EP0575526A1 - 6,9 BIS(SUBSTITUTED-AMINO)BENZO g]ISOQUINOLINE-5,10-DIONES - Google Patents

6,9 BIS(SUBSTITUTED-AMINO)BENZO g]ISOQUINOLINE-5,10-DIONES

Info

Publication number
EP0575526A1
EP0575526A1 EP92908816A EP92908816A EP0575526A1 EP 0575526 A1 EP0575526 A1 EP 0575526A1 EP 92908816 A EP92908816 A EP 92908816A EP 92908816 A EP92908816 A EP 92908816A EP 0575526 A1 EP0575526 A1 EP 0575526A1
Authority
EP
European Patent Office
Prior art keywords
amino
benzo
dione
bis
isoquinoline
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.)
Pending
Application number
EP92908816A
Other languages
German (de)
French (fr)
Other versions
EP0575526A4 (en
Inventor
A. Paul Krapcho
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.)
University of Vermont
Original Assignee
University of Vermont
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 University of Vermont filed Critical University of Vermont
Publication of EP0575526A1 publication Critical patent/EP0575526A1/en
Publication of EP0575526A4 publication Critical patent/EP0575526A4/xx
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • C07D221/06Ring systems of three rings
    • C07D221/08Aza-anthracenes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • This invention is directed to 6,9 bis (substitutedamino)benzo[g]isoquinoline-5,10-diones, and more particularly, to 6,9-substituents which are (aminoalkyl)amino substituents. These compounds have been shown to have antitumor activity in vitro and in vivo.
  • Mitoxantrone is a broad spectrum oncolytic agent, whose activity is similar to that of the anthracycline antibiotic doxorubicin. Clinical trials have demonstrated that mitoxantrone has particularly promising activity in the treatment of advanced breast cancer, acute leukemia and lymphoma (Legha, Drugs of Today, (1984), 20, 629).
  • Ametantrone has been reported to be, in animals, about 10-fold less potent and cardiotoxic than mitoxantrone. Because a delayed toxicity is observed only with mitoxantrone after administration of the two drugs by the i.p. route to non-tumor bearing rats at equieffective antitumor dosages, it is suggested that the presence of the 5,8-dihydroxy substitution in mitoxantrone might be implicated in the delayed deaths (Corbett et al., Cancer Chemother. Pharmacol., (1981), 6, 161).
  • both mitoxantrone and ametantrone have a remarkable myelodepressive toxicity and both compounds show cross-resistance to cell histotypes developing resistance against doxorubicin mediated by overexpression of glycoprotein P.
  • Such a resistance which is named multidrug resistance, involves a number of antitumor antibiotics, among which amsacrine and podophyllotoxinic derivatives, and it is one of the main reasons for therapeutical failures in the treatment of solid tumors with said antibiotics.
  • novel anthracenedione antitumor agents having a higher therapeutical index than mitoxantrone and being effective both in inhibiting or delaying the growth of those solid tumors which are more refractory to chemotherapeutic treatment (such as lung, breast and colon tumors) and against tumor histotypes developing multidrug resistance.
  • Aza- and diaza anthracene-9,10-diones such as 6,9- bis(ethoxycarbonylamino)benzo[g]quinoline-5,10-dione
  • the compounds 5a and 5b are less cytotoxic than the analogues 6a and 6b.
  • the compound 5a which is poorly cytotoxic in vitro, is inactive in vivo, and it is both less active and less potent than the carbocyclic analogue 6a. Even though the introduction of an heteroatom is a common process in the medicinal chemistry, its effect must be evaluated case by case.
  • mitoxantrone has shown good activity in other significative experimental tumors such as murine Lewis lung carcinoma and MXl human mammary carcinoma.
  • the compounds of the invention have the formula
  • R is C 1 -C 10 alkyl, phenyl or C 1 -C 10 aralkyl
  • C 1 -C 10 alkyl having one or two substituents selected from the group consisting of OR 1 and -NR 2 R 3 ;
  • R 1 is selected from the group consisting of hydrogen, C 1 -C 6 alkyl, phenyl, C 7 -C 10 aralkyl, -CHO, -COR 5 ,
  • R 4 is selected from the group consisting of hydrogen, C 1 -C 10 alkyl, C 2 -C 10 hydroxyalkyl, C 2 -C 10 alkyl substituted by -NR 2 R 3 , C 7 -C 10 aralkyl, phenyl, -COR 5 , -COOR 5 or -S(O 2 )R 5 ;
  • R 5 is selected from the group consisting of C 1 -C 10 alkyl, C 6 -C 10 aralkyl, ⁇ -, ß-, or ⁇ -naphthyl, phenyl o-, m- , or p-tolyl;
  • the present invention also concerns the tautomeric forms, the single enantiomers and diastereoisomers of the compounds of formula (I), as well as mixtures thereof.
  • the present invention also concerns the non-toxic salts of the compounds of formula (I) with acids acceptable for pharmaceutical and veterinary use such as those obtained by addition of inorganic acids like hydrochloric, hydrobromic, sulfuric, phosphoric, pyrophosphoric acid and/or of organic acids such as acetic, propionic, citric, benzoic, lactic, maleic, fumaric, succinic, tartaric, glutamic, aspartic, gluconic, ascorbic acids and the like.
  • acids acceptable for pharmaceutical and veterinary use such as those obtained by addition of inorganic acids like hydrochloric, hydrobromic, sulfuric, phosphoric, pyrophosphoric acid and/or of organic acids such as acetic, propionic, citric, benzoic, lactic, maleic, fumaric, succinic, tartaric, glutamic, aspartic, gluconic, ascorbic acids and the like.
  • phenyl means phenyl rings which can optionally contain substituents such as
  • (C 1 -C 4 )alkyl groups CF 3 , halogen atoms, nitro, amino, acetylamino, formylamino, dimethylamino, diethylamino, hydroxy, methoxy and ethoxy groups.
  • C 1 -C 10 alkyl groups are methyl, ethyl, n-propyl, sec-propyl, n-butyl, secbutyl, tert-butyl, n-pentyl, n-hexyl.
  • C 7 -C 10 aralkyl are benzyl and 4-methoxybenzyl.
  • substituent is a 5-6 member aromatic or not aromatic heterocyclic ring which may contain another heroatom such as sulfur, oxygen and nitrogen
  • preferred examples of said heterocyclic rings are 1-imidazolyl, 1-pyrrolyl, 1-tetrahydropyrrolyl, 1-pyrazolyl, 4-mor ⁇ holinyl, 1-piperidinyl, 1-piperazinyl, 1-(4-raethyl)-piperazinyl, 1-(4-benzyl)-piperazinyl.
  • R is a C 2 -C 10 alkyl selected from the group consisting of: - residue of formula -(CH 2 ) p -NH 2 wherein p is 2, 3 or
  • R 2 and R 3 are a C 1 -C 6 alkyl or taken together with the nitrogen atom, they form an heterocyclic ring selected from the group consisting of 1-ethyleneimine, 1-pyrrolidine, 4-morpholine, 1-piperazine, 4-methyl-1-piperazine, 4-benzyl-1-piperazine, 1-piperidine,
  • the compounds of this invention can be prepared by reaction of 6,9-difluorobenzo[g]isoquinoline-5,10-dione of formula (II)
  • R' when R' is one of the groups defined above for R in compounds of formula (I), and in which case the compounds of formula (la) are the same as the compounds of formula (I), R' can be optionally converted into another R group to give another compound of formula c) optional salification and/or solvation of the obtained compounds of formula (I) or separation of the isomers thereof.
  • reaction of the compound of formula (II) with a compound of formula (III) is generally carried out in the presence of a stoichiometric amount or a slight molar excess of a compound of formula (III) in a solvent such as methylene chloride, chloroform, 1,1,1-trichloroethane, dimethoxyethane, tetrahydrofuran, dimethylsulfoxide, dimethylformamide, pyridine, picoline, and mixtures thereof, or, if it is desired, using compound (III) itself as the solvent, optionally in the presence of an inorganic base such as an alkaline or alkalineearth carbonate or hydrogen carbonate or an organic base such as a trialkylamine, at a temperature from 0oC to the reflux temperature of the solvent.
  • a solvent such as methylene chloride, chloroform, 1,1,1-trichloroethane, dimethoxyethane, tetrahydrofuran, dimethylsulfoxide,
  • reaction is carried out in a solvent such as pyridine, chloroform or dimethylsulfoxide, using from 2 to 10 equivalents of compound (III) for 1 equivalent of compound (II) and working at a temperature ranging from room temperature to 50oC.
  • a solvent such as pyridine, chloroform or dimethylsulfoxide
  • E is a hydroxy protective group such as a trialkylsilane, (dialkyl)arylsilane, formyl, acetyl, which reaction may be optionally followed by removal of the protective group E.
  • the 6,9-difluorobenzo(g ⁇ isoquinoline-5,10-dione of formula (II) may be prepared by a multistep procedure involving the Friedel-Crafts acylation of 1,4-difluorobenzene with pyridine-3,4-dicarboxylic acid anhydride, which results in compounds having the structure according to formulas (VIIa) and (VIIb):
  • the compounds of formula (V) may be prepared according to the procedures described in Synth. Comm., (1990), 20, 2559 and in J. Med. Chem., (1990), 33, 97.
  • the evaluation of the "in vitro" cytotoxic activity of the compounds of the invention was performed using a human colon adenocarcinoma cell line (Lovo) isolated from a metastatic nodule and a subline with aquired resistance to a number of antitumor agents, among which doxorubicin, VP-16 and vincristine.
  • This subline (named Lovo/DX) shows reduced accumulation of doxorubicin and overexpression of a protein (Grandi, M., Geroni, C., Giuliani, F.C., British, J. Cancer, (1986), 54, 515).
  • the "in vitro" cytotoxic evaluation was also performed on L 1210 murine leukemia cells using the above mentioned cells maintained in suspension cultures (McCoy's 5A medium supplemented with 10% horse serum, glutamine, penicillin and streptomycin) grown in an humidified environment of 10% carbon dioxide and 90% air at 37oC.
  • the compounds were dissolved in dimethyIsulfoxide (DMSO) and added to the suspended cells in appropriate concentrations. After 72 hours of continuous exposure, the cell concentration was counted using a Coulter Counter and growth inhibition calculated using the formula:
  • % growth inhibition 1 - (cell number treated/cell number DMSO alone] ⁇ 100.
  • the IC 50 was calculated from the growth inhibition data and they are reported in table VII in comparison to the prior art compound 5a (see table I for structure of 5a).
  • P 388 murine leukemia cells were intraperitoneally
  • ip intravenously
  • iv intravenously injected in CD2F1 mice.
  • Treatment was initiated approximately 24 hours after tumor transplantation and dosages of the drug were admi nistered ip (P388 ip/ip) or iv (P388 iv/iv) according to preestablished protocols, usually at 3-day (P388 iv/iv) or 4-day (P388 ip/ip) intervals.
  • the studies were done over a 60-day period and the date of death for each animal was recorded.
  • the % T/C was determined using the mean survival time (MST) for each group according to the formula
  • % T/C [(MST treated)/(MST control)] ⁇ 100
  • 6 representative compounds of this invention namely 6,9-bis ⁇ [2-(amino)ethyl]aminojbenzo- [g]isoquinoline-5,10-dione 10i as the dimaleate salt (10i maleate) described in example 12 and 6,9-bis[[(2- dimethylamino)ethyl]amino)benzo[g]isoquinoline-5,10- dione 10a described in example 4 showed an activity superior to that of mitoxantrone in the P 388 iv/iv model.
  • Compound 10i of the present invention both as the hydrochloride (10i.HCl) and the dimaleate salt (10i.maleate) described in example 12, was superior to mitoxantrone also in the P 388 ip/ip model. Moreover the above representative compounds of the invention showed antileukemic activity over a wide range of well tolerated dosages and in particular they were active at dosages which were lower than the maximum tolerated dose, providing indication for a more favourable therapeutic index in comparison to mitoxantrone. The results are shown in table VIII and table IX.
  • the antitumor activity of representative compounds of this invention was evaluated also in the L 1210 murine leukemia model.
  • L 1210 leukemia cells were intraperitoneally (ip) injected in CDF1 mice and treatment was initiated ap proximately 24 hours after tumor transplantation. Dosages of the drugs were administered ip according to preestablished protocols, usually at 4-day intervals. The studies were done over a 60-day period and the date of death for each animal was recorded. The % T/C was determined using the mean survival time (MST) for each group according to the formula
  • table XII shows activity data against these two solid tumors of one representative compound of this invention, 6,9-bis ⁇ [(2-amino)ethyl]amino]benzo[g]isoquinoline-5,10-dione dimaleate (10i maleate; example 12).
  • the compounds of the present invention may therefore be used as active ingredients of therapeutic compositions to induce regression and/or palliation of cancers in mammals when administered in amounts ranging from about 1 mg to about 0.4 g per kilogram of body weight.
  • a preferred dosage regimen would be from about 1 mg to about 50 mg per kilogram of body weight per day.
  • Unit dosages may be employed so that from about 70 mg to about 3.5 g of the active compound for a subject of about 70 kg of body weight are administered in a 24-hour period.
  • the dosage may be adjusted to be compatible to other treatment regimens, such as radiation therapy.
  • the pharmaceutical composition may be in the form of tablets, capsules, gel capsules, suppositories, lyophilized powders and solutions for intravenous administration.
  • N,N-diisopropylethylenediamine (588 mg, 4.1 mmol) was added to compound 9 (100 mg, 0.41 mmol) in methanol (1 ml) and water (1 ml). The mixture was allowed to stir at room temperature for 88 h and then poured into ice water. The blue precipitate was recovered by filtration. The solid was purified by column chromatography over silica gel. The initial eluant was chloroform followed by 2% and 20% methanol in chloroform. Concentration of the latter eluants led to 163 mg (81%) of product 10c.
  • the crude material obtained (6.07 g) was recrystallized by dissolution in methanol (20 ml) at 40oC and reprecipitation with methylene chloride (100 ml) and n-hexane (300 ml) to give 5.12 g of 10i as a blue solid.
  • Example 12 The procedure of Example 12 using compound !9 and 1,2-diaminoethane, was repeated and the crude reaction mixture was applied to a silica gel column. Acetic anhydride (30 ml) was added to the column and it was allowed to stand for 15 minutes. A major blue fraction 10j eluted with 1:4 CH 3 OH:CHCl 3 which was crystallized from a CHCl 3 :CH 3 OH mixture to yield a blue solid (0.240 g, 35%) m.p.
  • reaction mixture was allowed to reach room temperature and left at this temperature for three hours.
  • the reaction mixture was applied to a silica gel (100 g) column chromatography and eluted first with CHCl 3 : CH 3 OH 90:10, then with CHCl 3 :CH 3 OH 85:15 and finally with CHCl 3 :CH 3 OH:NH 4 OH 85:15:1.
  • the fractions containing the product were pooled, the solvents were removed and the residue was subjected to a second purification hy silica gel (95 g) column chromatography, eluting with CHCl 3 :CH 3 OH:NH 4 OH cone. from 95:5:0 to 80:20:2.
  • the initial eluant was 5% methanol/95% chloroform followed by increasing the methanol amounts gradually to 10%, 20%, 30%, 40% and 50%.
  • the desired compound coluld be eluted using 60% methanol/40% chloroform containing some ammonium hydroxide. Removal of the eluant led to 50 mg (30%) of the product (10o). mp 105-106oC.
  • the resultant blue solid was purified by column chromatography over silica gel using chloroform as the initial eluant followed by gradual changes to 2%, 5%, 10%, 20%, 40% and 50% methanol in chloroform.
  • the desired product was eluted from the column with 50% methanol/49% chloroform/1% ammonium hydroxide. Removal of the eluants led to 56 mg (30%) of the desired product (10p). mp 136-137oC.
  • the MTT assay was performed according to Mosmann, T., J. Immunol. Methods, (1983), 65, 55-63 and Green, L. M., J. Immunol. Methods, (1984), 70, 257-268.
  • Murine leukemia cells were routinely maintained in suspension cultures in McCoy's 5A medium suppiemented with 10% horse serum glutamine, penicillin and streptomycin and grown in a humidified environment of 10% carbon dioxide and 90% air at 37oC.
  • each compound was dissolved in dimethylsulfoxide and added to 1 ml of L1210 cells (10 cells/tube) to attain final concentrations of 0.01, 0.1 and 1 pg of drug/ml of culture. After 72 hours of continues exposure to the drug, the cell concentration was determined with a Coulter Counter. Growth inhibition was calculated for each drug using the following formula:
  • % growth inhibition 1-[cell number treated/cell number DMSO alone] ⁇ 100.
  • the growth inhibition data was then used to calculate the IC 50 value (the calculated drug concentration required to inhibit cell growth by 50% of control.
  • P388 Murine Leukemia cells were maintained in vivo by serial intraperitoneal (i.p.) injections of 10 6 cells in DBA2 mice.
  • CDF1 mice were inoculated intravenously (i.v.) with 10 P388 cells and treatment was initiated 24 hr later.
  • the i.v. dose of drug was administered on days 1, 4 and 7. Mice were observed daily for signs of toxicity and survival. The date of death was recorded for each animal that died or was sacrificed during the 60 day study.
  • the median survival time (MST) for each treatment group was calculated and the % T/C was determined using the following formula:
  • CDF1 Bale mice were transplanted iv with 10 cells/mouse. Treataent was given iv on days 1,4,7 after tumor transplantation (day 0).
  • P388 Murine Leukemia cells were maintained in vivo by serial intraperitoneal (ip) injections of 10 cells in DBA2 mice.
  • ip serial intraperitoneal
  • CDF1 mice were inoculated i.p. with 10 P388 cells and treatment was initiated 24 hours later.
  • the i.p. dose of drug was administered on days 1, 5 and 9. Mice were observed daily for signs of toxicity and survival. The date of death was recorded for each animal that died or was sacrificed during the 60-day study.
  • the median survival time (MST) for each treatment group was calculated and the % T/C was determined using the following formula:
  • MITOX 1.5 186(160-225) 6/26 1/26
  • CDF1 mice were injected ip with 10 cells/mouse; treatment was given ip
  • L1210 murine leukemia cells were maintained in vivo by weekly intraperitoneal (ip) injections of 10 cells in BDF. mice.
  • mice were inoculated i.p. with 10 L1210 cells and treatment was initiated 24 hours later.
  • the desired dose of drug was administered on days 1, 5 and 9.
  • Mice were observed daily for signs of toxicity and survival. The date of death was recorded for each animal that died or was sacrificed during the 60-day study.
  • the mean survival time (MST) for each treatment group was calculated and the % T/C was determined using the following formula:
  • L1210 murine leukemia cells were maintained in vivo by weekly intraperitoneal (ip) injections of 10 cells in DBA2 mice.
  • ip intraperitoneal
  • CDFl mice were inoculated i.p. with 10 L1210 cells and treatment was initiated 24 hours later.
  • the desired dose of drug was administered on days 1, 5 and 9.
  • Mice were observed daily for signs of toxicity and survival. The date of death was recorded for each animal that died or was sacrificed during the 60-day study.
  • the median survival time (MST) for each treatment group was calculated and the % T/C was determined using the following formula:
  • CDF1 mice were injected ip with 10 cells/mouse; treatment was given
  • C57bl/6 female mice were trasplantated im (intralimbs) with 10 cells. Treatment was given iv (intravenously) on days 1, 7, 15, after tumor transplantation (day 0). The mean tumor weight for each treatment group was calculated according to Geran, R.I. et al., Cancer Chemother. Rep., (1972), 3 , 51-61 and the TWI% was calculated 7 days after the last drug treatment using the formula:
  • TWI% 100-[(Mean tumor weight of treated mice) / (mean tumor weight of controls)] ⁇ 100.
  • CD1 nu/nu female mice were transplantated sc (subcutaneously) with tumor fragments (about 1 mm ⁇ 1 mm ⁇ 1 mm). Treatment was given iv once a week for three weeks, when tumor weight reached an average of 150 mg. For all individual tumors the weight change (relative tumor weight) for the start of treatment (V o ) was expressed as V t /V o at each day of measurement (V t ). TWI% was calculated 7 days after the last drug treatment using the formula:
  • TWI% 100-[(mean relative tumor weight of treated mice) /(mean relative tumor weight of controls)] ⁇ 100.
  • Table XII 6,9-bis ⁇ [2-(amino)ethyl]amino)benzo[g]isoquinoline-5,10-dione (10i) as the dimaleate salt (10i maleate) of the example 12.

Abstract

A compound according to formula (I): <CHEM> wherein R is C1-C10 alkyl; phenyl or C7-C10 aralkyl; C2-C10 aklyl substituted with one or two substituents selected from the group consisting of OR1 and -NR2R3; C2-C10 alkyl interrupted by one or two oxygen atoms or by a member selected from the group consisting of -NR4-, cis -CH=CH, trans -CH=CH- and -C=C-, and optionally substituted with one or two hydroxy (OH) or -NR2R3 groups; and wherein R1 is selected from the group consisting of hydrogen, C1-C6 alkyl, phenyl, C7-C10 aralkyl, -CHO, -COR5-, -COOR5, -S(O2)R5 and C2-C6 alkyl optionally substituted with -NR2R3; R2 and R3 are the same or different and are selected from the group consisting of hydrogen, C1-C10 alkyl, C7-C10 aralkyl, phenyl, C2-C10 alkyl substituted with one or two hydroxy (OH) groups, -CHO, -COR5, -COOR5, and -S(O2)R5, R2 and R3 taken together with the nitrogen atom to which they are bound form an ethyleneimine ring or a 5- or 6-membered aromatic or non-aromatic heterocyclic ring optionally containing another heteroatom selected from the group consisting of sulfur, oxygen and nitrogen, R2 is H and R3 is -C(=NH)NH2 or R2 is -C(=NH)NH2 and R3 is H; R4 is selected from the group consisting of hydrogen, C1-C10 alkyl, C2-C10 hydroxyalkyl, C2-C10 alkyl substituted with -NR2R3, C7-C10 aralkyl, phenyl, -COR5, -COOR5 and -S(O2)R5; R5 is selected from the group consisting of C1-C10 alkyl, C7-C10 aralkyl, alpha -, beta -, or gamma -naphthyl, phenyl, o-, m-, or p-tolyl as free bases and their salts with pharmaceutically acceptable acids, have been found to have cytostatic and anti-tumor activity.

Description

6,9 BIS(SUBSTITUTED-AMINO)BENZO[g]ISOQUINOLINE-5,10-DI- ONES
BACKGROUND OF THE INVENTION
Field of the invention
This invention is directed to 6,9 bis (substitutedamino)benzo[g]isoquinoline-5,10-diones, and more particularly, to 6,9-substituents which are (aminoalkyl)amino substituents. These compounds have been shown to have antitumor activity in vitro and in vivo.
Background
Certain 1,4-bis[(aminoalkyl)amino]anthracene-9,10-diones have been reported which show antitumor activity in clinical trials. Of particular interest has been ametantrone, 1,4-bis {[2-(2-hydroxyethylamino)ethyl ]amino} anthracene-9,10-dione and mitoxantrone, 5,8-dihydroxy-1,4-bis{[2-(2-hydroxyethylamino)ethyl]amino}anthracene-9,10-dione. (Zee-Cheng et al., J. Med. Chem., (1978), 21, 291-4; Cheng et al., "Progress in Medicinal Chemistry", Ellis, G.P. and West, G.B., eds.; Elsevier: Amsterdam, 1983; pp. 20, 83 and references cited therein). Mitoxantrone is a broad spectrum oncolytic agent, whose activity is similar to that of the anthracycline antibiotic doxorubicin. Clinical trials have demonstrated that mitoxantrone has particularly promising activity in the treatment of advanced breast cancer, acute leukemia and lymphoma (Legha, Drugs of Today, (1984), 20, 629). Although animal studies have demonstrated a diminished cardiotoxicity in comparison to doxorubicin, some clinical cardiotoxicity has been observed also with mitoxantrone, mostly in patients previously treated with doxorubicin (R. Stuart Harris et al., Lancet, (1984), 219, and references cited therein).
Ametantrone has been reported to be, in animals, about 10-fold less potent and cardiotoxic than mitoxantrone. Because a delayed toxicity is observed only with mitoxantrone after administration of the two drugs by the i.p. route to non-tumor bearing rats at equieffective antitumor dosages, it is suggested that the presence of the 5,8-dihydroxy substitution in mitoxantrone might be implicated in the delayed deaths (Corbett et al., Cancer Chemother. Pharmacol., (1981), 6, 161).
In addition, both mitoxantrone and ametantrone have a remarkable myelodepressive toxicity and both compounds show cross-resistance to cell histotypes developing resistance against doxorubicin mediated by overexpression of glycoprotein P. Such a resistance, which is named multidrug resistance, involves a number of antitumor antibiotics, among which amsacrine and podophyllotoxinic derivatives, and it is one of the main reasons for therapeutical failures in the treatment of solid tumors with said antibiotics.
Therefore, search is required for novel anthracenedione antitumor agents, having a higher therapeutical index than mitoxantrone and being effective both in inhibiting or delaying the growth of those solid tumors which are more refractory to chemotherapeutic treatment (such as lung, breast and colon tumors) and against tumor histotypes developing multidrug resistance.
In the search for safer, active analogues of anthracenediones, compounds bearing hydroxy substituents and/or (aminoalkyl)amino side chains at various posi tions on the anthracene-9,10-dione nucleus have been studied (Cheng et al.. Drugs of the Future, (1983), 8, 229) without notable improvement.
Aza- and diaza anthracene-9,10-diones such as 6,9- bis(ethoxycarbonylamino)benzo[g]quinoline-5,10-dione
(1), 6,9-bis (ethoxycarbonylamino)benzo[g]isoquinoline- 5,10-dione ( 2 ) , 6,9-bis(ethoxycarbonylamino)benzo[g]- quinazoline-5,10-dione (3), were disclosed by Potts et al. (Synthesis, 1983, 31). These compounds were repor- ted to be related to the antitumor 1,4- bis[(aminoalkyl)amino]anthracene-9,10-diones; however, no antitumor activity data was reported for any of the above compounds. 1-((aminoalkyl) amino] derivatives (4) of 6,9-dihydroxybenzo[g]isoquinoline-5,10-dione related to mitoxantrone have been disclosed as DNA intercalators but they were completely devoid of any "in vitro" or "in vivo" antitumor activity (Croisy-Delcey et al., Eur. J. Med. Chem., (1988), 23 , 101-106).
The 6,9-bis[(aminoalkyl)amino]benzo[g]quinoline- 5,10-diones of formulas 5a-d (see Table I) were disclosed in J. Med. Chem. (1985), 28, 1124-26, where they were referred to as 5,8-bis[(aminoalkyl)amino]-1-azaanthtacenediones.
As reported in table II herein below reported the prior art compounds 5 are clearly less active than the corresponding carbocyclic analogues (6).
In fact, as reported in table II, the compounds 5a and 5b are less cytotoxic than the analogues 6a and 6b. Moreover the compound 5a, which is poorly cytotoxic in vitro, is inactive in vivo, and it is both less active and less potent than the carbocyclic analogue 6a. Even though the introduction of an heteroatom is a common process in the medicinal chemistry, its effect must be evaluated case by case.
In this particular case of aza-analogues of anthracenediones the prior art teachings clearly show that the introduction of a nitrogen atom into the anthracenedione skeleton is detrimental for antitumor activity: In fact the aza-substituted anthracenediones are less cytotoxic than the corresponding anthracene- diones and inactive "in vivo".
Thus a person skilled in the art, would have not considered the introduction of heteroatoms into the anthracenedione skeleton as a possible way to obtain more effective antitumor agents.
It is well known that the screening for the discovery of the antitumor agent mitoxantrone (J. Med. Chem., (1978), 21, 291-4) among a number of analogues has been carried out by using the experimental model of murine leukemia P388: this model is then predictive of antitumor activity in humans at least for this class of antitumor antibiotics. Many other clinically active antitumor antibiotics are active on murine leukemias P388 and L1210 such as m-amsacrine and doxorubicin.
Moreover mitoxantrone has shown good activity in other significative experimental tumors such as murine Lewis lung carcinoma and MXl human mammary carcinoma.
We have now discovered that the compounds of the invention 6,9-bis[(aminoalkyl)amino]benzo[g]isoquinoline-5,10-diones are active as antitumor agents: they are highly effective against murine leukemias L1210 and
P388 and against Lewis lung carcinoma and MXl human mammary carcinoma.
Table II
Antitumor activity of prior art compounds 5 and 6 against murine L 1210 leukemia (from J. Med. Chem. (1985), 28, 1124-1126)
in vitro in vivo
ID50 (pg/ml) Dose (mg/kg) T/C
5a 0.16 50 130
5b 2.4 100 toxic
5c 1.7
6a 0.08 30 150
6b 0.30
BRIEF SDMMARY OF THE INVENTION
The compounds of the invention have the formula
( I )
wherein R is C1-C10 alkyl, phenyl or C1-C10 aralkyl;
C1-C10 alkyl having one or two substituents selected from the group consisting of OR1 and -NR2R3;
C2-C10 alkyl interrupted by one or two oxygen atoms or by one member selected from the group consisting of -NR4-, cis -CH=CH, trans -CH=CH-, -C≡C-, and optionally substituted by one or two hydroxy (OH) or -NR2R3 groups;
R1 is selected from the group consisting of hydrogen, C1-C6 alkyl, phenyl, C7-C10 aralkyl, -CHO, -COR5,
-COOR5, -S(O2)R5, C2-C6 alkyl optionally substituted by -NR2R3;
R2 and R3 may be the same or different and are selected from the group consisting of hydrogen, C1-C10 alkyl, C7-C10 aralkyl, phenyl, C2-C10 alkyl substituted with one or two hydroxy (OH) groups, -CHO, -COR5, -COOR5, -S(O2)R5, one of R2 or R3 is H and the other is -C(=NH)NH2, or R2 and R3 taken together with the nitrogen atom to which they are bound form an ethyleneimine ring or a 5- or 6-membered aromatic or non-aromatic heterocyclic ring which might contain another heteroatom such as sulfur, oxygen or nitrogen;
R4 is selected from the group consisting of hydrogen, C1-C10 alkyl, C2-C10 hydroxyalkyl, C2-C10 alkyl substituted by -NR2R3, C7-C10 aralkyl, phenyl, -COR5, -COOR5 or -S(O2)R5;
R5 is selected from the group consisting of C1-C10 alkyl, C6-C10 aralkyl, α-, ß-, or ɣ-naphthyl, phenyl o-, m- , or p-tolyl;
as free bases and their salts with pharmaceutically acceptable acids.
The present invention also concerns the tautomeric forms, the single enantiomers and diastereoisomers of the compounds of formula (I), as well as mixtures thereof.
The present invention also concerns the non-toxic salts of the compounds of formula (I) with acids acceptable for pharmaceutical and veterinary use such as those obtained by addition of inorganic acids like hydrochloric, hydrobromic, sulfuric, phosphoric, pyrophosphoric acid and/or of organic acids such as acetic, propionic, citric, benzoic, lactic, maleic, fumaric, succinic, tartaric, glutamic, aspartic, gluconic, ascorbic acids and the like.
DETAILED DESCRIPTION OF THE INVENTION
In compounds (I) the term "phenyl" means phenyl rings which can optionally contain substituents such as
(C1-C4)alkyl groups, CF3, halogen atoms, nitro, amino, acetylamino, formylamino, dimethylamino, diethylamino, hydroxy, methoxy and ethoxy groups.
Preferred examples of C1-C10 alkyl groups are methyl, ethyl, n-propyl, sec-propyl, n-butyl, secbutyl, tert-butyl, n-pentyl, n-hexyl.
Preferred examples of C7-C10 aralkyl are benzyl and 4-methoxybenzyl. When in compounds of formula (I) R is a C1-C10 alkyl interrupted by one or two oxygen atoms or by one member selected from the group consisting of cis-CH=CH-, trans-CH=CH-, -C≡C- and optionally substituted by one or two hydroxy or -NR2R3 groups, at least two carbon atoms are preferably interposed between said oxygen atoms and/or the -NR.- and -NR2R3 groups.
When in compounds of formula (I) the -NR2R, substituent is a 5-6 member aromatic or not aromatic heterocyclic ring which may contain another heroatom such as sulfur, oxygen and nitrogen, preferred examples of said heterocyclic rings are 1-imidazolyl, 1-pyrrolyl, 1-tetrahydropyrrolyl, 1-pyrazolyl, 4-morρholinyl, 1-piperidinyl, 1-piperazinyl, 1-(4-raethyl)-piperazinyl, 1-(4-benzyl)-piperazinyl.
Particularly preferred are compounds of formula (I) in which R is a C2-C10 alkyl selected from the group consisting of: - residue of formula -(CH2)p-NH2 wherein p is 2, 3 or
4;
- residue of formula -(CH2)p-NR2R3 wherein p is as above defined and R2 and R3 are a C1-C6 alkyl or taken together with the nitrogen atom, they form an heterocyclic ring selected from the group consisting of 1-ethyleneimine, 1-pyrrolidine, 4-morpholine, 1-piperazine, 4-methyl-1-piperazine, 4-benzyl-1-piperazine, 1-piperidine,
- residue of formula -(CH2)p-NR2R3 wherein p is as aabboovvee defined and R2 is hydrogen and R3 is a C1-C6 alkyl,
- residue of formula wherein p is as
above defined
- residue of formula -(CH 2)p-NH-(CH2)q-OH wherein p and q are independently an integer selected from the group consisting of 2, 3, 4,
- residue of formula -(CH 2)p-OH wherein p is as above defined
- residue of formula -(CH 2)p-O-(CH2)q-OH wherein p and q are as above defined.
Specific examples of the preferred compounds of this invention are as follows:
6,9-bis[[(2-(amino)ethyl]amino)benzo[g]isoquinoline- 5,10-dione;
6,9-bis[[2-(4'-morpholino)ethylJaτnino)benzo[g]isoquinoline-5,10-dione;
6,9-bis[[2-(dimethylamino)ethyl]amino)benzo[g]isoquinoline-5,10-dione;
6,9-bis([2-(diethylamino)ethyl]amino)benzo[g]isoquino line-5,10-dione;
6,9-bis{[ 2-[di(sec-propyl)amino]ethyl]amino)benzo[g]- isoquinoline-5,10-dione;
6,9-bis{[2-(1'-pyrrolidino)ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-(1'-aziridino)ethyl}amino)benzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-(methanesulfonyloxy)ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[4-(amino)butyl]amino}benzo(g]isoquinoline5,10-dione;
6,9-bis{[3-(amino)propyl)]amino}benzo[g]isoquinoline- 5,10-dione;
6,9-bis{(2-[(2-hydroxyethyl)amino]ethyl]amino}benzo[g]- isoquinoline-5,10-dione;
6,9-bis{[3-(dimethylamino)propyl]aminojbenzotg]isoquinoline-5,10-dione;
6,9-bis{[(2-hydroxyJethyl]amino}benzo[g]isoquinoline- 5,10-dione;
6,9-bis{[(2-(2-hydroxyethoxy)ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-(methylamino)ethyl]amino}benzo[g)isoquinoline-5,10-dione;
6,9-bis{[2-(ethylamino)ethyl]amino}benzolgjisoquinoline-5,10-dione;
6,9-bis{t2-(n-propylamino)ethyl]aminojbenzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-(sec-propylamino)ethyl]amino)benzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-(guanidino)ethyl]amino}benzo[g]isoquinoline- 5,10-dione; 6,9-bis{[(2-amino-2,2-dimethyl)ethyl]amino)benzo[g]isoquinoline-5,10-dione.
The compounds of this invention can be prepared by reaction of 6,9-difluorobenzo[g]isoquinoline-5,10-dione of formula (II)
with a compound of formula (III)
R'-NH2 (III) wherein R' has the same meanings as defined in formula (I) for R or it is a group which may be converted to R, to give a compound of formula (Ia):
and then optionally performing one or more of the following steps:
a) when R' is other than R, converting R' into R to obtain a compound of formula (I);
b) when R' is one of the groups defined above for R in compounds of formula (I), and in which case the compounds of formula (la) are the same as the compounds of formula (I), R' can be optionally converted into another R group to give another compound of formula c) optional salification and/or solvation of the obtained compounds of formula (I) or separation of the isomers thereof.
The reaction of the compound of formula (II) with a compound of formula (III) is generally carried out in the presence of a stoichiometric amount or a slight molar excess of a compound of formula (III) in a solvent such as methylene chloride, chloroform, 1,1,1-trichloroethane, dimethoxyethane, tetrahydrofuran, dimethylsulfoxide, dimethylformamide, pyridine, picoline, and mixtures thereof, or, if it is desired, using compound (III) itself as the solvent, optionally in the presence of an inorganic base such as an alkaline or alkalineearth carbonate or hydrogen carbonate or an organic base such as a trialkylamine, at a temperature from 0ºC to the reflux temperature of the solvent.
Preferably the reaction is carried out in a solvent such as pyridine, chloroform or dimethylsulfoxide, using from 2 to 10 equivalents of compound (III) for 1 equivalent of compound (II) and working at a temperature ranging from room temperature to 50ºC.
If it is desired the compounds of formula (I) in which R is an hydroxyalkylamino alkyl group of formula -(CH2)p-NH-(CH2)q-OH, wherein p and q are as defined above, can be obtained by reaction of a compound of formula (I) in which R is a group of formula
-(CH2)p-OS(O2)R5 with a compound of formula (IV)
H- 2N-(CH2)q-O-E (IV) wherein E is a hydroxy protective group such as a trialkylsilane, (dialkyl)arylsilane, formyl, acetyl, which reaction may be optionally followed by removal of the protective group E.
If it is desired the compounds of formula (I) in which R is a group of formula -(CH2)p-NR2R3 wherein one of R. or R3 is hydrogen and the other is hydrogen or a C1-C6 alkyl may be obtained by reaction of compound (II) with a monoprotected diamine of formula (V):
H2N-(CH2)p-N(COOR5)R3 (V) wherein R3 is hydrogen or a C1-C6 alkyl and p and R5 are as defined in formula (I), to give a compound of formula (la) in which R' is a group of formula
-(CH2)p-N(COOR5)R3, which reaction may be followed by removal of the protective group COOR5.
Useful teachings for the removal of the above mentioned protective groups can be found in Green, T.W., Wuts, P.G.M., "Protective Groups in Organic Syntesis", second Edition, John Wiley and sons, (1991).
The compounds of formula (I) in which R is a group of formula -(CH2)p-NH-C(=NH)NH2 can be prepared by reaction of a compound of formula (I) in which R is a group of formula -(CH2)p-NH with the reagent of formula (VI):
H2N-C(=NH)SO3H (VI) for example following the procedure described in Tetra. Lett. (1988), 29, 3183-3186.
The 6,9-difluorobenzo(g}isoquinoline-5,10-dione of formula (II) may be prepared by a multistep procedure involving the Friedel-Crafts acylation of 1,4-difluorobenzene with pyridine-3,4-dicarboxylic acid anhydride, which results in compounds having the structure according to formulas (VIIa) and (VIIb):
Compounds according to formulas (VIIa) and (VIIb) may be then subjected to a cyclization reaction in 30% fuming sulfuric acid at approximately 140ºC to give the compound of formula (II).
The compounds of formula (III), (IV) and (V) are known, commercially available or they can be prepared accordingly to known procedures.
For example, the compounds of formula (V) may be prepared according to the procedures described in Synth. Comm., (1990), 20, 2559 and in J. Med. Chem., (1990), 33, 97.
The evaluation of the biological activity for the compounds of this invention was performed "in vitro" and "in vivo" following the protocols developed by the U.S. National Cancer Institute.
The evaluation of the "in vitro" cytotoxic activity of the compounds of the invention was performed using a human colon adenocarcinoma cell line (Lovo) isolated from a metastatic nodule and a subline with aquired resistance to a number of antitumor agents, among which doxorubicin, VP-16 and vincristine. This subline (named Lovo/DX) shows reduced accumulation of doxorubicin and overexpression of a protein (Grandi, M., Geroni, C., Giuliani, F.C., British, J. Cancer, (1986), 54, 515). The compounds were tested according to the MTT assay (Mosman, T., "Rapid Colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assay", J. Immunol. Methods, (1983), 155, 55-63; Green, L.M., "Rapid colorimetric assay for cell viability; application to the quantitation of cytotoxic and growth inhibitory lymphokines", J. Immunol. Methods, (1984), 70, 257-268) in comparison with mitoxantrone, ametantrone and doxorubicin. The data are reported in table VI.
In general representative compounds of this invention were equally cytotoxic as ametantrone and mitoxantrone in the Lovo cell line, the highest cytotoxicity (superior to that of mitoxantrone) being shown by the compound 6,9-bis([2-(dimethylamino)ethyl]amino}- benzo[g]isoquinoline-5,10-dione (10a) described in example 4 of the experimental section. When ametantrone or mitoxantrone were tested in the Lovo/DX cell line, a resistance index R.I. (defined as the ratio of the IC50 for the resistant cell line to the IC50 for the sensible cell line) as high as 101.2 and 29.0 respectivelly was found, showing that this subline did have an aquired resistance to mitoxantrone and ametantrone. On the other hand, when representative compounds of this invention were tested in the same resistant subline no cross resistance with mitoxantrone and ametantrone occured, as can be observed for compounds 10a, 12c, 10b, 10e and 10d described in the examples 4, 22, 5, 8 and 7 of the experimental section, respectively. These compounds, in general, showed on this resistant subline the same IC50 shown by ametantrone on the sensible Lovo cell line. Compound 10a, in particular, was more active than mitoxantrone in the Lovo/DX cell line.
These "in vitro" data suggest that representative compounds of this invention may be useful in order to overcome the multidrug resistance mediated mechanism of tumor resistance.
The "in vitro" cytotoxic evaluation was also performed on L 1210 murine leukemia cells using the above mentioned cells maintained in suspension cultures (McCoy's 5A medium supplemented with 10% horse serum, glutamine, penicillin and streptomycin) grown in an humidified environment of 10% carbon dioxide and 90% air at 37ºC. The compounds were dissolved in dimethyIsulfoxide (DMSO) and added to the suspended cells in appropriate concentrations. After 72 hours of continuous exposure, the cell concentration was counted using a Coulter Counter and growth inhibition calculated using the formula:
% growth inhibition = 1 - (cell number treated/cell number DMSO alone] × 100.
The IC50 was calculated from the growth inhibition data and they are reported in table VII in comparison to the prior art compound 5a (see table I for structure of 5a).
Studies of the biological activity "in vivo"
of representative compounds of the invention were performed using the P 388 and L 1210 murine leukemia models.
P 388 murine leukemia cells were intraperitoneally
(ip) or intravenously (iv) injected in CD2F1 mice. Treatment was initiated approximately 24 hours after tumor transplantation and dosages of the drug were admi nistered ip (P388 ip/ip) or iv (P388 iv/iv) according to preestablished protocols, usually at 3-day (P388 iv/iv) or 4-day (P388 ip/ip) intervals. The studies were done over a 60-day period and the date of death for each animal was recorded. The % T/C was determined using the mean survival time (MST) for each group according to the formula
% T/C = [(MST treated)/(MST control)] × 100 For example two representative compounds of this invention, namely 6,9-bis {[2-(amino)ethyl]aminojbenzo- [g]isoquinoline-5,10-dione 10i as the dimaleate salt (10i maleate) described in example 12 and 6,9-bis[[(2- dimethylamino)ethyl]amino)benzo[g]isoquinoline-5,10- dione 10a described in example 4 showed an activity superior to that of mitoxantrone in the P 388 iv/iv model. Compound 10i of the present invention, both as the hydrochloride (10i.HCl) and the dimaleate salt (10i.maleate) described in example 12, was superior to mitoxantrone also in the P 388 ip/ip model. Moreover the above representative compounds of the invention showed antileukemic activity over a wide range of well tolerated dosages and in particular they were active at dosages which were lower than the maximum tolerated dose, providing indication for a more favourable therapeutic index in comparison to mitoxantrone. The results are shown in table VIII and table IX.
The antitumor activity of representative compounds of this invention was evaluated also in the L 1210 murine leukemia model.
L 1210 leukemia cells were intraperitoneally (ip) injected in CDF1 mice and treatment was initiated ap proximately 24 hours after tumor transplantation. Dosages of the drugs were administered ip according to preestablished protocols, usually at 4-day intervals. The studies were done over a 60-day period and the date of death for each animal was recorded. The % T/C was determined using the mean survival time (MST) for each group according to the formula
% T/C - [(MST treated)/(MST control)] × 100 The results are shown in table X and XI
From table X the surprisingly superior antileukemic activity of representative compounds of the invention (namely compound 10a of example 4, compound 10i. HC1 of example 12 and 6, 9-bis{[2-(2-hydroxyethylamino)- ethyl]amino}benzo[g]isoquinoline-5,10-dione 101 prepared in example 17 (dimaleate salt)) in comparison to the prior art compound Sa is evident.
From table XI a clearly favourable comparison again emerges between the excellent antileukemic activity of compound 10i . HC1 of the invention and mitoxantrone.
Activity against solid tumors of the compounds of the invention was demonstrated using the murine Lewis Lung carcinoma and the human mammary carcinoma MX-1 models. These tumor models are enclosed in the panel of eight transplanted tumors used by the U.S.-NC1 as screen panel for selection of clinically useful antitumor agents (R.K.Y. Zee Cheng and C.C. Cheng, "Screening and evaluation of anticancer agents", Meth. and Find. Expl. Clin. Pharmacol., (1988), 10(2), 67-101). As an example, table XII shows activity data against these two solid tumors of one representative compound of this invention, 6,9-bis {[(2-amino)ethyl]amino]benzo[g]isoquinoline-5,10-dione dimaleate (10i maleate; example 12).
Since representative compounds of this invention show good results against "in vivo" models of murine P 388 and L 1210 leukemias, murine Lewis Lung carcinoma and human mammary carcinoma MX-1, which as noted above are predictive of good results in humans, the compounds disclosed herein are expected to be operative against human leukemias and solid tumors.
The compounds of the present invention may therefore be used as active ingredients of therapeutic compositions to induce regression and/or palliation of cancers in mammals when administered in amounts ranging from about 1 mg to about 0.4 g per kilogram of body weight. A preferred dosage regimen would be from about 1 mg to about 50 mg per kilogram of body weight per day. Unit dosages may be employed so that from about 70 mg to about 3.5 g of the active compound for a subject of about 70 kg of body weight are administered in a 24-hour period. The dosage may be adjusted to be compatible to other treatment regimens, such as radiation therapy.
The pharmaceutical composition may be in the form of tablets, capsules, gel capsules, suppositories, lyophilized powders and solutions for intravenous administration.
The invention is illustrated by the following non-limiting examples, and variations which are readily apparent to those skilled in the art. The pertinent formulae are illustrated in the tables I, III, IV and V. Example 1
Pyridine-3,4-dicarboxylic acid anhydride (7)
A mixture of pyridine-3,4-dicarboxylic acid (15.0 g, 0.09 mol) and acetic anhydride (30 ml) was refluxed for 2 hours. The excess acetic anhydride was removed by distillation and the anhydride was collected and purified by sublimation (123ºC at 3 mm Hg) to yield 7 as a white solid (10.1 g, 76%), m.p. 74-76ºC. 1H NMR (CDCl3) 9.39 (s, 1H), 9.24 (d, 1H), 7.94 (d, 1H).
Example 2
4-(2',5'-Difluorobenzoyl)nicotinic acid (8a) and 3-(2'- 5'-difluorobenzoyl)isonicotinic acid (8b)
A mixture of 2 (5.0 g, 0.033 mol) and aluminum chloride (17.5 g, 0.131 mol) in 1,4-difluorobenzene was heated in an oil bath at 110ºC for 22 hours. The excess of 1,4-difluorobenzene was recovered by distillation. The residue was cooled in an ice bath and quenched with ice water (75 ml) and cone, hydrochloric acid (6.3 ml). The precipitated solid was filtered and dried to yield a white powder (7.7 g, 87%). This material could be crystallized from acetonitrile and water; m.p. 214-217ºC; 1H NMR (DMSO-d6), 9.15 (s), 8.90 (d), 8.80 (d), 7.90 (d), 7.5 (m), 7.4 (m). Anal. Calcd for C13H7F2NO3: C, 59.32; H, 2.69; N, 5.32. Found: C, 59.03; H, 2.55; N, 5.18.
Example 3
6,9-Difluorobenzo[g]isoquinoline-5,10-dione (9)
The keto acid mixture of 8a and 8b (3.0 g, 0.011 mol) in fuming sulfuric acid (7.5 ml, 30% SO3) was heated in an oil bath at 135-140ºC for 3 hours. After cooling to room temperature, the mixture was poured into ice (200 ml) and neutralized with sodium bicarbonate. Extraction with methylene chloride gave 9 as a yellow solid (2.0 g, 72%); m.p. 199-200'C; 1H NMR (CDCl3) 9.54 (s, 1H), 9.12 (d, 1H), 8.03 (d, 1H), 7.57 (m, 2H). Anal. Calcd. for C13H5F2NO2: C, 63.67; H, 2.04; N, 5.71. Found: C, 63.86; H, 1.66; N, 5.39.
Example 4
6,9-Bis{[2-(dimethylamino)ethyl]amino)benzo[g]isoquinoline-5,10-dione (10a)
A mixture of 6,9-difluorobenzo[g]isoquinoline- 5,10-dione, 9 (20 mg, 0.08 mmol) and N,N-dimethylethylenediamine (0.8 mmol, 0.8 ml of a 1 M solution in pyridine) was stirred at room temperature for 48 hours. Most of the pyridine was allowed to evaporate, ether was added, and the product 10a removed by filtration (24 mg, 79%), m.p. 167-168ºC; 1H NMR (CDCl3) 11.06 (br t, 1H), 10.97 (br t, 1H), 9.63 (s, 1H), 8.92 (d, 1H), 8.13 (d, 1H), 7.32 (m, 2H), 3.54 (q, 4H), 2.7 (t, 4H), 2.38 (s, 12H); mass spectrum m/z (relative intensity) 381 (2.0, M+), 59 (5.4), 59 (100); U.V. X max (2- methoxyethanol) 580 (sh, 6200), 614 (10,600), 661
(12,700). Anal. Calcd. for C21H27N5O2: C, 66.12; H, 7.13; N, 18.36. Found: C, 66.22; H, 7.10, N, 18.20.
Example 5
6,9-Bis(12-(diethylamino)ethyl]amino}benzo[g]isoquinoline-5,10-dione (10b)
A solution of N,N-diethylethylenediamine (684 mg,
5.9 mmol) in pyridine (2 ml) was added to 9 (202 mg,
0.82 mmol) in pyridine (2 ml). The purple mixture was stirred at room temperature for 48 hours (TLC analysis at this point showed one blue component: silica gel, 5% MeOH/CHCl3). The excess base and pyridine were removed by evaporation with a slow stream of nitrogen gas. The crude material was then placed under vacuum overnight. Upon addition of ice-water to the residue the solid was collected by filtration and dried (330 mg, 84%); m.p. 142-144ºC. 1H NMR (CDCl3) 11.10 (t, 1H), 11.00 (t, 1H), 9.60 (s, 1H), 8.92 (d, 1H), 8.10 (d, 1H), 7.25 (m, 3H), 3.50 (m, 1H), 2.82 (m, 1H), 2.35 (m, 3H), 1.10 (m, 12H). Anal. Calcd. for C25H35N5O2: C, 68.62; H, 16.00; N, 7.31. Found: C, 67.95; H, 16.05; N, 7.28.
Example 6
6,9-Bis{[2-[di(sec-propyl)amino]ethyl]amino}benzo[g]- isoquinoline-5,10-dione (10c)
N,N-diisopropylethylenediamine (588 mg, 4.1 mmol) was added to compound 9 (100 mg, 0.41 mmol) in methanol (1 ml) and water (1 ml). The mixture was allowed to stir at room temperature for 88 h and then poured into ice water. The blue precipitate was recovered by filtration. The solid was purified by column chromatography over silica gel. The initial eluant was chloroform followed by 2% and 20% methanol in chloroform. Concentration of the latter eluants led to 163 mg (81%) of product 10c. mp 134-136ºC; 1H NMR (CDCl3): 10.95-11.20 (m, 2H), 9.63 (s, 1H), 8.92 (d, 1H), 8.13 (d, 1H), 7.33 (m, 2H), 3.45 (2q, 4H), 3.10 (h, 4H), 2.82 (t, 4H), 1.08 (d, 24H), Anal. Calcd. for C29H43N5O2 : C, 70.55; H, 8.78; N, 14.19. Found: C, 69.23; H, 8.71; N, 13.43.
Example 7
6,9-Bis(12-(1' pyrrolidino)ethyl]amino}benzo[g]isoquinoline-5,10-dione (10d) A solution of 1-(2-aminoethyl)pyrrolidine (690 mg, 6.0 mmol) in pyridine (2 ml) was added to 9 (202 mg, 0.82 mmol) in pyridine (2 ml). The mixture was stirred at room temperature for 49 hours. The excess base and pyridine were removed using a slow stream of nitrogen. The residue was placed under vacuum overnight. Cold water was added to the residue and a tacky solid separated. The product was extracted with chloroform (3 × 35 ml), the extracts dried over sodium sulfate and the chloroform removed by rotary evaporation to yield the title compound (260 mg, 73%); TLC, silica gel, 5%
MeOH/CHCl3, one blue spot; m.p. 141-142ºC. 1H NMR
(CDCl3) 11.15 (t, 1H), 11.00 (t, 1H), 9.65 (s, 1H), 8.90 (d, 1H), 8.05 (d, 1H), 7.35 (m, 2H), 3.65 (m, 4H), 2.90 (m, 4H), 2.70 (m, 8H), 1.85 (m, 8H). Anal. Calcd. for C25H31N5O2: C, 69.26; H, 7.21; N, 16.15. Found: C, 69.02; H, 7.25; N, 16.00.
Maleate salt. Under a nitrogen atmosphere a solution of maleic acid (271 mg; 2.34 mmol) in ethanol (5 ml) was added to a stirred suspension of compound 10d (450 mg; 1.04 mmol) in ethanol (20 ml). After stirring for two hours diethyl ether (25 ml) was slowly added and the precipitate was filtered and dried under vacuum at 40ºC to give the hygroscopic dimaleate salt of 10d (580 mg); m.p. 164-166ºC.
Anal. Calcd. for C33H39N5O10 · H2O: C, 57.96; H, 6.04; N, 10.24. Found: C, 57.85, H, 5.99; N 10.14.
Example 8
6,9-Bis(12-(4'-morpholino)ethyl]amino}benzo[g]isoquinoline-5,10-dione (10e)
A solution of 4-(2-aminoethyl)morpholine (220 mg, 1.7 mmol) in pyridine (2 ml) was added to 9 (102 mg, 0.42 mmol) in pyridine (1 ml). The purple mixture was stirred at room temperature for 120 hours. The excess amine and pyridine were evaporated using a slow nitrogen stream and the residue placed under vacuum overnight. The mixture was taken up into chloroform and then filtered. Removal of the chloroform left a blue solid which on TLC (silica gel, 25% MeOH/75% CHCl3/few drops of aq. ammonium hydroxide) showed one blue spot (140 mg, 72%); 1H NMR (CDCl3) 11.015 (m, 1H) , 10.90 (m, 1H)
9.62 (d, 1H), 8.90 (d, 1H), 8.10 (d, 1H) , 7.25 (m, 2H), 3.80 (m, 8H), 3.55 (m, 4H), 2.75 (t, 4H), 2.50 (m, 8H). Anal. Calcd. for C25H31N5O4: C, 64.22; H, 6.68; N, 11.98. Found: C, 63.95; H, 6.78; N, 11.89.
Example 9
6,9-Bis(12-(1'-aziridino)ethyl]amino}benzo[g]isoquinoline-5,10-dione (10f)
The procedure of example 4 was repeated reacting compound 9 with N-(2-aminoethyl)aziridine and the residue obtained after evaporation of pyridine was purified by silica gel column chromatography.
A major blue band eluted with 10% CH3OH in CHCl3 was collected. Crystallization from a mixture of methylene chloride and ligroine yielded a dark blue solid, m.p. 100-103ºC; 1H NMR (CDCl3) 11.23 (br, 1H), 11.15 (br, 1H), 9.73 (s, 1H), 8.91 (d, 1H), 8.12 (d, 1H), 7.40 (d, 1H), 7.38 (d, 1H) , 3.69 (q, 4H), 2.60 (t, 4H), 1.85 (m, 4H), 1.24 (m, 4H); mass spectrum m/z (relative intensity) 377 (100, M+), 278 (72.7), 56 (12.6). Anal. Calcd. for C21H23N5O2: C, 66.82; H, 6.14; N, 18.55. Found: C, 66.50; H, 6.00; N, 18.20. Example 10
6,9-Bis{[2-1N-(t-butoxycarbonyl)amino]ethyl]amino}benzo[g ]isoquinoline-5 , 10-dione (10g )
A solution of 9 (0.10 g, 0.41 mmol) and N-(t-butoxycarbonyl)ethylenediamine (Synth. Comm., (1990), 20, 2559; 0.65 g, 4.10 mmol) in pyridine (4.0 ml) was stirred at room temperature for 24 hours. Addition of water (20 ml) led to 10g which was filtered, washed with water and dried (0.17 g, 80%), m.p. 213-216ºC. Crystallization from a CHCl3 : CCl4 mixture gave a dark blue solid m.p. 215-216ºC; 1H NMR (CDCl3) 11.07 (br, 1H), 10.96 (br, 1H), 9.49 (s, 1H), 8.90 (d, 1H), 7.99 (d, 1H), 7.32 (s, 2H), 5.30 (br, 2H), 3.58 (m, 4H), 3.47
(m, 4H), 1.56 (s, 18H); mass spectrum m/z (relative intensity) 525 (100, M+) 339 (51.9), 57 (87.7). Anal. Calcd. for C27H35N5O6: C, 61.70; H, 6.73; N, 13.32. Found: C, 61.18; H, 6.29; N, 12.88.
Example 11
6,9-Bis{[2-(N-t-butoxycarbonyl-N-methylamino)ethyl]amino}benzo[g]isoquinoline-5,10-dione (10h)
6,9-difluorobenzo[g]isoquinoline-5,10-dione (0.29 g, 1.18 mmol) was added to a stirred solution of N-(t-butoxycarbonyl)-N-methylethylenediamine (0.824 g, 4.73 mmol) (J. Med. Chem. 1990, 33, 97) in dry pyridine (5 ml). The reaction mixture was stirred in a nitrogen atmosphere for 24 hours at room temperature and then for further 8 hours at 50ºC. The solvent was removed under reduced pressure and the blue residue obtained was taken up with CH2Cl2 (50 ml), washed with a 5% NaHCO3 solution (2 × 30 ml) and with water (30 ml). The combined organic phases were dried over Na2SO4 and the sol vent evaporated under reduced pressure.
The blue residue was purified by column chromatography (silica gel 230-400 mesh, 50 g, eluent CH2Cl2:
AcOEt: MeOH 93:5:2) yielding 400 mg (61%) of blue crystals, m.p. 161.5 - 162.5ºC after recrystallization from CH2Cl2: hexane.
1H NMR (CDCl3) 1.49 (s, 18H); 2.94 (s, 6H); 3.45-3.75 (m, 8H); 7.25-7.55 (m, 2H); 8.12 (d, 1H); 8.95 (d, 1H); 9.62 (s, 1H); 10.95-11.23 (m, 2H, exchangeable with D2O).
Example 12
6,9-Bis{[2-(amino)ethyl]amino}benzo[g]isoquinoline¬
5,10-dione (10i)
From difluoride 9: Under a nitrogen atmosphere 1,2-diaminoethane (8.72 ml; 130.5 mmol) was added to a stirred solution of compound 9 (4.00 g, 16.3 mmol) in pyridine (80 ml). A slightly exotermic reaction ensued that was moderated by cooling in a water bath at 20ºC. The reaction mixture was stirred at room temperature for 34 hours, then it was cooled to -5ºC for 30 minutes, the solid was separated by suction under a nitrogen flow and dried overnight under vacuum at 30ºC. The crude material obtained (6.07 g) was recrystallized by dissolution in methanol (20 ml) at 40ºC and reprecipitation with methylene chloride (100 ml) and n-hexane (300 ml) to give 5.12 g of 10i as a blue solid.
1H NMR (CDCl3): 11.10-11.30 (2m, 2H), 9.53 (s, 1H), 8.93 (d, 1H)), 8.13 (d, 1H), 7.35 (m, 2H), 3.55 (q, 4H), 3.10 (t, 4H).
HPLC Lichrospher 100 RP18, 5 μm; eluent: sodium heptanesulfonate 2 g/l in H2O: CH3CN: dioxane 75:20:5, pH 3 with H3PO4;λ = 245 nm; flow 1 ml/min; R = 8.80 minutes.
Maleate salt: Under a nitrogen atmosphere the free base 10i (3.50 g; 10.8 mmol) was dissolved in a mixture of ethanol (170 ml) and methanol (30 ml) by heating at 50ºC for 10 minutes, then a solution of maleic acid (2.87 g, 24.7 mmol) in ethanol (30 ml) was added. After heating for five more minutes at 50ºC the mixture was allowed to cool to room temperature and then left at 4ºC for 2 hours. The precipitate was separated by suction under a nitrogen flow and dried at 40ºC under vacuum to give 5.75 g of the crude dimaleate.
To a stirred suspension of this crude material (5.71 g) in water (90 ml) at 50ºC ethanol was added dropwise until a complete solution was obtained (about 300 ml), then more ethanol was added (400 ml) and the mixture was allowed to cool to room temperature. After 12 hours the precipitate was separated by suction, washed with ethanol (15 ml) and diethyl ether (10 ml) and then dried under vacuum at 30ºC for 5 hours to give 4.22 g of the dimaleate salt of 10i; m.p. 176-178ºC shrinking, it does not melt up to 245ºC; Anal. Calcd. for C17H19N5O2.2C4H4O4.1.5 H2O: C, 51.37; H, 5.17; N, 11.98. Found: C, 51.31; H 4.99; N 11.93.
1H NMR (DMSO-d6) 10.92 (br, 1H), 10.87 (br, 1H), 9.45 (s, 1H), 9.01 (m, 1H), 8.08 (m, 1H), 7.59 (s, 2H), 6.03 (s, 4H), 3.76 (br, 4H) , 3.08 (m, 4H). U.V., 1.528 mg in water (50 ml), nm (E 1%) : 641 (271), 597 (245), 313 (98), 273 (257), 246 (479).
Hydrochloride salt:
a) From amine 10i: Hydrogen chloride gas was bubbled through a solution of the crude amine in chloroform until the solution was no longer blue. The solid was filtered and dried to yield a dark blue hygroscopic solid (0.174 g, 26%), m.p. 209-212ºC; 1H NMR (DMSO-d6) 10.91 (br, 2H), 9.44 (s, 1H), 9.01 (d, 1H), 8.23 (br, 4H), 8.09 (d, 1H), 7.74 (s, 2H), 3.84 (m, 4H), 3.02 (m, 4H). Anal. Calcd. for C17H19N5O2.2HCl.4H2O; C, 44.94; H, 6.43; N, 15.41. Found: C, 44.52; H, 5.29; N 14.53. The free amine could be regenerated by adding solid potassium carbonate to the NMR sample. A 1H NMR analysis indicated the presence of the free amine.
b) From deprotection of t-Boc Analogue 10g: Hydrogen chloride gas was bubbled through a solution of 10g (0.160 g, 0.30 mmol) in dry chloroform (10 ml) for 30 minutes. A dark blue solid was filtered and dried (0.115 g, 95%); m.p. 2l3-215ºC whose structure was confirmed as the hydrochloride salt of 10i by 1H NMR analysis.
Example 13
6,9-Bis{[2-(acetylamino)ethyl]amino}benzo[g]isoquinoli¬ne-5,10-dione (10j)
The procedure of Example 12 using compound !9 and 1,2-diaminoethane, was repeated and the crude reaction mixture was applied to a silica gel column. Acetic anhydride (30 ml) was added to the column and it was allowed to stand for 15 minutes. A major blue fraction 10j eluted with 1:4 CH3OH:CHCl3 which was crystallized from a CHCl3:CH3OH mixture to yield a blue solid (0.240 g, 35%) m.p. 155-156ºC; 2H NMR (DMSO-d6) 11.12 (t, 1H), 11.02 (t, 1H), 9.42 (s, 1H), 8.95 (d, 1H), 8.15 (br, 2H), 8.03 (d, 1H), 7.62 (s, 2H), 3.57 (m, 8H), 1.83 (s, 6H); mass spectrum m/z (relative intensity) 409 (2.5 M+), 337 (6.4), 86 (100). Anal. Calcd. for C12H23N5O4 C, 61.60; H, 5.67; N, 17.10. Found: C, 61.37; H, 5.42 N, 16.89.
Example 14
6,9-Bis{[2-(methylamino)ethyl]amino)benzo(g]isoquinoline-5,10-dione trihydrochloride (10k)
Ethanolic HCl (6.7 N, 2.0 ml) was added to a solution of 6,9-bis{[2-(N-t-butoxycarbonyl-N-methylamino)ethyljamino}benzo(gjisoquinoline-5,10-dione (10h) (440 mg, 0.795 mmol) in CHCl3 (40 ml). The reaction mixture was stirred at room temperature in a nitrogen atmosphere for 24 hours, then the dark green crystals formed were collected by filtration, washed with absolute ethanol and dried at 40ºC in vacuo to give 945 rag (94% yield) of pure product, m.p. 188ºC dec. (DSC evaluation).
Elemental Analysis Calcd. for C19H26Cl3N5O2.2H2O: C, 46.51; H, 6.03; N, 14.14; Cl 23.60. Found: C, 45.75; H 6.06; N, 14.04, Cl 21.32. HPLC (Lichrosper 100 RP18, 5 pm; Eluent K2HPO4 25 mM, n-C7H15SO3Na 2 mg/ml in H2O: CH3CN: dioxane 80:15:5, pH 2.7 with HClO4; λ 275 nm, flow 1 ml/mih., RT 20.11 minutes.
U.V., 1.07 mg in 20.0 ml of water: nm (E 1%): 641 (290); 597 (267); 313 (106); 273 (278); 247 (486).
1H NMR (D2O) 2.75 (s, 6H), 3.30-3.45 (m, 4H), 3.80-3.95 (m, 4H), 7.38 (a, 2H); 8.17 (d, 1H), 8.85 (d, 1H), 9.27 (s, 1H).
Example 15
6,9-Bis[(2-hydroxyethyl)amino]benzo[g]isoquinoline¬5,10-dione (11a) Difluoride !9 (1.0 g, 4.1 mmol) and 2-aminoethanol (2.5 g, 40.8 mmol) in pyridine (7 ml) were stirred at room temperature for 18 hours. The mixture was poured into water (50 ml) and the product 11a was filtered and dried (1.26 g, 94%), m.p. 236-239ºC. Recrystallization from methanol yielded dark blue needles m.p. 237-239ºC; 1H NMR (DMSO-d6) 11.28 (t, 1H), 11.19 (t, 1H), 9.43 (s, 1H), 8.94 (d, 1H), 8.03 (d, 1H), 7.56 (s, 2H), 5.01 (t, 2H), 3.69 (m, 4H), 3.54 (m, 4H). Anal. Calcd. for C17H17N3O4: C, 62.38; H, 5.25; N, 12.83. Found: C, 62.21; H, 5.12; N, 12.50.
Example 16
6,9-BisI(2-methanesulfonyloxyethyl)aminoJbenzo[g]iso- quinoline-5,10-dione (11b)
A solution of 11a (0.30 g, 0.92 mmol) in dry pyridine (4 ml) was stirred at room temperature under nitrogen for 10 minutes. Methanesulfonyl chloride (0.24 g, 2.07 mmol) was added and the mixture stirred for 20 minutes. The mixture was quenched into ice-water (20 ml) and the solid was filtered. Crystallization from a chloroform-ligroine mixture gave a blue solid (0.294 g, 66%); m.p. 116-118ºC; 1H NMR (CDCl3) 10.98 (br, 2H), 9.59 (s, 1H), 8.97 (d, 1H), 8.09 (d, 1H), 7.38 (d, 1H), 7.34 (d, 1H), 4.50 (t, 4H), 3.83 (q, 4H), 3.09 (s, 6H). Anal. Calcd. for C19H21N3O8S2 : C, 47.20; H, 4.39; N,
8.69. Found: C, 46.80; H, 4.31; N, 8.48.
Example 17
6,9-Bis((2-[(2-hydroxyethyl)amino]ethyl]amino}benzo[g]-isoquinoline-5,10-dione (101)
a) From mosylate (11b)
A solution of (11b) (0.40 g, 0.83 mmol) and 2- (trimethylsilyloxy)ethylamine (2.21 g, 16.5 mmol) in pyridine (5.0 ml) was stirred at room temperature under a nitrogen atmosphere for 48 hours. The pyridine was removed under a nitrogen flow and the residue was taken up in methylene chloride. This solution was washed with saturated sodium bicarbonate and dried over sodium sulfate. Removal of the solvent left a blue oil which was dried under vacuum overnight. Chromatography on a silica gel column resulted in a cleavage of the Si-O bond to yield 101. This product was eluted with a 1:5:5 Et3N:CH3OH:CHCl3 mixture. Concentration gave 101 as an oil: 1H NMR (CDCl3) 11.19 (br, 1H), 11.06 (br, 1H), 9.46 (s, 1H), 8.83 (d, 1H), 7.98 (d, 1H), 6.98 (s, 2H), 3.82 (m, 4H), 3.52 (m, 4H), 3.07 (m, 4H), 2.96 (m, 4H); U.V. (2-methoxyethanol): 570 (sh, ε = 7200), 612 ( ε = 13,200), 658 (ε= 14,000).
b) From difluoride (9)
Under a nitrogen atmosphere 2-[(2-hydroxyethyl)- aminojethylamine, (6.19 ml; 61.2 mmol) was added to a stirred suspension of compound 9 (1.00 g; 4.08 mmol) in pyridine (30 ml) at 0ºC.
After one hour the reaction mixture was allowed to reach room temperature and left at this temperature for three hours. The reaction mixture was applied to a silica gel (100 g) column chromatography and eluted first with CHCl3: CH3OH 90:10, then with CHCl3:CH3OH 85:15 and finally with CHCl3:CH3OH:NH4OH 85:15:1. The fractions containing the product were pooled, the solvents were removed and the residue was subjected to a second purification hy silica gel (95 g) column chromatography, eluting with CHCl3:CH3OH:NH4OH cone. from 95:5:0 to 80:20:2. A final purification over grade I basic alumina (25 g) eluting with CH2Cl2: EtOH 96:4 gave a residue of 500 mg that was crystallized from a mixture of ethanol (0.5 ml) and methylene chloride (10 ml) to give 101 (142 mg).
Maleate salt: The free amine 101 (138 mg; 0.334 mmol) was dissolved in ethanol (6 ml) under a nitrogen atmosphere and a solution of maleic acid (87 mg; 0.75 mmol) in ethanol (3 ml) was added at room temperature. After cooling for 3 hours at 4ºC the blue crystals obtained were separated by suction under a nitrogen flow, washed with ethanol (1 ml) and diethyl ether (2 ml) and dried under vacuum at 40ºC to give the dimaleate salt of 101 (160 mg), m.p. 132-133ºC; 1H NMR (DMSO-d6) 10.94 (br, 1H), 10.88 (br, 1H), 9.47 (s, 1H), 9.02 (d, 1H), 8.60 (br, 2H), 8.09 (d, 1H), 7.63 (s, 2H), 6.01 (s, 4H), 5.32 (br, 2H), 3.89 (m, 4H), 3.71 (m, 4H), 3.26 (m, 4H), 3.12 (m, 4H). U.V. 1.355 mg in H2O (25 ml): nm (E 1%) : 641 (220), 598 (208), 312 (85), 272 (221), 246 (406).
Anal. Calcd. for C21H27N5O4 · 2C4H4O4. H2O: C, 52.52; H, 5.57; N, 10.64. Found: C, 52.49; H, 5.56; N, 10.61.
Example 18
6,9-Bis{[2-(2-methoxyethylamino)ethyl]amino}benzo[g]-isoquinoline-5,10-dione (10m)
A solution of 11b (0.15 g, 0.21 mmol) and 2-methoxyethylamine (0.47 g, 6.20 mmol) in pyridine (2.0 ml) was stirred at room temperature under a nitrogen blanket for 48 hours. The piridine and excess amine were removed under vacuum. The residue was dissolved in methylene chloride, washed with a saturated sodium bi carbonate solution and dried over sodium sulfate. Upon removal of the solvent the residue was purified by column chromatography on silica gel by eluting with 1:1 CH3OH:CHCl3. Crystallization from a ethanol and pentane mixture led to 10m (0.091 g, 66%); m.p. 174-175ºC; 1H NMR (CDCl3) 11.08 (br, 1H), 10.97 (br, 1H), 9.56 (s, 1H), 8.90 (d, 1H), 8.05 (d, 1H), 7.31 (s, 2H), 3.62 (m, 8H), 3.43 (s, 6H), 3.08 (t, 4H), 2.94 (t, 4H), 2.66 (br s, 2H); mass spectrum m/z (relative intensity) 441 (100, M+), 354 (57.4), 266 (50.5). Anal. Calcd. for
C23H31N5O4: C, 62.57; H, 7.09; N, 15.86. Found: C, 62.51; H, 6.92; N, 15.47.
Example 19
6,9-Bis{[2-(2-hydroxyethoxy)ethyl]amino}benzo[g]isoquinoline-5,10-dione (11c)
A solution of 2-(2-aminoethoxy)ethanol (446 rag, 4.25 mmol) in pyridine (1 ml) was added to compound 9 (100 mg, 0.41 mmol) in pyridine (1 ml). The mixture was stirred at room temperature for 45 h. The excess amine and pyridine were removed via a slow stream of nitrogen gas and the residual solid was placed under vacuum overnight. A cold saturated NaCl solution was added to the solid and the product was extracted with chloroform (6 × 25 ml). The extracts were dried over magnesium sulfate and concentrated on the rotary evaporator to yield 140 mg (82%) of product 11c; mp 97-99ºC. 1H NMR (CDCl3): 11.15-11.35 (2m, 2H), 9.60 (s, 1H), 8.85 (d, 1H), 8.08 (d, 1H), 7.30 (m, 2H), 3.85 (m, 8H), 3.57-3.75 (2m, 9H), 2.75-3.25 (br s, 1H).
Example 20
6,9-Bis{[3-(amino)propyl]amino}benzo[g]isoquinoline- 5 , 10-dione ( 12a)
1) Free Amine: A solution of 1,3-diaminopropane (870 mg, 11.76 mmol) in chloroform (3 ml) was added to 9 (300 mg, 1.22 mmol) in chloroform (6 ml). The purple mixture was allowed to stir for 166 hours at room temperature. The mixture was filtered and the salts washed with chloroform. The solvent was removed by rotary evaporation and the residue was placed under vacuum overnight to yield 290 mg (93%) of 12a; TLC (silica gel, 25% MeOH/75% chloroform/few drops of aq. ammonium hydroxide) indicated one blue spot; m.p. 105ºC (softens) 112-115ºC. 1H NMR (DMSO-d6) 11.15 (m, 1H), 11.05 (m, 1H), 9.45 (s, 1H), 8.90 (d, 1H), 8.0 (d, 1H), 7.55 (br s, 2H), 3.55 (br m, 4H), 4.65 (t, 4H), 3.75 (t, 4H), 1.75 (t, 4H). Anal. Calcd. for C19H23N5O2: C,
64.57; H, 6.56; N, 19.81. Found: C, 65.00; H, 6.50; N,
19.78.
2) Maleate Salt: A solution of maleic acid (11 mg, 0.86 mmol) in methanol was added to 12a (100 mg, 0.28 mmol) dissolved in methanol (2 ml) and ethyl acetate (2 ml). The addition of more ethyl acetate gave a blue precipitate which was filtered and dried under vacuum (120 mg, 79%), 1H NMR (DMSO-d6) 11.10 (t, 1H), 11.00 (t, 1H), 9.45 (s, 1H), 8.95 (d, 1H), 8.05 (d, 1H), 7.75 (br, 4H), 7.60 (s, 2H), 6.0 (s, 4H), 3.60 (m, 4H), 2.95 (t, 4H), 1.95 (m, 4H). Anal. Calcd. for C23H27N5O6: C, 58.84; H, 5.80; N, 14.92. Found: C, 58.75; H, 5.70; N, 14.85.
Example 21
6,9-Bis{[4-(amino)butyl]amino}benzo[g]isoquinoline-5,10-dione (12b) 1) Free Amine: A solution of 1,4-diaminobutane (960 mg, 10.9 mmol) in chloroform (3 ml) was added to 9 (319 mg, 1.3 mmol) in chloroform (6 ml). The purple reaction mixture was stirred at room temperature for 217 hours (9 days). The precipitate salts were removed by filtration amd the filtrate concentrated under a slow nitrogen stream to yield the free amine 12b (400 mg, 80%); TLC (Silica gel, 25% MeOH/75% CHCl3/few drops of aq. ammonium hydroxide); m.p. 80ºC, (softens) 90-92ºC. H NMR (DMSO-d6) 11.15 (m, 1H), 11.05 (m, 1H), 9.42 (s, 1H), 8.90 (d, 1H), 8.0 (d, 1H), 7.5 (br s, 2H), 3.45 (m, 4H), 2.60 (m, 4H), 1.70 (m, 4H), 1.45 (m, 4H). Anal. Calcd. for C21H27N5O2: C, 66.12; H, 7.13; N, 18.36. Found: C, 64.26; H, 6.95; N, 17.42.
2) Maleate Salt: A solution of maleic acid (116 mg, 1 mmol) in ethyl acetate (4 ml) was added to 12b (124 mg, 0.40 mmol) in a methanol (6 ml) and ethyl acetate (8 ml) solution. A blue oil separated and the mixture was placed in the refrigerator overnight. The solvents were removed by deeantation and the remaining solid washed thoroughly with ethyl acetate to yield a blue, very hygroscopic solid, 125 mg (63%). 1H NMR (DMSO-d6) 11.20 (m, 1H), 11.10 (m, 1H), 9.45 (s, 1H), 8.95 (d, 1H), 8.05 (d, 1H), 7.70 (br s, 4H), 6.00 (s, 4H), 3.55 (m, 4H), 2.85 (m, 4H), 1.70 (m, 8H). Anal. Calcd. for
C29H35N5O10: C, 56.76; H, 5-75; N, 11.41. Found: C, 53.65; H, 6.10; N, 10.05.
Example 22
6,9-Bis{[3-(dimethylamino)propyl]amino}benzo[g]isoquinoline-5,10-dicne (12c)
A solution of 3-dimethylaminopropylamine (700 mg, 6.9 mmol) in pyridine (2 ml) was added to compound 9 (100 mg, 0.41 mmol) in pyridine (2 ml). The mixture was stirred at room temperature for 120 h. The excess diamine and pyridine were removed under a slow stream of nitrogen gas and the residue placed under vacuum overnight. The residue was added to cold water and extracted with chloroform (4 × 20 ml). The extract was dried over Na2SO4 and concentrated by rotary evaporation. The blue solid was purified by column chromatography over silica gel. The initial eluant was 10% methanol/90% chloroform followed by 25% methanol/75% chloroform. The product was eluted by addition of small amounts of ammonium hydroxide to 25% methanol/75% chloroform. Concentration of these eluants led to 92 mg (55%) of the desired product 12c mp 107-109ºC. 1H NMR (CDCl3): 11.00-11.20 (2m, 2H), 9.63 (s, 1H), 8.93 (d, 1H), 8.12 (d, 1H), 7.37 (s, 2H), 3.55 (q, 4H), 2.55 (m, 4H), 2.35 (s, 12H), 2.00 (m, 4H).
Example 23
6,9-bis(12-(ethylamino)ethyl)amino}benzo[g]isoquinoline-5,10-dione (10n)
A solution of N-ethylethylenediamine (400 mg, 4.5 mmol) in pyridine (1 ml) was added to compound 9 (98 mg, 0.40 mmol) in pyridine (1 ml). The mixture was stirred at room temperature for 66 h. The pyridine and excess diamine were removed under a slow nitrogen stream and the residual material was placed under vacuum overnight. Chloroform was then added and the chloroform washed twice with cold water. The chloroform extract was dried over MgSO4 and the solvent removed by rotary evaporation. The resultant blue solid was purified by column chromatography over silica gel usin 5% methanol/95% chloroform as the initial eluant. The eluant was gradually changed to 5%, 10% and then 50% methanol in chloroform. The desired product was eluted using 50% methanol/48% chloroform/2% ammonium hydroxide. Removal of the solvent led to 32 mg (21%) of product (10n). mp 101-102ºC.
1H NMR (CDCl3) 11.10 (m, 1H), 11.00 (m, 1H), 9.6 (s, 1H), 8.9 (d, 1H), 8.05 (d, 1H), 7.3 (m, 2H), 3.55 (m, 4H), 3.0 (t, 4H), 2.8 (q, 4H), 1.15 (t, 6H).
Example 24
6,9-bis{[2-(propylamino)ethyl]amino}benzo[g]isoquinoline-5,10-dione (10o)
A solution of N-propylethylenediamine (403 mg, 4.0 mmol) in pyridine (1 ml) was added to compound 9 (98 mg, 0.40 mmol) in pyridine (1 ml). The reaction mixture was stirred at room temperature for 24 h and concentrated under a slow stream of nitrogen. The residual materaial was placed under vacuum overnight. Ice water was added to the residual and the aqueous layer was extracted with chloroform (5 × 40 ml). The extracts were dried over magnesium sulfate and the chloroform removed by rotary evaporation. The blue solid was purified by column chromatography over silica gel. The initial eluant was 5% methanol/95% chloroform followed by increasing the methanol amounts gradually to 10%, 20%, 30%, 40% and 50%. The desired compound coluld be eluted using 60% methanol/40% chloroform containing some ammonium hydroxide. Removal of the eluant led to 50 mg (30%) of the product (10o). mp 105-106ºC.
1H NMR (CDCl3) 11.15 (m, 1H), 11.05 (m, 1H), 9.6 (s. 1H), 8.9 (d, 1H), 8.10 (d, 1H), 7.3 (m, 2H), 3.6 (m, m, 4H), 3.0 (t, 4H), 2.75 (t, 4H), 1.6 (m, 4H, H2O peak superimposed), 0.95 (t, 6H).
Example 25
6,9-bis {(2-(isopropylamino)ethyl]amino}benzo(g]isoquinoline-5,10-dione (10p)
A solution of N-isopropylethylenediamine (450 mg, 4.5 mmol) in pyridine (2 ml) was added to compound 9 (110 mg, 0.45 mmol) in pyridine (1 ml). An instantaneous permanganate-like coloration resulted. The mixture was stirred at room temperature for 40 h. The pyridine and excess diamine were removed under a slow nitrogen stream and the residue was placed under vacuum overnight. Chloroform (20 ml) was added followed by ice water. Since the blue coloration appeared int he aqueous layer, this was extracted with chloroform (4 × 25 ml). The combined extracts were dried over MgSO4 and the chloroform removed by rotary evaporation. The resultant blue solid was purified by column chromatography over silica gel using chloroform as the initial eluant followed by gradual changes to 2%, 5%, 10%, 20%, 40% and 50% methanol in chloroform. The desired product was eluted from the column with 50% methanol/49% chloroform/1% ammonium hydroxide. Removal of the eluants led to 56 mg (30%) of the desired product (10p). mp 136-137ºC.
1H NMR (CDCl3) 11.1 (t, 1H), 11.0 (t, 1H), 9.6 (s, 1H), 8.9 (d, 1H), 8.1 (d, 1H), 7.30 (m, 2H), 3.6 (m, 4H), 3.0 (t, 4H), 2.9 (m, 2H), 1.05 (d, 6H).
Example 26
In vitro biological evaluation: MTT assay on human co Ion adenocarcinoma Lovo cells.
The MTT assay was performed according to Mosmann, T., J. Immunol. Methods, (1983), 65, 55-63 and Green, L. M., J. Immunol. Methods, (1984), 70, 257-268.
Immediately before use, compounds and reference standards are dissolved in proper solvent and subsequently further diluted in complete culture medium. Human colon adenocarcinoma cells Lovo and the subline resistant to doxorubicin (Lovo/DX) (2.5. 104 cells/ml) are plated in 96-well plates and preincubated for 24 hours in culture medium. After this time, the cells are exposed to drugs for 144 hours, then the thiazolyl blue tetrazolium bromide solution (MTT solution) is added and the cells are reincubated for 4 hours. The supernatant is removed and 150 μl of dimethylsulfoxide are added to solubilize formazan crystals. The plate is read at 570 nm with microplate reader and the inhibiting concentration of 50% cellular growth (IC50; μg/ml) is calculated.
The results are reported in Table VI
Example 27
In vitro biological evaluation: L1210 murine leukemia
L1210 Murine leukemia cells were routinely maintained in suspension cultures in McCoy's 5A medium suppiemented with 10% horse serum glutamine, penicillin and streptomycin and grown in a humidified environment of 10% carbon dioxide and 90% air at 37ºC.
To assess the in vitro toxicity, each compound was dissolved in dimethylsulfoxide and added to 1 ml of L1210 cells (10 cells/tube) to attain final concentrations of 0.01, 0.1 and 1 pg of drug/ml of culture. After 72 hours of continues exposure to the drug, the cell concentration was determined with a Coulter Counter. Growth inhibition was calculated for each drug using the following formula:
% growth inhibition = 1-[cell number treated/cell number DMSO alone] × 100.
The growth inhibition data was then used to calculate the IC50 value (the calculated drug concentration required to inhibit cell growth by 50% of control.
The results are reported in Table VII
TABLE VII IN VITRO CYTOTOXIC ACTIVITIES OF REPRESENTA- TIVE COMPOUNDS OF THE INVENTION AGAINST L1210 LEUKEMIA IN COMPARISON TO THE PRIOR ART COMPOUND 5a
Compound Example IC50 (μg/ml)
10a 4 0.006
10f 9 0.002
10i ( a ) 12 0.05
101 ( b ) 17 0.06
5a ( c ) / 0.155
(a) Hydrochloride Salt: 10i.HCl
(b) Dimaleate Salt: 101.maleate
(c) Prior art compound, 6,9-bis {[2-(dimethylamino)-ethyl]amino}benzo[g]quinoline-5,10-dione: A.P. Krapcho et al., J. Med. Chem. (1985), 28, 1124-1126.
Example 28
In vivo biological studies: P388 Murine Leukemia (iv/iv, d 1, 4, 7)
P388 Murine Leukemia cells were maintained in vivo by serial intraperitoneal (i.p.) injections of 106 cells in DBA2 mice. For test purposes, CDF1 mice were inoculated intravenously (i.v.) with 10 P388 cells and treatment was initiated 24 hr later. The i.v. dose of drug was administered on days 1, 4 and 7. Mice were observed daily for signs of toxicity and survival. The date of death was recorded for each animal that died or was sacrificed during the 60 day study. The median survival time (MST) for each treatment group was calculated and the % T/C was determined using the following formula:
% T/C = ((MST treated)/(MST control)] × 100 The results of the antileukemic activity of 6,9-bis{[2-(dimethylamino)ethyl]amino)benzo[g]isoquinoline-5,10-dione (10a) of example 4 and 6,9-bis{[2-(amino)-ethyl]amino)benzo[g]isoquinoline-5,10-dione dimaleate
(10i maleate) of example 12 in comparison to mitoxantrone are reported in table VIII
Table VIII ANTITUMOR ACTIVITY OF COMPOUNDS 10a, 10i.taleate AND MITOXANTRONE
1)
(MITOX) AGAINST P388 MURINE LEUKEMIA (iv/iv 1,4,7)
2) 3) 4) 5)
COMPOUND DOSE T/C% TOX LTS
(mg/kg/day) (range)
Controls - 100 0/73 0/73
10i. maleate 18 200,188 0/16 0/16
27 203(181-227) 2/32 0/32
10a 18 172,156 0/14 0/14
27 188,167 3/17 0/17
40 183,156 1/16 0/16
MITOX 2 162(154-167) 0/42 0/42
3 185(167-217) 4/78 0/78
4 118(89-133) 20/24 0/24
1) CDF1 Bale mice were transplanted iv with 10 cells/mouse. Treataent was given iv on days 1,4,7 after tumor transplantation (day 0).
2) 10i. maleate and MITOX were dissolved in sterile water; 10a in citric acid 1%.
3) Median survival time of treated mice/median survival time of controls × 100: 4 ) Number of toxic deaths/total number of mice.
5) Long term survivors: animals tumor free at the end of the experiment (60 days)
Example 29
In vivo biological studies: P388 Murine Leukemia
(ip/ip, d 1, 5, 9)
P388 Murine Leukemia cells were maintained in vivo by serial intraperitoneal (ip) injections of 10 cells in DBA2 mice. For test purposes, CDF1 mice were inoculated i.p. with 10 P388 cells and treatment was initiated 24 hours later. The i.p. dose of drug was administered on days 1, 5 and 9. Mice were observed daily for signs of toxicity and survival. The date of death was recorded for each animal that died or was sacrificed during the 60-day study. The median survival time (MST) for each treatment group was calculated and the % T/C was determined using the following formula:
% T/C = ((MST treated)/(MST control)] × 100
The results of the antileukemic activity of 6,9-bis{[2-(amino)ethyl]amino)benzo[g]isoquinoline-5,10-dione (10i) as the dihydrochloride (10i.HCl) and dimaleate (10i.maleate) salts (example 12) in comparison to mitoxantrone are reported in table IX
Tabl e IX. ANTITUMOR ACTIVITY OF 10i.HCl , 10i. maleate AND MITOXANTEONE (MITOX ) AGAINST
1)
ASCITIC P3B8 MURINE LEUKEMIA (ip/ip 1,5,9)
2) 3) 4) 5)
COMPOUNDS DOSE % T/C TOX LTS
(mg/kg/day) (range) (day 60)
10i.HCl 3.1 144 0/8 0/8
6.25 196 1/8 0/8
12.5 239 0/8 0/8
25 367 0/8 2/8
10i.maleate 25 185 0/8 0/8
37.5 260 1/8 1/8
MITOX 1.5 186(160-225) 6/26 1/26
3 173(142-205) 21/42 1/42
1) CDF1 mice were injected ip with 10 cells/mouse; treatment was given ip
on days 1,5,9 after tumor transplantation (day 0)
2) 10i.HCl and 10i.maleate were dissolved in sterile water and MITOX in saline.
3) Median survival time of treated mice/median survival time of controls × 100
4) No. of toxic deaths/ total No. of mice
5) Long term survivors it the end of the experiment (60 days).
Example 30
In vivo biological studies: L1210 Murine Leukemia (ip/ip, d 1, 5, 9)
a) L1210 murine leukemia cells were maintained in vivo by weekly intraperitoneal (ip) injections of 10 cells in BDF. mice. For test purposes, mice were inoculated i.p. with 10 L1210 cells and treatment was initiated 24 hours later. The desired dose of drug was administered on days 1, 5 and 9. Mice were observed daily for signs of toxicity and survival. The date of death was recorded for each animal that died or was sacrificed during the 60-day study. The mean survival time (MST) for each treatment group was calculated and the % T/C was determined using the following formula:
% T/C = [(MST treated)/(MST control)] × 100
The results are reported in table X
b) L1210 murine leukemia cells were maintained in vivo by weekly intraperitoneal (ip) injections of 10 cells in DBA2 mice. For test purposes, CDFl mice were inoculated i.p. with 10 L1210 cells and treatment was initiated 24 hours later. The desired dose of drug was administered on days 1, 5 and 9. Mice were observed daily for signs of toxicity and survival. The date of death was recorded for each animal that died or was sacrificed during the 60-day study. The median survival time (MST) for each treatment group was calculated and the % T/C was determined using the following formula:
% T/C = [(MST treated) /(MST control)] × 100
The results are reported in table XI TABLE X IN VIVO ANTITUMOR ACTIVITIES OF COMPOUNDS 10a, - 10i . HCl AND 101 AGAINST L1210 LEUKEMIA (ip/ip, d 1, 5, 9) IN COMPARISON TO THE PRIOR ART COMPOUND 5a.
Compound Example Dosea %T/C LTSf 10a(b) 4 50 151 -
10i.HCl(c) 12 25 265 5/6
12.5 265 4/6
101.maleate (d) 17 25 415 1/6
12.5 313 2/6 5a(e)'(b) / 50 129 -
(a) mg/kg, days 1, 5 and 9
(b) Compound 10a and 5a were dissolved in DMSO (<35%) and water
(c) Hydrochloride salt dissolved in water
(d) Dimaleate salt dissolved in water
(e) Prior art compound, 6,9-bis{[2-(dimethylamino)- ethyl]amino)benzo[g]quinoline-5,10-dione: A.P. Krapcho et al., J. Med. Chem., (1985), 28, 1124-1126.
(f) Long Term Survivors: animals tumor-free at the end of the experiment (60 days)
Table XI. ANTITUMOR ACTIVITY OF COMPOUND 10i.HCl AND MITOXANTRONE (MITOX) AGAINST
1)
ASCITIC MURINE LEUKEMIA L1210 (ip/ip d.1,5,9)
2) 3) 4) 5)
COMPOUNDS DOSE % T/C TOX LTS
(mg/kg/day)
10i.HCl 6.25 156 0/8 0/8
12.5 175 0/8 1/8
25 250* 1/7 4/7
MITOX 1.5 188 0/8 1/8
3 194 1/8 1/8
5
1) CDF1 mice were injected ip with 10 cells/mouse; treatment was given
ip on days 1,5,9 after tumor transplantation (day 0).
2) 10i.HCl was dissolved in sterile water and MITOX in saline.
3) Median survival time of treated mice/median survival time of controls x 100
4) No. of toxic deaths/ total No. of mice at the end of the experiment (60 days).
5) Long term survivors: animals tumor free at the end of the experiment ( 60 days). * This value was calculated on dead mice.
Example 31
In vivo biological studies: antitumor activity against murine Lewis Lung carcinoma and human MX-1 mammary carcinoma
a) Murine Lewis Lung carcinoma
C57bl/6 female mice were trasplantated im (intralimbs) with 10 cells. Treatment was given iv (intravenously) on days 1, 7, 15, after tumor transplantation (day 0). The mean tumor weight for each treatment group was calculated according to Geran, R.I. et al., Cancer Chemother. Rep., (1972), 3 , 51-61 and the TWI% was calculated 7 days after the last drug treatment using the formula:
TWI% = 100-[(Mean tumor weight of treated mice) / (mean tumor weight of controls)] × 100.
The results are reported in table XII for the representative compound of the invention 6, 9-bis {[2-(amino)ethyl]amino)benzo[g]isoquinoline-5,10-dione (10i) as the dimaleate salt (10i maleate) of the example 12.
b) Human MX-1 mammary carcinoma
CD1 nu/nu female mice were transplantated sc (subcutaneously) with tumor fragments (about 1 mm × 1 mm × 1 mm). Treatment was given iv once a week for three weeks, when tumor weight reached an average of 150 mg. For all individual tumors the weight change (relative tumor weight) for the start of treatment (Vo) was expressed as Vt/Vo at each day of measurement (Vt). TWI% was calculated 7 days after the last drug treatment using the formula:
TWI% = 100-[(mean relative tumor weight of treated mice) /(mean relative tumor weight of controls)] × 100. The results are reported in table XII for the representative compound of the invention 6,9-bis{[2-(amino)ethyl]amino)benzo[g]isoquinoline-5,10-dione (10i) as the dimaleate salt (10i maleate) of the example 12.

Claims

IN THE CLAIMS:
1. A compound according to formula (I) :
wherein R is
C1-C10 alkyl; phenyl or C7-C10 aralkyl;
C2-C10 aklyl substituted with one or two substituents selected from the group consisting of OR1 and -NR2R3;
C2-C10 alkyl interrupted by one or two oxygen atoms or by a member selected from the group consisting of -NR4-, cis -CH=CH, trans
-CH=CH- and -C≡C-, and optionally substituted with one or two hydroxy (OH) or -NR2R3 groups; and wherein
R1 is selected from the group consisting of hydrogen, C1-C6 alkyl, phenyl, C6-C10 aralkyl, -CHO, -COR5-, -COOR5, -S(O2)R5 and C2-C6 alkyl optionally substituted with -NR2R3;
R2 and R3 are the same or different and are selected from the group consisting of hydrogen, C1-C10 alkyl, C6-C10 aralkyl, phenyl, C2-C10 aikyl substituted with one or two hydroxy (OH) groups, -CHO, -COR5, -COOR5, and -S(O2)R5, R2 and R3 taken together with the nitrogen atom to which they are bound form an ethyleneimine ring or a 5- or 6-membered aromatic or non-aromatio heterocyclio ring optionally containing another heteroatom selected from the group consisting of sulfur, oxygen and nitrogen, R2 is H and R3 is -C(=NH)NH2 or R2 is -C(=NH)NH2 and R3 is H; R4 is selected from the groupconsisting of hydrogen, C1-C10 alkyl, C2-C10 hydroxyalkyl, C2-C10 alkyl substituted with -NR2R3, C6-C10 aralkyl, phenyl, -COR5, -COOR5 and -S(O2)R5;
R5 is selected from the group consisting of C1-C10 alkyl, C7-C10 aralkyl, α-, ß-, or γ-naphthyl, phenyl, o-, m-, or p-tolyl as free bases and their salts with pharmaceutically acceptable acids.
2. A compound according to claim 1, wherein R is -(CH2)p-NH2 and p is 2, 3 or 4.
3. A compound according to claim 1, wherein R is -(CH2)p-NR2R3 and p is 2, 3 or 4 and Wherein R2 and R3 are a C1-C6 alkyl or taken together with the nitrogen atom form a heterooyclic ring selected from the group consisting of 1-ethyleneimine, 1-pyrrolldine, 4-morpholine, 1-piperazine, 4-methyl-1-piperazine, 4-benzyl-1-piperazlne and 1-piperidine.
4. A compound according to claim 1, wherein R is -(CH2)p-MR2R3 and p is 2, 3 or 4 and wherein R2 is hydrogen and R3 is C1-C6 alkyl.
5. A compound according to claim 1, wherein R is and p is 2, 3 or 4.
6. A compound according to claim 1, wherein R is -(CH2)p-NH-(CH2)qOH and p and q are independently 2, 3 or 4.
7. A compound according to claim 1, wherein R is -(CH2)p-OH and p is 2, 3 or 4.
8. A compound according to claim 1, wherein R is -(CH2)p-o-(CH2)q-OH and p and q are independently 2, 3 or 4.
9. A compound according to claim 1 selected from the group consisting of:
6,9-bis{[2-(amino)ethyl]amino}benzo[g]isoquinoline-5,10-dione; 6,9-bis{[2-(4'-morpholino}ethyl]amino}benzo[g]isoguinoline-5,10- dione;
6,9-bis{[2-(dimethylamino}ethyl]amino}benzo[g]isoquinoline-5,10- dione;
6,9-bis{[2-(diethylamino}ethyl]amino}benzo[g]isoquinoline-5,10- dione;
6,9-bis{[2-(di(sec-propyl)amino]ethyl]amino}benzo[g]isoguinoline- 5,10-dione;
6,9-bis{[2-(1'-pyrrolidino}ethyl]amino}benzo[g]isoguinoline-5,10- dione;
6,9-bis{[2-(1,-aziridino}ethyl]amino}benzo[g]isoguinoline-5,10-dione;
6,9-bis([ 2-(methanesulfonyloxy)ethyl]amino}benzo[g]isoqulnoline-5,10-dione;
6,9-bis{[4-(amino}butyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[3-(amino}propyl]amino}benzo[g]isoquinoline-5,10-dione; 6,9-bis{[2-[(2-hydroxyethyl)amino]ethyl}amino}benzo[g]-isoquinoline-5,10-dionet 6 ,9-bis([3-(dimethylamino)propyl]amino)benzo[g]isoquinoline-5,10- dione;
6,9-bis{[(2-hydroxy)ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[(2-(2-hydroxyethoxy)ethyl]amino}benzo[g]isoquinoline-5,10- dione;
6,9-bis([2-(methylamino)ethyl]amino)benzo[g]isoquinoline-5,10- dione;
6,9-bis{[2-(ethylamino}ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-(n-propylamino}ethyl]amino}benzo[g]isoquinoline-5,10- dione;
6,9-bis([2-(sec-propylamino)ethyl]amino)benzo[g]isoquinoline-5,10- dione;
6,9-bis([2-(guanidino)ethyl]amino)benzo[g]isoqulnoline-5,10-dione;
6,9-bis([(2-amino-2,2-dimethyl)ethyl)amino)benzo[g]isoquinoline- 5,10-dione.
10. A compound according to claim 1 selected from the group consisting of;
6,9-bis{(2-(amino)ethyl]amino)benzo[g]isoquinoline-5,10-dione, 6,9-bis((2-(1'-pyrrolidino)ethyl]amino)benzo[g]isoquinoline-5,10-dione,
6,9-bis{[4-(amino)butyl]amino)benzo[g]isoquinollne-5,10-dione, 6,9-bis([3-(amino)propyl]amino)benzo[g]isoquinoline-5,10-dione, 6,9-bis([2-[(2-hydroxyethyl)aminojethyl]amino)benzo(g)-isoqulnoline-5,10-dione, and a salt of any one of said compounds.
11. A compound according to claim 1 which is 6,9-bis{[2- (amino}ethyl]amino}benzo(g]isoquinoline-5,10-dione or a salt thereof.
12. The compound according to claim 11, wherein said salt is a maleate salt.
13. The compound according to claim 11, wherein said salt is a hydrochloride salt.
14. A pharmaceutical composition comprising a compound according to claim 1 and a pharmaceutically acceptable diluent or exoipient.
15. A pharmaceutical composition comprising a compound according to claim 2 and a pharmaceutically acceptable diluent or exclpient.
16. A pharmaceutical composition comprising a compound according to claim 3 and a pharmaceutically acceptable diluent or excipient.
17. A pharmaceutical composition comprising a compound according to claim 4 and a pharmaceutically acceptable diluent or excipient.
18. A pharmaceutical composition comprising a compound according to claim 5 and a pharmaceutically acceptable diluent or excipient.
19. A pharmaceutical composition comprising a compound according to claim 6 and a pharmaceutically acceptable diluent or excipient.
20. A pharmaceutical composition comprising a compound according to claim 7 and a pharmaceutically acceptable diluent or excipient.
21. A pharmaceutical composition comprising a compound according to claim 8 and a pharmaceutically acceptable diluent or excipient.
22. A pharmaceutical composition comprising a compound selected from the group consisting of!
6,9-bis{[2-(amino}ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-(4'-morpholino}ethyl]amino}benzo[g]isoquinoline-5,10-dioney
6,9-bis{[2-(dimethylamino}ethyl]amino}benzo[g]lsoquinoline-5,10-dione;
6,9-bis{[2-(diethylamino}ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-tdi(seo-propyl]amino]ethyl]amino}benzo(g]isoquinoline- 5,10-dione; 6,9-bis((2-(1'-pyrrolidino)ethyl]amino}benzo[g]isoquinoline-5,10- dione;
6,9-bis({2-(1'-aziridino)ethyl]amino}benzo[g]isoquinoline-5,10- dione.
6,9-bis{[2-(methanesulfonyloxy)ethyl]amino}benzo[g]isoquinoline- 5,10-dione;
6,9-bis([4-(amino)butyl]amino)benzo[g]isoquinoline-5,10-dione; 6,9-bis([3-(amino)propyl]amino)benzo[g]isoquinoline-5,10-dione. 6,9-bisf [2-[(2-hydroxyethyl)amino]ethyl]amino)benzo[g]- isoquinoline-5,10-dione;
6,9-bis({3-(dimethylamino)propyl]amino)benzo[g]isoquinoline-5,10- dione;
6,9-bis([(2-hydroxy)ethyl]amino)benzo[g]isoquinoline-5,10-dione;
6,9-bis([(2-(2-hydroxyethoxy)ethyl]amino]benzo[g]isoquinoline-5,10- dione;
6,9-bis{[2-(methylamino}ethyl]amino)benzo[g]isoquinoline-5,10- dione;
6,9-bis({2-(ethylamino}ethyl]amino)benzo[g]isoqulnoline-5,10-dione;
6,9-bis{[2-(n-propylamino}ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-(sec-propylamino}ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis{[2-(guanidino}ethyl]amino]benzo(g)isoquinoline-5,10-dione,
6,9-bis{((2-amino-2,2-dimethyl)ethyl]amino}benzofg]isoqulnoline- 5,10-dione; and
a pharmaceutically acceptable diluent or excipient.
23. A pharmaceutical composition comprising a compound selected from the group consisting of:
6,9-bis([ 2-(amino}ethyl]amino}benzo[g]isoquinoline-5,10-dione, 6,9-bis{[2-(1'-pyrrolidino}ethyl]amino}benzo[g]isoquinoline-5,10 dione,
6,9-bis{(4-(amino}butyl]amino}benzo[g]isoquinoline-5,10-dione, 6,9-bis{[3-(amino}propyl]amlno}benzo[g]isoquinoline-5,10-dione, 6,9-bis{[2-[(2-hydroxyethyl)aminojethyl]amino}benzo[g]- isoquinoline-5,10-dione, and a salt of any one of said compounds; and a pharmaceutically acceptable diluent or excipient.
24. A pharmaceutical composition comprising 6,9-bis{[2- (amino}ethyl]amino}benzo[g]isoquinoline-5,10-dione or a sal thereof and a pharmaceutically acceptable diluent or excipient.
25. The pharmaceutical composition according to claim 24, wherein said salt is a maleate salt.
26. The pharmaceutical composition according to claim 24, wherein said salt is a hydrochloride salt.
27. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound according to claim 1.
28. The method according to claim 27, wherein said compound is administered daily in a dosage range of about 1 mg to about 50 mg per kilogram of body weight.
29. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound according to claim 2.
30. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound according to claim 3.
31. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound according to claim 4.
32. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound according to claim 5.
33. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound according to claim 6.
34. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound according to claim 7.
35. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound according to claim 8.
36. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound selected from the group consisting of;
6,9-bis([2-(amino}ethyl]amino}benzo[g]isoquinoline-5,10-dione;
6,9-bis((2-(4'-morpholino}ethyl]amlno}benzo[g]isoquinoline-5,10- dione;
6,9-bis((2-(dimethylamino}ethyl]amino}benzo[g]isoquinoline-5,10- dione;
6,9-bis((2-(diethylamino}ethyl]amino}benzo[g]isoquinoline-5,10- dione;
6,9-bis{[2-(di(sec-propyl)amino]ethyl]amino}benzo[g]isoquinoline- 5,10-dione;
6,9-bis((2-(1'-pyrrolidino}ethyl]amino}benzo[g]isoqulnoline-5,10-dione;
6,9-bls((2-(1'-aziridino}ethyl]amino]benzo[g]isoquinoline-5,10-dlone;
6,9-bls((2-(methanesulfonyloxy)ethyl]amino}benzo[g]isoquinoline- 5,10-dione;
6,9-bis([4-(amino)butyl]amino)benzo[g]isoquinoline-5,10-dlonef
6,9-bis{(3-(amlno)propyl]amino)benzo[g]isoqulnoline-5,10-dione;
6,9-bis([2-((2-hydroxyethyl)aminojethyl]amino]benzo[g]-isoquinoline-5,10-dionet 6,9-bis((3-(dimethylamino}propyl]amino}benzo[g]isoquinoline-5,10- dione;
6,9-bis([(2-hydroxy)ethyl]amino]benzo[g]isoquinoline-5,10-dione;
6,9-bis{((2-(2-hydroxyethoxy)ethyl]amino]benzo[g]isoquinoline-5,10- dione;
6,9-bis((2-(methyiamino)ethyl]amino)benzo[g]isoquinoline-5,10- dione;
6,9-bls{{2-(ethylamino)ethyl]amino)benzo[g]isoquinoline-5,10-dionef
6,9-bis((2-(n-propylamino)ethyl]amino)benzo[g]isoquinoline-5,10- dione.
6,9-bis([2-(sec-propylamino)ethyl]amino)benzo[g]isoquinoline-5,10- dione;
6,9-bis([2-(guanidino)ethyl]amino)benzo[g]isoquinoline-5,10-dione;
6,9-bls([(2-amino-2,2-dimethyl)ethyl]amino]benzo[g]isoquinoline- 5,10-dione.
37. A method of treatment of tumors in a mammal requiring such treatment comprising administration of an effective anti-tumor amount of a compound selected from the group consisting of!
6,9-bis((2-(amino)ethyl]amino]benzo[g]isoquinoline-5,10-dione, 6,9-bis((2-(1'-pyrrolidino)ethyl]amino)benzo[g]isoquinoline-5,10-dione,
6,9-bis((4-(amlno)butyl]amino)benzo[g]isoquinoline-5,10-dione, 6,9-bis([3-(amino)propyl]amino)benzo[g]isoquinoline-5,10-dione, 6,9-bls{(2-[(2-hydroxyethyl)amino]ethyl]amino)benzo[g]-isoquinoline-5,10-dione, and a salt of any one of said compounds. such treatment comprising administration of an effective anti-tumor amount of 6,9-bis((2-(amino}ethyl]amino}benzotg]isoquinollne-5,10- dione or a salt thereof.
39. The method according to claim 38, wherein said salt is a maleate salt.
40. The method according to claim 38, wherein said salt is a hydrochloride salt.
41. A process for the synthesis of a compound according to formula I.
wherein R is
C1-C10 alkyl; phenyl or C7-C10 aralkyl;
C1-C10 aklyl substituted with one or two substituents selected from the group consisting of OR1 and -NR2R3;
C1-C10 aklyl interrupted by one or two oxygen atoms or by a member selected from the group consisting of -NR4-, cis -CH=CH, trans
-CH=CH- and -C≡C-, and optionally substituted with one or two hydroxy (OH) or -MR2R3 groups; and wherein R1 s selec ed from e group cons s ng of y rogen, C1-C6 alkyl, phenyl, C7-C10 aralkyl, -CHO, -COR5-, -COOR5, -S(O2)R5 and C2-C6 alkyl optionally substituted with -HR2R3;
R2 and R3 are the same or different and are selected from the group consisting of hydrogen, C1-C10 alkyl, C7-C10 aralkyl, phenyl, C2-C10 aklyl substituted with one or two hydroxy (Off) groups, -CHO,
-COR5, -COOR5, and -S(O2)R5, R2 and R3 taken together with the nitrogen atom to which they are bound form an ethyleneimine ring or a 5- or 6-membered aromatic or non-aromatic heterocyclic ring optionally containing another heteroatom selected from the group consisting of sulfur, oxygen and nitrogen, R2 is If and R3 is
-C(=NH)NH2 or R2 is -C(=NH)NH2 and R3 is H.
R4 is selected from the group consisting of hydrogen, C1-C10 alkyl, C2-c10 hydroxyalkyl, C2-C10 alkyl substituted with -MR2R3, C7-C10 aralkyl, phenyl, -COR5, -COOR5 and -S(O2)R5;
R5 is selected from the group consisting of C1-C10 alkyl, C7-C10 aralkyl, or-, ß-, or γ-naphthyl, phenyl, o-, m-, or p-tolyl as free bases and their salts with pharmaceutically acceptable acids, comprising the steps ofs
a) reacting pyridine -3,4-diearboxylic anhydride with 1,4-difluorobenzene;
b) cyclizing the product of step a) in fuming sulfuric acid at elevated temperature; and
c) displacement of fluoride by addition of amine of formula R'-HH2, wherein R' is the same as that defined by R. obtained in step (c) has the formula (la):
and wherein R' has a meaning other than that defined by R and further comprising the step of converting R' to R.
43. The process according to claim 41, further comprising the step of converting R' in the compound obtained in step (c) to another compound defined by R.
44. The process according to claim 4l, further comprising the step of subjecting the compound obtained in step (c) to saliflcation and/or solvation or separating isoraers thereof.
45. A process for the synthesis of a compound according to formula I :
wherein R is
C1-C10 alkyl; phenyl or C7-C10 aralkyl;
C2-C10 aklyl substituted with one or two substituents selected from the group consisting of OR1 and -NR2R3;
C2-C10 aklyl interrupted by one or two oxygen atoms or by a member selected from the group consisting of -MR4-, cis -CH=CH, trans
-CH=CH- and -C≡C-, and optionally substituted with one or two hydroxy (OH) or -HR2R3 groups; and wherein
R1 is selected from the group consisting of hydrogen, C1-C6 alkyl, phenyl, C6-C10 aralkyl, -CHO, -COR5-, -COOR5, -S(O2)R5 and C2-C8 alkyl optionally substituted with -HR2R3;
R2 and R3 are the same or different and are selected from the group consisting of hydrogen, C1-C10 alRyl, C7-C10 aralkyl, phenyl, C1-C10 aklyl substituted with one or two hydroxy (OH) groups, -CHO,
-COR5, -COOR5, and -S(O2)R5, R2 and R3 taken together with the nitrogen atom to which they are bound form an ethyleneimine ring or a 5- or 6-membered aromatic or non-aromatic heterocycllc ring optionally containing another heteroatom selected from the group consisting of sulfur, oxygen and nitrogen, R2 is H and R3 is
-C(=NH)NH2 or R2 is -C(=NH)NH2 and R3 is If.
R4 is selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 hydroxyalkyl, C2-C10 alkyl substituted with -NR2R3, C7-C10 aralkyl, phenyl, -COR5, -COOR5 and -S(O2)R5;
R5 is selected from the group consisting of C1-C10 alkyl, C6-C10 aralkyl, α-, β-, or γ-naphthyl, phenyl, o-, m-, or p-tolyl as free bases and their salts with pharmaceutically acceptable acids, comprising the step oft reacting a compound of formula (I), wherein R is -(CH2)p-OS(O2)R5 and wherein R5 is as previously defined and p is 2, 3 or 4, with a compound having the formula H2N-(CH2)q-O-E, wherein E is selected from the group consisting of trialkylsilane, (dialkyl)arylsilane, formyl and aoetyl, and q is 2, 3 or 4.
46. The process according to claim 45, further comprising the step of removing the E group.
47. A process for the synthesis of a compound according to formula I.
wherein R is
C1-C10 alkyl; phenyl or C7-C10 aralkyl;
C2-C10 aklyl substituted with one or two substituents selected from the group consisting of OR1 and -NR2R3;
C2-C10 alkyl interrupted by one or two oxygen atoms or by a member selected from the group consisting of -NR4-, cis -CH=CH, trans
-CH=CH- and -C≡C-, and optionally substituted with one or two hydroxy (OH) or -NR2R3 groups; and wherein R1 is selected from the group consisting of hydrogen, C1-C6 alkyl, phenyl, C7-C10 aralkyl, -CHO, -COR5-, -COOR5, -S(O2)R5 and C2-C8 alkyl optionally substituted with -NR2R3;
R2 and R3 are the same or different and are selected from the group consisting of hydrogen, C1-C10 alkyl, C7-C10 aralkyl, phenyl,
C2-C10 alkyl substituted with one or two hydroxy (OH) groups, -CHO,
-COR5, -COOR5, and -S(O2)R5, R2 and R3 taken together with the nitrogen atom to which they are bound form an ethyleneimine ring or a 5- or 6-membered aromatic or non-aromatic heterocycllc ring optionally containing another heteroatom selected from the group consisting of sulfur, oxygen and nitrogen, R2 is If and R3 is
-C(=NH)NH2 or R2 is -C(=NH)NH2 and R3 is H;
R4 is selected from the group consisting of hydrogen,. C1-C10 alkyl,
C2-C10 hydroxyalkyl, C2-C10 alkyl substituted with -HR2R3, C7-C10 aralkyl, phenyl, -COR5, -COOR5 and -S(O2)R5;
R5 is selected from the group consisting of C1-C10 alkyl, C7-C10 aralkyl, o-, ß-, or γ-naphthyl, phenyl, o-, m-, or p-tolyl as free bases and their salts with pharmaceutically acceptable acids, comprising the steps of!
a) reacting 6,9-difluorobenzo(gjisoqulnoline-5,10-dione with a compound having the formula H2N-(CH2)p-N(COOR5)R3,
wherein R3 is hydrogen or a C1-C6 alkyl, R5 is as previously defined and p is 2, 3 or 4 to obtain a compound having the formula:
wherein R ' is - ( CH2 )p-N(COOR5 )R3 ;
b) removing the group COOR5.
48. A process for the synthesis of a compound according to formula I :
wherein R is
C1-C10 alkyl; phenyl or C7-C10 aralkyl;
C2-C10 aklyl substituted with one or two substituents selected from the group consisting of OR1 and -NR2R3;
C2-C10 alkyl interrupted by one or two oxygen atoms or by a member selected from the group consisting of -HR4-, cis -CH=CH, trans -CH=CH- and -C≡C-, and optionally substituted with one or two hydroxy (OH) or -MR2R3 groups; and wherein
R1 is selected from the group consisting of hydrogen, C1-C6 alkyl, phenyl, C7-C10 aralkyl, -CHO, -COR5-, -COOR5, -S(O2)R5 and C2-C6 alkyl optionally substituted with -HR2R3;
R2 and R3 are the same or different and are selected from the group consisting of hydrogen, C1-C10 alkyl, C7-C10 aralkyl, phenyl, C2-C10 alkyl substituted with one or two hydroxy (Off) groups, -CHO, -COR5, -COOR5, and -S(O2)R5, R2 and R3 taken together with the nitrogen atom to which they are bound form an ethyleneimine ring or a 5- or 6-membered aromatic or non-aromatio heterocycllc ring optionally containing another heteroatom selected from the group consisting of sulfur, oxygen and nitrogen, R2 is H and R3 is
-C(=NH)NH2 or R2 is -C(=NH)NH2 and R3 is H;
R4 is selected from the group consisting of hydrogen, C1-C10 alkyl,
C2-C10 hydroxyalkyl, C2-C10 alkyl substituted with -NR2R3, C7-C10 aralkyl, phenyl, -COR5, -COOR5 and -S(O2)R5;
R5 is selected from the group consisting of C1-C10 alkyl, C7-C10 aralkyl, α-, β-, or γ-naphthyl, phenyl, o-, m-, or p-tolyl as free bases and their salts with pharmaceutically acceptable acids, comprising the step of:
reacting a compound of formula (I), wherein R is (CH2)p-NH2 and p is 2, 3 or 4 with a compound having the formula H2N-C(=NH)SO3H.
EP92908816A 1991-03-08 1992-03-09 6,9 BIS(SUBSTITUTED-AMINO)BENZO g]ISOQUINOLINE-5,10-DIONES Pending EP0575526A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US66595491A 1991-03-08 1991-03-08
US665954 1991-03-08
US82730292A 1992-01-29 1992-01-29
US827302 1992-01-29

Publications (2)

Publication Number Publication Date
EP0575526A1 true EP0575526A1 (en) 1993-12-29
EP0575526A4 EP0575526A4 (en) 1994-03-30

Family

ID=27099336

Family Applications (2)

Application Number Title Priority Date Filing Date
EP92908816A Pending EP0575526A1 (en) 1991-03-08 1992-03-09 6,9 BIS(SUBSTITUTED-AMINO)BENZO g]ISOQUINOLINE-5,10-DIONES
EP92104004A Expired - Lifetime EP0503537B1 (en) 1991-03-08 1992-03-09 6,9 Bis(substituted-amino)benzo-[g]isoquinoline-5,10-diones

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP92104004A Expired - Lifetime EP0503537B1 (en) 1991-03-08 1992-03-09 6,9 Bis(substituted-amino)benzo-[g]isoquinoline-5,10-diones

Country Status (22)

Country Link
EP (2) EP0575526A1 (en)
JP (1) JP3009465B2 (en)
KR (1) KR100193593B1 (en)
AT (1) ATE153018T1 (en)
AU (1) AU663494B2 (en)
BR (1) BR9205740A (en)
CA (1) CA2104582A1 (en)
CZ (1) CZ286180B6 (en)
DE (1) DE69219646T2 (en)
DK (1) DK0503537T3 (en)
ES (1) ES2104753T3 (en)
FI (1) FI103575B (en)
GR (1) GR3024436T3 (en)
HU (1) HU215967B (en)
IE (1) IE920754A1 (en)
IL (1) IL102087A (en)
MX (1) MX9201020A (en)
NZ (1) NZ241868A (en)
RU (1) RU2129546C1 (en)
SG (1) SG66252A1 (en)
TW (1) TW201735B (en)
WO (1) WO1992015300A1 (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ZA936396B (en) * 1992-09-08 1994-06-13 Univ Vermont 4-Substituted 6,9-bis(substituted-amino)benzo[g] isoquinoline-5,10,diones
US5519029A (en) * 1992-09-08 1996-05-21 Boehringer Mannheim Italia, S.P.A. 2-aminoalkyl-5-aminoalkylamino substituted-isoquinoindazole-6(2H)-ones
US5587382A (en) * 1994-03-28 1996-12-24 Boehringer Mannheim Italia, Spa 6,9-bis[(2-aminoethyl) amino]benzo [g]isoquinoline-5,10- dione dimaleate; an aza-anthracenedione with reduced cardiotoxicity
US5506232A (en) * 1994-03-28 1996-04-09 Boehringer Mannheim Italia S.P.A. 6,9-bis[(2-aminoethyl)amino]benzo[g]isoquinoline-5,10-dione and its dimaleate salt
US6747039B2 (en) 2002-03-12 2004-06-08 Albany Molecular Research, Inc. Aza-benzothiopyranoindazoles with antitumor activity
ITMI20021040A1 (en) 2002-05-16 2003-11-17 Novuspharma Spa INJECTABLE PHARMACEUTICAL COMPOSITIONS OF AN ANTHROCENEDIONAL DERIVATIVE WITH ANTI-TUMOR ACTIVITY
US20050222190A1 (en) * 2004-03-30 2005-10-06 Curd John G 1,4-bis-N-oxide azaanthracenediones and the use thereof
WO2008103320A1 (en) * 2007-02-16 2008-08-28 Novacea, Inc. Methods of treating ophthalmic disorders with anthraquinones
US8173621B2 (en) 2008-06-11 2012-05-08 Gilead Pharmasset Llc Nucleoside cyclicphosphates
TW201031675A (en) 2008-12-23 2010-09-01 Pharmasset Inc Synthesis of purine nucleosides
CN102753563A (en) 2008-12-23 2012-10-24 吉利德制药有限责任公司 Nucleoside analogs
NZ593648A (en) 2008-12-23 2013-09-27 Gilead Pharmasset Llc Nucleoside phosphoramidates
CN102858790A (en) 2010-03-31 2013-01-02 吉利德制药有限责任公司 Nucleoside Phosphoramidates
CN104513201B (en) * 2013-09-28 2017-12-26 正大天晴药业集团股份有限公司 Crystal of pixantrone maleate
CN104557704B (en) * 2013-10-28 2017-05-10 北京凯莱天成医药科技有限公司 Preparation method of pixantrone maleate
CN103787970A (en) * 2014-01-14 2014-05-14 北京万全德众医药生物技术有限公司 Technology for preparing 6,9-difluoro benzisothiazol-5,10-diketone employing one-pot method

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2000029A (en) * 1977-07-22 1979-01-04 Allied Chem Anthraquinones for Cancer Therapy
US4197249A (en) * 1977-08-15 1980-04-08 American Cyanamid Company 1,4-Bis(substituted-amino)-5,8-dihydroxyanthraquinones and leuco bases thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2051005A (en) * 1933-09-19 1936-08-11 Gen Aniline Works Inc Process of producing n-substitution products of 1, 4-diaminoanthraquinones
US2552263A (en) * 1947-09-27 1951-05-08 Eastman Kodak Co alpha-anthrapyridinequinone compounds
EP0270306B1 (en) * 1986-12-01 1991-03-27 Sumitomo Chemical Company, Limited Anthrapyridone compounds, their production process and their use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2000029A (en) * 1977-07-22 1979-01-04 Allied Chem Anthraquinones for Cancer Therapy
US4197249A (en) * 1977-08-15 1980-04-08 American Cyanamid Company 1,4-Bis(substituted-amino)-5,8-dihydroxyanthraquinones and leuco bases thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY vol. 23, no. 1 , January 1988 , PARIS FR pages 101 - 106 M. CROISY-DELCEY ET AL 'Aminoalkylamino derivatives of dihydroxy-benzoÄgÜisoquinoline dione and of trihydroxy-naphtho[2,3-gÜisoquinoline dione:synthesis and anti-tumor evaluation' *
See also references of WO9215300A1 *

Also Published As

Publication number Publication date
HU215967B (en) 1999-03-29
FI103575B1 (en) 1999-07-30
SG66252A1 (en) 1999-07-20
ATE153018T1 (en) 1997-05-15
TW201735B (en) 1993-03-11
KR100193593B1 (en) 1999-06-15
WO1992015300A1 (en) 1992-09-17
AU1580392A (en) 1992-10-06
JPH06511230A (en) 1994-12-15
HUT68649A (en) 1995-07-28
FI933895A (en) 1993-09-07
EP0503537A1 (en) 1992-09-16
FI933895A0 (en) 1993-09-07
CZ184793A3 (en) 1994-06-15
BR9205740A (en) 1994-09-27
CA2104582A1 (en) 1992-09-17
DE69219646T2 (en) 1997-10-02
NZ241868A (en) 1995-05-26
DK0503537T3 (en) 1997-12-15
CZ286180B6 (en) 2000-02-16
IE920754A1 (en) 1992-09-09
RU2129546C1 (en) 1999-04-27
AU663494B2 (en) 1995-10-12
GR3024436T3 (en) 1997-11-28
IL102087A (en) 1996-09-12
JP3009465B2 (en) 2000-02-14
ES2104753T3 (en) 1997-10-16
MX9201020A (en) 1993-11-01
DE69219646D1 (en) 1997-06-19
FI103575B (en) 1999-07-30
EP0575526A4 (en) 1994-03-30
EP0503537B1 (en) 1997-05-14

Similar Documents

Publication Publication Date Title
EP0503537B1 (en) 6,9 Bis(substituted-amino)benzo-[g]isoquinoline-5,10-diones
KR20000036102A (en) Substituted benzocycloheptapyridine useful as inhibitors of farnesyl-protein transferase
HU217551B (en) 6-[(2-hydroxyethyl)-amino-alkyl]-5,11-dioxo-5,6-dihydro-11-h-indeno(1,2-c)isoquinolin derivatives, process for producing them and pharmaceutical compositions containing them
AU680431B2 (en) 2,5-disubstituted isoquindazole-6-(2H)-ones
US5519029A (en) 2-aminoalkyl-5-aminoalkylamino substituted-isoquinoindazole-6(2H)-ones
AU8050191A (en) 1,2-dihydro-3h-dibenzisoquinoline-1,3-dione anticancer agents
Yamato et al. Synthesis and Antitumor Activity of Fused Quinoline Derivatives. II. Novel 4-and 7-Hydroxyindolo-[3, 2-b] quinolines
US5763453A (en) Condensed indan derivatives and salts thereof
EP0639183B1 (en) Nitrogen oxides of aza- and diaza-anthracenedione derivatives, their preparation and their use as antitumor agents
US5604246A (en) 2-aminoalkyl-5-aminoalkylamino substituted-isoquinoindazole-6(2H)-ones
CA2143853C (en) 2-aminoalkyl-5-aminoalkylamino substituted isoquinoindazole-6-(2h)-ones with antitumour activity
NO180302B (en) 6,9-bis (substituted-amino) benzo [gÅisokinolin-5,10-diones
WO1994005641A1 (en) 4-SUBSTITUTED 6,9,-BIS (SUBSTITUTED-AMINO) BENZO [g] ISOQUINOLINE-5,10,DIONES
JP2001181281A (en) Quinolone derivative useful as antimicrobial agent
大和正利 et al. Synthesis and Antitumor Activity of Fused Quinoline Derivatives. II. Novel 4-and 7-Hydroxyindolo (3, 2-b) quinolines.
WO1991006540A1 (en) 1,4-bis(alkylamino)-2,3-diaza-anthracene-9,10-diones
WO1992015307A1 (en) 6,9-BIS(AMINO SUBSTITUTED)BENZO[g]PHTHALAZINE-5,10-DIONES AS ANTITUMOR AGENTS
WO1998025910A1 (en) NEW DERIVATIVES OF 6-[(AMINOALKYL)AMINO]-7H-BENZO[e]-PERIMIDIN-7-ONE, METHOD OF THEIR PREPARATION, AND THEIR USE AS A MEDICAMENT

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: 19930908

AK Designated contracting states

Kind code of ref document: A1

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

RHK1 Main classification (correction)

Ipc: C07D221/08

A4 Supplementary search report drawn up and despatched

Effective date: 19940210

AK Designated contracting states

Kind code of ref document: A4

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

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

Free format text: STATUS: PROCEEDINGS CLOSED FOLLOWING CONSOLIDATION WITH EP92104004.4

XX Miscellaneous (additional remarks)

Free format text: VERFAHREN ABGESCHLOSSEN INFOLGE VERBINDUNG MIT 92104004.4/0503537 (EUROPAEISCHE ANMELDENUMMER/VEROEFFENTLICHUNGSNUMMER) DURCH ENTSCHEIDUNG VOM 28.08.95.

RTI1 Title (correction)

Free format text: 6,9 BIS(SUBSTITUTED-AMINO)BENZO G ISOQUINOLINE-5,10-DIONES