EP1109796A1 - Inhibiteurs de telomerase et leurs procedes d'utilisation - Google Patents

Inhibiteurs de telomerase et leurs procedes d'utilisation

Info

Publication number
EP1109796A1
EP1109796A1 EP00950282A EP00950282A EP1109796A1 EP 1109796 A1 EP1109796 A1 EP 1109796A1 EP 00950282 A EP00950282 A EP 00950282A EP 00950282 A EP00950282 A EP 00950282A EP 1109796 A1 EP1109796 A1 EP 1109796A1
Authority
EP
European Patent Office
Prior art keywords
compound
mmol
composition
thiazolidine
dione
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP00950282A
Other languages
German (de)
English (en)
Inventor
Allison C. Chin
Ryan Holcomb
Mieczyslaw A. Piatyszek
Upinder Singh
Richard L. Tolman
Tsutomu Akama
Yutaka Kanda
Akira Asai
Yoshinori Yamashita
Kaori Endo
Hiroyuki Yamaguchi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
KH Neochem Co Ltd
Geron Corp
Original Assignee
Kyowa Hakko Kogyo Co Ltd
Geron Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from JP30757699A external-priority patent/JP2001072592A/ja
Application filed by Kyowa Hakko Kogyo Co Ltd, Geron Corp filed Critical Kyowa Hakko Kogyo Co Ltd
Publication of EP1109796A1 publication Critical patent/EP1109796A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/34Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Definitions

  • telomeric repeat sequences possibly due to crosslinking of the telomeric repeat sequences, is also known (Burger et al., (1997) Eur. J. Cancer 33: 638-644).
  • Thiazolidinediones comprise a group of structurally related antidiabetic compounds that increases the insulin sensitivity of target tissues (skeletal muscle, liver, adipose) in insulin resistant animals. In addition to these effects on hyperglycemia, thiazolidinediones also reduce lipid and insulin levels in animal models of NIDDM. Recently, the thiazolidinedione troglitazone was shown to have these same beneficial effects in human patients suffering from impaired glucose tolerance, a metabolic condition that precedes the development of NIDDM, as in patients suffering from NIDDM (Nolan et al., (1994) N. Eng. J. Med. 331, 1188-1193).
  • thiazolidinediones have been implicated in appetite regulation disorders (see WO 94/25026 Al), and in increase of bone marrow fat content.
  • thiazolidinedione compounds have been suggested for use in the treatment of psoriasis (U.S. Patent No.
  • telomere activity provides important benefits to efforts at treating human disease.
  • Compounds that inhibit telomerase activity can be used to treat telomerase-mediated disorders, such as cancer, since cancer cells express telomerase activity and normal human somatic cells do not possess telomerase activity at biologically relevant levels (i.e., at levels sufficient to maintain telomere length over many cell divisions).
  • telomerase activity can be used to treat telomerase-mediated disorders, such as cancer, since cancer cells express telomerase activity and normal human somatic cells do not possess telomerase activity at biologically relevant levels (i.e., at levels sufficient to maintain telomere length over many cell divisions).
  • Unfortunately few such compounds, especially compounds with high potency or activity and compounds that are orally bioavailable, have been identified and characterized.
  • compounds that act as telomerase inhibitors that have relatively high potency or activity and that are orally bioavailable, and for compositions and methods for treating cancer and other diseases in which telomerase activity is present abnormally.
  • the present invention provides methods, compounds and compositions that are specific and effective for treating telomerase-mediated disorders, such as malignant conditions by targeting cells having telomerase activity.
  • the methods, compounds, and compositions of the invention can be applied to a wide variety of malignant cell types and avoid the problems inherent in current cancer treatment modalities which are non-specific and excessively toxic.
  • the present invention is based on the finding that certain known thiazolidinone compounds, as well as new thiazolidinone derivatives disclosed herein, are effective in the inhibition of telomerase enzyme activity, in vitro, ex vivo and in vivo.
  • the present invention provides methods of inhibiting telomerase by contacting telomerase with the compounds described herein.
  • the telomerase to be inhibited is a mammalian telomerase, such as a human telomerase.
  • a related aspect of the present invention is the discovery that thiazolidinone compounds inhibit the proliferation of cells that have telomerase activitiy, such as many cancer cells.
  • this aspect of the present invention provides methods of inhibiting telomerase activity in a patient, preferably a mammal, suffering from a telomerase-mediated condition or disease, comprising administering to the patient a therapeutically effective amount of a telomerase inhibiting thiazolidinone compound, or a pharmaceutically acceptable salt thereof.
  • the present invention provides methods, compounds and compositions for inhibiting a telomerase enzyme, comprising contacting the telomerase enzyme with a composition or a compound of formula (I):
  • X is oxygen or sulfur, ⁇ is a single or double bond;
  • A is aryl or heteroaryl;
  • Ri is hydrogen or lower alkyl;
  • R 2 , R 3 and R are independently selected from the group consisting of hydrogen, halo, alkyl, aryl, hydroxyl, alkoxyl, aryloxyl, aralkoxyl, cyano, nitro, alkylcarbamido, arylcarbamido, dialkylcarbamido, diarylcarbamido, alkylarylcarbamido, alkylthiocarbamido, arylthiocarbamido, dialkylthiocarbamido, diarylthiocarbamido, alkylarylthiocarbamido, amino, alkylamino, arylamino, dialkylamino, diarylamino, arylalkylamino, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, dialky
  • the present invention provides methods, compounds and compositions for inhibiting a telomerase enzyme, comprising contacting the telomerase enzyme with a composition or a compound of formula (V):
  • the new compounds of the invention have many valuable uses as inhibitors of deleterious telomerase activity, such as, for example, in the treatment of cancer in mammals, such as humans.
  • the pharmaceutical compositions of this invention can be employed in treatment regimens in which cancer cells are killed, in vivo, or can be used to kill cancer cells ex vivo.
  • this invention provides therapeutic compounds and compositions for treating cancer, and methods for treating cancer and other telomerase-mediated conditions or diseases in humans and other mammals (e.g., cows, horses, sheep, steer, pigs and animals of veterinary interest such as cats and dogs).
  • thiazolidinone or "thiazolidinone derivative” as used herein refers to compounds of the general formula:
  • X is O or S.
  • the derivatives are thiazolidinedione derivatives.
  • the derivatives are the thiazolidinonethione derivatives also known as rhodanines (see Examples 25-28, below).
  • alkyl refers to a straight, branched, or cyclic hydrocarbon chain fragment or radical containing between about one and about twenty carbon atoms, more preferably between about one and about ten carbon atoms (e.g., methyl, ethyl, n-propyl, iso- propyl, cyclopropyl, n-butyl, iso-butyl, tert-butyl, cyclobutyl, adamantyl, noradamantyl and the like).
  • Straight, branched, or cyclic hydrocarbon chains having eight or fewer carbon atoms will also be referred to herein as "lower alkyl".
  • the hydrocarbon chains may further include one or more degrees of unsaturation, i.e., one or more double or triple bonds (e.g., vinyl, propargyl, allyl, 2-buten-l-yl, 2-cyclopenten-l-yl, 1,3-cyclohexadien-l-yl, 3-cyclohexen-l-yl and the like).
  • Alkyl groups containing double bonds such as just described will also be referred to herein as "alkenes”.
  • alkyl groups having triple bonds will also be referred to herein as "alkynes”.
  • alkynes alkynes
  • alkyl as used herein further includes one or more substitutions at one or more carbon atoms of the hydrocarbon fragment or radical.
  • substitutions include, but are not limited to: aryl; heterocycle; halogen (to form, e.g., trifluoromethyl, ⁇ CF 3 ); nitro ( ⁇ NO 2 ); cyano (--CN); hydroxyl (also referred to herein as "hydroxy"), alkoxyl (also referred herein as alkoxy) or aryloxyl (also referred to herein as "aryloxy")(— OR); thio or mercapto, alkyl- or arylthio (--SR); amino, alkylamino, arylamino, dialkyl- or diarylamino, or arylalkylamino (— NRR'); aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, dialkylaminocarbonyl, diarylaminocarbonyl or aryl
  • heterocyclic groups i.e., heterocycle, heteroaryl, and heteroaralkyl
  • heterocycleoxy refers to the group -OR, where R is heterocycle as defined below.
  • alkyl moiety of "lower alkanoyl”, “lower alkoxy”, “lower alkanoyloxy”, “lower alkylthio”, is the same as “alkyl” defined above.
  • methylene refers to the group — CH 2 ⁇ .
  • metal refers to a methylene group for which one hydrogen atom has been replaced by a substituent as described above.
  • halo or halogen as used herein refers to the substituents fluoro, bromo, chloro, and iodo.
  • carbonyl refers to the functional group ⁇ C(O) ⁇ .
  • thiocarbonyl — C(S) ⁇
  • sulfinyl ⁇ S(O) ⁇
  • sulfonyl ⁇ SO 2 ⁇
  • phosphonyl --PO 2 --
  • aryl refers to cyclic aromatic hydrocarbon chains having twenty or fewer carbon atoms, e.g., phenyl, naphthyl, biphenyl and anthracenyl.
  • One or more carbon atoms of the aryl group may also be substituted with, e.g.: alkyl; aryl; heterocycle; formyl; halogen; nitro; cyano; hydroxyl, alkoxyl or aryloxyl; thio or mercapto, alkyl-, or arylthio; amino, alkylamino, arylamino, dialkyl-, diaryl-, or arylalkylamino; aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, dialkylaminocarbonyl, diarylaminocarbonyl or arylalkylaminocarbonyl; carboxyl, or alkyl- or aryloxycarbonyl; carboxaldeh
  • heterocyclic groups e.g., heterocycleoxy, heteroaryloxy, and heteroaralkylthio
  • heterocycleoxy, heteroaryloxy, and heteroaralkylthio are defined by analogy to the above- described terms.
  • aralkyl refers to an aryl group that is joined to a parent structure by an alkyl group as described above, e.g., benzyl, ⁇ -methylbenzyl, phenethyl, and the like.
  • alkyl group e.g., benzyl, ⁇ -methylbenzyl, phenethyl, and the like.
  • the aralkyl moiety of "aralkylsulfonyl" aralkyloxy is the same as “aralkyl” defined above.
  • heterocycle refers to a cyclic alkyl group or aryl group as defined above in which one or more carbon atoms have been replaced by a non-carbon atom, especially nitrogen, oxygen, or sulfur.
  • Non-aromatic heterocycles will also be referred to herein as "cyclic heteroalkyl”.
  • Aromatic heterocycles are also referred to herein as "heteroaryl".
  • such groups include furyl, tetrahydrofuryl, pyrrolyl, pyrrolidinyl, thienyl, tetrahydrothienyl, oxazolyl, isoxazolyl, triazolyl, thiazolyl, isothiazolyl, pyrazolyl, pyrazolidinyl, oxadiazolyl, thiadiazolyl, imidazolyl, imidazolinyl, pyridyl, pyridazinyl, triazinyl, piperidinyl, morpholinyl, thiomorpholinyl, pyrazinyl, piperazinyl, pyrimidinyl, naphthyridinyl, benzofuranyl, benzothienyl, indolyl, indolinyl, indolizinyl, indazolyl, quino
  • heteroarylalkyl The heteroaryl moiety of "heteroarylalkyl”, “heteroarylalkenyl”, “heteroarylalkynyl”, “heteroarylsulfonyl”, “heteroarylalkylsulfonyl”, “heteroarylalkenylsulfonyl", “heteroarylalkynylsulfonyl", “heteroaryloxy”, “heteroarylalkyloxy” is the same as “heteroaryl” defined above.
  • heterocyclic groups may further include one or more substituents at one or more carbon and/or non-carbon atoms of the heteroaryl group, e.g.: alkyl; aryl; heterocycle; halogen; nitro; cyano; hydroxyl, alkoxyl or aryloxyl; thio or mercapto, alkyl- or arylthio; amino, alkyl-, aryl-, dialkyl- diaryl-, or arylalkylamino; aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, dialkylaminocarbonyl, diarylaminocarbonyl or arylalkylaminocarbonyl;
  • heterocyclealkyl refers to a heterocycle group that is joined to a parent structure by one or more alkyl groups as described above, e.g., 2-piperidylmethyl, and the like.
  • heteroarylkyl refers to a heteroaryl group that is joined to a parent structure by one or more alkyl groups as described above, e.g., 2-thienylmethyl, and the like.
  • the compounds of the present invention may be used to inhibit or reduce telomerase enzyme activity and/or proliferation of cells having telomerase activity.
  • inhibition and reduction of the enzyme or cell proliferation refers to a lower level of the measured activity relative to a control experiment in which the enzyme or cells are not treated with the test compound.
  • the inhibition or reduction in the measured activity is at least a 10% reduction or inhibition.
  • reduction or inhibition of the measured activity of at least 20%, 50%, 75%, 90% or 100% may be preferred for particular applications.
  • telomere activity As noted above, the immortalization of cells involves inter alia the activation of telomerase. More specifically, the connection between telomerase activity and the ability of many tumor cell lines, including skin, connective tissue, adipose, breast, lung, stomach, pancreas, ovary, cervix, uterus, kidney, bladder, colon, prostate, central nervous system (CNS), retina and blood tumor cell lines, to remain immortal has been demonstrated by analysis of telomerase activity (Kim et al.). This analysis, supplemented by data that indicates that the shortening of telomere length can provide the signal for replicative senescence in normal cells (see WO 93/23572), demonstrates that inhibition of telomerase activity can be an effective anti-
  • inhibitor is simply meant a reagent, drug or chemical which is able to decrease the activity of the telomerase enzyme in vitro or in vivo.
  • Such inhibitors can be readily identified using standard screening protocols in which a cellular extract or other preparation having telomerase activity is placed in contact with a potential inhibitor, and the level of telomerase activity measured in the presence or absence of the inhibitor, or in the presence of varying amounts of inhibitor. In this way, not only can useful inhibitors be identified, but the optimum level of such an inhibitor can be determined in vitro for further testing in vivo.
  • the invention proves a method for inhibiting the ability of a cell to proliferate or replicate.
  • one or more of the thiazolidinone compounds of the invention that are capable of inhibiting telomerase enzyme activity, are provided during cell replication.
  • telomeres play a critical role in allowing the end of the linear chromosomal DNA to be replicated completely without the loss of terminal bases at the 5'-end of each strand.
  • Immortal cells and rapidly proliferating cells use telomerase to add telomeric DNA repeats to chromosomal ends. Inhibition of telomerase will result in the proliferating cells not being able to add telomeres and they will eventually stop dividing.
  • this method for inhibiting the ability of a cell to proliferate is useful for the treatment of a condition associated with an increased rate of proliferation of a cell, such as in cancer (telomerase-activity in malignant cells), and hematopoiesis (telomerase activity in hematopoietic stem cells), for example.
  • the present invention provides compositions and compounds for the prevention or treatment of many types of malignancies.
  • the compounds of the present invention can provide a highly general method of treating many, if not most, malignancies, as demonstrated by the highly varied human tumor cell lines and tumors having telomerase activity.
  • the thiazolidinone compounds of the present invention can be effective in providing treatments that discriminate between malignant and normal cells to a high degree, avoiding many of the deleterious side-effects present with most current chemotherapeutic regimes which rely on agents that kill dividing cells indiscriminately.
  • thiazolidinedione compounds include the glitazones, such as, for example, troglitazone (also known as CS-045 (Sankyo) and CI-991 (Park-Davis)), pioglitazone
  • rosiglitazone also known as BRL49653
  • englitazone also known as CP-68,722
  • ciglitazone also known as CP-68,72
  • the present invention provides new compounds, pharmaceutical compositions and methods relating to the new compounds, or their pharmaceutically acceptable salts, for inhibiting a telomerase enzyme, comprising contacting the telomerase enzyme with a compound, or its pharmaceutically acceptable salt, having the formula (I):
  • X oxygen or sulfur
  • is a single or double bond
  • A is aryl or heteroaryl
  • R ⁇ is hydrogen or lower alkyl
  • R 2 , R 3 and j are independently selected from the group consisting of hydrogen, halo, alkyl, aryl, hydroxyl, alkoxyl, aryloxyl, aralkoxyl, cyano, nitro, alkylcarbamido, arylcarbamido, dialkylcarbamido, diarylcarbamido, alkylarylcarbamido, alkylthiocarbamido, arylthiocarbamido, dialkylthiocarbamido, diarylthiocarbamido, alkylarylthiocarbamido, amino, alkylamino, arylamino, dialkylamino, diarylamino, arylalkylamino, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, dialkylamino
  • the new compounds of the present invention have the general structure shown as formula II below:
  • A may be aryl to form, for example, a phenyl moiety.
  • A may be heteroaryl, such as, for example, pyridine, quinoline, isoquinoline, thiophene, furan, imidazole, benzimidazole, pyrazole, and the like.
  • A is phenyl, as in formula (II).
  • when n is 1, Rj can not be hydrogen.
  • at least one of R2 and R 3 is other than hydrogen.
  • at least one of R 2 and R 3 is halo, and most preferably both R2 and R 3 are halo to form a dihalo-substituted phenyl moiety.
  • L may be a direct bond, or may be a 1 to 3 atom linking group wherein the atoms of the linking group independently selected from unsubstituted or substituted carbon, N, O or S.
  • Representative linking groups useful in the compounds of the invention include, for example -O-, -S-, -NH-, -CH 2 -, -OCH 2 -, -OC(O)-, -C0 2 -, -NHC(O)-, -C(O)NH-, -OC(O)CH 2 -, -OC(O)NH-, and -NHC(O)NH-.
  • representative compounds include, without limitation, 5-(2- (3,4-Dichlorophenyl)benzylidene)thiazolidine-2,4-dione, 5-(3-(3,4-Dichlorophenyl)- benzylidene)thiazolidine-2,4-dione, 5-(4-(3,4-Dichlorobenzyloxy)benzylidene)thiazolidine-2,4- dione, 5-(2-(3,4-Dichlorobenzyloxy)benzylidene)thiazolidine-2,4-dione, 5-(4-(3,4-
  • the new compounds of the present invention have the general structure shown as formula III below:
  • the present invention provides methods, compounds and compositions for inhibiting a telomerase enzyme, comprising contacting the telomerase enzyme with a compound, or its pharmaceutically acceptable salt, having the formula (IV):
  • Compounds of formula (IV) having the double bond can be obtained by reacting a thiazolidine derivative with an aromatic carbonyl compound. The reaction can be carried out optionally in the presence of a base catalyst and optionally in a solvent.
  • the base catalyst may be piperidine, piperidinium acetate, diethylamine, pyridine, sodium acetate, potassium carbonate, sodium carbonate, and the like.
  • the solvent may be an alcohol, such as methanol, ethanol, propanol, or the like, an ether, such as diethyl ether, tetrahydrofuran, dioxane, or the like, or a hydrocarbon, such as benzene, roluene, xylene, or the like, and mixtures thereof.
  • the reaction is carried out at a temperature of about room temperature to about 200°C, preferably about 50-100°C, and completes in about one hour to about 50 hours.
  • the present invention provides methods, compounds and compositions for inhibiting a telomerase enzyme, comprising contacting the telomerase enzyme with a compound, or its pharmaceutically acceptable salt, having the formula (V):
  • the compounds of the present invention can be synthesized using techniques and materials known to those of skill in the art, such as described, for example, in March,
  • the procedures described herein for synthesizing the compounds of the invention may include one or more steps of protection and deprotection (e.g., the formation and removal of acetal groups).
  • the synthetic procedures disclosed below can include various purifications, such as column chromatography, flash chromatography, thin-layer chromatography (TLC), recrystallization, distillation, high-pressure liquid chromatography (HPLC) and the like.
  • various techniques well known in the chemical arts for the identification and quantification of chemical reaction products such as proton and carbon- 13
  • the compounds of formula IV, where L is SO or SO2 can be synthesised by oxidizing the corresponsing S compound in an inert solvent.
  • the inert solvent may be dichloromethane, methanol, tetrahydrofuran, ether, hexane, toluene, cyclohexane, or the like, and mixtures thereof.
  • the oxidizing agent may be m- chloroperbenzoic acid, hydrogen peroxide, or the like.
  • the reaction is carried out at a temperature in the range of about -78 °C to the boiling point of the solvent, preferably from about 0 °C to about 30 °C for about 0.5 to about 12 hours.
  • the compounds of the present invention demonstrate inhibitory activity against telomerase activity in vivo, as has been and can be demonstrated as described below.
  • the in vitro activities of the compounds of the invention can also be demonstrated using the methods described herein.
  • the term "ex vivo" refers to tests performed using living cells in tissue culture.
  • One method used to identify compounds of the invention that inhibit telomerase activity involves placing cells, tissues, or preferably a cellular extract or other preparation containing telomerase in contact with several known concentrations of a test compound in a buffer compatible with telomerase activity. The level of telomerase activity for each concentration of test compound is measured and the ICs 0 (the concentration of the test compound at which the observed activity for a sample preparation was observed to fall one-half of its original or a control value) for the compound is determined using standard techniques. Other methods for
  • IC 5 0 values for several of the compounds of the present invention were determined, and found to be below 100 ⁇ M.
  • compounds of the present invention are expected to induce crisis in telomerase-positive cell lines.
  • Treatment of telomerase-positive cell lines, such as HEK-293 and HeLa cells, with a compound of the invention is also expected to induce a reduction of telomere length in the treated cells.
  • Compounds of the invention are also expected to induce telomere reduction during cell division in human tumor cell lines, such as the ovarian tumor cell lines OVCAR-5 and SK-OV- 3.
  • telomere length is expected to be no different from cells treated with a control substance, e.g., dimethyl sulfoxide (DMSO).
  • DMSO dimethyl sulfoxide
  • the compounds of the invention also are expected to demonstrate no significant cytotoxic effects at concentrations below about 5 ⁇ M in the normal cells.
  • the specificity of the compounds of the present invention for telomerase can be determined by comparing their activity (IC 5 o) with respect to telomerase to other enzymes having similar nucleic acid binding or modifying activity similar to telomerase in vitro.
  • enzymes include DNA Polymerase I, HeLa RNA Polymerase II, T3 RNA Polymerase, MMLV Reverse Transcriptase, Topoisomerase I, Topoisomerase II, Terminal Transferase and Single- Stranded DNA Binding Protein (SSB).
  • SSB Single- Stranded DNA Binding Protein
  • mice treated with a compound of the invention are expected to have tumor masses that, on average, may increase for a period following the initial dosing, but will begin to shrink in mass with continuing treatment.
  • mice treated with a control e.g., DMSO are expected to have tumor masses that continue to increase.
  • TRF terminal restriction fragment
  • telomere length By measuring the length of telomeric DNA, one can estimate how long a telomerase inhibitor should be administered and whether other methods of therapy (e.g., surgery, chemotherapy and/or radiation) should also be employed. In addition, during treatment, one can test cells to determine whether a decrease in telomere length over progressive cell divisions is occurring to demonstrate treatment efficacy.
  • other methods of therapy e.g., surgery, chemotherapy and/or radiation
  • the present invention also provides pharmaceutical compositions for inhibiting cell proliferation of telomerase positive cells, and treating cancer and other conditions in which inhibition of telomerase is an effective therapy.
  • compositions include a therapeutically effective amount of a telomerase inhibiting compound of the invention in a pharmaceutically acceptable carrier or salt.
  • the present invention provides methods, compounds and compositions for inhibiting a telomerase enzyme, inhibiting proliferation of telomerase postive cells, and for treating cancer in a mammal.
  • the compositions of the invention include a therapeutically effective amount of a compound of formulas I to V (or a pharmaceutically acceptable salt thereof) in a pharmaceutically acceptable carrier.
  • the compounds and compositions of the present invention may also be used for the treatment of other telomerase mediated conditions or diseases, such as, for example, other hyperproliferative or autoimmune disorders such as psoriasis, rheumatoid arthritis, immune system disorders requiring immune system suppression, immune system reactions to poison ivy or poison oak, and the like.
  • telomerase inhibitor of the invention with other anti-cancer agents, including other inhibitors of telomerase such as described in U.S. Patent Nos. 5,656,638, 5,760,062, 5,767,278, 5,770,613 and 5,863,936.
  • Other inhibitors of telomerase such as described in U.S. Patent Nos. 5,656,638, 5,760,062, 5,767,278, 5,770,613 and 5,863,936.
  • the choice of such combinations will depend on various factors including, but not limited to, the type of disease, the age and general health of the patient, the aggressiveness of disease progression, the TRF length and telomerase activity of the diseased cells to be treated and the ability of the patient to tolerate the agents that comprise the combination.
  • telomerase inhibiting compound of the invention in cases where tumor progression has reached an advanced state, it may be advisable to combine a telomerase inhibiting compound of the invention with other agents and therapeutic regimens that are effective at reducing tumor size (e.g. radiation, surgery, chemotherapy and/or hormonal treatments).
  • a telomerase inhibiting agent of the invention with one or more agents that treat the side effects of a disease, e.g., an analgesic, or agents effective to stimulate the patient's own immune response (e.g., colony stimulating factor).
  • a pharmaceutical formulation comprises a telomerase inhibitor of the invention with an anti-angiogenesis agent, such as fumagillin, fumagillin derivatives, or AGM-1470.
  • telomerase inhibitor of the invention in addition to one or more antineoplastic agents or adjuncts (e.g., folinic acid or mesna).
  • Antineoplastic agents suitable for combination with the compounds of the present invention include, but are not limited to, alkylating agents including alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines, such as a benzodizepa, carboquone, meturedepa and uredepa; ethylenimines and methylmelamines such as altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylolmelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cyclophosphamide, estramustine, iphosphamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichine, phenesterine, prednimustine, trofosfamide, and uracil mustard; nitroso ureas, such as carmustine, chlorozotocin, fotemustine
  • ranimustine Additional agents include dacarbazine, mannomustine, mitobronitol, mitolactol and pipobroman. Still other classes of relevant agents include antibiotics, hormonal antineoplastics and antimetabolites. Yet other combinations will be apparent to those of skill in the art.
  • Additional agents suitable for combination with the compounds of the present invention include protein synthesis inhibitors such as abrin, aurintricarboxylic acid, chloramphenicol, colicin E3, cycloheximide, diphtheria toxin, edeine A, emetine, erythromycin, ethionine, fluoride, 5-fluorotryptophan, ftisidic acid, guanylyl methylene diphosphonate and guanylyl imidodiphosphate, kanamycin, kasugamycin, kirromycin, and O-methyl threonine.
  • protein synthesis inhibitors such as abrin, aurintricarboxylic acid, chloramphenicol, colicin E3, cycloheximide, diphtheria toxin, edeine A, emetine, erythromycin, ethionine, fluoride, 5-fluorotryptophan, ftisidic acid, gu
  • Additional protein synthesis inhibitors include modeccin, neomycin, norvaline, pactamycin, paromomycine, puromycin, ricin, _-sarcin, shiga toxin, showdomycin, sparsomycin, spectinomycin, streptomycin, tetracycline, thiostrepton and trimethoprim.
  • Inhibitors of DNA synthesis including alkylating agents such as dimethyl sulfate, mitomycin C, nitrogen and sulfur mustards, MNNG and NMS; intercalating agents such as acridine dyes, actinomycins, adriamycin, anthracenes, benzopyrene, ethidium bromide, propidium diiodide-intertwining, and agents such as distamycin and netropsin, can also be combined with compounds of the present invention in pharmaceutical compositions.
  • alkylating agents such as dimethyl sulfate, mitomycin C, nitrogen and sulfur mustards, MNNG and NMS
  • intercalating agents such as acridine dyes, actinomycins, adriamycin, anthracenes, benzopyrene, ethidium bromide, propidium diiodide-intertwining, and agents such as distamycin and netropsin
  • DNA base analogs such as acyclovir, adenine ⁇ -1- D-arabinoside, amethopterin, aminopterin, 2-aminopurine, aphidicolin, 8-azaguanine, azaserine, 6-azauracil, 2'-azido-2'-deoxynucleosides, 5-bromodeoxycytidine, cytosine ⁇ -l-D- arabinoside, diazooxynorleucine, dideoxynucleosides, 5-fluorodeoxycytidine,
  • 5-fluorodeoxyuridine, 5-fluorouracil, hydroxyurea and 6-mercaptopurine also can be used in combination therapies with the compounds of the invention.
  • Topoisomerase inhibitors such as coumermycin, nalidixic acid, novobiocin and oxolinic acid, inhibitors of cell division, including colcemide, colchicine, vinblastine and vincristine; and RNA synthesis inhibitors including actinomycin D, ⁇ -amanitine and other fungal amatoxins, cordycepin (3'- deoxyadenosine), dichlororibofuranosyl benzimidazole, rifampicine, streptovaricin and streptolydigin also can be combined with the compounds of the invention to provide pharmaceutical compositions.
  • the present invention includes compounds and compositions in which a telomerase inhibitor is either combined with or covalently bound to a cytotoxic agent
  • a targeting agent such as a monoclonal antibody (e.g., a murine or humanized monoclonal antibody).
  • a targeting agent such as a monoclonal antibody (e.g., a murine or humanized monoclonal antibody).
  • a monoclonal antibody e.g., a murine or humanized monoclonal antibody
  • the telomerase inhibitors of the invention may also be combined with monoclonal antibodies that have therapeutic activity against cancer.
  • telomerase inhibitors of the present invention can be applied to agricultural phytopathogenic organisms that are characterized by telomerase activity. These organisms include nematodes such as
  • telomere-inhibiting compounds of the invention can be administered to plants and soil infected with phytopathogenic organisms having telomerase activity alone, or in combination with other telomerase-inhibiting agents and/or other agents used to control plant diseases.
  • the determination of the compositions used to control such phytopathogenic organisms and the appropriate modes of delivering such compositions will be known to those having skill in the agricultural arts.
  • telomerase inhibitors provided by the present invention can be used to treat nematode infections in humans and animals of veterinary interest such as dogs and cats. Nematode infection in humans and animals often is in the form of hookworm or roundworm infection and leads to a host of deadly secondary illnesses such as meningitis, myocarditis, and various neurological diseases.
  • administration of the telomerase-inhibiting compounds such as those of the invention, alone, or in combination with other telomerase-inhibiting agents and/or other therapeutic agents, can be used to control nematode, protozoan and fungal infections in humans and animals.
  • a suitable effective dose of a compound of the invention will be in the range of 0.001 to 1000 milligram (mg) per kilogram (kg) of body weight of the recipient per day, preferably in the range of 0.001 to 100 mg per kg of body weight per day, more preferably between about 0.1 and 100 mg per kg of body weight per day and still more preferably in the range of between 0.1 to 10 mg per kg of body weight per day.
  • the desired dosage is preferably presented in one, two, three, four, or more subdoses administered at appropriate intervals throughout the day, or by the action of a continuous pump. These subdoses can be administered as unit dosage form, for example, containing 5 to 10,000 mg, preferably 10 to 1000 mg of active ingredient per unit dosage from.
  • the dosage is presented once per day at a dosing at least equal to TID, or is administered using a continuous pump delivery system.
  • compositions used in these therapies can be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as tablets, pills, powders, liquid solutions or suspensions, liposomes, and injectable and infusible solutions. The preferred form depends on the intended mode of administration and therapeutic application.
  • the compositions also preferably include conventional pharmaceutically acceptable carriers and adjuvants, as is well known to those of skill in the art. See, e.g., REMINGTON'S PHARMACEUTICAL SCIENCES, Mack Publishing Co.: Easton, Pa., 17th Ed. (1985).
  • administration will be by oral or parenteral (including subcutaneous, intramuscular, intravenous, and intradermal) routes. More preferably, the route of administration will be oral.
  • the therapeutic methods and agents of this invention can of course be used concomitantly or in combination with other methods and agents for treating a particular disease or disease condition.
  • telomerase activity-inhibiting compound of this invention in a therapeutically or pharmaceutically effective dose together with one or more pharmaceutically or therapeutically acceptable carriers and optionally other therapeutic ingredients.
  • Various considerations for preparing such formulations are described, e.g., in Gilman et al. (eds.) GOODMAN AND GILMAN'S: THE PHARMACOLOGICAL BASES OF THERAPEUTICS, 8th Ed., Pergamon Press (1990); and
  • compositions for administration are discussed therein, e.g., for oral, intravenous, intraperitoneal, intramuscular, and other forms of administration.
  • methods for administering pharmaceutical compositions will be either topical, parenteral, or oral administration methods for prophylactic and/or therapeutic treatment.
  • Oral administration is preferred.
  • the pharmaceutical compositions can be administered in a variety of unit dosage forms depending upon the method of administration. As noted above, unit dosage forms suitable for oral administration include powders, tablets, pills, and capsules.
  • the skin sites include anatomic regions for transdermally administering the drug, such as the forearm, abdomen, chest, back, buttock, and mastoidal area.
  • the compound is administered to the skin by placing on the skin either a topical formulation comprising the compound or a transdermal drug delivery device that administers the compound.
  • the delivery vehicle is designed, shaped, sized, and adapted for easy placement and comfortable retention on the skin.
  • transdermal drug delivery devices can be employed with the compounds of this invention.
  • a simple adhesive patch comprising a backing material and an acrylate adhesive can be prepared.
  • the drug and any penetration enhancer can be formulated into the adhesive casting solution.
  • the adhesive casting solution can be cast directly onto the backing material or can be applied to the skin to form an adherent coating. See, e.g., U.S. Pat. Nos. 4,310,509; 4,560,555; and 4,542,012.
  • the compound of the invention will be delivered using a liquid reservoir system drug delivery device.
  • a liquid reservoir system drug delivery device typically comprise a backing material, a membrane, an acrylate based adhesive, and a release liner.
  • the membrane is sealed to the backing to form a reservoir.
  • the drug or compound and any vehicles, enhancers, stabilizers, gelling agents, and the like are then incorporated into the reservoir. See, e.g., U.S. Pat. Nos. 4,597,961; 4,485,097; 4,608,249; 4,505,891; 3,843,480; 3,948,254; 3,948,262; 3,053,255; and 3,993,073.
  • Matrix patches comprising a backing, a drug/penetration enhancer matrix, a membrane, and an adhesive can also be employed to deliver a compound of the invention transdermally.
  • the matrix material typically will comprise a polyurethane foam.
  • the drug, any enhancers, vehicles, stabilizers, and the like are combined with the foam precursors.
  • the foam is allowed to cure to produce a tacky, elastomeric matrix which can be directly affixed to the backing material. See, e.g., U.S. Pat. Nos. 4,542,013; 4,460,562; 4,466,953; 4,482,534; and 4,533,540.
  • preparations for topical application to the skin comprising a compound of the invention, typically in concentrations in the range from about 0.001% to 10%, together with a non-toxic, pharmaceutically acceptable topical carrier.
  • Topical preparations can be prepared by combining an active ingredient according to this invention with conventional pharmaceutical diluents and carriers commonly used in topical dry, liquid, and cream formulations.
  • Ointment and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • bases may include water and/or an oil, such as liquid paraffin or a vegetable oil, such as peanut oil or castor oil.
  • Thickening agents that may be used according to the nature of the base include soft paraffin, aluminum stearate, cetostearyl alcohol, propylene glycol, polyethylene glycols, woolfat, hydrogenated lanolin, beeswax, and the like.
  • Lotions may be formulated with an aqueous or oily base and will, in general, also include one or more of the following: stabilizing agents, emulsifying agents, dispersing agents, suspending agents, thickening agents, coloring agents, perfumes, and the like. Powders may be formed with the aid of any suitable powder base, e.g., talc, lactose, starch, and the like. Drops may be formulated with an aqueous base or non-aqueous base also comprising one or more dispersing agents, suspending agents, solubilizing agents, and the like. Topical administration of compounds of the invention may also be preferred for treating diseases such as skin cancer and fungal infections of the skin (pathogenic fungi typically express telomerase activity).
  • the topical pharmaceutical compositions according to this invention may also include one or more preservatives or bacteriostatic agents, e.g., methyl hydroxybenzoate, propyl hydroxybenzoate, chlorocreosol, benzalkonium chlorides, and the like.
  • the topical pharmaceutical compositions also can contain other active ingredients such as antimicrobial agents, particularly antibiotics, anesthetics, analgesics, and antipruritic agents.
  • the compounds of the present invention can also be delivered through mucosal membranes.
  • Transmucosal (i.e., sublingual, buccal, and vaginal) drug delivery provides for an
  • Transmucosal drug dosage forms e.g., tablet, suppository, ointment, pessary, membrane, and powder
  • Transmucosal drug dosage forms are typically held in contact with the mucosal membrane and disintegrate and/or dissolve rapidly to allow immediate systemic absorption.
  • certain such routes may be used even where the patient is unable to ingest a treatment composition orally.
  • delivery of a telomerase inhibitor of the invention would be enhanced, one can select a composition for delivery to a mucosal membrane, e.g., in cases of colon cancer one can use a suppository to deliver the telomerase inhibitor.
  • an oral formulation such as a lozenge, tablet, or capsule
  • the method of manufacture of these formulations is known in the art, including, but not limited to, the addition of the pharmacological agent to a pre-manufactured tablet; cold compression of an inert filler, a binder, and either a pharmacological agent or a substance containing the agent (as described in U.S. Pat. No. 4,806,356); and encapsulation.
  • Another oral formulation is one that can be applied with an adhesive, such as the cellulose derivative hydroxypropyl cellulose, to the oral mucosa, for example as described in U.S. Pat. No. 4,940,587.
  • compositions for intravenous administration that comprise a solution of a compound of the invention dissolved or suspended in an acceptable carrier.
  • injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Suitable excipients are, for example, water, buffered water, saline, dextrose, glycerol, ethanol, or the like.
  • compositions will be sterilized by conventional, well known sterilization techniques, such as sterile filtration.
  • sterilization techniques such as sterile filtration.
  • the resulting solutions can be packaged for use as is or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.
  • the pharmaceutical compositions to be administered may also contain minor amounts of non-toxic auxiliary substances, such as
  • Another method of parenteral administration employs the implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained. See, e.g., U.S. Pat. No. 3,710,795.
  • Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc., an active compound as defined above and optional pharmaceutical adjuvants in an excipient, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, olive oil, and other lipophilic solvents, and the like, to form a solution or suspension.
  • an excipient such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, olive oil, and other lipophilic solvents, and the like
  • the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, etc.
  • wetting or emulsifying agents such as wetting or emulsifying agents, pH buffer
  • composition or formulation to be administered will contain an effective amount of an active compound of the invention.
  • nontoxic solid carriers can be used and include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally 0.1-95%) of active ingredient, preferably about 20%.
  • compositions containing the compounds of the invention can be administered for prophylactic and/or therapeutic treatments.
  • compositions are administered to a patient already suffering from a disease, as described above, in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications.
  • An amount adequate to accomplish this is defined as a "therapeutically effective amount or dose.” Amounts effective for this use will depend on the severity of the disease and the weight and general state of the patient.
  • the compounds and compositions of the invention may be applied ex vivo to achieve therapeutic effects, as for example, in the case of a patient suffering from leukemia.
  • cells to be treated e.g., blood or bone marrow cells
  • the cells are returned to the patient following treatment.
  • Such a procedure can allow for exposure of cells to concentrations of therapeutic agent for longer periods or at higher concentrations than otherwise available.
  • a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, can be reduced, as a function of the systems, to a level at which the improved condition is retained. When the symptoms have been alleviated to the desired level, treatment can cease. Patients can, however, require additional treatment upon any recurrence of the disease symptoms.
  • compositions containing the compounds of the invention are administered to a patient susceptible to or otherwise at risk of a particular disease.
  • a patient susceptible to or otherwise at risk of a particular disease is defined to be a "prophylactically effective amount or dose.”
  • prophylactically effective amount or dose the precise amounts again depend on the patient's state of health and weight.
  • the present invention provides valuable reagents relating to human and mammalian telomerase.
  • the above description of necessity provides a limited and merely illustrative sampling of specific compounds, and should not be construed as limiting the scope of the invention.
  • Other features and advantages of the invention will be apparent from the following examples and claims.
  • the acidified solution was extracted with chloroform or ethyl acetate, organic phase washed with water, dried over Na 2 SO 4 and concentrated under reduced pressure to yield crude product as a solid that was purified either by column chromatography or recrystallization from appropriate solvent system. Reactions were generally run on a 0.5 mmolar scale.
  • the requisite aldehyde was prepared from 3-bromobenzaldehyde using the procedure of Example 1, Step A.
  • Step A Preparation of aldehyde To a solution of 4-hydroxybenzaldehyde in acetonitrile was added K 2 CO 3 (1.5 eq.) followed by addition of 3,4-dichlorobenzylchloride (3 eq.). The resulting reaction mixture was heated to 90 °C for 2-16 h at which time the precipitate was filtered off. The filtrate was diluted with EtOAc, washed with water, dried over Na 2 SO 4 and concentrated under reduced pressure to provide crude product. This product was purified by recrystallization from CH 2 Cl 2 /hexane solvent system to yield pure aldehyde.
  • 5-(2-carboxybenzylidene)thiazolidine-2,4-dione was prepared by the method of General Procedure 1 from 2-carboxybenzaldehyde.
  • the 5-(2-carboxybenzylidene)thiazolidine-2,4-dione was dissolved in SO 2 Cl 2 followed by addition of 1-2 drops of DMF. This resulting mixture was heated to -80 °C for 15-30 min at which time the reaction was concentrated under reduced pressure. The residue was dissolved in THF and added drop wise to a solution of 3,4-dichloroaniline (1.5 eq) and TEA (2 eq). This mixture was stirred for additional 1-2 h at which time the solid in the reaction mixture was filtered. The filtrate was concentrated under reduced pressure to yield solid that was washed with water and ether to afford pure product.
  • 5-(4-hydroxybenzylidene)thiazolidine-2,4-dione was prepared by the method of General Procedure 1 from 4-hydroxybenzaldehyde.
  • Step A Preparation of 5-(2-hydroxybenzylidene)thiazolidine-2.4-dione 5-(2-Hydroxybenzylidene)thiazolidine-2,4-dione was prepared by General Procedure 1 from 2-hydroxybenzaldehyde.
  • Step B Elaboration of hydroxy group 3,4-Dichlorophenylacetic acid was dissolved in SO2CI2 followed by addition of a few drops of DMF. This resulting mixture was heated to 80 °C for 15-30 min. followed by concentration of the reaction mixture under reduced pressure. The residue was dissolved in THF and slowly added to a solution of 5-(hydroxybenzylidene)thiazolidine-2,4-dione (1.5 eq.)
  • Step A Preparation of 5-(2-hydroxybenzylidene)thiazolidine-2.4-dione 5-(2-Hydroxybenzylidene)thiazolidine-2,4-dione was prepared by General Procedure 1 from 2-hydroxybenzaldehyde.
  • Step B Acylation of the hydroxy group
  • Step A Preparation of aldehyde A mixture of 2-fluorobenzaldehyde (248.23 mg 210 ⁇ L, 2 mmol) and 3,4- dichlorophenol was stirred with potassium carbonate in 5 mL of dimethylacetamide at 90°C for 12 hours. The reaction was diluted with 20 mL of water and extracted with 25 mL of ethyl acetate. The organic layer was washed with saturated aqueous sodium bicarbonate and saturated sodium chloride solutions then dried over sodium sulfate and concentrated in vacuo to give a brown oil that was taken on without further purification.
  • Reactions were generally run on a 0.1 mmolar scale.
  • Step A Preparation of aldehyde.
  • Step A Preparation of aldehyde.
  • Step A Preparation of aldehyde.
  • Step A Preparation of aldehyde To a solution of 3 -hydroxy benzaldehyde in acetonitrile was added K 2 CO 3 (1.5 eq.) followed by addition of 3,4-dichlorobenzylchloride (3 eq.). The resulting reaction mixture was heated to 90°C for 2-16 h at which time the precipitate was filtered off. The filtrate was diluted with EtOAc, washed with water, dried over Na2SO 4 and concentrated under reduced pressure to provide crude product. This product was purified by recystallization from CH 2 Cl 2 /hexane solvent system to yield pure aldehyde. Step B. General Procedure 1.
  • Example 31 Preparation of Compound 2
  • Compound 1 200 mg, 0.538 mmol
  • acetone 30 mL
  • titanium trichloride 20%> aqueous solution, 4 mL
  • To the reaction solution was added an aqueous saturated sodium bicarbonate solution, and the mixture was extracted twice with ethyl acetate.
  • the organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate.
  • the solvent was evaporated under reduced pressure, and the residue was recrystallized from ethyl acetate/hexane to give Compound 2 (69 mg, 38%).
  • Example 33 Preparation of Compound 4 Under ice-cooling, Compound 1 (100 mg, 0.269 mmol) was dissolved in a mixed solvent of dichloromethane (20 mL) and methanol (4 mL), and the solution was mixed with m- chloroperbenzoic acid (50%) purity, 100 mg, 0.289 mmol), followed by stirring at room temperature for 3 hours.
  • the reaction solution was mixed with a 10% aqueous sodium hydrogen sulfite solution and extracted with chloroform.
  • the organic layer was washed with an aqueous saturated sodium bicarbonate solution and brine, and then dried over anhydrous sodium sulfate.
  • the solvent was evaporated under reduced pressure, and the residue was triturated with ethyl acetate to give Compound 4 (86 mg, 82%).
  • Example 36 Preparation of Compound 7 Under ice-cooling, Compound 6 (20 mg, 0.057 mmol) was suspended in dichloromethane (5 mL), and the suspension was mixed with m-chloroperbenzoic acid (50% purity, 22 mg, 0.063 mmol), followed by stirring for 20 minutes. To the reaction solution was added a 10% aqueous sodium hydrogen sulfite solution, and the mixture was extracted with chloroformmethanol (9:1). The organic layer was washed with an aqueous sodium bicarbonate solution, water and brine, and then dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the residue was triturated with diisopropyl ether to give Compound 7 (15 mg, 72%).
  • Example 37 Preparation of Compound 8 Under ice-cooling, Compound 6 (20 mg, 0.057 mmol) was suspended in dichloromethane (5 mL), and the suspension was mixed with m-chloroperbenzoic acid (50% purity, 200 mg, 0.57 mmol), followed by stirring for 3 hours. To the reaction solution was added a 10% aqueous sodium hydrogen sulfite solution, and the mixture was extracted with chloroformmethanol (9:1). The organic layer was washed with an aqueous saturated sodium bicarbonate solution, water and brine, and then dried over anhydrous sodium sulfate. The
  • Example 38 Preparation of Compound 9 4-(4-Methylphenylthio)-3-nitrobenzaldehyde (273mg, 1.00 mmol), 2,4-thiazolidinedione (176 mg, 1.50 mmol) and piperidine (0.40 mL, 0.40 mmol) were heated under reflux for 19 hours in ethanol (8 mL). The reaction solution was cooled down to room temperature, and the thus precipitated crystals were collected by filtration to give Compound 9 (175 mg, 47%).
  • Example 44 Preparation of Compound 15
  • 4-fluorobenzaldehyde (0.53 mL, 5.0 mmol) and p-cresol (648 mg, 6.0 mmol) were dissolved in dimethylacetamide (8 mL), and to the solution were added potassium carbonate (828 mg, 6.0 mmol) and cupric oxide (95 mg, 0.50 mmol) and the mixture was heated under reflux for 1.5 hours.
  • the reaction solution was cooled down to room temperature, and water was added, and then the mixture was extracted with ethyl acetate.
  • the organic layer was washed with a 0.5 N aqueous sodium hydroxide solution, water and brine, and then dried over anhydrous sodium sulfate.
  • 5-bromosalicylaldehyde (1.00 g, 5.00 mmol) was dissolved in dimethylformamide (10 mL), and to the solution were added 4-methylbenzyl bromide (925 mg, 5.00 mmol) and sodium hydride (220 mg, 5.50 mmol), followed by stirring at 70°C for 0.5 hour.
  • the reaction solution was cooled with ice, and water was added, and then the mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure.
  • 2,4-Thiazolidinedione (702 mg, 6.00 mmol), piperidine (0.50 mL, 5.0 mmol) and ethanol (40 mL) were added thereto, followed by heating under reflux for 4 hours.
  • the reaction solution was cooled down to room temperature, and water and 1 N HCl (5 mL) were added, and then the mixture was extracted with ethyl acetate. The organic layer was washed with water and
  • Example 55 Preparation of Compound 26 4-Diphenylaminobenzaldehyde (273 mg, 1.00 mmol), 2,4-thiazolidinedione (176 mg, 1.50 mmol) and piperidine (0.10 mL, 1.0 mmol) were heated under reflux for 4 hours in ethanol (8 mL). The reaction solution was cooled down to room temperature, and water and 1 N HCl (1 mL) were added, and then the mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the residue was triturated with ethyl acetate to give Compound 26 (293 mg, 79%).
  • Example 58 Preparation of Compound 29 4-Phenylbenzaldehyde (182 mg, 1.00 mmol), 2,4-thiazolidinedione (176 mg, 1.5 mmol) and piperidine (0.10 mL, 1.0 mmol) were heated under reflux for 3 hours in ethanol (6 mL). The reaction solution was cooled down to room temperature, and water and 1 N HCl (1 mL) were added, and then the mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate. The solvent was
  • Example 61 Preparation of Compound 32
  • Compound 30 (115 mg, 0.39 mmol) was dissolved in dichloromethane (15 mL), and to the solution were added trifluoroacetic acid (0.30 mL, 3.9 mmol) and triethylsilane (0.81 mL, 5.1 mmol) and the mixture was heated under reflux for 12 hours. The solvent was evaporated under reduced pressure, and the residue was recrystallized from acetone/hexane to give Compound 32 (70 mg, 61%).
  • Example 63 Preparation of Compound 34
  • Compound 33 (219 mg, 0.566 mmol) was dissolved in dichloromethane (15 mL), and to the solution were added trifluoroacetic acid (0.385 mL, 0.50 mmol) and triethylsilane (0.80 mL, 0.50 mmol), followed by stirring at room temperature for 10 minutes. The solvent was evaporated under reduced pressure, and the residue was triturated with hexane to give Compound 34 (198 mg, 94%).
  • diphenylamine (338 mg, 2.00 mmol) and 4-bromobenzyl bromide (500 mg, 2.00 mmol) were dissolved in dimethylformamide (8 mL), and to the solution was added sodium hydride (88 mg, 2.2 mmol) under ice -cooling, followed by stirring at room temperature for 4 hours.
  • sodium hydride 88 mg, 2.2 mmol
  • To the reaction solution was added water, and the mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the residue was purified by silica gel column chromatography (14:1 hexane/acetone) to obtain N-(4-bromobenzyl)diphenylamine (478 mg, 71%).
  • Example 65 Preparation of Compound 36
  • 4 -bromoaniline (344 mg, 2.00 mmol) was dissolved in dimethylformamide (5 mL), and to the solution were added sodium hydride (200 mg, 5.00 mmol) and benzyl bromide (0.52 mL, 4.4 mmol) under ice-cooling, followed by stirring at room temperature for 11 hours.
  • sodium hydride 200 mg, 5.00 mmol
  • benzyl bromide 0.52 mL, 4.4 mmol
  • the organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate.
  • the solvent was evaporated under reduced pressure, and the residue was recrystallized from ethyl acetate/hexane to obtain 4-bromo-N,N-dibenzylaniline (442 mg, 63%).
  • 2,4-Thiazolidinedione (176 mg, 1.5 mmol) , piperidine (0.10 mL, 1.0 mmol) and ethanol (6 mL) were added thereto, followed by heating under reflux for 13 hours.
  • the reaction solution was cooled down to room temperature, and water and 1 N HCl (5 mL) were added, and then the mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate.
  • 2,4-thiazolidinedione (145 mg, 1,24 mmol), and piperidine (0.083 mL, 0.83 mmol) were heated under reflux for four hours in ethanol (8 mL).
  • the reaction liquid was cooled to room temperature, mixed with water and 1 mol/L HCl (1 mL), and was extracted with chloroform.
  • the organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate.
  • Example 75 Preparation of Compound 48 2-Thienyl boric acid (26 mg, 0.20 mmol) was used instead of phenyl boric acid to obtain Compound 48 (7.5 mg, 18%>) from Compound 25 (40 mg, 0.10 mmol), using the same method as Example 74.
  • Example 79 Preparation of Compound 52 Under ice-cooling, Compound 43 (50 mg, 0.12. mmol) was dissolved in a mixed solvent of dichloromethane (8 mL) and methanol (2 mL). m-Chloroperbanzoic acid (50% purity, 398 mg, 1.2 mmol) was added thereto, followed by stirring at room temperature for 17 hours. To the reaction liquid a 5% sodium hydrogen sulfite aqueous solution was added and the product was extracted with chloroform. The organic layer was washed with sodium hydrogencarbonate aqueous solution and brine, and then dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and triturated with methanol to give Compound 52 (23 mg, 41%>).
  • Example 81 Preparation of Compound 54
  • Compound 53 (80 mg, 0.20 mmol) was dissolved in dimethylformamide (3 mL). l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (77 mg, 0.40 mmol) and diethylamine (0.041 mL, 0.40 mmol) were added thereto, followed by stirring at room temperature for 3 hours. Water was added to the reaction liquid and the product was extracted with ethyl acetate. The organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the residue was purified by preparative thin layer chromatography (9:1 chloroform/methanol) and was triturated with hexane to give Compound 54 (22 mg, 24%).
  • Example 82 Preparation of Compound 55 Aniline (0.037 mL, 0.40 mmol) was used instead of diethylamine to obtain Compound 55 (37 mg, 39%), using the same method as Example 81.
  • Example 83 Preparation of Compound 56 1 -Methylpiperazine (0.044 mL, 0.40 mmol) was used instead of diethylamine to obtain Compound 56 (11 mg, 12%>), using the same method as Example 81. Compound 56 was obtained in the form of hydrochloride.
  • Example 84 Preparation of Compound 57 Morpholine (0.035 mL, 0.40 mmol) was used instead of diethylamine to obtain Compound 57 (22 mg, 24%), using the same method as Example 81.
  • Example 86 Preparation of Compound 59
  • Compound 59 (260 mg, 86%) was obtained from commercially available 3-phenoxythiophene-2-carboxaldehyde (204 mg, 1.00 mmol) (MAYBRIDGE, Catalog Number: KM05428), 2,4-thiazolidinedione (176 mg, 1.5 mmol), and piperidine (0.099 mL, 1.0 mmol), using the same method as Example 70.
  • the organic layer was washed with water and brine and then dried over anhydrous sodium sulfate, and the solvent was evaporated under reduced pressure.
  • the residue was dissolved in tetrahydrofuran (6 mL) and 1 mol/L hydrochloric acid was added thereto, followed by stirring at room temperature for 1 hour.
  • Sodium hydrogencarbonate aqueous solution was added to the reaction liquid and the product was extracted with ethyl acetate.
  • the organic layer was washed with water and brine, and then dried over anhydrous sodium sulfate.
  • Example 89 Preparation of Compound 62
  • Compound 62 (113 mg, 62%) was obtained from 5-(4-chlorophenylthio)thiophene- 2-carboxyaldehyde (132 mg, 0.520 mmol) obtained in Example 87, 2,4-thiazolidinedione (73 mg, 0.62 mmol), and piperidine (0.052 mL, 0.52 mmol), using the same method as Example 87.
  • Compound 65 (211 mg, 88%) was obtained from 3-(4-chlorobenzoyl)-2- (4-chlorophenylthio)thiophen-5-carboxyaldehyde (193 mg, 0.490 mmol), 2,4-thiazolidinedione (69 mg, 0.59 mmol), and piperidine (0.049 mL, 0.49 mmol), using the same method as Example 70.
  • Example 93 Preparation of Compound 66
  • Compound 66 14 mg, 28%) was obtained from Compound 65 (50 mg, 0.10 mmol) and m-chloroperbenzoic acid (50% purity, 53 mg, 0.15 mol), using the same method as Example 78.
  • Example 95 Preparation of Compound 68
  • Compound 68 (0.0137 g, 65.7%>) was obtained by using Compound 67 (0.02 g, 0.051 mmol), using the same method as Example 78.
  • Example 99 Preparation of Compound 72 2-[(3,4-Dichlorophenyl)thio]-5-nitrobenzaldehyde (0.16 g, 79%) was obtained from 3,4-dichlorobenzenethiol (0.12 g, 0.68 mmol), a 2.5 mol/L sodium hydroxide aqueous solution (1.2 mL, 2.9 mmol), tetrabutylammonium bromide (0.011 g, 0.034 mmol) and a toluene (1.2 mL) solution of 2-fluoro-5-nitrobenzaldehyde (0.12 g, 0.68 mmol) using the same method as Example 97.
  • Example 100 Preparation of Compound 73 2- [(4-Bromophenyl)thio]- 5-nitrobenzaldehyde (0.28 g, 82%) was obtained from 4-bromobenzenethiol (0.19 g, 1.0 mmol), a 2.5 mol/L sodium hydroxide aqueous solution (1.7 mL, 4.3 mmol), tetrabutylammonium bromide (0.016 g, 0.051 mmol) and a toluene (1.7 mL) solution of 2-fluoro-5-nitrobenzaldehyde (0.17 g, 1.0 mmol) using the same method as Example 97.
  • Example 101 Preparation of Compound 74 2-[(4-Methoxyphenyl)thio]-5-nitrobenzaldehyde (0.22 g, 76%) was obtained from 4-methoxybenzenethiol (0.14 g, 0.99 mmol), a 2.5 mol/L sodium hydroxide aqueous solution (1.8 mL, 4.4 mmol), tetrabutylammonium bromide (0.016 g, 0.050 mmol) and a toluene (1.8 mL) solution of 2-fluoro-5-nitrobenzaldehyde (0.17 g. 0.99 mmol) using the same method as Example 98.
  • Compound 75 (0.14 g, 51%>) was obtained from 2-[(4-ethylphenyl)thio]-5- nitrobenzaldehyde (0.20 g, 0.69 mmol), toluene (9.9 mL), 2,4-thiazolidinedione (0.32 g, 2.7 mmol), piperidine (0.027 mL, 0.27 mmol), acetic acid (0.016 mL, 0.27 mmol) and a molecular sieve 4A (0.99 g) using the same method as Example 97.
  • Example 104 Preparation of Compound 77
  • Compound 77 (0.0055 g, 17.6%) was obtained by using Compound 76 (0.03 g, 0.081 mmol) using the same method as Example 96.
  • Example 105 Preparation of Compound 78
  • Compound 78 (0.0192 g, 59.0%) was obtained by using Compound 76 (0.03 g, 0.081 mmol) using the same method as Example 96.
  • Example 106 Preparation of Compound 79 2-[(4-Chlorobenzyl)thio]-5-nitrobenzaldehyde (0.17 g, 67%) was obtained from 4-chlorobenzylthiol (0.11 g, 0.83 mmol), a 2.5 mol/L sodium hydroxide aqueous solution (1.4 mL, 3.5 mmol), tetrabutylammonium bromide (0.013 g, 0.041 mmol) and a toluene (1.4 mL) solution of 2-fluoro-5-nitrobenzaldehyde (0.14 g, 0.83 mmol) using the same method as Example 97.
  • Compound 79 (0.11 g, 70%) was obtained from
  • Example 109 Preparation of Compound 82
  • Compound 82 (0.0195 g, 62.0%) was obtained by using Compound 9 (0.03 g, 0.081 mmol) using the same method as Example 96.
  • Example 111 Preparation of Compound 84
  • Compound 84 (0.152 g, 41.8%) was obtained by using commercially available (MAYBRIDGE, Catalog Number: XAX00135) 2-(cyclohexylthio)-5-nitrobenzaldehyde (0.26 g, 1.0 mmol) using the same method as Example 94.
  • Example 112 Preparation of Compound 85
  • Compound 85 (0.0149 g, 71.3%) was obtained by using Compound 84 (0.02 g, 0.055 mmol) using the same method as Example 96.
  • Example 116 Preparation of Compound 89
  • the reaction was performed in accordance with a document (Tetrahedron Lett. Vol. 36, No. 50, pp. 9085-9088, 1995) and the product was treated by using 5-bromo-2-[(4- methylphenyl)thio]-benzaldehyde (0.06 g, 0.2 mmol) and 2-pyridyl trifluoromethanesulfonate (0.03 g, 0.2 mmol).
  • Tri(dibenzylideneacetone)-dipalladium (0.18 g, 0.2 mmol), and triphenylphosphine (0.21 g, 0.8 mmol) were dissolved in tetrahydrofuran (60 mL), followed by stirring at room temperature for 30 minutes. Then, 5-bromo-2-fluorobenzaldehyde (0.4 g, 2.0 mmol) and 2-(tributylstannyl)-furan (1.25 mL, 4. 0 mmol) were added thereto, followed by heating under reflux for 10 hours.
  • Example 118 Preparation of Compound 91
  • Compound 91 (0.069 g, 66.5%) was obtained by using compound 74 (0.1 g, 0.25 mmol) using the same method as Example 96.
  • Example 120 Preparation of Compound 93
  • Compound 93 (0.051 g, 49.4%) was obtained by using Compound 92 (0.1 g, 0.24 - mmol) using the same method as Example 96.
  • Example 123 Preparation of Compound 96 3-Bromo-4-[(4-methylphenyl)thio]benzaldehyde (0.18 g, 59.0%) was obtained by using 3-bromo-4-fuluorobenzaldehyde (0.12 g, 1.0 mmol) using the same method an Example 114 (synthesis of 5-bromo-2-[(4-methylphenyl)thio]benzaldehyde).
  • Compound 96 (0.067 g, 70.4%) was obtained by using 5-hydroxymethyl-2-[(4-methyl- phenyl)thio]benzaldehyde (0.1 g, 0.27 mmol) using the same method as Example 94.
  • Example 126 Preparation of Compound 98 2-[(4-Methylphenyl)thio]-6-(trifluoromethyl)benzaldehyde (0.44 g, 100%) was obtained by using 2-fluoro-6-(trifluoromethyl)benzaldehyde (0.19 g, 1.5 mmol) using the same method as Example 114 (synthesis of 5-bromo-2-[(4-methylphenyl)thio]benzaldehyde).
  • Example 127 Preparation of Compound 100 2-[(4-Methylphenyl)thio]-5-(trifluoromethyl)benzaldehyde (0.45 g, 86.4%>) was obtained by using 2-fluoro-5(trifluoromethyl)benzaldehyde (0.1 g , 0.5 mmol) using the same method as Example 114 (synthesis of 5-bromo-2-[(4-methylphenyl)thio]benzaldehyde).
  • Compound 100 (0.068 g, 50.9%) was obtained by using 2-[(4-methylphenyl)thio]- 5-(trifluoromethyl)benzaldehyde (0.1 g, 0.34 mmol) using the same method as Example 94.
  • Example 131 Preparation of Compound 104
  • 2-fluoro-5-methoxybenzaldehyde 0.25 g, 2.0 mmol
  • Example 133 Preparation of Compound 106 5-Chloro-2-fluorobenzaldehyde (0.55 g, 77.6%) was obtained by using 4-chlorofluorobenzene (0.48 mL, 4.5 mmol) using the same method an Example 121 (synthesis of 2-fluoro-5-cyanobenzaldehyde).
  • Compound 106 (0.096 g,70.1%) was obtained by using 5-chloro-2-[(4-methylphenyl)- thiojbenzaldehyde (0.1 g, 0.38 mmol) using the same method as Example 94.
  • Compound 107 (0.094 g, 90.0%) was obtained by using Compound 106 (0.1 g, 0.28 mmol) using the same method as Example 96.
  • Compound 108 (0.052 g, 75.1%) was obtained by using 4-chloro-2-[(4- methylphenyl)thio]benzaldehyde (0.05 g, 0.2 mmol) using the same method as Example 94.
  • Example 138 Preparation of Compound 111 2-Fluoro-5-nitrobenzaldehyde (0.13 g, 0.79 mmol) was dissolved in N,N- dimethylformamide (6.7 mL), and p-cresol (0.22 g, 2.0 mmol) and potassium carbonate (0.27 g, 2.0 mmol) were added thereto, followed by stirring at 25°C for 1.5 hours. After a conventional treatment, the residue was purified by silica gel chromatography (eluted with chloroform), to obtain 5-nitro-2-(4-methylphenoxy)benzaldehyde (0.20 g, 98%).
  • Example 143 Preparation of Compound 116
  • Compound 120 (0.0044 g, 21.2%) was obtained by using Compound 119 (0.02 g, 0.056 mmol) obtained in Example 146 using the same method as Example 96.
  • N,N-diphenylbenzylamine (935 mg, 3.60 mmol) was suspended in acetic acid (20 mL), and hexamethylenetriamine (1.12 g, 7.96 mmol) was added thereto, followed by stirring at 90°C for 12 hours.
  • the reaction liquid was cooled to room temperature, a 6 mol/L sodium hydroxide aqueous solution and water were added thereto, and then the product was extracted with chloroform. The organic layer was washed with brine, and then dried over anhydrous sodium sulfate.
  • Example 151 Preparation of Compound 131 4- ⁇ 4-[(2-Formyl-4-nitrophenyl)thio]benzoyl ⁇ morpholine (0.13 g, 78%) was obtained from 5-nitro-2-[(4-carboxylphenyl)thio]benzaldehyde (0.13 g, 0.42 mmol) obtained in
  • Example 149 N,N-dimethylformamide (7.1 mL), morpholine (0.073 mL, 0.83 mmol), 1-ethyl- 3-(3-dimethylaminopropyl)carbodiimide hydrochloride (0.16 g, 0.83 mmol), and 1- hydroxybenzotriazole monohydrate (0.11 g, 1.7 mmol), using the same method as Example 150.
  • Example 138 - Compound 133 (0.12 g, 100%) was obtained from Compound 75 (0.11 g, 0.27 mmol) obtained in Example 102, dichloromethane (21 mL), methanol (4.2 mL) and m- chloroperbenzoic acid (0.10 g, 0.30 mmol), using the same method as Example 134.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Thiazole And Isothizaole Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)

Abstract

L'invention porte sur des composés de thiazolidinedione, sur des compositions, et sur des procédés d'inhibition de l'activité de la télomérase in vitro et sur le traitement des états ou maladies induits par la télomérase ex vivo et in vivo. Les procédés, composés et compositions de l'invention peuvent être utilisés seuls, ou en combinaison avec d'autres agents actifs d'un point de vue pharmacologique dans le traitement d'états ou maladies induits par l'activité de la télomérase, par exemple dans les traitements anticancéreux. L'invention porte également sur de nouvelles méthodes de dosage ou recherche systématique des inhibiteurs de l'activité de la télomérase.
EP00950282A 1999-07-01 2000-06-30 Inhibiteurs de telomerase et leurs procedes d'utilisation Withdrawn EP1109796A1 (fr)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US14217399P 1999-07-01 1999-07-01
JP18761699 1999-07-01
US142173P 1999-07-01
JP18761699 1999-07-01
JP30757699A JP2001072592A (ja) 1999-07-01 1999-10-28 テロメラーゼ阻害剤
JP30757699 1999-10-28
PCT/US2000/018211 WO2001002377A1 (fr) 1999-07-01 2000-06-30 Inhibiteurs de telomerase et leurs procedes d'utilisation

Publications (1)

Publication Number Publication Date
EP1109796A1 true EP1109796A1 (fr) 2001-06-27

Family

ID=27325921

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00950282A Withdrawn EP1109796A1 (fr) 1999-07-01 2000-06-30 Inhibiteurs de telomerase et leurs procedes d'utilisation

Country Status (9)

Country Link
EP (1) EP1109796A1 (fr)
JP (1) JP2003507473A (fr)
KR (1) KR20010099634A (fr)
CN (1) CN1321153A (fr)
AU (1) AU6340600A (fr)
CA (1) CA2341253A1 (fr)
HK (1) HK1043113A1 (fr)
ID (1) ID29875A (fr)
WO (1) WO2001002377A1 (fr)

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1376150A (zh) * 1999-07-26 2002-10-23 盐野义制药株式会社 具有血栓形成素受体拮抗作用的药物组合物
DE60115132T2 (de) * 2000-04-21 2006-07-06 Pfizer Products Inc., Groton Thyroid-Rezeptorliganden
US6620830B2 (en) 2000-04-21 2003-09-16 Pfizer, Inc. Thyroid receptor ligands
EP1276730A2 (fr) * 2000-04-24 2003-01-22 ARYx Therapeutics Compositions et methodes destinees au traitement du diabete, de l'hyperlipidemie, de l'hypercholesterolemie et de l'atherosclerose
US6958355B2 (en) 2000-04-24 2005-10-25 Aryx Therapeutics, Inc. Materials and methods for the treatment of diabetes, hyperlipidemia, hypercholesterolemia, and atherosclerosis
US6768008B2 (en) * 2000-04-24 2004-07-27 Aryx Therapeutics Materials and methods for the treatment of diabetes, hyperlipidemia, hypercholesterolemia, and atherosclerosis
US6784199B2 (en) 2000-09-21 2004-08-31 Aryx Therapeutics Isoxazolidine compounds useful in the treatment of diabetes, hyperlipidemia, and atherosclerosis in mammals
US6680387B2 (en) 2000-04-24 2004-01-20 Aryx Therapeutics Materials and methods for the treatment of diabetes, hyperlipidemia, hypercholesterolemia, and atherosclerosis
DE10133665A1 (de) * 2001-07-11 2003-01-30 Boehringer Ingelheim Pharma Carbonsäurederivate, diese Verbindungen enthaltende Arzneimittel, deren Verwendung und Herstellung
WO2003018011A1 (fr) 2001-08-24 2003-03-06 Wyeth Holdings Corporation 5-(substitue)-5-(substitue sulfonyl ou sulfanyl)thiazolidine-2,4-diones utiles pour l'inhibition de la farnesyl-proteine transferase
US7045523B2 (en) 2001-10-18 2006-05-16 Novartis Ag Combination comprising N-{5-[4-(4-methyl-piperazino-methyl)-benzoylamido]-2-methylphenyl}-4-(3-pyridyl)-2-pyrimidine-amine and telomerase inhibitor
GB0204252D0 (en) * 2002-02-23 2002-04-10 Oxford Glycosciences Uk Ltd Novel compounds
WO2004028535A1 (fr) * 2002-09-26 2004-04-08 Pintex Pharmaceuticals, Inc. Composes de modulation de pin-1 et leurs procedes d'utilisation
US7781464B2 (en) * 2003-01-17 2010-08-24 Bexel Pharmaceuticals, Inc. Heterocyclic diphenyl ethers
US6794401B2 (en) 2003-01-17 2004-09-21 Bexel Pharmaceuticals, Inc. Amino acid phenoxy ethers
US7521465B2 (en) 2003-01-17 2009-04-21 Bexel Pharmaceuticals, Inc. Diphenyl ether derivatives
WO2004093803A2 (fr) * 2003-04-16 2004-11-04 Pintex Pharmaceuticals, Inc. Composes photochimiotherapeutiques utilises dans le traitement d'etats associes a pin1
US7087576B2 (en) 2003-10-07 2006-08-08 Bexel Pharmaceuticals, Inc. Dipeptide phenyl ethers
FI20041129A0 (fi) * 2004-08-30 2004-08-30 Ctt Cancer Targeting Tech Oy Tioksotiatsolidinoniyhdisteitä lääkkeinä käytettäviksi
CA2580811C (fr) 2004-09-28 2014-04-08 Otsuka Pharmaceutical Co., Ltd. Derive de carbostyrile
FR2887444A1 (fr) * 2005-06-28 2006-12-29 Oreal Composes benzylidene-1,3-thiazolidine-2,4-diones, leurs utilisations et compositions pour stimuler ou induire la pousse des fibres keratiniques et/ou freiner leur chute et/ou augmenter leur densite.
KR100814109B1 (ko) 2006-01-09 2008-03-14 한국생명공학연구원 로다닌 유도체, 이의 제조방법 및 이를 유효성분으로함유하는 약학적 조성물
TWI315304B (en) * 2006-08-31 2009-10-01 Univ Taipei Medical Indoline-sulfonamides compounds
WO2008109731A2 (fr) * 2007-03-07 2008-09-12 Jassen Pharmaceutica N.V. Phénoxythiazolidinediones n-alkylées en tant que modulateurs du récepteur-α œstrogénique
EP2217235A4 (fr) * 2007-11-15 2011-01-12 Musc Found For Res Dev Inhibiteurs de protéines kinases pim, compositions et procédés pour traiter le cancer
TWI461423B (zh) * 2008-07-02 2014-11-21 Astrazeneca Ab 用於治療Pim激酶相關病狀及疾病之噻唑啶二酮化合物
CN101973893B (zh) * 2010-09-07 2013-07-17 黑龙江省科学院石油化学研究院 4-[2-(1-苯乙烯基)]-n,n-二(4-溴苯基)苯胺的合成方法
KR101817287B1 (ko) * 2016-01-08 2018-01-10 조선대학교 산학협력단 티아졸리딘디온 유도체 및 그의 용도
CN116217511B (zh) * 2023-03-22 2024-04-19 沈阳药科大学 一种双靶点化合物及其制备方法和在制备sEH抑制剂与PPARs激动剂中的应用

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5522636A (en) * 1978-08-04 1980-02-18 Takeda Chem Ind Ltd Thiazoliding derivative
JPS5697277A (en) * 1980-01-07 1981-08-05 Takeda Chem Ind Ltd Thiazolidine derivative
JPS58118577A (ja) * 1982-01-07 1983-07-14 Takeda Chem Ind Ltd チアゾリジン誘導体
CN1003445B (zh) * 1984-10-03 1989-03-01 武田药品工业株式会社 噻唑烷二酮衍生物,其制备方法和用途
AR240698A1 (es) * 1985-01-19 1990-09-28 Takeda Chemical Industries Ltd Procedimiento para preparar compuestos de 5-(4-(2-(5-etil-2-piridil)-etoxi)benzil)-2,4-tiazolidindiona y sus sales
US4738972A (en) * 1985-05-21 1988-04-19 Pfizer Inc. Hypoglycemic thiazolidinediones
US4812570A (en) * 1986-07-24 1989-03-14 Takeda Chemical Industries, Ltd. Method for producing thiazolidinedione derivatives
WO1989008650A1 (fr) * 1988-03-08 1989-09-21 Pfizer Inc. Agents hypoglycemiques de thiazolidinedione
WO1989008651A1 (fr) * 1988-03-08 1989-09-21 Pfizer Inc. Derives de thiaxolidinedione hypoglycemiques
IE940525L (en) * 1988-05-25 1989-11-25 Warner Lambert Co Known and selected novel arylmethylenyl derivatives of¹thiazolidinones, imidazolidinones and oxazolidinones useful¹as antiallergy agents and antiinflammatory agents
US5143929A (en) * 1991-05-09 1992-09-01 Warner-Lambert Company 2-substituted thiazolidinone, oxazolidinone, and imidazolidinone derivatives of fenamates as antiinflammatory agents
US6251928B1 (en) * 1994-03-16 2001-06-26 Eli Lilly And Company Treatment of alzheimer's disease employing inhibitors of cathepsin D
US5814647A (en) * 1997-03-04 1998-09-29 Board Of Regents, The University Of Texas System Use of troglitazone and related compounds for the treatment of the climacteric symptoms

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0102377A1 *

Also Published As

Publication number Publication date
CA2341253A1 (fr) 2001-01-11
WO2001002377A9 (fr) 2001-10-11
JP2003507473A (ja) 2003-02-25
WO2001002377A1 (fr) 2001-01-11
HK1043113A1 (zh) 2002-09-06
CN1321153A (zh) 2001-11-07
AU6340600A (en) 2001-01-22
KR20010099634A (ko) 2001-11-09
ID29875A (id) 2001-10-18

Similar Documents

Publication Publication Date Title
US6518268B1 (en) Telomerase inhibitors and methods of their use
WO2001002377A1 (fr) Inhibiteurs de telomerase et leurs procedes d'utilisation
US5863936A (en) Telomerase inhibitors
US5767278A (en) Telomerase inhibitors
US5656638A (en) Telomerase inhibitors
US6452014B1 (en) Telomerase inhibitors and methods of their use
US5760062A (en) Telomerase inhibitors
US5770613A (en) Telomerase inhibitors
US5703116A (en) Telomerase Inhibitors
WO1992009279A1 (fr) Derive de 2-arylthiazole et composition pharmaceutique contenant ce derive
WO2001072745A1 (fr) 4-heteroaryle-pyrimidines substituees en 2 et leur utilisation dans le traitement de troubles proliferants
JP2003155274A (ja) 置換されたインドールアルカノール酸
CA2408448A1 (fr) Indazoles substitues avec de la 1,1-dioxoisothiazolidine utiles comme inhibiteurs de la proliferation cellulaire
JPH09188677A (ja) 医薬有効物質としての置換されたイミダゾリジン− 2,4− ジオン− 化合物
US7329682B2 (en) Method for inhibiting 5-lipoxygenase using a benzoxazole derivative
NZ528347A (en) Inhibitors of cyclin dependent kinases in the treatment of proliferative disorders
US20230039679A1 (en) Compounds for inhibiting tnik and medical uses thereof
US6372742B1 (en) Substituted indole compounds and methods of their use
RU2324685C2 (ru) Производные тиазола, способ их получения и применение, фармацевтическая композиция, обладающая свойствами антагонистов рецепторов npy
WO1999065875A1 (fr) Inhibiteurs de la telomerase
GB2387172A (en) [(Aryl-/arylthio-)aryl]methylene substituted azole & azine derivatives and their therapeutic use as antibacterials
US7563905B2 (en) Triazole derivatives and method of using the same to treat HIV infections
CN109574920B (zh) 3-腈基-6环丙基吡啶类ido1抑制剂及其用途
EP0881878A1 (fr) Derives d'arylhydrazones utiles comme agents antibacteriens
WO2002076397A2 (fr) Inhibiteurs de telomerase et leurs procedes d'utilisation

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010226

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

RIN1 Information on inventor provided before grant (corrected)

Inventor name: CHIN, ALLISON, C.

Inventor name: ASAI, AKIRA

Inventor name: PIATYSZEK, MIECZYSLAW, A.

Inventor name: AKAMA, TSUTOMU

Inventor name: TOLMAN, RICHARD, L.

Inventor name: SINGH, UPINDER

Inventor name: KANDA, YUTAKA

Inventor name: HOLCOMB, RYAN

17Q First examination report despatched

Effective date: 20030130

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20030611