US20100331355A1 - Substituted Quinolines for the Treatment of Cancer - Google Patents

Substituted Quinolines for the Treatment of Cancer Download PDF

Info

Publication number
US20100331355A1
US20100331355A1 US12/760,408 US76040810A US2010331355A1 US 20100331355 A1 US20100331355 A1 US 20100331355A1 US 76040810 A US76040810 A US 76040810A US 2010331355 A1 US2010331355 A1 US 2010331355A1
Authority
US
United States
Prior art keywords
indolo
quinoline
radical
carboxamide
methyl
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.)
Abandoned
Application number
US12/760,408
Inventor
Juan Aymami Bofarull
Miquel Coll Capella
Amadeo Llebaria Soldevila
Isabel Navarro Muñoz
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.)
Consejo Superior de Investigaciones Cientificas CSIC
Universitat Politecnica de Catalunya UPC
Oryzon Genomics SA
Original Assignee
Consejo Superior de Investigaciones Cientificas CSIC
Universitat Politecnica de Catalunya UPC
Crystax Pharmaceuticals SL
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 Consejo Superior de Investigaciones Cientificas CSIC, Universitat Politecnica de Catalunya UPC, Crystax Pharmaceuticals SL filed Critical Consejo Superior de Investigaciones Cientificas CSIC
Priority to US12/760,408 priority Critical patent/US20100331355A1/en
Publication of US20100331355A1 publication Critical patent/US20100331355A1/en
Assigned to UNIVERSITAT POLITENCNICA DE CATALUNYA, CRYSTAX PHARMACEUTICALS, S.L., CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS reassignment UNIVERSITAT POLITENCNICA DE CATALUNYA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AYMAMI BOFARULL, JUAN, COLL CAPELLA, MIQUEL, LLEBARIA SOLDEVILA, AMADEO, NAVARRO MUNOZ, ISABEL
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to new compounds which intercalate between DNA base pairs. Such compounds can be used for the treatment of cancer.
  • Intercalators are a group of compounds that bind between the DNA base pairs. This intercalation can produce an interruption of transcription, replication and/or topoisomerase activity. It has been found that many compounds that bind to DNA by intercalation have antiproliferative properties and in vivo antitumour effects. Intercalators have been used as anticancer drugs. Cryptolepine is a naturally occurring alkaloid and it is used in traditional medicine against malaria. Its formula, shown below, is structurally close to some compounds of the present invention. Cryptolepine is able to bind CG-rich sequences of DNA containing non-alternating CC sites, and it has been reported as potential anticancer drug (cf. e.g. J. N.
  • -G 1 is a radical selected from (IIa) y (IIb);
  • R′ is an electron pair or a (C 1 -C 3 )-alkyl radical
  • radicals (IIa) and (IIb) are respectively (IIa′) and (II′), and
  • radicals (IIa) and (IIb) are respectively (IIa′′) and (IIb′′);
  • —R 1 to —R 12 represent radicals, same or different, selected from the group consisting of H, (C 1 -C 4 )-alkyl, (C 1 -C 4 )-alkoxy, (C 1 -C 4 )-alkylamino, phenyl, F, Cl, Br, amino, hydroxy, and nitro;
  • —B— is a biradical selected from the group consisting of —CONH—
  • (C 1 -C 4 )-alkylamino; —R′′ are side chains radicals, same or different, corresponding to natural aminoacids; n is an integer from 1 to 3; m is an integer from 1 to 3; u is an integer from 1 to 3, and —Z— is a biradical of a oligonucleotide phosphate between 4 and 23 bases in length, linked to the methylene group at the 5′ end or at the 3′ end;
  • -L- is a single covalent bond or a covalent linking biradical selected from the following ones;
  • R′′′ and —R′′′′ are radicals, same or different, selected from the group consisting of H and (C 1 -C 3 )-alkyl; r is an integer from 1 to 3; s is an integer from 1 to 3; t is an integer from 1 to 3; and
  • -G 2 is a radical selected from H, a radical of formula (IIa), a radical of formula (IIb), the N-radical of 1,8-naphthalimide, the C4-radical of 2-phenylquinoline, and the C9-radical of acridine;
  • compounds of formula (I) are those where (IIa) is the radical (IIa′).
  • Preferred compounds are those of formula (I) where (IIa) is (IIa′) and —B— is
  • —Z— is -TTCCGGAA- linked at 3′ end or at 5′ end, or -CTTCTTCTTCT- linked at 3′ end.
  • -L- is a single covalent bond.
  • -L- is a covalent linking biradical selected from the following ones.
  • compounds of formula (I) are those where (IIa) is the following radical.
  • Preferred compounds are those where (IIa) is (IIa′′) and —B— is —CONH— or
  • the most preferred compounds of formula (I) are those of the following list, whose preparation is described in the accompanying examples for the first time: N-[3-[[3-[(9-acridinecarbonyl)amino]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ia); N,N′-(4-methyl-4-azaheptamethylene)-di-(10H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Ib);
  • Another aspect of the present invention refers to the use of the compound of formula (I), defined above, for the preparation of a medicament for the treatment of cancer.
  • a further aspect of the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising as active ingredient a therapeutically effective amount of the compound of formula (I), defined above, together with suitable pharmaceuticalexcipients or carriers.
  • the 11-aminocarboxylic radical selected from formula (IIa) or the 4-aminocarboxylic radical selected from formula (IIb) is coupled by benzotriazol-1-yloxytripyrrolidinophosphonium hexafluoro-phosphate (PyBOP) and diisopropylamine, followed by acidolysis.
  • PyBOP benzotriazol-1-yloxytripyrrolidinophosphonium hexafluoro-phosphate
  • Compounds of formula (I) may be converted into pharmaceutically acceptable acid addition salts, and salts may be converted into free compounds, by conventional methods.
  • the acid addition salts may be prepared by contacting the free base with an appropriate amount of the desired acid in a conventional manner.
  • Compounds of formula (I) are intercalators (i.e. compounds which intercalate between DNA base pairs) and, as illustrated by biological results in example 30 are active for cancer treatment.
  • 9-acridinecarboxylic acid (0.036 g, 0.16 mmol) was dissolved in 3 ml of refluxing SOCl 2 . After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 4 ml of CH 2 Cl 2 . A solution of 10H-indolo[3,2-b]quinoline-[3-[(3-aminopropyl)methyl-amino]propyl]-11-carboxamide (0.076 g, 0.19 mmol) and triethylamine (0.2 ml) in 4 ml of CH 2 Cl 2 was added at room temperature, and the reaction mixture was stirred for 14 hours.
  • 1,8-naphthalic anhydride (0.18 g, 0.9 mmol) was added to a solution of [3-[(3-aminopropyl)methylamino]propyl]carbamic acid tert-butyl ester previously prepared (Spicer at el., Bioorg. Med. Chem. 2002, vol. 10, p. 19) (0.2 g, 0.8 mmol) in absolute ethanol (20 ml). The reaction mixture was heated at reflux overnight.
  • 9-acridinecarboxylic acid (0.15 g, 0.67 mmol) was dissolved in 15 ml of refluxing SOCl 2 . After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 15 ml of CH 2 Cl 2 . A solution of 10H-indolo[3,2-b]quinoline-[2-[[3-[(2-aminoethyl)methylamino]propyl]methylamino]ethyl]-11-carboxamide (0.4 g, 0.92 mmol) and triethylamine (1 ml) in 15 ml of CH 2 Cl 2 was added at room temperature. The reaction mixture was stirred for 14 hours.
  • the 5-methyl-5H-indolo[3,2-b]quinoline-11-carboxylic acid is known in the literature as part of extract plant Cryptolepis sanguinolenta (cf. Paulo et al., Planta Medica 2000, vol. 66, p. 30). It can also be prepared by the following process:
  • the compound of the title was prepared by solid phase methods. After assembling the Boc-glicine-arginine sequence on a p-methylbenzhydrylamine resin, the N-terminal was deprotected and 10H-indolo[3,2-b]quinoline-11-carboxylic acid was coupled by means of benzotriazol-1-yloxytripyrrolidinophosphonium hexafluoro-phosphate (PyBOP) and diisopropylethylamine (5, 5 and 10 equivalents, respectively) in 1:1 DMSO-DMF for 1 h. Acidolysis with HF/anisole (9:1, 0° C., 1 h) provided the desired compound in highly homogeneous form (>98% by HPLC). MS(MALDI-TOF): EM(expected): 474.1; EM(found): 475.5 (M+H + ).
  • 5′TTCCGGAA-NH 2 -3′ oligonucleotide sequence carrying an amino group at the 3′ end was prepared using 3-O-dimethoxytrityloxy-2-(N-9-fluorenphenylmethoxycarbonyl-4-aminobutyl)-1-propanyl ester succinamidyl-controlled pore glass (3′ amino C7 modifier CPG from Glen Research).
  • the solid support obtained after the assembly of the sequence was treated with a 0.1 M solution of 1,8-diazabiciclo[5.4.0]undece-7-ene (DBU) in dry acetonitrile (2 min, room temperature). In this way, the Fmoc group that protects the amino group is removed selectively with a non-nucleophillic base.
  • DBU 1,8-diazabiciclo[5.4.0]undece-7-ene
  • 3′ oligonucleotide sequence carrying an amino group at the 3′ end was prepared using the 3′ amino C7 modifier CPG (Glen Research).
  • the solid support obtained after the assembly of the sequence was treated with concentrated ammonia for 1 hr at 50° C.
  • the mixture was filtered and the ammonia solution was concentrated to dryness.
  • the residue was passed over a Dowex 50 ⁇ 4 (Na+ form) column to exchange ammonium ions for sodium ions.
  • the resulting amino-oligonucleotide was dissolved in 0.1 ml of water, and mixed with 0.1 ml of 1M sodium carbonate aqueous buffer pH 8-9.
  • HPLC solutions were as follows: Solvent A: 5% ACN in 100 mM triethylammonium acetate pH 6.5 and solvent B: 70% ACN in 100 mM triethylammonium acetate pH 6.5.
  • the desired product was characterized by the UV-spectra. UVmax: 258, 348 and mass spectrometry (MALDI-TOF): found 2861 (M+H + ) expected 2862. Yield: 18%.
  • the compound of the title was obtained in a similar way to example 16 but ammonia treatment was run for 2 hours at room temperature.
  • the compound of the title eluted at 13.8 min. It was characterized by the UV-spectra. UVmax: 273, 349. Yield: 5%.
  • the compound of the title was obtained in a similar way to example 17.
  • the compound of the title eluted at 13.9 min. It was characterized by the UV-spectra. UVmax: 273, 349, and mass spectrometry (MALDI-TOF): found 3646 (M+H + ) expected 3647. Yield: 20%.
  • 5′NH 2 -TTCCGGAA 3′ oligonucleotide sequence carrying an amino group at the 5′ end was prepared using the phosphoramidite of the N6-monomethoxytrityl (MMT) protected derivative of 6-aminohexanol (Glen Research).
  • MMT N6-monomethoxytrityl
  • the solid support obtained after the assembly of the sequence was treated with concentrated ammonia for 1 hr at 50° C.
  • Reaction of the 5′ amino-oligonucleotide with 10H-indolo[3,2-b]quinoline-11-carboxylic acid was carried out as described in example 17.
  • the desired product was characterized by the UV-spectra UVmax: 268, 349. Yield: 12%.
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid (0.066 g, 0.25 mmol) was dissolved in 3 ml of refluxing SOCl 2 . After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 2 ml of CH 2 Cl 2 . A solution of 10H-indolo[3,2-b]quinoline-[3-[(3-aminopropyl)methyl-amino]propyl]-4-carboxamide (0.14 g, 0.36 mmol) and triethylamine (0.2 ml) in 2 ml of CH 2 Cl 2 was added at room temperature, and the reaction mixture was stirred for 14 hours.
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid (0.009 g, 0.034 mmol) was dissolved in 2 ml of refluxing SOCl 2 . After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 2 ml of CH 2 Cl 2 . A solution of 10H-indolo[3,2-b]quinoline-11-carboxylic acid(2-[3-(2-amino-ethylamino)-propylamino]-ethyl]-amide (0.02 g, 0.049 mmol) and triethylamine (0.2 ml) in 2 ml of CH 2 Cl 2 was added at room temperature.
  • the in vitro cytotoxicity of the compounds of the present invention was evaluated by colorimetric assays with tetrazol salts (MTT) on Jurkat clon E6-1 and on GLC-4, human leukemia and carcinoma cell lines, respectively.
  • This assay is based on the capacity of live cells to incorporate and reduce the MTT, with yellow colour in a red derivative. This reduction is done by the action of the mitochondrial enzyme succinate hydrogenase, only active in live cells. The intensity of observed colour is directly correlated with the quantity of live cells in the sample as it is described by Mosmann et al., J. Immunol. Methods 1983, vol. 65, p. 55. In all cases the concentrations used were up to 100 mikroM and times of incubation until 72 h. Table 1 shows the biological activity (IC 50 ) of some compounds of formula (I)

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

Compounds of formula G1-L-G2, where G1, is a radical structurally close to cryptolepine, -L- is a single covalent bond or a covalent linking biradical selected from —(CH2)rNR′″(CH2)s— and —(CH2)rNR′″(CH2)sNR″″(CH2)t—, —R′″ and —R″″ are radicals, same or different, selected from the group consisting of H and (C1-C3)-alkyl; r, s and t are an integer from 1 to 3 and -G2 is H or a radical structurally close to -G1, are intercalators. They are compounds which intercalate between DNA base pairs, and are useful as therapeutic agents against cancer, as assess by an in vitro test of citotoxicity with human leukemia cells Jurkat E6-1 and human carcinoma cells GLC-4. Preferred compounds are those where -G1 is bonded to -L- through a carbonyl amino and -L- is —(CH2)3NCH3(CH2)3— or —(CH2)2NCH3(CH2)sNCH3(CH2)2— where s=2 or 3.
-G1 is a radical selected from (IIa) and (IIb); -G2 is a radical selected from H, a radical of formula (IIa), a radical of formula (IIb), the N-radical of 1,8-naphthalimide, the C4-radical of 2-phenylquinoline and the C9-radical of acridine.
Figure US20100331355A1-20101230-C00001

Description

  • This invention relates to new compounds which intercalate between DNA base pairs. Such compounds can be used for the treatment of cancer.
  • BACKGROUND ART
  • Intercalators are a group of compounds that bind between the DNA base pairs. This intercalation can produce an interruption of transcription, replication and/or topoisomerase activity. It has been found that many compounds that bind to DNA by intercalation have antiproliferative properties and in vivo antitumour effects. Intercalators have been used as anticancer drugs. Cryptolepine is a naturally occurring alkaloid and it is used in traditional medicine against malaria. Its formula, shown below, is structurally close to some compounds of the present invention. Cryptolepine is able to bind CG-rich sequences of DNA containing non-alternating CC sites, and it has been reported as potential anticancer drug (cf. e.g. J. N. Lisgarten et al., “The antimalarial and cytotoxic drug cryptolepine intercalates into DNA at cytosine-cytosine sites”, Nature Structural Biology 2002, vol. 9, pp. 57-60). Thus, the provision of new intercalators with anticancer activity is highly desirable.
  • Figure US20100331355A1-20101230-C00002
  • SUMMARY OF THE INVENTION
  • According to an aspect of the present invention, there is provided a compound of formula (I),

  • G1-L-G2   1.
      • (ii) (I)
  • or a pharmaceutically acceptable salt thereof, wherein:
  • -G1 is a radical selected from (IIa) y (IIb);
  • Figure US20100331355A1-20101230-C00003
  • wherein —R′ is an electron pair or a (C1-C3)-alkyl radical; with the condition that
      • (iii) when —R′ is an electron pair, a is a N═C double bond and the
  • Figure US20100331355A1-20101230-C00004
      •  fused ring
  • is the biradical
  • Figure US20100331355A1-20101230-C00005
  • thus radicals (IIa) and (IIb) are respectively (IIa′) and (II′), and
  • Figure US20100331355A1-20101230-C00006
      • (iv) when —R′ is a (C1-C3)-alkyl radical, a is a N—C single bond and the fused ring
  • is the triradical
  • Figure US20100331355A1-20101230-C00007
  • thus radicals (IIa) and (IIb) are respectively (IIa″) and (IIb″);
  • Figure US20100331355A1-20101230-C00008
  • wherein —R1 to —R12 represent radicals, same or different, selected from the group consisting of H, (C1-C4)-alkyl, (C1-C4)-alkoxy, (C1-C4)-alkylamino, phenyl, F, Cl, Br, amino, hydroxy, and nitro;
  • and wherein —B— is a biradical selected from the group consisting of —CONH—,
  • —NR13—, —O—, —(CH2)nNH—, —(CH2)nO—, —CONH(CH2)uZ—,
  • —CONH(CH2)uCH(CH2OH)CH2Z— and —CO[NHCHR″CO]mO—; wherein —R13 is selected from the group consisting of H, (C1-C4)-alkyl, (C1-C4)-alkoxy and
  • (C1-C4)-alkylamino; —R″ are side chains radicals, same or different, corresponding to natural aminoacids; n is an integer from 1 to 3; m is an integer from 1 to 3; u is an integer from 1 to 3, and —Z— is a biradical of a oligonucleotide phosphate between 4 and 23 bases in length, linked to the methylene group at the 5′ end or at the 3′ end;
  • -L- is a single covalent bond or a covalent linking biradical selected from the following ones;

  • —(CH2)rNR′″(CH2)s

  • —(CH2)rNR′″(CH2)sNR″″(CH2)t
  • wherein —R′″ and —R″″ are radicals, same or different, selected from the group consisting of H and (C1-C3)-alkyl; r is an integer from 1 to 3; s is an integer from 1 to 3; t is an integer from 1 to 3; and
  • -G2 is a radical selected from H, a radical of formula (IIa), a radical of formula (IIb), the N-radical of 1,8-naphthalimide, the C4-radical of 2-phenylquinoline, and the C9-radical of acridine;
  • with the proviso that (I) is not one of the compounds of the following list, which have been previously described (the Chemical Abstracts Registry number, CAS RN, is given) but not for the industrial applicability of the present invention. The corresponding values for radicals in formula (I) are also included:
  • 10H-quindoline-11-carboxamide (CAS RN 367911-30-6; -G1=IIa′; —B—═—CONH—; —R′=electron pair; —R1 to R8═H; -L-=single covalent bond; -G2=—H);
  • 2-bromo-10H-quindoline-11-carboxamide (CAS RN 241470-56-4; -G1=IIa′; —B—═—CONH—; —R′=electron pair; —R2═—Br; —R1, —R3 to —R4, —R5 to —R8═H; -L-=single covalent bond; -G2=—H);
  • N-10H-quindolin-11-yl-1,3-propanediamine (CAS RN 188630-48-0; -G1=IIa′; —B—═—NH—, —R′=electron pair; —R1 to —R4, —R5 to —R8═—H; -L-=—(CH2)3NH—; -G2=—H);
  • 10H-quindolin-11-amine monohydrochloride (CAS RN 164406-49-9; G1=IIa′, —B—═—NH; —R′=electron pair; —R1 to —R4, —R5to —R8═—H; -L-=single covalent bond; -G2=—H);
  • 10H-quindolin-11-methanol (CAS RN 241470-44-0; G1=IIa′; —B—═—CH2O—; —R′=electron pair; —R1 to —R4, —R5 to —R8═H, -L-=single covalent bond, -G2=—H); or
  • N-[2-(dimethylamino)ethyl]-10-H-quindoline-4-carboxamide (CAS RN 191172-25-5; G1=IIb′; —B—═—CONH—; —R′=electron pair; —R1 to —R4, —R5 to —R8═—H, -L-=—(CH3)2N(CH3)CH2—, -G2=—H).
  • In a preferred embodiment, compounds of formula (I) are those where (IIa) is the radical (IIa′).
  • Figure US20100331355A1-20101230-C00009
  • Preferred compounds are those of formula (I) where (IIa) is (IIa′) and —B— is
  • —CONH— or —NR13—. Also preferred are those of formula (I) where (IIa) is (IIa′) and —B— is —CO[NHCHR″CO]mO— and more preferred are the compounds where m=2, the leftward —R″ is a glicine side chain, and the rightward —R″ is a arginine side chain . Also preferred compounds are those of formula (I) where (IIa) is (IIa′) and —B— is —CONH(CH2)uZ— or —CONH(CH2)uCH(CH2OH)CH2Z—. More preferred are the compounds where —Z— is -TTCCGGAA- linked at 3′ end or at 5′ end, or -CTTCTTCTTCT- linked at 3′ end. Specially preferred are those where -L- is a single covalent bond. Also specially preferred are those where -L- is a covalent linking biradical selected from the following ones.

  • —(CH2)rNR′″(CH2)s

  • —(CH2)rNR′″(CH2)sNR″″(CH2)t
  • The most preferred are the compounds where -L- is the biradical —(CH2)rNR′″(CH2)s—, —R′″ is methyl and both r and s are 3, and the compounds where -L- is the biradical —(CH2)rNR′″(CH2)sNR″″(CH2)t—, —R′″ and —R′″ are methyl; both r and t are 2 and s is an integer from 2 to 3.
  • In another preferred embodiment, compounds of formula (I) are those where (IIa) is the following radical.
  • Figure US20100331355A1-20101230-C00010
  • Preferred compounds are those where (IIa) is (IIa″) and —B— is —CONH— or
  • —NR13—. Also preferred are those where —B— is —CO[NHCHR″CO]mO—, and more preferred are the compounds where m=2, the leftward —R″ is a glicine side chain, and the rightward —R″ is a arginine side chain. Also preferred compounds are those of formula (I) where (IIa) is (IIa″) and —B— is —CONH(CH2)uZ— or —CONH(CH2)uCH(CH2OH)CH2Z—. More preferred are the compounds where —Z— is -TTCCGGAA- linked at 3′ or 5′ end or -CTTCTTCTTCT- linked at 3′ end. Even more preferred are those where —R′ is methyl. Specially preferred are those where -L- is a single covalent bond. Also specially preferred are those where -L- is a covalent linking biradical selected from the following ones.

  • —(CH2)rNR′″(CH2)s

  • —(CH2)rNR′″(CH2)sNR″″CH2)t
  • The most preferred are the compounds where -L- is the biradical —(CH2)rNR′″(CH2)s—, —R′″ is methyl and both r and s are 3, and the compounds where -L- is the biradical —(CH2)rNR′″(CH2)sNR″″(CH2)t—, —R′″ and —R′″ are methyl; both r and t are 2, and s is an integer from 2 to 3.
  • The most preferred compounds of formula (I) are those of the following list, whose preparation is described in the accompanying examples for the first time: N-[3-[[3-[(9-acridinecarbonyl)amino]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ia); N,N′-(4-methyl-4-azaheptamethylene)-di-(10H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Ib);
  • N-[3-[3-[[2-(1,3-dioxo-(2,3-dihydro)-1H-benzo[de]isoquinolinyl)propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ic); N-[3-[[3-[2-phenyl-4-quinolinecarbonyl)amino]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Id); N,N′-(3,7-dimethyl-3,7-diazanonamethylene)-di-(10H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Ie);
  • N-[(9-acridinecarbonyl)-3,7,10-triaza-3,7-dimethyldecyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (If); N,N′-(3,6-dimethyl-3,6-diazaoctamethylene)-di-(10H-indolo[3,2-b]quinoline-11-11′-carboxamide) (Ig); N-[(9-acridinecarbonyl)-3,6-dimethyl-3,6-diazaoctamethylene]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ih); N-[[1,3-dioxo-(2,3-dihydro)-1H-benzo[b]isoquinolyl]-3,6-dimethyl-3,6-diazaoctamethylene]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ii); N-[[1,3-dioxo-(2,3-dihydro)-1H-benzo[b]isoquinolyl]-3,7,10-triaza-3,7-dimethyldecyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ij); N,N′-(4-methyl-4-azaheptamethylene)-di-(methyl-5H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Im); 10H-indolo[3,2-b]quinoline-11-carbonyl-glicine-arginine (Io); N,N-dimethyl-N′-(5-methyl-5H-indolo[3,2-b]quinolin-11-yl)-ethane-1,2-diamine (Ip); N,N′-(4-methyl-4-azaheptamethylen)-di-(10H-indolo[3,2-b]quinoline-11,11′-amine (Iq); N-1-[(5-hydroxymethyl-6-(5′-TTCCGGAA-3′-phosphate)-hexyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ir); N-1-[(5-hydroxymethyl-6-(5′-CTTCCTCCTCT-3′-phosphate)-hexyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Is); and N-1-[6-(5′-phosphate-TTCCGGAA)-hexyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (It); 10H-Indolo[3,2-b]quinoline-11-carboxylic acid(2-dimethylamino-ethyl)amide (Iu); 10H-Indolo[3,2-b]quinoline-11-carboxylic acid(2-dimethylamino-propyl)amide (Iv); N,N-dimethyl-N′-(5-methyl-5H-indolo[3,2-b]quinolin-11-yl)-propane-1,2-diamine (Iw); N-[3-[[3-[(10H-indolo[3,2-b]quinoline-4-carboxamide]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ix); N,N′-(3,7-dimethyl-3,7-diazanonamethylene)-di-(methyl-5H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Iy); N,N′-(3,6-dimethyl-3,6-diazaoctamethylene)-di-(methyl-5H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Iz); (3,7-diazanonamethylene)-di-(10H-indolo[3,2-b]guinoline-11,11′-carboxamide (Iaa); N,N′-(3,7-dimethyl-3,7-diazanonamethylene)-di-(10H-indolo[3,2-b]guinoline-11,11′-amine (lab); N,N′-(3,6-dimethyl-3,6-diazaoctamethylene)-di-(10H-indolo[3,2-b]guinoline-11,11′-amine (lac).
  • Another aspect of the present invention refers to the use of the compound of formula (I), defined above, for the preparation of a medicament for the treatment of cancer.
  • A further aspect of the present invention relates to a pharmaceutical composition comprising as active ingredient a therapeutically effective amount of the compound of formula (I), defined above, together with suitable pharmaceuticalexcipients or carriers.
  • Compounds of formula (I) where biradical —B— in -G1 is —CONH— and -G2 is not the N-radical of 1,8-naphtalimide can be prepared according to the process summarized in Scheme I, where GP represents an amino protecting group. The process involves a coupling reaction of a compound of formula (Vb) with a compound of formula (III). The compound of formula (Vb) is prepared by reaction of a compound of formula (III) with a monoprotected bis-amine of formula (IV), followed by the removal of the amino protective group. Monoprotected bis-amines of formula (IV) are commercially available compounds or they can be prepared by methods known in the art (cf. Spicer et al., Bioorg. Med. Chem. 2002, vol. 10, p. 19; Deady et al., Bioorg. Med. Chem. 2000, vol. 8, p. 977). An example of amino protective group is the t-butoxycarbonyl (BOC).
  • Figure US20100331355A1-20101230-C00011
  • When biradical —B— in -G1 is —CONH— and -G2 is 1,8-naphthalamide, compounds of formula (I) can be obtained by the process summarized in Scheme II. This process involves the coupling reaction of a compound of formula (VIIb) with a compound of formula (III). The compound of formula (VIIb) can be prepared by reaction of 1,8-naphthalic anhydride (VI) with a monoprotected bis-amine of formula (IV), followed by the removal of the amino protective group.
  • Figure US20100331355A1-20101230-C00012
  • Compounds of formula (I) where —B— is a biradical selected from other groups as —NR13—, —O—, —(CH2)nNH—, —(CH2)nO—, can be obtained analogously by using standards processes of organic chemistry, well known in the art for the production of such type of compounds. For example, for —B—═—NH— the process of Scheme III can be followed, as it has been done for the compound of the Example 15.
  • Figure US20100331355A1-20101230-C00013
  • Compounds of formula (I) where biradical —B— is —CO[NHCHR″CO]mO—, —R″ is side chain radicals, same or different, corresponding to a natural aminoacid and m is an integer selected from 1 to 3, can be obtained by solid phase methods. The synthesis is carried out assembling a N-protected aminoacid or peptide in a p-methylbenzhydrilamine resine. After removal of the protective group, the 11-aminocarboxylic radical selected from formula (IIa) or the 4-aminocarboxylic radical selected from formula (IIb) is coupled by benzotriazol-1-yloxytripyrrolidinophosphonium hexafluoro-phosphate (PyBOP) and diisopropylamine, followed by acidolysis.
  • Compounds of formula (I) where —B— is a birradical selected from —CONH(CH2)uZ— or —CONH(CH2)uCH(CH2OH)CH2Z—, where u is an integer from 1 to 6, and —Z— is a biradical of a oligonucleotide phosphate between 4 and 23 bases in length, linked to the methylene group at 5′ end or at 3′ end can be prepared by conventional methods of peptide coupling in solid phase or solution phase.
  • Compounds of formula (I) may be converted into pharmaceutically acceptable acid addition salts, and salts may be converted into free compounds, by conventional methods. For instance, the acid addition salts may be prepared by contacting the free base with an appropriate amount of the desired acid in a conventional manner.
  • Compounds of formula (I) are intercalators (i.e. compounds which intercalate between DNA base pairs) and, as illustrated by biological results in example 30 are active for cancer treatment.
  • Throughout the description and claims the word “comprise” and variations of the word, are not intended to exclude other technical features, additives, components, or steps. The abstract of this application is incorporated herein as reference. Additional objects, advantages and features of the invention will become apparent to those skilled in the art upon examination of the description or may be learned by practice of the invention. The following examples are provided by way of illustration, and they are not intended to be limiting of the present invention
  • EXAMPLES Example 1 Preparation of N-[3-[[3-[(9-acridinecarbonyl)amino]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ia)
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid previously prepared (cf. Bierer et al., J. Med. Chem. 1998, vol. 41, p. 894 (0.2 g, 0.76 mmol) was dissolved in 5 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 5 ml of CH2Cl2. A solution of [3-[(3-aminopropyl)methylamino]propyl]carbamic acid tert-butyl ester previously prepared (Spicer at el., Bioorg. Med. Chem. 2002, vol. 10, p. 19) (0.26 g, 1.1 mmol) and triethylamine (0.4 ml) in 5 ml of CH2Cl2 was added at room temperature, and the mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 90:10) yielded [2-[[2-[(10H-Indolo[3,2-b]quinoline-11-carbonyl)amino]ethyl]methylamino)ethyl]carbamic acid tert-butyl ester (0.26 g, 70%) as a viscous oil. 1H-NMR [MeOD, δ, ppm]: 8.48 (d, J=7.8 Hz, 1H), 7.25 (m, 2H), 7.64 (m, 4H), 7.35 (dd, J=8.0 Hz, 1H), 3.67 (t, J=6.9 Hz, 2H, CH2NO), 3.07 (t, J=6.9 Hz, 2H, CH2NHBOC), 2.56 (m, 2H, CH2N(CH3)), 2.43 (m, 2H, CH2N(CH3)), 2.26 (s, 3H, N(CH3)), 1.66 (m, 4H, 2CH2CN(CH3)), 1.48 (s, 9H, C(CH3)3).
  • To a solution of [2-[[2-[(10H-indolo[3,2-b]quinoline-11-carbonyl)amino]-ethyl]methylamino)ethyl]-carbamic acid tert-butyl ester (0.13 g, 0.27 mmol) in CH2Cl2 (15 ml), was added trifluoroacetic acid (3 ml). The reaction mixture was stirred at room temperature for 16 h at which point the reaction was completed by TLC. All solvents were removed under reduced pressure to give 10H-indolo[3,2-b]quinoline-[3-[(3-aminopropyl)methylamino]propyl]-11-carboxamide (0.09 g, 92%) as an oil, which was used directly in the next reaction. 1H-NMR [MeOD, δ, ppm]: 8.51 (d, J=7.8 Hz, 1H), 8.32 (m, 2H), 8.02 (m, 1H), 7.84 (m, 2H), 7.67 (d, J=8.0 Hz, 1H), 7.44 (m, 1H), 3.76 (t, J=6.9 Hz, 2H, CH2NO), 3.31 (m, 2H, CH2NH2), 2.96 (s, 3H, N(CH3)), 2.27 (m, 2H, CH2N(CH3)), 2.15 (m, 2H, CH2N(CH3)), 1.26 (m, 4H, 2CH2CN(CH3)).
  • 9-acridinecarboxylic acid (0.036 g, 0.16 mmol) was dissolved in 3 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 4 ml of CH2Cl2. A solution of 10H-indolo[3,2-b]quinoline-[3-[(3-aminopropyl)methyl-amino]propyl]-11-carboxamide (0.076 g, 0.19 mmol) and triethylamine (0.2 ml) in 4 ml of CH2Cl2 was added at room temperature, and the reaction mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 95:5) yielded the desired compound (55 mg, 50%) as a solid. mp 108-110° C. 1H-NMR [CDCl3, δ, ppm]: 8.38 (m, 1H), 8.05 (m, 7H), 7.58 (m, 7H), 7.26 (m, 1H), 3.59 (m, 4H, 2CH2NO), 2.55 (m, 4H, 2CH2NO), 2.26 (s, 3H, N(CH3)), 1.91 (m, 4H, 2CH2CN(CH3)); MS (EI, m/z) 595 (M++1).
  • Example 2 Preparation of N,N′-(4-methyl-4-azaheptamethylene)-di-(10H-indolo[3,2-b]quinoline-11,11′-carboxamide (Ib)
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid (0.02 g, 0.08 mmol) was dissolved in 2 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 2 ml of CH2Cl2. A solution of 10H-indolo[3,2-b]quinoline-[3-[(3-aminopropyl)methylamino]propyl]-11-carboxamide (0.037 g, 0.09 mmol) prepared as in Example 1, and triethylamine (0.2 ml) in 2 ml of CH2Cl2 was added at room temperature, and the mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 95:5) yielded the desired compound (30 mg, 50%) as a solid. mp 124-126° C. 1H NMR [CDCl3, δ, ppm]: 8.47 (m, 2H), 8.17 (m, 4H), 7.59 (m, 8H), 7.25 (m, 2H), 3.65 (m, 4H, 2CH2NO), 2.63 (m, 4H, 2CH2NO), 2.33 (s, 3H, N(CH3)), 1.99 (m, 4H, 2CH2CN(CH3)); MS (EI, m/z) 634 (M++1).
  • Example 3 Preparation of N-[3-[3-[2-[1,3-dioxo-(2,3-dihydro)-1H-benzo[de]isoquinolinyl]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ic)
  • 1,8-naphthalic anhydride (0.18 g, 0.9 mmol) was added to a solution of [3-[(3-aminopropyl)methylamino]propyl]carbamic acid tert-butyl ester previously prepared (Spicer at el., Bioorg. Med. Chem. 2002, vol. 10, p. 19) (0.2 g, 0.8 mmol) in absolute ethanol (20 ml). The reaction mixture was heated at reflux overnight. Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 50:1) yielded [3-[[3-(1,3-dioxo-3a,9b-dihydro-1H,3H-benzo[de]isoquinolin-2-yl)propyl]methylamino]propyl]carbamic acid tert-butyl ester (0.37 g, 90%) as a foam. 1H-NMR [CDCl3, δ, ppm]: 8.62 (m, 2H), 8.22 (m, 2H), 7.72 (m, 2H), 4.23 (m, 2H, 2CH2NO), 3.20 (m, 2H, CH2NH), 2.55 (m, 4H, 2(CH2)), 2.26 (s, 3H, N(CH3)), 1.93 (m, 2H, (CH2)), 1.68 (m, 2H, (CH2)), 1.48 (s, 9H, C(CH3)3).
  • To a solution of [3-[[3-(1,3-dioxo-3a,9b-dihydro-1H,3H-benzo[de]isoquinolin-2-yl)propyl]methylamino]propyl]carbamic acid tert-butyl ester (0.34 g, 0.81 mmol) in CH2Cl2 (24 ml) was added trifluoroacetic acid (6 ml). The reaction mixture was stirred at room temperature for 16 h at which point the reaction was completed by TLC. All solvents were removed under reduced pressure to give 2-[3-[(3-aminopropyl)methylamino]propyl]-3a,9b-dihydro-benzo[de]isoquinoline-1,3-dione (0.24 g, 92%) as an oil, which was used directly in the next reaction. 1H-NMR [MeOD, δ, ppm]: 8.64 (m, 2H), 8.54 (m, 2H), 7.92 (m, 2H), 4.45 (m, 2H, 2CH2NO), 3.55 (m, 4H, 2CH2), 3.33 (m, 2H, CH2), 3.22 (s, 3H, N(CH3)), 2.44 (m, 4H, 2(CH2)).
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid (0.24 g, 0.9 mmol) was dissolved in 7 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 10 ml of CH2Cl2. A solution of 2-[3-[(3-aminopropyl)methyl-amino]propyl]-3a,9b-dihydro-benzo[de]isoquinoline-1,3-dione (0.24 g, 0.73 mmol) and NEt3 (1 ml) in 10 ml of CH2Cl2 was added at room temperature, and the reaction mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and chromatography on alumina (CH2Cl2/MeOH, 95:5) yielded the desired compound (0.13 g, 30%) as a solid. mp 200-204° C. 1H-NMR [CDCl3, δ, ppm]: 8.38-8.02 (m, 7H), 7.61-7.37 (m, 7H), 3.86 (m, 4H, 2CH2NO), 2.60 and 2.40 (m, 4H, 2CH2NO), 2.22 (s, 3H, N(CH3)), 1.91 and 1.61 (m, 4H, 2CH2CN(CH3)).
  • Example 4 Preparation of N-[3-[[3-[2-phenyl-4-quinolinecarbonyl)amino]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Id)
  • Activation and coupling of 10H-indolo[3,2-b]quinoline-11-carboxylic acid with 2-phenyl-quinoline-[3-[(3-aminopropyl)methylamino]propyl]-4-carboxamide (prepared analogously to Example 1 from 2-phenyl-4-quinolinecarboxylic acid), gave the desired compound as a solid, 36%, mp 144-148° C. 1H-NMR [MeOD, δ, ppm]: 8.18 (m, 2H), 8.08 (m, 4H), 7.61-7.44 (m, 10H), 7.28 (m, 1H), 3.68 (m, 2H, 2CH2NO), 3.54 (m, 2H, 2CH2NO), 3.34 (s, 3H, N(CH3)), 2.64 (m, 4H, 2CH2CN(CH3)); 1.98 (m, 4H, 2CH2); MS (EI, m/z) 619 (M+−1).
  • Example 5 Preparation of N,N′-(3,7-dimethyl-3,7-diazanonamethylene)-di-(10H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Ie)
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid (0.25 g, 0.95 mmol) was dissolved in 5 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 5 ml of CH2Cl2. A solution of [2-[[3-[(2-aminoethyl)methylamino]propyl]methylamino]ethyl]carbamic acid tert-butyl ester (Deady et al., Bioorg. Med. Chem. 2000, vol. 8, p. 977) (0.38 g, 1.3 mmol) and triethylamine (0.4 ml) in 5 ml of CH2Cl2 was added at room temperature. The reaction mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 90:10) yielded [2-[[3-[[2-[(10H-indolo[3,2-b]quinoline-11-carbonyl)-amino]ethyl]methylamino]propyl]methyl-amino]ethyl]carbamic acid tert-butyl ester (0.31 g, 62%) as a viscous oil . 1H-NMR [CDCl3, δ, ppm]: 8.45 (d, J=7.8 Hz, 1H), 8.25 (m, 2H), 7.62 (m, 4H), 7.35 (dd, J=8.0, 8.0 Hz, 1H), 3.85 (m, 2H, CH2NO), 3.21 (m, 2H, CH2), 3.07 (m, 2H, CH2NHBOC), 2.79 (m, 4H, 2CH2N(CH3)), 2.37-2.19 (m, 4H, 2(CH2)), 2.15 (s, 6H, 2N(CH3)), 1.48 (s, 9H, C(CH3)3).
  • To a solution [2-[[3-[[2-[(10H-indolo[3,2-b]quinoline-11-carbonyl)amino]ethyl]methylamino]propyl]methylamino]ethyl]carbamic acid tert-butyl ester (0.3 g, 0.57 mmol) in CH2Cl2 (20 ml), was added trifluoroacetic acid (6 ml). The reaction mixture was stirred at room temperature for 16 h, at which point the reaction was completed by TLC. All solvents were removed under reduced pressure to give 10H-indolo[3,2-b]quinoline-[2-[[3-[(2-aminoethyl)methylamino]propyl]methylamino]ethyl]-11-carboxamide (0.23 g, 92%) as a oil, which was used directly. 1H-NMR [MeOD, δ, ppm]: 8.59 (d, J=7.8 Hz, 1H), 8.44 (m, 2H), 8.09 (m,1H), 7.94 (m, 2H), 7.87 (d, J=8.0 Hz, 1H), 7.51 (m, 1H), 3.68 (m, 2H, CH2NO), 3.46 (m, 8H, 4CH2), 3.11 (m, 4H, 2CH2N(CH3)), 2.97 (s, 6H, 2N(CH3)).
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid (0.11 g, 0.41 mmol) was dissolved in 10 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 15 ml of CH2Cl2. A solution of 10H-indolo[3,2-b]quinoline-11-carboxylic acid[2-[[3-[(2-aminoethyl)methylamino]propyl]methylamino]-ethyl]-amide (0.23 g, 0.5 mmol) and triethylamine (0.5 ml) in 15 ml of CH2Cl2 was added at room temperature. The reaction mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 95:5) yielded the desired compound (0.14 g, 52%) as a foam. 1H-NMR [CDCl3, δ, ppm]: 8.48 (m, 2H), 8.30 (m, 4H), 7.73-7.54 (m, 8H), 7.25 (m, 2H) 3.77 (m, 4H, 2CH2NO), 2.58 (m, 4H, 2CH2N(CH3)), 2.38 (m, 6H, 4CH2), 2.15 (s, 6H, 2N(CH3)).
  • Example 6 Preparation of N-[(9-acridinecarbonyl)-3,7,10-triaza-3,7-dimethyldecyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (If)
  • 9-acridinecarboxylic acid (0.15 g, 0.67 mmol) was dissolved in 15 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 15 ml of CH2Cl2. A solution of 10H-indolo[3,2-b]quinoline-[2-[[3-[(2-aminoethyl)methylamino]propyl]methylamino]ethyl]-11-carboxamide (0.4 g, 0.92 mmol) and triethylamine (1 ml) in 15 ml of CH2Cl2 was added at room temperature. The reaction mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 95:5) yielded the desired compound (0.38 g, 88%) as a foam. 1H-NMR [CDCl3, δ, ppm]: 8.12 (m, 1H), 8.02 (m, 7H), 7.58 (m, 7H), 7.33 (m, 1H), 3.77 (m, 4H, 2CH2NO), 2.58 (m, 4H, 2CH2N(CH3)), 2.48 (m, 6H, 4CH2), 2.28 (s, 6H, 2N(CH3)).
  • Example 7 Preparation of N,N′-(3,6-dimethyl-3,6-diazaoctamethylene)-di-(10H-indolo[3,2-b]quinoline-11-11′-carboxamide (Ig)
  • Activation and coupling of 10H-indolo[3,2-b]quinoline-11-carboxylic acid with 10H-indolo[3,2-b]quinoline-[2-[[2-[(2-aminoethyl)methylamino]-ethyl]-methylamino]thyl]-11-carboxamide (prepared analogously to Example 5 from [2-[[2-[(2-aminoethyl)methylamino]ethyl]methylamino]ethyl]carbamic acid tert-butyl ester) gave the desired compound as a foam, 38%. 1H-NMR [CDCl3, δ, ppm]: 8.46 (m, 2H), 8.30 (m, 4H), 7.73-7.54 (m, 8H), 7.18 (m, 2H) 3.62 (m, 4H, 2CH2NO), 2.58 (m, 4H, 2CH2N(CH3)), 2.38 (m, 4H, 4CH2), 2.35 (s, 6H, 2N(CH3)).
  • Example 8 Preparation of N-[(9-acridinecarbonyl)-3,6-dimethyl-3,6-diazaoctamethylene)-10H-indolo[3,2-b]quinoline-11-carboxamide (Ih)
  • Activation and coupling of 9-acridinecarboxylic acid with 10H-indolo[3,2-b]quinoline-[2-[[2-[(2-aminoethyl)methylamino]ethyl]methylamino]ethyl]-11-carboxamide (prepared analogously to Example 5 from [2-[[2-[(2-aminoethyl)methylamino]ethyl]methylamino]ethyl]carbamic acid tert-butyl ester) gave the desired compound as a foam, 48%. 1H-NMR [CDCl3, δ, ppm]: 8.12 (m, 1H), 8.02 (m, 7H), 7.58 (m, 7H), 7.33 (m, 1H), 4.02 (m, 4H, 2CH2NO), 3.98 (m, 4H, 2CH2N(CH3)), 3.01 (m, 4H, 4CH2), 2.70 (s, 6H, 2N(CH3)).
  • Example 9 Preparation of N-[[1,3-dioxo-(2,3-dihydro)-1H-benzo[de]isoquinolyl]-3,6-dimethyl-3,6-diazaoctamethylene]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ii)
  • Activation and coupling of 10H-indolo[3,2-b]quinoline-11-carboxylic acid with 2-[2-[[2-[(2-aminoethyl)methylamino]ethyl]methylamino]ethyl]-benzo[de]isoquinoline-1,3-dione (prepared analogously to Example 3 from 2-[[2-[(2-amino-ethyl)methylamino]ethyl]methylamino]ethyl]carbamic acid tert-butyl ester) gave the desired compound as a foam, 37%, 1H-NMR [CDCl3, δ, ppm]: 8.55-8.11 (m, 7H), 7.72-7.17 (m, 7H), 4.03 (m, 4H, 2CH2NO), 3.80 (m, 4H, 2CH2N(CH3)), 2.93-2.64 (m, 4H, 4CH2), 2.55 (s, 6H, 2N(CH3)).
  • Example 10 Preparation of N-[[1,3-dioxo-(2,3-dihydro)-1H-benzo[de]isoquinolyl]-3,7,10-triaza-3,7-dimethyldecyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ij)
  • Activation and coupling of 10H-indolo[3,2-b]quinoline-11-carboxylic acid with 2-[2-[[3-[(2-aminoethyl)methylamino]propyl]methylamino]ethyl]-benzo[de]isoquinoline-1,3-dione (prepared analogously to Example 3 from 2-[[3-[(2-aminoethyl)methylamino]propyl]methylamino]ethyl]carbamic acid tert-butyl ester) gave the desired compound as a foam, 40%. 1H-NMR [CDCl3 δ, ppm]: 8.59-8.02 (m, 7H), 7.77-7.25 (m, 7H), 4.05 (m, 4H, 2CH2NO), 3.82 (m, 4H, 2CH2N(CH3)), 2.78-2.34 (m, 6H, 4CH2), 2.25 (s, 6H, 2N(CH3)).
  • Example 11 Preparation of 5-methyl-5H-indolo[3,2-b]quinoline-11-carboxylic acid, Intermediate for the Preparation of (Im)
  • The 5-methyl-5H-indolo[3,2-b]quinoline-11-carboxylic acid is known in the literature as part of extract plant Cryptolepis sanguinolenta (cf. Paulo et al., Planta Medica 2000, vol. 66, p. 30). It can also be prepared by the following process:
  • 5,11-dimethyl-5H-indolo[3,2-b]quinoline (cf. Yang et al., J. Nat. Prod, 1999, vol. 62, pp. 976] (0.1 g, 0.4 mmol) was added to 10 ml of water. KMnO4 (64 mg. 0.4 mmol) is added, and the solution is heated to reflux for 1 h. Then a second portion of KMnO4 (64 mg. 0.4 mmol) is introduced, followed by 10 ml of water, and the heating is continued for 1 h. The reaction mixture is allowed to cool slightly, and the precipitated oxide of manganese is filtered and washed with hot water. The filtrated is concentrated under reduced pressure to half of the volume, filtered and acidified with concentrated hydrochloric acid. This acid solution is then evaporated to dryness under reduced pressure to give the desired compound (70 mg, 64%), which was used directly. 1H-NMR [MeOD δ, ppm]: 8.24 (m, 2H), 7.79 (m, 2H), 7.54 (m, 4H), 5.15 (s, 3H) MS (EI, m/z) 277 (M++1).
  • Example 12 Preparation of N,N′-(4-methyl-4-azaheptamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Im)
  • Activation and coupling of 5-methyl-5H-indolo[3,2-b]quinoline-11-carboxylic acid, prepared as in Example 11, with 5-methyl-5H-indolo[3,2-b]quinoline-[3-[(3-aminopropyl)methylamino]propyl]-11carboxamide (prepared analogously to Example 1 from 5-methyl-5H-indolo[3,2-b]quinoline-11-carboxylic acid) gave the desired compound as a foam, 50%, 1H NMR [CDCl3 δ, ppm]: 8.47 (m, 2H), 8.17 (m, 4H), 7.59 (m, 8H), 7.25 (m, 2H), 5.15 (s, 6H), 3.65 (m, 4H, 2CH2NO), 2.63 (m, 4H, 2CH2NO), 2.33 (s, 3H, N(CH3)), 1.99 (m, 4H, 2CH2CN(CH3)).
  • Example 13 Preparation of 10H-indolo[3,2-b]quinoline-11-carbonyl-glicine-arginine (Io)
  • The compound of the title was prepared by solid phase methods. After assembling the Boc-glicine-arginine sequence on a p-methylbenzhydrylamine resin, the N-terminal was deprotected and 10H-indolo[3,2-b]quinoline-11-carboxylic acid was coupled by means of benzotriazol-1-yloxytripyrrolidinophosphonium hexafluoro-phosphate (PyBOP) and diisopropylethylamine (5, 5 and 10 equivalents, respectively) in 1:1 DMSO-DMF for 1 h. Acidolysis with HF/anisole (9:1, 0° C., 1 h) provided the desired compound in highly homogeneous form (>98% by HPLC). MS(MALDI-TOF): EM(expected): 474.1; EM(found): 475.5 (M+H+).
  • Example 14 Preparation of N,N-dimethyl-N′-(5-methyl-5H-indolo[3,2-b]quinolin-11-yl)-ethane-1,2-diamine (Ip)
  • A solution of 11-cloro-5-metil-5H-indolo[3,2-b]quinoline previously prepared (cf. Bierer et al., J. Med. Chem. 1998, vol. 41, p. 2754) (66 mg, 0.25 mmol) and N,N-dimethylethylenediamine (94 μl, 1 mmol) in 2-ethoxyethanol (10 ml) was heated at 120° C. for 10 min. Removal of the solvent and chromatography on alumina (CHCl3 100%) yielded the desired compound (36 mg, 46%) as a yellow solid. m. p. 162-164° C. 1H NMR [CDCl3 δ, ppm]: 8.28 (d, J=8.5 Hz, 1H), 7.89 (m, 2H), 7.69-7.61 (m, 3H), 7.31 (m, 2H), 4.58 (s, 3H, NCH3), 4.32 (m, 2H, CH2), 3.14 (m, 2H, CH2), 2.51 (s, 6H, N(CH3)2).
  • Example 15 Preparation of N,N′-(4-methyl-4-azaheptamethylen)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-amine (Iq)
  • A solution of 11-chloro-5-methyl-5H-indolo[3,2-b]quinoline (cf. Bierer et al., J. Med. Chem. 1998, vol. 41, pp. 2754) (61 mg, 0.23 mmol) and [[3-[(3-amino-propyl)methylamino]propyl]carbamic acid tert-butyl ester previously prepared (Spicer at el., Bioorg. Med. Chem. 2002, vol. 10, p. 19) (84 μl, 0.34 mmol) in 2-ethoxyethanol (10 ml) was heated at 120° C. for 10 min. Removal of the solvent and purification by chromatography on alumina (CHCl3: MeOH 2%) yielded [3-[methyl-[3-(5-methyl-5H-indolo[3,2-b]quinolin-11-yl-amino)propyl]-amino]propyl]carbamic acid tert-butyl ester (49 mg, 49%) as a foam. 1H NMR [CDCl3 δ, ppm]: 8.16 (d, J=8.5 Hz, 1H), 7.81 (m, 4H), 7.55-7.48 (m, 2H), 7.21 (m, 1H), 4.51 (s, 3H, NCH3), 4.27 (m, 2H, CH2NO), 3.12 (m, 2H, CH2NHBOC), 2.76 (m, 2H, CH2N(CH3)), 2.63 (m, 2H, CH2N(CH3)), 2.39 (s, 3H, N(CH3)), 2.34 (m, 2H, CH2CN(CH3)), 1.73 (m, 2H, CH2CN(CH3)), 1.37 (s, 9H, C(CH3)3).
  • To a solution of [3-[methyl-[3-(5-methyl-5H-indolo[3,2-b]quinolin-11-ylamino)-propyl]amino]propyl]-carbamic acid tert-butyl ester (49 mg, 0.1 mmol) in CH2Cl2 (10 ml), was added trifluoroacetic acid (12 μl). This mixture was stirred at room temperature for 16 h, at which point the reaction was completed by TLC. All solvents were removed under reduced pressure to give N1-methyl-N1-[3-(5-methyl-5H-indolo[3,2-b]quinolin-11-ylamino)propyl]propane-1,3-diamine (38 mg, 92%) as an oil, which was used directly. 1H-NMR [MeOD, δ, ppm]: 8.52 (d, J=8.5 Hz, 1H), 8.35-7.73 (m, 4H), 7.63-7.60 (m, 2H), 7.31 (m, 1H), 4.45 (s, 3H, NCH3), 3.60 (m, 2H, CH2NO), 3.02 (m, 2H, CH2N(CH3)), 2.92 (s, 3H, N(CH3)), 2.36 (m, 4H, 2CH2CN(CH3)), 2.14 (m, 2H, CH2N(CH3)), 1.90 (m, 2H, CH2CN(CH3)).
  • A solution of 11-chloro-5-methyl-5H-indolo[3,2-b]quinoline, previously prepared, (cf. Bierer et al., J. Med. Chem. 1998, vol. 41, p. 2754) (40 mg, 0.15 mmol) and N1-methyl-N1-[3-(5-methyl-5H-indolo[3,2-b]quinolin-11-ylamino)-propyl]propane-1,3-diamine (38 mg, 0.1 mmol) in 2-ethoxyethanol (10 ml) was heated at 120° C. for 10 min. Removal of the solvent and purification by chromatography on alumina (CHCl3 100%) yielded the desired compound (17 mg, 46%) as a foam. 1H NMR [CDCl3 δ, ppm]: 8.16-8.21 (m, 4H), 8.01 (m, 2H), 7.93-7.71 (m, 4H), 7.50-7.30 (m, 2H), 7.23-7.25 (m, 4H), 4.26 (s, 3H, NCH3), 4.24 (s, 3H, NCH3), 4.12 (m, 2H, CH2NO), 3.03 (m, 2H, CH2), 2.80 (m, 2H, CH2N(CH3)), 2.70 (m, 2H, CH2N(CH3)), 2.44 (s, 3H, N(CH3)), 2.16 (m, 2H, CH2CN(CH3)), 1.72 (m, 2H, CH2CN(CH3)).
  • Example 16 Preparation of N-1-[5-hydroxymethyl-6-(5′-TTCCGGAA-3′-phosphate)-hexyl]-10H-indolo[3,2-b]quinoline-11-carboxamide by Solid Phase Coupling (Ir)
  • 5′TTCCGGAA-NH2-3′ oligonucleotide sequence carrying an amino group at the 3′ end was prepared using 3-O-dimethoxytrityloxy-2-(N-9-fluorenphenylmethoxycarbonyl-4-aminobutyl)-1-propanyl ester succinamidyl-controlled pore glass (3′ amino C7 modifier CPG from Glen Research). The solid support obtained after the assembly of the sequence was treated with a 0.1 M solution of 1,8-diazabiciclo[5.4.0]undece-7-ene (DBU) in dry acetonitrile (2 min, room temperature). In this way, the Fmoc group that protects the amino group is removed selectively with a non-nucleophillic base. The resulting support was washed with acetonitrile and reacted with 10H-indolo[3,2-b]quinoline-11-carboxylic acid as follows. A mixture containing 10 molar excess of 10H-indolo[3,2-b]quinoline-11-carboxylic acid, 20 molar excess of diisopropylethylamine (DIEA) as base catalyst and 10 molar excess of (benzotriazol-1-yloxy)trispyrrolidinophosphonium hexafluorophosphate (PyBOP) as carboxyl activator was prepared in dry dimethylformamide (DMF) (0.2 ml). The mixture was left for 2 min at room temperature and added to the support. After 1 hour at room temperature, the mixture was filtered and washed with DMF. The support was dried and concentrated ammonia (1 ml) was added. The ammonia solution was left for 1 hr at 50° C. The mixture was filtered and the ammonia solution was concentrated to dryness. The residue was dissolved in water and desalted by NAP-10 (Sephadex G-25) eluted with water. Finally, the oligonucleotide fractions were analyzed by HPLC. HPLC solutions were as follows. Solvent A: 5% ACN in 100 mM triethylammonium acetate pH 6.5 and solvent B: 70% ACN in 100 mM triethylammonium acetate pH 6.5. Columns: PRP-1 (Hamilton), 250×10 mm. Flow rate: 3 ml/min. A 20 min linear gradient from 5-35%. The compound of the title eluted at 14.0 min. Unreacted amino-oligonucleotide eluted around 9 min. The desired product was characterized by the UV-spectra. UVmax: 258,348. Yield: 7%.
  • Example 17 Preparation of N-1-[5-hydroxymethyl-6-(5′-TTCCGGAA-3′-phosphate)-hexyl]-10H-indolo[3,2-b]quinoline-11-carboxamide by Solution-Phase Coupling (Ir)
  • 5′TTCCGGAA-NH2 3′ oligonucleotide sequence carrying an amino group at the 3′ end was prepared using the 3′ amino C7 modifier CPG (Glen Research). The solid support obtained after the assembly of the sequence was treated with concentrated ammonia for 1 hr at 50° C. The mixture was filtered and the ammonia solution was concentrated to dryness. The residue was passed over a Dowex 50×4 (Na+ form) column to exchange ammonium ions for sodium ions. The resulting amino-oligonucleotide was dissolved in 0.1 ml of water, and mixed with 0.1 ml of 1M sodium carbonate aqueous buffer pH 8-9. In a separate container 10H-indolo[3,2-b]quinoline-11-carboxylic acid (10 molar excess) was dissolved in 0.1 ml of DMF, and mixed with N-hydroxysuccinimide (10 molar excess) and N,N-diisopropylcarbodiimide (10 molar excess). The mixture was left for 10 min at room temperature and added to the aqueous solution of the amino-oligonucleotide. The reaction mixture was kept at room temperature overnight. The mixture was concentrated to dryness and the residue was dissolved in water. The solution was passed through a NAP-10 column. The fractions containing oligonucleotide were analyzed by HPLC. HPLC solutions were as follows: Solvent A: 5% ACN in 100 mM triethylammonium acetate pH 6.5 and solvent B: 70% ACN in 100 mM triethylammonium acetate pH 6.5. Columns: PRP-1 (Hamilton), 250×10 mm. Flow rate: 3 ml/min. A 20 min linear gradient from 5-35%. The compound of the title eluted at 14.0 min Unreacted amino-oligonucleotide eluted around 9 min. The desired product was characterized by the UV-spectra. UVmax: 258, 348 and mass spectrometry (MALDI-TOF): found 2861 (M+H+) expected 2862. Yield: 18%.
  • Example 18 Preparation of N-1-[5-hydroxymethyl-6-(5′-CTTCCTCCTCT-3′-phosphate)-hexyl]-10H-indolo[3,2-b]quinoline-11-carboxamide by Solid Phase Coupling (Is)
  • The compound of the title was obtained in a similar way to example 16 but ammonia treatment was run for 2 hours at room temperature. The compound of the title eluted at 13.8 min. It was characterized by the UV-spectra. UVmax: 273, 349. Yield: 5%.
  • Example 19 Preparation of N-1-[5-hydroxymethyl-6-(5′-CTTCCTCCTCT-3′-phosphate)-hexyl]-10H-indolo[3,2-b]quinoline-11-carboxamide by Solution-Phase Coupling (Is)
  • The compound of the title was obtained in a similar way to example 17. The compound of the title eluted at 13.9 min. It was characterized by the UV-spectra. UVmax: 273, 349, and mass spectrometry (MALDI-TOF): found 3646 (M+H+) expected 3647. Yield: 20%.
  • Example 20 Preparation of N-1-[6-(5′-phosphate-TTCCGGAA)-hexyl]-10H-indolo[3,2-b]quinoline-11-carboxamide by Solution-Phase Coupling (It)
  • 5′NH2-TTCCGGAA 3′ oligonucleotide sequence carrying an amino group at the 5′ end was prepared using the phosphoramidite of the N6-monomethoxytrityl (MMT) protected derivative of 6-aminohexanol (Glen Research). The solid support obtained after the assembly of the sequence was treated with concentrated ammonia for 1 hr at 50° C. Reaction of the 5′ amino-oligonucleotide with 10H-indolo[3,2-b]quinoline-11-carboxylic acid was carried out as described in example 17. The compound of the title eluted at 16 min. The desired product was characterized by the UV-spectra UVmax: 268, 349. Yield: 12%.
  • Example 21 Preparation of 10H-Indolo[3,2-b]quinoline-11-carboxylic acid(2-dimethylamino-ethyl)amide (Iu)
  • 10H-Indolo[3,2-b]quinoline-11-carboxylic acid (0.5 g, 1.9 mmol) was dissolved in 20 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 10 ml of CH2Cl2. A solution of N,N-dimethylethylendiamine (0.2 mL, 2.26 mmol) and triethylamine (2.2 ml) in 10 ml of CH2Cl2 was added at room temperature, and the mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 90:10) yielded the desired compound (0.38 g, 61%) as a solid. m. p. 175-176° C. 1H-NMR [MeOD, δ, ppm]: 8.16 (d, J=7.8 Hz, 1H), 7.92 (m, 2H), 7.35-7.20 (m, 3H), 7.04 (m, 2H), 3.62 (m, 2H, CH2NO), 2.55 (m, 2H, CH2N(CH3)2), 2.29 (s, 6H, N(CH3)2). MS (EI, m/z) 277 (M++1).
  • Example 22 Preparation of 10H-Indolo[3,2-b]quinoline-11-carboxylic acid(2-dimethylamino-propyl)amide (Iv)
  • 10H-Indolo[3,2-b]quinoline-11-carboxylic acid (0.2 g, 0.76 mmol) was dissolved in 8 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 10 ml of CH2Cl2. A solution of N,N-dimethylproylendiamine (0.1 mL, 0.97 mmol) and triethylamine (1 ml) in 10 ml of CH2Cl2 was added at room temperature, and the mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 90:10) yielded the desired compound (80 mg, 31%) as a solid. m. p. 188-189° C. 1H-NMR [MeOD, δ, ppm]: 8.16 (d, J=7.8 Hz, 1H), 7.92 (m, 2H), 7.35-7.20 (m, 3H), 7.04 (m, 2H), 3.62 (m, 2H, CH2NO), 2.55 (m, 2H, CH2N(CH3)2), 2.29 (s, 6H, N(CH3)2), 1.88 (m, 2H, CH2). MS (EI, m/z) 347 (M++1).
  • Example 23 Preparation of N,N-dimethyl-N′-(5-methyl-5H-indolo[3,2-b]quinolin-11-yl)-propane-1,2-diamine (Iw)
  • A solution of 11-chloro-5-metil-5H-indolo[3,2-b]quinoline (47 mg, 0.17 mmol) and N,N-dimethylpropylenediamine (78 μl, 0.76 mmol) in 2-ethoxyethanol (5 ml) was heated at 120° C. for 10 min. Removal of the solvent and chromatography on alumina (CHCl3 100%) yielded the desired compound (20 mg, 35%) as a foam. 1H NMR [CDCl3 δ, ppm]: 8.28 (d, J=8.5 Hz, 1H), 7.89 (m, 2H), 7.69-7.61 (m, 3H), 7.31 (m, 2H), 4.58 (s, 3H, NCH3), 4.23 (m, 2H, CH2), 3.86 (m, 2H, CH2), 3.55 (m, 2H, CH2), 2.55 (s, 6H, N(CH3)2). MS (EI, m/z) 333 (M++1).
  • Example 24 Preparation of N-[3-[[3-[(10H-indolo[3,2-b]quinoline-4-carboxamide]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ix)
  • 10H-indolo[3,2-b]quinoline-4-carboxylic acid (0.2 g, 0.76 mmol) was dissolved in 5 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 5 ml of CH2Cl2. A solution of [3-[(3-aminopropyl)methylamino]propyl]carbamic acid tert-butyl ester (0.26 g, 1.1 mmol) and triethylamine (0.4 ml) in 5 ml of CH2Cl2 was added at room temperature, and the mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine, HCl 1N) and dried (MgSO4). All solvents were removed under reduced pressure to give 10H-indolo[3,2-b]quinoline-[3-[(3-aminopropyl)methylamino]propyl]-4-carboxamide (0.14 g, 48%) as an oil, which was used directly in the next reaction. 1H-NMR [MeOD, δ, ppm]: 8.72 (d, J=7.8 Hz, 1H), 8.63 (d, J=7.8 Hz, 1H), 8.51 (d, J=7.8 Hz, 1H), 7.97 (m, 2H), 7.79 (d, J=8.0 Hz, 1H), 7.43 (m, 2H), 3.75 (t, J=6.9 Hz, 2H, CH2NO), 3.31 (m, 2H, CH2NH2), 2.96 (s, 3H, N(CH3)), 2.27 (m, 2H, CH2N(CH3)), 2.15 (m, 2H, CH2N(CH3)), 1.35 (m, 4H, 2CH2CN(CH3)).
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid (0.066 g, 0.25 mmol) was dissolved in 3 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 2 ml of CH2Cl2. A solution of 10H-indolo[3,2-b]quinoline-[3-[(3-aminopropyl)methyl-amino]propyl]-4-carboxamide (0.14 g, 0.36 mmol) and triethylamine (0.2 ml) in 2 ml of CH2Cl2 was added at room temperature, and the reaction mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 95:5) yielded the desired compound (27 mg, 17%) as an oil. 1H-NMR [CDCl3, 6, ppm]: 8.43 (m, 1H), 8.28 (m, 7H), 7.58 (m, 7H), 7.26 (m, 1H), 3.20 (m, 4H, 2CH2NO), 2.49 (m, 4H, 2CH2NO), 2.05 (s, 3H, N(CH3)), 1.64 (m, 4H, 2CH2CN(CH3)).
  • Example 25 Preparation of N,N′-(3,7-dimethyl-3,7-diazanonamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Iy)
  • Activation and coupling of 5-methyl-5H-indolo[3,2-b]quinoline-11-carboxylic acid with 5-methyl-5H-indolo[3,2-b]quinoline[2-[[2-[(2-aminoethyl)methylamino]-ethyl]-methylamino]ethyl]-11-carboxamide (prepared analogously to Example 5 from [2-[[3-[(2-aminoethyl)methylamino]propyl]methylamino]ethyl]carbamic acid tert-butyl ester and 5-methyl-5H-indolo[3,2-b]quinoline-11-carboxylic acid gave the desired compound as a foam, (4 mg, 15%), 1H NMR [CDCl3 δ, ppm]: 8.47 (m, 2H), 8.17 (m, 4H), 7.59 (m, 8H), 7.25 (m, 2H), 5.15 (s, 6H), 3.77 (m, 4H, 2CH2NO), 2.58 (m, 4H, 2CH2N(CH3)), 2.38 (m, 6H, 4CH2), 2.15 (s, 6H, 2N(CH3)).
  • Example 26 Preparation of N,N′-(3,6-dimethyl-3,6-diazaoctamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Iz)
  • Activation and coupling of 5-methyl-5H-indolo[3,2-b]quinoline-11-carboxylic acid with 5-methyl-5H-indolo[3,2-b]quinoline[2-[[2-[(2-aminoethyl)methylamino]-ethyl]-methyl-amino]ethyl]-11-carboxamide (prepared analogously to Example 7 from [2-[[2-[(2-aminoethyl)methylamino]ethyl]methylamino]ethyl]carbamic acid tert-butyl ester and 5-methyl-5H-indolo[3,2-b]quinoline-11-carboxylic acid gave the desired compound as a foam, (28 mg, 10%), 1H NMR [CDCl3 δ, ppm]: 8.47 (m, 2H), 8.17 (m, 4H), 7.59 (m, 8H), 7.25 (m, 2H), 5.15 (s, 6H), 3.65 (m, 4H, 2CH2NO), 2.58 (m, 4H, 2CH2N(CH3)), 2.38 (m, 4H, 4CH2), 2.35 (s, 6H, 2N(CH3)).
  • Example 27 Preparation of (3,7-diazanonamethylene)-di-(10H-indolo[3,2-b]quinoline-11,11′-carboxamide (Iaa)
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid (0.056 g, 0.21 mmol) was dissolved in 2 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 2 ml of CH2Cl2. A solution of (2-[3-(2-amino-ethylamino]-ethyl)-carbamic acid tert-butyl ester (0.079 g, 0.3 mmol) and triethylamine (0.2 ml) in 2 ml of CH2Cl2 was added at room temperature. The reaction mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 90:10) yielded [2-(3-{2-[10H-indolo[3,2-b]quinoline-11-carbonyl)-amino]-ethylamino}-propylamino)-ethyl]carbamic acid tert-butyl ester (0.025 g, 25%) as a viscous oil . 1H-NMR [CDCl3, δ, ppm]: 8.46 (d, J=7.8 Hz, 1H), 8.30 (m, 2H), 7.68 (m, 4H), 7.53 (dd, J=8.0, 8.0 Hz, 1H), 3.65 (m, 2H, CH2NO), 3.12 (m, 2H, CH2), 3.07 (m, 2H, CH2NHBOC), 2.06 (m, 4H, 2CH2NH), 1.67-1.55 (m, 4H, 2(CH2)), 1.48 (s, 9H, C(CH3)3).
  • To a solution [2-(3-{2-[10H-indolo[3,2-b]quinoline-11-carbonyl)-amino]-ethylamino}-propylamino)-ethyl]carbamic acid tert-butyl ester (0.025 g, 0.049 mmol) in CH2Cl2 (5 ml), was added trifluoroacetic acid (1 ml). The reaction mixture was stirred at room temperature for 16 h, at which point the reaction was completed by TLC. All solvents were removed under reduced pressure to give 10H-indolo[3,2-b]quinoline-11-carboxylic acid(2-[3-(2-amino-ethylamino)-propylamino]-ethyl]-amide (0.02 g , 95%) as a oil, which was used directly. 1H-NMR [MeOD, δ, ppm]: 8.55 (d, J=7.8 Hz, 1H), 8.26 (m, 2H), 8.00 (m,1H), 7.80 (m, 2H), 7.64 (d, J=8.0 Hz, 1H), 7.26 (m, 1H), 4.19 (m, 2H, CH2NO), 3.56 (m, 8H, 4CH2), 3.20 (m, 4H, 2CH2NH).
  • 10H-indolo[3,2-b]quinoline-11-carboxylic acid (0.009 g, 0.034 mmol) was dissolved in 2 ml of refluxing SOCl2. After 6 hours a clear solution was obtained, and all volatiles were removed in vacuo. The crude acid chloride was suspended in 2 ml of CH2Cl2. A solution of 10H-indolo[3,2-b]quinoline-11-carboxylic acid(2-[3-(2-amino-ethylamino)-propylamino]-ethyl]-amide (0.02 g, 0.049 mmol) and triethylamine (0.2 ml) in 2 ml of CH2Cl2 was added at room temperature. The reaction mixture was stirred for 14 hours. The organic phase was washed (NaHCO3, brine) and dried (MgSO4). Removal of the solvent and purification by chromatography on alumina (CH2Cl2/MeOH, 95:5) yielded the desired compound (0.015 g, 68%) as a foam. 1H-NMR [CDCl3, δ, ppm]: 8.56 (m, 2H), 8.30 (m, 4H), 7.88-7.65 (m, 8H), 7.15 (m, 2H) 3.62 (m, 4H, 2CH2NO), 2.37 (m, 4H, 2CH2NH), 2.17 (m, 6H, 4CH2).
  • Example 28 Preparation of N,N′-(3,7-dimethyl-3,7-diazanonamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-amine (Iab)
  • Activation and coupling of 11-chloro-5-methyl-5H-indolo[3,2-b]quinoline with N-(2-amino-ethyl)-(N,N′-dimethyl-N′-[2-(5-methyl-5H-indolo[3,2-b]quinolin-11-ylamino)-ethyl]-propane-1,3-diamine (prepared analogously to Example 15 from [2-[[3-[(2-aminoethyl)methylamino]propyl]methylamino]ethyl]carbamic acid tert-butyl ester and 11-chloro-5-methyl-5H-indolo[3,2-b]quinoline gave the desired compound as a foam, (13 mg, 16%), 1H NMR [CDCl3 δ, ppm]: 8.58-8.40 (m, 4H), 8.01 (m, 2H), 7.84-7.64 (m, 4H), 7.63-7.55 (m, 2H), 7.42-7.27 (m, 4H), 4.61 (s, 6H, NCH3), 4.12 (m, 4H, 2CH2NH), 2.58 (m, 4H, 2CH2N(CH3)), 2.38 (m, 6H, 4CH2), 2.15 (s, 6H, 2N(CH3)).
  • Example 29 Preparation of N,N′-(3,6-dimethyl-3,6-diazaoctamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-amine (Iac)
  • Activation and coupling of 11-chloro-5-methyl-5H-indolo[3,2-b]quinoline with N-(2-amino-ethyl)-(N,N′-dimethyl-N′-[2-(5-methyl-5H-indolo[3,2-b]quinolin-11-ylamino)-ethyl]-ethane-1,2-diamine (prepared analogously to Example 15 from [2-[[2-[(2-aminoethyl)methylamino]ethyl]methylamino]ethyl]carbamic acid tert-butyl ester and 11-chloro-5-methyl-5H-indolo[3,2-b]quinoline gave the desired compound as a foam, (23 mg, 56%), 1H NMR [CDCl3 δ, ppm]: 8.66-8.21 (m, 4H), 8.01 (m, 2H), 7.84-7.64 (m, 4H), 7.63-7.55 (m, 2H), 7.42-7.27 (m, 4H), 4.91 (s, 6H, NCH3), 4.12 (m, 4H, 2CH2NH), 2.58 (m, 4H, 2CH2N(CH3)), 2.38 (m, 4H, 4CH2), 2.35 (s, 6H, 2N(CH3)).
  • Example 30 In Vitro Biological Test
  • The in vitro cytotoxicity of the compounds of the present invention was evaluated by colorimetric assays with tetrazol salts (MTT) on Jurkat clon E6-1 and on GLC-4, human leukemia and carcinoma cell lines, respectively. This assay is based on the capacity of live cells to incorporate and reduce the MTT, with yellow colour in a red derivative. This reduction is done by the action of the mitochondrial enzyme succinate hydrogenase, only active in live cells. The intensity of observed colour is directly correlated with the quantity of live cells in the sample as it is described by Mosmann et al., J. Immunol. Methods 1983, vol. 65, p. 55. In all cases the concentrations used were up to 100 mikroM and times of incubation until 72 h. Table 1 shows the biological activity (IC50) of some compounds of formula (I)
  • TABLE 1
    Biological activities.
    Compound (I) IC50 (μM) Jurkat clon E6-1 IC50 (μM) GLC-4
    Ia 1.42 1.63
    Ib 0.59 1.10
    Ic 0.95 4.70
    Id 3.37 3.57
    Ie 5.13 2.88
    If 7.16 3.64
    Ip 0.26 0.67
    Iu 18.7  15.70
    Iv Not tested 16.73
    Iw Not tested 1.08
    Ix 1.21 19.57
    Iy 7.94 19.08
    Iz 5.02 11.53
    Iac Not tested 5.88

Claims (22)

1. A compound of formula (I)

G1-L-G2   (I)
or a pharmaceutically acceptable salt thereof, wherein:
-G1 is a radical (II)
Figure US20100331355A1-20101230-C00014
wherein —R1 is an electron pair or a (C1-C3)-alkyl radical; with the condition that (i) when —R′ is an electron pair, a is a N═C double bond and the fused ring
Figure US20100331355A1-20101230-C00015
is the biradical
Figure US20100331355A1-20101230-C00016
thus radical (II) is (IIa′), and
Figure US20100331355A1-20101230-C00017
(ii) when —R′ is a (C1-C3)-alkyl radical, a is a N—C single bond and the fused ring is the triradical
Figure US20100331355A1-20101230-C00018
thus radical (II) is (IIa″);
Figure US20100331355A1-20101230-C00019
wherein —R1 to —R12 represent radicals, same or different, selected from the group consisting of H, (C1-C4)-alkyl, (C1-C4)-alkoxy, (C1-C4)-alkylamino, phenyl, F, Cl, Br, amino, hydroxy, and nitro;
and wherein —B— is a biradical selected from the group consisting of —CONH—, —NR13—, —O—, —(CH2)nNH—, —(CH2)nO—, and —CO[NHCHR″CO]mO—; wherein —R13 is selected from the group consisting of H, (C1-C4)-alkyl, (C1-C4)-alkoxy and (C1-C4)-alkylamino; —R″ are side chains radicals, same or different, corresponding to natural aminoacids; n is an integer from 1 to 3 and m is an integer from 1 to 3;
-L- is a single covalent bond or a covalent linking biradical selected from the following ones;

—(CH2)rNR′″(CH2)s

—(CH2)rNR′″(CH2)sNR″″(CH2)t
wherein —R′″ and —R″″ are radicals, same or different, selected from the group consisting of H and (C1-C3)-alkyl; r is an integer from 1 to 3; s is an integer from 1 to 3; t is an integer from 1 to 3; and
-G2 is a radical selected from a radical of formula (II), the N-radical of 1,8-naphthalimide, the C4-radical of 2-phenylquinoline, and the C9-radical of acridine.
2. The compound according to claim 1, wherein (II) is the radical (IIa′)
Figure US20100331355A1-20101230-C00020
3. The compound according to claim 2, wherein —B— is selected from the group consisting of —CONH— and —NR13—.
4. The compound according to claim 2, wherein —B— is —CO[NHCHR″CO]mO—.
5. The compound according to claim 4, wherein m=2, the leftward —R″ is a glicine side chain, and the rightward —R″ is an arginine side chain.
6. The compound according to claim 2, wherein -L- is a single covalent bond.
7. The compound according to claim 2, wherein -L- is a covalent linking biradical selected from the following ones

—(CH2)rNR′″(CH2)s

—(CH2)rNR′″(CH2)sNR″″(CH2)t
8. The compound according to claim 7, wherein -L- is the biradical
—(CH2)rNR′″(CH2)s—, —R′″ is methyl, and both r and s are 3.
9. The compound according to claim 7, wherein -L- is the covalent linking biradical —(CH2)rNR′″(CH2)sNR″″(CH2)t—, both —R′″ and —R″″ are methyl; both r and t are 2, and s is 2 or 3.
10. The compound according to claim 1, wherein (II) is the radical (IIa″)
Figure US20100331355A1-20101230-C00021
11. The compound according to claim 10, wherein —B— is selected from the group consisiting of —CONH— and —NR13—.
12. The compound according to claim 10, wherein —B— is —CO[NHCHR″CO]mO—.
13. The compound according to claim 12, wherein m=2, the leftward —R″ is a glicine side chain, and the rightward —R″ is the arginine side chain.
14. The compound according to claim 10, wherein —R′ is methyl.
15. The compound according to claim 14, wherein -L- is a single covalent bond.
16. The compound according to claim 14, wherein -L- is a biradical selected from the following ones.

—(CH2)rNR′″(CH2)s

—(CH2)rNR′″(CH2)sNR″″(CH2)t
17. The compound according to claim 16, wherein -L- is the biradical —(CH2)rNR′″(CH2)s—, R′″ is methyl, and both r and s are 3.
18. The compound according to claim 16, wherein -L- is the biradical —(CH2)rNR′″(CH2)sNR′″(CH2)t—, both —R′″ and —R″″ are methyl; both r and t are 2, and s is an integer from 2 to 3.
19. The compound according to claim 1, which is selected from the group consisting of:
N-[3-[[3-[(9-acridinecarbonyl)amino]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ia);
N,N′-(4-methyl-4-azaheptamethylene)-di-(10H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Ib);
N-[3-[3-[[2-(1,3-dioxo-(2,3-dihydro)-1H-benzo[de]isoquinolinyl]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ic);
N-[3-[[3-[(2-phenyl-4-quinolinecarbonyl)amino]propyl]methylamino]propyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Id);
N,N′-(3,7-dimethyl-3,7-diazanonamethylene)-di-(10H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Ie);
N-[(9-acridinecarbonyl)-3,7,10-triaza-3,7-dimethyldecyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (If);
N,N′-(3,6-dimethyl-3,6-diazaoctamethylene)-di-(10H-indolo[3,2-b]quinoline-11-11′-carboxamide (Ig);
N-[(9-acridinecarbonyl)-3,6-dimethyl-3,6-diazaoctamethylene]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ih);
N-[[1,3-dioxo-(2,3-dihydro)-1H-benzo[de]isoquinolyl]-3,6-dimethyl-3,6-diazaoctamethylene]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ii);
N-[[1,3-dioxo-(2,3-dihydro)-1H-benzo[de]isoquinolyl]-3,7,10-triaza-3,7-dimethyldecyl]-10H-indolo[3,2-b]quinoline-11-carboxamide (Ij);
N,N′-(4-methyl-4-azaheptamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Im);
N,N′-(4-methyl-4-azaheptamethylen)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-amine (Iq);
N,N′-(3,7-dimethyl-3,7-diazanonamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Iy);
N,N′-(3,6-dimethyl-3,6-diazaoctamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-carboxamide) (Iz);
(3,7-diazanonamethylene)-di-(10H-indolo[3,2-b]quinoline-11,11′-carboxamide (Iaa);
N,N′-(3,7-dimethyl-3,7-diazanonamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-amine (Iab); and
N,N′-(3,6-dimethyl-3,6-diazaoctamethylene)-di-(5-methyl-5H-indolo[3,2-b]quinoline-11,11′-amine (Iac).
20. A method for the preparation of a medicament for the treatment of cancer which comprises administering to a subject a therapeutically effective amount of a compound of formula (I)

G1-L-G2   (I)
or a pharmaceutically acceptable salt thereof, wherein:
-G1 is a radical (II)
Figure US20100331355A1-20101230-C00022
wherein —R′ is an electron pair or a (C1-C3)-alkyl radical; with the condition that (i) when —R′ is an electron pair, a is a N═C double bond and the fused ring
Figure US20100331355A1-20101230-C00023
is the biradical
Figure US20100331355A1-20101230-C00024
thus radical (II) is (IIa′), and
Figure US20100331355A1-20101230-C00025
(ii) when —R′ is a (C1-C3)-alkyl radical, a is a N—C single bond and the fused ring is the triradical
Figure US20100331355A1-20101230-C00026
thus radical (II) is (IIa″);
Figure US20100331355A1-20101230-C00027
wherein —R1 to —R12 represent radicals, same or different, selected from the group consisting of H, (C1-C4)-alkyl, (C1-C4)-alkoxy, (C1-C4)-alkylamino, phenyl, F, Cl, Br, amino, hydroxy, and nitro;
and wherein —B— is a biradical selected from the group consisting of —CONH—, —NR13—, —O—,—(CH2)nNH—, —(CH2)nO—, and —CO[NHCHR″CO]mO—; wherein —R13 is selected from the group consisting of H, (C1-C4)-alkyl, (C1-C4)-alkoxy and (C1-C4)-alkylamino; —R″ are side chains radicals, same or different, corresponding to natural aminoacids; n is an integer from 1 to 3 and m is an integer from 1 to 3;
-L- is a single covalent bond or a covalent linking biradical selected from the following ones.

—(CH2)rNR′″(CH2)s

—(CH2)rNR′″(CH2)sNR′″(CH2)t
wherein —R′″ and —R′″ are radicals, same or different, selected from the group consisting of H and (C1-C3)-alkyl; r is an integer from 1 to 3; s is an integer from 1 to 3; t is an integer from 1 to 3; and
-G2 is a radical selected from a radical of formula (II), the N-radical of 1,8-naphthalimide, the C4-radical of 2-phenylquinoline, and the C9-radical of acridine.
21. A pharmaceutical composition comprising a therapeutically effective amount of the compound as defined in claim 1, together with appropriate amounts of pharmaceutical excipients or carriers.
22. A method of producing a composition of matter of formula (I) comprising one of the following processes:
Figure US20100331355A1-20101230-C00028
when biradical —B— in -G1 is —CONH— and -G2 is not an N-radical of 1,8-naphtalimide; and
wherein GP represents an amino protective group and wherein formula (IV) is a monoprotected bis-amine; or
Figure US20100331355A1-20101230-C00029
when biradical —B— in -G1 is —CONH— and -G2 is 1,8-naphtalimide; and
wherein GP represents an amino protective group and wherein formula (IV) is a monoprotected bis-amine; or
Figure US20100331355A1-20101230-C00030
when biradical —B— is a biradical selected from a group of: —NR13—, —O—, —(CH2)nNH, and —(CH2)nO—; and
wherein GP represents an amino protective group.
US12/760,408 2003-11-20 2010-04-14 Substituted Quinolines for the Treatment of Cancer Abandoned US20100331355A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/760,408 US20100331355A1 (en) 2003-11-20 2010-04-14 Substituted Quinolines for the Treatment of Cancer

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
ESP200302821 2003-11-20
ES200302821 2003-11-20
US10/580,140 US7728000B2 (en) 2003-11-20 2004-11-18 Substituted quinolines for the treatment of cancer
PCT/EP2004/013106 WO2005054236A1 (en) 2003-11-20 2004-11-18 Substituted quinolines for the treatment of cancer
US12/760,408 US20100331355A1 (en) 2003-11-20 2010-04-14 Substituted Quinolines for the Treatment of Cancer

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2004/013106 Continuation WO2005054236A1 (en) 2003-11-20 2004-11-18 Substituted quinolines for the treatment of cancer
US11/580,140 Continuation US20070088412A1 (en) 2005-10-13 2006-10-11 System and device for heating or cooling shape memory surgical devices

Publications (1)

Publication Number Publication Date
US20100331355A1 true US20100331355A1 (en) 2010-12-30

Family

ID=34639541

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/580,140 Expired - Fee Related US7728000B2 (en) 2003-11-20 2004-11-18 Substituted quinolines for the treatment of cancer
US12/760,408 Abandoned US20100331355A1 (en) 2003-11-20 2010-04-14 Substituted Quinolines for the Treatment of Cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/580,140 Expired - Fee Related US7728000B2 (en) 2003-11-20 2004-11-18 Substituted quinolines for the treatment of cancer

Country Status (4)

Country Link
US (2) US7728000B2 (en)
EP (1) EP1687304A1 (en)
JP (1) JP2007511573A (en)
WO (1) WO2005054236A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101324781B1 (en) 2011-04-29 2013-11-08 (주)씨에스엘쏠라 Noble photosensitizer for photovoltaic cell and photovoltaic cell prepared from the same
US10008017B2 (en) 2006-06-29 2018-06-26 Intuitive Surgical Operations, Inc. Rendering tool information as graphic overlays on displayed images of tools

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005054236A1 (en) * 2003-11-20 2005-06-16 Crystax Pharmaceuticals S.L. Substituted quinolines for the treatment of cancer
ATE454149T1 (en) * 2005-05-20 2010-01-15 Crystax Pharmaceuticals S L HALOGENATED QUINDOLINE DIMERS FOR THE TREATMENT OF CANCER
EP2014698A1 (en) * 2007-07-12 2009-01-14 Crystax Pharmaceuticals S.L. Polymers and their use as fluorescent labels
JP6108696B2 (en) * 2011-10-28 2017-04-05 国立大学法人 岡山大学 Indolequinoline derivative, method for producing the derivative, and antimalarial agent and anticancer agent containing the derivative
CN115160310B (en) * 2022-05-10 2023-06-23 中国人民解放军北部战区总医院 Alternanthene derivatives as CDK2/Topo I inhibitors, preparation method thereof and antitumor application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7728000B2 (en) * 2003-11-20 2010-06-01 Crystax Pharmaceuticals Substituted quinolines for the treatment of cancer

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH01228965A (en) * 1988-03-10 1989-09-12 Mect Corp Condensed quinoline and condensed acridine, their production and anticancer agent containing said compounds
JPH02256667A (en) * 1988-12-27 1990-10-17 Masatoshi Yamato Condensed quinoline-based compound and production thereof
US5362726A (en) * 1993-02-12 1994-11-08 Healthsearch, Inc. Compound and method of treatment for falciparum malaria
JPH10193807A (en) * 1997-01-09 1998-07-28 Konica Corp Thermal transfer recording material and thermal transfer recording method
WO1998045272A1 (en) * 1997-04-07 1998-10-15 Latrobe University Topoisomerase inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7728000B2 (en) * 2003-11-20 2010-06-01 Crystax Pharmaceuticals Substituted quinolines for the treatment of cancer

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10008017B2 (en) 2006-06-29 2018-06-26 Intuitive Surgical Operations, Inc. Rendering tool information as graphic overlays on displayed images of tools
KR101324781B1 (en) 2011-04-29 2013-11-08 (주)씨에스엘쏠라 Noble photosensitizer for photovoltaic cell and photovoltaic cell prepared from the same

Also Published As

Publication number Publication date
WO2005054236A1 (en) 2005-06-16
JP2007511573A (en) 2007-05-10
WO2005054236B1 (en) 2005-08-18
US20070105784A1 (en) 2007-05-10
US7728000B2 (en) 2010-06-01
EP1687304A1 (en) 2006-08-09

Similar Documents

Publication Publication Date Title
US20100331355A1 (en) Substituted Quinolines for the Treatment of Cancer
US7223833B1 (en) Peptide nucleic acid conjugates
US8859490B2 (en) Peptide nucleic acid monomers and oligomers
EP3152223B1 (en) Mitomycin conjugate
EP0804456A1 (en) Peptide nucleic acid conjugates
WO1996011205A9 (en) Peptide nucleic acid conjugates
JP4805917B2 (en) Camptothecin conjugated with a cyclic peptide at position 7 as a cell division inhibitor
Bailly et al. Design, synthesis, DNA binding, and biological activity of a series of DNA minor‐groove‐binding intercalating drugs
US7049061B1 (en) Stereochemical control of the DNA binding affinity, sequence specificity, and orientation-preference of chiral hairpin polyamides in the minor groove
TW201004647A (en) Novel dual targeting antitumoural conjugates
US11319341B2 (en) Immune-stimulating soluble doxorubicin-conjugated complex
US8008316B2 (en) Azonafide derived tumor and cancer targeting compounds
US20120220537A1 (en) 9-aminoacridine derivatives, their preparation and uses
WO2019047941A1 (en) Amanitin antibody conjugate
EP0646596A1 (en) Nucleic acid binding oligomers having C-branches for therapy and diagnosis
US9056887B2 (en) Minor groove binder phosphoramidites and methods of use
CA2172958A1 (en) Anthracene derivatives for use as anticancer agent or dye
WO1997046551A1 (en) Substituted quinolylmethylen-oxindole analogues as tyrosine kinase inhibitors
RU2697519C1 (en) Peptide agent comprising a psma-binding ligand based on a urea derivative, a method for production thereof and use thereof for preparing a conjugate with a drug and diagnostic agent
KR20080091103A (en) Compound for the inhibition of apoptosis
Mehiri et al. An efficient biodelivery system for antisense polyamide nucleic acid (PNA)
US7999101B2 (en) Cationic lipids for the transfection of nucleic acids
Patino et al. Modelling, synthesis and biological evaluation of an ethidium–arginine conjugate linked to a ribonuclease mimic directed against TAR RNA of HIV-1
JP2007537243A (en) 7-t-Butoxyiminomethylcamptothecin conjugated with an integrin antagonist at position 20
Aymamí et al. Substituted Quinolines for the Treatment of Cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: CRYSTAX PHARMACEUTICALS, S.L., SPAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AYMAMI BOFARULL, JUAN;COLL CAPELLA, MIQUEL;LLEBARIA SOLDEVILA, AMADEO;AND OTHERS;REEL/FRAME:026731/0556

Effective date: 20060425

Owner name: UNIVERSITAT POLITENCNICA DE CATALUNYA, SPAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AYMAMI BOFARULL, JUAN;COLL CAPELLA, MIQUEL;LLEBARIA SOLDEVILA, AMADEO;AND OTHERS;REEL/FRAME:026731/0556

Effective date: 20060425

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS, S

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AYMAMI BOFARULL, JUAN;COLL CAPELLA, MIQUEL;LLEBARIA SOLDEVILA, AMADEO;AND OTHERS;REEL/FRAME:026731/0556

Effective date: 20060425

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION