Compounds useful for inhibition of farnesyl proteins transferase

Information

  • Patent Grant
  • 5861395
  • Patent Number
    5,861,395
  • Date Filed
    Thursday, September 11, 1997
    26 years ago
  • Date Issued
    Tuesday, January 19, 1999
    25 years ago
Abstract
Novel compounds of the formula: ##STR1## are disclosed. Compounds of Formula 1.0 are represented by the compounds of formulas: ##STR2## wherein R.sup.1, R.sup.3 and R.sup.4 are each independently selected from halo. Also disclosed are methods of inhibiting farnesyl protein transferase and the growth of abnormal cells, such as tumor cells.
Description

This application claims priority under 35 U.S.C. .sctn. 119(e) of provisional applications 60/026,008, filed Sep. 13, 1996 and 60/050,009, filed Jun. 17, 1997.
BACKGROUND
WO 95/10516, published Apr. 20, 1995 discloses tricyclic compounds useful for inhibiting farnesyl protein transferase.
In view of the current interest in inhibitors of farnesyl protein transferase, a welcome contribution to the art would be compounds useful for the inhibition of farnesyl protein transferase. Such a contribution is provided by this invention.
SUMMARY OF THE INVENTION
This invention provides compounds useful for the inhibition of farnesyl protein transferase (FPT). The compounds of this invention are represented by the formula: ##STR3## or a pharmaceutically acceptable salt or solvate thereof, wherein: one of a, b, c and d represents N or NR.sup.9 wherein R.sup.9 is O--, --CH.sub.3 or --(CH.sub.2).sub.n CO.sub.2 H wherein n is 1 to 3, and the remaining a, b, c and d groups represent CR.sup.1 or CR.sup.2 ; or
each of a, b, c, and d are independently selected from CR.sup.1 or CR.sup.2 ;
each R.sup.1 and each R.sup.2 is independently selected from H, halo, --CF.sub.3, --OR.sup.10 (e.g., --OCH.sub.3), --COR.sup.10, --SR.sup.10 (e.g., --SCH.sub.3 and --SCH.sub.2 C.sub.6 H.sub.5), --S(O).sub.t R.sup.11 (wherein t is 0, 1 or 2, e.g., --SOCH.sub.3 and --SO.sub.2 CH.sub.3), --SCN, --N(R.sup.10).sub.2, --NR.sup.10 R.sup.11, --NO.sub.2, --OC(O)R.sup.10, --CO.sub.2 R.sup.10, --OCO.sub.2 R.sup.11, --CN, --NHC(O)R.sup.10, --NHSO.sub.2 R.sup.10, --CONHR.sup.10, --CONHCH.sub.2 CH.sub.2 OH, --NR.sup.10 COOR.sup.11, ##STR4## --SR.sup.11 C(O)OR.sup.11 (e.g., --SCH.sub.2 CO.sub.2 CH.sub.3), --SR.sup.11 N(R.sup.75).sub.2 wherein each R.sup.75 is independently selected from H and --C(O)OR.sup.11 (e.g., --S(CH.sub.2).sub.2 NHC(O)O-t-butyl and --S(CH.sub.2).sub.2 NH.sub.2), benzotriazol-1-yloxy, tetrazol-5-yl-thio, or substituted tetrazol-5-ylthio (e.g., alkyl substituted tetrazol-5-ylthio such as 1-methyl-tetrazol-5-ylthio), alkynyl, alkenyl or alkyl, said alkyl or alkenyl group optionally being substituted with halo, --OR.sup.10 or --CO.sub.2 R.sup.10 ;
R.sup.3 and R.sup.4 are the same or different and each independently represents H, any of the substituents of R.sup.1 and R.sup.2, or R.sup.3 and R.sup.4 taken together represent a saturated or unsaturated C.sub.5 -C.sub.7 fused ring to the benzene ring (Ring III);
R.sup.5, R.sup.6, R.sup.7 and R.sup.8 each independently represents H, --CF.sub.3, --COR.sup.10, alkyl or aryl, said alkyl or aryl optionally being substituted with --OR.sup.10, --SR.sup.10, --S(O).sub.t R.sup.11, --NR.sup.10 COOR.sup.11, --N(R.sup.10).sub.2, --NO.sub.2, --COR.sup.10, --OCOR.sup.10, --OC.sub.2 R.sup.11, --CO.sub.2 R.sup.10, OPO.sub.3 R.sup.10, or R.sup.5 is combined with R.sup.6 to represent .dbd.O or .dbd.S and/or R.sup.7 is combined with R.sup.8 to represent .dbd.O or .dbd.S;
R.sup.10 represents H, alkyl, aryl, or aralkyl (e.g., benzyl);
R.sup.11 represents alkyl or aryl;
X represents N, CH or C, which C may contain an optional double bond (represented by the dotted line) to carbon atom 11;
the dotted line between carbon atoms 5 and 6 represents an optional double bond, such that when a double bond is present, A and B independently represent --R.sup.10, halo, --OR.sup.11, --OCO.sub.2 R.sup.11 or --OC(O)R.sup.10, and when no double bond is present between carbon atoms 5 and 6, A and B each independently represent H.sub.2, --(OR.sup.11).sub.2 ; H and halo, dihalo, alkyl and H, (alkyl).sub.2, --H and --OC(O)R.sup.10, H and --OR.sup.10, .dbd.O, aryl and H, .dbd.NOR.sup.10 or --O--(CH.sub.2).sub.p --O-- wherein p is 2, 3 or 4; and
W is selected from the group consisting of:
(1) cyano (i.e., CN);
(2) --C(O)R.sup.12 wherein R.sup.12 is selected from:
(a) a heteroaryl group, for example, pyridyl (e.g., 3-pyridyl), indolyl (e.g., 2-indolyl), pyrrolyl (e.g., 2-pyrrolyl) and N-substituted pyrrolyl (e.g., N-alkylpyrrolyl such as N-alkylpyrrolyl such as, N-alkylpyrrol-2-yl, such as, N-methylpyrrol-2-yl);
(b) H;
(c) alkyl (e.g., --CH.sub.3); or
(d) a substituent of the formula: ##STR5## wherein R.sup.28 is selected from --OC(O)R.sup.29, --OH, --OC(O)NHC(O)CCl.sub.3, or --OC(O)NH.sub.2, wherein R.sup.29 is alkyl (e.g., --CH.sub.3);
(3) an imidate represented by the formula: ##STR6## wherein R.sup.13 is selected from the group consisting of: (a) H, (b) CN, (c) --SO.sub.2 -alkyl (e.g., --SO.sub.2 CH.sub.3), (d) --C(O)-aryl (e.g., --C(O)C.sub.6 H.sub.5, i.e., --C(O)phenyl), (e) --SO.sub.2 NR.sup.10 R.sup.15 (e.g., --SO.sub.2 NH.sub.2), (f) --C(O)NR.sup.10 R.sup.15 (e.g., --C(O)NH.sub.2), (g) --OR.sup.10 (e.g., --OH and --OCH.sub.3); and (h) --C(O)NR.sup.10 C(O)NR.sup.10 R.sup.15 (e.g., --C(O)NHC(O)NH.sub.2); R.sup.14 is aryl; and R.sup.10 and R.sup.15 are independently selected from the group consisting of: H, alkyl, aryl and aralkyl;
(4) an imidamido (amidino) represented by the formula: ##STR7## wherein R.sup.13 is selected from the group consisting of (a)H, (b) CN, (c) --SO.sub.2 -allyl (e.g., --SO.sub.2 CH.sub.3), (d) --C(O)-aryl (e.g., --C(O)C.sub.6 H.sub.5, i.e., --C(O)phenyl), (e) --SO.sub.2 NR.sup.10 R.sup.15 (e.g., --SO.sub.2 NH.sub.2), (f) --C(O)NR.sup.10 R.sup.15 (e.g., --C(O)NH.sub.2), (g) --OR.sup.10 (e.g., --OH and --OCH.sub.3); and (h) --C(O)NR.sup.10 C(O)NR.sup.10 R.sup.15 (e.g., --C(O)NHC(O)NH.sub.2); R.sup.16 is selected from the group consisting of: alkyl, aralkyl, aryl, cycloalkyl, heteroaryl, heteroaralkyl and heterocycloalkyl; R.sup.10 and R.sup.15 are as defined above; and R.sup.10 and R.sup.16 are independently selected from the above defined groups;
(5) 1-amino-2-nitroethylene derivatives of the formula: ##STR8## wherein R.sup.10 is as defined above; and (6) a substituent of the formula: ##STR9##
The compounds of this invention: (i) potently inhibit farnesyl protein transferase, but not geranylgeranyl protein transferase I, in vitro; (ii) block the phenotypic change induced by a form of transforming Ras which is a farnesyl acceptor but not by a form of transforming Ras engineered to be a geranylgeranyl acceptor; (iii) block intracellular processing of Ras which is a farnesyl acceptor but not of Ras engineered to be a geranylgeranyl acceptor; and (iv) block abnormal cell growth in culture induced by transforming Ras.
The compounds of this invention inhibit farnesyl protein transferase and the farnesylation of the oncogene protein Ras. Thus, this invention further provides a method of inhibiting farnesyl protein transferase, (e.g., ras farnesyl protein transferase) in mammals, especially humans, by the administration of an effective amount of the tricyclic compounds described above. The administration of the compounds of this invention to patients, to inhibit farnesyl protein transferase, is useful in the treatment of the cancers described below.
This invention provides a method for inhibiting or treating the abnormal growth of cells, including transformed cells, by administering an effective amount of a compound of this invention. Abnormal growth of cells refers to cell growth independent of normal regulatory mechanisms (e.g., loss of contact inhibition). This includes the abnormal growth of: (1) tumor cells (tumors) expressing an activated Ras oncogene; (2) tumor cells in which the Ras protein is activated as a result of oncogenic mutation in another gene; and (3) benign and malignant cells of other proliferative diseases in which aberrant Ras activation occurs.
This invention also provides a method for inhibiting or treating tumor growth by administering an effective amount of the tricyclic compounds, described herein, to a mammal (e.g., a human) in need of such treatment. In particular, this invention provides a method for inhibiting or treating the growth of tumors expressing an activated Ras oncogene by the administration of an effective amount of the above described compounds. Examples of tumors which may be inhibited or treated include, but are not limited to, lung cancer (e.g., lung adenocarcinoma), pancreatic cancers (e.g., pancreatic carcinoma such as, for example, exocrine pancreatic carcinoma), colon cancers (e.g., colorectal carcinomas, such as, for example, colon adenocarcinoma and colon adenoma), myeloid leukemias (for example, acute myelogenous leukemia (AML)), thyroid follicular cancer, myelodysplastic syndrome (MDS), bladder carcinoma, epidermal carcinoma, breast cancer and prostate cancer.
It is believed that this invention also provides a method for inhibiting or treating 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 or treatment being accomplished by the administration of an effective amount of the tricyclic compounds described herein, to a mammal (e.g., a human) in need of such treatment. For example, the benign proliferative disorder neurofibromatosis, or tumors in which Ras is activated due to mutation or overexpression of tyrosine kinase oncogenes (e.g., neu, src, abl, lck, and fyn), may be inhibited or treated by the tricyclic compounds described herein.
The tricyclic compounds useful in the methods of this invention inhibit or treat the abnormal growth of cells. Without wishing to be bound by theory, it is believed that these compounds may function through the inhibition of G-protein function, such as ras p21, by blocking G-protein isoprenylation, thus making them useful in the treatment of proliferative diseases such as tumor growth and cancer. Without wishing to be bound by theory, it is believed that these compounds inhibit ras farnesyl protein transferase, and thus show antiproliferative activity against ras transformed cells.





DETAILED DESCRIPTION OF THE INVENTION
As used herein, the following terms are used as defined below unless otherwise indicated:
Ac--represents acetyl;
MH.sup.+ --represents the molecular ion plus hydrogen of the molecule in the mass spectrum;
M.sup.+ --represents the molecular ion of the molecule in the mass spectrum;
benzotriazol-1-yloxy represents ##STR10## 1-methyl-tetrazol-5-ylthio represents ##STR11## acyl-represents represents --C(O)-alkyl, --C(O)-alkenyl, --C(O)-alkynyl, --C(O)-cycloalkyl, --C(O)-cycloalkenyl or --C(O)-cycloalknyl;
alkenyl--represents straight and branched carbon chains having at least one carbon to carbon double bond and containing from 2 to 12 carbon atoms, preferably from 2 to 6 carbon atoms and most preferably from 3 to 6 carbon atoms;
alkynyl-represents straight and branched carbon chains having at least one carbon to carbon triple bond and containing from 2 to 12 carbon atoms, preferably from 2 to 6 carbon atoms;
alkyl-(including the alkyl portions of alkoxy, aralkyl and heteroarylalkyl)-represents straight and branched carbon chains and contains from one to twenty carbon atoms, preferably one to six carbon atoms;
aralkyl-represents an aryl group, as defined below, bound to an alkyl group, as defined above, preferably the alkyl group is --CH.sub.2 --, (e.g., benzyl);
aryl (including the aryl portion of aralkyl, aryloxy and arylalkyloxy)-represents a carbocyclic group containing from 6 to 15 carbon atoms and having at least one aromatic ring (e.g., aryl is a phenyl ring), with all available substitutable carbon atoms of the carbocyclic group being intended as possible points of attachment, said carbocyclic group being optionally substituted (e.g., 1 to 3) with one or more of halo, alkyl, hydroxy, alkoxy, phenoxy, CF.sub.3, amino, alkylamino, dialkylamino, --COOR.sup.10 or --NO.sub.2 ;
cycloalkyl-represents saturated carbocyclic rings branched or unbranched of from 3 to 20 carbon atoms, preferably 3 to 7 carbon atoms;
Et--represents ethyl;
halo--represents fluoro, chloro, bromo and iodo;
heteroaryl--represents cyclic groups, optionally substituted with R.sup.3 and R.sup.4, having at least one heteroatom selected from O, S or N, said heteroatom interrupting a carbocyclic ring structure and having a sufficient number of delocalized pi electrons to provide aromatic character, with the aromatic heterocyclic groups preferably containing from 2 to 14 carbon atoms, e.g., (1) thienyl (e.g., 2- or 3-thienyl), (2) imidazolyl (e.g., (2-, 4- or 5-) imidazolyl), (3) triazolyl (e.g., 3- or 5- ��1,2,4-triazolyl!), (4) tetrazolyl, (5) substituted tetrazolyl, such as ##STR12## �wherein R.sup.27 represents (a) aryl (e.g., phenyl), (b) alkyl (e.g., --CH.sub.3) or (c) arylalkyl (aralkyl) (e.g., benzyl)!, (6) furyl (e.g., 2- or 3-furyl), (7) thiazolyl (or thiazyl) (e.g., 2-, 4- or 5-thiazolyl), (8) pyrimidinyl (e.g., 2-, 4- or 5-pyrimidinyl), (9) pyrazinyl (e.g., 2-pyrazinyl), (10) pyridazinyl (e.g., 3- or 4-pyridazinyl), (11) triazinyl (e.g., 2-, 4- or 6-�1,3,5-triazinyl!), (12) 3- or 5-�1,2,4-thiadiazolyl!, (13) 2-, 3-, 4-, 5-, 6- or 7-benzofuranyl, (14) benzoxazolyl (e.g., 2-, 4-, 5-, 6- or 7-benzoxazolyl), (15) indolyl (benzopyrrolyl) (e.g., 2-, 3-, 4-, 5-, 6- or 7-indolyl), (16) pyrazolyl (e.g., 3-, 4- or 5-pyrazolyl), (17) oxazolyl (e.g., 2-, 4- or 5-oxazolyl), (18) 2-, 3- or 4-pyridyl or pyridyl N-oxide (optionally substituted with R.sup.3 and R.sup.4), wherein pyridyl N-oxide can be represented as: ##STR13## (19) benzisoxazolyl, (20) pyrrolyl, (21) benzimidazolyl, (22) isoquinolinyl, (23) quinolinyl, (24) pyridopyrazinyl, (25) naphthyridinyl, (26) pyranyl, (27) benzothienyl, (28) isobenzofuranyl, (29) isothiazolyl, (30) isoxazolyl and (31) an N-substituted pyrrolyl ##STR14## wherein R.sup.30 is selected from: alkyl (e.g., C.sub.2 H.sub.5), aralkyl (e.g., --CH.sub.2 C.sub.6 H.sub.5), --CH.sub.2 C(O)NH.sub.2, --SO.sub.2 CH.sub.3, or --CH.sub.2 C(O)OR.sup.31 wherein R.sup.31 is H or alkyl (e.g., --C(CH.sub.3).sub.3);
heteroarylalkyl (heteroaralkyl)--represents a heteroaryl group, as defined above, bound to an alkyl group, as defined above, preferably the alkyl group is --CH.sub.2 --, for example, --CH.sub.2 --(4-or 5-)imidazolyl;
heterocycloalkyl--represents a saturated, branched or unbranched carbocylic ring containing from 3 to 15 carbon atoms, preferably from 4 to 6 carbon atoms, which carbocyclic ring is interrupted by 1 to 3 hetero groups selected from --O--, --S--, --NR.sup.10 -- (wherein R.sup.10 is as defined above); suitable heterocycloalkyl groups include: (1) tetrahydrofuranyl (e.g., 2- or 3-tetrahydrofuranyl), (2) tetrahydrothienyl e.g., (2- or 3-tetrahydrothienyl), (3) piperidinyl (e.g., 2-, 3- or 4-piperidinyl), (4) pyrrolidinyl (e.g., 2- or 3-pyrrolidinyl), (5) 2- or 3-piperizinyl, (6) 2- or 4-dioxanyl, (7) tetrahydopyranyl, and (8) substituted tetrahydropyranyl wherein said substituents are selected from hydroxy and hydroxyalkyl (e.g., hydroxymethyl), for example ##STR15## Ph--represents phenyl.
The following solvents and reagents are referred to herein by the abbreviations indicated: tetrahydrofuran (THF); isopropanol (iPrOH); ethanol (EtOH); methanol (MeOH); acetic acid (HOAc or AcOH); ethyl acetate (EtOAc); N,N-dimethylformamide (DMF); trifluoroacetic acid (TFA); trifluoroacetic anhydride (TFAA); 1-hydroxybenzo-triazole (HOBT); 1-(3-dimethylaminopropyl)-3-ethyl carbodiimide hydrochloride (DEC); diisobutylaluminum hydride (DIBAL); and 4-methylmorpholine (NMM).
Reference to the position of the substituents R.sup.1, R.sup.2, R.sup.3, and R.sup.4 is based on the numbered ring structure: ##STR16##
Those skilled in the art will also appreciate that the S and R stereochemistry at the C-11 bond are: ##STR17##
Compounds of Formula 1.0 include compounds wherein the bottom piperidinyl group is a 4- or 3-piperidinyl group, i.e., ##STR18##
Compounds of Formula 1.0 include compounds wherein R.sup.2 and R.sup.4 are H, and R.sup.1 and R.sup.3 are halo (preferably independently selected from Br or Cl). For example, R.sup.1 is Br and R.sup.3 is Cl. These compounds include compounds wherein R.sup.1 is in the 3-position and R.sup.3 is in the 8-position, e.g., 3-Br and 8-Cl. Compounds of Formula 1.0 also include compounds wherein R.sup.2 is H, and R.sup.1, R.sup.3 and R.sup.4 are halo (preferably independently selected from Br or Cl).
Preferably, compounds of Formula 1.0 are represented by compounds of Formula 1.1: ##STR19## wherein all substituents are as defined for Formula 1.0.
Preferably, R.sup.2 is H and R.sup.1, R.sup.3 and R.sup.4 are halo; a is N and b, c and d are carbon; A and B are each H.sub.2 ; the optional bond between C5 and C6 is absent; X is CH; and R.sup.5, R.sup.6, R.sup.7 and R.sup.8 are H. More preferably, R.sup.1, R.sup.3 and R.sup.4 are independently selected from Br or Cl. Most preferably, R.sup.1 is Br, and R.sup.3 and R.sup.4 are independently selected from Cl and Br.
More preferably, compounds of Formula 1.0 are represented by compounds of Formula 1.2 and Formula 1.3: ##STR20## and most preferably, compounds of Formulas 1.4 and 1.5 ##STR21## wherein R.sup.1, R.sup.3 and R.sup.4 are each independently selected from halo, preferably, Br or Cl; and A, B, X and W are as defined for Formula 1.0. More preferably, A and B are each H.sub.2 ; the optional bond between C5 and C6 is absent; and X is CH. Most preferably, R.sup.1 is Br; R.sup.3 and R.sup.4 are independently Br or Cl, and still more preferably R.sup.3 is Cl and R.sup.4 is Br; A and B are each H.sub.2 ; the optional bond between C5 and C6 is absent; X is CH; and R.sup.5, R.sup.6, R.sup.7 and R.sup.8 are H.
Examples of --C(O)R.sup.12 substituents for W include groups wherein R.sup.12 is selected from the group consisting of: ##STR22##
Examples of imidates for substituent W include groups wherein R.sup.13 is selected from the group consisting of: (1) CN; (2) H; (3) --SO.sub.2 NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are selected from the group consisting of: H and alkyl (e.g., methyl); (4) --C(O)NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are selected from the group consisting of: H and alkyl (e.g., methyl); (5) --SO.sub.2 -alkyl; and (6) --C(O)-aryl. Examples of imidates also include groups wherein R.sup.14 is aryl (e.g., phenyl).
For example, imidates for substituent W include groups wherein R.sup.13 is selected from the group consisting of: CN, --C(O)NH.sub.2, H, --SO.sub.2 NH.sub.2, --SO.sub.2 NHCH.sub.3, --SO.sub.2 N(CH.sub.3).sub.2, --C(O)NHCH.sub.3, --SO.sub.2 CH.sub.3 and --C(O)C.sub.6 H.sub.5. Examples of imidates also include groups wherein R.sup.14 is phenyl; and R.sup.13 is selected from the group consisting of: CN, --C(O)NH.sub.2, H, --SO.sub.2 NH.sub.2, --SO.sub.2 NHCH.sub.3, --SO.sub.2 N(CH.sub.3).sub.2, --C(O)NH.sub.2, --C(O)NHCH.sub.3, --SO.sub.2 CH.sub.3 and --C(O)C.sub.6 H.sub.5.
Examples of imidamidos for substituent W include groups wherein R.sup.13 is selected from the group consisting of: (1) CN; (2) H; (3) --OR.sup.10 ; (4) --C(O)NR.sup.10 C(O)NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are H; (5) --SO.sub.2 NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl (e.g., methyl), (6) --C(O)NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl (e.g., methyl); (7) --SO.sub.2 -alkyl; and (8) --C(O)-aryl. Examples of the imidamidos also include groups wherein R.sup.10 and R.sup.15 are selected from the group consisting of: H and alkyl (e.g., methyl); and wherein R.sup.16 is selected from the group consisting of: H and heteroaralkyl (e.g., 3-pyridylmethyl).
For example, imidamidos for substituent W include groups wherein R.sup.13 is selected from the group consisting of: CN, H, --OCH.sub.3, --OH, --C(O)NHC(O)NH.sub.2, --SO.sub.2 NH.sub.2, --SO.sub.2 NHCH.sub.3, --SO.sub.2 N(CH.sub.3).sub.2, --C(O)NH.sub.2, --C(O)NHCH.sub.3, --SO.sub.2 CH.sub.3 and --C(O)C.sub.6 H.sub.5. Examples of imidamidos also include groups wherein R.sup.10 and R.sup.16 are selected from the group consisting of: H and ##STR23## (i.e., 3-pyridylmethyl).
Examples of the imidamido substituents additionally include groups wherein: R.sup.10 and R.sup.16 are selected from the group consisting of: H and 3-pyridylmethyl; and R.sup.13 is selected from the group consisting of: CN, H, --OCH.sub.3, --OH, --C(O)NHC(O)NH.sub.2, --SO.sub.2 NH.sub.2, --SO.sub.2 NHCH.sub.3, --SO.sub.2 N(CH.sub.3).sub.2, --C(O)NH.sub.2, --C(O)NHCH.sub.3, --SO.sub.2 CH.sub.3 and --C(O)C.sub.6 H.sub.5.
In addition, examples of the imidamido substituents additionally include groups wherein: (1) R.sup.13 and R.sup.10 are H, and R.sup.16 is 3-pyridylmethyl; and (2) R.sup.10 and R.sup.16 are H, and R.sup.13 is selected from the group consisting of: CN, H, --OCH.sub.3, --OH, --C(O)NHC(O)NH.sub.2, --SO.sub.2 NH.sub.2, -and --C(O)NHCH.sub.3, --SO.sub.2 CH.sub.3 and --C(O)C.sub.6 H.sub.5.
Examples of 1-amino-2-nitroethylene derivatives for substituent W include groups wherein R.sup.10 is alkyl, e.g., methyl.
Compounds of Formulas 1.2A and 1.3A: ##STR24## are preferred when X is CH or N, and R.sup.1, R.sup.3 and R.sup.4 are halo.
The preferred compounds of this invention are represented by the compounds of Formulas: ##STR25## wherein R.sup.1, R.sup.3 and R.sup.4 are halo and the remaining substituents are as defined above, with the compounds of Formula 1.5A being more preferred.
Representative compounds of Formula 1.0 wherein W is an imidate include: ##STR26##
Representative compounds of Formula 1.0 wherein W is an imidate also include: ##STR27##
Representative compounds of formula 1.0 wherein W is an imidamido include: ##STR28##
Representative compounds of Formula 1.0 wherein W is an imidamido also include: ##STR29##
Representative compounds of Formula 1.0 wherein W is cyano include: ##STR30##
Representative compounds of Formula 1.0 wherein W is cyano also include: ##STR31##
Representative compounds of Formula 1.0 wherein W is a 1-amino-2-nitroethylene derivative include: ##STR32##
Representative compounds of Formula 1.0 wherein W is a 1-amino-2-nitroethylene derivative also include: ##STR33##
Compounds of Formula 1.0 wherein W is a --C(O)R.sup.12 group include: ##STR34##
Compounds of Formula 1.0 also include a compound of the formula: ##STR35##
The compounds of this invention also include the 1-N-oxides--i.e, for example, compounds of the the formula: ##STR36## wherein ##STR37## represents the remainder of the compound, or pharmaceutically acceptable salts or solvates thereof.
Optical rotation of the compounds ((+)- or (-)-) are measured in methanol or ethanol at 25.degree. C.
This invention includes the above compounds in the amorphous state or in the cyrstalline state.
Lines drawn into the ring systems indicate that the indicated bond may be attached to any of the substitutable ring carbon atoms.
Certain compounds of the present invention may exist in different isomeric forms (e.g., enantiomers or diastereoisomers) including atropisomers (i.e., compounds wherein the 7-membered ring is in a fixed conformation such that the 11-carbon atom is positioned above or below the plane of the fused beznene rings due to the presence of a 10-bromo substituent). The invention contemplates all such isomers both in pure form and in admixture, including racemic mixtures. Enol forms are also included.
Certain tricyclic compounds will be acidic in nature, e.g. those compounds which possess a carboxyl or phenolic hydroxyl group. These compounds may form pharmaceutically acceptable salts. Examples of such salts may include sodium, potassium, calcium, aluminum, gold and silver salts. Also contemplated are salts formed with pharmaceutically acceptable amines such as ammonia, alkyl amines, hydroxyalkylamines, N-methylglucamine and the like.
Certain basic tricyclic compounds also form pharmaceutically acceptable salts, e.g., acid addition salts. For example, the pyrido-nitrogen atoms may form salts with strong acid, while compounds having basic substituents such as amino groups also form salts with weaker acids. Examples of suitable acids for salt formation are hydrochloric, sulfuric, phosphoric, acetic, citric, oxalic, malonic, salicylic, malic, fumaric, succinic, ascorbic, maleic, methanesulfonic and other mineral and carboxylic acids well known to those in the art. The salts are prepared by contacting the free base form with a sufficient amount of the desired acid to produce a salt in the conventional manner. The free base forms may be regenerated by treating the salt with a suitable dilute aqueous base solution such as dilute aqueous NaOH, potassium carbonate, ammonia and sodium bicarbonate. The free base forms differ from their respective salt forms somewhat in certain physical properties, such as solubility in polar solvents, but the acid and base salts are otherwise equivalent to their respective free base forms for purposes of the invention.
All such acid and base salts are intended to be pharmaceutically acceptable salts within the scope of the invention and all acid and base salts are considered equivalent to the free forms of the corresponding compounds for purposes of the invention.
Compounds of the invention may be prepared according to the procedures described in WO 95/10516 published Apr. 20, 1995, application Ser. No. 08/410,187 filed Mar. 24, 1995, application Ser. No. 08/577,951 filed Dec. 22, 1995 (now abandoned), application Ser. No. 08/615,760 filed Mar. 13, 1996 (now abandoned), WO 97/23478 published Jul. 3, 1997 which discloses the subject matter of Ser. Nos. 08/577,951 and 08/615,760, application Ser. No. 08/713297 filed Sep. 13, 1996, and application Ser. No. 08/877,453 filed Jun. 17, 1997; the disclosures of each being incorporated herein by reference thereto; and according to the procedures described below.
Compounds of the invention can be prepared by reacting a compound of the formula: ##STR38## wherein all substituents are as defined for Formula 1.0, with the appropriate protected piperidinyl acetic acid (e.g., 1-N-t-butoxy-carbonylpiperidinyl acetic acid) together with DEC/HOBT/NMM in DMF at 25.degree. C. for about 18 hours to produce a compound of the formula: ##STR39## The compound of Formula 44.0 is then reacted either with TFA or 10% sulfuric acid in dioxane and methanol, followed by NaOH to produce the compound of formula: ##STR40##
For example, the compound of formula ##STR41## can be prepared by reaction of a compound of Formula 43.0 with 1-N-t-butoxy-carbonylpiperidinyl-4-acetic acid as described above.
For Example, compounds of Formula 46.0 include: ##STR42## The preparation of these compounds are described in Preparative Examples 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 and 13, respectively, below.
The compounds of the invention can be prepared by reacting a compound of the formula: ##STR43## with the appropriate protected piperidinyl acetic acid (e.g., 1-N-t-butoxycarbonylpiperidinyl acetic acid together with DEC/HOBT/NMM in DMF at about 25.degree. C. for about 18 hours to produce a compound of the formula: ##STR44## The compound of Formula 44.1 is then reacted either with TFA or 10% sulfuric acid in dioxane and methanol followed by NaOH to produce the compound of Formula 46.1 ##STR45##
The amide compounds of this invention, represented by Formula 1.7 ##STR46## can be prepared by reacting the compound of Formula 46.1 with the appropriate carboxylic acid in the presence of a coupling agent such as DEC and HOBT in dimethylformamide. Alternatively, the compound of Formula 46.1 can be reacted with an acid chloride or anhydride in a solvent such as pyridine.
Compounds having an 1-N-O group: ##STR47## can be prepared from the corresponding pyridyl compounds: ##STR48## by oxidation with meta-chloroperoxybenzoic acid. This reaction is conducted in a suitable organic solvent, e.g., dichloromethane (usually anhydrous) or methylene chloride, at a suitable temperature, to produce the compounds of the invention having the N-O substituent at position 1 of Ring I of the tricyclic ring system.
Generally, the organic solvent solution of the starting tricyclic reactant is cooled to about 0.degree. C. before the m-chloroper-oxybenzoic acid is added. The reaction is then allowed to warm to room temperature during the reaction period. The desired product can be recovered by standard separation means. For example, the reaction mixture can be washed with an aqueous solution of a suitable base, e.g., saturated sodium bicarbonate or NaOH (e.g., 1N NaOH), and then dried over anhydrous magnesium sulfate. The solution containing the product can be concentrated in vacuo. The product can be purified by standard means, e.g., by chromatography using silica gel (e.g., flash column chromatography).
Alternatively, N-O compounds can be made from intermediate: ##STR49## by the above oxidation procedure with m-chloroperoxybenzoic acid and ##STR50## wherein Q is a protecting group, e.g., BOC. After oxidation the protecting group is removed by techniques well known in the art. The N-O intermediate are then reacted further to produce the compounds of the invention.
Compounds of Formula 43.0 include the compound of Formula 43.1: ##STR51## The compound of Formula 43.1 is prepared by methods known in the art, for example by methods disclosed in WO 95/10516, in U.S. Pat. No. 5,151,423 and those described below. The above intermediate compound can also be prepared by a procedure comprising the following steps:
(a) reacting an amide of the formula ##STR52## wherein R.sup.11a is Br, R.sup.5a is hydrogen and R.sup.6a is C.sub.1 -C.sub.6 alkyl, aryl or heteroaryl; R.sup.5a is C.sub.1 -C.sub.6 alkyl, aryl or heteroaryl and R.sup.6a is hydrogen; R.sup.5a and R.sup.6a are independently selected from the group consisting of C.sub.1 -C.sub.6 alkyl and aryl; or R.sup.5a and R.sup.6a, together with the nitrogen to which they are attached, form a ring comprising 4 to 6 carbon atoms or comprising 3 to 5 carbon atoms and one hetero moiety selected from the group consisting of --O-- and --NR.sup.9a --, wherein R.sup.9a is H, C.sub.1 -C.sub.6 alkyl or phenyl;
with a compound of the formula ##STR53## wherein R.sup.1a, R.sup.2a, R.sup.3a and R.sup.4a are are independently selected from the group consisting of hydrogen and halo and R.sup.7a is Cl or Br, in the presence of a strong base to obtain a compound of the formula ##STR54## (b) reacting a compound of step (a) with (i) POCl.sub.3 to obtain a cyano compound of the formula ##STR55## (ii) DIBALH to obtain an aldehyde of the formula ##STR56## (c) reacting the cyano compound or the aldehyde with a a piperidine derivative of the formula ##STR57## wherein L is a leaving group selected from the group consisting of Cl and Br, to obtain a ketone or an alcohol of the formula below, respectively: ##STR58## (d)(i) cyclizing the ketone with CF.sub.3 SO.sub.3 H to obtain a compound of Formula 13.0a wherein the dotted line represents a double bond; or
(d)(ii) cyclizing the alcohol with polyphosphoric acid to obtain an Intermediate compound wherein the dotted line represents a single bond.
Methods for preparing the Intermediate compounds disclosed in WO 95/10516, U.S. Pat. No. 5,151,423 and described below employ a tricyclic ketone intermediate. Such intermediates of the formula ##STR59## wherein R.sup.11b, R.sup.1a, R.sup.2a, R.sup.3a and R.sup.4a are independently selected from the group consisting of hydrogen and halo, can be prepared by the following process comprising:
(a) reacting a compound of the formula ##STR60## (i) with an amine of the formula NHR.sup.5a R.sup.6a, wherein R.sup.5a and R.sup.6a are as defined in the process above; in the presence of a palladium catalyst and carbon monoxide to obtain an amide of the formula: ##STR61## (ii) with an alcohol of the formula R.sup.10a OH, wherein R.sup.10a is C.sub.1 -C.sub.6 lower alkyl or C.sub.3 -C.sub.6 cycloalkyl, in the presence of a palladium catalyst and carbon monoxide to obtain the ester of the formula ##STR62## followed by reacting the ester with an amine of formula NHR.sup.5a R.sup.6a to obtain the amide;
(b) reacting the amide with an iodo-substituted benzyl compound of the formula ##STR63## wherein R.sup.1a, R.sup.2a, R.sup.3a, R.sup.4a and R.sup.7a are as defined above, in the presence of a strong base to obtain a compound of the formula ##STR64## (c) cyclizing a compound of step (b) with a reagent of the formula R.sup.8a MgL, wherein R.sup.8a is C.sub.1 -C.sub.8 alkyl, aryl or heteroaryl and L is Br or Cl, provided that prior to cyclization, compounds wherein R.sup.5a or R.sup.6a is hydrogen are reacted with a suitable N-protecting group.
(+)-Isomers of compounds of Formula 43.2 ##STR65## can be prepared with high enantioselectivity by using a process comprising enzyme catalyzed transesterification. Preferably, a racemic compound of Formula 43.3 ##STR66## is reacted with an enzyme such as Toyobo LIP-300 and an acylating agent such as trifluoroethly isobutyrate; the resultant (+)-amide is then isolated from the (-)-enantiomeric amine by techniques well known in the art, and then the (+)-amide is hydrolyzed, for example by refluxing with an acid such as H.sub.2 SO.sub.4, and the resulting compound is then reduced with DIBAL by techniques well known in the art to obtain the corresponding optically enriched (+)-isomer of Formula 43.2. Alternatively, a racemic compound of Formula 43.3, is first reduced to the corresponding racemic compound of Formula 43.2 and then treated with the enzyme (Toyobo LIP-300) and acylating agent as described above to obtain the (+)-amide, which is hydrolyzed to obtain the optically enriched (+)-isomer.
Those skilled in the art will appreciate that compounds of Formula 1.0 having other R.sup.1, R.sup.2, R.sup.3, and R.sup.4 substituents may be made by the above enzyme process.
To produce compounds of Formula 1.0 wherein W is an imidate, a compound of Formula 45.0 or 46.0 is reacted with ##STR67## in the presence of a suitable solvent (e.g., isopropanol) and at a suitable temperature (e.g., about 80.degree. C.) for a suitable period of time (e.g., about 24 hours).
For example, a compound of Formula 46.0, wherein X is N, can be reacted with ##STR68## in isopropanol at 80.degree. C. for 24 hours to produce a compound of Formula 1.0 wherein R.sup.13 is CN and R.sup.14 is phenyl. Similarly, reaction with ##STR69## (M. Haake and B. Schummelfeder, Synthesis, pp. 753-758, September 1991) can produce compounds of Formula 1.0 wherein R.sup.13 is --SO.sub.2 NH.sub.2 and R.sup.14 is phenyl.
Compounds of Formula 1.0 wherein R.sup.13 is H and R.sup.14 is phenyl can be produced by reacting a compound of Formula 45.0 or 46.0 with (e.g., wherein X is CH) with CNOC.sub.6 H.sub.5 in diisopropylethylamine according to the procedure disclosed in Zweifel et al., Synthesis, p. 150 (1980).
Compounds of Formula 1.0 wherein W is --CN can be prepared by reacting a compound of Formula 1.0, wherein W is an imidate, with a strong base, such as NaH, in a suitable solvent, such as dichloromethane. For example, compounds of Formula 1.0 wherein W is an imidate and R.sup.13 is H and R.sup.14 is phenyl and X is CH can be reacted with NaH in dichloromethane to produce compounds of Formula 1.0 wherein W is --CN.
Compounds of Formula 1.0, wherein W is an imidamido group, can be produced by reacting a compound of Formula 1.0, wherein W is an imidate group, with concentrated NH.sub.4 OH in a suitable solvent (e.g., isopropanol) at a suitable temperature (e.g., about 25.degree. C.) for a suitable amount of time (e.g., about 24 hours). For example, an imidate compound of Formula 1.0, wherein R.sup.13 is --CN and R.sup.14 is phenyl and X is N, can be reacted with concentrated NH.sub.4 OH in isopropanol at 25.degree. C. for 24 hours to produce the corresponding imidamido wherein R.sup.13 is --CN and both R.sup.10 and R.sup.16 are H.
Compounds of Formula 1.0, wherein W is an imidamido group, can also be produced as a hydrochloride salt by reacting a compound of Formula 1.0, wherein W is an imidate group, with concentrated NH.sub.4 OH, NH.sub.4 Cl, and water in a suitable pressure vessel at a suitable temperature (e.g., about 87.degree. C.). For example, an imidate of Formula 1.0 wherein X is CH, R.sup.13 is H and R.sup.14 is phenyl can be reacted with concentrated NH.sub.4 OH, NH.sub.4 Cl, and water in a suitable pressure vessel at about 87.degree. C. to produce the corresponding imidamido hydrochloride salt wherein R.sup.13 is H and both R.sup.10 and R.sup.16 are H.
Compounds of Formula 1.0, wherein W is an imidamido group, can also be produced by reacting a compound of Formula 1.0, wherein W is an imidate group, with an amine (e.g., HNR.sup.10 R.sup.16) in a suitable solvent (e.g., THF) at a suitable temperature (e.g., about 70.degree. C.). For example, an imidate of Formula 1.0 wherein X is CH and R.sup.13 is H and R.sup.14 is phenyl can be reacted with ##STR70## (3-aminomethylpyridine) in THF at about 70.degree. C. to produce the corresponding guanidine wherein R.sup.13 is H and R.sup.10 is H and R.sup.16 is 3-methylpyridyl.
Compounds of Formula 1.0, wherein W is an imidamido group, can also be prepared by fusing an imidate of Formula 1.0 with sulfamide. For example, an imidate of Formula 1.0 wherein R.sup.13 is H and R.sup.14 is phenyl and X is N can be fused with sulfamide to produce the corresponding guanidine wherein R.sup.17 is --SO.sub.2 NH.sub.2 and both R.sup.18 and R.sup.19 are H.
Compounds of Formula 1.0, wherein W is an imidamido group, can also be prepared by fusing a cyanide of Formula 1.0 (e.g., X is N) with sulfamide to produce the corresponding imidamido wherein R.sup.13 is --SO.sub.2 NH.sub.2 and both R.sup.10 and R.sup.16 are H.
Compounds of Formula 1.0, wherein W is an imidate group and R.sup.13 is an N-methylsulfamoyl group, can also be prepared by reacting an imidate of Formula 1.0, wherein R.sup.13 is H, with a sulfamoyl chloride in a suitable solvent (e.g., acetonitrile, benzene or toluene) with a suitable base (e.g., triethylamine) at a suitable temperature (e.g., 0.degree. to about 25.degree. C.). For example, an imidate of Formula 1.0 wherein R.sup.13 is H and R.sup.14 is phenyl and X is N can be reacted with N-methylsulfamoyl chloride or dimethylsulfamoyl chloride to produce the corresponding imidate wherein R.sup.13 is --SO.sub.2 NHCH.sub.3 or --SO.sub.2 N(CH.sub.3).sub.2, respectively, and R.sup.14 is phenyl.
Compounds of Formula 1.0, wherein W is an imidamido can also be prepared by reacting an imidate of Formula 1.0 with an aqueous solution of hydroxylamine hydrochloride and sodium hydroxide, or an aqueous solution of methoxylamine and sodium hydroxide, to produce an imidamido wherein R.sup.13 is --OH or --OCH.sub.3, respectively, and both R.sup.10 and R.sup.16 are H. The imidate of Formula 1.0 used, for example, can be one wherein X can be N, R.sup.13 can be H and R.sup.14 can be phenyl.
Compounds of Formula 1.0, wherein W is an imidate group and R.sup.13 is --C(O)NH.sub.2 or --C(O)NHCH.sub.3, can also be prepared by reacting an imidate of Formula 1.0, wherein R.sup.13 is H, with trimethylsilylisocyanate or methylisocyanate, respectively, in a suitable solvent (e.g., anhydrous dichloromethane) at a suitable temperature (e.g., about 25.degree. C.) for a suitable time period (about 48 hours). The imidate of Formula 1.0 used, for example, can be one wherein X can be N, R.sup.13 can be H and R.sup.14 can be phenyl.
Compounds of Formula 1.0, wherein W is an imidamido can also be prepared by fusing an imidate of Formula 1.0 with urea to produce an imidamido wherein R.sup.13 is --C(O)NH.sub.2, and both R.sup.10 and R.sup.16 are H. The imidate of Formula 1.0 used, for example, can be one wherein X can be N, R.sup.13 can be H and R.sup.14 can be phenyl.
The compounds of Formula 1.0 wherein W is 1-amino-2-nitroethylene can be prepared by reacting a compound of Formula 45.0 or 46.0 with 1-methylthio-1-methylamino-2-nitroethene, N(C.sub.2 H.sub.5).sub.3 and CuCl according to the procedures disclosed in Canada Patent No. 1178289 (1984).
The compounds of Formula 1.0 wherein W is -amino-2-nitroethylene can also be prepared by reaction of a compound of ##STR71## in methanol with methyl iodide at reflux, and then refluxing the resulting product with nitromethane (see Indian J. Chem., 15B, 297 (1977)).
The compounds of Formula 1.0 wherein W is --C(O)R.sup.12 may be prepared by reacting a compound of formula 45.0 or 46.0 with the appropriate carboxylic acid, HOOCR.sup.12, and HOBT/DEC/DMF.
Compounds of the invention are exemplified by the following examples, which should not be construed to limit the scope of the disclosure.
PREPARATIVE EXAMPLE 1 ##STR72## Step A: ##STR73##
Combine 10 g (60.5 mmol) of ethyl 4-pyridylacetate and 120 mL of dry CH.sub.2 Cl.sub.2 at -20.degree. C., add 10.45 g (60.5 mmol) of MCPBA and stir at -20.degree. C. for 1 hour and then at 25.degree. C. for 67 hours. Add an additional 3.48 g (20.2 mmoles) of MCPBA and stir at 25.degree. C. for 24 hours. Dilute with CH.sub.2 Cl.sub.2 and wash with saturated NaHCO.sub.3 (aqueous) and then water. Dry over MgSO.sub.4, concentrate in vacuo to a residue, and chromatograph (silica gel, 2%-5.5% (10% NH.sub.4 OH in MeOH)/CH.sub.2 Cl.sub.2)to give 8.12 g of the product compound. Mass Spec.: MH.sup.+ =182.15
Step B: ##STR74##
Combine 3.5 g (19.3 mmol) of the product of Step A, 17.5 mL of EtOH and 96.6 mL of 10% NaOH (aqueous) and heat the mixture at 67.degree. C. for 2 hours. Add 2N HCl (aqueous) to adjust to pH=2.37 and concentrate in vacuo to a residue. Add 200 mL of dry EtOH, filter through celite.RTM. and wash the filter cake with dry EtOH (2.times.50 ml). Concentrate the combined filtrates in vacuo to give 2.43 g of the title compound.
PREPARATIVE EXAMPLE 2 ##STR75##
The title compound is prepared via the process disclosed in PCT International Publication No. WO95/10516.
PREPARATIVE EXAMPLE 3 ##STR76## Step A: ##STR77##
Combine 14.95 g (39 mmol) of 8-chloro-11-(1-ethoxy-carbonyl-4-piperidinyl)-11H-benzo�5,6!cyclohepta�1,2-b!pyridine and 150 mL of CH.sub.2 Cl.sub.2, then add 13.07 g (42.9 mmol) of (nBu).sub.4 NNO.sub.3 and cool the mixture to 0.degree. C. Slowly add (dropwise) a solution of 6.09 mL (42.9 mmol) of TFAA in 20 mL of CH.sub.2 Cl.sub.2 over 1.5 hours. Keep the mixture at 0.degree. C. overnight, then wash successively with saturated NaHCO.sub.3 (aqueous), water and brine. Dry the organic solution over Na.sub.2 SO.sub.4, concentrate in vacuo to a residue and chromatograph the residue (silica gel, EtOAc/hexane gradient) to give 4.32 g and 1.90 g of the two product compounds 3A(i) and 3A(ii), respectively.
Mass Spec. for compound 3A(i): MH.sup.+ =428.2;
Mass Spec. for compound 3A(ii): MH.sup.+ =428.3.
Step B: ##STR78##
Combine 22.0 g (51.4 mmol) of the product 3A(i) from Step A, 150 mL of 85% EtOH (aqueous), 25.85 g (0.463 mole) of Fe powder and 2.42 g (21.8 mmol) of CaCl.sub.2, and heat at reflux overnight. Add 12.4 g (0.222 mole) of Fe powder and 1.2 g (10.8 mmol) of CaCl.sub.2 and heat at reflux for 2 hours. Add another 12.4 g (0.222 mole) of Fe powder and 1.2 g (10.8 mmol) of CaCl.sub.2 and heat at reflux for 2 hours more. Filter the hot mixture through celite.RTM., wash the celite.RTM. with 50 mL of hot EtOH and concentrate the filtrate in vacuo to a residue. Add 100 mL of anhydrous EtOH, concentrate to a residue and chromatograph the residue (silica gel, MeOH/CH.sub.2 Cl.sub.2 gradient) to give 16.47 g of the product compound.
Step C: ##STR79##
Combine 16.47 g (41.4 mmol) of the product from Step B, and 150 mL of 48% HBr (aqueous) and cool to -3.degree. C. Slowly add (dropwise) 18 mL of bromine, then slowly add (dropwise) a solution of 8.55 g (0.124 mole) of NaNO.sub.2 in 85 mL of water. Stir for 45 minutes at -3.degree. to 0.degree. C., then adjust to pH=10 by adding 50% NaOH (aqueous). Extract with EtOAc, wash the extracts with brine and dry the extracts over Na.sub.2 SO.sub.4. Concentrate to a residue and chromatograph (silica gel, EtOAc/hexane gradient) to give 10.6 g and 3.28 g of the two product compounds 3C(i) and 3C(ii), respectively.
Mass Spec. for compound 3C(i): MH.sup.+ =461.2;
Mass Spec. for compound 3C(ii): MH.sup.+ =539.
Step D: ##STR80##
Hydrolyze the product 3C(i) of Step C by dissolving in concentrated HCl and heating to about 100.degree. C. for @ 16 hours. Cool the mixture, the neutralize with 1M NaOH (aqueous). Extract with CH.sub.2 Cl.sub.2, dry the extracts over MgSO.sub.4, filter and concentrate in vacuo to the title compound. Mass Spec.: MH.sup.+ =466.9.
Step E: ##STR81##
Dissolve 1.160 g (2.98 mmol) of the title compound from Step D in 20 mL of DMF, stir at room temperature, and add 0.3914 g (3.87 mmol) of 4-methyl-morpholine, 0.7418 g (3.87 mmol) of DEC, 0.5229 g (3.87 mmol) of HOBT, and 0.8795 g (3.87 mmol) of 1-N-t-butoxycarbonyl-piperidinyl-4-acetic acid. Stir the mixture at room temperature for 2 days, then concentrate in vacuo to s residue and partition the residue between CH.sub.2 Cl.sub.2 and water. Wash the organic phase successively with saturated NaHCO.sub.3 (aqueous), 10% NaH.sub.2 PO.sub.4 (aqueous) and brine. Dry the organic phase over MgSO.sub.4, filter and concentrate in vacuo to a residue. Chromatograph the residue (silica gel, 2% MeOH/CH.sub.2 Cl.sub.2 +NH.sub.3) to give 1.72 g of the product. m.p.=94.0.degree.-94.5.degree. C., Mass Spec.: MH.sup.+ =616.3,
elemental analysis: calculated--C, 60.54; H, 6.06; N, 6.83 found--C, 59.93; H, 6.62; N, 7.45.
Step F: ##STR82##
Combine 1.67 g (2.7 mmol) of the product of Step E and 20 mL of CH.sub.2 Cl.sub.2 and stir at 0.degree. C. Add 20 mL of TFA, stir the mixture for 2 hours, then basify the mixture with 1N NaOH (aqueous). Extract with CH.sub.2 Cl.sub.2, dry the organic phase over MgSO.sub.4, filter and concentrate in vacuo to give 1.16 g of the product. m.p.=140.2.degree.-140.8.degree. C., Mass Spec.: MH.sup.+ =516.2.
PREPARATIVE EXAMPLE 4 ##STR83## Step A: ##STR84##
Combine 25.86 g (55.9 mmol) of 4-(8-chloro-3-bromo-5,6-dihydro- 11H-benzo�5,6!cyclohepta�1,2-b!pyridin-11-ylidene)-1-piperidine-1-carboxylic acid ethyl ester and 250 mL of concentrated H.sub.2 SO.sub.4 at -5.degree. C., then add 4.8 g (56.4 mmol) of NaNO.sub.3 and stir for 2 hours. Pour the mixture into 600 g of ice and basify with concentrated NH.sub.4 OH (aqueous). Filter the mixture, wash with 300 mL of water, then extract with 500 mL of CH.sub.2 Cl.sub.2. Wash the extract with 200 mL of water, dry over MgSO.sub.4, then filter and concentrate in vacuo to a residue. Chromatograph the residue (silica gel, 10% EtOAc/CH.sub.2 Cl.sub.2) to give 24.4 g (86% yield) of the product. m.p.=165.degree.-167.degree. C., Mass Spec.: MH.sup.+ =506 (CI),
elemental analysis: calculated--C, 52.13; H, 4.17; N, 8.29 found--C, 52.18; H, 4.51; N, 8.16.
Step B: ##STR85##
Combine 20 g (40.5 mmol) of the product of Step A and 200 mL of concentrated H.sub.2 SO.sub.4 at 20.degree. C., then cool the mixture to 0.degree. C. Add 7.12 g (24.89 mmol) of 1,3-dibromo-5,5-dimethyl-hydantoin to the mixture and stir for 3 hours at 20.degree. C. Cool to 0.degree. C., add an additional 1.0 g (3.5 mmol) of the dibromohydantoin and stir at 20.degree. C. for 2 hours. Pour the mixture into 400 g of ice, basify with concentrated NH.sub.4 OH (aqueous) at 0.degree. C., and collect the resulting solid by filtration. Wash the solid with 300 mL of water, slurry in 200 mL of acetone and filter to provide 19.79 g (85.6% yield) of the product. m.p.=236.degree.-237.degree. C., Mass Spec.: MH.sup.+ =584 (CI),
elemental analysis: calculated--C, 45.11; H, 3.44; N, 7.17 found--C, 44.95; H, 3.57; N, 7.16.
Step C: ##STR86##
Combine 25 g (447 mmol) of Fe filings, 10 g (90 mmol) of CaCl.sub.2 and a suspension of 20 g (34.19 mmol) of the product of Step B in 700 mL of 90:10 EtOH/water at 50.degree. C. Heat the mixture at reflux overnight, filter through Celite.RTM. and wash the filter cake with 2.times.200 mL of hot EtOH. Combine the filtrate and washes, and concentrate in vacuo to a residue. Extract the residue with 600 mL of CH.sub.2 Cl.sub.2, wash with 300 mL of water and dry over MgSO.sub.4. Filter and concentrate in vacuo to a residue, then chromatograph (silica gel, 30% EtOAc/CH.sub.2 Cl.sub.2) to give 11.4 g (60% yield) of the product. m.p.=211.degree.-212.degree. C., Mass Spec.: MH.sup.+ =554 (CI),
elemental analysis: calculated--C, 47.55; H, 3.99; N, 7.56 found--C, 47.45; H, 4.31; N, 7.49.
Step D: ##STR87##
Slowly add (in portions) 20 g (35.9 mmol) of the product of Step C to a solution of 8 g (116 mmol) of NaNO.sub.2 in 120 mL of concentrated HCl (aqueous) at -10.degree. C. Stir the resulting mixture at 0.degree. C. for 2 hours, then slowly add (dropwise) 150 mL (1.44 mole) of 50% H.sub.3 PO.sub.2 at 0.degree. C. over a 1 hour period. Stir at 0.degree. C. for 3 hours, then pour into 600 g of ice and basify with concentrated NH.sub.4 OH (aqueous). Extract with 2.times.300 mL of CH.sub.2 Cl.sub.2, dry the extracts over MgSO.sub.4, then filter and concentrate in vacuo to a residue. Chromatograph the residue (silica gel, 25% EtOAc/hexanes) to give 13.67 g (70% yield) of the product. m.p.=163.degree.-165.degree. C., Mass Spec.: MH.sup.+ =539 (CI),
elemental analysis: calculated--C, 48.97; H, 4.05; N, 5.22 found--C, 48.86; H, 3.91; N, 5.18.
Step E: ##STR88##
Combine 6.8 g (12.59 mmol) of the product of Step D and 100 mL of concentrated HCl (aqueous) and stir at 85.degree. C. overnight. Cool the mixture, pour it into 300 g of ice and basify with concentrated NH.sub.4 OH (aqueous). Extract with 2.times.300 mL of CH.sub.2 Cl.sub.2, then dry the extracts over MgSO.sub.4. Filter, concentrate in vacuo to a residue, then chromatograph (silica gel, 10% MeOH/EtOAc+2% NH.sub.4 OH (aqueous)) to give 5.4 g (92% yield) of the title compound. m.p.=172.degree.-174.degree. C., Mass Spec.: MH.sup.+ =467 (FAB),
elemental analysis: calculated--C, 48.69; H, 3.65; N, 5.97found--C, 48.83; H, 3.80; N, 5.97.
Step F:
Following essentially the same procedure as Step C of Preparative Example 5 below, the title compound from Step E above is reacted with 1-N-t-butoxycarbonylpiperidinyl-4-acetic acid to produce the compound ##STR89## Step G:
Following essentially the same procedure as Step D of Preparative Example 5 below, the title compound from Step F above is deprotected to yield the title compound of Preparative Example 4.
PREPARATIVE EXAMPLE 5 ##STR90## Step A: ##STR91##
Hydrolyze 2.42 g of 4-(8-chloro-3-bromo-5,6-dihydro-11H-benzo�5,6!cyclohepta�1,2-b!pyridin- 11-ylidene)-1-piperidine- 1-carboxylic acid ethyl ester via substantially the same procedure as described in Preparative Example 3, Step D, to give 1.39 g (69% yield) of the product.
Step B: ##STR92##
Combine 1 g (2.48 mmol) of the product of Step A and 25 mL of dry toluene, add 2.5 mL of 1M DIBAL in toluene and heat the mixture at reflux. After 0.5 hours, add another 2.5 mL of 1M DIBAL in toluene and heat at reflux for 1 hour. (The reaction is monitored by TLC using 50% MeOH/CH.sub.2 Cl.sub.2 +NH.sub.4 OH (aqueous).) Cool the mixture to room temperature, add 50 mL of 1N HCl (aqueous) and stir for 5 min. Add 100 mL of 1N NaOH (aqueous), then extract with EtOAc (3.times.150 mL). Dry the extracts over MgSO.sub.4, filter and concentrate in vacuo to give 1.1 g of the title compound.
Step C: ##STR93##
Combine 0.501 g (1.28 mmol) of the title compound of Step B and 20 mL of dry DMF, then add 0.405 g (1.664 mmol) of 1-N-t-butoxycarbonylpiperidinyl-4-acetic acid, 0.319 g (1.664 mmol) of DEC, 0.225 g (1.664 mmol) of HOBT, and 0.168 g (1.664 mmol) of 4-methylmorpholine and stir the mixture at room temperature overnight. Concentrate the mixture in vacuo to a residue, then partition the residue between 150 mL of CH.sub.2 Cl.sub.2 and 150 mL of saturated NaHCO.sub.3 (aqueous). Extract the aqueous phase with another 150 mL of CH.sub.2 Cl.sub.2. Dry the organic phase over MgSO.sub.4, and concentrate in vacuo to a residue. Chromatograph the residue (silica gel, 500 mL hexane, 1 L of 1% MeOH/CH.sub.2 Cl.sub.2 +0.1% NH.sub.4 OH (aqueous), then 1 L of 2% MeOH/CH.sub.2 Cl.sub.2 +0.1% NH.sub.4 OH (aqueous)) to give 0.575 g of the product. m.p.=115.degree.-125.degree. C.; Mass Spec.: MH.sup.+ =616.
Step D: ##STR94##
Combine 0.555 g (0.9 mmol) of the product of Step C and 15 mL of CH.sub.2 Cl.sub.2 and cool the mixture to 0.degree. C. Add 15 mL of TFA and stir at 0.degree. C. for 2 hours. Concentrate in vacuo at 40.degree.-45.degree. C. to a residue, then partition the residue between 150 mL of CH.sub.2 Cl.sub.2 and 100 mL of saturated NaHCO.sub.3 (aqueous). Extract the aqueous layer with 100 mL of CH.sub.2 Cl.sub.2, combine the extracts and dry over MgSO.sub.4. Concentrate in vacuo to give 0.47 g of the product.
m.p.=140.degree.-150.degree. C.; Mass Spec.: MH.sup.+ =516.
PREPARATIVE EXAMPLE 6 ##STR95## Step A: ##STR96##
Combine 16.6 g (0.03 mole) of the product of Preparative Example 4, Step D, with a 3:1 solution of CH.sub.3 CN and water (212.65 mL CH.sub.3 CN and 70.8 mL of water) and stir the resulting slurry overnight at room temperature. Add 32.833 g (0.153 mole) of NaIO.sub.4 and then 0.31 g (2.30 mmol) of RuO.sub.2 and stir at room temperature give 1.39 g (69% yield) of the product. (The addition of RuO is accompanied by an exothermic reaction and the temperature climbs from 20.degree. to 30.degree. C.) Stir the mixture for 1.3 hrs. (temperature returned to 25.degree. C. after about 30 min.), then filter to remove the solids and wash the solids with CH.sub.2 Cl.sub.2. Concentrate the filtrate in vacuo to a residue and dissolve the residue in CH.sub.2 Cl.sub.2. Filter to remove insoluble solids and wash the solids with CH.sub.2 Cl.sub.2. Wash the filtrate with water, concentrate to a volume of about 200 mL and wash with bleach, then with water. Extract with 6N HCl (aqueous). Cool the aqueous extract to 0.degree. C. and slowly add 50% NaOH (aqueous) to adjust to pH=4 while keeping the temperature <30.degree. C. Extract twice with CH.sub.2 Cl.sub.2, dry over MgSO.sub.4 and concentrate in vacuo to a residue. Slurry the residue in 20 mL of EtOH and cool to OIC. Collect the resulting solids by filtration and dry the solids in vacuo to give 7.95 g of the product. .sup.1 H NMR (CDCl.sub.3, 200 MHz): 8.7 (s, 1 H); 7.85 (m, 6 H);
Step B: ##STR97##
Combine 21.58 g (53.75 mmol) of the product of Step A and 500 mL of an anhydrous 1:1 mixture of EtOH and toluene, add 1.43 g (37.8 mmol) of NaBH.sub.4 and heat the mixture at reflux for 10 min. Cool the mixture to 0.degree. C., add 100 mL of water, then adjust to pH.apprxeq.4-5 with 1M HCl (aqueous) while keeping the temperature <10.degree. C. Add 250 mL of EtOAc and separate the layers. Wash the organic layer with brine (3.times.50 mL) then dry over Na.sub.2 SO.sub.4. Concentrate in vacuo to a residue (24.01 g) and chromatograph the residue (silica gel, 30% hexane/CH.sub.2 Cl.sub.2) to give the product. Impure fractions were purified by rechromatography. A total of 18.57 g of the product was obtained. .sup.1 H NMR (DMSO-d.sub.6, 400 MHz): 8.5 (s, 1H); 7.9 (s, 1 H); 7.5 (d of d, 2 H); 6.2 (s, 1 H); 6.1 (s, 1 H); 3.5 (m, 1 H); 3.4 (m, 1 H); 3.2 (m, 2 H).
Step C: ##STR98##
Combine 18.57 g (46.02 mmol) of the product of Step B and 500 mL of CHCl.sub.3, then add 6.70 mL (91.2 mmol) of SOCl.sub.2, and stir the mixture at room temperature for 4 hrs. Add a solution of 35.6 g (0.413 mole) of piperazine in 800 mL of THF over a period of 5 min. and stir the mixture for 1 hr. at room temperature. Heat the mixture at reflux overnight, then cool to room temperature and dilute the mixture with 1 L of CH.sub.2 Cl.sub.2. Wash with water (5.times.200 mL), and extract the aqueous wash with CHCl.sub.3 (3.times.100 mL). Combine all of the organic solutions, wash with brine (3.times.200 mL) and dry over MgSO.sub.4. Concentrate in vacuo to a residue and chromatograph (silica gel, gradient of 5%, 7.5%, 10% MeOH/CH.sub.2 Cl.sub.2 +NH.sub.4 OH) to give 18.49 g of the title compound as a racemic mixture.
Step D--Separation of Enantiomers: ##STR99##
The racemic title compound of Step C is separated by preparative chiral chromatography (Chiralpack AD, 5 cm.times.50 cm column, flow rate 100 mL/min., 20% iPrOH/hexane+0.2% diethylamine), to give 9.14 g of the (+)-isomer and 9.30 g of the (-)-isomer.
Physical chemical data for (+)-isomer: m.p.=74.5.degree.-77.5.degree. C.; Mass Spec. MH.sup.+ =471.9; �.alpha.!.sub.D.sup.25 +97.4.degree. (8.48 mg/2 mL MeOH).
Physical chemical data for (-)-isomer: m.p.=82.9.degree.-84.5.degree. C.; Mass Spec. MH.sup.+ =471.8; �.alpha.!.sub.D.sup.25 =97.4.degree. (8.32 mg/2 mL MeOH).
Step E: ##STR100##
Combine 3.21 g (6.80 mmol) of the (-)-isomer product of Step D and 150 mL of anhydrous DMF. Add 2.15 g (8.8 mmol) of 1-N-t-butoxycarbonylpiperidinyl-4-acetic acid, 1.69 g (8.8 mmol) of DEC, 1.19 g (8.8 mmol) of HOBT and 0.97 mL (8.8 mmol) of N-methylmorpholine and stir the mixture at room temperature overnight. Concentrate in vacuo to remove the DMF and add 50 mL of saturated NaHCO.sub.3 (aqueous). Extract with CH.sub.2 Cl.sub.2 (2.times.250 mL), wash the extracts with 50 mL of brine and dry over MgSO.sub.4. Concentrate in vacuo to a residue and chromatograph (silica gel, 2% MeOH/CH.sub.2 Cl.sub.2 +10% NH.sub.4 OH) to give 4.75 g of the product. m.p.=75.7.degree.-78.5.degree. C.; Mass Spec.: MH.sup.+ =697; �.alpha.!.sub.D.sup.25 =5.5.degree. (6.6 mg/2 mL MeOH).
Step F: ##STR101##
Combine 4.70 g (6.74 mmol) of the product of Step E and 30 mL of MeOH, then add 50 mL of 10% H.sub.2 SO.sub.4 /dioxane in 10 mL aliquots over a 1 hr. period. Pour the mixture into 50 mL of water and add 15 mL of 50% NaOH (aqueous) to adjust to pH.apprxeq.10-11. Filter to remove the resulting solids and extract the filtrate with CH.sub.2 Cl.sub.2 (2.times.250 mL). Concentrate the aqueous layer in vacuo to remove the MeOH and extract again with 250 mL of CH.sub.2 Cl.sub.2. Dry the combined extracts over MgSO.sub.4 and concentrate in vacuo to give the product. m.p.=128.1.degree.-131.5.degree. C.; Mass Spec.: MH.sup.+ =597; �.alpha.!.sub.D.sup.25 -6.02.degree. (9.3 mg/2 mL MeOH).
PREPARATIVE EXAMPLE 7 ##STR102## Step A: ##STR103##
Combine 15 g (38.5 mmol) of 4-(8-chloro-3-bromo-5,6-dihydro- 11 H-benzo�5,6!cyclohepta�1,2-b!pyridin-11-ylidene)-1-piperidine-1-carboxylic acid ethyl ester and 150 mL of concentrated H.sub.2 SO.sub.4 at -5.degree. C., then add 3.89 g (38.5 mmol) of KNO.sub.3 and stir for 4 hours. Pour the mixture into 3 L of ice and basify with 50% NaOH (aqueous). Extract with CH.sub.2 Cl.sub.2, dry over MgSO.sub.4, then filter and concentrate in vacuo to a residue. Recrystallize the residue from acetone to give 6.69 g of the product. .sup.1 H NMR (CDCl.sub.3, 200 MHz): 8.5 (s, 1 H); 7.75 (s, 1 H); 7.6 (s, 1 H); 7.35 (s, 1 H); 4.15 (q, 2 H); 3.8 (m, 2 H); 3.5-3.1 (m, 4 H); 3.0-2.8 (m, 2 H); 2.6-2.2 (m, 4 H); 1.25 (t, 3 H).
Step B: ##STR104##
Combine 6.69 g (13.1 mmol) of the product of Step A and .sub.100 mL of 85% EtOH/water, then add 0.66 g (5.9 mmol) of CaCl.sub.2 and 6.56 g (117.9 mmol) of Fe and heat the mixture at reflux overnight. Filter the hot reaction mixture through celite.RTM. and rinse the filter cake with hot EtOH. Concentrate the filtrate in vacuo to give 7.72 g of the product. Mass Spec.: MH.sup.+ =478.0.
Step C: ##STR105##
Combine 7.70 g of the product of Step B and 35 mL of HOAc, then add 45 mL of a solution of Br.sub.2 in HOAc and stir the mixture at room temperature overnight. Add 300 mL of 1N NaOH (aqueous) , then 75 mL of 50% NaOH (aqueous) and extract with EtOAc. Dry the extract over MgSO.sub.4 and concentrate in vacuo to a residue. Chromatograph the residue (silica gel, 20%-30% EtOAc/hexane) to give 3.47 g of the product (along with another 1.28 g of partially purified product). Mass Spec.: MH.sup.+ =555.9.
.sup.1 H NMR (CDCl.sub.3, 300 MHz): 8.5 (s, 1 H); 7.5 (s, 1 H); 7.15 (s, 1 H); 4.5 (s, 2 H); 4.15 (m, 3 H); 3.8 (br s, 2 H); 3.4-3.1 (m, 4 H); 9-2.75 (m, 1 H); 2.7-2.5 (m, 2 H); 2.4-2.2 (m, 2 H); 1.25 (m, 3 H).
Step D: ##STR106##
Combine 0.557 g (5.4 mmol) of t-butylnitrite and 3 mL of DMF, and heat the mixture at to 60.degree.-70.degree. C. Slowly add (dropwise) a mixture of 2.00 g (3.6 mmol) of the product of Step C and 4 mL of DMF, then cool the mixture to room temperature. Add another 0.64 mL of t-butylnitrite at 40.degree. C. and reheat the mixture to 60.degree.-70.degree. C. for 0.5 hrs. Cool to room temperature and pour the mixture into 150 mL of water. Extract with CH.sub.2 Cl.sub.2, dry the extract over MgSO.sub.4 and concentrate in vacuo to a residue. Chromatograph the residue (silica gel, 10%-20% EtOAc/hexane) to give 0.74 g of the product. Mass Spec.: MH.sup.+ =541.0.
.sup.1 H NMR (CDCl.sub.3, 200 MHz): 8.52 (s, 1 H); 7.5 (d, 2 H); 7.2 (s, 1 H); 4.15 (q, 2 H); 3.9-3.7 (m, 2 H); 3.5-3.1 (m, 4 H); 3.0-2.5 (m, 2 H); 2.4-2.2 (m, 2 H); 2.1-1.9 (m, 2 H); 1.26 (t, 3 H).
Step E: ##STR107##
Combine 0.70 g (1.4 mmol) of the product of Step D and 8 mL of concentrated HCl (aqueous) and heat the mixture at reflux overnight. Add 30 mL of 1N NaOH (aqueous), then 5 mL of 50% NaOH (aqueous) and extract with CH.sub.2 Cl.sub.2. Dry the extract over MgSO.sub.4 and concentrate in vacuo to give 0.59 g of the title compound. Mass Spec.: M.sup.+ =468.7. m.p.=123.9.degree.-124.2.degree. C.
Step F: ##STR108##
React 6.0 g (12.8 mmol) of the title compound from Step E and with 3.78 g (16.6 mmol) of 1-N-t-butoxycarbonylpiperidinyl-4-acetic acid using substantially the same procedures as described for Preparative Example 5, Step C, to give 8.52 g of the product. Mass Spec.: MH.sup.+ =694.0 (FAB). .sup.1 H NMR (CDCl.sub.3, 200 MHz): 8.5 (d, 1 H); 7.5 (d, 2 H); 7.2 (d, 1 H); 4.15-3.9 (m, 3 H); 3.8-3.6 (m, 1 H); 3.5-3.15 (m, 3 H); 2.9 (d, 2 H); 2.8-2.5 (m, 4 H); 2.4-1.8 (m, 6 H); 1.8-1.6 (br d, 2 H); 1.4 (s, 9 H); 1.25-1.0 (m, 2 H).
Step G: ##STR109##
Combine 8.50 g of the product of Step F and 60 mL of CH.sub.2 Cl.sub.2, then cool to 0.degree. C. and add 55 mL of TFA. Stir the mixture for 3 h at 0.degree. C., then add 500 mL of 1N NaOH (aqueous) followed by 30 mL of 50% NaOH (aqueous). Extract with CH.sub.2 Cl.sub.2, dry over MgSO.sub.4 and concentrate in vacuo to give 7.86 g of the product. Mass Spec.: M.sup.+ =593.9 (FAB). .sup.1 H NMR (CDCl.sub.3, 200 MHz): 8.51 (d, 1 H); 7.52 (d of d, 2 H); 7.20 (d, 1 H); 4.1-3.95 (m, 2 H); 3.8-3.65 (m, 2 H); 3.5-3.05 (m, 5 H); 3.0-2.5 (m, 6 H); 2.45-1.6 (m, 6 H);1.4-1.1 (m, 2 H).
PREPARATIVE EXAMPLE 8 ##STR110## Step A: ##STR111##
Prepare a solution of 8.1 g of the title compound from Preparative Example 7, Step E, in toluene and add 17.3 mL of a 1M solution of DIBAL in toluene. Heat the mixture at reflux and slowly add (dropwise) another 21 mL of 1M DIBAL/toluene solution over a period of 40 min. Cool the reaction mixture to about 0.degree. C. and add 700 mL of 1M HCl (aqueous). Separate and discard the organic phase. Wash the aqueous phase with CH.sub.2 Cl.sub.2, discard the extract, then basify the aqueous phase by adding 50% NaOH (aqueous). Extract with CH.sub.2 Cl.sub.2, dry the extract over MgSO.sub.4 and concentrate in vacuo to give 7.30 g of the title compound, which is a racemic mixture of enantiomers.
Step B--Separation of Enantiomers: ##STR112##
The racemic title compound of Step A is separated by preparative chiral chromatography (Chiralpack AD, 5 cm.times.50 cm column, using 20% iPrOH/hexane+0.2% diethylamine), to give the (+)-isomer and the (-)-isomer of the title compound.
Physical chemical data for (+)-isomer: m.p.=148.8.degree. C.; Mass Spec. MH.sup.+ =469; �.alpha.!.sub.D.sup.25 65.6.degree. (12.93 mg/2 mL MeOH).
Physical chemical data for (-)-isomer: m.p.=112.degree. C.; Mass Spec. MH.sup.+ =469; �.alpha.!.sub.D.sup.25 65.2.degree. (3.65 mg/2 mL MeOH).
Step C: ##STR113##
React 1.33 g of the (+)-isomer of the title compound of Preparative Example 8, Step B, with 1.37 g of 1-N-t-butoxy-carbonylpiperidinyl-4-acetic acid using substantially the same procedures as described for Preparative Example 5, Step C, to give 2.78 g of the product. Mass Spec.: MH.sup.+ =694.0 (FAB); �.alpha.!.sub.D.sup.25 =34.1.degree. (5.45 mg/2 mL, MeOH).
Step D: ##STR114##
Treat 2.78 g of the product of Step C via substantially the same procedure as described for Preparative Example 5, Step D, to give 1.72 g of the product. m.p.=104.1.degree. C.; Mass Spec.: MH.sup.+ =594; �.alpha.!.sub.D.sup.25 53.4.degree. (11.42 mg/2 mL, MeOH).
PREPARATIVE EXAMPLE 9 ##STR115## Step A: ##STR116##
Combine 40.0 g (0.124 mole) of the starting ketone and 200 mL of H.sub.2 SO.sub.4 and cool to 0.degree. C. Slowly add 13.78 g (0.136 mole) of KNO.sub.3 over a period of 1.5 hrs., then warm to room temperature and stir overnight. Work up the reaction using substantially the same procedure as described for Preparative Example 4, Step A. Chromatograph (silica gel, 20%, 30%, 40%, 50% EtOAc/hexane, then 100% EtOAc) to give 28 g of the 9-nitro product, along with a smaller quantity of the 7-nitro product and 19 g of a mixture of the 7-nitro and 9-nitro compounds.
Step B: ##STR117##
React 28 g (76.2 mmol) of the 9-nitro product of Step A, 400 mL of 85% EtOH/water, 3.8 g (34.3 mmol) of CaCl.sub.2 and 38.28 g (0.685 mole) of Fe using substantially the same procedure as described for Preparative Example 4, Step C, to give 24 g of the product
Step C: ##STR118##
Combine 13 g (38.5 mmol) of the product of Step B, 140 mL of HOAc and slowly add a solution of 2.95 mL (57.8 mmol) of Br.sub.2 in 10 mL of HOAc over a period of 20 min. Stir the reaction mixture at room temperature, then concentrate in vacuo to a residue. Add CH.sub.2 Cl.sub.2 and water, then adjust to pH=8-9 with 50% NaOH (aqueous). Wash the organic phase with water, then brine and dry over Na.sub.2 SO.sub.4. Concentrate in vacuo to give 11.3 g of the product.
Step D: ##STR119##
Cool 100 mL of concentrated HCl (aqueous) to 0.degree. C., then add 5.61 g (81.4 mmol) of NaNO.sub.2 and stir for 10 min. Slowly add (in portions) 11.3 g (27.1 mmol) of the product of Step C and stir the mixture at 0.degree.-3.degree. C. for 2.25 hrs. Slowly add (dropwise) 180 mL of 50% H.sub.3 PO.sub.2 (aqueous) and allow the mixture to stand at 0.degree. C. overnight. Slowly add (dropwise) 150 mL of 50% NaOH over 30 min., to adjust to pH=9, then extract with CH.sub.2 Cl.sub.2. Wash the extract with water, then brine and dry over Na.sub.2 SO.sub.4. Concentrate in vacuo to a residue and chromatograph (silica gel, 2% EtOAc/CH.sub.2 Cl.sub.2) to give 8.6 g of the product.
Step E: ##STR120##
Combine 8.6 g (21.4 mmol) of the product of Step D and 300 mL of MeOH and cool to 0.degree.-2.degree. C. Add 1.21 g (32.1 mmol) of NaBH.sub.4 and stir the mixture at .about.0.degree. C. for 1 hr. Add another 0.121 g (3.21 mmol) of NaBH.sub.4, stir for 2 hr. at 0.degree. C., then let stand overnight at 0.degree. C. Concentrate in vacuo to a residue then partition the residue between CH.sub.2 Cl.sub.2 and water. Separate the organic phase and concentrate in vacuo (50.degree. C.) to give 8.2 g of the product.
Step F: ##STR121##
Combine 8.2 g (20.3 mmol) of the product of Step E and 160 mL of CH.sub.2 Cl.sub.2, cool to 0.degree. C., then slowly add (dropwise) 14.8 mL (203 mmol) of SOCl.sub.2 over a 30 min. period. Warm the mixture to room temperature and stir for 4.5 hrs., then concentrate in vacuo to a residue, add CH.sub.2 Cl.sub.2 and wash with 1N NaOH (aqueous) then brine and dry over Na.sub.2 SO.sub.4. Concentrate in vacuo to a residue, then add dry THF and 8.7 g (101 mmol) of piperazine and stir at room temperature overnight. Concentrate in vacuo to a residue, add CH.sub.2 Cl.sub.2, and wash with 0.25N NaOH (aqueous), water, then brine. Dry over Na.sub.2 SO.sub.4 and concentrate in vacuo to give 9.46 g of the crude product. Chromatograph (silica gel, 5% MeOH/CH.sub.2 Cl.sub.2 +NH.sub.3) to give 3.59 g of the title compound, as a racemate. .sup.1 H NMR (CDCl.sub.3, 200 MHz): 8.43 (d, 1 H); 7.55 (d, 1 H); 7.45 (d, 1 H); 7.11 (d, 1 H); 5.31 (s, 1 H); 4.86-4.65 (m, 1 H); 3.57-3.40 (m, 1 H); 2.98-2.55 (m, 6 H); 2.45-2.20 (m, 5 H).
Step G--Separation of Enantiomers: ##STR122##
The racemic title compound from Step F (5.7 g) is chromatographed as described for Preparative Example 6, Step D, using 30% iPrOH/hexane+0.2% diethylamine, to give 2.88 g of the R-(+)-isomer and 2.77 g of the S-(-)-isomer of the title compound.
Physical chemical data for the R-(+)-isomer: Mass Spec. MH.sup.+ =470.0; �.alpha.!.sub.D.sup.25 =12.1.degree. (10.9 mg/2 mL MeOH).
Physical chemical data for the S-(-)-isomer: Mass Spec. MH.sup.+ =470.0; �.alpha.!.sub.D.sup.25 -13.20.degree. (11.51 mg/2 mL MeOH).
Step H:
Following essentially the same procedure as Preparative Example 5, Steps C and D, the racemic title compound of Preparative Example 9 is obtained from the racemic compound of Step F. Similarly, using the (-)- or (+)- isomer from Step G, the (-)- or (+)-isomer of the title compound of Preparative Example 9 is obtained, respectively.
PREPARATIVE EXAMPLE 10 ##STR123## Step A: ##STR124##
Combine 13 g (33.3 mmol) of the title compound from Preparative Example 4, Step E, and 300 mL of toluene at 20.degree. C., then add 32.5 mL (32.5 mmol) of a 1M solution of DIBAL in toluene. Heat the mixture at reflux for 1 hr., cool to 20.degree. C., add another 32.5 mL of 1M DIBAL solution and heat at reflux for 1 hr. Cool the mixture to 20.degree. C. and pour it into a mixture of 400 g of ice, 500 mL of EtOAc and 300 mL of 10% NaOH (aqueous). Extract the aqueous layer with CH.sub.2 Cl.sub.2 (3.times.200 mL), dry the organic layers over MgSO.sub.4, then concentrate in vacuo to a residue. Chromatograph (silica gel, 12% MeOH/CH.sub.2 Cl.sub.2 +4% NH.sub.4 OH) to give 10.4 g of the title compound as a racemate. Mass Spec.: MH.sup.+ =469 (FAB). partial .sup.1 H NMR (CDCl.sub.3, 400 MHz): 8.38 (s, 1 H); 7.57 (s, 1 H); 7.27 (d, 1 H); 7.06 (d, 1 H); 3.95 (d, 1 H).
Step B--Separation of Enantiomers: ##STR125##
The racemic title compound of Step A is separated by preparative chiral chromatography (Chiralpack AD, 5 cm.times.50 cm column, using 5% iPrOH/hexane+0.2% diethylamine), to give the (+)-isomer and the (-)-isomer of the title compound.
Physical chemical data for (+)-isomer: Mass Spec. MH.sup.+ =469 (FAB); �.alpha.!.sub.D.sup.25 =43.5.degree. (c=0.402, EtOH); partial .sup.1 H NMR (CDCl.sub.3, 400 MHz): 8.38 (s, 1 H); 7.57 (s, 1 H); 7.27 (d, 1 H); 7.05 (d, 1 H); 3.95 (d, 1 H).
Physical chemical data for (-)-isomer: Mass Spec. MH.sup.+ =469 (FAB); �.alpha.!41.8.degree. (c=0.328 EtOH); partial .sup.1 H NMR (CDCl.sub.3, 400 MHz): 8.38 (s, 1 H); 7.57 (s, 1 H); 7.27 (d, 1 H); 7.05 (d, 1 H); 3.95 (d, 1 H).
Step C:
Following the procedure of Preparative Example 9, Step H, the racemic compound, the (+)-isomer or the (-)-isomer of the title compound of Preparative Example 10 can be obtained.
PREPARATVE EXAMPLE 11 ##STR126##
The compound ##STR127## is prepared according to the procedures of Preparative Example 40 of WO 95/10516 (published Apr. 20, 1995), by following the procedures described in Example 193 of WO 95/10516.
The (+)- and (-)-isomers can be separated by following essentially the same procedure as Step D of Preparative Example 6.
Physical chemical data for the R-(+)-isomer: .sup.13 C NMR (CDCl.sub.3): 155.8 (C); 146.4 (CH); 140.5 (CH); 140.2 (C); 136.2 (C); 135.3 (C); 133.4 (C); 132.0 (CH); 129.9 (CH); 125.6 (CH); 119.3 (C); 79.1 (CH); 52.3 (CH.sub.2); 52.3 (CH); 45.6 (CH.sub.2); 45.6 (CH.sub.2); 30.0 (CH.sub.2); 29.8 (CH.sub.2). �.alpha.!.sub.D.sup.25 25.8.degree. (8.46 mg/2 mL MeOH).
Physical chemical data for the S-(-)-isomer: .sup.13 C NMR (CDCl.sub.3): 155.9 (C); 146.4 (CH); 140.5 (CH); 140.2 (C); 136.2 (C); 135.3 (C); 133.3 (C); 132.0 (CH); 129.9 (CH); 125.5 (CH); 119.2 (C); 79.1 (CH); 52.5 (CH.sub.2); 52.5 (CH); 45.7 (CH.sub.2); 45.7 (CH.sub.2); 30.0 (CH.sub.2); 29.8 (CH.sub.2). �.alpha.!.sub.D.sup.25 27.90.degree. (8.90 mg/2 mL MeOH).
Following essentially the same procedure as Preparative Example 5, Steps C and D, the racemic compound, (+)-isomer or (-)-isomer of the title compound of Preparative Example 11 can be obtained from the corresponding racemic compound, (+)-isomer or (-)-isomer of the compound ##STR128##
PREPARATIVE EXAMPLE 12 ##STR129##
Combine 14.73 g (27.3 mmol) of the compound from Example 193 of WO 95/10516 and 125 mL of anhydrous MeOH, and add (in portions) 300 mL of a 10% solution of concentrated H.sub.2 SO.sub.4 in dioxane. Stir the mixture at 25.degree. C. for 2 hours, then pour into water and adjust to pH=13 with 50% NaOH (aqueous). Extract with CH.sub.2 Cl.sub.2, wash the extract with water and dry over MgSO.sub.4. Concentrate in vacuo to a residue and chromatograph (silica gel, 10% (10% NH.sub.4 OH in MeOH)/CH.sub.2 Cl.sub.2) to give 8.9 g of the title compound. Mass Spec.: MH.sup.+ =539
EXAMPLE 1
PHENYL 4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL!-1-PIPERAZINYL!-2-OXOETHYL!-N-CYANO-1-PIPERIDINE-CARBOXIMIDATE ##STR130##
1-(3-Bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!-cyclohepta�1,2-b! pyridin-11-yl)-4-�(4-piperidinyl)acetyl!-piperazine (Formula 47.0) (prepared as described in Preparative Example 11) (2.5 g) (1 equivalent) and diphenylcyanocarbon-imidate (1.38 g) (1.2 equivalents) were dissolved in 2-propanol (65 ml) and the solution was heated at 80.degree. C. under reflux and under nitrogen for 24 h. The mixture was evaporated to dryness and the product was chromatographed on a silica gel column (60.times.2.5 cm) using neat ethyl acetate as the eluant to give the title compound (Formula 2.0) (2.7921 g; 87%), FABMS: m/z 661 (MH+).
______________________________________ .delta..sub.c (CDCl.sub.3) for Formula______________________________________ 2.0Tricyclic CH.sub.2 : 30.5, 30.6 CH: 147.1, 141.4, 132.5, 126.3, 130.5, 79.0 C: 120.1, 140.9, 134.3, 135.3, 136.8, 155.5Piperazine CH.sub.2 : 41.7, 51.4, 51.9, 45.6Piperazine CH.sub.2 : 46.4, 32.1, 32.1, 46.4, 38.8,N-substituent CH: 32.5, 118.5, 118.5, 130.1, 130.1, 125.7 C: 113.5, 152.5, 157.4, 169.3______________________________________
EXAMPLE 2
4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL!-N-CYANO-1-PIPERIDINE-CARBOXIMIDAMIDE ##STR131##
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6! cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-N-cyano-1-piperidinecarboximidate (2.62 g) (Formula 2.0, prepared as described in Example 1 above) was dissolved in 2-propanol (50 ml) and concentrated ammonium hydroxide (4 ml) was added. The mixture was stirred at 25.degree. C. for 24 h and then evaporated to dryness. The residue was triturated with diethyl ether (2.times.250 ml) and the ether was discarded. The remaining product was chromatographed on a silica gel column (60.times.2.5 cm) using 4% (10% concentrated ammonium hydroxide in methanol)-dichloromethane as the eluant to give the title compound (Formula 3.0) (1.967 g;85%), FABMS: m/z 584.2 (MH.sup.+).
______________________________________ .delta..sub.c (d.sub.6 -DMSO) for Formula______________________________________ 3.0Tricyclic CH.sub.2 : 29.0, 29.4 CH: 146.1, 140.7, 132.4, 125.4, 129.9, 77.7 C: 119.1, 141.1, 132.4, 135.4, 136.7, 155.5Piperazine CH.sub.2 : 40.6, 50.8, 51.3, 44.6Piperazine CH.sub.2 : 44.3, 44.3, 30.9, 30.9, 38.3N-substituent CH: 31.9 C: 118.1, 159.3, 168.8______________________________________
EXAMPLE 3
PHENYL 4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL-1-PIPERIDINECARBOXIMIDATE ##STR132##
1-(3-Bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6! cyclo-hepta�1,2-b!pyridin-11-yl)-4-�(4-piperidinyl)acetyl!piperazine (6 g) (1 equivalent) (Formula 47.0 from Preparative Example 11 above) was dissolved in anhydrous dichloromethane (71.1 ml). Phenyl cyanate (2.2752 ml) (2 equivalents) and diisopropyl-ethylamine (100 drops) were added and the mixture was stirred at 25.degree. C. for 15 minutes. The reaction mixture was directly introduced onto a silica gel column (15.times.5 cm) and eluted with 10% increasing to 20% (10% concentrated ammonium hydroxide in methanol)-dichloromethane to give the title compound (6.66 g; 92%), FABMS: m/z 635.9 (MH.sup.+).
______________________________________ .delta..sub.c (CDCl.sub.3) for Formula______________________________________ 4.0Tricyclic CH.sub.2 : 30.6, 30.5 CH: 147.1, 141.3, 132.5, 126.3, 130.5, 79.0 C: 120.1, 140.9, 134.2, 135.3, 136.8, 155.6Piperazine CH.sub.2 : 41.6, 51.4, 51.9, 45.8Piperazine CH.sub.2 : 45.3, 45.3, 32.0, 32.0, 39.5,N-substituent CH: 32.2, 122.1, 122.1, 130.0, 130.0, 125.7 C: 151.4, 159.3, 170.0______________________________________
EXAMPLE 4
1-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-4-�4-(1-CYANO-4-PIPERIDINYL)ACETYL!PIPERAZINE ##STR133##
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6! cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-1-piperidinecarboximidate (0.3 g) (1 equivalent) (Formula 4.0, prepared as described in Example 3 above) was dissolved in anhydrous THF (11.2 ml). A 60% sodium hydride dispersion in oil (0.0753 g) (4 equivalents) was added and the mixture was stirred at 25.degree. C. for 2 h. The mixture was diluted with dichloromethane and washed with 1.0N sodium hydroxide. The dichloromethane layer was dried over magnesium sulfate, filtered and evaporated to dryness. The product was chromatographed on a silica gel column (30.times.2.5 cm) using 1.5% (10% concentrated ammonium hydroxide in methanol)-dichloromethane as the eluant to give the title compound (Formula 5.0) (0.1824 g; 71%), FABMS: m/z 542.2 (MH.sup.+).
______________________________________ .delta..sub.c (CDCl.sub.3) for Formula______________________________________ 5.0Tricyclic CH.sub.2 : 30.6, 30.5 CH: 147.1, 141.4, 132.5, 126.3, 130.6, 79.0 C: 120.1, 140.9, 134.3, 135.3, 136.9, 155.6Piperazine CH.sub.2 : 41.6, 51.4, 51.9, 45.6Piperazine CH.sub.2 : 49.7, 49.7, 31.1, 31.1, 39.1N-substituent CH: 31.8 C: 118.4, 169.3______________________________________
EXAMPLE 5
PHENYL 4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-SULFAMOYL-1-PIPERIDINE-CARBOXIMIDATE
Procedure 1: ##STR134##
If one were to follow Procedure I then one would obtain the compound of Formula 6.0.
1-(3-Bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclo-hepta�1,2-b! pyridin-11-yl)-4-�(4-piperidinyl)acetyl!piperazine (1equivalent) (Formula 47.0 of Preparative Example 11) and diphenylsulfamoylcarbonimidate (1.2 equivalents) �prepare as described in: M. Haake and B. Schummelfeder, Synthesis, 753-15 758 (1991)! are dissolved in 2-propanol and the mixture is heated as described in Example 1 above to give the title compound (Formula 6.0).
Procedure 2: ##STR135##
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6! cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-1-piperidinecarboximidate (0.05 g)(1 equivalent) (Formula 4.0, prepared as described in Example 3 above) was dissolved in anhydrous dichloromethane (2 ml) and triethylamine (0.0328 ml) (3 equivalents) was added. Sulfamoyl chloride (0.0091 g) (1 equivalent) �prepared as described in: R. Appel and G. Berger, Chem. Ber., 91, 1339-1341 (1958)!1 was added and the mixture was stirred under argon at 25.degree. C. for 18 h. Additional sulfamoyl chloride (0.0091 g) (1 equivalent) was added and the stirring was continued for a total of 90 h. The mixture was diluted with dichloromethane and extracted with 1N sodium hydroxide. The dichloromethane layer was dried over magnesium sulfate, filtered and evaporated to dryness. The product was chromatographed on a silica gel column (15.times.1 cm) using 2% increasing to 4% (10% concentrated ammonium hydroxide in methanol)-dichloro-methane as the eluant to give the title compound (Formula 6.0) (0.0054 g; 10%), FABMS: m/z 715.4 (MH.sup.+), .delta.H (CDCl.sub.3) 7.03 (2 H, m, ArH), 7.09-7.18 (4 H, m, ArH), 7.37 (1 H, m, ArH), 7.39 (1 H, m, ArH), 7.59 (1 H, s, ArH) and 8.37 ppm (1 H, s, ArH).
EXAMPLE 6
4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-SULFAMOYL-1-PIPERIDINE-CARBOXIMIDAMIDE ##STR136##
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5H-benzo�5,6!cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-N-sulfamoyl-1-piperidinecarboximidate (Formula 6.0) (prepared as described in Example 5above) (0.089 g, 0.1 mmoles) was dissolved in anhydrous THF (1 ml). A saturated solution of ammonia in anhydrous THF (4 ml) was added and the mixture was stirred in a sealed reaction vessel at 25.degree. C. for 19 h. The mixture was evaporated to dryness and chromatographed on silica gel using 2%-6% methanol in dichloromethane as the eluant to give the title compound (Formula 7.0) (0.0667 g, 84%), ESIMS: m/z 638.0 (MH.sup.+).
______________________________________ .delta..sub.c (CDCl.sub.3)______________________________________Tricyclic CH.sub.2 : 30.5, 30.6 CH: 78.9, 126.3, 130.5, 132.6, 141.4, 147.0 C: 120.1, 134.2, 135.3, 136.9, 140.9, 155.6Piperazine CH.sub.2 : 41.7, 45.7, 51.4, 51.9Piperazine CH.sub.2 : 31.8, 31.8, 39.0, 44.9, 44.9N- CH: 32.8substituent C: 169.8______________________________________
EXAMPLE 7
PHENYL 4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL!-N-(N-METHYLSULFAMOYL)-1-PIPERIDINECARBOXIMIDATE
Procedure 1: ##STR137##
If one were to follow Procedure 1 then one would obtain the compound of Formula 8.0.
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6! cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-1-piperidinecarboximidate (1 equivalent) (Formula 4.0, prepare as described in Example 3 above) is dissolved in an inert anhydrous solvent such as acetonitrile, benzene, or toluene and triethylamine (2 equivalents) is added. The solution is cooled to 0.degree. C. and N-methylsulfamoyl chloride (1.2 equivalents) �prepare as described in: J. A. Kloek and K. L. Leschinsky, J. Org. Chem., 41(25), 4028-4029 (1976)! is added. The mixture is stirred at 0.degree. C. to 25.degree. C. for 3 h and is worked up to give the title compound (Formula 8.0).
Procedure 2: ##STR138##
If one were to follow Procedure 2 then one would obtain the compound of Formula 8.0.
1-(3-Bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclo-hepta�1,2-b! pyridin-1 1-yl)-4-�(4-piperidinyl)acetyl!piperazine (1 equivalent) (Formula 47.0, prepare as described in Preparative Example 11) and diphenylmethylsulfamoylcarbonimidate (1.2 equivalents) �prepare by the same procedure, only using methylsulfamoyl chloride, as described in: A. Buschauer, Arch. Pharm., 377-378 (1987)! are dissolved in 2-propanol and the mixture is heated as described in Example 1 above to give the title compound.
EXAMPLE 8
4-�2-�4-(3-BROMO-8-CHLORO-6,1-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL!-N-(N-METHYLSULFAMOYL)-1-PIPERIDINECARBOXIMIDAMIDE ##STR139##
If one were to follow this procedure then the compound of Formula 9.0 would be obtained.
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,1-dihydro-5 H-benzo-�5, 6!cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-N-(N-methyl-sulfamoyl)-1-piperidinecarboxiimidate (Formula 8.0, prepare as described in Example 7 above) is dissolved in 2-propanol and concentrated ammonium hydroxide is added. The mixture is stirred at 25.degree. C. for 24 h and then is evaporated to dryness. The residue is triturated with diethyl ether (2.times.250 ml) and the ether is discarded. The remaining product is chromatographed on a silica gel column to give the title compound (Formula 9.0).
Alternatively, anhydrous ammonia in a suitable inert solvent such as methanol, or THF may be used in place of ammonium hydroxide in the above reaction.
EXAMPLE 9
PHENYL 4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL!-N-(N,N-DIMETHYLSULFAMOYLI-1-PIPERIDINECARBOXIMIDATE
Procedure 1: ##STR140##
If one were to follow Procedure 1 then the compound of Formula 10.0 would be obtained.
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6! cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-1-piperidinecarboximidate (1 equivalent) (Formula 4.0, prepare as described in Example 3 above) is dissolved in an inert anhydrous solvent such as acetonitrile, benzene, or toluene and triethylamine (2 equivalents) is added. The solution is cooled to 0.degree. C. and N,N-dimethylsulfamoyl chloride (1.2 equivalents) is added. The mixture is stirred at 0.degree. C. to 25.degree. C. for 3 h and worked up to give the title compound .
Procedure 2: ##STR141##
If one were to follow Procedure 2 then the compound of Formula 10.0 would be obtained.
1-(3-Bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclo-hepta�1,2-b! pyridin-11-yl)-4-�(4-piperidinyl)acetyl!piperazine (1 equivalent) (Formula 47.0, prepare as described in Preparative Example 11) and diphenyldimethylsulfamoylcarbonimidate (1.2 equivalents) �prepare by the same procedure, only using dimethylsulfamoyl chloride, as is described in: A. Buschauer, Arch. Pharm., 377-378 (1987)! are dissolved in 2-propanol and the mixture is heated as described in Example 1 above to give the title compound (Formula 10.0).
EXAMPLE 10
4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-(N,N-DIMETHYLSULFAMOYL)-1-PIPERIDINECARBOXIMIDAMIDE ##STR142##
If one were to follow this procedure then the compound of Formula 11.0 would be obtained.
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6! cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-N-(N,N-dimethyl-sulfamoyl)-l-piperidinecarboximidate (Formula 10.0, prepare as described in Example 9 above) is dissolved in 2-propanol and concentrated ammonium hydroxide is added. The mixture is stirred at 25.degree. C. for 24 h and then evaporated to dryness. The residue is triturated with diethyl ether (2.times.250 ml) and the ether is discarded. The remaining product is chromatographed on a silica gel column to give the title compound (Formula 11.0).
Alternatively, anhydrous ammonia in a suitable inert solvent such as methanol, or THF, may be used in place of ammonium hydroxide in the above reaction.
EXAMPLE 11
4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6CYCLOHEPTA�1,2-b!PYRIDIN-11-YL-1-PIPERAZINYL!-2-OXOETHYL!-N-HYDROXY-1-PIPERIDINE-CARBOXIMIDATE
PROCEDURE 1: ##STR143##
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo-�5,6!cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-1-piperidinecarboximidate (Formula 4.0) (prepared as described in Example 3 above) (0.15 g, 0.26 mmoles) was dissolved in methanol (0.75 ml). An aqueous solution of hydroxylamine �prepared by dissolving hydroxylamine hydrochloride (0.0164 g, 0.26 mmoles) in 50% (w/v) sodium hydroxide (0.0188 g, 0.26 mmoles) and water (0.258 ml)! was added and the mixture was stirred at 25.degree. C. for 3 h. Additional methanol (1.2 ml) was added and the mixture was stirred at 25.degree. C. for a total of 26 h. The solution was evaporated to dryness and the residue was chromatographed on silica gel using 2%-4%-5%-10%-25%-35% methanol in dichloromethane as the eluant to give the title compound (Formula 12.0) (0.0323 g, 24%), which was identical with that prepared in Procedure 2 below.
Procedure 2: ##STR144##
1-(3-Bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclo-hepta!1,2-b!pyridin-11-yl)-4-�4-(1-cyano-4-piperidinyl)acetyl!-piperazine (Formula 5.0) (prepared as described in Example 4 above) (0.4 g, 0.737 mmoles) was dissolved in methanol (2 ml). An aqueous solution of hydroxylamine �prepared by dissolving hydroxylamine hydrochloride (0.0512 g, 0.737 mmoles) in 50% (w/v) sodium hydroxide (0.0592 g, 0.737 mmoles) and water (0.8 ml)! was added and the mixture was stirred at 25.degree. C. for 3 h. Additional methanol (3.2 ml) was added and the mixture was stirred at 25.degree. C. for a total of 18 h. The solution was evaporated to dryness and the residue was chromatographed on silica gel using 5% methanol in dichloromethane as the eluant to give the title compound (Formula 12.0) (0.2965 g, 70%), FABMS: m/z 575.0 (MH.sup.+).
______________________________________ .delta..sub.c (CDCl.sub.3) for Formula______________________________________ 12.0Tricyclic CH.sub.2 : 30.6, 30.6 CH: 147.0, 141.4, 132.6, 126.3, 130.5, 79.0 C: 120.1, 140.9, 134.2, 135.3, 136.9, 155.6Piperazine CH.sub.2 : 41.7, 51.4, 51.9, 45.8Piperazine CH.sub.2 : 46.8, 46.8, 31.5, 31.5, 39.2N-substituent CH: 32.8 C: 157.6, 170.0______________________________________
EXAMPLE 12
4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL!-N-METHOXY-1-PIPERIDINE-CARBOXIMIDATE ##STR145##
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo-�5,6!cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-1-piperidinecarboximidate (Formula 4.0) (prepared as described in Example 3 above) (0.1 g, 0.157 mmoles) and methoxylamine hydrochloride (0.0154 g, 0.157 mmoles) were dissolved in anhydrous pyridine (1 ml) and the mixture was stirred at 25.degree. C. for 2 h. The solution was evaporated to dryness and the residue was chromatographed on silica gel using 2%-4%-10% methanol in dichloromethane as the eluant to give the title compound (Formula 13.0) (0.0741 g, 80%), FABMS: m/z 589.1 (MH.sup.+).
______________________________________ .delta..sub.c (CDCl.sub.3)______________________________________Tricyclic CH.sub.2 : 30.4, 30.5 CH: 79.0, 126.2, 130.4, 132.4, 141.2, 147.0 C: 120.0, 134.1, 135.3, 136.7, 140.8, 155.5Piperazine CH.sub.2 : 41.5, 45.7, 51.3, 51.8Piperazine CH.sub.3 : 61.0N- CH.sub.2 : 39.4, 31.7, 31.7, 46.8, 46.8substituent CH: 33.2 C: 156.7______________________________________
EXAMPLE 13
PHENYL N- ��(AMINOCARBONYL)AMINO!CARBONYL!4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6CYCLOHEPTA-�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL!-1-PIPERIDINECARBOXIMIDATE ##STR146##
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5H-benzo-�5,6!cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-1-piperidinecarboximidate (Formula 4.0) (prepared as described in Example 3 above) (0.35 g, 0.549 mmoles) was dissolved in anhydrous dichloromethane (7 ml) and trimethylsilylisocyanate (1.484 ml, 10.98 mmoles) was added. The mixture was stirred at 25.degree. C. for 21 h. The mixture was diluted with dichloromethane, washed with saturated aqueous sodium bicarbonate, water, dried over MgSO.sub.4, filtered and evaporated to dryness. The residue was chromatographed on silica gel using 2%-3% methanol in dichloromethane as the eluant to give the title compound (Formula 16.1) (0.1302 g, 33%), FABMS: m/z 721.9 (MH.sup.+).
______________________________________ .delta..sub.c (CDCl.sub.3)______________________________________Tricyclic CH.sub.2 : 30.5, 30.6 CH: 79.0, 126.4, 130.6, 132.5, 141.4, 147.1 C: 120.1, 134.3, 135.3, 136.8, 140.9, 155.5Piperazine CH.sub.2 : 41.7, 45.7, 51.4, 51.9Piperazine CH.sub.2 : 32.0, 32.0, 38.9, 46.3, 46.3N- CH: 32.7, 119.0, 119.0, 125.0, 129.6, 129.6substituent C: 153.0, 155.2, 157.6, 169.4______________________________________
EXAMPLE 14
N-��(AMINOCARBONYL)AMINO!CARBONYL!-4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!-PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-1-PIPERIDINECARBOXIMIDAMIDE ##STR147##
Phenyl N-��(aminocarbonyl)amino!carbonyl!-4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-1-piperidine-carboximidate (Formula 16.1) (0.2943 g, 0.462 mmoles) was dissolved in a saturated solution of ammonia in anhydrous THF (80 ml) and the mixture was stirred at 25.degree. C. for 19 h in a sealed vessel. The solution was evaporated to dryness and the residue was chromatographed on silica gel using 3% (10% conc. ammonium hydroxide in methanol)-dichloromethane as the eluant to give the title compound (Formula 17.1) (0.2663 g, 40%), ESIMS: m/z 645.2 (MH.sup.+).
______________________________________ .delta..sub.c (CDCl.sub.3)______________________________________Tricyclic CH.sub.2 : 30.5, 30.6 CH: 79.0, 126.3, 130.6, 132.5, 141.3, 147.1 C: 120.1, 134.3, 135.3, 136.8, 140.9, 155.5Piperazine CH.sub.2 : 41.7, 45.7, 51.4, 51.9Piperazine CH.sub.2 : 31.9, 31.9, 39.1, 44.3, 44.3N- CH: 32.9substituent C: 156.1, 159.4, 162.6, 169.6______________________________________
EXAMPLE 15
PHENYL 4-�2-�4-(3-BROMO-8-CHLORO-6,1 1-DIHYDRO-5 H-BENZO�5,6CYCLOHEPTA!1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-CARBOXAMIDO-1-PIPERIDINE-CARBOXIMIDATE ##STR148##
If one were to follow Procedure 1 then the compound of Formula 16.0 would be obtained.
1-(3-Bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclo-hepta�1,2-b!pyridin-11-yl)-4-�(4-piperidinyl)acetyl!piperazine (1 equivalent) (Formula 47.0, prepare as described in Preparative Example 11) and diphenylcarboxamidocarbonimidate (1.2 equivalents) �prepare, using urea in place of sulfamide, as is described in: M. Haake and B. Schummelfeder, Synthesis, 753-758 (1991)! is dissolved in 2-propanol and the solution is heated at 80.degree. C. under reflux and under nitrogen for 24 h. The mixture is evaporated to dryness and chromatographed on a silica gel column to give the title compound (Formula 16.0).
EXAMPLE 16
4-�2-�4-(3-BROMO-8-CHLORO-6.11-DIHYDRO-5 H-BENZO�5.6!CYCLOHEPTA�1 2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-CARBOXAMIDO-1-PIPERIDINECARBOXIMIDAMIDE ##STR149##
If one were to follow Procedure 1 then the compound of Formula 17.0 would be obtained.
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6! cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-N-carboxamido-1-piperidinecarboximidate (formula 16.0, prepare as described in Example 15 above) is dissolved in 2-propanol and concentrated ammonium hydroxide is added. The mixture is stirred at 25.degree. C. for 24 h and then evaporated to dryness. The residue is triturated with diethyl ether (2.times.250 ml) and the ether is discarded. The remaining product is chromatographed on a silica gel column to give the title compound (Formula 17.0).
Alternatively, anhydrous ammonia in a suitable inert solvent such as methanol, or THF, may be used in place of ammonium hydroxide in the above reaction.
EXAMPLE 17
PHENYL 4-�2- �4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-(N'-METHYLCARBOXAMIDO)-1-PIPERIDINECARBOXIMIDATE ##STR150##
If one were to follow this procedure then the compound of Formula 18.0 would be obtained.
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-l-piperidinecarboximidate (1 equivalent) (Formula 4.0, prepare as described in Example 3 above) is dissolved in anhydrous dichloromethane. Methylisocyanate (2 equivalents) is added and the mixture is stirred at 25.degree. C. for 48 h. The mixture is shaken with aqueous sodium bicarbonate and is extracted with dichloromethane. The latter is dried over MgSO.sub.4, is filtered and is evaporated to dryness to give the title compound (Formula 18.0) after chromatography on silica gel
EXAMPLE 18
4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-N'-METHYLCARBOXAMIDO)-1-PIPERID INECARBOXIMIDAMIDE ##STR151##
If one were to follow this procedure then the compound of Formula 19.0 would be obtained.
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-N-(N'-methylcarboxamido)-1-piperidinecarboximidate (Formula 18.0, prepare as described in Example 17 above) is dissolved in 2-propanol and concentrated ammonium hydroxide is added. The mixture is stirred at 25.degree. C. for 24 h and then evaporated to dryness. The residue is triturated with diethyl ether (2.times.250 ml) and the ether is discarded. The remaining product is chromatographed on a silica gel column to give the title compound (Formula 19.0).
Alternatively, anhydrous ammonia in a suitable inert solvent such as methanol, or THF, may be used in place of ammonium hydroxide in the above reaction.
EXAMPLE 19
N-�4-�2-�4-(-3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL!-1-PIPERIDINYL!-N'-METHYL-2-NITRO- 1-ETHENEAMINE ##STR152##
If one were to follow this procedure then the compound of Formula 20.0 would be obtained.
Copper(I)chloride (1 equivalent) is dissolved in anhydrous acetonitrile. To this solution, a solution of 1-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclohepta�1,2-b!pyridin- 11-yl)-4-�(4-piperidinyl)acetyl!-piperazine (1 equivalent) (Formula 47.0, prepare as described in Preparative Example 11), 1-methylthio-1-methyl-amino-2-nitroethene (1 equivalent) (prepare as described in Canadian Patent No. 1178289 (1984)) and triethylamine in anhydrous acetonitrile is added dropwise over 10 minutes with stirring. The solid is filtered off, the volume is reduced and dichloromethane is added. The mixture is washed with aqueous sodium bicarbonate and the dichloromethane layer is dried over magnesium sulfate, filtered and evaporated to dryness. The residue is purified on silica gel to give the title compound Formula 20.0).
EXAMPLE 20
N-�4-�2-�4-(8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL!1-1-PIPERIDINYL!-N'-METHYL-2-NITRO-1-ETHENEAMINE ##STR153##
If one were to follow this procedure then the compound of Formula 21.0 would be obtained.
STEP A:
N-�4-�2-�4-(8-CHLORO-6,11-DIHYDRO-5 H-BENZO �5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-1-PIPERIDINYL!-N'-METHYL-METHYLTHIO-CARBOXIMIDATE ##STR154##
4-�2-�4-(8-CHLORO-6,11-dihydro-5 H-benzo�5,6!cyclohepta-�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-N-methyl-1-piperidinecarbothioamide (1 equivalent) (Formula A-38) is dissolved in methanol and methyl iodide (1.2 equivalents) is added over 15 minutes. The mixture is then heated under reflux for 2 h. The solution is evaporated to dryness and the residue is dissolved in water and basified with 50% sodium hydroxide and then saturated with solid sodium chloride. The mixture is extracted with dichloromethane, dried over magnesium sulfate, filtered and evaporated to dryness to give the title compound (Formula B-38).
The compound of Formula A-38 is prepared by the reaction of the compound of Preparative Example 12 above with CH.sub.3 NCS. The reaction is conducted in anhydrous CH.sub.2 Cl.sub.2, with stirring, under argon at 25.degree. C. until the reaction is complete. The reaction mixture is diluted with CH.sub.2 Cl.sub.2 an is washed with saturated NaHCO.sub.3 (aqueous), then water. Dry over MgSO.sub.4, concentrate in vacuo to a residue and chromatograph (silica gel, 5% (10%NH.sub.4 OH in MeOH)/CH.sub.2 Cl.sub.2 to give the compound of Formula A-38.
STEP B:
N-�4-�2-�4-(8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-1-PIPERIDINYL!-N'-METHYL-2-NITRO- 1-ETHENEAMINE ##STR155##
The crude product from Step A above (Formula B-38) is refluxed with excess nitromethane for 16 h and then is evaporated to dryness to give the title compound (Formula 21.0).
EXAMPLE 21
PHENYL 4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-(METHYLSULFONYL)-1-PIPERIDINECARBOXIMIDATE ##STR156##
If one were to follow this procedure then the compound of Formula 22.0 would be obtained.
1-(3-Bromo-8-chloro-6,1 1-dihydro-5 H-benzo�5,6!cyclo-hepta�1,2-b! pyridin-11-yl)-4-�(4-piperidinyl)acetyl!piperazine (1 equivalent) (Formula 47.0, prepare as described in Preparative Example 11) and diphenylmethylsulfonylcarbonimidate (1.2 equivalents) �prepare as described in: A. Buschauer, Arch. Pharm., 377-378 (1987)! are dissolved in 2-propanol and the mixture is heated as described in Example 1 above to give the title compound (Formula 22.0).
EXAMPLE 22
4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-(METHYLSULFONYL)-1-PIPERIDINECARBOXIMIDAMIDE ##STR157##
If one were to follow this procedure then the compound of Formula 23.0 would be obtained.
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-N-(methylsulfonyl)-1-piperidinecarboximidate (Formula 22.0, prepare as described in Example 21 above) is dissolved in 2-propanol and concentrated ammonium hydroxide is added. The mixture is stirred at 25.degree. C. for 24 h and then is evaporated to dryness. The residue is triturated with diethyl ether (2.times.250 ml) and the ether is discarded. The remaining product is chromatographed on a silica gel column to give the title compound (Formula 23.0).
Alternatively, anhydrous ammonia in a suitable inert solvent such as methanol, or THF, may be used in place of ammonium hydroxide in the above reaction.
EXAMPLE 23
PHENYL 4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPTA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!-2-OXOETHYL!-N-BENZOYL-1-PIPERIDINE-CARBOXIMIDATE ##STR158##
If one were to follow this procedure then the compound of Formula 24.0 would be obtained.
1-(3-Bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6!cyclo-hepta�1,2-b! pyridin-11-yl)-4-�(4-piperidinyl)acetyl!piperazine (1 equivalent) (Formula 47.0, prepare as described in Preparative Example 11) and diphenylmethylbenzoylcarbonimidate (1.2 equivalents) �prepare as described in: A. Buschauer, Arch. Pharm., 377-378 (1987)! are dissolved in 2-propanol and the mixture is heated as described in Example 1 above to give the title compound (Formula 24.0).
EXAMPLE 24
4-�2-�4-(3-BROMO-8-CHLORO-6,11-DIHYDRO-5 H-BENZO�5,6!CYCLOHEPA�1,2-b!PYRIDIN-11-YL)-1-PIPERAZINYL!2-OXOETHYL!-N-BENZOYL-1-PIPERIDINE-CARBOXIMIDAMIDE ##STR159##
If one were to follow this procedure then the compound of Formula 25.0 would be obtained.
Phenyl 4-�2-�4-(3-bromo-8-chloro-6,11-dihydro-5 H-benzo�5,6! cyclohepta�1,2-b!pyridin-11-yl)-1-piperazinyl!-2-oxoethyl!-N-benzoyl-1-piperidinecarboximidate (Formula 24.0, prepare as described in Example 23 above) is dissolved in 2-propanol and concentrated ammonium hydroxide is added. The mixture is stirred at 25.degree. C. for 24 h and is then evaporated to dryness. The residue is triturated with diethyl ether (2.times.250 ml) and the ether is discarded. The remaining product is chromatographed on a silica gel column to give the title compound (Formula 25.0).
Alternatively, anhydrous ammonia in a suitable inert solvent such as methanol, or THF, may be used in place of ammonium hydroxide in the above reaction.
EXAMPLE 25 ##STR160##
A solution of ##STR161## (Formula B-10, (+) isomer, from Step D of Preparative Example 10 above) (0.15 g) in dichloromethane (1.5 ml) was reacted with phenyl cyanate ##STR162## (Zweifel, Synthesis 150(1980)) (0.048 ml) in the presence of di-isopropylethylamine (2 drops) at room temperature for one hour. The reaction mixture was then flash chromatographed on silica-gel (30 ml); elution with 10% methanol-methylene chloride followed by evaporation afforded the compound of Formula 27.0 as a colorless powder (0.15 g). MS (FAB): m/e 713 (MH.sup.+)
EXAMPLE 26 ##STR163##
A solution of the compound of Formula 27.0 (Example 25) (0.06 g) in dichloromethane (2.0 ml) under nitrogen was stirred with 60% sodium hydride ((0.016 g) for 2 hrs. at room temperature. The reaction mixture was then treated with ice-water and extracted with dichloromethane (5.times.5 ml); the combined extracts were dried, evaporated and the crude reaction product was chromatographed on silica-gel (20 ml). Elution with 5% methanol-methylene chloride followed by evaporation afforded the compound of Formula 29.0 as a colorless powder ((0.042 g). MS (FAB): m/e 619 (MH.sup.+)
EXAMPLE 27 ##STR164##
By following the procedure in Example 25 above, the amine ##STR165## (Formula B-13, racemic, see Preparative Example 10) (0.09 g) was reacted with phenyl cyanate (0.04 ml) in the presence of di-isopropylethylamine to afford the compound of Formula 26.0 (0.078 g) as a white powder. MS(FAB): m/e 713 (MH.sup.+)
EXAMPLE 28 ##STR166##
A solution of the compound of Formula 26.0 (Example 27) (0.24 g) and ammonium chloride (0.24 g) in tetrahydrofuran (2.0 ml) and ammonium hydroxide (2.0 ml) was heated in a pressure tube at 90.degree. C. for 1.5 hrs. The reaction mixture was then cooled in ice, diluted with water and extracted with methylene chloride (3.times.10 ml). The combined extracts were evaporated and the crude product was chromatographed on silica-gel (20 ml). Elution with 10%methanol-3%ammonia-methylene chloride afforded after evaporation the title compound (Formula 30.00 as a white solid (0.115 g). MS (FAB): m/e 636 (MH.sup.+)
EXAMPLE 29 ##STR167##
A solution of the compound of Formula 26.0 (Example 27) (0.25 g) in tetrahydrofuran (3.0 ml) was treated with a solution of 3-aminomethylpyridine (0.14 ml) in water (0.5 ml) containing 4M hydrochloric acid in dioxane (0.09 ml). The reaction mixture was heated at 70.degree. C. for 3.5 hrs, cooled to room temperature, diluted with ice-water; the pH of the mixture was adjusted to 10 with aqueous sodium hydroxide followed by extraction with methylene chloride (2.times.5 ml). The crude product was then chromatographed on silica-gel (10 ml); elution with 10%methanol-3%ammonia-methylene chloride afforded the title compound (Formula 31.0) as a colorless powder (0.28 g). MS (FAB): m/e 727 (MH.sup.+)
EXAMPLE 30 ##STR168##
By following the procedure for the preparation of the compound of Formula 29.0 in described above in Example 26, the amine 26.0 (Example 27) (0.2 g) was reacted with 60% sodium-hydride (0.022 g) to afford the title compound (Formula 28.0) (0.087 g) as a white powder. MS(FAB): m/e 619 (MH.sup.+)
EXAMPLE 31 ##STR169##
The compound of Formula B-10 ##STR170## (+-isomer, see Example 25 above) (100 mg, 1 eq), HOBT (25 mg, 1.1 eq), DEC (35 mg, 1.1 eq), DMF (5 ml) and the carboxylic acid ##STR171## (23 mg, 1.1 eq) were added to a flask and stirred at 25.degree. C. for 18 h. When TLC showed that the reaction was complete, water was added and the resulting mixture was stirred for about 5 minutes. The mixture was filtered and the precipitate was washed with water. The resulting solids were diluted in CH.sub.2 Cl.sub.2, washed with 1M NaOH (aqueous), dried over anhydrous MgSO.sub.4, filtered and the solvent was removed under vacuum. Formula 35.0 was obtained as a white solid (52 mg).
EXAMPLE 32 ##STR172##
Following the procedure of Example 31, but using ##STR173## as the carboxylic acid (21 mg, 1.1 eq), yielded the compound of Formula 36.0 as a white solid (86 mg).
EXAMPLE 33 ##STR174##
Following the procedure of Example 31, but using ##STR175## as the carboxylic acid (23 mg, 1.1 eq) yielded the compound of Formula 37.0 as a white solid (84.4 mg).
EXAMPLE 34 ##STR176##
Following the procedure of Example 31, but using ##STR177## as the carboxylic acid (30 mg, 1.1 eq) yielded the compound of Formula 38.0 as an off-white solid (0.105 g).
EXAMPLE 35 ##STR178##
Dissolve the (+) product of Preparative Example 8, Step D (0.139 g, 0.233 mmol) in 1.3 mL of DMF, stir at room temperature and add 55.2 mg (0.288 mmol) of DEC, 41.5 mg (0.307 mmol) of HOBT and 42.2 mg (0.324 mmole) of ##STR179## and 80 .mu.l (0.73 mmol) N-methylmorpholine. Stir the mixture at room temperature for 1 hr. Add the mixture to 10 mL H.sub.2 O, filter the resulting precipitate and wash with water. Dry at 10 mm Hg 40.degree. C. 18 h to give 123 mg of the product (m.p.=170.3.degree.-177.2.degree. C., heating 2.degree.-3.degree. C. per minute).
EXAMPLES 36-53
Follow the procedure of Example 35 but use the acid shown in Table 1 below instead of the acid used in Example 35 to obtain the compounds of Formula 1.7 ##STR180## wherein W is defined in Table 1. The Formula number of the compound formed is given in parenthesis below the W substituent.
TABLE 1__________________________________________________________________________EX. Acid W mp (.degree.C.)__________________________________________________________________________36 ##STR181## ##STR182## 183.7-185.937 ##STR183## ##STR184## 117.6-127.438 ##STR185## ##STR186## 154.5-157.139 ##STR187## ##STR188## 81.9-83.940 ##STR189## ##STR190## 117.1-12041 ##STR191## ##STR192## 123-12442 ##STR193## ##STR194## 151.343 ##STR195## ##STR196## 157.244 ##STR197## ##STR198## 184.8-190.045 ##STR199## ##STR200## 206.546 ##STR201## ##STR202## 120.3-127.147 ##STR203## ##STR204## 192-20348 ##STR205## ##STR206## 162-17249 ##STR207## ##STR208## 138-15150 ##STR209## ##STR210## 134-14851 ##STR211## ##STR212## 176-19152 ##STR213## ##STR214## 111-11953 ##STR215## ##STR216## 230 (d)__________________________________________________________________________
EXAMPLE 54 ##STR217##
Dissolve 1.0 equivalent of the product of Example 32 in anhydrous DMF and add 2.0 equivalents of NaH (60% in mineral oil). Stir for 0.5 hr and add 1.5 equivalent of ethyliodide and stir for 18 hr and adjust to pH7.0 with 1N HC1. Concentrate under vacuum and partition between water and dichloromethane. Dry the organic layer over magnesium sulfate and concentrate under vacuum. Chromatograph the residue on silica gel using methanol-dichloromethane, saturated with ammonia (5-95) to give the product as a white solid, mp=112.6.degree. C.
EXAMPLE 55 ##STR218##
Follow the procedure of Example 54 (Compound 40.0-B), but use benzylbromide instead of ethyliodide to obtain the product as a white solid, mp=108.degree. C.
EXAMPLE 56 ##STR219##
Follow the procedure of Example 54, but use bromoacetamide instead of ethyliodide to obtain the product as a white solid, mp=138.3.degree. C.
EXAMPLE 57 ##STR220##
Follow the procedure of Example 54, but use methane-sulfonylchloride instead of ethyliodide to obtain the product as a white solid, mp=136.3.degree. C.
EXAMPLE 58 ##STR221##
Follow the procedure of Example 54, but use tert-butyl-bromo acetate instead of ethyliodide to obtain the product as a yellow solid, mp=120.degree.-127.degree. C. EXAMPLE 59 ##STR222##
Dissolve the product of Example 58 in dichloromethane and add trifluoroacetic acid and allow to stir for 1 hr. Concentrate under vacuum. Chromatograph the residue by preparative silica gel TLC using methanol-dichloromethane (10-90) to obtain the product as a pink solid, mp=198.degree. C.
EXAMPLE 60 ##STR223##
Dissolve 1.0 equivalent of the (+) product of Preparative Example 8, Step D, in dichloromethane containing 1.0 equivalent of 2,3,4,6-tetra-O-acetyl-beta-D-glucopyranosyl isothiocyanate. Stir for 6 days then wash with 1N HCl followed by 1N NaOH. Dry the organic layer over magnesium sulfate and concentrate under vacuum. Chromatograph the residue on silica gel using methanol-dichloromethane saturated with ammonia (2-98) to obtain the product as a white solid, mp=156.7-157.9.degree. C.
EXAMPLE 61 ##STR224##
Dissolve 1.0 equivalent of the (+) product of Preparative Example 8, Step D, in DMF containing 2.5 equivalent of anhydrous sodium carbonate and 2.2 equivalents of 4-chloropyridine hydrochloride and heat at 100.degree. C. for 5 days. Cool to 25.degree. C., add water and filter the precipitate. Dissolve the precipitate in dichloromethane and wash with 1N HCl followed by 1N NaOH. Dry the organic layer over magnesium sulfate and concentrate under vacuum. Chromatograph the residue on silica gel to obtain the produce as a white solid, mp=128.6.degree. C.
EXAMPLE 62 ##STR225##
Dissolve 471.4 mg, (0.617 mmol) of the product of Example 46 in 2 mL of 6M HCl and the solution stirred at room temperature overnight. The reaction mixture was added to 20 mL water, and the resulting precipitate was filtered and washed with 0.1M HCl. Purify by flash chromatoghrapy (C-18 reverse phase silica, gradient of 50% MeOH/H.sub.2 O to 90% MeOH/H.sub.2 O). The resulting material was dissolved in MeOH and added to water to give the title compound as a white solid after filtration and drying (334.7 mg, mp 147.1.degree.-154.3.degree. C., heating 2.degree.-30.degree. C./min).
EXAMPLE 63 ##STR226##
The product of Example 62 (63.8 mg, 0.0884 mmol) was dissolved in CH.sub.2 Cl.sub.2 and trichloroacetyl isocyanate (12 .mu.L) was added. After 24 h trichloroacetyl isocyanate (12 .mu.L) was added. After 24 h the reaction mixture was purified by preparative TLC (5% EtOH/CH.sub.2 Cl.sub.2). The isolate material was dissolved in CH.sub.2 Cl.sub.2 and added to hexane and the resulting suspension evaporated to give the title compound as a white solid (51.6 mg, mp 165.4.degree.-178.50.degree. C., heating 2.degree.-3.degree. C./min).
EXAMPLE 64 ##STR227##
The product of Example 62 (151.4 mg, 0.2097 mmol) was dissolved in CH.sub.2 Cl.sub.2 and trichloroacetyl isocyanate (40 .mu.L) was added. After 15 min trichloroacetyl isocyanate (6 .mu.L) was added. After an additional 15 min MeOH was added and the mixture evaporated to dryness. The residue was suspended in MeOH/THF and K.sub.2 CO.sub.3 (anhydrous, 15.9 mg, 0.115 mmol) was added. After 3 h the reaction mixture was purified by flash chromatography (5% EtOH/CH.sub.2 Cl.sub.2). The resulting material was dissolved in MeOH and added to water to give the title compound as a white solid after filtration and drying (mp 165.4.degree.-178.5.degree. C., heating 2.degree.-3.degree. C./min).
Assays
FPT IC.sub.50 (inhibition of farnesyl protein transferase, in vitro enzyme assay), GGPT IC.sub.50 (inhibition of geranylgeranyl protein transferase, in vitro enzyme assay) and COS Cell IC.sub.50 (Cell-Based Assay) were determined following the assay procedures described in WO 95/10516, published Apr. 20, 1995. Cell Mat Assay, and anti-tumor activity (in vivo anti-tumor studies) could be determined by the assay procedures described in WO 95/10516. The disclosure of WO 95/10516 is incorporated herein by reference thereto.
Additional assays can be carried out by following essentially the same procedure as described above, but with substitution of alternative indicator tumor cell lines in place of the T24-BAG cells. The assays can be conducted using either DLD-1-BAG human colon carcinoma cells expressing an activated K-ras gene or SW620-BAG human colon carcinoma cells expressing an activated K-ras gene. Using other tumor cell lines known in the art, the activity of the compounds of this invention against other types of cancer cells could be demonstrated.
Soft Agar Assay:
Anchorage-independent growth is a characteristic of tumorigenic cell lines. Human tumor cells are suspended in growth medium containing 0.3% agarose and an indicated concentration of a farnesyl transferase inhibitor. The solution is overlayed onto growth medium solidified with 0.6% agarose containing the same concentration of farnesyl transferase inhibitor as the top layer. After the top layer is solidified, plates are incubated for 10-16 days at 37.degree. C. under 5% CO.sub.2 to allow colony outgrowth. After incubation, the colonies are stained by overlaying the agar with a solution of MTT (3-�4,5-dimethyl-thiazol-2-yl!-2,5-diphenyltetrazolium bromide, Thiazolyl blue) (1 mg/mL in PBS). Colonies can be counted and the IC.sub.50 's can be determined.
The results are given in Table 2. In Table 2 "Ex. No." stands for "Example Number". In Table 2, FPT IC.sub.50 results are less indicated otherwise.
TABLE 2______________________________________ COS Cell IC.sub.50Ex No. Formula No. FPT IC.sub.50 (.mu.M) (.mu.M)______________________________________1 2.0 44% @ 0.3 .mu.M --2 3.0 0.1 --4 5.0 0.0543 --6 7.0 0.12 --11 12.0 0.058 --12 13.0 0.11 --13 16.1 0.14 --14 17.1 0.078 --25 27.0 0.0088 0.02426 29.0 0.0023 0.01227 26.0 0.039 --28 30.0 0.023 0% @ 1 .mu.M29 31.0 0.011 >1.0 0.782 (K-ras)30 28.0 0.007 0.46 0.0387 (K-ras)______________________________________
The compound of Formula 31.0 (Example 29) had a GGPT IC.sub.50 of 7.5 .mu.M and the compound of Formula 28.0 (Example 30) had a GGPT IC.sub.50 of >9.6 .mu.M.
Additional results are given in Table 3. "Cmpd" represents "compound" and the number in that column is the compound or structure number. The FPT IC.sub.50 results are expressed as nanomolar (nM) in Table 3.
TABLE 3______________________________________Cmpd FPT IC.sub.50 Cmpd FPT IC.sub.50 Cmpd FPT IC.sub.50______________________________________ 5.0-B 3.7 15.0-B 2.0 25.0-B 0.827.0-B 4.0 28.0-B 2.3 29.0-B 2.231.0-B 3.0 36.0-B 2.6 40.0-B 7.841.0-B 1.7 42.0-B 3.2 43.0-B 3.845.0-B 2.5 46.0-B 1.9 47.0-B 2.948.0-B 2.1 61.0-B 11 62.0-B 2.866.0-B 7.3 73.0-B 4.5 78.0-B 2.680.0-B 2.6 83.0-B 3.2 84.0-B 8.591.0-B 2.5 93.0-B 5.7 94.0-B 4.0______________________________________
The following compounds had the following FPT IC.sub.50 (H-ras) results: 4.0, 290 nM; 4.0-B, 12 nM; 98.0-B, 2.3 and 2.4 nM; and 100.0-B, 5.5 nM.
The following compounds had the following FPT IC.sub.50 (K-ras) results: 27.0, 46 nM; 4.0-B, 29 nM; 5.0-B, 16 nM; and 98.0-B, 17 nM.
Additional results are given in Table 4. "Cmpd" represents "compound" and the number in that column is the compound or structure number. The Cos Cell and Soft Agar FPT IC.sub.50 results are expressed as nanomolar (nM) in Table
TABLE 4______________________________________Cmpd Cos Cell Soft Agar Cmpd Cos Cell Soft Agar______________________________________27.0 24 200 98.0-B 12 1005.0-B 8 210, 90 15.0-B 33 280, 37025.0-B >1000 >500 27.0-B 10 37028.0-B 7 120 29.0-B 21 50031.0-B 7 150 36.0-B 50 180, 17040.0-B 40 >500 41.0-B 30 >50042.0-B 27 310 43.0-B <10 20045.0-B 10 130 46.0-B 7 16547.0-B 35 450 48.0-B 47 >50062.0-B 700 >500 66.0-B 10 44073.0-B 24 360 78.0-B 30 17080.0-B -- >500 83.0-B 12 50084.0-B 11 100 91.0-B 500 >50093.0-B 30 410 94.0-B 25 210100.0-B <10 380 4.0-B 1000 --______________________________________
For preparing pharmaceutical compositions from the compounds described by this invention, inert, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, dispersible granules, capsules, cachets and suppositories. The powders and tablets may be comprised of from about 5 to about 70 percent active ingredient. Suitable solid carriers are known in the art, e.g. magnesium carbonate, magnesium stearate, talc, sugar, lactose. Tablets, powders, cachets and capsules can be used as solid dosage forms suitable for oral administration.
For preparing suppositories, a low melting wax such as a mixture of fatty acid glycerides or cocoa butter is first melted, and the active ingredient is dispersed homogeneously therein as by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool and thereby solidify.
Liquid form preparations include solutions, suspensions and emulsions. As an example may be mentioned water or water-propylene glycol solutions for parenteral injection.
Liquid form preparations may also include solutions for intranasal administration.
Aerosol preparations suitable for inhalation may include solutions and solids in powder form, which may be in combination with a pharmaceutically acceptable carrier, such as an inert compressed gas.
Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for either oral or parenteral administration. Such liquid forms include solutions, suspensions and emulsions.
The compounds of the invention may also be deliverable transdermally. The transdermal compositions can take the form of creams, lotions, aerosols and/or emulsions and can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose.
Preferably the compound is administered orally.
Preferably, the pharmaceutical preparation is in unit dosage form. In such form, the preparation is subdivided into unit doses containing appropriate quantities of the active component, e.g., an effective amount to achieve the desired purpose.
The quantity of active compound in a unit dose of preparation may be varied or adjusted from about 0.1 mg to 1000 mg, more preferably from about 1 mg. to 300 mg, according to the particular application.
The actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage for a particular situation is within the skill of the art. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day if desired.
The amount and frequency of administration of the compounds of the invention and the pharmaceutically acceptable salts thereof will be regulated according to the judgment of the attending clinician considering such factors as age, condition and size of the patient as well as severity of the symptoms being treated. A typical recommended dosage regimen is oral administration of from 10 mg to 2000 mg/day preferably 10 to 1000 mg/day, in two to four divided doses to block tumor growth. The compounds are non-toxic when administered within this dosage range.
The following are examples of pharmaceutical dosage forms which contain a compound of the invention. The scope of the invention in its pharmaceutical composition aspect is not to be limited by the examples provided.
PHARMACEUTICAL DOSAGE FORM EXAMPLES
EXAMPLE A--TABLETS
______________________________________No. Ingredients mg/tablet mg/tablet______________________________________1. Active compound 100 5002. Lactose USP 122 1133. Corn Starch, Food Grade, 30 40 as a 10% paste in Purified Water4. Corn Starch, Food Grade 45 405. Magnesium Stearate 3 7 Total 300 700______________________________________
Method of Manufacture
Mix Item Nos. 1 and 2 in a suitable mixer for 10-15 minutes. Granulate the mixture with Item No. 3. Mill the damp granules through a coarse screen (e.g., 1/4 ", 0.63 cm) if necessary. Dry the damp granules. Screen the dried granules if necessary and mix with Item No. 4 and mix for 10-15 minutes. Add Item No. 5 and mix for 1-3 minutes. Compress the mixture to appropriate size and weigh on a suitable tablet machine.
EXAMPLE B--CAPSULES
______________________________________No. Ingredient mg/capsule mg/capsule______________________________________1. Active compound 100 5002. Lactose USP 106 1233. Corn Starch, Food Grade 40 704. Magnesium Stearate NF 7 7 Total 253 700______________________________________
Method of Manufacture
Mix Item Nos. 1, 2 and 3 in a suitable blender for 10-15 minutes. Add Item No. 4 and mix for 1-3 minutes. Fill the mixture into suitable two-piece hard gelatin capsules on a suitable encapsulating machine.
While the present invention has been described in conjunction with the specific embodiments set forth above, many alternatives, modifications and variations thereof will be apparent to those of ordinary skill in the art. All such alternatives, modifications and variations are intended to fall within the spirit and scope of the present invention.
Claims
  • 1. A compound of the formula: ##STR228## or a pharmaceutically acceptable sat or solvate thereof, wherein: one of a, b, c and d represents N or NR.sup.9 wherein R.sup.9 is O.sup.13, --CH.sub.3 or --(CH.sub.2).sub.n CO.sub.2 H wherein n is 1 to 3, and the remaining a, b, c and d groups represent CR.sup.1 or CR.sup.2 ; or
  • each of a, b, c, and d are independently selected from CR.sup.1 or CR.sup.2 ;
  • each R.sup.1 and each R.sup.2 is independently selected from H, halo, --CF.sub.3, --OR.sup.10, --COR.sup.10, --SR.sup.10, --S(O).sub.t R.sup.11 (wherein t is 0, 1 or 2), --SCN, --N(R.sup.10).sub.2, --NR.sup.10 R.sup.11, --NO.sub.2, --OC(O)R.sup.10, --CO.sub.2 R.sup.10, --OCO.sub.2 R.sup.11, --CN, --NHC(O)R.sup.10, --NHSO.sub.2 R.sup.10, --CONHR.sup.10, --CONHCH.sub.2 CH.sub.2 OH, --NR.sup.10 COOR.sup.11, ##STR229## --SR.sup.11 C(O)OR.sup.11, --SR.sup.11 N(R.sup.75).sub.2 wherein each R.sup.75 is independently selected from H and --C(O)OR.sup.11, benzotriazol-1-yloxy, tetrazol-5-yl-thio, or substituted tetrazol-5-ylthio, alkynyl, alkenyl or alkyl, said alkyl or alkenyl group optionally being substituted with halo, --OR.sup.10 or --CO.sub.2 R.sup.10 ;
  • R.sup.3 and R.sup.4 are the same or different and each independently represents H, any of the substituents of R.sup.1 and R.sup.2, or R.sup.3 and R.sup.4 taken together represent a saturated or unsaturated C.sub.5 -C.sub.7 fused ring to the benzene ring (Ring III);
  • R.sup.5, R.sup.6, R.sup.7 and R.sup.8 each independently represents H, --CF.sub.3, --COR.sup.10, alkyl or aryl, said alkyl or aryl optionally being substituted with --OR.sup.10, --SR.sup.10, --S(O).sub.t R.sup.11, --NR.sup.10 COOR.sup.11, --N(R.sup.10).sub.2, --NO.sub.2, --COR.sup.10, --OCOR.sup.10, --OCO.sub.2 R.sup.11, --CO.sub.2 R.sup.10, --OPO.sub.3 R.sup.10, or R.sup.5 is combined with R.sup.6 to represent .dbd.O or .dbd.S and/or R.sup.7 is combined with R.sup.8 to represent .dbd.O or .dbd.S;
  • R.sup.10 represents H, alkyl, aryl, or aralkyl;
  • R.sup.11 represents alkyl or aryl;
  • X represents N, CH or C, which C may contain an optional double bond (represented by the dotted line) to carbon atom 11;
  • the dotted line between carbon atoms 5 and 6 represents an optional double bond, such that when a double bond is present, A and B independently represent --R.sup.10, halo, --OR.sup.11, --OCO.sub.2 R.sup.11 or --OC(O)R.sup.10, and when no double bond is present between carbon atoms 5 and 6, A and B each independently represent H.sub.2, --(OR.sup.11).sub.2 ; H and halo, dihalo, alkyl and H, (alkyl)2, --H and --OC(O)R.sup.10, H and --OR.sup.10, .dbd.O, aryl and H, .dbd.NOR.sup.10 or --O-- (CH.sub.2).sub.p --O-- wherein p is 2, 3 or 4; and
  • W is selected from the group consisting of:
  • (1) cyano;
  • (2) --C(O)R.sup.12 wherein R.sup.12 is selected from:
  • (a) a heteroaryl group;
  • (b) H;
  • (c) alkyl; or
  • (d) a substituent of the formula: ##STR230## wherein R.sup.28 is selected from --OC(O)R.sup.29, --OH, --OC(O)NHC(O)CCl.sub.3, or --OC(O)NH.sub.2, and wherein R.sup.29 is alkyl;
  • (3) an imidate represented by the formula: ##STR231## wherein R.sup.13 is selected from the group consisting of: (a) H, (b) CN, (c) --SO.sub.2 -alkyl, (d) --C(O)-aryl, (e) --SO.sub.2 NR.sup.10 R.sup.15, (f) --C(O)NR.sup.10 R.sup.15, (g) --OR.sup.10 and (h) --C(O)NR.sup.10 C(O)NR.sup.10 R.sup.15 ; R.sup.14 is aryl; and R.sup.10 and R.sup.15 are independently selected from the group consisting of: H, alkyl, aryl and aralkyl;
  • (4) an imidamido represented by the formula: ##STR232## wherein R.sup.13 is selected from the group consisting of (a)H, (b) CN, (c) --SO.sub.2 -alkyl, (d) --C(O)-aryl, (e) --SO.sub.2 NR.sup.10 R.sup.15, (f) --C(O)NR.sup.10 R.sup.15, (g) --OR.sup.10 and (h) --C(O)NR.sup.10 C(O)NR.sup.10 R.sup.15 ; R.sup.16 is selected from the group consisting of: alkyl, aralkyl, aryl, cycloalkyl, heteroaryl, heteroaralkyl and heterocycloalkyl; and R.sup.10 and R.sup.15 are as defined above; and R.sup.10 and R.sup.16 are independently selected from the above defined groups;
  • (5) 1-amino-2-nitroethylene derivatives of the formula: ##STR233## wherein R.sup.10 is as defined above; and (6) a substituent of the formula: ##STR234##
  • 2. The compound of claim 1 wherein R.sup.2 is H; R.sup.1 is selected from the group consisting of: Br and Cl; R.sup.3 is selected from the group consisting of: Br and Cl; R.sup.4 is selected from the group consisting of: H, Br and Cl; R.sup.5, R.sup.6, R.sup.7 and R.sup.8 are H; A and B are each H.sub.2 ; and the optional bond between C5 and C6 is absent.
  • 3. The compound of claim 2 wherein R.sup.4 is H.
  • 4. The compound of claim 2 wherein R.sup.4 is selected from the group consisting of: Cl or Br.
  • 5. The compound of claim 4 wherein X is CH.
  • 6. The compound of claim 5 wherein W is selected from the group consisting of:
  • (1) --CN;
  • (2) --C(O)R.sup.12 wherein R.sup.12 is selected from the group consisting of: H, alkyl, ##STR235## (3) an imidate wherein R.sup.13 is selected from the group consisting of: (a) --CN;
  • (b) H;
  • (c) --SO.sub.2 NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are selected from the group consisting of: H and alkyl;
  • (d) --C(O)NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are selected from the group consisting of: H and alkyl;
  • (e) --SO.sub.2 -alkyl; and
  • (f) --C(O)-aryl;
  • (4) an imidamido wherein R.sup.13 is selected from the group consisting of:
  • (a) CN;
  • (b) H;
  • (c) --OR.sup.14 ;
  • (d) --NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl;
  • (e) --SO.sub.2 NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl;
  • (f) --C(O)NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl;
  • (g) --SO.sub.2 -alkyl; and
  • (h) --C(O)-aryl; and
  • (5) a 1-amino-2-nitroethylene derivative wherein R.sup.10 is alkyl.
  • 7. The compound of claim 6 wherein R.sup.14 for said imidate group is phenyl; and R.sup.10 and R.sup.16 for said imidamido group is selected from the group consisting of: H and heteroaralkyl.
  • 8. The compound of claim 3 wherein X is N.
  • 9. The compound of claim 8 wherein W is selected from the group consisting of:
  • (1) --CN;
  • (2) --C(O)R.sup.12 wherein R.sup.12 is selected from the group consisting of: H, alkyl, ##STR236## (3) an imidate wherein R.sup.13 is selected from the group consisting of: (a) --CN;
  • (b) H;
  • (c) --SO.sub.2 NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are selected from the group consisting of: H and alkyl;
  • (d) --C(O)NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are selected from the group consisting of: H and alkyl;
  • (e) --SO.sub.2 -alkyl; and
  • (f) --C(O)-aryl;
  • (4) an imidamido wherein R.sup.13 is selected from the group consisting of:
  • (a) CN;
  • (b) H;
  • (c) --OR.sup.14 ;
  • (d) --NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl;
  • (e) --SO.sub.2 NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl;
  • (fl --C(O)NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl;
  • (g) --SO.sub.2 -alkyl; and
  • (h) --C(O)-aryl; and
  • (5) a 1-amino-2-nitroethylene derivative wherein R.sup.10 is alkyl.
  • 10. The compound of claim 9 wherein R.sup.1 is Br and R.sup.3 is Cl.
  • 11. The compound of claim 10 wherein R.sup.14 for said imidate group is phenyl; and R.sup.10 and R.sup.16 for said imidamido group is selected from the group consisting of: H and heteroaralkyl.
  • 12. The compound of claim 1 selected from: ##STR237## wherein R.sup.1, R.sup.3 and R.sup.4 are each independently selected from halo; and A, B, X and W are as defined in claim 1.
  • 13. The compound of claim 12 wherein R.sup.1 is selected from the group consisting of: Br and Cl; R.sup.3 and R.sup.4 are independently selected from the group consiting of: Br and Cl; A and B are each H.sub.2 ; and the optional bond between C5 and C6 is absent.
  • 14. The compound of claim 13 wherein R.sup.1 is Br; and R.sup.3 is Cl; and R.sup.4 is Br.
  • 15. The compound of claim 14 wherein X is CH.
  • 16. The compound of claim 15 wherein W is selected from the group consisting of:
  • (1) --CN;
  • (2) --C(O)R.sup.12 wherein R.sup.12 is selected from the group consisting of: H, alkyl, ##STR238## (3) an imidate wherein R.sup.13 is selected from the group consisting of: (a) --CN;
  • (b) H;
  • (c) --SO.sub.2 NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are selected from the group consisting of: H and alkyl;
  • (d) --C(O)NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are selected from the group consisting of: H and alkyl;
  • (e) --SO.sub.2 -alkyl; and
  • (f) --C(O)-aryl;
  • (4) an imidamido wherein R.sup.13 is selected from the group consisting of:
  • (a) CN;
  • (b) H;
  • (c) --OR.sup.14 ;
  • (d) --NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl;
  • (e) --SO.sub.2 NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl;
  • (f) --C(O)NR.sup.10 R.sup.15 wherein R.sup.10 and R.sup.15 are independently selected from the group consisting of: H and alkyl;
  • (g) --SO.sub.2 -alkyl; and
  • (h) --C(O)-aryl; and
  • (5) a 1-amino-2-nitroethylene derivative wherein R.sup.10 is alkyl.
  • 17. The compound of claim 16 wherein R.sup.14 for said imidate group is phenyl; and R.sup.10 and R.sup.16 for said imidamido group is selected from the group consisting of: H and heteroaralkyl.
  • 18. The compound of claim 16 wherein W is selected from the group consisting of:
  • (1) --C(O)R.sup.12 wherein R.sup.12 is selected from the group consisting of:H, --CH.sub.3, ##STR239## (2) an imidate wherein R.sup.13 is selected from the group consisting of: (a) CN; (b) --C(O)NH.sub.2 ; (c) H; (d) --SO.sub.2 NH.sub.2 ; (e) --SO.sub.2 NHCH.sub.3 ; (f) --SO.sub.2 N(CH.sub.3).sub.2 ; (g) --C(O)NHCH.sub.3 ; (h) --SO.sub.2 CH.sub.3 and (i) --C(O)C.sub.6 H.sub.5 ;
  • (3) an imidamido wherein R.sup.13 is selected from the group consisting of: (a) CN; (b) H; (c) --OCH.sub.3 ; (d) --OH; (e) --NH.sub.2 ; (f) --N(CH.sub.3).sub.2 ; (g) --SO.sub.2 NH.sub.2 ; (h) --SO.sub.2 NHCH.sub.3 ; (i) --SO.sub.2 N(CH.sub.3).sub.2 ; (j) --C(O)NH.sub.2 ; (k) --C(O)NHCH.sub.3 ; (l) --SO.sub.2 CH.sub.3 and (m) --C(O)C.sub.6 H.sub.5 ; and
  • (4) a 1-amino-2-nitroethylene derivative wherein R.sup.10 is --CH.sub.3.
  • 19. The compound of claim 18 wherein R.sup.14 for said imidate group is phenyl; and R.sup.10 and R.sup.16 for said imidamido group is selected from the group consisting of: H and heteroaralkyl.
  • 20. The compound of claim 19 wherein for said imidate group R.sup.14 is phenyl; and for said imidamido groups R.sup.10 and R.sup.16 are independently selected from the group consisting essentially of: H and ##STR240##
  • 21. The compound of claim 16 having the structure ##STR241##
  • 22. The imidate compound of claim 1 selected from the group consisting of: ##STR242##
  • 23. The imidamido compound of claim 1 selected from the group consisting of: ##STR243##
  • 24. The cyano compound of claim 1 selected from the group consisting of: ##STR244##
  • 25. The compound of claim 1 selected from the group consisting of: ##STR245##
  • 26. The compound of claim 1 selected from the group consisting of: ##STR246##
  • 27. The compound of claim 1 selected from -the group consisting of: ##STR247##
  • 28. A method of treating tumor cells expressing an activated ras oncogene comprising administering an effective amount of a compound of claim 1.
  • 29. The method of claim 28 wherein the cells treated are pancreatic tumor cells, lung cancer cells, myeloid leukemia tumor cells, thyroid follicular tumor cells, myelodysplastic tumor cells, epidermal carcinoma tumor cells, bladder carcinoma tumor cells, colon tumors cells, breast tumor cells or prostate tumor cells.
  • 30. A method of treating tumor cells wherein the Ras protein is activated as a result of oncogenic mutation in genes other than the Ras gene comprising administering an effective amount of a compound of claim 1.
  • 31. A method of inhibiting farnesyl protein transferase comprising the administration of an effective amount of the compound of claim 1.
  • 32. A pharmaceutical composition for inhibiting the abnormal growth of cells comprising an effective amount of compound of claim 1 in combination with a pharmaceutically acceptable carrier.
US Referenced Citations (15)
Number Name Date Kind
4826853 Piwinski et al. May 1989
5089496 Piwinski et al. Feb 1992
5151423 Piwinski et al. Sep 1992
5393890 Syoji et al. Feb 1995
5661152 Bishop et al. Aug 1997
5672611 Doll et al. Sep 1997
5684013 Afonso et al. Nov 1997
5696121 Bishop et al. Dec 1997
5700806 Doll et al. Dec 1997
5703090 Afonso et al. Dec 1997
5712280 Doll et al. Jan 1998
5714609 Bishop et al. Feb 1998
5719148 Bishop et al. Feb 1998
5721236 Bishop et al. Feb 1998
5728703 Bishop et al. Mar 1998
Foreign Referenced Citations (13)
Number Date Country
0270818 Jun 1988 EPX
0396083 Nov 1990 EPX
0495484 Jul 1992 EPX
WO9510515 Apr 1995 WOX
WO9510516 Apr 1995 WOX
WO9510514 Apr 1995 WOX
WO9515949 Jun 1995 WOX
WO9630362 Oct 1996 WOX
WO9631478 Oct 1996 WOX
WO9630363 Oct 1996 WOX
WO9630018 Oct 1996 WOX
WO9631477 Oct 1996 WOX
WO9723478 Jul 1997 WOX