are prepared. Scheme 11 describes the macrocyclic compounds of the instant invention wherein the macro- cyclic connection is through the heteroatom at the benzylic site between imidazole and fluorophenyl. Compound 43 was coupled to compounds 39 or 40 or 42 to give compound 44. Removal of the methyl group with BBr3 gives the phenol 45 which is cyclized to give the macrocyclic compounds 46 of the instant invention.
SCHEME 1
Figure imgf000051_0001
SCHEME 2
Figure imgf000052_0001
Et3N m = 1 or 2
Figure imgf000052_0002
SCHEME 3
Figure imgf000052_0003
aCNBH3 m = 1 or 2
Figure imgf000052_0004
SCHEME 4
Figure imgf000053_0001
m = 1 or 2
SCHEME 5
Figure imgf000053_0002
SCHEME 5 (CONTINUED)
Figure imgf000054_0001
Figure imgf000054_0002
19 SCHEME 6
Figure imgf000055_0001
Figure imgf000055_0002
R is alkyl
SCHEME 6 (CONTINUED)
pyr SOG reductive alkylation
Figure imgf000056_0001
Figure imgf000056_0002
SCHEME 7
Figure imgf000057_0001
Ra = H or CH3
SCHEME 8
Figure imgf000058_0001
SCHEME 9
Figure imgf000059_0001
PYBOP, DIEA CH2CI2
Figure imgf000059_0002
SCHEME 9 (CONTINUED)
Figure imgf000060_0001
x = 0 to 6
Figure imgf000060_0002
x = 0 to 6
SCHEME 10
Figure imgf000061_0001
SCHEME 11
fl
Figure imgf000062_0001
In a preferred embodiment of the instant invention the compounds of the invention are selective inhibitors of farnesyl-protein transferase. A compound is considered a selective inhibitor of farnesyl-protein transferase, for example, when its in vitro farnesyl-protein transferase inhibitory activity, as assessed by the assay described in Example 5, is at least 100 times greater than the in vitro activity of the same compound against geranylgeranyl-protein transferase-type I in the assay described in Example 6. Preferably, a selective compound exhibits at least 1000 times greater activity against one of the enzymatic activities when comparing geranylgeranyl-protein transferase-type I inhibition and farnesyl-protein transferase inhibition.
It is also preferred that the selective inhibitor of farnesyl-protein transferase is further characterized by: a) an IC50 (a measure of in vitro inhibitory activity) for inhibition of the prenylation of newly synthesized K-Ras protein more than about 100-fold higher than the EC50 for the inhibition of the farnesylation of hDJ protein.
When measuring such IC50S and EC50S the assays described in Example 10 may be utilized.
It is also preferred that the selective inhibitor of farnesyl-protein transferase is further characterized by: b) an IC50 (a measurement of in vitro inhibitory activity) for inhibition of
K4B-Ras dependent activation of MAP kinases in cells at least 100-fold greater than the EC50 for inhibition of the farnesylation of the protein hDJ in cells.
It is also preferred that the selective inhibitor of farnesyl-protein transferase is further characterized by: c) an IC50 (a measurement of in vitro inhibitory activity) against H-Ras dependent activation of MAP kinases in cells at least 1000 fold lower than the inhibitory activity (IC50) against H-rø-.-CNLL (SEQ.ID.ΝO.: 1) dependent activation of MAP kinases in cells. When measuring Ras dependent activation of MAP kinases in cells the assays described in Example 9 may be utilized.
In another preferred embodiment of the instant invention the compounds of the invention are dual inhibitors of farnesyl-protein transferase and geranylgeranyl-protein transferase type I. Such a dual inhibitor may be termed a Class II prenyl-protein transferase inhibitor and will exhibit certain characteristics when assessed in in vitro assays, which are dependent on the type of assay employed.
In a SEAP assay, such as described in Example 9, it is preferred that the dual inhibitor compound has an in vitro inhibitory activity (IC50) that is less than about 12μM against K4B-Ras dependent activation of MAP kinases in cells. The Class II prenyl-protein transferase inhibitor may also be characterized by: a) an IC50 (a measurement of in vitro inhibitory activity) for inhibiting
K4B-Ras dependent activation of MAP kinases in cells between 0.1 and 100 times the IC50 for inhibiting the farnesylation of the protein hDJ in cells; and b) an IC50 (a measurement of in vitro inhibitory activity) for inhibiting
K4B-Ras dependent activation of MAP kinases in cells greater than 5-fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with the pCMN-SEAP plasmid that constitutively expresses the SEAP protein.
The Class II prenyl-protein transferase inhibitor may also be characterized by: a) an IC50 (a measurement of in vitro inhibitory activity) against H-Ras dependent activation of MAP kinases in cells greater than 2 fold lower but less than 20,000 fold lower than the inhibitory activity (IC50) against
H-r -.-CNLL (SEQ.ID.ΝO.: 1) dependent activation of MAP kinases in cells; and b) an IC50 (a measurement of in vitro inhibitory activity) against H-ras-
CNLL dependent activation of MAP kinases in cells greater than 5-fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with the pCMN-SEAP plasmid that constitutively expresses the SEAP protein.
The Class II prenyl-protein transferase inhibitor may also be characterized by: a) an IC50 (a measurement of in vitro inhibitory activity) against H-Ras dependent activation of MAP kinases in cells greater than 10-fold lower but less than 2,500 fold lower than the inhibitory activity (IC50) against H-ras-
CNLL (SEQ.ID.ΝO.: 1) dependent activation of MAP kinases in cells; and b) an IC50 (a measurement of in vitro inhibitory activity) against H-ras- CNLL dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with the pCMN-SEAP plasmid that constitutively expresses the SEAP protein.
A method for measuring the activity of the inhibitors of prenyl-protein transferase, as well as the instant combination compositions, utilized in the instant methods against Ras dependent activation of MAP kinases in cells is described in Example 9.
In yet another embodiment, a compound of the instant invention may be a more potent inhibitor of geranylgeranyl-protein transferase-type I than it is an inhibitor of farnesyl-protein transferase.
The instant compounds are useful as pharmaceutical agents for mammals, especially for humans. These compounds may be administered to patients for use in the treatment of cancer. Examples of the type of cancer which may be treated with the compounds of this invention include, but are not limited to, colorectal carcinoma, exocrine pancreatic carcinoma, myeloid leukemias and neurological tumors. Such tumors may arise by mutations in the ras genes themselves, mutations in the proteins that can regulate Ras activity (i.e., neurofibromin (ΝF-1), neu, src, abl, lck, fyn) or by other mechanisms.
The compounds of the instant invention inhibit farnesyl-protein transferase and the farnesylation of the oncogene protein Ras. The instant compounds may also inhibit tumor angiogenesis, thereby affecting the growth of tumors (J. Rak et al. Cancer Research, 55:4575-4580 (1995)). Such anti-angiogenesis properties of the instant compounds may also be useful in the treatment of certain forms of vision deficit related to retinal vascularization. The compounds of this invention are also useful for inhibiting other proliferative diseases, both benign and malignant, wherein Ras proteins are aberrantly activated as a result of oncogenic mutation in other genes (i.e., the Ras gene itself is not activated by mutation to an oncogenic form) with said inhibition being accomplished by the administration of an effective amount of the compounds of the invention to a mammal in need of such treatment. For example, the composition is useful in the treatment of neurofibromatosis, which is a benign proliferative disorder. The instant compounds may also be useful in the treatment of certain viral infections, in particular in the treatment of hepatitis delta and related viruses (J.S. Glenn et al. Science, 256:1331-1333 (1992). The compounds of the instant invention are also useful in the prevention of restenosis after percutaneous transluminal coronary angioplasty by inhibiting neointimal formation (C. Indolfi et al. Nature medicine, 1:541-545(1995). The instant compounds may also be useful in the treatment and prevention of polycystic kidney disease (D.L. Schaffner et al. American Journal of Pathology, 142:1051-1060 (1993) and B. Cowley, Jr. et al., FASEB Journal, 2: A3160 (1988)).
The instant compounds may also be useful for the treatment of fungal infections. The instant compounds may also be useful as inhibitors of proliferation of vascular smooth muscle cells and therefore useful in the prevention and therapy of arteriosclerosis and diabetic vascular pathologies.
The compounds of the instant invention may also be useful in the prevention and treatment of endometriosis, uterine fibroids, dysfunctional uterine bleeding and endometrial hyperplasia.
In such methods of prevention and treatment as described herein, the prenyl-protein transferase inhibitors of the instant invention may also be co- administered with other well known therapeutic agents that are selected for their particular usefulness against the condition that is being treated. For example, the prenyl-protein transferase inhibitor may be useful in further combination with drugs known to supress the activity of the ovaries and slow the growth of the endometrial tissue. Such drugs include but are not limited to oral contraceptives, progestins, danazol and GnRH (gonadotropin-releasing hormone) agonists.
Administration of the prenyl-protein transferase inhibitor may also be combined with surgical treatment of endometriosis (such as surgical removal of misplaced endometrial tissue) where appropriate.
The instant compounds may also be useful as inhibitors of corneal inflammation. These compounds may improve the treatment of corneal opacity which results from cauterization-induced corneal inflammation. The instant compounds may also be useful in reducing corneal edema and neovascularization. (K. Sonoda et al., Invest. Ophthalmol. Vis. Sci., 1998, vol. 39, p 2245-2251).
The compounds of this invention may be administered to mammals, preferably humans, either alone or, preferably, in combination with pharmaceutically acceptable carriers, excipients or diluents, in a pharmaceutical composition, according to standard pharmaceutical practice. The compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
Additionally, the compounds of the instant invention may be administered to a mammal in need thereof using a gel extrusion mechanism (GEM) device, such as that described in US Serial No. 60/144,643, filed on July 20, 1999, which is hereby incorporated by reference.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specific amounts, as well as any product which results, directly or indirectly, from combination of the specific ingredients in the specified amounts.
The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, or alginic acid; binding agents, for example starch, gelatin, polyvinyl-pyrrolidone or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to mask the unpleasant taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a water soluble taste masking material such as hydroxypropyl-methylcellulose or hydroxypropyl- cellulose, or a time delay material such as ethyl cellulose, cellulose acetate buryrate may be employed.
Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water soluble carrier such as polyethyl- eneglycol or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellu- lose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethylene- oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as butylated hydroxyanisol or alpha-tocopherol.
Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
The pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsions. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring phospha- tides, for example soy bean lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening, flavoring agents, preservatives and antioxidants.
Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.
The pharmaceutical compositions may be in the form of a sterile injectable aqueous solutions. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
The sterile injectable preparation may also be a sterile injectable oil- in-water microemulsion where the active ingredient is dissolved in the oily phase. For example, the active ingredient may be first dissolved in a mixture of soybean oil and lecithin. The oil solution then introduced into a water and glycerol mixture and processed to form a microemulation.
The injectable solutions or microemulsions may be introduced into a patient's blood-stream by local bolus injection. Alternatively, it may be advantageous to administer the solution or microemulsion in such a way as to maintain a constant circulating concentration of the instant compound. In order to maintain such a constant concentration, a continuous intravenous delivery device may be utilized. An example of such a device is the Deltec CADD-PLUS™ model 5400 intravenous pump.
The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension for intramuscular and subcutaneous administration. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
Compounds of Formula A may also be administered in the form of a suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compound of Formula A are employed. (For purposes of this application, topical application shall include mouth washes and gargles.) The compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles and delivery devices, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen. Compounds of the present invention may also be delivered as a suppository employing bases such as cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
When a compound according to this invention is administered into a human subject, the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, sex and response of the individual patient, as well as the severity of the patient's symptoms.
In one exemplary application, a suitable amount of compound is administered to a mammal undergoing treatment for cancer. Administration occurs in an amount between about 0.1 mg/kg of body weight to about 60 mg/kg of body weight per day, preferably of between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day.
The compounds of the instant invention may also be co-administered with other well known therapeutic agents that are selected for their particular useful- ness against the condition that is being treated. For example, the compounds of the instant invention may also be co-administered with other well known cancer therapeutic agents that are selected for their particular usefulness against the condition that is being treated. Included in such combinations of therapeutic agents are combinations of the instant prenyl-protein transferase inhibitors and an antineoplastic agent. It is also understood that such a combination of antineoplastic agent and inhibitor of prenyl-protein transferase may be used in conjunction with other methods of treating cancer and/or tumors, including radiation therapy and surgery. It is further understood that any of the therapeutic agents described herein may also be used in combination with a compound of the instant invention and an antineoplastic agent. Examples of an antineoplastic agent include, in general, microtubule- stabilizing agents such as paclitaxel (also known as Taxol®), docetaxel (also known as Taxotere®), epothilone A, epothilone B, desoxyepothilone A, desoxyepothilone B or their derivatives); microtubule-disruptor agents; alkylating agents, for example, nitrogen mustards, ethyleneimine compounds, alkyl sulfonates and other compounds with an alkylating action such as nitrosoureas, cisplatin, and dacarbazine; anti-metabolites, for example, folic acid, purine or pyrimidine antagonists; epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes; biological response modifiers and growth inhibitors; mitotic inhibitors, for example, vinca alkaloids and derivatives of podophyllotoxin; cytotoxic antibiotics; hormonal/anti-hormonal therapeutic agents, haematopoietic growth factors and antibodies (such as trastuzumab, also known as Herceptin™).
Example classes of antineoplastic agents include, for example, the anthracycline family of drugs, the vinca drugs, the mitomycins, the bleomycins, the cytotoxic nucleosides, the taxanes, the epothilones, discodermolide, the pteridine family of drugs, diynenes and the podophyllotoxins. Particularly useful members of those classes include, for example, doxorubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloro-methotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine arabinoside, podophyllotoxin or podo-phyllotoxin derivatives such as etoposide, etoposide phosphate or teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine, leurosine, paclitaxel and the like. Other useful antineoplastic agents include estramustine, cisplatin, carboplatin, cyclophosphamide, bleomycin, tamoxifen, ifosamide, melphalan, hexamethyl melamine, thiotepa, cytarabin, idatrexate, trimetrexate, dacarbazine, L-asparaginase, dactinomycin, mechlorethamine (nitrogen mustard), streptozocin, cyclophosphamide, carmustine (BCNU), lomustine (CCNU), procarbazine, mitomycin, cytarabine, etoposide, methotrexate, bleomycin, chlorambucil, camptothecin, CPT-11, topotecan, ara-C, bicalutamide, flutamide, leuprolide, pyridobenzoindole derivatives, interferons and interleukins. Particular examples of antineoplastic, or chemotherapeutic, agents are described, for example, by D. J. Stewart in "Nausea and Vomiting: Recent Research and Clinical Advances", Eds. J. Kucharczyk, et al., CRC Press Inc., Boca Raton, Florida, USA (1991), pages 177-203, especially page 188. See also, R. J. Gralla, et al., Cancer Treatment Reports, 68(1), 163-172 (1984).
The preferred class of antineoplastic agents is the taxanes and the preferred antineoplastic agent is paclitaxel.
The compounds of the instant invention may also be co-administered with antisense oligonucleotides which are specifically hybridizable with RNA or DNA deriving from human ras gene. Such antisense oligonucleotides are described in U.S. Patent. No. 5,576,208 and PCT Publication No. WO 99/22772. The instant compounds are particularly useful when co-administered with the antisense oligonucleotide comprising the amino acid sequence of SEQ.ID.NO: 2 of U.S. Patent No. 5,576,208. Certain compounds of the instant invention may exhibit very low plasma concentrations and significant inter-individual variation in the plasma levels of the compound. It is believed that very low plasma concentrations and high inter- subject variability achieved following administration of certain prenyl-protein transferase inhibitors to mammals may be due to extensive metabolism by cytochrome P450 enzymes prior to entry of drug into the systemic circulation. Prenyl-protein transferase inhibitors may be metabolized by cytochrome P450 enzyme systems, such as CYP3A4, CYP2D6, CYP2C9, CYP2C19 or other cytochrome P450 isoform. If a compound of the instant invention demonstrates an affinity for one or more of the cytochrome P450 enzyme systems, another compound with a higher affinity for the P450 enzyme(s) involved in metabolism should be administered concomitantly. Examples of compounds that have a comparatively very high affinity for CYP3 A4, CYP2D6, CYP2C9, CYP2C19 or other P450' isoform include, but are not limited to, piperonyl butoxide, troleandomycin, erythromycin, proadifen, isoniazid, allyliso- propylacetamide, ethinylestradiol, chloramphenicol, 2-ethynylnaphthalene and the like. Such a high affinity compound, when employed in combination with a compound of formula A, may reduce the inter-individual variation and increase the plasma concentration of a compound of formula A to a level having substantial therapeutic activity by inhibiting the metabolism of the compound of formula A. Additionally, inhibiting the metabolism of a compound of the instant invention prolongs the pharmacokinetic half-life, and thus the pharmacodynamic effect, of the compound.
A compound of the present invention may be employed in conjunction with antiemetic agents to treat nausea or emesis, including acute, delayed, late-phase, and anticipatory emesis, which may result from the use of a compound of the present invention, alone or with radiation therapy. For the prevention or treatment of emesis a compound of the present invention may be used in conjunction with other anti-emetic agents, especially neurokinin-1 receptor antagonists, 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, or a corticosteroid such as Decadron (dexa- methasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U.S.Patent Nos. 2,789,118, 2,990,401, 3,048,581, 3,126,375, 3,929,768, 3,996,359, 3,928,326 and 3,749,712. For the treatment or prevention of emesis, conjunctive therapy with a neurokinin-1 receptor antagonist, a 5HT3 receptor antagonist and a corticosteroid is preferred. Neurokinin-1 receptor antagonists of use in conjunction with the compounds of the present invention are fully described, for example, in U.S. Patent Nos. 5,162,339, 5,232,929, 5,242,930, 5,373,003, 5,387,595, 5,459,270, 5,494,926, 5,496,833, 5,637,699, 5,719,147; European Patent Publication Nos. EP 0360390, 0 394 989, 0428434, 0429 366, 0430771, 0436 334, 0443 132, 0482 539, 0498 069, 0 499 313, 0512 901, 0 512902, 0514273, 0 514 274, 0 514275, 0 514276, 0 515 681, 0 517 589, 0 520 555, 0522 808, 0 528 495, 0532456, 0 533 280, 0 536 817, 0 545 478, 0 558 156, 0 577 394, 0 585 913,0 590 152, 0 599 538, 0 610793, 0634402, 0686629, 0693 489, 0694 535, 0699655, 0699 674, 0 707006, 0708 101, 0709 375, 0709 376, 0714 891, 0723 959, 0733 632 and 0776 893; PCT International Patent Publication Nos. WO 90/05525, 90/05729, 91/09844, 91/18899, 92/01688, 92/06079, 92/12151, 92/15585, 92/17449, 92/20661, 92/20676, 92/21677, 92/22569, 93/00330, 93/00331, 93/01159, 93/01165, 93/01169, 93/01170, 93/06099, 93/09116, 93/10073, 93/14084, 93/14113, 93/18023, 93/19064, 93/21155, 93/21181, 93/23380, 93/24465, 94/00440, 94/01402, 94/02461, 94/02595, 94/03429, 94/03445, 94/04494, 94/04496, 94/05625, 94/07843, 94/08997, 94/10165, 94/10167, 94/10168, 94/10170, 94/11368, 94/13639, 94/13663, 94/14767, 94/15903, 94/19320, 94/19323, 94/20500, 94/26735, 94/26740, 94/29309, 95/02595, 95/04040, 95/04042, 95/06645, 95/07886, 95/07908, 95/08549, 95/11880, 95/14017, 95/15311, 95/16679, 95/17382, 95/18124, 95/18129, 95/19344, 95/20575, 95/21819, 95/22525, 95/23798, 95/26338, 95/28418, 95/30674, 95/30687, 95/33744, 96/05181, 96/05193, 96/05203, 96/06094, 96/07649, 96/10562, 96/16939, 96/18643, 96/20197, 96/21661, 96/29304, 96/29317, 96/29326, 96/29328, 96/31214, 96/32385, 96/37489, 97/01553, 97/01554, 97/03066, 97/08144, 97/14671, 97/17362, 97/18206, 97/19084, 97/19942 and 97/21702; and in British Patent Publication Nos. 2266 529, 2 268 931, 2 269 170, 2 269590, 2 271 774, 2292 144, 2 293 168, 2293 169, and 2302689. The preparation of such compounds is fully described in the aforementioned patents and publications.
A particularly preferred neurokinin-1 receptor antagonist for use in conjunction with the compounds of the present invention is 2-(R)-(l-(R)-(3,5-bis (trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-4-(3-(5-oxo-lH,4H-l,2,4- triazolo)methyl)morpholine, or a pharmaceutically acceptable salt thereof, which is described in U.S. Patent No. 5,719,147.
For the treatment of cancer, it may be desirable to employ a compound of the present invention in conjunction with another pharmacologically active agent(s). A compound of the present invention and the other pharmacologically active agent(s) may be administered to a patient simultaneously, sequentially or in combination. For example, the present compound may employed directly in combination with the other active agent(s), or it may be administered prior, concurrent or subsequent to the administration of the other active agent(s). In general, the currently available dosage forms of the known therapeutic agents for use in such combinations will be suitable.
For example, a compound of the present invention may be presented together with another therapeutic agent in a combined preparation, such as with an antiemetic agent for simultaneous, separate, or sequential use in the relief of emesis associated with employing a compound of the present invention and radiation therapy. Such combined preparations may be, for example, in the form of a twin pack. A preferred combination comprises a compound of the present invention with antiemetic agents, as described above.
Radiation therapy, including x-rays or gamma rays which are delivered from either an externally applied beam or by implantation of tiny radioactive sources, may also be used in combination with the instant inhibitor of prenyl-protein transferase alone to treat cancer.
Additionally, compounds of the instant invention may also be useful as radiation sensitizers, as described in WO 97/38697, published on October 23, 1997, and herein incorporated by reference. The instant compounds may also be useful in combination with other inhibitors of parts of the signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation. Thus, the instant compounds may be utilized in combination with farnesyl pyrophosphate competitive inhibitors of the activity of farnesyl-protein transferase or in combination with a compound which has Raf antagonist activity. The instant compounds may also be co-administered with compounds that are selective inhibitors of geranylgeranyl protein transferase.
In particular, if the compound of the instant invention is a selective inhibitor of farnesyl-protein transferase, co-administration with a compound(s) that is a selective inhibitor of geranylgeranyl protein transferase may provide an improved therapeutic effect.
In particular, the compounds disclosed in the following patents and publications may be useful as farnesyl pyrophosphate-competitive inhibitor component of the instant composition: U.S. Serial Nos. 08/254,228 and 08/435,047. Those patents and publications are incorporated herein by reference.
In practicing methods of this invention, which comprise administering, simultaneously or sequentially or in any order, two or more of a protein substrate- competitive inhibitor and a farnesyl pyrophosphate-competitive inhibitor, such administration can be orally or parenterally, including intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration. It is preferred that such administration be orally. It is more preferred that such administration be orally and simultaneously. When the protein substrate-competitive inhibitor and farnesyl pyrophosphate-competitive inhibitor are administered sequentially, the administration of each can be by the same method or by different methods.
The instant compounds may also be useful in combination with an integrin antagonist for the treatment of cancer, as described in U.S. Serial No. 09/055,487, filed April 6, 1998, and WO 98/44797, published on October 15, 1998, which are incorporated herein by reference. As used herein the term an integrin antagonist refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to an integrin(s) that is involved in the regulation of angiogenisis, or in the growth and invasiveness of tumor cells. In particular, the term refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the vβ3 integrin, which selectively antagonize, inhibit or counteract binding of a physiological ligand to the αvβ5 integrin, which antagonize, inhibit or counteract binding of a physiological ligand to both the αvβ3 integrin and the αvβ5 integrin, or which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells. The term also refers to antagonists of the αlβl, 2βl, α5βl, 6βl and α6β4 integrins. The term also refers to antagonists of any combination of vβ3 integrin, ocvβ5 integrin, lβl, 2βl, cc5βl, α6βl and α6β4 integrins. The instant compounds may also be useful with other agents that inhibit angiogenisis and thereby inhibit the growth and invasiveness of tumor cells, including, but not limited to angiostatin and endostatin.
The instant compounds may also be useful in combination with an inhibitor of 3-hydroxy-3-methyIglutaryl-CoA reductase (HMG-CoA reductase) for the treatment of cancer. Compounds which have inhibitory activity for HMG-CoA reductase can be readily identified by using assays well-known in the art. For example, see the assays described or cited in U.S. Patent 4,231,938 at col. 6, and WO 84/02131 at pp. 30-33. The terms "HMG-CoA reductase inhibitor" and "inhibitor of HMG-CoA reductase" have the same meaning when used herein. Examples of HMG-CoA reductase inhibitors that may be used include but are not limited to lovastatin (MENACOR®; see US Patent No. 4,231,938; 4,294,926; 4,319,039), simvastatin (ZOCOR®; see US Patent No. 4,444,784; 4,820,850; 4,916,239), pravastatin (PRAVACHOL®; see US Patent Nos. 4,346,227; 4,537,859; 4,410,629; 5,030,447 and 5,180,589), fluvastatin (LESCOL®; see US Patent Nos. 5,354,772; 4,911,165; 4,929,437; 5,189,164; 5,118,853; 5,290,946; 5,356,896), atorvastatin (L-PITOR®; see US Patent Nos. 5,273,995; 4,681,893; 5,489,691; 5,342,952) and cerivastatin (also known as rivastatin and BAYCHOL®; see US Patent No. 5,177,080). The structural formulas of these and additional HMG-CoA reductase inhibitors that may be used in the instant methods are described at page 87 of M. Yalpani, "Cholesterol Lowering Drugs", Chemistry & Industry, pp. 85-89 (5 February 1996) and US Patent Nos. 4,782,084 and 4,885,314. The term HMG-CoA reductase inhibitor as used herein includes all pharmaceutically acceptable lactone and open-acid forms (i.e., where the lactone ring is opened to form the free acid) as well as salt and ester forms of compounds which have HMG-CoA reductase inhibitory activity, and therefor the use of such salts, esters, open-acid and lactone forms is included within the scope of this invention. An illustration of the lactone portion and its corresponding open-acid form is shown below as structures I and II.
Figure imgf000077_0001
Lactone Open-Acid
I II
In HMG-CoA reductase inhibitors where an open-acid form can exist, salt and ester forms may preferably be formed from the open-acid, and all such forms are included within the meaning of the term "HMG-CoA reductase inhibitor" as used herein. Preferably, the HMG-CoA reductase inhibitor is selected from lovastatin and simvastatin, and most preferably simvastatin. Herein, the term "pharmaceutically acceptable salts" with respect to the HMG-CoA reductase inhibitor shall mean non- toxic salts of the compounds employed in this invention which are generally prepared by reacting the free acid with a suitable organic or inorganic base, particularly those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc and tetramethylammonium, as well as those salts formed from amines such as ammonia, ethylenediamine, N-methylglucamine, lysine, arginine, ornithine, choline, N,N'-dibenzylethylenediamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, l-p-chlorobenzyl-2-pyrrolidine-l'-yl-methyl- benzimidazole, diethylamine, piperazine, and tris(hydroxymethyl) aminomethane. Further examples of salt forms of HMG-CoA reductase inhibitors may include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynapthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylsulfate, mucate, napsylate, nitrate, oleate, oxalate, pamaote, palmitate, panthothenate, phosphate/ diphosphate, polygalacturonate, salicylate, stearate, subacetate, succinate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate.
Ester derivatives of the described HMG-CoA reductase inhibitor compounds may act as prodrugs which, when absorbed into the bloodstream of a warm-blooded animal, may cleave in such a manner as to release the drug form and permit the drug to afford improved therapeutic efficacy.
Similarly, the instant compounds may be useful in combination with agents that are effective in the treatment and prevention of NF-1, restenosis, polycystic kidney disease, infections of hepatitis delta and related viruses and fungal infections.
If formulated as a fixed dose, such combination products employ the combinations of this invention within the dosage range described above and the other pharmaceutically active agent(s) within its approved dosage range. Combinations of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a multiple combination formulation is inappropriate.
The instant compounds may also be useful in combination with prodrugs of antineoplastic agents. In particular, the instant compounds may be co-administered either concurrently or sequentially with a conjugate (termed a "PSA conjugate") which comprises an oligopeptide, that is selectively cleaved by enzymatically active prostate specific antigen (PSA), and an antineoplastic agent. Such co-administration will be particularly useful in the treatment of prostate cancer or other cancers which are characterized by the presence of enzymatically active PSA in the immediate surrounding cancer cells, which is secreted by the cancer cells. Compounds which are PSA conjugates and are therefore useful in such a co-administration, and methods of synthesis thereof, can be found in the following patents, pending patent applications and publications which are herein incorporated by reference:
U.S. Patent No. 5,599,686, granted on February 4, 1997;
WO 96/00503 (January 11, 1996); USSN 08/404,833, filed on March 15, 1995; USSN 08/468,161, filed on June 6, 1995;
U.S. Patent No. 5,866,679, granted on February 2, 1999; WO 98/10651 (March 19, 1998); USSN 08/926,412, filed on September 9, 1997;
WO 98/18493 (May 7, 1998); USSN 08/950,805, filed on October 14, 1997;
WO 99/02175 (January 21, 1999); USSN 09/112,656, filed on July 9, 1998; and
WO 99/28345 (June 10, 1999); USSN 09/193,365, filed on November 17, 1998.
Compounds which are described as prodrugs wherein the active therapeutic agent is released by the action of enzymatically active PSA and therefore may be useful in such a co-administration, and methods of synthesis thereof, can be found in the following patents, pending patent applications and publications, which are herein incorporated by reference: WO 98/52966 (November 26, 1998). All patents, publications and pending patent applications identified are herein incorporated by reference.
The compounds of the instant invention are also useful as a component in an assay to rapidly determine the presence and quantity of farnesyl-protein transferase (FPTase) in a composition. Thus the composition to be tested may be divided and the two portions contacted with mixtures which comprise a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl pyrophosphate and, in one of the mixtures, a compound of the instant invention. After the assay mixtures are incubated for an sufficient period of time, well known in the art, to allow the FPTase to farnesylate the substrate, the chemical content of the assay mixtures may be determined by well known immuno-logical, radiochemical or chromatographic techniques. Because the compounds of the instant invention are selective inhibitors of FPTase, absence or quantitative reduction of the amount of substrate in the assay mixture without the compound of the instant invention relative to the presence of the unchanged substrate in the assay containing the instant compound is indicative of the presence of FPTase in the composition to be tested.
It would be readily apparent to one of ordinary skill in the art that such an assay as described above would be useful in identifying tissue samples which contain farnesyl-protein transferase and quantitating the enzyme. Thus, potent inhibitor compounds of the instant invention may be used in an active site titration assay to determine the quantity of enzyme in the sample. A series of samples composed of aliquots of a tissue extract containing an unknown amount of farnesyl- protein transferase, an excess amount of a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl pyrophosphate are incubated for an appropriate period of time in the presence of varying concentrations of a compound of the instant invention. The concentration of a sufficiently potent inhibitor (i.e., one that has a Ki substantially smaller than the concentration of enzyme in the assay vessel) required to inhibit the enzymatic activity of the sample by 50% is approximately equal to half of the concentration of the enzyme in that particular sample.
EXAMPLES
Examples provided are intended to assist in a further understanding of the invention. Particular materials employed, species and conditions are intended to be further illustrative of the invention and not limitative of the reasonable scope thereof.
EXAMPLE 1
Preparation of 5-[(17R)-10-cyano-14R-methyl-2,2-dioxido-16-oxo-8-oxa-2-thia-l, 15- diazatetracyclo[15.3.1.13'7.l9,13]tricosa-3(23),4,6,9(22),10,12-hexaen-14-yl]-l- methyl-lH-imidazol-1-ium trifluoroacetate and 5-[(17S)-10-cyano-14R-methyl-2,2- dioxido-16-oxo-8-oxa-2-thia-l,15-diazatetracyclo[15.3.1.13'7.l9,13]tricosa-
3r23).4.6.9r22 .10.12-hexaen-14-yll-l-methyl-lH-imidazol-l-ium trifluoroacetate
Step A: Preparation of 4-Bromo-3-fluorobenzoic acid
4-Bromo-3-fluorotoluene (40.0 g, 0.212 mol) was heated at 90°C in H2O (200 mL)-pyridine (200 mL) with mechanical stirring under Ar. Potassium permanganate (KMnO-j.) (67 g, 0.424 mol) was added portionwise over 3 hours.
After 4 hours, an HPLC of a filtered sample indicated 50% conversion to the acid. An additional 30 g of KMnθ4 was added and heating continued overnight. HPLC indicated 81% conversion. Further KMnθ4 was added portionwise with reaction monitoring by HPLC until >95% conversion was obtained. The reaction mixture was filtered through Celite, the filter pad washed with H2O, aq NaOH and EtOH. The filtrate was concentrated to a small volume, then partitioned between 3N NaOH solution and diethyl ether. The aqueous basic layer was separated, cooled in an ice- H2O bath and acidified slowly with 6N HCI solution to precipitate the white solid product. This was collected by suction filtration and dried at 40°C. in a vacuum oven overnight to give the title compound, mp 190 -192°C. *H NMR (CDCI3) δ 7.83
(dd, 1H, J = 2, 9 Hz), 7.78 (dd, 1H, J = 2, 8 Hz), 7.67 - 7.71 (m, 1H).
Step B: Preparation of 4-bromo-3-fluorobenzyl alcohol 4-Bromo-3-fluorobenzoic acid (40.8 g, 0.187 mol) was dissolved in THF (250 ml) with magnetic stirring under Ar in an ice-H2θ bath. The cloudy solution was treated dropwise with borane-THF complex (1 M) (374 mL, 0.374 mol) over a 1 hour period maintaining the internal temperature at <10°C. The reaction mixture was left to warm to ambient temperature overnight, then cooled in an ice-H2θ bath and treated dropwise with H2O (150 mL). The THF was removed on a rotary evaporator, and the residue partitioned between EtOAc and H2O. The aqueous layer was extracted with EtOAc (3 x 100 mL), the organic layers combined, washed with brine, and dried (Na2SO4), filtered, and concentrated to give the title compound as an oil which solidified on standing. 1H NMR (CDCI3) δ 7.52 (t, 1H, J = 8 Hz), 7.16 (d, 1H, J = 9 Hz), 7.02 (d, 1H, J = 8 Hz), 4.67 (s, 2H), 1.47 (br s, 1H).
Step C: Preparation of 2-fluoro-4-hydroxymethylbenzonitrile
4-Bromo-3-fluorobenzyl alcohol (20 g, 0.097 mol) was dissolved in DMF (100 mL) then placed under high vacuum for 15 min. The solution was then purged with Ar for 15 min. While purging continued, zinc cyanide (8 g, 0.068 mol) and the catalyst, Pd[(PPh3)]4, (5.63 g, 0.0049 mol) were added. The reaction mixture was heated at 95°C. under Ar for 18 h, then cooled to ambient temperature and added to H2O. The mixture was extracted with EtOAc, then washed with IM HCI, H2O, brine, and dried (Na2SO4). Filtration and concentration to dryness gave the title compound as a white solid after chromatography (silica gel, hexane: EtOAc, 6.5:3.5). !H NMR (CDCI3) δ 7.61 (t, 1H, J = 8 Hz), 7.23 - 7.29 (m, 2H), 4.80 (d, 2H,
J = 6 Hz), 1.93 (t, 1H, J = 6Hz). Step D: Preparation of 2-Fluoro-4-formylbenzonitrile
2-Fluoro-4-hydroxymethylbenzonitrile (10 g, 0,066 mol) and triethyl- amine (32.3 mL, 0.231 mol) were dissolved in CH2CI2 (100 mL)- DMSO (20 mL) at <5°C with stirring and treated dropwise with a solution of pyridine»S03 complex (31.5 g, 0.198 mol) in DMSO (70 mL) maintaining the reaction mixture temperature at <10°C. The reaction mixture was stirred at 5°C for 1 hour after the addition, then at 20°C for 1 hour, then partitioned between CH2CI2 and H2O. The organic layer was separated, washed well with H2O, brine, and dried (Na2SO4). Filtration and concentration gave the title compound after purification by chromatography (silica gel, hexane: EtOAc, 3:1). lR NMR (CDCI3) δ 10.06 (d, IH, J = 2 Hz), 7.86 (dd, IH,
J = 5,8 Hz), 7.798 (dd, IH, J = 1, 8 Hz), 7.728 (dd, IH, J = 1, 8 Hz).
Step E: Preparation of 2-Fluoro-4-[hydroxy-(3-methyl-3H-imidazol-4-yl)- methyll -benzonitrile
1-Methylimidazole (15.88 mL, 0.199 mol), dissolved in anhydrous TΗF (500 mL) in flame-dried glassware under Ar, was cooled to -78°C and treated with n-butyl lithium (1.6M in hexane)(124 mL, 0.199 mol) via syringe. After stirring for 1 hour chlorotriethylsilane (33.4 mL, 0.199 mol) was added and the reaction mixture was left to warm to ambient temperature overnight. The TΗF was removed in vacuo with gentle warming, and the residue was redissolved in dry TΗF (500 mL), cooled to -78°C, and treated with sec-butyl lithium (1.3M in cyclohexane) (153 mL, 0.199 mol) dropwise. After 1 hour this solution was cannulated into a solution of 2-fluoro-4-formylbenzonitrile (27 g, 0.181 mol) in TΗF (200 mL). After 15 minutes the cooling bath was removed, the mixture was stirred for 2 hr at ambient temperature, then was quenched with saturated NΗ4CI solution. After 15 minutes 10% HCI was added to pH = 3. After 0.5 hour the THF was removed in vacuo, the mixture was made basic with solid Na2CO3 and extracted with EtOAc (3 x 200 mL). The organics were combined, washed with 10% HCI (3x), the aqueous acidic layers combined, made basic with solid Na2CO3, extracted with EtOAc (3x), the organics combined, washed with brine, and dried (MgSO4). Filtration and concentration to dryness gave the title compound.
Step F: Preparation of 2-Fluoro-4-(3-methyl-3H-imidazole-4-carbonyl)- benzonitrile 2-Fluoro-4-[hydroxy-(3-methyl-3H-imidazol-4-yl)-methyl]- benzonitrile (0.655 g, 2.83 mmol) and Mnθ2 (1.23 g, 14.2 mmol) were stirred in CΗ2CI2 (50 mL) and CH3CN (5 mL) for 3 hours. The solution was filtered and concentrated to yield the title compound.
Step G: Preparation of N-[(4-cyano-3-fluoro-phenyl)-(3-methyl-3H-imidazol-
4-yl>methylene]-2-methylpropanesulfinamide
2-Fluoro-4-(3-methyl-3H-imidazole-4-carbonyl)-benzonitrile (2.56 g, 11.2 mmol), titanium(JN) ethoxide (7.02 mL, 33.5 mmol) and commercially available (R)-(+)-2-methyl-2-propanesulfinamide (1.35 g, 11.17 mmol) were dissolved in anhydrous TΗF (100 mL) and heated at 75°C for 7 days. The solution was cooled, diluted with brine (100 mL), filtered through a celite pad and washed generously with EtOAc and Η2O. The filtrate was separated, dried (MgSO4), and purified using Siθ2 chromatography (0-3% MeOH/CH2Cl2) to give the title compound.
Step H: Preparation of N-[l-(4-cyano-3-fluoro-phenyl)-l-(3-methyl-
3H-imidazol-4-yl. -ethyl! - 2-methylpropanesulf inamide
N-[(4-Cyano-3-fluoro-phenyl)-(3-methyl-3H-imidazol-4-yl)- methylene]-2-methylpropanesulfinamide (1.50 g, 4.51 mmol) was dissolved in anhydrous TΗF (30 mL) at 0°C and treated with a 3.0M solution of MeMgBr (4.50 mL, 13.5 mmol) in E-2O. After 15 minutes the reaction was quenched with aq. ΝΗ4CI solution, diluted with saturated NaHCO3 solution and extracted with CH2CI2 (3X). The combined organic layers were dried (MgSO4), filtered, concentrated to give the title compound.
Step I: Preparation of 4-[(lS)-amino-l-(3-methyl-3H-imidazol-4-yl)- ethyll -2-fluoro-benzoni trile
A cold methanolic HCI solution (50 mL) was added to N-[l-(4-cyano- 3-fluoro-phenyl)-l-(3-methyl-3H-in idazol-4-yl)-ethyl]- 2-methylpropanesulfinamide (0.880 g, 2.51 mmol) dissolved in MeOΗ (50 mL) and stirred for 1 hour at room temperature. After concentration, the crude product was chromatographed using a Chiralcel OD column eluting with 60-100% EtOΗ in hexane (0.1% DEA) to give the title compound as the major enantiomer.
Using the procedure described above, but substituting (S)-(-)-2- methyl-2-propanesulfinamide for (R)-(+)-2-methyl-2-propanesulfinamide in Step G, 4-[(lR)-amino-l-(3-methyl-3H-imidazol-4-yl)-ethyl]-2-fluoro-benzonitrile was obtained as the major enantiomer.
Step J: Preparation of l-(3-methoxy-benzenesulfonyl)-piperidine-3-carboxylic acid ethyl ester
A solution of ethyl pipecolate (1.02 g, 6.46 mmol) and Et3N (1.98 mL, 14.2 mmol) in CΗ2CI2 (13 mL) is treated with 3-methoxybenzenesulfonyl chloride (1.33 g, 6.46 mmol) dropwise with stirring under N2 at 0°C. After 3 hours the reaction was quenched with H2O, extracted with CH2CI2 (2x), the organics combined, washed with brine, dried (Na2SO4), filtered and concentrated to give the title compound as a yellow oil.
Step K: Preparation of l-(3-methoxy-benzenesulfonyl)-piperidine-3-carboxylic acid To a solution of l-(3-methoxy-benzenesulfonyl)-piperidine-3R- carboxylic acid ethyl ester (2.02 g, 6.16 mmol) in absolute ethanol (20 mL) at ambient temperature was added 50% w/w NaOH (0.26 mL, 6.47 mmol) and H2O
(1.25 mL). The reaction was stirred overnight, then treated with 12N HCI (0.53 mL) to a pH = 4. The ethanol was removed in vacuo and the residue partitioned between EtOAc and H2O. The aqueous layer was washed with EtOAc (2x), the organics combined, washed with brine, dried (Na2SO4), filtered, and concentrated in vacuo. Under the basic hydrolysis conditions partial racemization resulted in a 3: 1 mixture of R:S enantiomers, respectively. LC/MS (M+l) 300.
Step L: Preparation of 3-N-[l-(4-cyano-3-fluorophenyl)-l-(l-methyl-lH- imidazol-5-yl)ethyl]-l-[(3-methoxyphenyl)sulfonyl] piperidine-3- carboxamide
4-[(lR)-amino-l-(3-methyl-3H-imidazol-4-yl)-ethyl]-2-fluoro- benzonitrile (Step I) (0.192 g, 0.78 mmol), l-(3-methoxy-benzenesulfonyl)- piperidine-3-carboxylic acid, (0.235 g, 0.78 mmol), EDC (0.284 g, 1.5 mmol), ΗOBt (0.230 g, 1.5 mmol) and Ν-methylmorpholine (0.172 mL, 1.5 mmol) were stirred in DMF (4 mL) at ambient temperature overnight under Ν2. The reaction mixture was concentrated in vacuo, then partitioned between EtOAc and Η2O, the aqueous layer washed with EtOAc (2x), the organics combined, washed with brine, and dried (Na2SO4), filtered, and concentrated in vacuo to provide the title compound after normal phase Siθ2 chromatography on an ISCO combiflash eluting with 1-5% CH3OH:CH2Cl2 (0.1% NH4OH). LC/MS (M+l) 525.
Step M: Preparation of 3-N-[l-(4-cyano-3-fluorophenyl)-l-(l-methyl-lH- imidazol-5-yl)ethyl]-l-[(3-hydroxyphenyl)sulfonyl] piperidine-3- carboxamide
3-N-[l -(4-cyano-3-fluorophenyl)- 1-( 1 -methyl- lH-imidazol-5-yl) ethyl]- l-[(3-methoxyphenyl)sulf onyl] piperidine-3-carboxamide (0.265 g, 0.42 mmol) was dissolved in CΗ2CI2 (13 mL) at 0°C. and treated with IM BBr3 in CH2CI2 (8.42 mL, 8.42 mmol). After 1 hour aqueous saturated ΝaHCθ3 solution was added and, the layers were separated. The aqueous layer extracted with CH2CI2 (3x), and the organics combined, washed with brine, and dried (Na2SO4). Filtration and concentration to dryness gave crude product which was combined with a gummy residue which was insoluble in both phases and chromatographed on RPLC PrepPak eluting with 0.1%TFA-H2θ:0.1%TFA-CH3CN, 95:5 to 5:95 to give the title compound as the TFA salt after lyophilization. LC/MS (M+l) 511.
Step N: Preparation of 5-[(17i?)-10-cyano-14R-methyl-2,2-dioxido-16- oxo-8-oxa-2-thia-l,15-diazatetracyclo[15.3.1.13'7.l9'13]tricosa- 3(23),4,6,9(22),10,12-hexaen-14-yl]-l-methyl-lH-imidazol-l- ium trifluoroacetate
To a solution of 3-N-[l-(4-cyano-3-fluorophenyl)-l-(l-methyl-lH- imidazol-5-yl)ethyl]-l-[(3-hydroxyphenyl)sulfonyl] piperidine-3-carboxamide (0.243 g, 0.39 mmol) in DMF (10 mL) was added cesium carbonate (0.569 g, 1.75 mmol). The mixture was stirred at 60°C for 17 hours, taken up in water, concentrated to a small volume, and purified on a Waters Delta PrepPak ΗPLC column using a 0.1% trifluoroacetic acid water: 0.1% trifluoracetic acid/acetonitrile, 95:5 to 5:95 gradient over 1 hour. The pure fractions were pooled, concentrated to remove the acetonitrile and lyophilized overnight to yield 5-[(17R)-10-cyano-14R-methyl-2,2- dioxido-16-oxo-8-oxa-2-thia-l,15-diazatetracyclo[15.3.1.l3,7.l9,l3]tricosa-3(23),4,6, 9(22),10,12-hexaen-14-yl]-l-methyl-lH-imidazol-l-ium trifluoroacetate as the major diastereomer, FAB mass spectrum m/e 491 (M+l), and 5-[(17S)-10-cyano-14R- methyl-2,2-dioxido-16-oxo-8-oxa-2-thia-l,15-diazatetracyclo[15.3.1.13,7.19,13] tricosa-3(23),4,6, 9(22),10,12-hexaen-14-yl]-l-methyl-lH-imidazol-l-ium trifluoroacetate as the minor diastereomer, FAB mass spectrum m/e 491 (M+l).
These compounds, as free bases, are also named as (14/?, 17R or S)-14- methyl-14-(l -methyl- lH-imidazol-5-yl)-16-oxo-8-oxa-2-thia- 1,15- diazatetracyclo[15.3.1.13'7.l9'13] tricosa-3(23),4,6,9(22),10,12-hexaene-10-carbonitrile 2,2-dioxide.
EXAMPLE 2
Preparation of 5-[(18S)-l 0-cyano- 14-methyl-2,2-dioxido- 16-oxo-8-oxa-2-thia- 1,15- diazatetracyclo[16.3.1.l3,7.i9,13]tetracosa-3(24),4,6,9(23),10,12-hexaen-14-yl]-l- meth yl- 1 Η-imidazol- 1 -ium trifluoroacetate Using the procedures described in Example 1, but substituting l-(3-methoxy-benzenesulfonyl)-piperidine-3-acetic acid for l-(3-methoxy- benzenesulfonyl)-piperidine-3-carboxylic acid in Step L, the title compound was prepared. LC/MS M+l 506.
This compound, as free bases, is also named as (18S) -14-methyl- 14-(l-methyl-lΗ-imidazol-5-yl)-16-oxo-8-oxa-2-thia-l,15-diazatetracyclo
[16.3.1.13'7. l9,13jtetracosa-3(24),4,6,9(23), 10, 12-hexaene-10-carbonitrile 2,2-dioxide trifluoroacetate salt.
EXAMPLE 3
Preparation of 5- [( 19S)- 10-cyano- 14-methyl-2,2-dioxido- 16-oxo-8-oxa-2-thia- 1,15- diazatetracyclo[17.3.1.l3,7.l9,13]pentacosa-3(25),4,6,9(24),10,12-hexaen-14-yl]- 1 -methyl- lH-imidazol-1 -ium trifluoroacetate and 5-[(19R)-10-cyano-14-methyl- 2,2-dioxido-16-oxo-8-oxa-2-thia-l,15-diazatetracyclo[17.3.1.13.7. l9,13]pentacosa- 3(25),4,6.9r24).10.12-hexaen-14-vn-l-methyl-lH-imidazol-l-ium trifluoroacetate Using the procedures described in Example 1, but substituting l-(3-methoxy-benzenesulfonyl)-piperidine-3-propionic acid for l-(3-methoxy- benzenesulfonyl)-piperidine-3-carboxylic acid in Step L, the title compounds were prepared. LC/MS M+l 520. These compounds, as free bases, are also named as (19S)-14- methyl- 14-(1 -methyl- lH-imidazol-5-yl)- 16-oxo-8-oxa-2-thia- 1 , 15-diazatetracyclo [17.3.1. l3,7.l9'13]pentacosa-3(25),4,6,9(24),10,12-hexaene-10-carbonitrile 2,2-dioxide and (192?)- 14-methyl- 14-(1 -methyl- lH-imidazol-5-yl)- 16-oxo-8-oxa-2-thia- 1,15- diazatetracyclo[17.3.1.13,7.l9,13]ρentacosa-3(25),4,6,9(24),10,12-hexaene-10- carbonitrile 2,2-dioxide.
EXAMPLE 4
Preparation of (6S)-1 lS-amino-1 l-(l-methyl-lH-imidazol-5-yl)-17-oxa-2-thia-3,7- diazatetracyclo[16.3.1.l3,6.ll2,16]tetracosa-l(22),12(23),13,15,18,20-hexaene-15- carbonitrile 2.2-dioxide
Step A: Preparation of Benzyl 3-bromophenyl ether
To a stirred solution of 3-bromophenol (9.50 g, 54.9 mmol) in degassed DMF (150 mL) at 0°C, under argon, was added CS2CO3 (35.8 g, 109 mmol). The resulting mixture was stirred for 1 hour, then benzyl bromide (10.3 g, 60.2 mmol) was added and stirring was continued for 2 hours at 0°C. The solvent was removed under reduced pressure, and the residue was partitioned between 20% aqueous NaOH (250 mL) and CHCI3 (500 mL). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica, eluting first with two column volumes of hexane, then with hexane - 2% EtOAc to yield the desired product as a white solid.
Step B: Preparation of Magnesium 3-benzyloxyphenylsulfinate bromide To a stirred suspension of Rieke Mg (0.5 g, 21 mmol) in refluxing dry THF (45 mL), under argon, was added benzyl 3-bromophenyl ether, as described above in Step A, (5.0 g, 19 mmol) in dry THF (50 mL) dropwise, at a rate that maintained reflux with the heat source removed. The resulting mixture was heated to reflux for 30 min, then cooled to -78°C and SO2 was bubbled into the solution slowly for 1 hr. The reaction mixture was allowed to warm slowly to ambient temperature and stirred for 18 hours. Argon was bubbled into the mixture for 1 hour, then the solution was concentrated in vacuo to give the desired product as a solid.
Step C: Preparation of 3-Benzyloxyphenylsulfonyl chloride Magnesium 3-benzyloxyphenylsulfinate bromide, as described above in Step B, (4.5 g, 14 mmol) was dissolved in dry CH2CI2 (175 mL) and cooled to 0°C under argon. Sulfuryl chloride (19 mL of a 1.0 M solution in CH2CI2, 19 mmol) was added dropwise, and the mixture was stirred for 18 hrs at ambient temperature, during which time a precipitate formed. The precipitate was removed by filtration and the filtrate was concentrated under reduced pressure to give the product as a yellow oil.
Step D: Preparation of (R)- 1 -(3-Benzyloxyphenylsulf onyl)-3-(tert- butoxycarbonylamino')pyrrolidine
To a stirred solution of 3-benzyloxyphenylsulfonyl chloride, as described above in Step C, (1.8 g, 7.2 mmol) in CH2CI2 (50 mL) was added NJV- diisopropylethylamine (1.8 mL, 10.3 mmol) followed by (R)-3-(tβrt-butoxycarbonyl- amino)pyrrolidine (1.77 g, 9.5 mmol) and the reaction mixture was stirred at ambient temperature for 4 hours. The mixture was washed with 10% aqueous citric acid (75 mL), then water (50 mL), then saturated aqueous NaHCO3 (50 mL). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica, eluting with hexane - 20% EtOAc to yield the desired product as a white solid.
Step E: Preparation of (R)-3-(tert-Butoxycarbonylamino)-l-(3- hydroxyphenylsulfonyPpyrrolidine
To a solution of (7?)-l-(3-benzyloxyphenylsulfonyl)-3-(tert-butoxy- carbonylamino)pyrrolidine, as described above in Step D, (3.1 g, 7.17 mmol) in EtOH (150 mL) was added 20% Pd(OH)2 on carbon (900 mg) and the reaction mixture was stirred under an atmosphere of hydrogen (ca. 1 atm) at ambient temperature for 18 hours. The mixture was filtered through a pad of celite, washing with EtOH, and the filtrate was concentrated in vacuo to give the titled product as a white solid.
Step F: Preparation of (R)-3-Amino-l-(3-hydroxyphenyl-sulfonyl)pyrrolidine
Hydrochloride
A solution of (R)-3-(tert-butoxycarbonylamino)-l-(3-hydroxyphenyl- sulfonyl)pyrrolidine, as described above in Step E, (2.4 g, 7.0 mmol) in EtOAc (100 mL) at 0°C was saturated with HCI (g). After 15 minutes, the mixture was concentrated in vacuo to yield the amine hydrochloride as a white solid. Using the methods outlined above, but substituting (S)-3-(tert- butoxycarbonylamino)pyrrolidine for (i?)-3-(tert-butoxycarbonyl-amino)pyrrolidine in Step D, (S)-3-Amino-l-(3-hydroxyphenylsulfonyl)pyrrolidine hydrochloride was prepared.
Step G: Preparation of N-[4-{ [tert-butyl(dimethyl)silyl]oxy }-l-(4-cyano-3- fluorophenyl)- 1 -( 1 -methyl- lH-imidazol-5-yl)butyl] -2-methylpropane-
2-sulfinamide
To a flame-dried flask containing magnesium (0.317 g, 12.03 mmol) under N2 was added dry THF (3 mL), a pinch of iodine, and Rieke magnesium (1 mL of a IM THF solution). 3-tert-Butyl(dimethyl)silyloxypropylbromide (3.048 g, 12.03 mmol) in THF (5 mL) was added in a small portion to the magnesium suspension with gentle warming to initiate the reaction. Once the reaction had started, the rest of the bromide solution was added dropwise over 15 minutes. After stirring for 15 minutes, this solution was added to a solution of N-[(4-cyano-3-fluoro-phenyl)-(3- methyl-3H-imidazol-4-yl)-methylene]-2-methylpropanesulfinamide (Example 1, Step G) (1.00 g, 3.01 mmol) in dry TΗF (12 mL) with stirring in an ice-Η2θ bath under
Ν2. Grignard solution was added until HPLC indicated loss of starting i ine. The reaction mixture was quenched with H2O and partitioned between aqueous saturated NaHCO3 solution and CH2CI2. The organic layer was separated, the aqueous layer washed with CH2CI2 (2x), the organics combined, washed with brine, dried (MgSO4), filtered and concentrated to give the title compound after purification on an ISCO combiflash column eluting with 1-3% CH3OH/CH2CI2 (NH4OH).
Step H: Preparation of N-[l-(4-cyano-3-fluorophenyl)-4-hydroxy-l(S and R)-
('l-methyl-lH-imidazol-5-yl)butyl1-2-methylpropane-2-sulfinamide N- [4- { [tert-Butyl(dimethyl)silyl] oxy } - 1 -(4-cyano-3 -fluorophenyl)- l-(l-methyl-lH-imidazol-5-yl)butyl]-2-methylpropane-2-sulfinamide (0.111 g, 0.219 mmol) was dissolved in CH3CN (80 mL): H2O (20 mL) at ambient temperature and treated with CF3CO2H (1 mL) with stirring. After 0.5 hour the mixture was neutralized with aqueous saturated NaHCO3 solution and concentrated to remove the CH3CN. The crude product was chromatographed on a Chiralcel AD HPLC column eluting with 90/10 hexane/ethanol to give both enantiomers: N-[l-(4-cyano-3- fluorophenyl)-4-hydroxy-l(S)-(l-methyl-lH-imidazol-5-yl)butyl]-2-methylpropane- 2-sulfinamide MS (M+l) 393; and N-[l-(4-cyano-3-fluorophenyl)-4-hydroxy-l(R)- (l-methyl-lH-imidazol-5-yl)butyl]-2-methylproρane-2-sulfinamide MS (M+l) 393.
Step I: Preparation of N-[l-(4-cyano-3-fluorophenyl)-l-(l-methyl-lH- imidazol-5-ylV4-oxobutyll-2-methylpropane-2-sulfinamide
Pyridine-SO3 complex (0.174 g, 1.10 mmol) in DMSO (2 mL) was added to a solution of N-[l-(4-cyano-3-fluorophenyl)-4-hydroxy-l(S)-(l-methyl- lH-imidazol-5-yl)butyl]-2-methylρropane-2-sulfinamide (0.108 g, 0.275 mmol) and Et3N (0.222 g, 2.2 mmol) in CH2CI2 (5 mL) with cooling in an ice-H2θ bath. The reaction mixture was allowed to warm to ambient temperature, stirred overnight, then partitioned between aqueous saturated NaHCO3 solution and CH2CI2. The aqueous layer was washed with CH2CI2 (3x), organics combined, washed with brine, dried
(MgSO ), filtered and concentrated to dryness to give the title compound after purification on an ISCO combiflash eluting with 1-5% CH3OH CH2CI2. LC/MS = 391.
Step J: Preparation of N-[l-(4-cyano-3-fluorophenyl)-4-({ (3S)-l-[(3- hydroxyphenyl)sulfonyl]pyrrolidin-3-yl } amino)- 1 -( 1 -methyl- 1H- imidazol-5-yl butyll-2-methylpropane-2-sulfinamide N-[l-(4-Cyano-3-fluorophenyl)-l-(l-methyl-lH-imidazol-5-yl)-4- oxobutyl]-2-methylpropane-2-sulfinamide (0.028 g, 0.0717 mmol) and (S)-3-Amino- l-(3-hydroxyphenylsulfonyl) pyrrolidine hydrochloride (0.021 g, 0.0753 mmol) were dissolved in CH3OH (5 mL) with stirring at ambient temperature, the pH of the solution adjusted to 5 with acetic acid, and the solution stirred for 0.5 hour. Sodium cyanoborohydride (0.0068 g, 0.1076 mmol) was added, and the mixture stirred for 16 hours. The mixture was concentrated in vacuo and partitioned between aqueous saturated N-1HCO3 solution and CH2CI2. The aqueous layer was washed with CH2CI2 (3x), organics combined, washed with brine, dried (MgSO4), filtered and concentrated to dryness to give the title compound.
Step K: Preparation of N-[(6S)-15-cyano-l 1-(1 -methyl- lH-imidazol-5-yl)-
2,2-dioxido-17-oxa-2-thia-3,7-diazatetracyclo [16.3.1.13,6.112,16] tetracosa- 1 (22), 12(23), 13 , 15, 18,20-hexaen- 11 -yl]-2-methylpropane- 2-sulfinamide A solution of N-[l-(4-cyano-3-fluorophenyl)-4-({(3S)-l-[(3- hydroxyphenyl)sulfonyl]pyιτolidin-3-yl}amino)-l-(l-methyl-lH-imidazol-5- yl)butyl]-2-methylproρane-2-sulfinamide (0.030 g, 0.0486 mmol) in DMF (5 mL) was treated with CS2CO3 (0.0475 g, 0.1458 mmol) and heated overnight at 80°C. The reaction mixture was concentrated in vacuo, partitioned between aqueous saturated NaHCO3 solution and CH2CI2. The aqueous layer was washed with CH2CI2 (2x), organics combined, washed with brine, dried (MgSO ), filtered and concentrated to dryness to give the title compound.
Step L: Preparation of (6S)-ll-amino-ll-(l-methyl-lH-imidazol-5-yl)-
17-oxa-2-thia-3,7-diazatetracyclo [16.3.1. l3.6.ll2,l6]tetracosa- l(22..12(23).13.15.18.20-hexaene-15-carbonitrile 2.2-dioxide N-[(6S)-15-cyano-ll-(l-methyl-lH-imidazol-5-yl)-2,2-dioxido-17- oxa-2-thia-3,7-diazatetracyclo [16.3.1.13,6.il2,16]tetracosa-l(22),12(23),13,15,18,20- hexaen-1 l-yl]-2-methylpropane-2-sulfinamide (0.0486 mmol) was dissolved in
CH3OH (5 mL) - 4M HCI in dioxane (4 mL), stirred for 15 min at ambient temperature, concentrated in vacuo, then chromatographed on RPLC PrepPak eluting with 95:5 to 5:95 H2O:CH3CN with 0.1% TFA over 60 minutes. Like fractions were combined, concentrated, and lyophilized to give the title compound. MS (M+l) = 493.
Anal, calcd for C25H28N6O3S . 1.00 H2O • 3.2 CF3CO2H:
C, 43.08; H, 3.82; N, 9.60; Found: C, 42.76; H, 4.07; N, 9.65. «
Using (R)-3-Amino-l-(3-hydroxyphenylsulfonyl) pyrrolidine hydrochloride in place of the S-enantiomer in Step J, (6R)-llR-amino-ll-(l-methyl- lH-imidazol-5-yl)-17-oxa-2-thia-3,7-diazatetracyclo [16.3.1. l3,6.l12,16]tetracosa- l(22),12(23),13,15,18,20-hexaene-15-carbonitrile 2,2-dioxide was prepared in a similar manner. MS (M+l) 493.
EXAMPLE 5
In vitro inhibition of ras farnesyl transferase Transferase Assays. Isoprenyl-protein transferase activity assays are carried out at 30°C unless noted otherwise. A typical reaction contains (in a final volume of 50 μL): [3H]farnesyl diphosphate, Ras protein, 50 mM HEPES, pH 7.5, 5 mM MgCl2, 5 mM dithiothreitol, 10 μM ZnCl2, 0.1% polyethyleneglycol (PEG) (15,000-20,000 mw) and isoprenyl-protein transferase. The FPTase employed in the assay is prepared by recombinant expression as described in Omer, C.A., Krai, A.M., Diehl, R.E., Prendergast, G.C., Powers, S., Allen, CM., Gibbs, J.B. and Kohl, N.E. (1993) Biochemistry 32:5167-5176. After thermally pre-equilibrating the assay mixture in the absence of enzyme, reactions are initiated by the addition of isoprenyl- protein transferase and stopped at timed intervals (typically 15 min) by the addition of 1 M HCI in ethanol (1 mL). The quenched reactions are allowed to stand for 15 minutes (to complete the precipitation process). After adding 2 mL of 100% ethanol, the reactions are vacuum-filtered through Whatman GF/C filters. Filters are washed four times with 2 mL aliquots of 100% ethanol, mixed with scintillation fluid (10 mL) and then counted in a Beckman LS3801 scintillation counter.
For inhibition studies, assays are run as described above, except inhibitors are prepared as concentrated solutions in 100% dimethyl sulfoxide and then diluted 20-fold into the enzyme assay mixture. Substrate concentrations for inhibitor IC50 determinations are as follows: FTase, 650 nM Ras-CNLS (SEQ.ID.ΝO.: 2), 100 nM farnesyl diphosphate.
The compounds of the instant invention are tested for inhibitory activity against human FPTase by the assay described above.
The compounds of the instant case were tested by the assay above and were found to have an IC50 of <lμM.
EXAMPLE 6
Modified In vitro GGTase inhibition assay
The modified geranylgeranyl-protein transferase inhibition assay is carried out at room temperature. A typical reaction contains (in a final volume of
50 μL): [^Hjgeranylgeranyl diphosphate, biotinylated Ras peptide, 50 mM HEPES, pH 7.5, a modulating anion (for example 10 mM glycerophosphate or 5mM ATP), 5 mM MgCl2, 10 μM ZnCl2, 0.1% PEG (15,000-20,000 mw), 2 mM dithiothreitol, and geranylgeranyl-protein transferase type I(GGTase). The GGTase-type I enzyme employed in the assay is prepared as described in U.S. Patent No. 5,470,832, incorporated by reference. The Ras peptide is derived from the K4B-Ras protein and has the following sequence: biotinyl-G-O-XKKKSKTKCNIM (single amino acid code) (SEQ.-D.ΝO.: 3). Reactions are initiated by the addition of GGTase and stopped at timed intervals (typically 15 min) by the addition of 200 μL of a 3 mg mL suspension of streptavidin SPA beads (Scintillation Proximity Assay beads, Amersham) in 0.2 M sodium phosphate, pH 4, containing 50 mM EDTA, and 0.5% BSA. The quenched reactions are allowed to stand for 2 hours before analysis on a Packard TopCount scintillation counter. For inhibition studies, assays are run as described above, except inhibitors are prepared as concentrated solutions in 100% dimethyl sulfoxide and then diluted 25 fold into the enzyme assay mixture. IC50 values are determined with Ras peptide near KM concentrations. Enzyme and substrate concentrations for inhibitor IC50 determinations are as follows: 75 pM GGTase-I, 1.6 μM Ras peptide, 100 nM geranylgeranyl diphosphate.
The compounds of the instant invention are tested for inhibitory activity against human GGTase-type I by the assay described above.
EXAMPLE 7
Cell-based in vitro ras farnesylation assay
The cell line used in this assay is a v-ras line derived from either Ratl or ΝIH3T3 cells, which expressed viral Ha-ras p21. The assay is performed essentially as described in DeClue, J.E. et al., Cancer Research 51:712-717, (1991). Cells in 10 cm dishes at 50-75% confluency are treated with the test compound (final concentration of solvent, methanol or dimethyl sulfoxide, is 0.1%). After 4 hours at 37°C, the cells are labeled in 3 ml methionine-free DMEM supple-mented with 10% regular DMEM, 2% fetal bovine serum and 400 μCi[35s]methionine (1000 Ci/mmol). After an additional 20 hours, the cells are lysed in 1 ml lysis buffer (1% NP40/20 mM HEPES, pH 7.5/5 mM MgCl2/lmM DTT/10 mg/ml aprotinen/2 mg/ml leupeptin/2 mg/ml antipain/0.5 mM PMSF) and the lysates cleared by centrifugation at 100,000 x g for 45 minutes. Aliquots of lysates containing equal numbers of acid- precipitable counts are bought to 1 ml with IP buffer (lysis buffer lacking DTT) and immuno-precipitated with the ras-specific monoclonal antibody Y13-259 (Furth, M.E. et al., J. Virol. 43:294-304, (1982)). Following a 2 hour antibody incubation at 4°C, 200 μl of a 25% suspension of protein A-Sepharose coated with rabbit anti rat IgG is added for 45 minutes. The immuno-precipitates are washed four times with IP buffer (20 nM HEPES, pH 7.5/1 mM EDTA/1% Triton X-100.0.5% deoxycholate/ 0.1%/SDS/O.l M NaCl) boiled in SDS-PAGE sample buffer and loaded on 13% acrylamide gels. When the dye front reached the bottom, the gel is fixed, soaked in Enlightening, dried and autoradiographed. The intensities of the bands corresponding to farnesylated and nonfarnesylated ras proteins are compared to determine the percent inhibition of farnesyl transfer to protein.
EXAMPLE 8
Cell-based in vitro growth inhibition assay
To determine the biological consequences of FPTase inhibition, the effect of the compounds of the instant invention on the anchorage-independent growth of Ratl cells transformed with either a v-ras, v-raf, or v-mos oncogene is tested. Cells transformed by v-Raf and v-Mos maybe included in the analysis to evaluate the specificity of instant compounds for Ras-induced cell transformation. Rat 1 cells transformed with either v-ras, v-raf, or v-mos are seeded at a density of 1 x 10^ cells per plate (35 mm in diameter) in a 0.3% top agarose layer in medium A (Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum) over a bottom agarose layer (0.6%). Both layers contain 0.1% methanol or an appropriate concentration of the instant compound (dissolved in methanol at 1000 times the final concentration used in the assay). The cells are fed twice weekly with 0.5 ml of medium A containing 0.1% methanol or the concentra- tion of the instant compound. Photomicrographs are taken 16 days after the cultures are seeded and comparisons are made.
EXAMPLE 9
Construction of SEAP reporter plasmid pDSElOO
The SEAP reporter plasmid, pDSElOO was constructed by ligating a restriction fragment containing the SEAP coding sequence into the plasmid pCMV- RE-AKI. The SEAP gene is derived from the plasmid pSEAP2-Basic (Clontech, Palo Alto, CA). The plasmid pCMN-RE-AKI was constructed by Deborah Jones (Merck) and contains 5 sequential copies of the 'dyad symmetry response element' cloned upstream of a 'CAT-TATA' sequence derived from the cytomegalovirus immediate early promoter. The plasmid also contains a bovine growth hormone poly-A sequence. The plasmid, pDSElOO was constructed as follows. A restriction fragment encoding the SEAP coding sequence was cut out of the plasmid pSEAP2- Basic using the restriction enzymes EcoRl and Hpal. The ends of the linear DNA fragments were filled in with the Klenow fragment of E. coli DNA Polymerase I. The 'blunt ended' DNA containing the SEAP gene was isolated by electrophoresing the digest in an agarose gel and cutting out the 1694 base pair fragment. The vector plasmid pCMV-RE-AKI was linearized with the restriction enzyme Bgl-II and the ends filled in with Klenow DNA Polymerase I. The SEAP DNA fragment was blunt end ligated into the pCMN-RE-AKI vector and the ligation products were transformed into DH5-alpha E. coli cells (Gibco-BRL). Transformants were screened for the proper insert and then mapped for restriction fragment orientation. Properly oriented recombinant constructs were sequenced across the cloning junctions to verify the correct sequence. The resulting plasmid contains the SEAP coding sequence downstream of the DSE and CAT-TATA promoter elements and upstream of the BGH poly-A sequence.
Alternative Construction of SEAP reporter plasmid, pDSElOl
The SEAP repotrer plasmid, pDSElOl is also constructed by ligating a restriction fragment containing the SEAP coding sequence into the plasmid pCMN- RE-AKI. The SEAP gene is derived from plasmid pGEM7zf (-)/SEAP. The plasmid pDSElOl was constructed as follows: A restriction fragment containing part of the SEAP gene coding sequence was cut out of the plasmid pGEM7zf(-)/SEAP using the restriction enzymes Apa I and Kpnl. The ends of the linear DΝA fragments were chewed back with the Klenow fragment of E. coli DΝA Polymerase I. The "blunt ended" DΝA containing the truncated SEAP gene was isolated by electrophoresing the digest in an agarose gel and cutting out the 1910 base pair fragment. This 1910 base pair fragment was ligated into the plasmid pCMN-RE-AKI which had been cut with Bgl-II and filled in with E. coli Klenow fragment DΝA polymerase. Recombinant plasmids were screened for insert orientation and sequenced through the ligated junctions. The plasmid pCMN-RE-AKI is derived from plasmid pCMNIE-AKI-DHFR (Whang , Y., Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E. (1987) J. Virol., 61, 1796- 1807) by removing an EcoRI fragment containing the DHFR and Neomycin markers. Five copies of the fos promoter serum response element were inserted as described previously (Jones, R.E., Defeo-Jones, D., McAvoy, E.M., Nuocolo, G.A., Wegrzyn, R.J., Haskell, K.M. and Oliff, A. (1991) Oncogene, 6, 745-751) to create plasmid pCMV-RE-AKI.
The plasmid pGEM7zf(-)/SEAP was constructed as follows. The SEAP gene was PCRed, in two segments from a human placenta cDΝA library (Clontech) using the following oligos.
Sense strand Ν-terminal SEAP : 5' GAGAGGGAATTCGGGCCCTTCCTGCAT GCTGCTGCTGCTGCTGCTGCTGGGC 3' (SEQ.ID.ΝO. :4)
Antisense strand Ν-terminal SEAP: 5' GAGAGAGCTCGAGGTTAACCCGGGT GCGCGGCGTCGGTGGT 3' (SEQ.ID.ΝO.:5)
Sense strand C-terminal SEAP: 5' GAGAGAGTCTAGAGTTAACCCGTGGTCC CCGCGTTGCTTCCT 3' (SEQ.ID.NO.: 6)
Antisense strand C-terminal SEAP: 5' GAAGAGGAAGCTTGGTACCGCCACTG GGCTGTAGGTGGTGGCT 3' (SEQ.ID.NO.:7)
The N-terminal oligos (SEQ.ID.NO.: 4 and SEQ.ID.NO.: 5) were used to generate a 1560 bp N-terminal PCR product that contained EcoRI and Hpal restriction sites at the ends. The Antisense N-terminal oligo (SEQ.ID.NO.: 5) introduces an internal translation STOP codon within the SEAP gene along with the Hpal site. The C-terminal oligos (SEQ.ID.NO.: 6 and SEQ.ID.NO.: 7) were used to amplify a 412 bp C-terminal PCR product containing Hpal and Hind-XT restriction sites. The sense strand C-terminal oligo (SEQ.ID.NO.: 6) introduces the internal STOP codon as well as the Hpal site. Next, the N-terminal amplicon was digested with EcoRI and Hpal while the C-terminal amplicon was digested with Hpal and Hindlll. The two fragments comprising each end of the SEAP gene were isolated by electro-phoresing the digest in an agarose gel and isolating the 1560 and 412 base pair fragments. These two fragments were then co-ligated into the vector pGEM7zf (-) (Promega) which had been restriction digested with EcoRI and Hind-XT and isolated on an agarose gel. The resulting clone, pGEM7zf(-)/SEAP contains the coding sequence for the SEAP gene from amino acids.
Construction of a constitutively expressing SEAP plasmid pCMV-SEAP-A
An expression plasmid constitutively expressing the SEAP protein was created by placing the sequence encoding a truncated SEAP gene downstream of the cytomegalovirus (CMV) IE-1 promoter. The expression plasmid also includes the CMV intron A region 5' to the SEAP gene as well as the 3' untranslated region of the bovine growth hormone gene 3' to the SEAP gene.
The plasmid pCMVIE-AKI-DHFR (Whang , Y., Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E. (1987) J. Virol., 61:1796-1807) containing the CMV immediate early promoter was cut with EcoRI generating two fragments. The vector fragment was isolated by agarose electro- phoresis and reli gated. The resulting plasmid is named pCMV-AKI. Next, the cytomegalovirus intron A nucleotide sequence was inserted downstream of the CMV IEl promter in pCMV-AKI. The intron A sequence was isolated from a genomic clone bank and subcloned into pBR322 to generate plasmid pl6T-286. The intron A sequence was mutated at nucleotide 1856 (nucleotide numbering as in Chapman, B.S., Thayer, R.M., Vincent, K.A. and Haigwood, N.L., Nuc.Acids
Res. 19, 3979-3986) to remove a Sad restriction site using site directed mutagenesis. The mutated intron A sequence was PCRed from the plasmid pl6T-287 using the following oligos.
Sense strand: 5' GGCAGAGCTCGTTTAGTGAACCGTCAG 3' (SEQ.ID.NO.: 8)
Antisense strand: 5' GAGAGATCTCAAGGACGGTGACTGCAG 3' (SEQ.ID.NO.: 9)
These two oligos generate a 991 base pair fragment with a Sad site incorporated by the sense oligo and a Bgl-II fragment incorporated by the antisense oligo. The PCR fragment is trimmed with Sad and Bgl-II and isolated on an agarose gel. The vector pCMV-AKI is cut with Sad and Bgl-II and the larger vector fragment isolated by agarose gel electrophoresis. The two gel isolated fragments are ligated at their respective Sad and Bgl-II sites to create plasmid pCMV-AKI-InA. The DNA sequence encoding the truncated SEAP gene is inserted into the pCMV-AKI-InA plasmid at the Bgl-II site of the vector. The SEAP gene is cut out of plasmid ρGEM7zf(-)/SEAP (described above) using EcoRI and HindllL The fragment is filled in with Klenow DNA polymerase and the 1970 base pair fragment isolated from the vector fragment by agarose gel electrophoresis. The pCMV-AKI- InA vector is prepared by digesting with Bgl-II and filling in the ends with Klenow DNA polymerase. The final construct is generated by blunt end ligating the SEAP fragment into the pCMV-AKI-InA vector. Transformants were screened for the proper insert and then mapped for restriction fragment orientation. Properly oriented recombinant constructs were sequenced across the cloning junctions to verify the correct sequence. The resulting plasmid, named pCMV-SEAP-A (deposited in the ATCC under Budapest Treaty on August 27, 1998, and designated ATCC), contains a modified SEAP sequence downstream of the cytomegalovirus immediately early promoter IE-1 and intron A sequence and upstream of the bovine growth hormone poly-A sequence. The plasmid expresses SEAP in a constitutive manner when transfected into mammalian cells.
Alternative construction of a constitutively expressing SEAP plasmid pCMV-SEAP-B An expression plasmid constitutively expressing the SEAP protein can be created by placing the sequence encoding a truncated SEAP gene downstream of the cytomegalovirus (CMV) IE-1 promoter and upstream of the 3' unstranslated region of the bovine growth hormone gene.
The plasmid pCMVIE-AKI-DHFR (Whang , Y., Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E. (1987) J. Virol., 61:1796-1807) containing the CMV immediate early promoter and bovine growth hormone poly-A sequence can be cut with EcoRI generating two fragments. The vector fragment can be isolated by agarose electrophoresis and religated. The resulting plasmid is named pCMV-AKI. The DNA sequence encoding the truncated SEAP gene can be inserted into the pCMV-AKI plasmid at a unique Bgl-II in the vector. The SEAP gene is cut out of plasmid pGEMzf(-)/SEAP (described above) using EcoRI and Hindlll. The fragments are filled in with Klenow DNA polymerase and the 1970 base pair fragment is isolated from the vector fragment by agarose gel electrophoresis. The pCMV-AKI vector is prepared by digesting with Bgl-II and filling in the ends with Klenow DNA polymerase. The final construct is generated by blunt end ligating the SEAP fragment into the vector and transforming the ligation reaction into E. coli DH5 cells. Transformants can then be screened for the proper insert and mapped for restriction fragment orientation. Properly oriented recombinant constructs would be sequenced across the cloning junctions to verify the correct sequence. The resulting plasmid, named pCMN-SEAP-B contains a modified SEAP sequence downstream of the cytomegalovirus immediate early promoter, IE1, and upstream of a bovine growth hormone poly-A sequence. The plasmid would express SEAP in a constitutive nammer when transfected into mammalian cells.
Cloning of a Myristylated viral-H-ras expression plasmid pSMS600
A DΝA fragment containing viral-H-ras can be PCRed from plasmid "HB-11 (deposited in the ATCC under Budapest Treaty on August 27, 1997, and designated ATCC 209,218) using the following oligos.
Sense strand:
5'TCTCCTCGAGGCCACCATGGGGAGTAGCAAGAGCAAGCCTAAGGACCC CAGCCAGCGCCGGATGACAGAATACAAGCTTGTGGTGG3'. (SEQ.ID.ΝO.: 10)
Antisense:
5'CACATCTAGATCAGGACAGCACAGACTTGCAGC 3'. (SEQ.ID.ΝO.: 11)
A sequence encoding the first 15 aminoacids of the v-src gene, containing a myristylation site, is incorporated into the sense strand oligo. The sense strand oligo also optimizes the 'Kozak' translation initiation sequence immediately 5' to the ATG start site. To prevent prenylation at the viral-r -? C-terminus, cysteine 186 would be mutated to a serine by substituting a G residue for a C residue in the C-terminal antisense oligo. The PCR primer oligos introduce an Xhol site at the 5' end and a Xbal site at the 3 'end. The Xhol-Xbal fragment can be ligated into the mammalian expression plasmid pCI (Promega) cut with Xhol and Xbal. This results in a plasmid, pSMS600, in which the recombinant myr-viral-H-ras gene is constitutively transcribed from the CMV promoter of the pCI vector.
Cloning of a viral-H-ras-CNLL expression plasmid pSMSόOl A viral-H-r-w clone with a C-terminal sequence encoding the amino acids CVLL can be cloned from the plasmid "HB-11" by PCR using the following oligos.
Sense strand:
5'TCTCCTCGAGGCCACCATGACAGAATACAAGCTTGTGGTGG-3' (SEQ.ID.NO.: 12)
Antisense strand: 5'CACTCTAGACTGGTGTCAGAGCAGCACACACTTGCAGC-3' (SEQ.ID.NO.:
13)
The sense strand oligo optimizes the 'Kozak' sequence and adds an Xhol site. The antisense strand mutates serine 189 to leucine and adds an Xbal site. The PCR fragment can be trimmed with Xhol and Xbal and ligated into the Xhol- Xbal cut vector pCI (Promega). This results in a plasmid, pSMS601, in which the mutated viral-H-rαs-CVLL gene is constitutively transcribed from the CMV promoter of the pCI vector.
Cloning of cellular-H-ras-Leuό! expression plasmid pSMS620
The human cellular-H-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligonucleotide primers.
Sense strand: 5'-GAGAGAATTCGCCACCATGACGGAATATAAGCTGGTGG-3' (SEQ.ID.NO.: 14)
Antisense strand:
5'-GAGAGTCGACGCGTCAGGAGAGCACACACTTGC-3' (SEQ.ID.NO.: 15)
The primers will amplify a c-H-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, an EcoRI site at the N-terminus and a Sal I site at the C-terminal end. After trimming the ends of the PCR product with EcoRI and Sal I, the c-H-ras fragment can be ligated ligated into an EcoRI -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of glutamine-61 to a leucine can be accomplished using the manufacturer's protocols and the following oligonucleotide:
5'-CCGCCGGCCTGGAGGAGTACAG-3' (SEQ.ID.NO.: 16)
After selection and sequencing for the correct nucleotide substitution, the mutated c-H-ras-Leuόl can be excised from the pAlter-1 vector, using EcoRI and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with EcoRI and Sal I. The new recombinant plasmid, pSMS620, will constitutively transcribe c-H-ras-Leuόl from the CMV promoter of the pCI vector.
Cloning of a c-N-ra.s-Nal-12 expression plasmid pSMS630
The human c-N-ras gene can be PCRed from a human cerebral cortex cDΝA library (Clontech) using the following oligonucleotide primers.
Sense strand:
5'-GAGAGAATTCGCCACCATGACTGAGTACAAACTGGTGG-3' (SEQ.ID.ΝO.: 17)
Antisense strand:
5'-GAGAGTCGACTTGTTACATCACCACACATGGC-3' (SEQ.ID.NO.: 18)
The primers will amplify a c-N-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, an EcoRI site at the N-terminus and a Sal I site at the C-terminal end. After trimming the ends of the PCR product with EcoRI and Sal I, the c-N-ras fragment can be ligated into an EcoRI -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of glycine-12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide:
5'-GTTGGAGCAGTTGGTGTTGGG-3' (SEQ.ID.NO.: 19)
After selection and sequencing for the correct nucleotide substitution, the mutated c-N-rø.s-Nal-12 can be excised from the pAlter-1 vector, using EcoRI and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with EcoRI and Sal I. The new recombinant plasmid, pSMS630, will constitutively transcribe c-N-r--_>-Val-12 from the CMV promoter of the pCI vector.
Cloning of a c-K4B-ra,f-Val-12 expression plasmid pSMS640 The human c-K4B-rø.s gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligo-nucleotide primers.
Sense strand:
5'-GAGAGGTACCGCCACCATGACTGAATATAAACTTGTGG-3' (SEQ.ID.NO.: 20)
Antisense strand:
5'-CTCTGTCGACGTATTTACATAATTACACACTTTGTC-3' (SEQ.ID.NO.: 21)
The primers will amplify a c-K4B-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, a Kpnl site at the N-terminus and a Sal I site at the C-terminal end. After trimming the ends of the PCR product with Kpn I and Sal I, the c-K4B-rø-? fragment can be ligated into a Kpnl -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of cysteine-12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide:
5'-GTAGTTGGAGCTGTTGGCGTAGGC-3' (SEQ.ID.NO.: 22)
After selection and sequencing for the correct nucleotide substitution, the mutated c-K4B-rα-ϊ-Val-12 can be excised from the pAlter-1 vector, using Kpnl and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with Kpnl and Sal I. The new recombinant plasmid will constitutively transcribe c-K4B-ra-?-Val-12 from the CMV promoter of the pCI vector.
Cloning of c-K-rø-.4A-Val-12 expression plasmid pSMS650
The human c-K4A-rα-? gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligo-nucleotide primers.
Sense strand: 5'-GAGAGGTACCGCCACCATGACTGAATATAAACTTGTGG-3' (SEQ.ID.NO.: 23)
Antisense strand: 5'-CTCTGTCGACAGATTACATTATAATGCATTTTTTAATTTTCACAC-3' (SEQ.ID.NO.: 24)
The primers will amplify a c-K4A-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, a Kpnl site at the N-terminus and a Sal I stite at the C-terminal end. After trimming the ends of the PCR product with Kpn I and Sal I, the c-K-ras4A fragment can be ligated into a Kpnl -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of cysteine-12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide:
5'-GTAGTTGGAGCTGTTGGCGTAGGC-3' (SEQ.ID.NO.: 25)
After selection and sequencing for the correct nucleotide substitution, the mutated c-K4A-rα_?-Val-12 can be excised from the pAlter-1 vector, using Kpnl and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with Kpnl and Sal I. The new recombinant plasmid, pSMS650, will constitutively transcribe c-K4A-ra-?-Val-12 from the CMV promoter of the pCI vector.
SEAP assay
Human C33A cells (human epitheial carcenoma - ATTC collection) are seeded in 10cm tissue culture plates in DMEM + 10% fetal calf serum + IX Pen/Strep + IX glutamine + IX NEAA. Cells are grown at 37°C in a 5% CO2 atmosphere until they reach 50-80% of confluency. The transient transfection is performed by the CaPO4 method
(Sambrook et al., 1989). Thus, expression plasmids for R-ras, N-ras, K-ras, Myr- ras or H-ras-CVLL are co-precipitated with the DSE-SEAP reporter construct. (A ras expression plasmid is not included when the cell is transfected with the pCMV- SEAP plasmid.) For 10 cm plates 600 μl of CaCl2-DNA solution is added dropwise while vortexing to 600 μl of 2X HBS buffer to give 1.2 ml of precipitate solution (see recipes below). This is allowed to sit at room temperature for 20 to 30 minutes. While the precipitate is forming, the media on the C33 A cells is replaced with DMEM (minus phenol red; Gibco cat. No. 31053-028)+ 0.5% charcoal stripped calf serum + IX (Pen Strep, Glutamine and nonessential aminoacids). The CaPO4-DNA precipitate is added dropwise to the cells and the plate rocked gently to distribute. DNA uptake is allowed to proceed for 5-6 hrs at 37°C under a 5% CO2 atmosphere.
Following the DNA incubation period, the cells are washed with PBS and trypsinized with 1ml of 0.05% trypsin. The 1 ml of trypsinized cells is diluted into 10 ml of phenol red free DMEM + 0.2% charcoal stripped calf serum + IX (Pen/Strep, Glutamine and NEAA). Transfected cells are plated in a 96 well microtiter plate (100 μl/well) to which drug, diluted in media, has already been added in a volume of 100 μl. The final volume per well is 200 μl with each drug concentration repeated in triplicate over a range of half-log steps.
Incubation of cells and drugs is for 36 hours at 37°C under CO2. At the end of the incubation period, cells are examined micro-scopically for evidence of cell distress. Next, 100 μl of media containing the secreted alkaline phosphatase is removed from each well and transferred to a microtube array for heat treatment at 65 °C for 1 hour to inactivate endogenous alkaline phosphatases (but not the heat stable secreted phosphatase). The heat treated media is assayed for alkaline phosphatase by a luminescence assay using the luminescence reagent CSPD® (Tropix, Bedford, Mass.). A volume of 50 μl media is combined with 200 μl of CSPD cocktail and incubated for 60 minutes at room temperature. Luminesence is monitored using an ML2200 microplate luminometer (Dynatech). Luminescence reflects the level of activation of the fos reporter construct stimulated by the transiently expressed protein.
DNA-CaPOj. precipitate for 10cm. plate of cells Ras expression plasmid (1 μg/μl) 10 μl DSE-SEAP Plasmid (1 μg/μl) 2 μl
Sheared Calf Thymus DNA (1 μg/μl) 8 μl 2M CaCl2 74 μl dH2θ 506 μl 2X HBS Buffer 280mM NaCl lOmM KCl 1.5mM Na2HPθ4 2H2θ 12mM dextrose 50mM HEPES Final pH = 7.05
Luminesence Buffer (26ml) Assay Buffer 20ml
Emerald Reagent™ (Tropix) 2.5ml lOOmM homoarginine 2.5ml
CSPD Reagent® (Tropix) 1.0ml
Assay Buffer
Add 0.05M Na2CO3 to 0.05M NaHCO3 to obtain pH 9.5. Make ImM in MgCl2
EXAMPLE 10
The processing assays employed are modifications of that described by DeClue et al rCancer Research 51. 712-717. 19911.
K4B-Ras processing inhibition assay PSN-1 (human pancreatic carcinoma) or viral-K4B-ras-transformed
Ratl cells are used for analysis of protein processing. Subconfluent cells in 100 mm dishes are fed with 3.5 ml of media (methionine-free RPMI supplemented with 2% fetal bovine serum or cysteine-free/methionine-free DMEM supplemented with 0.035 ml of 200 mM glutamine (Gibco), 2% fetal bovine serum, respectively) containing the desired concentration of test compound, lovastatin or solvent alone. Cells treated with lovastatin (5-10 μM), a compound that blocks Ras processing in cells by inhibiting a rate-limiting step in the isoprenoid biosynthetic pathway, serve as a positive control. Test compounds are prepared as lOOOx concentrated solutions in DMSO to yield a final solvent concentration of 0.1%. Following incubation at 37°C for two hours 204 μCi/ml [35s]Pro-Mix (Amersham, cell labeling grade) is added.
After introducing the label amino acid mixture, the cells are incubated at 37°C for an additional period of time (typically 6 to 24 hours). The media is then removed and the cells are washed once with cold PBS. The cells are scraped into 1 ml of cold PBS, collected by centrifugation (10,000 x g for 10 sec at room temperature), and lysed by vortexing in 1 ml of lysis buffer (1% Nonidet P-40, 20 mM HEPES, pH 7.5, 150 mM NaCl, 1 mM EDTA, 0.5% deoxycholate, 0.1% SDS, 1 mM DTT, 10 μg/ml AEBSF, 10 .g/ml aprotinin, 2 μg/ml leupeptin and 2 μg/ml antipain). The lysate is then centrifuged at 15,000 x g for 10 min at 4°C and the supernatant saved.
For immunoprecipitation of Ki4B-Ras, samples of lysate supernatant containing equal amounts of protein are utilized. Protein concentration is determined by the bradford method utilizing bovine serum albumin as a standard. The appropri- ate volume of lysate is brought to 1 ml with lysis buffer lacking DTT and 8 μg of the pan Ras monoclonal antibody, Y13-259, added. The protein/antibody mixture is incubated on ice at 4°C for 24 hours. The immune complex is collected on pansorbin (Calbiochem) coated with rabbit antiserum to rat IgG (Cappel) by tumbling at 4°C for 45 minutes. The pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in 100 μl elution buffer (10 mM Tris pH 7.4, 1% SDS). The Ras is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation (15,000 x g for 30 sec. at room temperature).
The supernatant is added to 1 ml of Dilution Buffer 0.1% Triton X- 100, 5 mM EDTA, 50 mM NaCl, 10 mM Tris pH 7.4) with 2 μg Kirsten-ras specific monoclonal antibody, c-K-ras Ab-1 (Calbiochem). The second protein/antibody mixture is incubated on ice at 4°C for 1-2 hours. The immune complex is collected on pansorbin (Calbiochem) coated with rabbit antiserum to rat IgG (Cappel) by tumbling at 4°C for 45 minutes. The pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in Laemmli sample buffer. The Ras is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation. The supernatant is subjected to SDS-PAGE on a 12% acrylamide gel (bis-acrylamide: acrylamide, 1:100), and the Ras visualized by fluorography. hDJ Processing Inhibition Assay
PSN-1 cells are seeded in 24- well assay plates. For each compound to be tested, the cells are treated with a minimum of seven concentrations in half-log steps. The final solvent (DMSO) concentration is 0.1%. A vehicle-only control is included on each assay plate. The cells are treated for 24 hours at 37°C / 5% CO2-
The growth media is then aspirated and the samples are washed with PBS. The cells are lysed with SDS-PAGE sample buffer containing 5% 2-mercaptoethanol and heated to 95°C for 5 minutes. After cooling on ice for 10 minutes, a mixture of nucleases is added to reduce viscosity of the samples.
The plates are incubated on ice for another 10 minutes. The samples are loaded onto pre-cast 8% acrylamide gels and electrophoresed at 15 mA/gel for 3-4 hours. The samples are then transferred from the gels to PNDF membranes by Western blotting. The membranes are blocked for at least 1 hour in buffer containing
2% nonfat dry milk. The membranes are then treated with a monoclonal antibody to hDJ-2 (Νeomarkers Cat. # MS-225), washed, and treated with an alkaline phosphatase-conjugated secondary antibody. The membranes are then treated with a fluorescent detection reagent and scanned on a phosphorimager. For each sample, the percent of total signal corresponding to the unprenylated species of hDJ (the slower-migrating species) is calculated by densitometry. Dose-response curves and EC50 values are generated using 4- parameter curve fits in SigmaPlot software.
EXAMPLE 11
Rapl processing inhibition assay
Protocol A: Cells are labeled, incubated and lysed as described in Example 10.
For immunoprecipitation of Rapl, samples of lysate supernatant containing equal amounts of protein are utilized. Protein concentration is determined by the bradford method utilizing bovine serum albumin as a standard. The appropriate volume of lysate is brought to 1 ml with lysis buffer lacking DTT and 2 μg of the Rapl antibody, Rapl/Krevl (121) (Santa Cruz Biotech), is added. The protein/ antibody mixture is incubated on ice at 4°C for 1 hour. The immune complex is collected on pansorbin (Calbiochem) by tumbling at 4°C for 45 minutes. The pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in 100 μl elution buffer (10 mM Tris pH 7.4, 1% SDS). The Rapl is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation (15,000 x g for 30 sec. at room temperature).
The supernatant is added to 1 ml of Dilution Buffer (0.1% Triton X-100, 5 mM EDTA, 50 mM NaCl, 10 mM Tris pH 7.4) with 2 μg Rapl antibody, Rapl/Krevl (121) (Santa Cruz Biotech). The second protein/antibody mixture is incubated on ice at 4°C for 1-2 hours. The immune complex is collected on pansorbin (Calbiochem) by tumbling at 4°C for 45 minutes. The pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in Laemmli sample buffer. The Rapl is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation. The supernatant is subjected to SDS-PAGE on a 12% acrylamide gel (bis-acrylamide: acrylamide, 1:100), and the Rapl visualized by fluorography.
Protocol B: PSN-1 cells are passaged every 3-4 days in 10cm plates, splitting near-confluent plates 1:20 and 1:40. The day before the assay is set up, 5x 10 cells are plated on 15cm plates to ensure the same stage of confluency in each assay. The media for these cells is RPM1 1640 (Gibco), with 15% fetal bovine serum and lx Pen/Strep antibiotic mix. The day of the assay, cells are collected from the 15 cm plates by trypsinization and diluted to 400,000 cells/ml in media. 0.5ml of these diluted cells are added to each well of 24-well plates, for a final cell number of 200,000 per well. The cells are then grown at 37°C overnight.
The compounds to be assayed are diluted in DMSO in 1/2-log dilutions. The range of final concentrations to be assayed is generally 0.1-100 μM. Four concentrations per compound is typical. The compounds are diluted so that each concentration is lOOOx of the final concentration (i.e., for a 10 μM data point, a 10 mM stock of the compound is needed). 2 μL of each lOOOx compound stock is diluted into 1 ml media to produce a 2X stock of compound. A vehicle control solution (2 μL DMSO to 1ml media), is utilized. 0.5 ml of the 2X stocks of compound are added to the cells.
After 24 hours, the media is aspirated from the assay plates. Each well is rinsed with 1ml PBS, and the PBS is aspirated. 180 μL SDS-PAGE sample buffer (Novex) containing 5% 2-mercapto-ethanol is added to each well. The plates are heated to 100°C for 5 minutes using a heat block containing an adapter for assay plates. The plates are placed on ice. After 10 minutes, 20 μL of an RNAse/DNase mix is added per well. This mix is lmg/ml DNasel (Worthington Enzymes), 0.25 mg/ml Rnase A (Worthington Enzymes), 0.5 M Tris-HCl pH 8.0 and 50 mM MgCl2.
The plate is left on ice for 10 minutes. Samples are then either loaded on the gel, or stored at -70°C until use.
Each assay plate (usually 3 compounds, each in 4-point titrations, plus controls) requires one 15-well 14% Novex gel. 25 μl of each sample is loaded onto the gel. The gel is run at 15 mA for about 3.5 hours. It is important to run the gel far enough so that there will be adequate separation between 21 kd (Rapl) and 29kd (Rab6).
The gels are then transferred to Novex pre-cut PVDF membranes for 1.5 hours at 30V (constant voltage). Immediately after transferring, the membranes are blocked overnight in 20ml Western blocking buffer (2% nonfat dry milk in
Western wash buffer (PBS + 0.1% Tween-20). If blocked over the weekend, 0.02% sodium azide is added. The membranes are blocked at 4°C with slow rocking.
The blocking solution is discarded and 20ml fresh blocking solution containing the anti Rap la antibody (Santa Cruz Biochemical SC1482) at 1:1000 (diluted in Western blocking buffer) and the anti Rab6 antibody (Santa Cruz Biochemical SC310) at 1:5000 (diluted in Western blocking buffer) are added. The membranes are incubated at room temperature for 1 hour with mild rocking. The blocking solution is then discarded and the membrane is washed 3 times with Western wash buffer for 15 minutes per wash. 20ml blocking solution containing 1 : 1000 (diluted in Western blocking buffer) each of two alkaline phosphatase conjugated antibodies (Alkaline phosphatase conjugated Anti-goat IgG and Alkaline phosphatase conjugated anti-rabbit IgG [Santa Cruz Biochemical]) is then added. The membrane is incubated for one hour and washed 3x as above.
About 2 ml per gel of the Amersham ECF detection reagent is placed on an overhead transparency (ECF) and the PVDF membranes are placed face-down onto the detection reagent. This is incubated for one minute, then the membrane is placed onto a fresh transparency sheet.
The developed transparency sheet is scanned on a phosphorimager and the Rapla Minimum Inhibitory Concentration is determined from the lowest concentration of compound that produces a detectable Rapla Western signal. The Rapla antibody used recognizes only unprenylated/unprocessed Rapla, so that the precence of a detectable Rapla Western signal is indicative of inhibition of Rapla prenylation.
Protocol C:
This protocol allows the determination of an EC50 for inhibition of processing of Rapla. The assay is run as described in Protocol B with the following modifications. 20 μl of sample is run on pre-cast 10-20% gradient acrylamide mini gels (Novex Inc.) at 15 mA/gel for 2.5-3 hours. Prenylated and unprenylated forms of Rapla are detected by blotting with a polyclonal antibody (Rapl/Krev-1 Ab#121 ; Santa Cruz Research Products #sc-65), followed by an alkaline phosphatase- conjugated anti-rabbit IgG antibody. The percentage of unprenylated Rapla relative to the total amount of Rapla is determined by peak integration using Imagequant™ software (Molecular Dynamics). Unprenylated Rapla is distinguished from prenyl- ated protein by virtue of the greater apparent molecular weight of the prenylated protein. Dose-response curves and EC50 values are generated using 4-parameter curve fits in SigmaPlot software.
EXAMPLE 12
In vivo tumor growth inhibition assay (nude mouse)
In vivo efficacy as an inhibitor of the growth of cancer cells may be confirmed by several protocols well known in the art. Examples of such in vivo efficacy studies are described by N. E. Kohl et al. (Nature Medicine, 1:792-797 (1995)) and N. E. Kohl et al. (Proc. Nat. Acad. Sci. U.S.A., 91:9141-9145 (1994)). Rodent fibroblasts transformed with oncogenically mutated human Ha-ras or Ki-ras (lθ6 cells/animal in 1 ml of DMEM salts) are injected subcutaneously into the left flank of 8-12 week old female nude mice (Harlan) on day 0. The mice in each oncogene group are randomly assigned to a vehicle or compound treatment group. Animals are dosed subcutaneously starting on day 1 and daily for the duration of the experiment. Alternatively, the farnesyl-protein transferase inhibitor may be administered by a continuous infusion pump. Compound or vehicle is delivered in a total volume of 0.1 ml. Tumors are excised and weighed when all of the vehicle-treated animals exhibited lesions of 0.5-1.0 cm in diameter, typically 11-15 days after the cells were injected. The average weight of the tumors in each treatment group for each cell line is calculated.

Claims

WHAT IS CLAIMED IS:
1. A compound of the formula A:
Figure imgf000112_0001
wherein:
Rla, Rib, Rlc and Rid are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) RlOO-, f) RllS(O)m-, g) Rl0C(O)NRl0-, h) (RlO)2NC(O)-, i) CN, j) NO2, k) RlOC(O)-,
1) RlOOC(O)-, m) -N(RlO)2, n) RHOC(O)NR10-, o) unsubstituted or substituted C1-C6 alkyl, p) unsubstituted or substituted C2-C6 alkenyl, or q) unsubstituted or substituted C2-C6 alkynyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted Ci-Cβ alkyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C3-C10 cycloalkyl, unsubstituted or unsubstituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, RlOO-, Rl lS(O)m-, Rl0C(O)NRl0-, (R10)2NC(O)-, CN,
RlOC(O)-, RlOθC(O)-, -N(RlO)2, and RHOC(O)-NRlO-,
or two Rl^s, two Ribs or two Rlcs, on the same carbon atom may be combined to form -(CH2)U-;
R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-8 alkyl, c) unsubstituted or substituted C2-8 alkenyl, d) unsubstituted or substituted C2-8 alkynyl, e) unsubstituted or substituted aryl, f) unsubstituted or substituted heterocycle, g)
R6R7
or h)
wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) Cι_4 alkyl,
Figure imgf000113_0001
d) halogen, e) CN,
2) C3-6 cycloalkyl, 3) OR6,
4) SR4, S(O)R4, SO2R4, 5) — NR6R7
Figure imgf000114_0001
11) — S02-NR6R7
Figure imgf000114_0002
13)
- 0 R6
Figure imgf000114_0003
15) N3, or
16) F; or R4 is selected from Cχ.6 alkyl, C3.6 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with:
1) Cι_6 alkoxy,
2) aryl or heterocycle,
3) halogen,
4) HO,
5)
Figure imgf000115_0001
6) S02 H,
7) N(R10)2, or
8) Ci-4 perfluoroalkyl;
R5, R6 and R7 are independently selected from: a) hydrogen, b) RlOC(O)-, c) RlOθC(O)-, or d) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3..6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
1) unsubstituted or substituted C1-C6 alkyl,
2) RlOO-,
3) unsubstituted or substituted aryl or heterocycle,
4) halogen, 5) R10C(O)NR10-,
6)
Figure imgf000115_0002
7) SO2Rll,
8) N(R10)2,
9) C3.6 cycloalkyl, 10) Cl-C6 perfluoroalkyl,
11) RlOθC(O)-,
12) RllOC(O)NRl0-,
13) CN, and
14) NO2; or
R6 and R7 may be joined in a ring; and independently,
R5 and R7 may be joined in a ring;
R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C 10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) C1-C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O)m-, k) Rl0C(O)NRl0-, 1) (RlO)2NC(O)-, m) CN, n) NO2, o) Rl0C(O)-, p) RlOθC(O)-, q) -N(RlO)2, r) RllOC(O)NRl0-, and s) unsubstituted or substituted Cχ-C6 alkyl, optionally substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C3-C10 cycloalkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, Cχ-C6 perfluoroalkyl, F, Cl, Br, RlOO- RllS(O)m-, Rl0C(O)NRl0-, (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, or Rl0θC(O)NRl0-;
R9 is selected from: a) hydrogen, b) unsubstituted or substituted C2-C6 alkenyl, c) unsubstituted or substituted C2-C6 alkynyl, d) Cχ-C6 perfluoroalkyl, e) halo,
D RlOO-, g) RπS(O)m-, h) RlOC(O)NRlO-, i) (RlO)2NC(O)-, j) CN, k) NO2,
1) Rl0C(O)-, m) RlOθC(O)-, n) -N(R10)2, o) RllOC(O)NRl0-, or
P) unsubstituted or substituted C1-C6 alkyl optionally substituted by aryl,
C3-C10 cycloalkyl, C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-,
RllS(O)m-, RlOC(O)NRlO-, (RlO)2NC(O)-, CN, Rl0C(O)-,
RlθQC(O)-, -N(RlO)2, or RllOC(O)NRl0-;
R 10 is independently selected from a) hydrogen, b) unsubstituted or substituted C1-C6 alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
Rl 1 is independently selected from a) unsubstituted or substituted C1-C6 alkyl, b) unsubstituted or substituted £ c) unsubstituted or substituted r
Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(Rl0)-, g) -S(O)2N(RlO)-,
Figure imgf000118_0001
i) S(O)m;
A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(Rl0)-, g) -S(O)2N(RlO)-,
Figure imgf000118_0002
i) -NRl0c(O)NRl0-, or j) S(O)m;
G is selected from H2 or O;
V is selected from: a) heterocycle, or b) aryl;
W is a heterocycle;
X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, ) -O-C(O)NRl0-, g) -NRl0C(O)NRl0-, h) O,
Figure imgf000119_0001
1) S(O)m;
Y is selected from a) a bond, b) -C(=O)-, c) -C(O)NRl0-, d) -C(O)O-, or e) -S(=O)m;
Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
1) Ci-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or ) Ci-4 perfluoroalkyl;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen,
4) OR6.
5) NR6R7,
6) CN,
7) NO2,
8) CF3;
9) -S(O)mR4,
Figure imgf000120_0001
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
Z is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
1) Ci-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or h) Cχ-4 perfluoroalkyl;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen,
4) OR6,
5) NR6R7,
6) CN, • ■ ■
7) NO2,
8) CF3,
9) -S(O)mR4,
Figure imgf000120_0002
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
kis 0, 1,2, 3 or 4; mis 0, 1 or 2; nis 0, 1,2, 3, 4 or 5; p is independently 0,1, 2, 3, 4 or 5; qis 1,2 or 3; ris 0,1,2, 3, 4 or 5; s is independently 0,1, 2, 3, 4 or 5; tis Oor 1; and u is independently 2, 3, 4, 5 or 6;
or a pharmaceutically acceptable salt or stereoisomer thereof.
The compound, according to Claim 1, of formula A:
^s
Figure imgf000121_0001
wherein: Rla, Rib, Rlc and Rid are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) RlOO-, f) RllS(O)m-, g) Rl0C(O)NRl0-, h) (RlO)2NC(O)-, i) CN, j) NO2, k) RlOC(O)-,
1) RlOθC(O)-, m) -N(RlO)2, n) RllOC(O)NRl0-, o) unsubstituted or substituted Cχ-C6 alkyl, p) unsubstituted or substituted C2-C6 alkenyl, or q) unsubstituted or substituted C2-C6 alkynyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted Cχ-C6 alkyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted C3-Cχo cycloalkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, RlOO-, RllS(O)m-, RlOC(O)NRlO-, (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, and Rl lOC(O)NRlO-,
or two Rlas, two Ri s or two Rlcs, on the same carbon atom may be combined to form -(CH2)uS
R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Ci-8 alkyl, c) unsubstituted or substituted C2-8 alkenyl, d) unsubstituted or substituted C2-8 alkynyl, e) unsubstituted or substituted aryl, f) unsubstituted or substituted heterocycle,
Figure imgf000122_0001
Figure imgf000123_0001
wherein the substituted group is substituted with one or more of: 1) aryl or heterocycle, unsubstituted or substituted with: a) Ci-4 alkyl,
Figure imgf000123_0002
d) halogen, e) CN, 2) C3.6 cycloalkyl,
3) OR6,
Figure imgf000123_0003
5) — NR6R7
Figure imgf000123_0004
Figure imgf000124_0001
11) — S02-NR6R7
Figure imgf000124_0002
13)
^ R6
0
14)
^τ 0 0R6
15) N3, or
16) F; or
R4 is selected from Ci-4 alkyl, C3.6 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with:
1) Cχ_4 alkoxy,
2) aryl or heterocycle,
3) halogen, 4) HO,
Figure imgf000124_0003
Figure imgf000125_0001
7) N(RlO)2, or
8) C 1-.4 perfluoroalkyl;
R5, R6 and R7 are independently selected from: a) hydrogen, b) Rl°C(O)-, c) Rl°OC(O)-, or d) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3.6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
1) RlOO-,
2) aryl or heterocycle,
3) halogen,
4) Rl0C(O)NRl0-,
5)
O
6) SO2Rll,
7) N(RlO)2,
8) C3-6 cycloalkyl,
9) C1-C6 perfluoroalkyl,
10) RlOθC(O)-,
11) RllOC(O)NRl0-,
12) CN, and
13) NO2; or
R6 and R7 may be joined in a ring; and independently,
R5 and R may be joined in a ring; R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) Cχ-C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O)m-. k) Rl0C(O)NRl0-, 1) (RlO)2NC(O)-, m) CN, n) NO2, o) RlOC(O)-, p) RlOθC(O)-, q) -N(RlO)2, r) RllOC(O)NRlO-, and s) unsubstituted or substituted Cχ-C6 alkyl, optionally substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cχ-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, Rl0C(O) NRlO., (RlO)2NC(O)-, CN, Rl0C(O)-, RlOθC(O)-, -N(RlO)2, or Rl0θC(O)NRl0-;
is selected from: a) hydrogen, b) unsubstituted or substituted C2-C6 alkenyl, c) unsubstituted or substituted C2-C6 alkynyl, d) C1-C6 perfluoroalkyl, e) halo, ) RlOO-, g) RllS(O)m-, h) RlOC(O)NRlO-, i) (RlO)2NC(O)-, j) CN, k) NO2, 1) RlOC(O)-, m) RlOθC(O)-, n) -N(RlO)2, o) RllOC(O)NRl0-, or p) unsubstituted or substituted Ci-Cg alkyl optionally substituted by C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, Rl lS(O)m-, Rl0c(O)
NRlO., (RlO)2NC(O)-, CN, Rl0c(O)-, RlOθC(O)-, -N(RlO)2, or RllOC(O)NRl0-;
RlO is independently selected from a) hydrogen, b) unsubstituted or substituted Cχ-C6 alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
Rll is independently selected from a) unsubstituted or substituted Cχ-C6 alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRlO-, d) -NRlOC(O)-, e) O,
-N(RlO)-, g) -S(O)2N(R10)-,
Figure imgf000128_0001
i) S(O)m;
A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(RlO)-, g) -S(O)2N(RlO)-,
Figure imgf000128_0002
i) -NRl0C(O)NRl0-, or j) S(O)m;
G is selected from H2 or O;
V is selected from pyridyl or phenyl;
W is a heterocycle;
X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, ) -O-C(O)NRl0-, g) -NRl0c(O)NRl0-, h) O, i) -N(Rl0)-, j) -S(O)2N(R10)-,
Figure imgf000128_0003
1) S(O)m; Y is selected from a) a bond, b) -C(=O)-, c) -C(O)NRl0-, d) -C(O)O-, or e) -S(=O)m;
Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
1) Cχ-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Cχ-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or h) Ci-4 perfluoroalkyl;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen,
4) OR6>
5) NR6R7,
6) CN,
7) NO2,
8) CF3;
9) -S(O)mR4,
Figure imgf000129_0001
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl; Z2 is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
1) Ci-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Cχ-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7 or h) C 1 -4 perfluoroalkyl ;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen,
4) OR6.
5) NR6R7,
6) CN,
7) NO2,
8) CF3,
9) -S(O)mR4,
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
k is 0, 1, 2, 3 or 4; m is 0, 1 or 2; n is 0, 1, 2, 3, 4 or 5; p is independently 0, 1, 2, 3, 4 or 5; q is 1, 2 or 3; r is 0, 1, 2, 3, 4 or 5; s is independently 0, 1, 2, 3, 4 or 5; t is O or l; and u is independently 2, 3, 4, 5 or 6;
or a pharmaceutically acceptable salt or stereoisomer thereof.
3. The compound, according to Claim 1, of formula B:
Figure imgf000131_0001
wherein:
Rla, Rib and Rlc are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) -N(RlO)2, f) unsubstituted or substituted C2-C6 alkenyl g) -ORlO, or h) unsubstituted or substituted C1-C6 alkyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, RlOO-, and -N(Rl )2, or two Rlas, two Ribs or two Rlcs, on the same carbon atom may be combined to form -(CH2)u-;
Ri is selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl e) RlOO-, f) RllS(O)m-, g) RlOC(O)NRlO-, h) (RlO)2NC(O)-, i) CN, j) NO2, k) RlOC(O)-,
1) RlOθC(O)-, m) -N(RlO)2, n) RllOC(O)NRlO-, o) unsubstituted or substituted Cχ-C6 alkyl,
P) unsubstituted or substituted C2-C6 alkenyl, or q) unsubstituted or substituted C2-C6 alkynyl,
wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, RlOO-, Rl lS(O)m-, RlOC(O)NRlO-, (RlO)2NC(O)-,
CN, RlOC(O)-, RlOOC(O)-, -N(RlO)2, and Rl lOC(O)NRlO-;
R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Ci_8 alkyl, c)
R6R7
or
Figure imgf000133_0001
wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) Cχ-4 alkyl,
Figure imgf000133_0002
d) halogen, e) CN,
2) OR6, 3) SR4, S(O)R4, SO2R4, or
4) -NR6R7;
R4 is selected from Ci-4 alkyl, or C3_6 cycloalkyl, unsubstituted or substituted with: 1) Ci-4 alkoxy,
2) aryl or heterocycle,
3) halogen,
4) HO,
Figure imgf000133_0003
7) N(RlO)2, or
8) C 1 -4 perfluoroalkyl ;
R6 and R7 are independently selected from: a) hydrogen, b) RlOC(O)-, c) RlOθC(O)-, or d) Cχ-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
1) RlOO-,
2) aryl or heterocycle,
3) halogen,
4) Rl0C(O)NRl0-,
Figure imgf000134_0001
6) SO2R11,
7) N(R10)2,
8) C3-6 cycloalkyl,
9) Cχ-C6 perfluoroalkyl,
10) RlOθC(O)-,
11) RllOC(O)NRl0-,
12) CN, and
13) NO2; or
R6 and R7 may be joined in a ring;
R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl,
D unsubstituted or substituted C2-C6 alkynyl, g) C 1 -C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O)m-, k) Rl0C(O)NRl0-,
1) (RlO)2NC(O)-, m) CN, n) NO2, o) Rl C(O)-, p) RlOOC(O)-, q) -N(R10)2, r) RllOC(O)NRl0-, or s) unsubstituted or substituted Cχ-C6 alkyl, optionally substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, RlOC(O)
NRlO., (Rl0)2NC(O)-, CN, RlOC(O)-, RlOOC(O)-, -N(RlO)2, or RlOθC(O) NRlO-;
R9 is selected from: a) hydrogen, b) Cl -Cβ perfluoroalkyl, c) RlOO-, d) Rl0C(O)NRl0-, e) (RlO)2NC(O)-, or f) unsubstituted or substituted Ci-Cβ alkyl optionally substituted by
C1-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, RlOC(O) NRlO., (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, or RllOC(O)NRlO-;
RlO is independently selected from a) hydrogen, b) unsubstituted or substituted C 1 -C alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
R l is independently selected from a) unsubstituted or substituted Ci-Cβ alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(R10)-, g) -S(O)2N(R10)-,
Figure imgf000136_0001
i) S(O)m;
A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NR10-, d) -NRl C(O)-, e) O,
-N(R10)-, g) -S(O)2N(RlO)-,
Figure imgf000136_0002
i) -NRl0C(O)NRl0-, or j) S(O)m;
G is selected from H2 or O;
W is a heterocycle selected from imidazolyl, pyridyl, or triazolyl;
X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, ) -O-C(O)NRl -, g) -NRl0C(O)NRl0-, h) O, i) -N(RlO)-, j) -S(O)2N(RlO)-,
Figure imgf000137_0001
1) S(O)m;
Y is selected from a) a bond, b) -C(=O)-, c) -C(O)NRl0-, d) -C(O)O-, or e) -S(=O)m;
Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of: 1) Cχ-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) NR6R7 c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or h) C 1 -4 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen,
4) ©Ro.
5) NR6R7, 6) CN,
7) NO2,
8) CF3;
Figure imgf000138_0001
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
kis 0,1, 2, 3 or 4; mis 0, 1 or 2; nis 0,1, 2, 3, 4 or 5; p is independently 0, 1,2, 3, 4 or 5; qis 1,2 or 3; ris 0,1,2, 3, 4 or 5; sisi ndependently 0,1,2, 3, 4 or 5; tis 0 or 1; and uis; independ. ϊntly 2, 3, 4, 5 or 6;
or a pharmaceutically acceptable salt or stereoisomer thereof.
4. The compound, according to Claim 1, of formula C
Figure imgf000138_0002
wherein:
Rla, Rib and Rlc are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) -N(Rl0)2, f) unsubstituted or substituted C2-C6 alkenyl g) -ORlO, or h) unsubstituted or substituted Cχ-C6 alkyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, RlOO-, and -N(RlO)2, or two Rlas, two Ribs or two Rlcs, on the same carbon atom may be combined to form -(CH2)u-;
Rid is selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) RlOO-, f) -N(RlO)2, or g) unsubstituted or substituted Cχ-C6 alkyl, wherein the substituent on the substituted group is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, RlOo-, Rl0c(O)NRl0-, (RlO)2NC(O)-, Rl0C(O)-, RlOθC(O)-, -N(RlO)2, and RllOC(O)-NRl0-;
R2 is independently selected from a) hydrogen, b) unsubstituted or substituted Cχ-8 alkyl, c)
R ϊ6°Rr,7'
O or
Figure imgf000140_0001
wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) Cχ-4 alkyl,
Figure imgf000140_0002
d) halogen, e) CN,
2) OR6,
3) SR4, S(O)R4, SO2R4, or
4) -NR6R7;
R4 is selected from Cχ_4 alkyl, or C3-6 cycloalkyl, unsubstituted or substituted with:
1) Cχ-4 alkoxy,
2) aryl or heterocycle,
3) halogen, or 4) -OH;
R6 and R are independently selected from: a) hydrogen, or b) Cχ-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-6 perfluoroalkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from:
1) RlOO-,
2) aryl or heterocycle, 3) halogen,
4) Rl0C(O)NRl0-, 5)
Figure imgf000141_0001
7) N(RlO)2, or 8) C3-6 cycloalkyl, or
R6 and R7 may be joined in a ring;
R8 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted C3-C10 cycloalkyl, e) unsubstituted or substituted C2-C6 alkenyl, f) unsubstituted or substituted C2-C6 alkynyl, g) Cχ-C6 perfluoroalkyl, h) halo, i) RlOO-, j) RllS(O)m-, k) Rl0C(O)NRl0-,
1) (RlO)2NC(O)-, m) CN, n) NO2, o) Rl0C(O)-, p) RlOθC(O)-,
Figure imgf000141_0002
r) RllOC(O)NRl0-, and s) unsubstituted or substituted Cχ-C6 alkyl, optionally substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C alkenyl, C2-C6 alkynyl,
Cχ-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, Rl0C(O)NRl0-, (RlO)2NC(O)-, CN, Rl0C(O)-, RlOθC(O)-, -N(RlO)2, or Rl0θC(O)NRl0-; R9 is selected from: a) hydrogen, b) C1-C6 perfluoroalkyl, or c) unsubstituted or substituted C1-C6 alkyl optionally substituted by
Cχ-C6 perfluoroalkyl, F, Cl, Br, RlOO-, RllS(O)m-, Rl0C(O)NRl0-, (RlO)2NC(O)-, CN, RlOC(O)-, RlOθC(O)-, -N(RlO)2, or RllOC(O)NRl0-;
Rl is independently selected from a) hydrogen, b) unsubstituted or substituted C1-C6 alkyl, c) unsubstituted or substituted C1-C3 perfluoroalkyl, d) unsubstituted or substituted benzyl, e) unsubstituted or substituted aryl, or f) unsubstituted or substituted heterocycle;
Rll is independently selected from a) unsubstituted or substituted Cχ-C6 alkyl, b) unsubstituted or substituted aryl, or c) unsubstituted or substituted heterocycle;
Al and A3 are independently selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, f) -N(Rl0)-, g) -S(O)2N(RlO)-,
Figure imgf000142_0001
i) S(O)m;
A2 is selected from a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) O, ) -N(RlO)-, g) -S(O)2N(R10)-, h) -N(RlO)S(O)2-, i) -NRl0C(O)NRl0-, or j) S(O)m;
G is selected from H2 or O;
X is selected from: a) a bond, b) -C(O)-, c) -C(O)NRl0-, d) -NRlOC(O)-, e) -NRl0C(O)-O-, ) -O-C(O)NRlO-, g) -NRl0C(O)NRl0-, h) O, i) -N(Rl0)-, j) -S(O)2N(R10)_,
Figure imgf000143_0001
1) S(O)m;
Y is selected from a) a bond, b) -C(=O)-, c) -C(O)NRl0-, d) - -C(O)O-, or e) -S(=O)m;
Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of:
1) Cχ-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with: a) Cχ-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f) -S(O)mR4, g) -C(O)NR6R7, or h) Cχ-4 perfluoroalkyl;
2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
3) halogen, 4) OR6,
5) NR6R7,
6) CN,
7) NO2,
8) CF3; 9) -S(O)mR4,
Figure imgf000144_0001
11) -C(O)NR6R7,
12) -C(O)OR6, or
13) C3-C6 cycloalkyl;
kis 0,1,2, 3 or 4; m is 0, 1 or 2; nis 0,1,2, 3, 4 or 5; p is independently 0, 1, 2, 3, 4 or 5; qis 1,2 or 3; ris 0,1,2, 3, 4 or 5; s is independently 0, 1, 2, 3, 4 or 5; tis Oorl; and u is independently 2, 3, 4, 5 or 6; or a pharmaceutically acceptable salt or stereoisomer thereof.
A compound which is selected from:
Figure imgf000145_0001
6(R or S)-l 1(R or S)-amino-l l-(l-methyl-lH-imidazol-5-yl)-17-oxa-2-thia-3,7- diazatetracyclo [16.3.1.13,6.ll2,16]tetracosa-l(22),12(23),13,15,18,20-hexaene-15- carbonitrile 2,2-dioxide,
Figure imgf000145_0002
(14R or S,19S or R)-14-methyl- 14-(1 -methyl- lH-imidazol-5-yl)- 16-oxo-8-oxa-2- thia-l,15-diazatetracyclo[17.3.1.13,7.l9,13]pentacosa-3(25),4,6,9(24),10,12-hexaene- 10-carbonitrile 2,2-dioxide
Figure imgf000146_0001
(14R ør S,18R or S) -14-methyl-14-(l-methyl-lH-imidazol-5-yl)-16-oxo-8-oxa-2- thia-l,15-diazatetracyclo[16.3.1.13,7.l9,13]tetracosa-3(24),4,6,9(23),10,12-hexaene-10- carbonitrile 2,2-dioxide
Figure imgf000146_0002
(14R or S, 17R or S)-14-methyl-14-(l-methyl-lH-imidazol-5-yl)-16-oxo-8-oxa-2-thia- l,15-diazatetracyclo[15.3.1.13,7.l9,13]tricosa-3(23),4,6,9(22),10,12-hexaene-10- carbonitrile 2,2-dioxide
or a pharmaceutically acceptable salt or stereoisomer thereof.
6. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 1.
7. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 4.
8. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 5.
9. A method for inhibiting prenyl-protein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 1.
10. A method for inhibiting prenyl-protein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 4.
11. A method for inhibiting prenyl-protein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 5.
12. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 1.
13. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 4.
14. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim
5.
15. A method for treating neurofibromin benign proliferative disorder which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 1.
16. A method for treating blindness related to retinal vascularization which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 1.
17. A method for treating infections from hepatitis delta and related viruses which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 1.
18. A method for preventing restenosis which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 1.
19. A method for treating polycystic kidney disease which comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 1.
20. A method of conferring radiation sensitivity on a tumor cell using a therapeutically effective amount of a compound of Claim 1 in combination with radiation therapy.
21. A method of using a therapeutically effective amount of a compound of Claim 1 in combination with an antineoplastic to treat cancer.
22. A method according to Claim 21 wherein the antineoplastic is paclitaxel.
23. A pharmaceutical composition made by combining the compound of Claim 1 and a pharmaceutically acceptable carrier.
24. A process for making a pharmaceutical composition comprising combining a compound of Claim 1 and a pharmaceutically acceptable carrier.
PCT/US2001/027013 2000-09-05 2001-08-30 Inhibitors of prenyl-protein transferase WO2002020015A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2001290588A AU2001290588A1 (en) 2000-09-05 2001-08-30 Inhibitors of prenyl-protein transferase

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US23010500P 2000-09-05 2000-09-05
US60/230,105 2000-09-05

Publications (1)

Publication Number Publication Date
WO2002020015A1 true WO2002020015A1 (en) 2002-03-14

Family

ID=22863969

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/027013 WO2002020015A1 (en) 2000-09-05 2001-08-30 Inhibitors of prenyl-protein transferase

Country Status (2)

Country Link
AU (1) AU2001290588A1 (en)
WO (1) WO2002020015A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005105783A1 (en) * 2004-05-03 2005-11-10 Janssen Pharmaceutica N.V. Diastereoselective synthesis process with 6-bromo-4-(3-chlorophenyl)-2-methoxy-quinoline
JP2007536333A (en) * 2004-05-03 2007-12-13 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Diastereoselective addition of lithiated N-methylimidazole to sulfimines
US7507760B2 (en) * 2004-01-22 2009-03-24 Neuromed Pharmaceuticals Ltd. N-type calcium channel blockers
JP2009519233A (en) * 2005-11-23 2009-05-14 ブリストル−マイヤーズ スクイブ カンパニー Heterocyclic CETP inhibitor

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6316436B1 (en) * 1998-12-08 2001-11-13 Merck & Co., Inc. Inhibitors of prenyl-protein transferase

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6316436B1 (en) * 1998-12-08 2001-11-13 Merck & Co., Inc. Inhibitors of prenyl-protein transferase

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7507760B2 (en) * 2004-01-22 2009-03-24 Neuromed Pharmaceuticals Ltd. N-type calcium channel blockers
WO2005105783A1 (en) * 2004-05-03 2005-11-10 Janssen Pharmaceutica N.V. Diastereoselective synthesis process with 6-bromo-4-(3-chlorophenyl)-2-methoxy-quinoline
JP2007536334A (en) * 2004-05-03 2007-12-13 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Diastereoselective synthesis method using 6-bromo-4- (3-chlorophenyl) -2-methoxy-quinoline
JP2007536333A (en) * 2004-05-03 2007-12-13 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Diastereoselective addition of lithiated N-methylimidazole to sulfimines
US7572916B2 (en) 2004-05-03 2009-08-11 Janssen Pharmaceutica Nv Diastereoselective synthesis process with 6-bromo-4-(3-chlorophenyl)-2-methoxy-quinoline
JP4917022B2 (en) * 2004-05-03 2012-04-18 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Diastereoselective synthesis method using 6-bromo-4- (3-chlorophenyl) -2-methoxy-quinoline
JP4917021B2 (en) * 2004-05-03 2012-04-18 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Diastereoselective addition of lithiated N-methylimidazole to sulfimines
JP2009519233A (en) * 2005-11-23 2009-05-14 ブリストル−マイヤーズ スクイブ カンパニー Heterocyclic CETP inhibitor
US7888376B2 (en) * 2005-11-23 2011-02-15 Bristol-Myers Squibb Company Heterocyclic CETP inhibitors
AU2006318310B2 (en) * 2005-11-23 2012-05-03 Bristol-Myers Squibb Company Heterocyclic CETP inhibitors

Also Published As

Publication number Publication date
AU2001290588A1 (en) 2002-03-22

Similar Documents

Publication Publication Date Title
AU2002211828A1 (en) Inhibitors of prenyl-protein transferase
WO2000001382A1 (en) Inhibitors of prenyl-protein transferase
US6284755B1 (en) Inhibitors of prenyl-protein transferase
US20020052380A1 (en) Inhibitors of prenyl-protein transferase
WO2001060368A1 (en) Inhibitors of prenyl-protein transferase
US6441017B1 (en) Inhibitors of prenyl-protein transferase
WO2001017992A1 (en) Inhibitors of prenyl-protein transferase
US6350755B1 (en) Inhibitors of prenyl-protein transferase
WO2001045707A1 (en) Inhibitors of prenyl-protein transferase
US20030134860A1 (en) Inhibitors of prenyl-protein transferase
US6525074B2 (en) Inhibitors of prenyl-protein transferase
EP1332134A2 (en) Inhibitors of prenyl-protein transferase
US6316436B1 (en) Inhibitors of prenyl-protein transferase
US6566385B2 (en) Inhibitors of prenyl-protein transferase
WO2002079147A2 (en) Inhibitors of prenyl-protein transferase
WO2002020015A1 (en) Inhibitors of prenyl-protein transferase
US6413964B1 (en) Inhibitors of prenyl-protein transferase
US6610722B2 (en) Inhibitors of prenyl-protein transferase
AU762440B2 (en) Inhibitors of prenyl-protein transferase
US20040110764A1 (en) Inhibitors of prenyl-protein transferase
WO2002078702A1 (en) Inhibitors of prenyl-protein transferase
US20020022633A1 (en) Inhibitors of prenyl-protein transferase
US6534506B2 (en) Inhibitors of prenyl-protein transferase
WO2001045704A1 (en) Inhibitors of prenyl-protein transferase
US6380228B1 (en) Inhibitors of prenyl-protein transferase

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PH PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP