WO1998020874A1 - Synthetic methods for the preparation of indolylquinones and mono- and bis-indolylquinones prepared therefrom - Google Patents

Synthetic methods for the preparation of indolylquinones and mono- and bis-indolylquinones prepared therefrom Download PDF

Info

Publication number
WO1998020874A1
WO1998020874A1 PCT/US1997/020557 US9720557W WO9820874A1 WO 1998020874 A1 WO1998020874 A1 WO 1998020874A1 US 9720557 W US9720557 W US 9720557W WO 9820874 A1 WO9820874 A1 WO 9820874A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
aryl
compound
alkylaryl
alkyl
Prior art date
Application number
PCT/US1997/020557
Other languages
French (fr)
Inventor
Peng C. Tang
G. David Harris
Original Assignee
Sugen, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sugen, Inc. filed Critical Sugen, Inc.
Priority to DE69734246T priority Critical patent/DE69734246T2/en
Priority to EP97946641A priority patent/EP0959881B1/en
Priority to CA002271737A priority patent/CA2271737A1/en
Priority to JP52274698A priority patent/JP2001504820A/en
Priority to AU51770/98A priority patent/AU5177098A/en
Priority to AT97946641T priority patent/ATE305000T1/en
Publication of WO1998020874A1 publication Critical patent/WO1998020874A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/12Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/42Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals

Definitions

  • the present invention relates to novel synthetic methods for the preparation of both known and novel indolylquinones.
  • Many indolylquinones in particular the class of indolylquinones known as the asterriquinones, have utility in the treatment of cell proliferative disorders such as cancer.
  • many indolylquinones are known to be useful as dyes.
  • indolylquinones are also known to exhibit antifungal and antibacterial properties.
  • indolylquinones are isolated from natural sources and are not prepared synthetically.
  • the present invention is directed to a synthetic method for the preparation of indolylquinones, and to novel compounds prepared using said method.
  • the methods of the present invention are directed to synthetic reactions involving indoles and halo-quinones in solvent and in the presence of a metal carbonate. These methods provide a direct and simple means of preparing the compounds of interest, indolylquinones.
  • indolylquinones Another class of indolylquinones known as the asterriquinones in which the nitrogen of the indole ring is substituted, has been shown to exhibit antitumor activity.
  • Arai et al proposed the general name "asterriquinones” for the class of indolylquinones based upon asterriquinone (1981, "Metabolic Products of Aspergillus terreu ⁇ IV. Metabolites of the Strain IFO 8835. (2) The Isolation and Chemical Structure of Indolyl Benzoquinone Pigments," Chem. Phar . Bull. 29(4): 961-969).
  • the term “asterriquinone” has a more general meaning, and is used interchangeably with the term “indolylquinone.
  • Yama oto et.. l . disclose the antitumor activity of asterriquinone, i.e., 2 , 5-bis[N- (1" , l"-dimethyl-2"- propenyl) indol-3 '-yl] -3 , 6-dihydroxy-l,4-benzoquinone, and its isolation from the fungus Aspergillu ⁇ terreus (1976, "Antitumor Activity of Asterriquinone, a Metabolic Product from Aspergillus terreus ,” Gann 67:623-624).
  • Arai et al disclose the isolation and characterization of 11 different kinds of bisindolyl-dimethoxyl-p- benzoquinones from Aspergillus terreus . Id. The isolation and structural determination of a number of other asterriquinones have also been reported. (Arai et al . 1981, "Metabolic Products of Aspergillu ⁇ terreus VI. Metabolites of the Strain IFO 8835. (3) the Isolation and Chemical Structures of Colorless Metabolites," Chem. Pharm. Bull. 29(4): 1005-1012; Kaji et al . , 1994, “Four New Metabolites of Aspergillu ⁇ Terreus", Chem. Pharm. Bull. 42(8): 1682-1684).
  • benzoquinone derivatives having aziridinyl groups in the molecule such as itomycin C, carbazilquinone or "E 39" are well known potent anticancer agents
  • replacement of the functional groups at the 3 and 6 positions in the benzoquinone moiety of asterriquinone failed to enhance its antitumor potency. Id.
  • the introduction of an ethyleneimino group into the molecule did not increase antitumor activity.
  • a dimethylallyl derivative of asterriquinone showed moderate activity against the ascites and solid tumors of Ehrlich carcinoma, while an allyl derivative did not. It was suggested that in order to enhance the antitumor activity, it may be necessary not only to alter the pKa value by alkylation, but also to introduce hydrophilic groups into the molecule.
  • asterriquinone and some of its analogues have also been shown to be strong inhibitors of HIV-reverse transcriptase (Ono et al . , 1991, "Inhibition of HIV-Reverse Transcriptase Activity by Asterriquinone and its Analogues," Biochem. Biophys. Res. Commun. 174(1): 56-62).
  • indolylquinones have utility as antitumor agents for the treatment of cancer and other cell proliferative disorders. These compounds are believed to arrest the growth of tumors by interfering with the signal transduction pathways that regulate cell proliferation and differentiation.
  • Protein phosphorylation is a common regulatory mechanism used by cells to selectively modify proteins carrying signals that regulate cell proliferation and differentiation.
  • the proteins that execute these biochemical modifications are a group of enzymes known as protein kinase ⁇ . They may further be defined by the amino acid that they target for phosphorylation.
  • protein kinase ⁇ are the tyrosine kinases (PTKs) which selectively pho ⁇ phorylate a target protein on its tyrosine residues.
  • Protein tyrosine kinases comprise a large family of proteins, including many growth factor receptors and potential oncogenes. Tyrosine kinases can be cytoplasmic, non-receptor-type enzymes and act as a key component of a signal transduction pathway which regulates cell functions such as cell division, differentiation and survival.
  • Adaptor proteins are intracellular proteins having characteristic conserved peptide domains (SH2 and/or SH3 domains, as described below) which are critical to the signal transduction pathway. Such adaptor proteins serve to link protein tyrosine kinases, especially receptor-type protein tyrosine kinases to downstream intracellular signalling path- ways such as the RAS signalling pathway. It is thought that such adaptor proteins may be involved in targeting signal transduction proteins to the correct site in the plasma membrane or subcellular compartments, and may also be involved in the regulation of protein movement within the cell. The profound cellular effects mediated by tyrosine kinases and adaptor molecules have made them attractive targets for the development of new therapeutic molecules.
  • indolylquinone ⁇ such as asterriquinones
  • research into the therapeutic activities of indolylquinones and efforts to obtain indolylquinones with enhanced therapeutic activity have both been limited by the lack of reliable sources for these compounds. Indeed, isolation of indolylquinones from natural sources requires multiple steps and produces only milligram quantities of the target molecules. Further, evaluation of the activities of novel indolylquinones has necessarily been confined to those compounds which can be obtained by chemical modification of known compounds that can be isolated from natural sources. Clearly, a synthetic routine to these compounds would be invaluable to the art.
  • This solid state reaction yields about 11% of 2,5-dibromo- 5,6-bis(5-bromo-3-indolyl) -1, 4-quinone.
  • the 2 , 5-dibromo-5, 6- bis (5-bromo-3-indolyl) -1,4-quinone is then treated with benzalcohol and sodium hydroxide to give 2 , 5-bis (benzyloxy) - 3,6-bis (5-bromo-3-indolyl) -1,4-quinone in 45% yield.
  • This product is then reacted with hydrogen gas in the presence of a 10% Pd on activated charcoal catalyst, followed by treatment with acetic anhydride in pyridine to give 1,2,4,5- tetracetoxy-3,6-bis(5-bromo-3-indolyl) benzene.
  • Reaction of this compound with a complex of isopentenyl bromide and tetracarbonyl nickel gives 1, 2 , 4,5-tetracetoxy-3, 6-bis[5- (3- methyl-2-butenyl) 3-indolyl] benzene.
  • This compound is then reacted with sodium hydroxide and oxygen to give cochliodinol. According to Horcher et al .
  • Horcher et al . report that this method resulted in isolation of only milligram quantities of cochliodinol in a very low overall yield.
  • Horcher et al . indicate that conducting the initial reaction of bromanil with 5- bromoindole in smaller batches results in better yields. This suggests that the methods of Horcher et al . are unsuited for production of bis-indolylquinones on a large scale.
  • the methods of Horcher et al . would be prohibitively ultistep, and would likely result in isolation of only milligram quantities of the target indolylquinones.
  • these methods require high temperature and manipulation in dry box.
  • the present invention provides a method for the synthesis of indolylquinones which comprise reacting a substituted or unsubstituted 2 , 5-dihalo-l, 4-benzoquinone with one or more substituted or unsubstituted indoles in a polar organic solvent and in the presence of metal carbonate.
  • the present invention provides a method for preparing a symmetrical indolylquinone compound of the formula I:
  • Rl and R2 are each independently Br, Cl, F, I, H, OH or -OCOR, wherein R is, lower alkyl, aryl or alkylaryl;
  • R"l is H, C ⁇ C- alkyl, C 2 -C 7 alkenyl, C 2 -C 7 alkynyl, arylalkyl or aryl;
  • R3 to R7 are each independently hydrogen, branched or unbranched C ⁇ C,, alkyl, alkylcarboxy, C 2 -C m alkenyl, C 2 -C m alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C ⁇ C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n
  • Rl and R2 are preferably Br, Cl, F or H; and least preferably OH.
  • the method comprises reacting a substituted or unsubstituted 2,5- dihalo-l,4-benzoquinone, preferably a 2,5-dibromo-l,4- benzoquinone compound of the formula II:
  • Rl and R2 are as defined above; with at least one indole of the formula III:
  • R"l and R3-R7 are a ⁇ defined above.
  • the reaction i ⁇ carried out in a polar organic ⁇ olvent and in the presence of metal carbonate under mild conditions which are further discussed below.
  • the method may further comprise reacting the indolylquinone compound of formula I with an alkali metal hydroxide to produce a compound of the formula IV:
  • the invention ' encompasses reacting the indolylquinone compound of formula I wherein Rl and R2 are Br with an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula:
  • R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
  • the present invention provides a method for preparing an asymmetrical indolylquinone compound of the formula VI:
  • Rl and R2 are each independently Br, Cl, F, I, H, OH, or —OCOR, wherein R is lower alkyl, aryl or alkylaryl; R"l and R"2 are each independently H, C ⁇ C, alkyl, C 2 -C 7 alkenyl, C 2 -C 7 alkynyl, arylalkyl or aryl; and R3 to R12 are each independently hydrogen, branched or unbranched C 1 -C n alkyl, alkylcarboxy, C 2 -C m alkenyl, C 2 -C m alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C x -C n alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2
  • R"l and R3-R7 are as defined above; in a polar organic solvent and in the presence of metal carbonate;
  • step (b) reacting the intermediate product of step (a) with one equivalent of a second indole of the formula IV:
  • R"2 and R8-R12 are as defined above. Both reactions are carried out in a polar organic solvent and in the presence of metal carbonate under mild conditions which are further discussed below.
  • the invention also encompas ⁇ es further reacting the indolylquinone compound of formula VI with an alkali metal hydroxide to produce a compound of the formula VIII:
  • R"l, R"2 and R3-R12 are as defined above.
  • the invention encompasses reacting the indolylquinone of formula VI wherein Rl and R2 are Br, F, Cl or I with an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula IX:
  • R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
  • the present invention provides a method for preparing an indolylquinone compound of the formula X:
  • R"l is H, C 1 -C 7 alkyl, C 2 -C 7 alkenyl, C 2 -C 7 alkynyl, arylalkyl or aryl; and R3 to R7 are each independently hydrogen, branched or unbranched C ⁇ C,, alkyl, alkylcarboxy, C 2 -C m alkenyl, C 2 -C m alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C ⁇ C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfona ido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer of 0 to 12, preferably 1-7, and m is an integer of 0 to 12 , preferably 1-7.
  • This embodiment of the inventive method involves reacting 2, 3
  • R"l and R3-R7 are as defined above.
  • the reaction is carried out in a polar organic solvent in the presence of metal carbonate.
  • the method also encompasses further reacting the indolylquinone compound of formula X with an alkali metal hydroxide to produce a compound of the formula:
  • the invention also encompasses a method which further comprises reacting the indolylquinone compound of formula X with an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula:
  • R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
  • the present invention provides a method for preparing an indolylquinone compound of the formula XIV:
  • R"l and R"2 are each independently H, C ⁇ C, alkyl, C 2 -C 7 alkenyl, C 2 -C 7 alkynyl, arylalkyl or aryl; and R3 to R12 are each independently hydrogen, branched or unbranched C 1 -C n alkyl, alkylcarboxy, C 2 -C m alkenyl, C 2 -C ra alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C ⁇ C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfona ido, amino, ercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer of 0 to 12, preferably 1-7, and m is an integer of 0 to 12, preferably 1- 7.
  • R"l and R3-R7 are as defined above; in a polar organic solvent and in the presence of metal carbonate;
  • R"2 and R8-R12 are as defined above. Both reactions are carried out in a polar organic solvent and in the presence of metal carbonate under mild conditions which are further discussed below.
  • the invention also encompasses further reacting the indolylquinone compound of formula XIV with an alkali metal hydroxide to produce a compound of the formula VIII:
  • R"l, R"2 and R3-R12 are as defined above.
  • the invention encompas ⁇ es reacting the indolylquinone of formula VI wherein Rl and R2 are Br, F, Cl or I with an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula IX:
  • R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
  • the present invention provides a method for preparing a mono-indolylquinone compound of the formula XI:
  • Rl, R2 and R30 are each independently Br, Cl, F, I, H, OH or -OCOR, wherein R is lower alkyl, aryl or alkylaryl;
  • R"l is H, C x -C 7 alkyl, C 2 -C 7 alkenyl, C 2 -C 7 alkynyl, arylalkyl or aryl;
  • R3 to R7 are each independently hydrogen, branched or unbranched C ⁇ C,, alkyl, alkylcarboxy, C 2 -C m alkenyl, alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C 1 -C n alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer from 0 to 12 and m is an integer from 0 to 12.
  • This method comprises reacting a substituted or unsubstituted 2,5-dibromo-l,4-benzoquinone compound of the formula:
  • R"l and R3-R7 are as defined above.
  • the reaction is carried out in a polar organic ⁇ olvent and in the presence of metal carbonate.
  • the method of the present invention may further comprise reacting the indolylquinone compound of formula XI with an alkali metal hydroxide to produce a compound of the formula:
  • the method may further comprise reacting the indolylquinone compound of formula XI wherein Rl, R2 and R30 are Br, F, Cl or I with a mixture of an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula: wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
  • the present invention further encompas ⁇ e ⁇ methods for producing large quantitie ⁇ of known, naturally occurring a ⁇ terriquinone ⁇ in high purity and in high yield.
  • the present invention is directed to known, synthetically prepared naturally occurring a ⁇ terriquinone ⁇ of high purity which are obtainable in large quantitie ⁇ and in high yield.
  • the invention also encompasses the preparation of novel monoindolylquinones, i.e., compounds ⁇ ub ⁇ tituted with only one indole, and the monoindolylquinone compound ⁇ , as de ⁇ cribed below.
  • the pre ⁇ ent invention relates to a synthetic route for the preparation of a wide variety of indolylquinones including asterriquinone compounds having interesting antitumor activity.
  • the invention provides a major contribution to the art in that naturally occurring compounds with important therapeutic properties can be prepared in large quantities, e.g., quantities of about 1 gram or more, with purity in excess of about 95% and in high yield.
  • the availability of large quantities allows the skilled artisan to more quickly and easily test these naturally occurring compounds.
  • Prior to the present invention such compounds were isolated from natural sources in limited quantities (e.g., about 750 mg or less) and low purity (e.g., les ⁇ than about 95%).
  • the present invention provides a means for preparing novel analogues of the naturally occurring indolylquinones, which analogues may prove to be of greater interest than the naturally occurring compounds for their therapeutic activity or other properties.
  • the pre ⁇ ent invention provides a solvent based reaction of indole ⁇ and haloquinone ⁇ in the presence of metal carbonate. It has been discovered that the reaction of indoles and halo-quinone ⁇ u ⁇ ing a polar organic solvent and metal carbonate provides a rapid and efficient reaction under mild conditions, which include but are not limited to mild temperatures, short reaction times and standard/ambient pres ⁇ ure ⁇ . Thu ⁇ , the pre ⁇ ent method i ⁇ well ⁇ uited for large ⁇ cale and commercial production of indolylquinones.
  • the present invention provides methods for the preparation of indolylquinone ⁇ which involve the reaction of a 2 , 5-dihalo-l, 4-benzoquinone of the formula II with at lea ⁇ t one indole of the formula III.
  • a ⁇ y metrical bi ⁇ -indolylquinone of the formula I is obtained as the product.
  • step-wise or concurrent addition of two different indoles may be u ⁇ ed to obtain an asymmetrical bis-indolylquinone of the formula VI.
  • an asymmetrical bis-indolylquinone may be made by the reaction of a 2 , 5-dihalo-l, 4-benzoquinone with about one equivalent of a first'indole, followed by addition of about 1.5 equivalents of a second indole.
  • the preparation of asymmetrical bis- indolylquinones is carried out in the presence of about 3 equivalents of metal carbonate.
  • mixtures of two or more indoles may be reacted with the starting quinone to give a mixture of symmetrical and asymmetrical bis-indolylquinones.
  • the controlled addition of one equivalent of at least one indole to the starting quinone in the presence of about one equivalent of metal carbonate can be used to obtain one or more monoindolylquinones.
  • the preferred 2 , 5-dihalo-l, 4-quinones u ⁇ eful in the pre ⁇ ent invention are the 2 ,5-dibromo-l,4-benzoquinones of formula II, which may be substituted or unsub ⁇ tituted, wherein Rl and R2 are each independently Br, Cl, F, I, OH, H or -OCOR, wherein R is lower alkyl, aryl or alkylaryl.
  • the 2 , 5-dichloro-, difluoro- or diiodo-1,4- benzoquinone ⁇ may be u ⁇ ed in the method ⁇ of the invention.
  • the indole ⁇ u ⁇ eful in the pre ⁇ ent invention may be ⁇ ubstituted at the 1, 2, 3, 4, 5 or 7 positions with hydrogen, branched or unbranched C ⁇ C,, alkyl, alkylcarboxy, C 2 -C ra alkenyl, C 2 -C m alkynyl, alkenylcarboxy, aryl, akylaryl, hydroxy, hydroxyalkyl, C ⁇ C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, preferably 1-7, and m is an integer from 0 to 12, preferably 1-7.
  • the indoles u ⁇ ed in the present invention are substituted at the 2 position.
  • Least preferred indoles are the unsubstituted indoles.
  • Certain indoles useful in the present invention are available from commercial sources such as the Aldrich Chemical Company, Milwaukee, WI.
  • the indoles may be prepared via a cyclization procedure according to the method of Verley and Bedure, 1925, Bull. Soc. Chim. Fr. (37): 190.
  • any non-nucleophilic, aprotic solvent may be used in the methods of the invention. Mixtures of solvents may also be used.
  • Preferred ⁇ olvent ⁇ are inert or non-reactive, polar organic solvent ⁇ including but not limited to acetonitrile, dimethyl formamide (DMF) and tetrahydrofuran (THF) .
  • a particularly preferred solvent is acetonitrile.
  • the solvent volume will depend upon the scale of the reactors, and may range from a few milliliters up to a multi-liter volumes useful in large-scale production. Reactant concentrations are set forth below.
  • the metal carbonate used in the methods of the present invention assi ⁇ ts the reaction of the 2,5-dihalo-l,4-quinone and the indole by scavenging the hydrogen bromide by-product formed in the reaction.
  • Any metal carbonate or mixture of metal carbonates may be u ⁇ ed; however, cesium carbonate, potas ⁇ iu carbonate, sodium carbonate, lithium carbonate and mixture ⁇ thereof are preferred.
  • a particularly preferred metal carbonate i ⁇ cesium carbonate.
  • the amount of metal carbonate used in the method ranges from about 2 to about 10 equivalent ⁇ based on the haloquinone; preferably 2 to 5 equivalent ⁇ ; and most preferably 3 to 4 equivalents.
  • the reaction may be run at any concentration ranging from about 0.1M to about 5M (molarity based on the haloquinone) .
  • reaction to produce a mono- indolylquinone i ⁇ preferably carried out in the presence of about two equivalents of metal carbonate.
  • reactions to produce symmetrical or asymmetrical bis-indolylquinones are preferably carried out in the presence of about 3 equivalents of metal carbonate.
  • the reaction of the 2,5-dihalo-l,4-quinone with the indole may be run at temperatures ranging from about -10°C to about 100°C.
  • a particularly beneficial aspect of the present invention is that harsh conditions and high temperatures are not required to effect this reaction.
  • the reaction of the 2 , 5-dihalo-l, 4-quinone is run at a temperature in the range of about 0°C to about 30°C. More preferably, the reaction is run at about room temperature.
  • the reaction of the 2, 5-dihalo-l,4-quinone with the indole may be conducted under an inert atmo ⁇ phere such as nitrogen or argon; however, the reaction may al ⁇ o be run in atmospheric air.
  • the reaction may be run at any pressure up to 500 psig; however, it is preferable to conduct the reaction at atmospheric pressure.
  • the reaction time will vary according to the specific reactants and reaction conditions used, but generally will be from about 2 hours to about 72 hours.
  • the product indolylquinone ⁇ are typically i ⁇ olated according to standard workup procedures.
  • the crude reaction mixture may be diluted with IN hydrochloric acid, followed by extraction with an organic solvent such as ethyl acetate. Typically, the organic layer is wa ⁇ hed with brine and then dried over anhydrou ⁇ ⁇ odium sulfate.
  • the crude reaction mixture may simply be filtered to remove solid ⁇ .
  • the solvent is removed under reduced pressure, and the crude residue is purified by recrystallization, flash chromatography, High Pressure Liquid Chromatography (HPLC) or a combination thereof.
  • the residue i ⁇ purified u ⁇ ing flash chromatography and/or High Pres ⁇ ure Liquid Chromatography (HPLC) .
  • the ⁇ ymmetrical bi ⁇ - indolylquinone of formula I is further reacted with an alkali metal hydroxide to give a bi ⁇ -indol-2, 5-dihydroxy-l,4-quinone of formula IV.
  • alkali metal hydroxides are sodium and potassium hydroxide, or mixtures thereof.
  • this reaction is carried out in a mixture of ethanol and tetrahydrofuran using concentrated aqueou ⁇ KOH at a reflux temperature of about 85°C for up to 20 hour ⁇ .
  • the bis-indol- 2, 5-dihydroxy-1,4-quinone of formula IV maybe isolated according to standard workup and purification procedures as described above.
  • the methods of the present invention comprise reacting a symmetrical compound of formula I wherein Rl and R2 are Br, or an asymmetrical compound of formula VI wherein Rl and R2 are Br, with an alkali metal hydroxide and an alcohol of the formula R'OH wherein R' i ⁇ lower alkyl or alkylaryl, to a produce symmetrical indolylquinone of the formula V or an asymmetrical indolylquinone of formula XI .
  • ⁇ ubstituted or un ⁇ ubstituted means that the group in question can be substituted with one or more sub ⁇ tituent ⁇ as desired; for example, ⁇ ubstituents other than hydrogen such as Br, Cl, F, I, NH 2 , NR2 , N0 2 , CN, COR, OH, OR, S0 2 , alkyl, aryl, alkylaryl and the like may be used. On the contrary, unsub ⁇ tituted group ⁇ do not contain any substituent ⁇ .
  • alkyl as u ⁇ ed herein i ⁇ meant a straight or branched chain ⁇ aturated hydrocarbon group having from 1 to 12 carbon ⁇ ⁇ uch a ⁇ methyl, ethyl, isopropyl, n-butyl, s- butyl, t-butyl, n-amyl, isoamyl, n-hexyl, n-octyl and n- decyl; "alkenyl” and “alkynyl” are used to mean straight or branched chain hydrocarbon groups having from 2 to 12 carbons and unsaturated by a double or triple bond respectively, such as vinyl, allyl, propargyl, 1-methylvinyl, but-1-enyl, but-2- enyl, but-2-ynyl, 1 methylbut-2-enyl, pent-1-enyl, pent-3- enyl, 3-methylbut-l-ynyl, 1, 1-dimethylally
  • R3-R10 hydrogen
  • R1-R7 and R30 can be as listed in Table II below.
  • R3-R6 hydrogen
  • the compounds of Tables I and II have therapeutic activity for example as antifungal agents, antibacterial agents, and antitumor agents. In addition, these compounds may be useful in the dye industry.
  • N- (2-methylphenyl) -4-methylvaleramide (20.5 g) , sodium amide (90%) (11.0 g) , and tetralin (100 ml) were mixed in a 500 ml round bottom flask equipped with a magnetic stirrer and reflux conden ⁇ er and heated at reflux for 2 hour ⁇ . After cooling to room temperature, ethanol (10 ml) was added, followed by H 2 0 (150 ml) .
  • Example 2 The layers were separated, the organic layer was filtered through a pad of anhydrous magne ⁇ ium ⁇ ulfate, and the ⁇ olution was placed in a 200 ml round bottom flask equipped with a 10-inch vacuum jacketed Vigreux column. Tetralin was distilled at 35- 45°C/0.5 mm Hg. The residue was transferred to a 50 ml round bottom flask equipped with a 4-inch Vigreux column, and distillation at 118-129°C/0.5 mm Hg provided 2-(3-methyl-n- butyl) -indole 13.1 g (70%) as a slightly yellow solid.
  • Example 2 Example 2
  • This compound was synthesized in the same manner as Example 2 except the starting indole was 2-n-butyl-indole.
  • This compound may be synthesized as follows: A mixture of 100 mg of 2 , 5-diacetoxy-3 , 6-dibromo-l, 4- quinone, 180 mg of 3-[2- (2-methylbut-2-en-4-yl) indole, prepared by the Fisher indole synthesis, 10 ml of anhydrous dimethylforamide, and powdered potassium carbonate, is heated at 100°C for 24 hours. The cooled mixture is partitioned between ethyl acetate and water. The ethyl acetate layer is then washed with brine, dried over sodium sulfate, filtered and concentrated.
  • the crude product is then purified on a medium pressure liquid chromatography column in a solvent mixture of dichloromethane and methanol to provide 25 mg of 2 , 5-diacetoxy-3 , 6-di- [ 2- (2-methylbut-2-en-4-yl) indol-3- yl] 1 , 4-quinone.
  • 2 5-Diacetoxy-3 , 6-di- [ 2 (2-methylbut-2-en-4- yl) indol-3-yl]l,4-quinone is then hydrolysed with 1 N aqueous sodium hydroxide ⁇ olution in methanol. Acidification of the above mixture produces the crude product after filtration. Further crystallization in ethanol and water produces the title compound.
  • quinones such as 3 , 6-dibromo- 2,5-ditrimethyl ⁇ iloxy-l,4-quinone, 3 , 6-dibromo-2 , 5-di- (t- butyldimethylsiloxy-1 , 4-quinone, 2 , 5-dibenzoxy-3 , 6-dibromo- 1,4-quinone, 3 , 6-dibromo-2 , 5-diisobutyroxy-l, 4-quinone, 2,5- dibenzyloxy-3,6-dibromo-l,4-quinone or 2,5- diallyoxycarbonyloxy-3 , 6-dibromo-l, 4-quinone which can be prepared from commercially available 2,4-dibromo-3 , 6- dihydroxy-l,4-quinone may also be used as starting materials.
  • protecting groups can be removed by conventional deprotection methods such as diluted acid, potas ⁇ ium fluoride or palladium (0) complex or palladium on carbon with hydrogen or by method ⁇ de ⁇ cribed by Greene and Wut ⁇ (1991, "Protective Group ⁇ In Organic Synthe ⁇ i ⁇ ,” John Wiley and Son).
  • Other solvents such as pyridine or dimethylsulfoxide (DMSO) may be used in place of dimethyl formamide.
  • This compound was prepared under condition ⁇ similar to those described in Examples 5 to 8.
  • This compound was prepared under conditions similar to those described in Examples 5 to 8.
  • Example 5 Methylation of Example 5 with methyl iodide and potas ⁇ ium carbonate in dimethylforamide followed by purification produces the title compound.
  • This compound may also be prepared by heating 2 , 5-dibromo-3, 6-di [2- (2-methylbut-2-en-4- yl) indol-3-y]l,4-quinone in methanol in the presence of powdered potassium carbonate.
  • Example 30 4- quinone Hydrogenation of Example 30 under conditions as those in Example 3 produced the title compound.
  • This compound is preferably synthesized by the method of Example 2 using 2-ethylindole as the starting indole. However, this compound may also be prepared by the method of
  • Example 32 using 2-ethylindole as the starting indole Preparation of 2-ethylindole: Refer to 32a) using methyl iodide as the alkylating agent.
  • Example 32 using 2-butylindole as the starting indole.
  • Preparation of 2- (but-l-en-4-yl), indole Refer to 32a) using allyl bromide as the alkylating agent.
  • Preparation of 2- butylindole Refer to 32b) using 2- (but-l-en-4-yl) indole as the starting material.
  • Example 36
  • This compound is preferably synthesized by the method of Example 2 but may also be prepared according to the method of Example 32 using 2- (but-l-en-4-yl) indole as the starting indole.
  • Example 37 Preparation of 2 , 5-Dihydroxy-3 , 6-di- [2- (4-methyl-n-pentyl) indol-3-yl]-l,4-quinone:
  • This compound i ⁇ preferably synthesized by the method of Example 2 but may also be prepared according to Example 32 using 2- (4-methyl-n-pentyl) indole as the ⁇ tarting indole.
  • Preparation of 2- (2-methyl-2- penten-5-yl) indole Refer to Example 1 using 5- methylhexanoic acid as the starting acid.
  • This indole may also be prepared according to Example 32a) using 4-bromo-2- methyl-2-butene as the alkylating reagent.
  • Preparation of 2-(4-methyl-n-pentyl) indole Refer to 32b) using 2- (2- methyl-2-penten-5-yl) indole as the starting material.
  • 3-yl]-l, 4-quinone This compound is preferably ⁇ ynthesized by the method of Example 2 but may also be prepared according to Example 32 using 2- (2-phenylethyl) indole as the starting indole.
  • Preparation of 2- (2-phenylethyl) indole Refer to Example 3 using 3-phenylpropionyl chloride as the starting acid chloride.
  • This indole may also be prepared according to Example 32a) using benzyl bromide a ⁇ the alkylating agent.
  • Thi ⁇ ⁇ ynthesi ⁇ may also be accomplished beginning with 5-chloro-2- methylindole, which is alkylated with methyl indole.
  • the product chloroindole is converted to its Grignard specie ⁇ and exposed to carbon dioxide to fini ⁇ h the synthesis.
  • Example 32 using 5-carboxy-2-propylindole as the starting indole.
  • Preparation of 5-carboxy-2- propylindole Refer to Example 1 using methyl 4-amino-3- methyl-benzoate instead of o-toluidine or refer to 40 u ⁇ ing ethyl iodide as the alkylating agent.
  • this synthesis may be accomplished with a standard nitration of 2-ethylindole using sodium nitrate and sulfuric acid similar to that cited in Chem. Lett. (7): 1125-1128 (1991).
  • the resultant 5-nitro-2- ethylindole is reduced to the de ⁇ ired amino compound u ⁇ ing catalytic hydrogenation as in 32b) .
  • Example 32 using 5-amino-2- (n-propyl) indole as the ⁇ tarting indole.
  • Preparation of 5-amino-2- (n-propyl) indole Refer to Example 45 using butyryl chloride. In the alternative, refer to the ⁇ ynthesis cited in Chem. Lett. (7) : 1125-1128 (1991) cited in Example 45 using 2-n-propylindole.
  • 2- (3-methyl-n-butyl) -indole is reduced to the de ⁇ ired amino compound a ⁇ in 32b.
  • the synthesi ⁇ may al ⁇ o be accomplished according to Example 45 using 2- (3-methyl-n-butyl) indole.
  • This synthe ⁇ i ⁇ wa ⁇ accomplished by treating 2,5-hydroxy- 3,6-di-[2-(3-methyl-n-butyl) indol-3-yl]-l,4-quinone with acetic anhydride in the presence of pyridine.
  • Example 32 u ⁇ ing 5- (4- methylphenyl ⁇ ulfonyla ino) -2- (n-propyl) indole as the starting indole.
  • Preparation of 5- (4- methylphenylsulfonylamino) -2- (n-propyl) indole Refer to 49 using 5-amino-2-propylindole.
  • the aqueou ⁇ layer was extracted with ethyl acetate (200 ml) .
  • the organic layer wa ⁇ wa ⁇ hed with brine (100 ml) and dried with sodium sulfate.
  • the crude residue was purified by flash chromatography (30% ethyl acetate/hexane) to provide 2 , 5-dibromo-3- (2-ethylcarboxyindol-3-yl) -6- (2- methylindol-3-yl) -1,4-quinone (0.37 g) as a blue crystalline solid.
  • the mono-indolylquinone 6- (2- ethylcarboxyindol-3-yl) -2,3, 5-tribromo-l, 4-quinone may isolated separately and used in further reactions with other indoles.
  • the compounds disclosed herein have utility, inter alia , at therapeutically effective doses to treat or ameliorate cell proliferative disorders involving PTK/adaptor protein interactions.
  • the compounds prepared according to the present invention may be tested by a variety of method ⁇ for determining the ability of the compound ⁇ to inhibit kinase activity or to disrupt PTK/adaptor protein complexes.
  • any assay currently used for screening compound ⁇ that act on cells containing PTK ⁇ can be u ⁇ ed.
  • such as ⁇ ays involve exposing cells that expre ⁇ s the PTK to a test substance and either: (a) scoring phenotypic changes in the cell culture as compared to control cells that were not exposed to the test substance; or (b) biochemically analyzing cell lysates to as ⁇ ess the level and/or identity of tyrosine phosphorylated proteins.
  • a common technique involves incubating cells with ligand and radiolabeled phosphate, lysing the cells, separating cellular protein components of the lysate using an SDS-polyacrylamide gel (SDS-PAGE) technique, in either one or two dimensions, and detecting the presence of pho ⁇ phorylated protein ⁇ by expo ⁇ ing X-ray film.
  • SDS-PAGE SDS-polyacrylamide gel
  • the phosphorylated proteins are detected by immunoblotting technique ⁇ , in which case the phosphate that is detected is not radiolabeled.
  • the cellular components separated by SDS-PAGE are transferred to a nitrocellulose membrane, where the presence of phosphorylated tyrosines is detected using an antiphosphotyrosine antibody (anti-PY) .
  • the anti-PY can be detected by labeling it with a radioactive substance, or an enzyme, such a ⁇ horseradish peroxidase.
  • a further alternative involves detecting the anti-PY by reacting with a second antibody which recognize ⁇ the anti-PY, thi ⁇ second antibody being labeled with either a radioactive moiety or an enzyme as previously described. Examples of these and similar techniques are described in Hansen et al . , 1993, Electrophoresis 14:112-126; Campbell et al . 1993, J ⁇ . Biol. Chem. 268:7427-7434; Donato et al . , 1992, Cell Growth and Diff . 3:258-268; and Katagiri et al . , 1993, J_j_ Immunol . 150:585-593.
  • ELISA-type as ⁇ ays in microtitre plates can also be used to test purified substrates. See for example Peraldi et al . , 5 1992, J,; . Biochem. 285: 71-78; Schraag et al . , 1993, Analytical Biochemistry 211:233-239; Cleavland, 1990, Analytical Biochemistry 190:249-253; Farley, 1992, Analytical Biochemistry 203:151-157; and Lczaro, 1991, Analytical Biochemistry 192:257-261. Examples of assay methods are 0 described in U.S. Application Serial No. 08/279,321, filed July 22, 1994 and U.S. Application Serial No. 08/488,156 filed June 7, 1995 which are hereby incorporated in their entiretie ⁇ by reference.
  • a variety of method ⁇ may be u ⁇ ed to a ⁇ ay the ability of 5 the compound ⁇ prepared according to the invention to disrupt PTK/adaptor protein complexes.
  • in vitro complex formation may be assayed by, first, immobilizing one component, or a functional portion thereof, of the complex of interest to a solid support.
  • the immobilized complex 0 component may be expo ⁇ ed to a compound prepared according to the present invention, and to the second component, or a functional portion thereof, of the complex of interest.
  • it may be determined whether or not the second component is still capable of forming a complex with the 5 immobilized component in the presence of the compound.
  • in vivo complex formation may be assayed by utilizing co-immunoprecipitation techniques well known to those of skill in the art. Briefly, a cell line capable of forming a PTK/adaptor complex of interest may be exposed to 0 one or more of the compound ⁇ prepared according to the pre ⁇ ent invention, and a cell ly ⁇ ate may be prepared from this exposed cell line. An antibody raised against one of the components of the complex of intere ⁇ t may be added to the cell lysate, and subjected to standard immunoprecipitation 5 techniques. In cases where a complex is still formed, the immunoprecipitation will precipitate the complex, whereas in cases where the complex has been disrupted, only the complex component to which the antibody is raised will be precipitated.
  • the effect of a compound of the invention on the transformation capability of the PTK/adaptor protein of interest may be directly a ⁇ sayed.
  • one or more of the compounds prepared according to the invention may be administered to a cell such as a fibroblast or hematopoietic cell capable of forming a PTK/adaptor complex which, in the absence of a compound of the invention, would lead to the cell's transformation (Muller, A.J. et al . , 1991, Mol. Cell. Biol. 11:1785-1792; McLaughlin, J. et al . , 1987, Proc. Natl. Acad. Sci. USA 84:6558-6562).
  • the transformation state of the cell may then be measured in vitro, by monitoring, for example, its ability to form colonies in soft agar (Lugo and Witte, 1989, Mol. Cell. Biol. 9:1263-1270; Gishizky, M.L. and Witte, O.N., 1992, Science 256:836-839).
  • a cell's transformation state may be monitored in vivo by determining it ⁇ ability to form tumors in immunodeficient nude or severe combined immunodeficiency (SCID) mice (Sawyers, CL. et al . , 1992, Blood 79:2089-2098).
  • the pre ⁇ ent invention al ⁇ o provide ⁇ a method of ameliorating ⁇ ymptoms of a cell proliferative disorder wherein the cell proliferative disorder involves a protein tyrosine kinase polypeptide/adaptor polypeptide complex, with an amount of a compound of either of the formula ⁇ I or XI, sufficient to disrupt protein tyrosine kinase polypeptide/adaptor polypeptide complexes of the cell ⁇ o that symptoms of the cell proliferative disorder are ameliorated.
  • the present invention also provides a method of ameliorating a cell proliferative disorder using a compound described herein, particularly a compound of the formula I, IV, V, VI, VIII, IX, X, XI, XII or XIII, wherein the cell proliferative disorder occurs in a mammal and the compound contacts the cell within a mammal so that the symptoms of the cell proliferative disorder in the mammal are ameliorated.
  • the compounds, i.e., indolylquinones, of the present invention may be u ⁇ ed alone or in combination with other drugs or therapies to treat cancer.
  • Cell proliferative disorders which are treatable according to the methods of the invention include BCR-ABL- associated cancers, gliomas, glioblastomas, melanomas, ovarian cancers, breast cancers, and prostate cancers.
  • the present invention provides a method of ameliorating symptoms of a cell proliferative disorder wherein the cell proliferative disorder involves a protein tyrosine kinase polypeptide/adaptor polypeptide complex, which involves contacting a cell capable of forming the protein tyrosine kinase polypeptide/adaptor polypeptide complex with an amount of a pharmaceutical composition comprising a compound of any one of the formulas I, IV, V, VI, VIII, IX, X, XI, XII or XIII, sufficient to disrupt protein tyro ⁇ ine kinase polypeptide/adaptor polypeptide complexes of the cell so that symptoms of the cell proliferative disorder are ameliorated.
  • the compounds prepared according to the pre ⁇ ent invention may be formulated into compo ⁇ itions comprising other drugs or pharmaceutical agent ⁇ .
  • the pharmaceutical compositions of compounds prepared according to the present invention also comprise additional cancer treatment agents.
  • the compound ⁇ prepared according to the present invention may be formulated into pharmaceutical compositions in a conventional manner using one or more physiologically acceptable carriers or excipients.
  • the compounds and their phy ⁇ iologically acceptable salts and solvates may also be formulated for administration by inhalation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration.
  • the pharmaceutical compositions may take the form of, for example, tablet ⁇ or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose) ; fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate) ; lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate) ; or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose
  • fillers e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate
  • lubricants e.g., magnesium stearate, talc or silica
  • disintegrants e
  • Liquid preparations for oral administration may take the form of, for example, solution ⁇ , ⁇ yrup ⁇ or ⁇ u ⁇ pensions, or they may be presented as a dry product for con ⁇ titution with water or other suitable vehicle before u ⁇ e.
  • Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such a ⁇ ⁇ uspending agents (e.g., sorbitol syrup, cellulose derivative ⁇ or hydrogenated edible fat ⁇ ) ; emulsifying agents (e.g.
  • non-aqueous vehicles e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils
  • preservatives e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid
  • the preparations may also contain buffer salts, flavoring, coloring and sweetening agents and other pharmaceutical agents as appropriate.
  • the compound ⁇ for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pres ⁇ urized pack ⁇ or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • Such formulations may also comprise other pharmaceutical agents as appropriate.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • Compound ⁇ prepared according to the present method may also be formulated for parenteral administration by injection, e.g., by bolu ⁇ injection or continuou ⁇ infu ⁇ ion.
  • Formulation ⁇ for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compo ⁇ itions may take such forms a ⁇ suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • EXAMPLE - In Vivo Activity of Compound #1 The following example illustrates the use of the compounds of the invention in an in vivo model of tumor growth.
  • mice Female athymic mice (BALB/c, nu/nu) were obtained from Simonsen Laboratories (Gilroy, CA) . All animals were maintained under clean-room conditions in Micro-isolator cages with Alpha-dri bedding. They received sterile rodent chow and water ad libitum.
  • Results The results are shown below as a percent tumor reduction compared to controls.
  • Administration of compound 1 inhibited tumor growth in a dose dependent manner. Tumor growth remained inhibited even after ce ⁇ sation of treatment with the compound.

Abstract

The present invention relates to novel synthetic methods for the preparation of indolylquinones. The methods of the present invention are directed to synthetic reactions involving indoles and halo-quinones in solvent and in the presence of a metal carbonate. The invention also relates to bis- and mono- indolylquinones of high purity and pharmaceutical compositions containing the same.

Description

SYNTHETIC METHODS FOR THE PREPARATION OF INDOLYLQUINONES AND MONO- AND BIS-INDOLYLQUINONES PREPARED THEREFROM
1. INTRODUCTION
The present invention relates to novel synthetic methods for the preparation of both known and novel indolylquinones. Many indolylquinones, in particular the class of indolylquinones known as the asterriquinones, have utility in the treatment of cell proliferative disorders such as cancer. In addition, many indolylquinones are known to be useful as dyes. Finally, indolylquinones are also known to exhibit antifungal and antibacterial properties. However, in general, indolylquinones are isolated from natural sources and are not prepared synthetically. The present invention is directed to a synthetic method for the preparation of indolylquinones, and to novel compounds prepared using said method. In particular, the methods of the present invention are directed to synthetic reactions involving indoles and halo-quinones in solvent and in the presence of a metal carbonate. These methods provide a direct and simple means of preparing the compounds of interest, indolylquinones.
2. BACKGROUND OF THE INVENTION
2.1 Isolation From Natural Sources And
Therapeutic Utility Of Indolylquinones
Research interest concerning indolylquinones grew out of early observations that extracts of Chaetomium exhibited antibiotic properties. These observations led researchers to attempt the isolation of active species from cultures of these microorganisms. For example, Brewer et al . disclose the isolation of a purple pigment, which was termed cochliodinol, from isolates of Chaetomium cochliodeε and
Chaetomium globosum (1968, "The Production of Cochliodinol and a Related Metabolite by Chaetomium Species," Can. J.
Microbiol. 14:861-866). Brewer et al . also disclose the synthetic conversion of cochliodinol to a diacetate compound.
Id. Further, the antifungal properties of cochliodinol have also been documented (Meiler et al . , 1971, "The Effect of Cochliodinol, a Metabolite of Chaetomium cochliodes on the Respiration of Microspores of Fusarium oxysporum, " Can. J. Microbiol. 17: 83-86).
The structure of cochliodinol was elucidated by Jerra et al . in 1975. (1975, "The Chemistry of Cochliodinol, a Metabolite of Chaetomium spp . , " Can. J. Chem. 53:727-737). Jerram et al . reported the structure of cochliodinol as: 2,5- dihydroxy-3 , 6-di (5 ' - (2 " -methylbut-Δ2 ' ' -ene) -indoly1-3 ' ) - cyclohexadiene-l,4-dione. The conversion of cochliodinol to various other derivatives, including its dimethyl and diacetyl analogues, was also disclosed. Id. Some of these derivatives were highly colored and suitable for use as dyes, while others were colorless. Id. Sekita discloses the isolation of other bis (3-indoly1) -dihydroxybenzoquinones, including isocochliodinol and neocochliodinol from Chaetomium muroum and C. amygdalisporum (1983, "Isocochliodinol and Neocochliodinol, Bis (indolyl) -benzoquinoneε from Chaetomium spp.," Chem. Phar . Bull. 31(9): 2998-3001).
Despite the therapeutic potential of cochliodinol and its derivatives, efficient methods suitable for large scale production of these compounds have remained elusive. U.S. Patent No. 3,917,820 to Brewer et al . discloses the purple pigment cochliodinol and a process for its production by culturing various types of Chaetomium under aerobic conditions. However, the methods of Brewer require long incubation periods for cochliodinol production (2-8 days) , the use of benzene, a known carcinogen, to effect chromatographic separation of cochliodinol from the culture and are limited to the few naturally occurring compounds. Moreover, Brewer discloses the isolation of only small quantities (0.75 grams) of cochliodinol from Chaetomium .
Another class of indolylquinones known as the asterriquinones in which the nitrogen of the indole ring is substituted, has been shown to exhibit antitumor activity. Arai et al . proposed the general name "asterriquinones" for the class of indolylquinones based upon asterriquinone (1981, "Metabolic Products of Aspergillus terreuε IV. Metabolites of the Strain IFO 8835. (2) The Isolation and Chemical Structure of Indolyl Benzoquinone Pigments," Chem. Phar . Bull. 29(4): 961-969). It should be noted that as used herein, the term "asterriquinone" has a more general meaning, and is used interchangeably with the term "indolylquinone. " Yama oto et.. l . disclose the antitumor activity of asterriquinone, i.e., 2 , 5-bis[N- (1" , l"-dimethyl-2"- propenyl) indol-3 '-yl] -3 , 6-dihydroxy-l,4-benzoquinone, and its isolation from the fungus Aspergilluε terreus (1976, "Antitumor Activity of Asterriquinone, a Metabolic Product from Aspergillus terreus ," Gann 67:623-624).
Arai et al . disclose the isolation and characterization of 11 different kinds of bisindolyl-dimethoxyl-p- benzoquinones from Aspergillus terreus . Id. The isolation and structural determination of a number of other asterriquinones have also been reported. (Arai et al . 1981, "Metabolic Products of Aspergilluε terreus VI. Metabolites of the Strain IFO 8835. (3) the Isolation and Chemical Structures of Colorless Metabolites," Chem. Pharm. Bull. 29(4): 1005-1012; Kaji et al . , 1994, "Four New Metabolites of Aspergilluε Terreus", Chem. Pharm. Bull. 42(8): 1682-1684). However, the separation of asterriquinones is troublesome because there are so many kinds of homologous pigments in the Aspergilluε extracts. Moreover, the chromatographic purification of asterriquinones is typically carried out using benzene, a known carcinogen, as a solvent. Finally, only milligram quantities of asterriquinones have actually been isolated from these natural sources.
In view of their potential as anticancer agents, research has been directed to determination of the relationship between structure and antitumor activity of asterriquinones. For example, Arai et al . reported a study in which hydroxyl benzoquinone derivatives obtained by demethylation of bisindolyl-dimethoxyl-p-benzoquinones were found to have greater antitumor activity than the ethoxyl derivatives (1981, "Metabolic Products of Aεpergilluε terreuε V. Demethylation of Asterriquinones," Chem. Pharm. Bull. 29(4): 991-999). Shimizu et al . noted that the presence of free hydroxyl groups in the benzoquinone moiety, as well the number and position of tert- , isopentenyl, or both pentyl groups, seems to have an effect on the antitumor activity of the compound (1982, "Antitumor Effect and Structure-Activity Relationship of Asterriquinone Analogs," Gann 73: 642-648). In an attempt to obtain information towards the development of more potent asterriquinone derivatives, Shimizu et al . conducted an investigation into the structure-activity relationship of asterriquinones in which the action mechanism of asterriquinone in its antitumor activity with reference to its interaction with DNA molecules and the plasma membrane of tumor cells was studied (1990, "Interaction of Asterriquinone with Deoxyribonucleic Acid in Vitro , " Chem. Pharm. Bull. 38(9): 2617-2619). It was reported that a correlation exists between the pKa value of the asterriquinone derivative and its antitumor activity. Id. Maximum antitumor activity was observed for compounds with pKa's in the range of 6-7. Id. Analysis of structure-activity relationships has led to attempts to obtain compounds with more potent antitumor activity by chemical modification of asterriquinone and related compounds isolated from natural sources (Shimizu et al . , 1982, "Antitumor Activity of Asterriquinones from Aspergillus Fungil IV. An Attempt to Modify the Structure of Asterriquinones to Increase the Activity," Chem. Pharm. Bull. 30(5): 1896-1899). Although benzoquinone derivatives having aziridinyl groups in the molecule such as itomycin C, carbazilquinone or "E 39" are well known potent anticancer agents, replacement of the functional groups at the 3 and 6 positions in the benzoquinone moiety of asterriquinone failed to enhance its antitumor potency. Id. Similarly, the introduction of an ethyleneimino group into the molecule did not increase antitumor activity. A dimethylallyl derivative of asterriquinone showed moderate activity against the ascites and solid tumors of Ehrlich carcinoma, while an allyl derivative did not. It was suggested that in order to enhance the antitumor activity, it may be necessary not only to alter the pKa value by alkylation, but also to introduce hydrophilic groups into the molecule.
Most recently, in addition to their demonstrated antitumor activity, asterriquinone and some of its analogues have also been shown to be strong inhibitors of HIV-reverse transcriptase (Ono et al . , 1991, "Inhibition of HIV-Reverse Transcriptase Activity by Asterriquinone and its Analogues," Biochem. Biophys. Res. Commun. 174(1): 56-62).
2.2 Cancer And Signal Transduction
As mentioned above, indolylquinones have utility as antitumor agents for the treatment of cancer and other cell proliferative disorders. These compounds are believed to arrest the growth of tumors by interfering with the signal transduction pathways that regulate cell proliferation and differentiation.
Protein phosphorylation is a common regulatory mechanism used by cells to selectively modify proteins carrying signals that regulate cell proliferation and differentiation. The proteins that execute these biochemical modifications are a group of enzymes known as protein kinaseε. They may further be defined by the amino acid that they target for phosphorylation. One group of protein kinaseε are the tyrosine kinases (PTKs) which selectively phoεphorylate a target protein on its tyrosine residues.
Protein tyrosine kinases comprise a large family of proteins, including many growth factor receptors and potential oncogenes. Tyrosine kinases can be cytoplasmic, non-receptor-type enzymes and act as a key component of a signal transduction pathway which regulates cell functions such as cell division, differentiation and survival.
Adaptor proteins are intracellular proteins having characteristic conserved peptide domains (SH2 and/or SH3 domains, as described below) which are critical to the signal transduction pathway. Such adaptor proteins serve to link protein tyrosine kinases, especially receptor-type protein tyrosine kinases to downstream intracellular signalling path- ways such as the RAS signalling pathway. It is thought that such adaptor proteins may be involved in targeting signal transduction proteins to the correct site in the plasma membrane or subcellular compartments, and may also be involved in the regulation of protein movement within the cell. The profound cellular effects mediated by tyrosine kinases and adaptor molecules have made them attractive targets for the development of new therapeutic molecules. It is known, for example, that the overexpresεion of tyrosine kinases, such as HER2 , can play a decisive role in the development of cancer (Slamon, D.J., et >al . , 1987, Science, 235:177-182) and that antibodieε capable of blocking the activity of this enzyme can abrogate tumor growth. (Drebin, et al . 1988, Oncogene 2:387-394). Blocking the signal transduction capability of tyrosine kinaseε such as Flk-1 and the PDGF receptor have been shown to block tumor growth in animal modelε (Millauer, B. , et al . 1994, Nature 367:577; Ueno, H., et al . 1991, Science 252:844-848).
Despite great interest in the various therapeutic and other utilitieε of indolylquinoneε such as asterriquinones, research into the therapeutic activities of indolylquinones and efforts to obtain indolylquinones with enhanced therapeutic activity have both been limited by the lack of reliable sources for these compounds. Indeed, isolation of indolylquinones from natural sources requires multiple steps and produces only milligram quantities of the target molecules. Further, evaluation of the activities of novel indolylquinones has necessarily been confined to those compounds which can be obtained by chemical modification of known compounds that can be isolated from natural sources. Clearly, a synthetic routine to these compounds would be invaluable to the art.
2.3 Synthesis Of Cochliodinol A synthetic route to an indolylquinone, cochliodinol, has been reported by Hδrcher et al . This route is a complex, multi-step, low-yield proceεε for the total synthesis of cochliodinol (1986, "Totalsynthese des Cochliodinols" , Liebigs. Ann. Chem. 1765-1771) . The Horcher method involveε an unusual solid state reaction of bromanil (2,3,5,6- tetrabromo-l,4-quinone) with 5-bromoindole in the presence of aluminum oxide and potassium carbonate in a dry box at 105 °C. This solid state reaction yields about 11% of 2,5-dibromo- 5,6-bis(5-bromo-3-indolyl) -1, 4-quinone. The 2 , 5-dibromo-5, 6- bis (5-bromo-3-indolyl) -1,4-quinone is then treated with benzalcohol and sodium hydroxide to give 2 , 5-bis (benzyloxy) - 3,6-bis (5-bromo-3-indolyl) -1,4-quinone in 45% yield. This product is then reacted with hydrogen gas in the presence of a 10% Pd on activated charcoal catalyst, followed by treatment with acetic anhydride in pyridine to give 1,2,4,5- tetracetoxy-3,6-bis(5-bromo-3-indolyl) benzene. Reaction of this compound with a complex of isopentenyl bromide and tetracarbonyl nickel gives 1, 2 , 4,5-tetracetoxy-3, 6-bis[5- (3- methyl-2-butenyl) 3-indolyl] benzene. This compound is then reacted with sodium hydroxide and oxygen to give cochliodinol. According to Horcher et al . , the reaction of bromanil with certain substituted indoles is problematic. Horcher et al . report that earlier attempts to react a p-benzoquinone with 2-methylindole resulted in only monoindolequinones in very low yields. Attempts to react bromanil with 5- (2- methylbut-2-en-4-yl) -indole were also reported by Horcher to be unsuccesεful due to the inεtability of the unsaturated side chain vis-a-vis the dehydrogenating bromanil. To overcome this difficulty, Horcher reacted bromanil with 5-bromoindole instead of 5- (2-methylbut-2-en-4-yl) , followed by introduction of the 2-methylbut-2-en-4-yl group at the end of the synthesis, requiring the additional step of reacting the 5,5 ' -dibromo-bis-indolylquinone with the complex of isopentenyl bromide and tetracarbonyl nickel, which substitutes the bromine atoms with 2-methylbut-2-en-4-yl groups.
Horcher et al . report that this method resulted in isolation of only milligram quantities of cochliodinol in a very low overall yield. However, Horcher et al . indicate that conducting the initial reaction of bromanil with 5- bromoindole in smaller batches results in better yields. This suggests that the methods of Horcher et al . are unsuited for production of bis-indolylquinones on a large scale. In addition, as applied to the production of bis-indolylquinones in general, the methods of Horcher et al . would be prohibitively ultistep, and would likely result in isolation of only milligram quantities of the target indolylquinones. Moreover, these methods require high temperature and manipulation in dry box.
Accordingly, despite the great interest in indolylquinones, there iε a lack of feasible large scale synthetic routes for obtaining these compounds. Thus, there is a need in the art for a fast, efficient synthetic method for making indolylquinones in preparative quantities. Further, there is a need for εynthetic eanε of producing known indolylquinones previously available only in milligram quantities from natural sources. Moreover, there is a need in the art for a synthetic method that may be manipulated easily to produce a wide variety of structurally diverse novel indolylquinones, so that structure-activity relationships may be further elucidated, and new, perhaps more therapeutically useful indolylquinones may be developed.
3. SUMMARY OF THE INVENTION
The present invention provides a method for the synthesis of indolylquinones which comprise reacting a substituted or unsubstituted 2 , 5-dihalo-l, 4-benzoquinone with one or more substituted or unsubstituted indoles in a polar organic solvent and in the presence of metal carbonate.
In one embodiment, the present invention provides a method for preparing a symmetrical indolylquinone compound of the formula I:
Figure imgf000011_0001
wherein: Rl and R2 are each independently Br, Cl, F, I, H, OH or -OCOR, wherein R is, lower alkyl, aryl or alkylaryl; R"l is H, C^C- alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and R3 to R7 are each independently hydrogen, branched or unbranched C^C,, alkyl, alkylcarboxy, C2-Cm alkenyl, C2-Cm alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12 , preferably 1-7 , and m is an integer from 0 to 12, preferably 1-7. Rl and R2 are preferably Br, Cl, F or H; and least preferably OH. The method comprises reacting a substituted or unsubstituted 2,5- dihalo-l,4-benzoquinone, preferably a 2,5-dibromo-l,4- benzoquinone compound of the formula II:
Figure imgf000011_0002
wherein Rl and R2 are as defined above; with at least one indole of the formula III:
Figure imgf000012_0001
wherein R"l and R3-R7 are aε defined above. The reaction iε carried out in a polar organic εolvent and in the presence of metal carbonate under mild conditions which are further discussed below.
The method may further comprise reacting the indolylquinone compound of formula I with an alkali metal hydroxide to produce a compound of the formula IV:
Figure imgf000012_0002
Further, the invention' encompasses reacting the indolylquinone compound of formula I wherein Rl and R2 are Br with an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula:
Figure imgf000013_0001
wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl. In another embodiment, the present invention provides a method for preparing an asymmetrical indolylquinone compound of the formula VI:
Figure imgf000013_0002
wherein: Rl and R2 are each independently Br, Cl, F, I, H, OH, or —OCOR, wherein R is lower alkyl, aryl or alkylaryl; R"l and R"2 are each independently H, C^C, alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and R3 to R12 are each independently hydrogen, branched or unbranched C1-Cn alkyl, alkylcarboxy, C2-Cm alkenyl, C2-Cm alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, Cx-Cn alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, preferably 1-7 , and m is an integer from 0 to 12 , preferably 1-7. Rl and R2 are preferably Br, Cl, F or H; and least preferably OH. The principal steps of the method comprise:
(a) reacting a substituted or unsubstituted 2,5- dibromo-1, 4-benzoquinone compound of the formula II:
Figure imgf000014_0001
wherein Rl and R2 are as defined above; with one equivalent of a first indole of the formula III:
Figure imgf000014_0002
wherein R"l and R3-R7 are as defined above; in a polar organic solvent and in the presence of metal carbonate;
(b) reacting the intermediate product of step (a) with one equivalent of a second indole of the formula IV:
Figure imgf000015_0001
wherein R"2 and R8-R12 are as defined above. Both reactions are carried out in a polar organic solvent and in the presence of metal carbonate under mild conditions which are further discussed below.
The invention also encompasεes further reacting the indolylquinone compound of formula VI with an alkali metal hydroxide to produce a compound of the formula VIII:
Figure imgf000015_0002
wherein R"l, R"2 and R3-R12 are as defined above.
Further, the invention encompasses reacting the indolylquinone of formula VI wherein Rl and R2 are Br, F, Cl or I with an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula IX:
Figure imgf000016_0001
wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
In yet another embodiment, the present invention provides a method for preparing an indolylquinone compound of the formula X:
Figure imgf000016_0002
wherein: R"l is H, C1-C7 alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and R3 to R7 are each independently hydrogen, branched or unbranched C^C,, alkyl, alkylcarboxy, C2-Cm alkenyl, C2-Cm alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfona ido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer of 0 to 12, preferably 1-7, and m is an integer of 0 to 12 , preferably 1-7. This embodiment of the inventive method involves reacting 2, 3 ,5, 6-tetrabromo- 1,4-benzoquinone with at least one indole of the formula:
Figure imgf000017_0001
wherein R"l and R3-R7 are as defined above. The reaction is carried out in a polar organic solvent in the presence of metal carbonate.
The method also encompasses further reacting the indolylquinone compound of formula X with an alkali metal hydroxide to produce a compound of the formula:
Figure imgf000017_0002
wherein R3-R7 are as defined above. Further, the invention also encompasses a method which further comprises reacting the indolylquinone compound of formula X with an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula:
Figure imgf000018_0001
wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl. In yet another embodiment, the present invention provides a method for preparing an indolylquinone compound of the formula XIV:
Figure imgf000018_0002
wherein: R"l and R"2 are each independently H, C^C, alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and R3 to R12 are each independently hydrogen, branched or unbranched C1-Cn alkyl, alkylcarboxy, C2-Cm alkenyl, C2-Cra alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfona ido, amino, ercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer of 0 to 12, preferably 1-7, and m is an integer of 0 to 12, preferably 1- 7. This embodiment of the inventive method involves reacting 2,3,5,6-tetrabromo-l,4-benzoquinone with one equivalent of a first indole of the formula III:
Figure imgf000019_0001
wherein R"l and R3-R7 are as defined above; in a polar organic solvent and in the presence of metal carbonate;
reacting the intermediate product of step (a) with one equivalent of a second indole of the formula VII:
Figure imgf000019_0002
wherein R"2 and R8-R12 are as defined above. Both reactions are carried out in a polar organic solvent and in the presence of metal carbonate under mild conditions which are further discussed below.
The invention also encompasses further reacting the indolylquinone compound of formula XIV with an alkali metal hydroxide to produce a compound of the formula VIII:
Figure imgf000020_0001
wherein R"l, R"2 and R3-R12 are as defined above. Further, the invention encompasεes reacting the indolylquinone of formula VI wherein Rl and R2 are Br, F, Cl or I with an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula IX:
Figure imgf000020_0002
wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl. In still another embodiment, the present invention provides a method for preparing a mono-indolylquinone compound of the formula XI:
Figure imgf000021_0001
wherein Rl, R2 and R30 are each independently Br, Cl, F, I, H, OH or -OCOR, wherein R is lower alkyl, aryl or alkylaryl;
R"l is H, Cx-C7 alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C^C,, alkyl, alkylcarboxy, C2-Cm alkenyl, alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C1-Cn alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer from 0 to 12 and m is an integer from 0 to 12. This method comprises reacting a substituted or unsubstituted 2,5-dibromo-l,4-benzoquinone compound of the formula:
Figure imgf000021_0002
wherein Rl and R2 are as defined above, with one indole of the formula
Figure imgf000022_0001
wherein R"l and R3-R7 are as defined above. The reaction is carried out in a polar organic εolvent and in the presence of metal carbonate.
The method of the present invention may further comprise reacting the indolylquinone compound of formula XI with an alkali metal hydroxide to produce a compound of the formula:
Figure imgf000022_0002
wherein R30 and R3-R7 are as defined above. The method may further comprise reacting the indolylquinone compound of formula XI wherein Rl, R2 and R30 are Br, F, Cl or I with a mixture of an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula:
Figure imgf000023_0001
wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
In another embodiment, the present invention further encompasεeε methods for producing large quantitieε of known, naturally occurring aεterriquinoneε in high purity and in high yield. In yet another embodiment, the present invention is directed to known, synthetically prepared naturally occurring aεterriquinoneε of high purity which are obtainable in large quantitieε and in high yield. The invention also encompasses the preparation of novel monoindolylquinones, i.e., compounds εubεtituted with only one indole, and the monoindolylquinone compoundε, as deεcribed below.
Other featureε and advantageε of the invention will be apparent from the following deεcription of the preferred embodimentε thereof, and from the claimε.
4. DETAILED DESCRIPTION OF THE INVENTION
As mentioned above, the preεent invention relates to a synthetic route for the preparation of a wide variety of indolylquinones including asterriquinone compounds having interesting antitumor activity. As a result, the invention provides a major contribution to the art in that naturally occurring compounds with important therapeutic properties can be prepared in large quantities, e.g., quantities of about 1 gram or more, with purity in excess of about 95% and in high yield. The availability of large quantities allows the skilled artisan to more quickly and easily test these naturally occurring compounds. Prior to the present invention, such compounds were isolated from natural sources in limited quantities (e.g., about 750 mg or less) and low purity (e.g., lesε than about 95%). With the present invention, large quantities can more easily be obtained and the purification of complex natural product mixtures is avoided. Finally, the present invention provides a means for preparing novel analogues of the naturally occurring indolylquinones, which analogues may prove to be of greater interest than the naturally occurring compounds for their therapeutic activity or other properties.
More specifically, the preεent invention provides a solvent based reaction of indoleε and haloquinoneε in the presence of metal carbonate. It has been discovered that the reaction of indoles and halo-quinoneε uεing a polar organic solvent and metal carbonate provides a rapid and efficient reaction under mild conditions, which include but are not limited to mild temperatures, short reaction times and standard/ambient presεureε. Thuε, the preεent method iε well εuited for large εcale and commercial production of indolylquinones.
More specifically, the present invention provides methods for the preparation of indolylquinoneε which involve the reaction of a 2 , 5-dihalo-l, 4-benzoquinone of the formula II with at leaεt one indole of the formula III. When one indole iε uεed, a εy metrical biε-indolylquinone of the formula I is obtained as the product. In the alternative, step-wise or concurrent addition of two different indoles may be uεed to obtain an asymmetrical bis-indolylquinone of the formula VI. For example, an asymmetrical bis-indolylquinone may be made by the reaction of a 2 , 5-dihalo-l, 4-benzoquinone with about one equivalent of a first'indole, followed by addition of about 1.5 equivalents of a second indole. Preferably, the preparation of asymmetrical bis- indolylquinones is carried out in the presence of about 3 equivalents of metal carbonate. As another alternative, mixtures of two or more indoles may be reacted with the starting quinone to give a mixture of symmetrical and asymmetrical bis-indolylquinones. Finally, the controlled addition of one equivalent of at least one indole to the starting quinone in the presence of about one equivalent of metal carbonate can be used to obtain one or more monoindolylquinones.
The preferred 2 , 5-dihalo-l, 4-quinones uεeful in the preεent invention are the 2 ,5-dibromo-l,4-benzoquinones of formula II, which may be substituted or unsubεtituted, wherein Rl and R2 are each independently Br, Cl, F, I, OH, H or -OCOR, wherein R is lower alkyl, aryl or alkylaryl. Alternatively, the 2 , 5-dichloro-, difluoro- or diiodo-1,4- benzoquinoneε may be uεed in the methodε of the invention. A particularly preferred 2 , 5-dibromo-l, 4-quinone iε 2,3,5,6- tetrabromo-1, 4-benzoquinone.
The indoleε uεeful in the preεent invention may be εubstituted at the 1, 2, 3, 4, 5 or 7 positions with hydrogen, branched or unbranched C^C,, alkyl, alkylcarboxy, C2-Cra alkenyl, C2-Cm alkynyl, alkenylcarboxy, aryl, akylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, preferably 1-7, and m is an integer from 0 to 12, preferably 1-7. Preferably, the indoles uεed in the present invention are substituted at the 2 position. Least preferred indoles are the unsubstituted indoles. Certain indoles useful in the present invention are available from commercial sources such as the Aldrich Chemical Company, Milwaukee, WI. Alternatively, the indoles may be prepared via a cyclization procedure according to the method of Verley and Bedure, 1925, Bull. Soc. Chim. Fr. (37): 190.
Any non-nucleophilic, aprotic solvent may be used in the methods of the invention. Mixtures of solvents may also be used. Preferred εolventε are inert or non-reactive, polar organic solventε including but not limited to acetonitrile, dimethyl formamide (DMF) and tetrahydrofuran (THF) . A particularly preferred solvent is acetonitrile. The solvent volume will depend upon the scale of the reactors, and may range from a few milliliters up to a multi-liter volumes useful in large-scale production. Reactant concentrations are set forth below.
It is believed that the metal carbonate used in the methods of the present invention assiεts the reaction of the 2,5-dihalo-l,4-quinone and the indole by scavenging the hydrogen bromide by-product formed in the reaction. Any metal carbonate or mixture of metal carbonates may be uεed; however, cesium carbonate, potasεiu carbonate, sodium carbonate, lithium carbonate and mixtureε thereof are preferred. A particularly preferred metal carbonate iε cesium carbonate. The amount of metal carbonate used in the method ranges from about 2 to about 10 equivalentε based on the haloquinone; preferably 2 to 5 equivalentε; and most preferably 3 to 4 equivalents.
The reaction may be run at any concentration ranging from about 0.1M to about 5M (molarity based on the haloquinone) . Preferably the reaction iε run at a concentration of about 1M.
Aε mentioned above, the reaction to produce a mono- indolylquinone iε preferably carried out in the presence of about two equivalents of metal carbonate. Reactions to produce symmetrical or asymmetrical bis-indolylquinones are preferably carried out in the presence of about 3 equivalents of metal carbonate.
According to the methods of the invention, the reaction of the 2,5-dihalo-l,4-quinone with the indole may be run at temperatures ranging from about -10°C to about 100°C. However, a particularly beneficial aspect of the present invention is that harsh conditions and high temperatures are not required to effect this reaction. Preferably the reaction of the 2 , 5-dihalo-l, 4-quinone is run at a temperature in the range of about 0°C to about 30°C. More preferably, the reaction is run at about room temperature. The reaction of the 2, 5-dihalo-l,4-quinone with the indole may be conducted under an inert atmoεphere such as nitrogen or argon; however, the reaction may alεo be run in atmospheric air. The reaction may be run at any pressure up to 500 psig; however, it is preferable to conduct the reaction at atmospheric pressure.
The reaction time will vary according to the specific reactants and reaction conditions used, but generally will be from about 2 hours to about 72 hours. After reaction of the 2 , 5-dihalo-l, 4-quinone and the indole, the product indolylquinoneε are typically iεolated according to standard workup procedures. For example, the crude reaction mixture may be diluted with IN hydrochloric acid, followed by extraction with an organic solvent such as ethyl acetate. Typically, the organic layer is waεhed with brine and then dried over anhydrouε εodium sulfate. As an alternative to extraction, the crude reaction mixture may simply be filtered to remove solidε. The solvent is removed under reduced pressure, and the crude residue is purified by recrystallization, flash chromatography, High Pressure Liquid Chromatography (HPLC) or a combination thereof. Preferably, the residue iε purified uεing flash chromatography and/or High Presεure Liquid Chromatography (HPLC) .
In a preferred embodiment, the εymmetrical biε- indolylquinone of formula I is further reacted with an alkali metal hydroxide to give a biε-indol-2, 5-dihydroxy-l,4-quinone of formula IV. Preferred alkali metal hydroxides are sodium and potassium hydroxide, or mixtures thereof. Preferably, this reaction is carried out in a mixture of ethanol and tetrahydrofuran using concentrated aqueouε KOH at a reflux temperature of about 85°C for up to 20 hourε. The bis-indol- 2, 5-dihydroxy-1,4-quinone of formula IV maybe isolated according to standard workup and purification procedures as described above. In other embodiments, the methods of the present invention comprise reacting a symmetrical compound of formula I wherein Rl and R2 are Br, or an asymmetrical compound of formula VI wherein Rl and R2 are Br, with an alkali metal hydroxide and an alcohol of the formula R'OH wherein R' iε lower alkyl or alkylaryl, to a produce symmetrical indolylquinone of the formula V or an asymmetrical indolylquinone of formula XI .
As used herein the term "εubstituted or unεubstituted" means that the group in question can be substituted with one or more subεtituentε as desired; for example, εubstituents other than hydrogen such as Br, Cl, F, I, NH2, NR2 , N02, CN, COR, OH, OR, S02, alkyl, aryl, alkylaryl and the like may be used. On the contrary, unsubεtituted groupε do not contain any substituentε .
By the term "alkyl" as uεed herein iε meant a straight or branched chain εaturated hydrocarbon group having from 1 to 12 carbonε εuch aε methyl, ethyl, isopropyl, n-butyl, s- butyl, t-butyl, n-amyl, isoamyl, n-hexyl, n-octyl and n- decyl; "alkenyl" and "alkynyl" are used to mean straight or branched chain hydrocarbon groups having from 2 to 12 carbons and unsaturated by a double or triple bond respectively, such as vinyl, allyl, propargyl, 1-methylvinyl, but-1-enyl, but-2- enyl, but-2-ynyl, 1 methylbut-2-enyl, pent-1-enyl, pent-3- enyl, 3-methylbut-l-ynyl, 1, 1-dimethylallyl, hex-2-enyl and 1-methyl-l-ethylallyl; "alkylaryl" means the aforementioned alkyl groupε substituted by a phenyl group such as benzyl, phenethyl, phenopropyl, 1-benzylethyl, phenobutyl and 2- benzylpropyl; "aryl" as used herein includes a monocyclic or bicyclic rings, wherein at least one ring is aromatic including aromatic or hetero-aromatic hydrocarbons; the term "hydroxy-alkyl" means the aforementioned alkyl groups substituted by a single hydroxyl group such as 2- hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 4- hydroxybutyl, 1-hydroxybutyl and 6-hydroxyhexyl.
Specific compounds which can be made according to the methods of the present invention are described by formula (XII) below. R1-R12 of the formula can be as listed in
Table I following the formula. Illustrative preparations of these compounds are found in the working examples.
Figure imgf000029_0001
TABLE I
Figure imgf000029_0002
Unless otherwise indicated, R3-R10 = hydrogen.
Figure imgf000030_0001
Figure imgf000031_0001
The mono-indolylquinone compounds which can be made according to the methods of the present invention are described by formula (XI) below. R1-R7 and R30 can be as listed in Table II below.
Figure imgf000032_0001
The following is a general experimental procedure for the synthesis of the trihalo-monoindolyl quinones of formula XI using the appropriate indoles. Aε discussed above, the indoles may be commercially available or may be prepared according to the method of Verley and Bedure, 1925, Bull. Soc. Chim. Fr. (37): 190.
Preparation of 6- (2-phenylindol-3-yl)-2,3,5-tribromo- 1,4-quinone [Compound (48a)]: To a 25 ml round bottom flask equipped with a magnetic stir bar and a drying tube was placed 2-phenylindole (2.28 g) , cesium carbonate (7.69 g) , bromanil (5 g) , and acetonitrile (11.8 ml). After stirring the mixture at room temperature for 3 hours, IN hydrochloric acid (150 ml) was added and the aqueous layer extracted with ethyl acetate (300 ml) . The organic layer was washed with brine (150 ml) and dried with sodium sulfate. Following removal of the solvent under reduced presεure, the crude residue was purified by flash chromatography (10% ethyl acetate/hexane) to yield 6-(2-phenylindol-3-yl) -2,3,5- tribromo-1, 4-quinone (1.88 g, 30%) as a blue crystalline solid. TABLE II
Figure imgf000033_0001
Unless otherwise indicated, R3-R6 = hydrogen.
Figure imgf000034_0001
As mentioned above, the compounds of Tables I and II have therapeutic activity for example as antifungal agents, antibacterial agents, and antitumor agents. In addition, these compounds may be useful in the dye industry.
5. WORKING EXAMPLES
In this section, examples of the methods described above are provided for illustration only and not by way of limitation. The reactants and starting materials are either readily synthesized or purchased from commercial sources. Example 1
2 , 5-Dihydroxy-3 , 6-di- [2- (3-methyl-n-butyl) indol-3-yl] -1, 4- quinone
1) Into a 250 ml round bottom flask, equipped with a magnetic stir bar, was placed 2- (3-methyl-n-butyl) indole (2.95 g) , cesium carbonate (10.3 g) bromanil (3.34 g) , and acetonitrile (79 ml) . The mixture was stirred at room temperature for 45 hourε. Following dilution with 1 N hydrochloric acid (250 ml) , the crude mixture was extracted with ethyl acetate (500 ml) . The organic layer was waεhed with brine (200 ml) and dried with εodium sulfate. After removal of solvent under reduced pressure, the crude residue was filtered through a short plug of flash silica, eluting with 20% ethyl acetate/hexane. The solvent was removed under reduced preεsure, and the residue was purified by flash chromatography (15% ethyl acetate/hexane) to yield 2,5- dibromo-3,6-di-[2-(3-methyl-n-butyl) indol-3-yl] -1,4-quinone (553 g, 11%) as a blue crystalline solid.
Note: When the reaction was performed on 27.2 mg of starting indole, 95 mg cesium carbonate, 31 mg bromanil and 0.72 ml acetonitrile, the reaction was complete after 1.5 hours, and the product yield was 28%.
2) To a stirred solution of 2 ,5-dibromo-3, 6-di-[2- (3-methyl- n-butyl) indol-3-yl]-l,4-quinone (553 mg) , ethanol (9 ml), and tetrahydrofuran (9 ml) in a 100 ml round bottom flask equipped with a reflux condenser was added 2N aqueous potassium hydroxide solution (9 ml) . The mixture was heated at 85°C for 13 hours, followed by dilution with IN aqueous sodium hydroxide solution (150 ml) . The mixture was washed with 3:1 hexane/ethyl acetate (400 ml). After setting aside the aqueous layer, the organic layer was washed with another portion of IN aqueous sodium hydroxide solution (150 ml) and then discarded. The basic aqueous layers were combined, acidified by adding 6N hydrochloric acid (60 ml) , and extracted with ethyl acetate (300 ml) . The organic layer was washed with brine (100 ml) and dried with sodium sulfate. Removal of solvent afforded 345 mg (78%) of 2 , 5-Dihydroxy- 3,6-di-[2-(3-methyl-n-butyl) indol-3-yl]-l,4-quinone as a reddish-purple crystalline solid.
3) Preparation of 2- (3-methyl-n-butyl) -indole. Carbonyldiimidazole (65 g) was mixed with 500 ml of dry dichloromethane in a 2-L round bottom flask and stirred magnetically. A solution of 4-methylvaleric acid in 200 ml of dichloromethane was added dropwise over 45 minutes and the mixture was stirred for another 1.25 hours. o-Toluidine (45 g) in 100 ml of dichloromethane was then added over about 20 minutes. After εtirring for 2 hourε the mixture was washed with water and then the solvent was stripped on a rotary evaporator. The reεidue waε mixed with 150 ml of methanol and 75 ml of water and put in the freezer. Filtration of the precipitate, dilution of the filtrate with water and refiltration of the precipitate gave 75 g (94%) of vacuum dried N- (2-methylphenyl) -4-methylvaleramide which was used without further treatment in the next step.
The following procedure iε cited in Bull. Soc. Chim. Fr. (37):190 (1925). N- (2-methylphenyl) -4-methylvaleramide (20.5 g) , sodium amide (90%) (11.0 g) , and tetralin (100 ml) were mixed in a 500 ml round bottom flask equipped with a magnetic stirrer and reflux condenεer and heated at reflux for 2 hourε. After cooling to room temperature, ethanol (10 ml) was added, followed by H20 (150 ml) . The layers were separated, the organic layer was filtered through a pad of anhydrous magneεium εulfate, and the εolution was placed in a 200 ml round bottom flask equipped with a 10-inch vacuum jacketed Vigreux column. Tetralin was distilled at 35- 45°C/0.5 mm Hg. The residue was transferred to a 50 ml round bottom flask equipped with a 4-inch Vigreux column, and distillation at 118-129°C/0.5 mm Hg provided 2-(3-methyl-n- butyl) -indole 13.1 g (70%) as a slightly yellow solid. Example 2
2,5-Dihydroxy-3,6-di-[2-(3-methyl-n-butyl) indol-3-yl]-l,4- quinone (larger scale)
1) Into a 250 ml round bottom flask, equipped with a magnetic stir bar, was placed 2- (3-methyl-n-butyl) indole (30.0 g) , cesium carbonate (62.6 g) bromanil (27.2 g) , and acetonitrile (64 ml) . The mixture was stirred at room temperature for 20 hours. Following dilution with IN hydrochloric acid (500 ml) , the crude mixture was extracted with ethyl acetate (1 L) . The organic layer was washed with brine (400 ml) and dried with sodium sulfate. The solvent was removed under reduced presεure, the reεidue was purified by flash chromatography (30% ethyl acetate/hexane) to yield a 1:1 mixture of the desired product, 2 , 5-dibromo-3 , 6-di-[2- (3- methyl-n-butyl) indol-3-yl]-l,4-quinone and the undesired byproduct, 2,6-dibromo-3,5-di-[2-(3-methyl-n-butyl) indol-3- yl]-l,4-quinone (42.3 g, 46% yield of desired product by HPLC) as a blue cryεtalline εolid.
2) To a stirred solution of 1:1 mixture of 2 ,5-dibromo-3 , 6- di- [2- (3-methyl-n-butyl) indol-3-yl] -1,4-quinone and 2,6- dibromo-3 , 5-di [2- (3-methyl-n-butyl) indol-3-yl] -1, 4-quinone (42.3 g) , ethanol (166 ml), and tetrahydrofuran (166 ml) in a 1 L 3-necked round bottom flask equipped with a reflux condenser was added 4N aqueous potassium hydroxide solution (166 ml) . The mixture was heated at 85°C for 10 hours, followed by dilution with IN hydrochloric acid (500 ml) . The mixture was extracted with ethyl acetate (1 L) . The organic layer was washed with brine (250 ml) and dried with sodium sulfate. Removal of solvent afforded 35.1 g of crude (about 50% pure) 2 , 5-Dihydroxy-3 , 6-di-[ 2-3-methyl-n-butyl) indol-3- yl]-l,4-quinone to be purified by HPLC.
In a εeparate experiment, purification of about 20 g of the crude product (about 50% pure) by HPLC resulted in 5 g of 2,5-Dihydroxy-3,6-di-[2-(3-methyl-n-butyl) indol-3-yl]-l,4- quinone with a purity of 97%. Example 3
2 , 5-Dihydroxy-3 , 6-di- (2-n-butyl-indol-3-yl) -l, 4-quinone
This compound was synthesized in the same manner as Example 2 except the starting indole was 2-n-butyl-indole.
Preparation of 2-n-butyl-indole o-Toluidine (55 g) was mixed with 100 ml dry pyridine and 200 ml dry tetrahydrofuran in a 1-L 3-necked round bottom flask fitted with a Trubore stirrer, thermometer and a dropping funnel, under nitrogen. Then, with cooling in a refrigerated bath, valeryl chloride (60.3 g) was added dropwise over 1 hour. The mixture was stirred for another hour at room temperature and then poured onto 500 g ice and water. The precipitate was washed repeatedly with water on a Buchner funnel. The precipitate (88.9 g, 93%) was cyclized according to Verley and Bedure, 1925, Bull. Soc. Chi . Fr. (37): 190 to afford 2-n-butyl indole (67.4 g, 84%) as a very slightly yellow oil.
Example 4
2 , 5-Dihydroxy-3 , 6-di-[2-methyl-indol-3-yl] -1, 4-quinone
This compound waε synthesized in the same manner as Example 2 except the reaction time was 24 hours.
Example 5
2 , 5-Dihydroxy-3 , 6-di- [2- (2-methylbut-2-en-4-yl) indol-3- yl] 1,4-quinone
This compound may be synthesized as follows: A mixture of 100 mg of 2 , 5-diacetoxy-3 , 6-dibromo-l, 4- quinone, 180 mg of 3-[2- (2-methylbut-2-en-4-yl) indole, prepared by the Fisher indole synthesis, 10 ml of anhydrous dimethylforamide, and powdered potassium carbonate, is heated at 100°C for 24 hours. The cooled mixture is partitioned between ethyl acetate and water. The ethyl acetate layer is then washed with brine, dried over sodium sulfate, filtered and concentrated. The crude product is then purified on a medium pressure liquid chromatography column in a solvent mixture of dichloromethane and methanol to provide 25 mg of 2 , 5-diacetoxy-3 , 6-di- [ 2- (2-methylbut-2-en-4-yl) indol-3- yl] 1 , 4-quinone. 2 , 5-Diacetoxy-3 , 6-di- [ 2 (2-methylbut-2-en-4- yl) indol-3-yl]l,4-quinone is then hydrolysed with 1 N aqueous sodium hydroxide εolution in methanol. Acidification of the above mixture produces the crude product after filtration. Further crystallization in ethanol and water produces the title compound. Other suitably protected quinones such as 3 , 6-dibromo- 2,5-ditrimethylεiloxy-l,4-quinone, 3 , 6-dibromo-2 , 5-di- (t- butyldimethylsiloxy-1 , 4-quinone, 2 , 5-dibenzoxy-3 , 6-dibromo- 1,4-quinone, 3 , 6-dibromo-2 , 5-diisobutyroxy-l, 4-quinone, 2,5- dibenzyloxy-3,6-dibromo-l,4-quinone or 2,5- diallyoxycarbonyloxy-3 , 6-dibromo-l, 4-quinone which can be prepared from commercially available 2,4-dibromo-3 , 6- dihydroxy-l,4-quinone may also be used as starting materials. These protecting groups can be removed by conventional deprotection methods such as diluted acid, potasεium fluoride or palladium (0) complex or palladium on carbon with hydrogen or by methodε deεcribed by Greene and Wutε (1991, "Protective Groupε In Organic Syntheεiε," John Wiley and Son). Other solvents such as pyridine or dimethylsulfoxide (DMSO) may be used in place of dimethyl formamide.
Example 6
2 , 5-Diacetoxy-3 , 6-di- [ 2- (2-methylbut-2-en-4-yl) indol-3- yl]l,4-quinone
2 , 5-Diacetoxy-3 , 6-di- [2- (2-methylbut-2-en-4-yl) indol-3- yl]l,4-quinone is prepared as in Example 5.
Example 7
2 , 5-Diacetoxy-3 , 6-di- [ 2 (3-methyl-n-butyl) indol-3-yl] 1 , 4- quinone Hydrogenation of 2,5-diacetoxy-3 , 6-di- [2- (2-methylbut-2-en-4- yl) indol-3-yl]l,4-quinone in methanol with 5% palladium on carbon under 1 atmosphere of hydrogen produced the title compound.
Example 8
2 , 5-Dihydroxy-3 , 6-di- [ 2 - ( 3 -methyl-n-butyl ) indol-3 -yl ] 1 , 4 - quinone
Base hydrolysiε of 2, 5-diacetoxy-3 , 6-di- [2- (3-methyl-n- butyl) indol-3-yl] 1,4-quinone aε described in Example 5 produced the title compound.
Under similar conditions as thoεe described in Examples 5 to 8, the following compounds are prepared using either 2,5- dibromo-3,6-dihydroxy-l,4-quinone or 2,3,5,6- tetrabromoquinone as starting materials:
Example 9 3 , 6-Di- [5- (bromo) indol-3-yl] -2 , 5-dihydroxy-l, 4-quinone
Example 10
3 , 6-Di- [ 2- (allyl) indol-3-y1] -2 , 5-dihydroxy-l , 4-quinone
Example 11
2 , 5-Dihydroxy-3 , 6-di- [2- (n-propyl) indol-3-yl] 1, 4-quinone
This compound was prepared under conditionε similar to those described in Examples 5 to 8.
Example 12
3 , 6-Di- [2- (aminocarbonyl) indol-3-yl] -2 , 5-dihydroxy-l, 4- quinone
Example 13
2 , 5-Diacetoxy-3 , 6-di- [ 2 (aminocarbonyl) indol-3-yl] -1, 4-quinone Example 14
3 , 6-Di-[2-allylindol-3-yl] -2 , 5-dibenzoyloxy-l, 4-quinone
Example 15 2 , 5-Dihydroxy-3 , 6-di- [2- (cyano) indol-3-yl] 1, 4-quinone
Example 16
2 , 5-Dihydroxy-3 , 6-di- [ 4- ( ethoxycarbonyl) indol-3-yl] 1 , 4- quinone
Example 17
2 , 5-Dihydroxy-3 , 6-di- [5,7- (dimethoxy) indol-3-yl] 1, 4-quinone
Example 18 2 , 5-Dihydroxy-3 , 6-di- [4,7- (dimethoxy) indol-3-yl] 1, 4-quinone
Example 19
2 , 5-Dihydroxy-3 , 6-di- [5- (nitro) indol-3-yl] 1, 4-quinone
Example 20
3, 6-di- [4 (4-chlorobenzoylamino) indol-3-yl]-2,5-dihydroxy-l, 4- quinone
Example 21 3, 6-di- [2- (4-chlorophenyl) indol-3-yl]-2, 5-dihydroxy-l, 4- quinone
Example 22
2 , 5-Dihydroxy-3 , 6-di- [2- (4-fluorophenyl) indol-3-yl] 1, 4- quinone
Example 23
2,5-Dihydroxy-3, 6-di- [4, 6- (dimethoxy) indol-3-yl] 1,4-quinone
Example 24
2 , 5-Dihydroxy-3 , 6-di- [ 2- ( 5-hydroxy-6-methoxy) indol-3-yl] l , 4- quinone Example 25
2,5-Dihydroxy-3,6-di-[4- (cyano) indol-3-yl] 1,4-quinone
Example 26 2,5-Dihydroxy-3,6-di-[5- (4- trif luoromethylphenylaminocarbonyl) indol-3-yl] 1, 4-quinone
Example 27
2 , 5-Dihydroxy-3 , 6-di- [2- (4- trifluoromethylphenylaminocarbonyl) indol-3-yl] 1,4-quinone
Example 28
2 , 5-Dihydroxy-3 , 6-di- [2- (ethyl) indol-3-yl] 1, 4-quinone
This compound was prepared under conditions similar to those described in Examples 5 to 8.
Example 29
3,6-di-[2-(5-bromo-6-nitro) indol-3-yl]-2,5-dihydroxy-l,4- quinone
Example 30
2 , 5-Dimethoxy-3 , 6-di- [2- (2-methylbut-2-en-4-yl) indol-3- yl] 1 , 4-quinone
Methylation of Example 5 with methyl iodide and potasεium carbonate in dimethylforamide followed by purification produces the title compound. This compound may also be prepared by heating 2 , 5-dibromo-3, 6-di [2- (2-methylbut-2-en-4- yl) indol-3-y]l,4-quinone in methanol in the presence of powdered potassium carbonate.
Example 31
2 , 5-Dimethoxy-3 , 6-di- [2 (3 -methyl-n-butyl) indol-3-yl] 1 , 4- quinone Hydrogenation of Example 30 under conditions as those in Example 3 produced the title compound.
Example 32 Preparation of 2, 5-Dihydroxy-3, 6-di-[2-(3-methyl-n-butyl) indol-3-yl] -1, 4-quinone
To a glass tube containing 2- (3-methyl-n-butyl) indole (400 mg) , bromanil (431 mg) and potasεium carbonate (703 mg) , equipped with a magnetic stir bar, was added dimethylformamide (10 ml) . The mixture was stirred at room temperature for 40 hours. Following dilution with IN hydrochloric acid (100 ml) , the crude mixture was extracted with ethyl acetate (200 ml) . The organic layer was washed with brine (100 ml) and dried with sodium sulfate. After removal of solvent under reduced presεure, the crude residue was filtered through a short plug of flash εilica, eluting with 30% ethyl acetate/hexane. The εolvent was removed under reduced preεεure, and the reεidue was purified by flash chromatography (15% ethyl acetate/hexane) to yield 2,5- dibromo-3 , 6-di-[2- (3-methyl-n-butyl) indol-3-yl]-l, 4-quinone (40 mg, 7%) as a blue crystalline solid.
To a stirred solution of 2,5-dibromo-3, 6-di- [2- (3- methyl-n-butyl) indol-3-yl]-l, 4-quinone (40 mg) in methanol (1.5 ml) was added 2N methanolic sodium hydroxide (0.251 ml). The solution was εtirred at room temperature for 24 hours, followed by dilution with water (50 ml) . The product was extracted with ethyl acetate (100 ml) , washed with brine (50 ml) and dried with sodium εulfate. Removal of solvent under reduced presεure provided 2 , 5-methoxy-3 , 6-di-[2- (3-methyl-n- butyl) indol-3-yl] -1,4-quinone (30 mg, 90%) as a yellow crystalline solid.
To a stirred solution of 2, 5-dimethoxy-3, 6-di-[2- (3- methyl-n-butyl) indol-3-yl]-l,4-quinone (9 mg) in ethanol (2 ml) was added 1 N aqueous potassium hydroxide (1 ml) . The mixture was heated at 85°C for 3.5 hours, then diluted with 1 N hydrochloric acid (25 ml) . The product was extracted with ethyl acetate (50 ml) , washed with brine (25 ml) and dried with sodium sulfate. The solvent was removed under reduced pressure to afford 2 , 5-dihydroxy-3 , 6-di- [2- (3-methyl- n-butyl) indol-3-yl] -1,4-quinone (8 mg) as a reddish-brown crystalline solid. 32a) Preparation of 2-(2-methyl-l-buten-4-yl) indole To a stirred solution of 2-methylindole (lg) in diethylether (76 ml) under nitrogen was added a 1.6 M solution of n-butyllithium in hexane (14.3 ml) slowly dropwise via syringe. Potassium tert-butoxide (1.711 g) was then added, producing a bright yellow mixture.
After stirring at room temperature under nitrogen for 50 minutes, the mixture waε cooled to -78 °C, whereupon 3- bromo-2-methylpropene (1.54 ml) was added dropwiεe via εyringe, giving a red-orange εolution. The reaction mixture was stirred at -78 °C for 2 hours, then quenched with water (10 ml) . After warming to room temperature, water (150 ml) and 1 N hydrochloric acid (1 ml) was added to neutralize the reaction mixture. The mixture was extracted with ethyl acetate (250 ml) , and the organic layer waε waεhed with brine (100 ml) and dried with sodium εulfate. The εolvent waε removed under reduced preεεure, and the crude residue was purified by flash chromatography (4% ethyl acetate/hexane) to afford 2-(2-methyl-l-butene-4-yl) indole (664 mg. 47%) as a waxy yellow εolid.
32b) Thiε indole iε preferably synthesized by the method of Example 1. However, this indole may also be prepared as follows: Preparation of 2- (3-methyl-n-butyl) indole
Into a 3-necked round bottom flask under a blanket of nitrogen was placed 5% palladium catalyst on charcoal (771 mg) . A solution of 2- (2-methyl-l-buten-4-yl) indole (671 mg) in ethanol (36 ml) was added to the flask, which was evacuated and charged with hydrogen twice. The mixture was stirred vigorously under hydrogen (1 atmosphere) for 2 hours, followed by filtration through a pad of Celite. The solvent was removed under reduced presεure and the crude residue was purified by flash chromatography (3% ethyl acetate/hexane) to give 2- (3-methyl-n-butyl) indole (400 mg, 59%) as a yellow cryεtalline solid.
Example 33
Preparation of 2 , 5-Dihydroxy-3 , 6-di- [2- (methyl) indol-3-yl]- 1, 4-quinone This compound iε preferably synthesized by the method of Example 2 using 2-methylindole as the starting indole. However, this compound may also be prepared by the method of Example 32 using 2-methylindole as the starting indole.
Example 34
Preparation of 3 , 6-Di- (2-ethylindol-3-yl) -2, 5-dihydroxy-l, 4- quinone
This compound is preferably synthesized by the method of Example 2 using 2-ethylindole as the starting indole. However, this compound may also be prepared by the method of
Example 32 using 2-ethylindole as the starting indole. Preparation of 2-ethylindole: Refer to 32a) using methyl iodide as the alkylating agent.
Example 35
Preparation of 3 , 6-Di- (2-butylindol-3-yl) 2 , 5-dihydroxy-l,4- quinone: This compound iε preferably synthesized by the method of Example 2 using 2-butylindole as the starting indole. Preparation of 2-butylindole: Refer to Example 3. However, this compound may also be prepared by the method of
Example 32 using 2-butylindole as the starting indole. Preparation of 2- (but-l-en-4-yl), indole: Refer to 32a) using allyl bromide as the alkylating agent. Preparation of 2- butylindole: Refer to 32b) using 2- (but-l-en-4-yl) indole as the starting material. Example 36
Preparation of 3,6-Di-[2-(but-l-en-4-yl) indol-3-yl] 2,5- dihydroxy-1, 4-quinone
This compound is preferably synthesized by the method of Example 2 but may also be prepared according to the method of Example 32 using 2- (but-l-en-4-yl) indole as the starting indole.
Example 37 Preparation of 2 , 5-Dihydroxy-3 , 6-di- [2- (4-methyl-n-pentyl) indol-3-yl]-l,4-quinone: This compound iε preferably synthesized by the method of Example 2 but may also be prepared according to Example 32 using 2- (4-methyl-n-pentyl) indole as the εtarting indole. Preparation of 2- (2-methyl-2- penten-5-yl) indole: Refer to Example 1 using 5- methylhexanoic acid as the starting acid. This indole may also be prepared according to Example 32a) using 4-bromo-2- methyl-2-butene as the alkylating reagent. Preparation of 2-(4-methyl-n-pentyl) indole: Refer to 32b) using 2- (2- methyl-2-penten-5-yl) indole as the starting material.
Example 38
Preparation of 2,5-Dihydroxy-3,6-di-[2-(2-phenylethyl) indol-
3-yl]-l, 4-quinone: This compound is preferably εynthesized by the method of Example 2 but may also be prepared according to Example 32 using 2- (2-phenylethyl) indole as the starting indole. Preparation of 2- (2-phenylethyl) indole: Refer to Example 3 using 3-phenylpropionyl chloride as the starting acid chloride. This indole may also be prepared according to Example 32a) using benzyl bromide aε the alkylating agent.
Example 39
Preparation of 2,5-Dihydroxy-6-(indol-3-yl) -3-[2- (3-methyl-n- butyl) indol-3-yl] -1,4-quinone This synthesis is achieved by treating 2- (3-methyl-n- butyl) indole with 2 equivalents of bromanil in the presence of cesium carbonate in dimethylformamide, followed by workup and purification similar to Example 32. The resultant mono- indolyl adduct iε optionally treated with 2 equivalents of indole under the same conditions aε above to provide the biε- indolyl product.
Example 40
Preparation of 3 , 6-Di- (5-carboxy-2-ethylindol-3-yl) -2,5- dihydroxy-1, 4-quinone: Refer to Example 32 uεing 5-carboxy- 2-ethylindole aε the starting indole. Preparation of 5- carboxy-2-ethylindole: Refer to Example 3 using methyl 4- amino-3-methylbenzoate and propionyl chloride as the starting compoundε. The methyl eεter iε hydrolyzed upon workup of the cyclization to give 5-carboxy-2-ethylindole. Thiε εynthesiε may also be accomplished beginning with 5-chloro-2- methylindole, which is alkylated with methyl indole. The product chloroindole is converted to its Grignard specieε and exposed to carbon dioxide to finiεh the synthesis.
Example 41 Preparation of 3 , 6-Di- [5-carboxy-2- (n-propyl) indol-3-yl]-
2 , 5-dihydroxy-l, 4-quinone:
Refer to Example 32 using 5-carboxy-2-propylindole as the starting indole. Preparation of 5-carboxy-2- propylindole: Refer to Example 1 using methyl 4-amino-3- methyl-benzoate instead of o-toluidine or refer to 40 uεing ethyl iodide as the alkylating agent.
Example 42
Preparation of 3, 6-Di-[5-carboxy-2-(3-methyl-n-butyl) indol- 3-yl]-2,5-dihydroxy-l, 4-quinone
Refer to Example 32 using 5-carboxy-2-(3-methyl-n-butyl) indole as the starting indole. Preparation of 5-carboxy-2- (2-methyl-l-buten-4-yl) indole:
Refer to 40 using 3-bromo-2-methylpropene as the alkylating agent. Preparation of 5-carboxy-2-(3- methyl-n-butyl) indole: Refer to Example 1 using methyl 4-amino-3-methyl-benzoate instead of o- toluidine, or refer to Example 32b) using 5- carboxy-2-(2-methyl-l-buten-4-yl) indole as the starting material.
Example 43
Preparation of 3 , 6-Di- [2- (4-carboxy-n-butyl) indol-3-yl] -2 , 5- dihydroxy-1 , 4-quinone:
Refer to Example 32 using 2- (4-carboxy-n-butyl) indole as the starting indole. Preparation of 2- (4-carboxy-3-buten- 1-yl) indole:
Refer to 32 (a) using 4-bromo-2-butenoic acid as the alkylating agent. Preparation of 2- (4-carboxy-n- butyl) indole: Refer to Example 3 using methyl adipyl chloride as the acid chloride. The methyl ester was hydrolyzed in the cyclization workup to provide the product carboxyindole. In the alternative, refer to Example 32b) using 2- (4- carboxy-3-buten-l-yl) indole as the starting material.
Example 44
Preparation of 3- [5-Carboxy-2-(3-methyl-n-butyl) indol-3-yl]-
2,5-dihydroxy-6- (indol-3-yl) -1,4-quinone
Refer to Example 39 using 5-carboxy-2-(3-methyl-n-butyl) indole in the first step.
Example 45
Preparation of 3 , 6-Di- (5-amino-2-ethylindol-3-yl) -2,5- dihydroxy-1 , 4-quinone Refer to Example 32 using 5-amino-2-ethylindole aε the starting indole. Preparation of 5-amino-2-ethylindole: Refer to Example 3 using 2-methyl-4-nitroaniline and propionyl chloride to give 5-nitro-2-ethylindole, which is reduced to the desired amino compound using catalytic hydrogenation as in 32b.
In the alternative, this synthesis may be accomplished with a standard nitration of 2-ethylindole using sodium nitrate and sulfuric acid similar to that cited in Chem. Lett. (7): 1125-1128 (1991). The resultant 5-nitro-2- ethylindole is reduced to the deεired amino compound uεing catalytic hydrogenation as in 32b) .
Example 46
Preparation. of 3, 6-Di- [5-amino-2- (n-propyl) indol-3-yl]-2 , 5- dihydroxy-1 , 4-quinone
Refer to Example 32 using 5-amino-2- (n-propyl) indole as the εtarting indole. Preparation of 5-amino-2- (n-propyl) indole: Refer to Example 45 using butyryl chloride. In the alternative, refer to the εynthesis cited in Chem. Lett. (7) : 1125-1128 (1991) cited in Example 45 using 2-n-propylindole.
Example 47
Preparation of 3, 6-Di-[5-amino-2- (3-methyl-n-butyl) indol-3- yl] 2,5-dihydroxy-l,4-quinone
Refer to Example 32 uεing 5-amino-2- (3-methyl-n-butyl) indole aε the starting indole. Preparation of 5-amino-2- (3- methyl-n-butyl) indole: Refer to Example 1 using 2-methyl-4- nitroaniline instead of o-toluidine. The resultant 5-nitro-
2- (3-methyl-n-butyl) -indole is reduced to the deεired amino compound aε in 32b. The synthesiε may alεo be accomplished according to Example 45 using 2- (3-methyl-n-butyl) indole.
Example 48
Preparation of 2, 5-Diacetoxy-3,6-di-[2-(3-methyl-n-butyl) indol-3-yl] -1 , 4-quinone
This syntheεiε waε accomplished by treating 2,5-hydroxy- 3,6-di-[2-(3-methyl-n-butyl) indol-3-yl]-l,4-quinone with acetic anhydride in the presence of pyridine.
Example 49
Preparation of 3, 6-Di-[2-ethyl-5-(4- methylphenylsulfonylamino) indol-3-yl]-2,5-dihydroxy-l,4- quinone Refer to Example 32 uεing 2-ethyl-5-(4- methylphenylεulfonylamino) indole aε the starting indole. Preparation of 2-ethyl-5- (4-methylphenylsulfonylamino) indole: The above compound iε synthesized by treating 5- amino-2-ethylindole with p-toluenesulfonyl chloride in the presence of triethylamine.
Example 50
Preparation of 2,5-Dihydroxy-3, 6-di-[5- (4- methylphenylsulfonyla ino) -2- (n-propyl) indol-3-yl]-l,4- quinone
Refer to Example 32 uεing 5- (4- methylphenylεulfonyla ino) -2- (n-propyl) indole as the starting indole. Preparation of 5- (4- methylphenylsulfonylamino) -2- (n-propyl) indole: Refer to 49 using 5-amino-2-propylindole.
Example 51
Preparation of 2 , 5-Dihydroxy-3 , 6-di-[2- (3-methyl-n-butyl) -5- (4-methylphenylsulfonylamino) indol-3-yl] -1,4-quinone
Refer to Example 32 uεing 2- (3-methyl-n-butyl) -5- (4- methylphenylεulfonylamino) indole aε the starting indole.
Preparation of 2- (3-methyl-n-butyl) -5- (4- methylphenylεulfonylamino) indole: Refer to 49 uεing 5- amino-2-(3-methyl-n-butyl) indole.
Example 52
Preparation of 2,5-Dihydroxy-3,6-di-[2-(2-methylbut-l-en-4- yl) indol-3-yl]-l,4-quinone Refer to Example 32 uεing 2- (2-methylbut-l-en-4-yl) indole aε the starting indole.
Example 53
2 , 5-dihydroxy-2 , 6-di- [2- (2-methylpent-2-en-5-yl) -indol-3-yl] 1,4-quinone Example 54
2 , 5-dihydroxy-3 , 6-di- (2-phenylindol-3-y-l) -1, 4-quinone: Refer to Example 2 uεing 2-phenylindole as the starting indole.
Example 55
2 , 5-dihydroxy-3 , 6-di- (2-carboxyindol-3-yl) -1, 4-quinone
Example 56 Preparation of 3- (2-carboxyindol-3-yl) -2,5-dihydroxy-6- (2- methylindol-3-yl) -1, 4-quinone:
1) Into a 10 ml screw-cap glass tube was placed bromanil (1 g) , cesium carbonate (2.3 g) , ethyl indole-2-carboxylate (446 mg) , and acetonitrile (2.36 ml). After stirring the mixture at room temperature for 3 hours, during which the mono-indolylquinone 6- (2-ethylcarboxyindol-3-yl) -2,3,5- tribromo-l,4-quinone waε formed, 2-methylindole (464 mg) waε added. The mixture waε stirred at room temperature for 24 hourε, after which 1 N hydrochloric acid (100 ml) waε added. The aqueouε layer was extracted with ethyl acetate (200 ml) . The organic layer waε waεhed with brine (100 ml) and dried with sodium sulfate. Following removal of the solvent under reduced presεure, the crude residue was purified by flash chromatography (30% ethyl acetate/hexane) to provide 2 , 5-dibromo-3- (2-ethylcarboxyindol-3-yl) -6- (2- methylindol-3-yl) -1,4-quinone (0.37 g) as a blue crystalline solid. Alternatively, the mono-indolylquinone 6- (2- ethylcarboxyindol-3-yl) -2,3, 5-tribromo-l, 4-quinone may isolated separately and used in further reactions with other indoles.
2) To a stirred solution of 2,5-dibromo-3- (2- ethylcarboxyindol-3-yl) -6- (2-methylindol-3-yl) -1, 4-quinone (0.37 g) , ethanol (1.6 ml), and tetrahydrofuran (1.6 ml) waε added 4 N aqueouε potassium hydroxide solution (1.6 ml). The mixture was heated at 85 °C for 10 hours, followed by dilution with 1 N hydrochloric acid (75 ml) . The mixture waε extracted with ethyl acetate (150 ml) . The organic layer was washed with brine (75 ml) and dried with sodium sulfate. Removal of solvent afforded 3- (2-carboxyindol-3-yl) -2 , 5- dihydroxy-6-(2-methylindol-3-yl) -1,4-quinone (0.258 g) aε a reddish brown crystalline solid.
Example 57
Preparation of 2 , 5-dihydroxy-6- (2-methylindol-3-yl) -3- (2- phenylindol-3-yl)-l,4-quinone. This compound was prepared according to the procedure of Example 56. It doesn't matter what order the two indoles are added in - the same product is obtained.
Example 58
Preparation of 2,5-dihydroxy-6-[2- (3-methyl-n-butyl) indol-3- yl]-3-(2-phenylindol-3-yl)-l,4-quinone. This compound waε prepared according to the procedure of Example 56. Aε in Example 57, the order of addition of the two indoles doeεn't matter .
Example 59
Preparation of 6-[2- (n-butyl) -indol-3-yl]-3- (2-carboxyindol- 3-yl) -2,5-dihydroxy-l,4-quinone. This compound waε prepared according to the procedure of Example 56.
Example 60
Preparation of 3- (2-carboxyindol-3-yl) -2, 5-dihydroxy-6-[2- (n- propyl) -indol-3-yl]-l,4-quinone. This compound waε prepared according to the procedure of Example 56.
Example 61
Preparation of 3,6-di(6-carboxy-2-n-propylindol-3-yl) -2,5- dihydroxy-1, 4-quinone: Refer to Example 2 using 6-carboxy-2- n-propylindole as the starting indole. Preparation of 6- carboxy-2-n-propylindole: Refer to Example 3 using methyl 3- amino-4-methylbenzoate and butyryl chloride as the starting compounds. The methyl ester was hydrolyzed upon workup of the cyclization to give 6-carboxy-2-n-propylindole. 6. FORMULATION AND USE OF INDOLYLQUINONES
The compounds disclosed herein have utility, inter alia , at therapeutically effective doses to treat or ameliorate cell proliferative disorders involving PTK/adaptor protein interactions. The compounds prepared according to the present invention may be tested by a variety of methodε for determining the ability of the compoundε to inhibit kinase activity or to disrupt PTK/adaptor protein complexes.
Any assay currently used for screening compoundε that act on cells containing PTKε can be uεed. In general, such asεays involve exposing cells that expreεs the PTK to a test substance and either: (a) scoring phenotypic changes in the cell culture as compared to control cells that were not exposed to the test substance; or (b) biochemically analyzing cell lysates to asεess the level and/or identity of tyrosine phosphorylated proteins.
A common technique involves incubating cells with ligand and radiolabeled phosphate, lysing the cells, separating cellular protein components of the lysate using an SDS-polyacrylamide gel (SDS-PAGE) technique, in either one or two dimensions, and detecting the presence of phoεphorylated proteinε by expoεing X-ray film. In a similar technique, the phosphorylated proteins are detected by immunoblotting techniqueε, in which case the phosphate that is detected is not radiolabeled. Instead, the cellular components separated by SDS-PAGE are transferred to a nitrocellulose membrane, where the presence of phosphorylated tyrosines is detected using an antiphosphotyrosine antibody (anti-PY) . The anti-PY can be detected by labeling it with a radioactive substance, or an enzyme, such aε horseradish peroxidase. A further alternative involves detecting the anti-PY by reacting with a second antibody which recognizeε the anti-PY, thiε second antibody being labeled with either a radioactive moiety or an enzyme as previously described. Examples of these and similar techniques are described in Hansen et al . , 1993, Electrophoresis 14:112-126; Campbell et al . 1993, J^. Biol. Chem. 268:7427-7434; Donato et al . , 1992, Cell Growth and Diff . 3:258-268; and Katagiri et al . , 1993, J_j_ Immunol . 150:585-593.
ELISA-type asεays in microtitre plates can also be used to test purified substrates. See for example Peraldi et al . , 5 1992, J,;. Biochem. 285: 71-78; Schraag et al . , 1993, Analytical Biochemistry 211:233-239; Cleavland, 1990, Analytical Biochemistry 190:249-253; Farley, 1992, Analytical Biochemistry 203:151-157; and Lczaro, 1991, Analytical Biochemistry 192:257-261. Examples of assay methods are 0 described in U.S. Application Serial No. 08/279,321, filed July 22, 1994 and U.S. Application Serial No. 08/488,156 filed June 7, 1995 which are hereby incorporated in their entiretieε by reference.
A variety of methodε may be uεed to aεεay the ability of 5 the compoundε prepared according to the invention to disrupt PTK/adaptor protein complexes. For example, in vitro complex formation may be assayed by, first, immobilizing one component, or a functional portion thereof, of the complex of interest to a solid support. Second, the immobilized complex 0 component may be expoεed to a compound prepared according to the present invention, and to the second component, or a functional portion thereof, of the complex of interest. Third, it may be determined whether or not the second component is still capable of forming a complex with the 5 immobilized component in the presence of the compound.
Additionally, in vivo complex formation may be assayed by utilizing co-immunoprecipitation techniques well known to those of skill in the art. Briefly, a cell line capable of forming a PTK/adaptor complex of interest may be exposed to 0 one or more of the compoundε prepared according to the preεent invention, and a cell lyεate may be prepared from this exposed cell line. An antibody raised against one of the components of the complex of intereεt may be added to the cell lysate, and subjected to standard immunoprecipitation 5 techniques. In cases where a complex is still formed, the immunoprecipitation will precipitate the complex, whereas in cases where the complex has been disrupted, only the complex component to which the antibody is raised will be precipitated.
The effect of a compound of the invention on the transformation capability of the PTK/adaptor protein of interest may be directly aεsayed. For example, one or more of the compounds prepared according to the invention may be administered to a cell such as a fibroblast or hematopoietic cell capable of forming a PTK/adaptor complex which, in the absence of a compound of the invention, would lead to the cell's transformation (Muller, A.J. et al . , 1991, Mol. Cell. Biol. 11:1785-1792; McLaughlin, J. et al . , 1987, Proc. Natl. Acad. Sci. USA 84:6558-6562). The transformation state of the cell may then be measured in vitro, by monitoring, for example, its ability to form colonies in soft agar (Lugo and Witte, 1989, Mol. Cell. Biol. 9:1263-1270; Gishizky, M.L. and Witte, O.N., 1992, Science 256:836-839). Alternatively, a cell's transformation state may be monitored in vivo by determining itε ability to form tumors in immunodeficient nude or severe combined immunodeficiency (SCID) mice (Sawyers, CL. et al . , 1992, Blood 79:2089-2098). Further, the ability of the compounds prepared according to the present invention, to inhibit various tumor cell lines, such as for example, melanoma, prostate, lung and mammary tumor cell lines established as SC xenografts can be examined. Thus, the preεent invention alεo provideε a method of ameliorating εymptoms of a cell proliferative disorder wherein the cell proliferative disorder involves a protein tyrosine kinase polypeptide/adaptor polypeptide complex, with an amount of a compound of either of the formulaε I or XI, sufficient to disrupt protein tyrosine kinase polypeptide/adaptor polypeptide complexes of the cell εo that symptoms of the cell proliferative disorder are ameliorated.
The present invention also provides a method of ameliorating a cell proliferative disorder using a compound described herein, particularly a compound of the formula I, IV, V, VI, VIII, IX, X, XI, XII or XIII, wherein the cell proliferative disorder occurs in a mammal and the compound contacts the cell within a mammal so that the symptoms of the cell proliferative disorder in the mammal are ameliorated. The compounds, i.e., indolylquinones, of the present invention may be uεed alone or in combination with other drugs or therapies to treat cancer.
Cell proliferative disorders which are treatable according to the methods of the invention include BCR-ABL- associated cancers, gliomas, glioblastomas, melanomas, ovarian cancers, breast cancers, and prostate cancers. Further, the present invention provides a method of ameliorating symptoms of a cell proliferative disorder wherein the cell proliferative disorder involves a protein tyrosine kinase polypeptide/adaptor polypeptide complex, which involves contacting a cell capable of forming the protein tyrosine kinase polypeptide/adaptor polypeptide complex with an amount of a pharmaceutical composition comprising a compound of any one of the formulas I, IV, V, VI, VIII, IX, X, XI, XII or XIII, sufficient to disrupt protein tyroεine kinase polypeptide/adaptor polypeptide complexes of the cell so that symptoms of the cell proliferative disorder are ameliorated.
Further, the compounds prepared according to the preεent invention may be formulated into compoεitions comprising other drugs or pharmaceutical agentε. In one embodiment, the pharmaceutical compositions of compounds prepared according to the present invention also comprise additional cancer treatment agents. For example, the compoundε prepared according to the present invention may be formulated into pharmaceutical compositions in a conventional manner using one or more physiologically acceptable carriers or excipients. The compounds and their phyεiologically acceptable salts and solvates may also be formulated for administration by inhalation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration. For oral administration, the pharmaceutical compositions may take the form of, for example, tabletε or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose) ; fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate) ; lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate) ; or wetting agents (e.g., sodium lauryl sulphate). The tabletε may be coated by methodε well known in the art. Liquid preparations for oral administration may take the form of, for example, solutionε, εyrupε or εuεpensions, or they may be presented as a dry product for conεtitution with water or other suitable vehicle before uεe. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such aε εuspending agents (e.g., sorbitol syrup, cellulose derivativeε or hydrogenated edible fatε) ; emulsifying agents (e.g. , lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils) ; and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid) . The preparations may also contain buffer salts, flavoring, coloring and sweetening agents and other pharmaceutical agents as appropriate.
For administration by inhalation, the compoundε for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from presεurized packε or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. Such formulations may also comprise other pharmaceutical agents as appropriate. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. Compoundε prepared according to the present method may also be formulated for parenteral administration by injection, e.g., by boluε injection or continuouε infuεion. Formulationε for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compoεitions may take such forms aε suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
The compounds that can be prepared according to the preεent methodε and their methodε of use can also be found in United Stateε patent application Serial Nos. 08/476,136, filed June 7, 1995 and 08/658,337, filed June 5, 1996, each of which is hereby incorporated by reference.
EXAMPLE - In Vivo Activity of Compound #1 The following example illustrates the use of the compounds of the invention in an in vivo model of tumor growth.
Materials and Methods Female athymic mice (BALB/c, nu/nu) were obtained from Simonsen Laboratories (Gilroy, CA) . All animals were maintained under clean-room conditions in Micro-isolator cages with Alpha-dri bedding. They received sterile rodent chow and water ad libitum. A cell line established from a human epidermoid carcinoma (A431, ATCC CRL 1555) was grown in DMEM with 10% FBS and 2 M GLN. All cell culture media, glutamine, and fetal bovine serum were purchased from Gibco Life Technologies (Grand Island, NY) unless otherwise specified. All cells were grown in a humid atmosphere of 90- 95% air and 5-10% C02 at 37°C. All cell lines were routinely subcultured twice a week and were negative for mycoplasma as determined by teh Mycotect method (Gibco) . Cells were harvested at or near confluency with 0.05% Trypsin-EDTA and pelleted at 450 x g for 10 in. Pellets were resuspended in sterile PBS or media (without FBS) to a particular concentration and the cells were implanted into the hindflank of the mice (8 - 1 mice per group) . Tumor growth waε measured over 3 to 6 weeks using venier calipers. Tumor volumes were calculated as a product of length x width x height unless otherwise indicated. P values were calculated using the Students' t-test. Compound 1 (from Example 1 and Table I) in 100 μL excipient (VPD-.D5W, 1:1 (VPD - 12% w/v polysorbate 80, 0.55% citric acid (anhydrous), 35% w/v polyenthlene glycol (MW = 300 daltons) and 26.3% v/v 190 proof ethanol diluted 1:22 in 5% dextrose in water (D5W) waε delivered by IP injection at different concentrations. Control animals received VPD:D5W alone. Animals were dosed daily on days 1-5 (high dose) , days 1-9 (mid-dose) or throughout the study (low dose) .
Results The results are shown below as a percent tumor reduction compared to controls. Administration of compound 1 inhibited tumor growth in a dose dependent manner. Tumor growth remained inhibited even after ceεsation of treatment with the compound.
Figure imgf000059_0001
The present invention iε not to be limited in scope by the specific examples or embodiments described herein. These examples are, thus, not to be construed as limiting the scope of the invention in any way. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the amended claims.

Claims

WHAT IS CLAIMED IS:
1. A method for preparing an indolylquinone compound of the formula:
Figure imgf000061_0001
wherein:
Rl and R2 are each independently Br, Cl, F, I, H, OH or
-OCOR, wherein R is lower alkyl, aryl or alkylaryl;
R"l iε H, Cx-C7 alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C1-Cn alkyl, alkylcarboxy, C2-Cm alkenyl, alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, a ido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer from 0 to 12 and m is an integer from 0 to 12, which comprises:
reacting a substituted or unsubstituted 2,5-dibromo-l,4- benzoquinone compound of the formula:
Figure imgf000062_0001
wherein Rl and R2 are as defined above;
with at least one indole of the formula
Figure imgf000062_0002
wherein R"l and R3-R7 are as defined above; in a polar organic solvent and in the presence of metal carbonate to produce said indolylquinone compound of formula I.
2. The method of claim 1 which further comprises reacting the indolylquinone compound of formula I with an alkali metal hydroxide to produce a compound of the formula:
Figure imgf000063_0001
wherein R3-R7 are as defined above.
3. The method of claim 1 which further comprises reacting the indolylquinone compound of formula I wherein Rl and R2 are Br with a mixture of an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' iε lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula:
Figure imgf000063_0002
wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
4. The method of claim 1 wherein the solvent is an aprotic solvent selected from the group consisting of acetonitrile, dimethyl formamide (DMF) , tetrahydrofuran (THF) and mixtures thereof.
5. The method of claim 1 wherein the initial concentration of the 2,5-dibromo-l,4-benzoquinone in the solvent is at least about 1 Molar.
6. The method of claim 1 wherein the metal carbonate is selected from the group consisting of cesium carbonate potassium carbonate, sodium carbonate, lithium carbonate and mixtures thereof.
7. The method of claim 1 wherein the metal carbonate is cesium carbonate.
8. The method of claim 1 wherein about 1 equivalent of the 2 , 5-dibromo-l,4-benzoquinone is reacted with at least 2 equivalents of at least one indole of the formula III.
9. The method of claim 1 wherein the 2,5-dibromo-l, 4- benzoquinone and the indole are reacted at a temperature of from about -10°C to about 100°C.
10. A method for preparing an indolylquinone compound of the formula:
Figure imgf000064_0001
wherein:
Rl and R2 are each independently Br, Cl, F, I, H, OH or
—OCOR, wherein R is lower alkyl, aryl or alkylaryl;
R"l and R"2 are each independently H, C^C, alkyl, C^C, alkenyl, C1-C7 alkynyl, arylalkyl or aryl; and
R3 to R12 are each independently hydrogen, branched or unbranched C^^ alkyl, alkylcarboxy, C2-Cra alkenyl, alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalo ethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer from 0 to 12 and m iε an integer from 0 to 12, which comprises:
(a) reacting a substituted or unsubstituted 2,5- dibromo-1, 4-benzoquinone compound of the formula:
Figure imgf000065_0001
wherein Rl and R2 are as defined above;
with one equivalent of a first indole of the formula
Figure imgf000065_0002
wherein R"l and R3-R7 are as defined above; in a polar organic solvent in the presence of metal carbonate;
(b) reacting the intermediate product of step (a) with one equivalent of a second indole of the formula:
Figure imgf000066_0001
wherein R"2 and R8-R12 are as defined above; in a polar organic solvent in the presence of metal carbonate to produce said indolylquinone compound of formula VI.
11. The method of claim 7 further comprising reacting the indolylquinone compound of formula VI with an alkali metal hydroxide to produce a compound of the formula:
Figure imgf000066_0002
wherein R"l, R"2 and R3-R12 are as defined above.
12. The method of claim 10 which further comprises reacting the indolylquinone compound of formula VI wherein Rl and R2 are Br with a mixture of an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula:
Figure imgf000067_0001
wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
13. The method of claim 10 wherein the solvent is an aprotic solvent selected from the group consisting of acetonitrile, dimethyl formamide (DMF) , tetrahydrofuran (THF) and mixtures thereof.
14. The method of claim 10 wherein the initial concentration of the 2,5-dibromo-l,4-benzoquinone in the solvent is at least about 1 Molar.
15. The method of claim 10 wherein the metal carbonate is selected from the group consisting of cesium carbonate potassium carbonate, sodium carbonate, lithium carbonate and mixtures thereof.
16. The method of claim 10 wherein the metal carbonate is cesium carbonate.
17. The method of claim 10 wherein two different indoles of the formula III are used.
18. The method of claim 10 wherein about l equivalent of the 2,5-dibromo-l,4-benzoquinone iε reacted with at least 2 equivalents of at least one indole of the formula III.
19. The method of claim 10 wherein steps (a) and (b) are each independently carried out at a temperature of from about -10°C to about 100°C.
20. A method for preparing an indolylquinone compound of the formula:
Figure imgf000068_0001
wherein:
R"l is H, Ci-G- alkyl, C2-C7 alkenyl or C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C^Cn alkyl, alkylcarboxy, C2-Cm alkenyl, alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer of 0 to 12 and m is an integer of 0 to 12, which comprises
reacting 2, 3,5, 6-tetrabromo-l, 4-benzoquinone with at least one indole of the formula:
Figure imgf000069_0001
wherein R"l and R3-R7 are as defined above; in a polar organic solvent in the presence of metal carbonate to produce said indolylquinone compound of Formula X.
21. The method of claim 20 which further comprises reacting the indolylquinone compound of formula X with an alkali metal hydroxide to produce a compound of the formula:
Figure imgf000069_0002
wherein R3-R7 are as defined above.
22. The method of claim 20 which further comprises reacting the indolylquinone compound of formula X with a mixture of an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula:
Figure imgf000070_0001
wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
23. The method of claim 20 wherein the solvent is an aprotic solvent selected from the group consisting of acetonitrile, dimethyl formamide (DMF) , tetrahydrofuran (THF) and mixtures thereof.
24. The method of claim 20 wherein the initial concentration of the tetrabromo-1, 4-benzoquinone in the solvent is at least about 1 Molar.
25. The method of claim 20 wherein the metal carbonate is selected from the group consisting of cesium carbonate, potassium carbonate, sodium carbonate, lithium carbonate and mixtures thereof.
26. The method of claim 20 wherein the metal carbonate is cesium carbonate.
27. The method of claim 20 wherein two different indoles of the formula III are used.
28. The method of claim 20 wherein about 1 equivalent of the 2 ,5-dibromo-l, 4-benzoquinone is reacted with at least 2 equivalents of at least one indole of the formula III.
29. The method of claim 13 wherein the tetrabromo-1,4- quinone and the indole are reacted at a temperature of from about -10°C to about 100°C.
30. The method of claim 2, 3, 11, 12, 21, or 22 wherein the alkali metal hydroxide is selected from the group consisting of sodium hydroxide, potassium hydroxide, and mixtures thereof.
31. A method for preparing an indolylquinone compound, which comprises:
reacting a substituted or unsubstituted 2,5-dibromo-l,4- benzoquinone compound of the formula:
Figure imgf000071_0001
wherein Rl and R2 are each independently Br, Cl, F, I, H, OH or -OCOR, wherein R is lower alkyl, aryl or alkylaryl; with at least one indole of the formula
Figure imgf000072_0001
wherein R"l is H, C1-C7 alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3-R7 are each independently hydrogen, branched or unbranched Ci-C-, alkyl, alkylcarboxy, C2-Cm alkenyl, alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, a ido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer from 0 to 12 and is an integer from 0 to 12; in a polar organic solvent in the presence of metal carbonate.
32. A method for preparing an indolylquinone compound which comprises:
reacting 2,3,5, 6-tetrabromo-l,4-benzoquinone with at least one indole of the formula
Figure imgf000072_0002
wherein R"l is H, C^C, alkyl, C2-C7 alkenyl or C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C^Cn alkyl, alkylcarboxy, C2-Cm alkenyl, alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^Cn alkoxy, nitro, halo, trihalo ethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or.2- methylbut-2-en-4-yl, wherein n is an integer of 0 to 12 and is an integer of 0 to 12; in a polar organic solvent in the presence of metal carbonate.
33. A method for preparing an indolylquinone compound of the formula:
Figure imgf000073_0001
wherein:
Rl, R2 and R30 are each independently Br, Cl, F, I, H, OH or
-OCOR, wherein R is lower alkyl, aryl or alkylaryl;
R"l is H, Ci-C, alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C1-Cn alkyl, alkylcarboxy, C2-Cm alkenyl, alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer from 0 to 12 and m is an integer from 0 to 12. This method comprises reacting a substituted or unsubstituted 2,5-dibromo-l,4-benzoquinone compound of the formula:
wherein Rl and R2 are as defined above, with one indole of the formula
Figure imgf000074_0002
wherein R"l and R3-R7 are as defined above. The reaction is carried out in a polar organic solvent and in the presence of metal carbonate.
34. The method of claim 33 which further comprises reacting the indolylquinone compound of formula XI with an alkali metal hydroxide to produce a compound of the formula:
Figure imgf000075_0001
wherein R30 and R3-R7 are aε defined above.
35. The method of claim 33 which further comprise reacting the indolylquinone compound of formula XI wherein Rl, R2 and R30 are Br, F, Cl or I with a mixture of an alkali metal hydroxide and an alcohol of the formula R'OH, wherein R' is lower alkyl or alkylaryl, to produce an indolylquinone compound of the formula:
Figure imgf000075_0002
wherein R'l and R'2 are each independently lower alkyl, aryl or alkylaryl.
36. The method of claim 33 wherein the solvent is an aprotic solvent selected from the group consisting of acetonitrile, dimethyl formamide (DMF) , tetrahydrofuran (THF) and mixtures thereof.
37. The method of claim 33 wherein the initial concentration of the 2,5-dibromo-l,4-benzoquinone in the solvent is at least about 1 Molar.
38. The method of claim 33 wherein the metal carbonate is selected from the group consisting of cesium carbonate potassium carbonate, sodium carbonate, lithium carbonate and mixtures thereof.
39. The method of claim 33 wherein the metal carbonate is cesium carbonate.
40. The method of claim 33 wherein about 1 equivalent of the 2,5-dibromo-l,4-benzoquinone is reacted with at least about 1 equivalent of one indole of the formula III.
41. The method of claim 33 wherein the 2,5-dibromo-l,4- benzoquinone and the indole are reacted at a temperature of from about -10°C to about 100°C.
42. Synthetic and highly pure compounds of the formula:
Figure imgf000076_0001
wherein: Rl and R2 are each independently Br, Cl, F, I, H, OH or -OCOR, wherein R is, lower alkyl, aryl or alkylaryl;
R"l is H, Ci-C, alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C^Cn alkyl, alkylcarboxy, C2-Cm alkenyl, C2-Cm alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, which are of about 95% or greater purity and which are synthetically prepared.
43. Synthetic and highly pure compounds of the formula:
Figure imgf000077_0001
wherein:
Rl and R2 are each independently Br, Cl, F, I, H, OH, or -OCOR, wherein R is lower alkyl, aryl or alkylaryl;
R"l and R"2 are each independently H, C^C? alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and R3 to R12 are each independently hydrogen, branched or unbranched C^Cn alkyl, alkylcarboxy, C2-Cm alkenyl, C2-Cm alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, preferably 1-7, and m is an integer from 0 to 12, which are of about 95% or greater purity and which are synthetically prepared.
44. Synthetic and highly pure compounds of the formula:
Figure imgf000078_0001
wherein: Rl and R2 are each independently Br, Cl, F, I, H, OH or -OCOR, wherein R is, lower alkyl, aryl or alkylaryl;
R"l is H, Ci-C- alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C^C,, alkyl, alkylcarboxy, C2-Cm alkenyl, C2-Cm alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C., alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, which are synthetically prepared in an amount of about 1 or more grams.
45. Synthetic and highly pure compounds of the formula:
Figure imgf000079_0001
wherein:
Rl and R2 are each independently Br, Cl, F, I, H, OH, or —OCOR, wherein R is lower alkyl, aryl or alkylaryl;
R"l and R"2 are each independently H, C^C, alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R12 are each independently hydrogen, branched or unbranched Cx-Cn alkyl, alkylcarboxy, C2-Cm alkenyl, C2-Cra alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, Cj-Cn alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, preferably 1-7, and m is an integer from 0 to 12, which are synthetically prepared in an amount of about 1. or more grams.
46. Compounds of the formula:
Figure imgf000080_0001
wherein:
Rl, R2 and R30 are each individually Br, Cl, F, I, H, OH or —OCOR, wherein R is lower alkyl, aryl or alkylaryl;
R"l is H, C1-C7 alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C^C,, alkyl, alkylcarboxy, C2-Cm alkenyl, alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^Cn alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2- methylbut-2-en-4-yl, wherein n is an integer from 0 to 12 and m is an integer of 0 to 12.
47. A compound of claim 46 in high purity and high yield.
48. A pharmaceutical composition for administration to humans which comprises a compound of the formula:
Figure imgf000081_0001
wherein:
Rl and R2 are each independently Br, Cl, F, I, H, OH or -OCOR, wherein R is, lower alkyl, aryl or alkylaryl;
R"l is H, Ci-C, alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C1-Cn alkyl, alkylcarboxy, C2-Cra alkenyl, C2-Cm alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C,, alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, or a pharmaceutically acceptable salt thereof, and a pharmaceutical carrier.
49. A pharmaceutical composition suitable for administration to humans which comprises a compound of the formula:
Figure imgf000082_0001
wherein:
Rl and R2 are each independently Br, Cl, F, I, H, OH, or -OCOR, wherein R is lower alkyl, aryl or alkylaryl;
R"l and R"2 are each independently H, C^C, alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R12 are each independently hydrogen, branched or unbranched C^C,, alkyl, alkylcarboxy, C2-Cm alkenyl, C2-Cm alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C^C., alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, preferably 1-7, and m is an integer from 0 to 12, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
50. A pharmaceutical composition which comprises a compound of claim 46 and a pharmaceutical excipient.
51. A method of ameliorating symptoms of a cell proliferative disorder wherein the cell proliferative disorder involves a protein tyrosine kinase polypeptide/adaptor polypeptide complex with an amount of a compound sufficient to disrupt protein tyrosine kinase polypeptide/adaptor polypeptide complexes of the cell so that symptoms of the cell proliferative disorder are ameliorated; wherein said compound has either of the following formulas:
Figure imgf000083_0001
Figure imgf000083_0002
wherein:
Rl and R2 are each independently Br, Cl, F, I, H, OH or -OCOR, wherein R is, lower alkyl, aryl or alkylaryl;
R"l is H, C-L-C, alkyl, C2-C7 alkenyl, C2-C7 alkynyl, arylalkyl or aryl; and
R3 to R7 are each independently hydrogen, branched or unbranched C^C,, alkyl, alkylcarboxy, C2-Cm alkenyl, C2-C_. alkynyl, alkenylcarboxy, aryl, alkylaryl, hydroxy, hydroxyalkyl, C1-Cn alkoxy, nitro, halo, trihalomethyl, amido, carboxamido, carboxy, sulfonyl, sulfonamido, amino, mercapto, or 2-methylbut-2-en-4-yl, wherein n is an integer from 0 to 12, and m is an integer of 0 to 12.
52. The method of claim 51 wherein the cell proliferative disorder occurs in a mammal and the compound contacts the cell within a mammal so that the symptoms of the cell proliferative disorder in the mammal are ameliorated.
53. The method of claim 51 wherein the cell proliferative disorder is a BCR-ABL-associated cancer, a glioma, a glioblastoma, a melanoma, an ovarian cancer, a breast cancer, or a prostate cancer.
54. A method of ameliorating symptoms of a cell proliferative disorder wherein the cell proliferative disorder involves a protein tyrosine kinase polypeptide/adaptor polypeptide complex, comprising: contacting a cell capable of forming the protein tyrosine kinase polypeptide/adaptor polypeptide complex with an amount of the pharmaceutical composition of claim 48, 49 or 50 sufficient to disrupt protein tyrosine kinase polypeptide/adaptor polypeptide complexes of the cell so that symptoms of the cell proliferative disorder are ameliorated.
55. The compound 2 , 5-dihydroxy-3 , 6-di-[2-(3-methyl-n- butyl) indol-3-yl]-l,4-quinone.
56. Substantially pure 2 , 5-dihydroxy-3 , 6-di- [2- (3- methyl-n-butyl) indol-3-yl]-l,4-quinone.
PCT/US1997/020557 1996-11-13 1997-11-12 Synthetic methods for the preparation of indolylquinones and mono- and bis-indolylquinones prepared therefrom WO1998020874A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
DE69734246T DE69734246T2 (en) 1996-11-13 1997-11-12 SYSNTHESIS PROCESS FOR THE PREPARATION OF INDOLYLQUINONES AND MONO AND BIS INDOLYLQUINONE PRODUCED BY THE METHOD
EP97946641A EP0959881B1 (en) 1996-11-13 1997-11-12 Synthetic methods for the preparation of indolylquinones and mono- and bis-indolylquinones prepared therefrom
CA002271737A CA2271737A1 (en) 1996-11-13 1997-11-12 Synthetic methods for the preparation of indolylquinones and mono- and bis-indolylquinones prepared therefrom
JP52274698A JP2001504820A (en) 1996-11-13 1997-11-12 Synthetic method for producing indolylquinone and mono- and bis-indolylquinones produced therefrom
AU51770/98A AU5177098A (en) 1996-11-13 1997-11-12 Synthetic methods for the preparation of indolylquinones and mono- and bis-indolylquinones prepared therefrom
AT97946641T ATE305000T1 (en) 1996-11-13 1997-11-12 SYSNTHESIC PROCESS FOR PRODUCING INDOLYLQUINONES AND MONO- AND BIS-INDOLYLQUINONES PRODUCED BY THE PROCESS

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US3060496P 1996-11-13 1996-11-13
US60/030,604 1996-11-13
US4298997P 1997-04-14 1997-04-14
US60/042,989 1997-04-14
US08/964,791 1997-11-05
US08/964,791 US5786488A (en) 1996-11-13 1997-11-05 Synthetic methods for the preparation of indolyquinones

Publications (1)

Publication Number Publication Date
WO1998020874A1 true WO1998020874A1 (en) 1998-05-22

Family

ID=27363680

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/020557 WO1998020874A1 (en) 1996-11-13 1997-11-12 Synthetic methods for the preparation of indolylquinones and mono- and bis-indolylquinones prepared therefrom

Country Status (9)

Country Link
US (3) US5786488A (en)
EP (1) EP0959881B1 (en)
JP (1) JP2001504820A (en)
AT (1) ATE305000T1 (en)
AU (1) AU5177098A (en)
CA (1) CA2271737A1 (en)
DE (1) DE69734246T2 (en)
ES (1) ES2251035T3 (en)
WO (1) WO1998020874A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1067925A1 (en) * 1998-04-02 2001-01-17 Merck & Co., Inc. Antidiabetic agents
EP1080066A1 (en) * 1998-05-22 2001-03-07 Shionogi Bioresearch Corporation Bioreductive cytotoxic agents
WO2001021589A2 (en) * 1999-09-24 2001-03-29 Sugen, Inc. Mono- and bis-indolylquinones and prophylactic and therapeutic uses thereof

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5786488A (en) * 1996-11-13 1998-07-28 Sugen, Inc. Synthetic methods for the preparation of indolyquinones
US6147106A (en) * 1997-08-20 2000-11-14 Sugen, Inc. Indolinone combinatorial libraries and related products and methods for the treatment of disease
US6316429B1 (en) * 1997-05-07 2001-11-13 Sugen, Inc. Bicyclic protein kinase inhibitors
US6486185B1 (en) 1997-05-07 2002-11-26 Sugen, Inc. 3-heteroarylidene-2-indolinone protein kinase inhibitors
US6051597A (en) * 1997-06-13 2000-04-18 Merck & Co., Inc. Indolylquinones as antidiabetic agents
US6114371A (en) * 1997-06-20 2000-09-05 Sugen, Inc. 3-(cyclohexanoheteroarylidenyl)-2-indolinone protein tyrosine kinase inhibitors
EA005032B1 (en) 1998-05-29 2004-10-28 Сьюджен, Инк. Pyrrole substituted 2-indolinone (variants), pharmaceutical composition (variants), method for modulation of catalytic activity method for treating and preventing a protein kinase related disorders in an organism
US6689806B1 (en) 1999-03-24 2004-02-10 Sugen, Inc. Indolinone compounds as kinase inhibitors
US6878733B1 (en) 1999-11-24 2005-04-12 Sugen, Inc. Formulations for pharmaceutical agents ionizable as free acids or free bases
ES2290117T3 (en) * 2000-02-15 2008-02-16 Sugen, Inc. PROTEIN QUINASE 2-INDOLIN INHIBITORS REPLACED WITH PIRROL.
AR042586A1 (en) 2001-02-15 2005-06-29 Sugen Inc 3- (4-AMIDOPIRROL-2-ILMETILIDEN) -2-INDOLINONE AS INHIBITORS OF PROTEIN KINASE; YOUR PHARMACEUTICAL COMPOSITIONS; A METHOD FOR THE MODULATION OF THE CATALYTIC ACTIVITY OF PROTEINQUINASE; A METHOD TO TREAT OR PREVENT AN AFFECTION RELATED TO PROTEINQUINASE
BR0213185A (en) 2001-10-10 2004-09-14 Sugen Inc 3- [4- (Substituted Heterocyclyl) -pyrrol-2-ylmethylidene] 2-indolinone derivatives as kinase inhibitors
US7057052B2 (en) * 2002-09-26 2006-06-06 Duke University Heterocyclic quinones as pharmaceutical agents

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3917820A (en) * 1969-05-29 1975-11-04 Canadian Patents Dev Antibiotic cochliodinol and production by chaetomium, cochliodes and chaetomium globsum

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE20680T1 (en) * 1982-04-30 1986-07-15 Ici Plc DELIGNIFICATION OF LIGNOCELLULOSIC MATERIAL.
JPS6360966A (en) * 1986-08-29 1988-03-17 Kyoto Yakuhin Kogyo Kk Quinone derivative
JPH11503519A (en) 1995-03-31 1999-03-26 メルク エンド カンパニー インコーポレーテッド Insulin mimetic and potentiator assays
IL122427A0 (en) * 1995-06-07 1998-06-15 Sugen Inc Pharmaceutical compositions and methods for inhibition of adaptor protein/tyrosine kinase interactions
US5786488A (en) * 1996-11-13 1998-07-28 Sugen, Inc. Synthetic methods for the preparation of indolyquinones
US6376529B1 (en) * 1995-06-07 2002-04-23 Peng Cho Tang Mono- and bis-indolylquinones and prophylactic and therapeutic uses thereof
US6051597A (en) * 1997-06-13 2000-04-18 Merck & Co., Inc. Indolylquinones as antidiabetic agents
EP1067925A4 (en) 1998-04-02 2003-04-16 Merck & Co Inc Antidiabetic agents
US20030073837A1 (en) * 1998-12-31 2003-04-17 Langecker Peter J. 3-heteroarylidenyl-2-indolinone compounds for modulating protein kinase activity and for use in cancer chemotherapy

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3917820A (en) * 1969-05-29 1975-11-04 Canadian Patents Dev Antibiotic cochliodinol and production by chaetomium, cochliodes and chaetomium globsum

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Chemical Abstracts Service (C A S); 1 January 1982 (1982-01-01), SHIMIZU SAKAE, ET AL: "ANTITUMOR EFFECT AND STRUCTURE-ACTIVITY RELATIONSHIP OF ASTERRIQUINONE ANALOGS", XP002945179, Database accession no. 97-174470W *
Chemical Abstracts Service (C A S); 1 January 1982 (1982-01-01), SHIMIZU SAKAE, YAMAMOTO YUZURI, KOSHIMURA SABURO: "ANTITUMOR ACTIVITY OF ASTERRIQUINONES FROM ASPERGILLUS FUNGI. IV. AN ATTEMPT TO MODIFY THE STRUCTURE OF ASTERRIQUINONES TO INCREASE THE ACTIVITY", XP002945178, Database accession no. 97-103877Z *
Chemical Abstracts Service (C A S); 1 January 1982 (1982-01-01), YOUNG T E, BABBITT B W: "2-(2-METHYL-3-INDOLYL)-1,4-BENZOQUINONE, A REVERSIBLE REDOX SUBSTRATE AT THE CARBON-PASTE-ELECTRODE IN ACIDIC AQUEOUS-ETHANOLIC MEDIA", XP002945169, Database accession no. 96-151191W *
Chemical Abstracts Service (C A S); 1 January 1988 (1988-01-01), YAMAMOTO YUZURU, ET AL: "PREPARATION OF INDOLYLBENZOQUINONES AS ANTICANCER AGENTS", XP002945157, Database accession no. 109-210891M *
HOERCHER U, SCHWENNER E, FRANCK B: "TOTALSYNTHESE DES COCHLIODINOLS TOTAL SYNTHESIS OF COCHLIODINOL", LIEBIGS ANNALEN DER CHEMIE., VERLAG CHEMIE GMBH. WEINHEIM., DE, 1 January 1986 (1986-01-01), DE, pages 1765 - 1771, XP002945156, ISSN: 0170-2041 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6376529B1 (en) 1995-06-07 2002-04-23 Peng Cho Tang Mono- and bis-indolylquinones and prophylactic and therapeutic uses thereof
US6660763B2 (en) 1996-11-13 2003-12-09 Sugen, Inc. Bis-indolylquinone compounds
EP1067925A1 (en) * 1998-04-02 2001-01-17 Merck & Co., Inc. Antidiabetic agents
EP1067925A4 (en) * 1998-04-02 2003-04-16 Merck & Co Inc Antidiabetic agents
EP1080066A1 (en) * 1998-05-22 2001-03-07 Shionogi Bioresearch Corporation Bioreductive cytotoxic agents
EP1080066A4 (en) * 1998-05-22 2004-12-22 Shionogi Biores Corp Bioreductive cytotoxic agents
WO2001021589A2 (en) * 1999-09-24 2001-03-29 Sugen, Inc. Mono- and bis-indolylquinones and prophylactic and therapeutic uses thereof
WO2001021589A3 (en) * 1999-09-24 2002-01-17 Sugen Inc Mono- and bis-indolylquinones and prophylactic and therapeutic uses thereof

Also Published As

Publication number Publication date
US20030060635A1 (en) 2003-03-27
AU5177098A (en) 1998-06-03
CA2271737A1 (en) 1998-05-22
JP2001504820A (en) 2001-04-10
ATE305000T1 (en) 2005-10-15
US6750240B2 (en) 2004-06-15
EP0959881A1 (en) 1999-12-01
EP0959881A4 (en) 2001-06-27
US6110957A (en) 2000-08-29
ES2251035T3 (en) 2006-04-16
US5786488A (en) 1998-07-28
EP0959881B1 (en) 2005-09-21
DE69734246T2 (en) 2006-06-22
DE69734246D1 (en) 2006-02-02

Similar Documents

Publication Publication Date Title
US6750240B2 (en) Methods of using bis-indolylquinones
RU2072989C1 (en) Derivatives of methylenehydroxyindole, method of their synthesis and pharmaceutical composition
ES2300132T3 (en) DERIVATIVES OF ARYLENE AND HETEROARYLIDEN-OXINDOLS AS INHIBITORS OF THYROSINE KINASE.
US6660763B2 (en) Bis-indolylquinone compounds
JPH0329069B2 (en)
HUT74875A (en) Substituted 3-ariliden-7-aza-oxindol compounds pharmaceutical compositions containing the same and process for producing them
EP1778616A1 (en) Alpha-ketoglutarates and their use as therapeutic agents
US6599938B1 (en) Compounds having MIF antagonist activity
HUT73811A (en) Substituted beta-aryl and beta-heteroaryl-alpha-cyanoacrylamide derivatives as tyrosine kinase inhibitors, pharmaceutical compns. contg. them and process to prepare them
WO2003082875A2 (en) Novel podophyllotoxin derivatives, the production thereof and the use of the same in therapeutics
EP0853614B1 (en) Substituted tetralylmethylen-oxindole analogues as tyrosine kinase inhibitors
WO1997046551A1 (en) Substituted quinolylmethylen-oxindole analogues as tyrosine kinase inhibitors
EP1911744B1 (en) Tryptophan derivative and use thereof
Oatis Jr et al. Synthesis of 4'-hydroxypropranolol sulfate, a major non-. beta.-blocking propranolol metabolite in man
JPH08506357A (en) Tryptoamine analogs having 5-HT1D selectivity
US4423073A (en) Fluorinated diaminopentene derivatives
EP0072762B1 (en) Fluorinated diaminoalkene derivatives
JP2006347940A (en) beta-AMYLOID FORMATION INHIBITOR
NZ201591A (en) Fluorinated alkane diamine derivatives
GB2104887A (en) Decarboxylase-inhibiting acetylenic diaminobutane derivatives
JP2004529110A (en) Indole derivatives with vascular damage activity

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CU CZ EE GE GH HU ID IL IS JP KG KP KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK SL TJ TM TR TT UA UZ VN YU AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH KE LS MW SD SZ UG ZW AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2271737

Country of ref document: CA

ENP Entry into the national phase

Ref country code: JP

Ref document number: 1998 522746

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1997946641

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1997946641

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1997946641

Country of ref document: EP