WO2006034574A1 - Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals - Google Patents

Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals Download PDF

Info

Publication number
WO2006034574A1
WO2006034574A1 PCT/CA2005/001467 CA2005001467W WO2006034574A1 WO 2006034574 A1 WO2006034574 A1 WO 2006034574A1 CA 2005001467 W CA2005001467 W CA 2005001467W WO 2006034574 A1 WO2006034574 A1 WO 2006034574A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
prodrug
compounds
solvate
Prior art date
Application number
PCT/CA2005/001467
Other languages
English (en)
French (fr)
Inventor
James B. Mcalpine
Arjun H. Banskota
Original Assignee
Ecopia Biosciences Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/951,436 external-priority patent/US7186713B2/en
Priority claimed from CA 2497031 external-priority patent/CA2497031A1/en
Application filed by Ecopia Biosciences Inc. filed Critical Ecopia Biosciences Inc.
Priority to US11/663,890 priority Critical patent/US20090029972A1/en
Priority to AU2005289317A priority patent/AU2005289317A1/en
Priority to EP05791575A priority patent/EP1809612A4/de
Priority to JP2007532739A priority patent/JP2008514551A/ja
Priority to CA002581658A priority patent/CA2581658A1/en
Publication of WO2006034574A1 publication Critical patent/WO2006034574A1/en
Priority to IL182214A priority patent/IL182214A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D243/00Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms
    • C07D243/06Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4
    • C07D243/10Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D243/38[b, e]- or [b, f]-condensed with six-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • This invention relates to dibenzodiazepinone analogues having improved properties, which are chemical derivatives of Compound 1.
  • the invention further relates to their pharmaceutically acceptable salts, solvates and prodrugs, and to methods for obtaining the compounds.
  • One method of obtaining the derivatives involves post- biosynthesis chemical modification of Compound 1.
  • the present invention further relates to the use of dibenzodiazepinone analogues, and their pharmaceutically acceptable salts, solvates and prodrugs as pharmaceuticals, in particular to their use as inhibitors of cancer cell growth, mammalian lipoxygenase, and for treating acute and chronic inflammation, and to pharmaceutical compositions comprising a dibenzodiazepinone analogue, or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • actinomycetes are a subset of a large and complex group of Gram- positive bacteria known as actinomycetes. Over the past few decades these organisms, which are abundant in soil, have generated significant commercial and scientific interest as a result of the large number of therapeutically useful compounds, particularly antibiotics, produced as secondary metabolites. The intensive search for strains able to produce new antibiotics has led to the identification of hundreds of new species.
  • 5,541 ,181 discloses a dibenzodiazepinone compound, specifically 5-farnesyl-4,7,9-trihydroxy-dibenzodiazepin-11-one (named "BU-4664L”), produced by a known euactinomycetes strain, Micromonospora sp. M990-6 (ATCC 55378).
  • ECO- 4601 Compound 1
  • Micromonospora sp. strains 046-ECO11 and [S01]046 are disclosed in CA 2,466,340. Its use for the treatment of cancer is disclosed in PCT/CA2005/000751.
  • the dibenzodiazepinone analogue is represented by a compound of Formula I as defined below, or a hydrogenated or hydroalkoxylated farnesyl derivative, or a pharmaceutically acceptable salt, solvate or prodrug of a compound of Formula I.
  • the dibenzodiazepinone analogue is represented by any one of Compounds 2 to 27 as defined below, or a pharmaceutically acceptable salt, solvate or prodrug, or salt of a prodrug of any one of Compounds 2 to 25.
  • the dibenzodiazepinone analogue is represented by any one of Compounds 2 to 5, 7, 13, 14, 15, 18, 21 , and 22 as defined below, or a pharmaceutically acceptable solvate or prodrug of any one of Compounds 2 to 5, 7, 13, 14, 15, 18, 21 , and 22.
  • the dibenzodiazepinone analogue is represented by any one of Compounds 23 to 25 as defined below, or a pharmaceutically acceptable solvate or prodrug of any one of Compounds 23 to 25.
  • the invention further encompasses a dibenzodiazepinone analogue obtained by a method comprising the steps of: (a) providing an isolated form of ECO-4601 , (b) chemically modifying Compound 1.
  • the dibenzodiazepinone analogue is a compound of Formula I, or a hydrogenated or hydroalkoxylated farnesyl derivative, or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • the chemical modification step (b) is one or more steps selected from N- alkylation, O-triacetylation, aryl bromination, and farnesyl hydrogenation or hydroalkoxylation.
  • the dibenzodiazepinone analogue is selected from Compounds 2 to 27.
  • the dibenzodiazepinone analogue is selected from Compounds 2 to 5, 7, 13, 14, 15, 18, 21 , and 22.
  • the dibenzodiazepinone analogue is selected from Compounds 23 to 25.
  • the invention further encompasses a method for making a dibenzodiazepinone compound, comprising chemically modifying the farnesyl dibenzodiazepinone Compound 1 , and optionally isolating and purifying the dibenzodiazepinone compound produced.
  • the chemical modification step comprises at least one step selected from ⁇ /-alkylations, O-acylations, aryl bromination and modifications of the double bonds of the farnesyl side chain including, hydrogenation, and hydroalkoxylation.
  • the farnesyl side chain modification reaction is partial (one or two double bonds modified) or complete (all three double bonds modified).
  • the invention further encompasses the use of a compound of Formula I or a hydrogenated or hydroalkoxylated farnesyl derivative, or a pharmaceutically acceptable salt, solvate or prodrug thereof as an antitumor agent for the treatment of a pre ⁇ cancerous or cancerous condition in a mammal.
  • the compound is selected from Compounds 2 to 27.
  • the compound is selected from Compounds 2 to 5, 7, 13, 14, 15, 18, 21 , and 22.
  • the compound is selected from Compounds 23 to 25.
  • the invention further encompasses the use of a compound of Formula I or a hydrogenated or hydroalkoxylated farnesyl derivative, or a pharmaceutically acceptable salt or prodrug thereof as an antineoplastic agent for the treatment of a proliferative disorder in a mammal.
  • the compound is selected from Compounds 2 to 27.
  • the compound is selected from Compounds 2 to 5, 7, 13, 14, 15, 18, 21 , and 22.
  • the compound is selected from Compounds 23 to 25.
  • the invention further encompasses the use of a compound of Formula I or a hydrogenated or hydroalkoxylated farnesyl derivative, or a pharmaceutically acceptable salt or prodrug thereof in the preparation of a medicament for the treatment of a pre ⁇ cancerous or cancerous condition in a mammal.
  • the compound is selected from Compounds 2 to 27.
  • the compound is selected from Compounds 2 to 5, 7, 13, 14, 15, 18, 21 , and 22.
  • the compound is selected from Compounds 23 to 25.
  • the invention further encompasses a commercial package comprising a compound of Formula I or a hydrogenated or hydroalkoxylated farnesyl derivative, or a pharmaceutically acceptable salt or prodrug thereof, together with instructions for use in the treatment of a neoplasm or a pre-cancerous or cancerous condition.
  • the compound is selected from Compounds 2 to 27.
  • the compound is selected from Compounds 2 to 5, 7, 13, 14, 15, 18, 21 , and 22.
  • the compound is selected from Compounds 23 to 25.
  • the cancer cell, neoplastic, pre-cancerous or cancerous condition, in the above-mentioned uses is selected from leukemia, melanoma, breast cancer, lung cancer, pancreatic cancer, ovarian cancer, renal cancer, colon or colorectal cancer, prostate cancer, and CNS cancer.
  • the cancer cell, and pre-cancerous or cancerous condition, in the above-mentioned methods and uses is selected from leukemia, breast cancer, prostate cancer, and CNS cancer.
  • Figure 1 shows inhibition of tumor growth resulting from bolus administration of 10 to 30 mg/kg of Compound 1 to C6 glioblastoma-bearing mice one day after tumor cell inoculation.
  • Figure 2 shows inhibition of tumor growth resulting from bolus administration of 20-30 mg/kg of Compound 1 to glioblastoma-bearing mice ten days after tumor cell inoculation.
  • Figure 3 shows micrographs of tumor sections from mice bearing glioblastoma tumors and treated with saline or Compound 1. The cell density of tumor treated with Compound 1 appears decreased and nuclei from tumor cells are larger and pycnotic suggesting a cytotoxic effect.
  • Figure 4 inhibition of tumor growth resulting from bolus administration of 20 to 75 mg/kg of Compound 2 to C6 glioblastoma-bearing mice from day 11 to day 20 of treatment.
  • Figure 5 shows the mean ( ⁇ SD) plasma concentrations of Compound 1 in Swiss mice following 30 mg/kg intravenous (iv), intraperitoneal (ip), subcutaneous (sc) and oral (po) bolus administrations.
  • Figure 6 shows the mean ( ⁇ SD) plasma concentrations of Compounds 1 and 2 in CD-1 mice following 30 mg/kg intravenous (iv) and intraperitoneal (ip) bolus administrations.
  • Figure 7 shows the mean concentration of Compound 1 in various tissues, 30 minutes after 30mg/kg intravenous (iv), intraperitoneal (ip) and subcutaneous (sc) bolus administrations.
  • the present invention relates to novel dibenzodiazepi ⁇ one analogues having improved properties, herein referred as the compounds of Formula I and hydrogenated or hydroalkoxylated famesyl derivatives, and Compounds 2 to 27.
  • the invention further relates to pharmaceutically acceptable salts, solvates and prodrugs of dibenzodiazepinone compounds.
  • the invention further relates to processes of obtaining the analogs by chemical modification of Compound 1.
  • Compound 1 is isolated from strains of actinomycetes, Micromonospora sp. 046-ECO11 (also as 046(ECO11 )) or [S01]046, as described in PCT/CA04/000069.
  • the invention also relates to a method for producing novel dibenzodiazepinone analogs, by chemical modification of the farnesyl dibenzodiazepinone obtained from fermentation and isolation.
  • the compound produced is a compound of Formula I or a hydrogenated or hydroalkoxylated farnesyl derivative thereof, or a compound selected from Compounds 23 to 27.
  • the present invention also relates to pharmaceutical compositions comprising a compound selected from the compounds of Formula I, and Compounds 2 to 27, and their pharmaceutically acceptable salts, solvates and derivatives.
  • the compounds are useful as pharmaceuticals, in particular for use as inhibitors of neoplastic cell growth.
  • compositions comprising the dibenzodiazepinone compounds of the present invention together with a pharmaceutically acceptable carrier, and methods of using the pharmaceutical compositions to treat pre-cancererous or cancerous conditions.
  • farnesyl dibenzodiazepinone refers to Compound 1 , namely 10-farnesyl-4,6,8-trihydroxy-5,10-dihydrodibenzo[b,e][1 ,4]diazepin-11-one, also referred to as ECO-4601.
  • the terms "compound(s) of the invention”, “dibenzodiazepinone analogue(s)”, “dibenzodiazepinone compound(s)”, and equivalent expressions refer to a class of dibenzodiazepinone compounds containing a farnesyl moiety or being derived from a farnesyl moiety, and pharmaceutically acceptable salts, solvates and prodrugs thereof.
  • the term includes a compound of Formula I, a compound selected from Compounds 2 to 27, or the exemplified compounds of the present invention, Compounds 2 to 5, 7, 13, 14, 15, 18, and 21 to 25, or a pharmaceutically acceptable salt, solvate or prodrug of any of the above compounds.
  • dibenzodiazepinone analogues includes compounds of this class that can be used as intermediates in chemical syntheses and variants containing different isotopes than the most abundant isotope of an atom (e.g, D replacing H, 13 C replacing 12 C, etc).
  • the compounds of the invention are also sometimes referred as "active ingredients”.
  • chemical modification refers to one or more steps of modifying a dibenzodiazepinone compound, referred to as "starting material", by chemical synthesis.
  • starting material Preferred compound used as starting materials in a chemical modification process is Compound 1.
  • chemical modification steps include ⁇ /-alkylations, O-acylations, aromatic bromination, and modifications of the double bonds of the farnesyl side chain including, hydrogenation and hydroalkoxylation.
  • Farnesyl side chain modification reaction can be partial (one or two double bonds modified) or complete (three double bonds modified).
  • Chemical modification steps are also defined in the Schemes of Section MIB, and exemplified in Examples 4 to 8.
  • ester refers to a dibenzodiazepinone analogue obtained by the replacement of a hydrogen atom from an alcohol by a C(O)R" replacement group by an O-acylation reaction as defined in Scheme 1 (b) below, wherein C(O)R" can also be a C- coupled amino acid.
  • ester also encompasses ester equivalents including, without limitation, carbonate, carbamate, and the like. More particularly, the term “ester” encompasses esters of the alcohols in positions 4, 6, and 8 (see Exampes 3-8 for atom numbering).
  • ⁇ /-alkylated derivative refers to a dibenzodiazepinone analogue obtained by the replacement of a hydrogen atom of a nitrogen atom by an R replacement group by an /V-alkylation reaction as defined in Scheme 2(a) below. More particularly, the term “ ⁇ /-alkylated derivative” encompasses substituted derivatives at the nitrogen in position 5 (see Exampes 3-8 for atom numbering).
  • hydroalkoxylated farnesyl derivative refers to a compound having a modified farnesyl side chain at one to three positions by either saturation (addition of two hydrogen atoms) or by addition of a molecule of alcohol (H and OCi- ⁇ alkyl) produced respectively by the procedures generally defined in Schemes 3(a) and (b) of Section IHB, and more specifically in Examples 4 and 7.
  • abbreviations have their common meaning. Unless otherwise noted, the abbreviations “Ac”, “Me”, “Et”, “Pr”, “i-Pr”, “Bu”, “Bz”, “Bn” and “Ph”, respectively refer to acetyl, methyl, ethyl, propyl (n- or /so-propyl), /so-propyl, butyl (n-, iso-, sec- or terf-butyl), benzoyl, benzyl and phenyl.
  • alkyl refers to linear, branched or cyclic, saturated hydrocarbon groups.
  • alkyl groups include, without limitation, methyl, ethyl, n-propyl, isopropyl, n-butyl, pentyl, hexyl, heptyl, cyclopentyl, cyclohexyl, cyclohexylmethyl, and the like.
  • Alkyl groups may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl, oxo, guanidino and formyl.
  • substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, aryloxy, sulfiny
  • Ci -n alkyl refers to an alkyl group having from 1 to the indicated "n" number of carbons.
  • the Ci -n alkyl can be cyclic or a straight or branched chain.
  • linear Ci -n alkyl refers to an alkyl group having from 1 to the indicated "n" number of carbons and being Near, i.e. not cyclic or branched in the vicinity of the attached atom (herein the nitrogen).
  • the C- I- n alkyl can optionally be substituted with groups such as amino, cyano, halo, hydroxyl, nitro, thio, and alkoxy.
  • alkenyl refers to linear, branched or cyclic unsaturated hydrocarbon groups containing, from one to six carbon-carbon double bonds. Examples of alkenyl groups include, without limitation, vinyl, 1-propene-2-yl, 1-butene-4-yl, 2-butene-4-yl, 1- pentene-5-yl and the like.
  • Alkenyl groups may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, ajkoxy, aryloxy, sulfinyl, sulfonyl, formyl, oxo and guanidino.
  • the double bond portion(s) of the unsaturated hydrocarbon chain may be either in the cis or trans configuration.
  • C 2 - n alkenyl refers to an alkenyl group having from 2 to the indicated "n" number of carbons.
  • the C- 2-n alkenyl can be cyclic or a straight or branched chain.
  • alkynyl refers to linear, branched or cyclic unsaturated hydrocarbon groups containing at least one carbon-carbon triple bond. Examples of alkynyl groups include, without limitation, ethynyl, 1-propyne-3-yl, 1-butyne-4-yl, 2-butyne-4-yl, 1- pentyne-5-yl and the like.
  • Alkynyl groups may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl, formyl, oxo and guanidine.
  • substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, aryloxy, sulfin
  • C 2-n alkynyr wherein n is an integer from 3 to 12, refers to an alkynyl group having from 2 to the indicated "n" number of carbons.
  • the C 2-n alkynyl can be cyclic or a straight or branched chain.
  • cycloalkyl or "cycloalkyl ring” refers to an alkyl group, as defined above, further comprising a saturated or partially unsaturated carbocyclic ring in a single or fused carbocyclic ring system having from three to fifteen ring members.
  • cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclopenten-1-yl, cyclopenten-2-yl, cyclopenten-3-yl, cyclohexyl, cyclohexen-1-yl, cyclohexen-2-yl, cyclohexen-3-yl, cycloheptyl, bicyclo[4,3,0]nonanyl, norbornyl, and the like.
  • Cycloalkyl groups may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • C 3-n cycloalkyl wherein n is an integer from 4 to 15, refers to a cycloalkyl ring or ring system or having from 3 to the indicated "n" number of carbons.
  • heterocycloalkyl refers to a cycloalkyl group, as defined above, further comprising one to four hetero atoms (e.g. N, O, S, P) or hetero groups (e.g. NH, NR X , PO 2 , SO, SO 2 ) in a single or fused heterocyclic ring system having from three to fifteen ring members (e.g. tetrahydrofuranyl has five ring members, including one oxygen atom).
  • hetero groups e.g. NH, NR X , PO 2 , SO, SO 2
  • heterocycloalkyl, heterocyclic or heterocycloalkyl ring examples include, without limitation, pyrrolidino, tetrahydrofuranyl, tetrahydrodithienyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1 ,2,3,6-t ⁇ trahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1 ,3-dioxolanyl, pyra
  • heterocycloalkyl groups may be C-attached or N-attached where such is possible.
  • Heterocycloalkyl, heterocyclic or heterocycloalkyl ring may optionally be substituted with substituents selected from acyl, amino, acylamino, acyloxy, oxo, thiocarbonyl, imino, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • C 3-n heterocycloalkyr' wherein n is an integer from 4 to 15, refers to an heterocycloalkyl group having from 3 to the indicated "n" number of atoms in the cycle and at least one hetero group as defined above.
  • halo or halogen refers to bromine, chlorine, fluorine or iodine substituents.
  • aryl refers to common aromatic groups having "4n+2" ⁇ (pi) electrons, wherein n is an integer from 1 to 3, in a conjugated monocyclic or polycyclic system and having from five to fourteen ring atoms.
  • Aryl may be directly attached, or connected via a C- ⁇ -3 alkyl group (also referred to as aralkyl).
  • Examples of aryl include, without limitation, phenyl, benzyl, phenethyl, 1-phenylethyl, tolyl, naphthyl, biphenyl, terphenyl, and the like.
  • Aryl groups may optionally be substituted with one or more substituent group selected from acyl, amino, acylamino, acyloxy, azido, alkythio, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • substituent group selected from acyl, amino, acylamino, acyloxy, azido, alkythio, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy,
  • C 5-n aryr wherein n is an integer from 5 to 14, refers to an aryl group having from 5 to the indicated "n" number of atoms, including carbon, nitrogen, oxygen and sulfur.
  • the C 5-n aryl can be mono or polycyclic.
  • heteroaryl refers to an aryl ring, as defined above, further containing one to four heteroatoms selected from oxygen, nitrogen, sulphur or phosphorus.
  • heteroaryl include, without limitation, pyridyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, tetrazolyl, furyl, thienyl, isooxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrollyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl,
  • Heteroaryl may optionally be substituted with one or more substituent group selected from acyl, amino, acylamino, acyloxy, azido, alkythio, carboalkoxy, carboxy, carboxyamido, cyano, halo, hydroxyl, nitro, thio, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, aryloxy, sulfinyl, sulfonyl and formyl.
  • Heteroaryl may be directly attached, or connected via a C h alky! group (also referred to as heteroaralkyl).
  • the foregoing heteroaryl groups as derived from the compounds listed above, may be C-attached or N-attached where such is possible.
  • C 5-n heteroaryl wherein n is an integer from 5 to 14, refers to an heteroaryl group having from 5 to the indicated "n" number of atoms, including carbon, nitrogen, oxygen and sulphur atoms.
  • the C 5-n heteroaryl can be mono or polycyclic.
  • amino acid refers to an organic acid containing an amino group.
  • the term includes both naturally occurring and synthetic amino acids; therefore, the amino group can be but is not required to be, attached to the carbon next to the acid.
  • a C- coupled amino acid substituent is attached to the heteroatom (nitrogen or oxygen) of the parent molecule via its carboxylic acid function.
  • C-coupled amino acid forms an ester with the parent molecule when the heteroatom is oxygen, and an amide when the heteroatom is nitrogen.
  • amino acids include, without limitation, alanine, valine, leucine, isoleucine, proline, phenylalanine, tryptophan, methionine, glycine, serine, threonine, cysteine, asparagine, glutamine, tyrosine, histidine, lysine, arginine, aspartic acid, glutamic acid, desmosine, ornithine, 2-aminobutyric acid, cyclohexylalanine, dimethylglycine, phenylglycine, norvaline, norleucine, hydroxylysine, allo-hydroxylysine, hydroxyproline, isodesmosine, allo-isoleucine, ethylglycine, beta- alanine, aminoadipic acid, aminobutyric acid, ethyl asparagine, and N-methyl amino acids.
  • Amino acids can be pure L or D isomers or mixtures of L and D is
  • the compounds of the present invention can possess one or more asymmetric carbon atoms and can exist as optical isomers forming mixtures of racemic or non- racemic compounds.
  • the compounds of the present invention are useful as single isomers or as a mixture of stereochemical isomeric forms.
  • Diastereoisomers, i.e., nonsuperimposable stereochemical isomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, including chiral chromatography (e.g. HPLC), immunoassay techniques, or the use of covalently (e.g. Mosher's esters) or non-covalently (e.g.
  • chiral salts bound chiral reagents to respectively form a diastereomeric ester or salt, which can be further separated by conventional methods, such as chromatography, distillation, crystallization or sublimation. The chiral ester or salt is then cleaved or exchanged by conventional means, to recover the desired isomer(s).
  • the invention encompasses isolated or purified compounds.
  • An "isolated” or “purified” compound refers to a compound which represents at least 10%, 20%, 50%,. 80% or 90% of the mixture by weight, provided that the mixture comprising the compound of the invention has demonstrable (i.e. statistically significant) biological activity including cytostatic, cytotoxic, enzyme inhibitory or receptor binding action when tested in conventional biological assays known to a person skilled in the art.
  • salts refers to nontoxic salts synthesized from a compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, methanol, ethanol, isopropanol, or acetonitrile are preferred.
  • Another method for the preparation of salts is by the use of ion exchange resins.
  • salt includes both acid addition salts and base addition salts, either of the parent compound or of a prodrug or solvate thereof.
  • the nature of the salt is not critical, provided that it is pharmaceutically acceptable.
  • Exemplary acids used in acid addition salts include, without limitation, hydrochloric, hydrobromic, hydroiodic, nitric, carbonic, sulfuric, sulfonic, phosphoric, formic, acetic, citric, tartaric, succinic, oxalic, malic, glutamic, propionic, glycolic, gluconic, maleic, embonic (pamoic), methanesulfonic, ethanesulfonic, 2-hydroxyethanesulfonic, pantothenic, benzenesulfonic, toluenesulfonic, sulfanilic, mesylic, cyclohexylaminosulfonic, stearic, algenic, ⁇ -hydroxybutyric, malonic,
  • Suitable pharmaceutically acceptable base addition salts include, without limitation, metallic salts made from aluminium, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts, such as those made from /V, ⁇ /'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine, lysine, procaine and the like. Additional examples of pharmaceutically acceptable salts are listed in Berge et al (1977), Journal of Pharmaceutical Sciences, vol 66, no 1 , pp 1-19, the content of which is incorporated herein by reference in its entirety.
  • solvate refers to a physical association of a compound of this invention with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate” encompasses both solution-phase and isolable solvates. Exemplary solvates include hydrates, ethanolates, methanolates, hemiethanolates, and the like.
  • pharmaceutically acceptable prodrug means any pharmaceutically acceptable ester, salt of an ester or any other derivative of a compound of this invention, which upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or a biologically active metabolite or residue thereof.
  • Particularly favored salts or prodrugs are those with improved properties, such as solubility, efficacy, or bioavailability of the compounds of this invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
  • a prodrug is a drug having one or more functional groups covalently bound to a carrier wherein metabolic or chemical release of the drug occurs in vivo when the drug is administered to a mammalian subject.
  • Pharmaceutically acceptable prodrugs of the compounds of this invention include derivatives of hydroxyl groups such as, without limitation, acyloxymethyl, acyloxyethyl and acylthioethyl ethers, esters, amino acid esters, phosphate esters, sulfonate and sulfate esters, and their metal salts, and the like.
  • the invention relates to novel dibenzodiazepinone analogues, referred to herein as the compounds of the invention, and to pharmaceutically acceptable salts, solvates and prodrugs thereof.
  • the compounds of the invention may be characterized by any one of their physicochemical and spectral properties, such as mass and NMR spectra, detailed in Example 4 through Example 8.
  • R 1 is a linear C-i-ioalkyl; or a farnesyl derivative thereof, wherein said farnesyl derivative has one, two or three hydrogenated or hydroalkoxylated double bonds; or a pharmaceutically acceptable salt or prodrug thereof.
  • R 1 is a linear C- M oalkyl, and all other groups are as previously disclosed. In a subclass of this embodiment, the alkyl group is optionally substituted with a substituent selected from halo, fluoro, amino and carboxy, or pharmaceutically acceptable salt thereof.
  • R 1 is methyl.
  • R 1 is ethyl.
  • R 1 is ⁇ -propyl.
  • R 1 is n-butyl.
  • R 1 is n-pe.ntyl.
  • R 1 is n-hexyl. ,ln another embodiment, R 1 is methyl, and the farnesyl is fully hydrogenated (i.e. saturated).
  • R 1 is methyl, and one double bond of the farnesyl is hydrogenated. In another embodiment, R 1 is methyl, and two double bonds of the farnesyl are hydrogenated. In another embodiment, R 1 is a linear C-i-ioalkyl, and one double bond of the farnesyl is hydrogenated. In another embodiment, R 1 is a linear C-
  • R 1 is a linear C- M oalkyl, and two double bonds of the farnesyl are hydromethoxylated.
  • R 1 is methyl, and one double bond of the farnesyl is hydroalkoxylated.
  • R 1 is methyl, and two double bonds of the farnesyl are hydroalkoxylated.
  • R 1 is ethyl, and one double bond of the farnesyl is hydroalkoxylated.
  • R 1 is ethyl, and two double bonds of the farnesyl are hydroalkoxylated.
  • the invention encompasses all esters or A/-alkylated derivatives, and pharmaceutically acceptable salts, solvates and prodrugs of the foregoing compounds.
  • the invention further provides esters and /V-alkylated derivatives of any of the foregoing compounds. Certain embodiments expressly exclude one or more of the compounds of Formula I.
  • Prodrugs of the compounds of the invention include compounds wherein one or more of the 4, 6 and 8-hydroxy groups, or any other hydroxyl group on the molecule is bounded to any group that, when administered to a mammalian subject, is cleaved to form the free hydroxyl group.
  • Examples of prodrugs include, but are not limited to, acetate, formate, hemisuccinate, benzoate, dimethylaminoacetate and phosphoryloxycarbonyl derivatives of hydroxy functional groups; dimethylglycine esters, aminoalkylbenzyl esters, aminoalkyl esters or carb ⁇ xyalkyl esters of hydroxy functional groups. Carbamate and carbonate derivatives of the hydroxy groups are also included.
  • Derivatizations of hydroxyl groups also encompassed, are (acyloxy)methyl and (acyloxy)ethyl ethers, wherein the acyl group contains an alkyl group optionally substituted with groups including, but not limited to, ether, amino and carboxylic acid functionalities, or where the acyl group is an amino acid ester.
  • phosphate and phosphonate esters, sulfate esters, sulfonate esters which are in alkylated (such as bis-pivaloyloxymethyl (POM) phosphate trimester or -P(O)O 2 Et 2 ) or in the salt form (such as sodium phosphate ester (-P(O)O " 2 Na + 2 )).
  • prodrugs used in anticancer therapy and their metabolism see Rooseboom et al (2004), Phamacol. Rev., vol 56, 53-102, incorporated herein by reference.
  • the prodrug may also be prepared as its pharmaceutically acceptable salt.
  • the compounds of this invention may be formulated into pharmaceutical compositions comprised of a compound of the invention, in combination with a pharmaceutically acceptable carrier, as discussed in Section IV below.
  • farnesyl dibenzodiazepinone-producing microorganism and “producer of farnesyl dibenzodiazepinone,” as used herein, refer to a microorganism that carries genetic information necessary to produce a farnesyl dibenzodiazepinone compound, whether or not the organism naturally produces the compound.
  • the terms apply equally to organisms in which the genetic information to produce the farnesyl dibenzodiazepinone compound is found in the organism as it exists in its natural environment, and to organisms in which the genetic information is introduced by recombinant techniques.
  • Compound is produced by isolation of the fermentation broth of
  • the farnesyl dibenzodiazepinone compound may be biosynthesized by various microorganisms.
  • Microorganisms that may synthesize the farnesyl dibenzodiazepinone compound include but are not limited to bacteria of the order Actinomycetales, also referred to as actinomycetes.
  • Non-limiting examples of members belonging to the genera of Actinomycetes include Nocardia, Geodermatophilus, Actinoplanes, Micromonospora, Nocardioides, Saccharothrix, Amycolatopsis, Kutzneria, Saccharomonospora, Saccharopolyspora, Kitasatospora, Streptomyces, Microbispora, Streptosporangium, and Actinomadura.
  • the taxonomy of actinomycetes is complex and reference is made to Goodfellow, Suprageneric Classification of Actinomycetes (1989); Bergey's Manual of Systematic Bacteriology, Vol. 4 (Williams and Wilkins, Baltimore, pp.
  • Famesyl dibenzodiazepinone-producing microorganisms are cultivated in culture medium containing known nutritional sources for actinomycetes. Such media having assimilable sources of carbon, nitrogen, plus optional inorganic salts and other known growth factors, at a pH of about 6 to about 9. Suitable media include, without limitation, the growth media provided in Table 1. Microorganisms are cultivated at incubation temperatures of about 18 0 C to about 40 0 C for about 3 to about 40 days.
  • the culture media inoculated with a famesy dibenzodiazepinone-producing microorganism may be aerated by incubating the inoculated culture media with agitation, for example, shaking on a rotary shaker, a shaking water bath, or in a fermentor. Aeration may also be achieved by the injection of air, oxygen or an appropriate gaseous mixture to the inoculated culture media during incubation.
  • the farnesyl dibenzodiazepinone compound can be extracted and isolated from the cultivated culture media by techniques known to a person skilled in the art and/or disclosed herein, including for example centrifugation, chromatography, adsorption, filtration.
  • the cultivated culture media can be optionally acidified and mixed with a suitable organic solvent such as methanol, ethanol, n- butanol, ethyl acetate, n-butyl acetate or 4-methyl-2-pentanone.
  • the organic layer can be separated from the mycelial cake for example, by centrifugation and decantation or filtration.
  • the mycelial cake is further optionally extracted with an organic solvent, and the organic extracts combined.
  • the organic layer is further optionally treated, for example by: aqueous washings, precipitation, filtration and the like, followed the removal of the solvent, for example, by evaporation to dryness under vacuum.
  • the resulting residue can optionally be reconstituted with for example water, ethyl ether, ethanol, ethyl acetate, methanol or a mixture thereof, and re-extracted in a two-phase system with a suitable organic solvent such as hexane, carbon tetrachloride, methylene chloride or a mixture thereof.
  • a suitable organic solvent such as hexane, carbon tetrachloride, methylene chloride or a mixture thereof.
  • the compound can be further purified by the use of standard techniques such as normal and reverse-phase liquid chromatography, crystallization, sublimation, adsorption, mass exclusion chromatography, and the like.
  • the farnesyl dibenzodiazepinone Compound 1 is biosynthesized by microorganisms and isolated as described herein, and in Canadian patent 2,466,340 (and PCT/CA04/000069). Compound 1 is subjected to random and/or directed chemical modifications to form compounds that are derivatives or structural analogues. Such derivatives or structural analogues having similar functional activities are within the scope of the present invention.
  • the farnesyl dibenzodiazepinone may be modified by one or more chemical modification steps, using methods known in the art and described herein. Examples of chemical modifications procedures are also provided in Examples 4 to 8.
  • Dibenzodiazepinone analogues that are derivatives of Compound 1 , for example those identified herein as the compounds of Formula I and their derivatives, and Compounds 2 to 27, are generated by standard organic chemistry approaches.
  • General principles of organic chemistry required for making and manipulating the compounds described herein, including functional moieties, reactivity and common protocols are described, for example, in "Advanced Organic Chemistry," 4 th Edition by Jerry March (1992), Wiley-lnterscience, USA, incorporated herein by reference in its entirety.
  • the synthetic methods described herein may use a variety of protecting groups, whether or not they are explicitly described.
  • a "protecting group” as used herein means a moiety used to block one or more functional moieties such as reactive groups including oxygen, sulfur or nitrogen, so that a reaction can be carried out selectively at another reactive site in a polyfunctional compound.
  • functional moieties such as reactive groups including oxygen, sulfur or nitrogen
  • General principles for the use of protective groups, their applicability to specific functional groups and their uses are described for example in T. H. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, New York (1999), incorporated herein by reference in its entirety.
  • Alcohols and phenols are protected, if necessary with, for example: silyl ethers (TMS: trimethylsilyl, TIPS: triisopropylsilyl), acetals (MOM: methyloxymethyl, BOM: benzyloxymethyl), esters (acetate, benzoyl) and ethers (Bn: benzyl).
  • TMS trimethylsilyl
  • TIPS triisopropylsilyl
  • MOM methyloxymethyl
  • BOM benzyloxymethyl
  • esters acetate, benzoyl
  • Bn benzyl
  • Alcohols are deprotected by conditions such as: TBAF (tetrabutylammonium fluoride) for silyl ethers, aqueous acid catalysis for acetals and esters, saponification for esters, and hydrogenolysis for Bn and BOM.
  • TBAF tetrabutylammonium fluoride
  • Amine is protected, if necessary, using standard amino acid protecting groups, for example, carbamates (such as t-butyl (BOC) and benzyl (CBZ)), fluorene derivatives (such as FMOC: /V-(9-fluorenylmethoxycarbonyl)-), etc.
  • Amine is deprotected by conditions such as: acid hydrolysis for BOC, hydrogenolysis for CBZ, or base treatment for FMOC. All protection and deprotection conditions are demonstrated in the Greene et al reference above.
  • R" is an acetate
  • phenolic alcohols are converted to esters when reacted with activated carboxylic acids (R 11 C(O)X) such as an acid halide, anhydride, N- hydroxysuccinimide ester, or a carboxylic acid activated by a coupling agent (e.g.: EDC (1-(3-dimethylaminopropyl)-3-d ' fi ' sopropylethyIcarbodiimide hydrochloride); or HATU (O- (7-azabenzotriazol-1-yl)- ⁇ /, ⁇ /, ⁇ /',/ ⁇ /-tetramethyluronium hexafluorophosphate)) with a base (e.g., pyridine or ⁇ /, ⁇ /-diisopropylethylamine (DIPEA)) and optional acatalysts such HOBt (1-hydroxybenzotriazole hydrate) and/or DMAP (4-(dimethylamino)pyr
  • Scheme 1 is used to obtain, for example, Compounds 25 from Compound 1 ; and to produce esters of the Compounds of the invention.
  • Scheme 2 Amine modifications ( ⁇ /-alkylations)
  • R is selected an optionally substituted linear C- M oalkyl.
  • amine group in position 5 (for position, see Example 3) is alkylated.
  • an amine is alkylated using an RX alkylating agent such as dialkyl sulfates and alkyl halides, preferably in the presence of a base (e.g., sodium bicarbonate, pyridine and the like).
  • RX alkylating agent such as dialkyl sulfates and alkyl halides, preferably in the presence of a base (e.g., sodium bicarbonate, pyridine and the like).
  • Scheme 2 is used to prepare, for example, Compounds 2 to 13 from Compound 1 , and Compound 14 from Compound 23; and to produce any of the Compounds of Formula I comprising an ⁇ /-alkyl group.
  • R z is OCi -6 alkyl
  • double bond is modified by: (a) hydrogenation; and (b) hydroalkoxylation.
  • hydrogenation is carried out using a hydrogen source (e.g. hydrogen, formic acid) and a catalyst (such as rhodium, platinum, or palladium).
  • a hydrogen source e.g. hydrogen, formic acid
  • a catalyst such as rhodium, platinum, or palladium.
  • electrophilic addition to the double bond is achieved by the formation of a carbocation from addition of a proton in acidic conditions (e.g., p-toluene sulfonic acid, alkyl sulfate/NaHCO 3 /MeOH, and the like), and trapping of the carbocation with an alcohol (C- ⁇ - 6 alkyl alcohol, hydroalkoxylation).
  • Scheme 3 is used to obtain, for example: in (a) Compound 23 from Compound 1 , and Compounds 14 to 16 from Compound 2; in (b) Compounds 17 to 19 from Compound 2, Compounds 20 to 22 from Compound 3.
  • Schemes 3 (a) and (b) are also used to produce any hydrogenated and hydroalkoxylated farnesyl derivative of the Compounds of Formula I.
  • brominating agents include bromine, /V-haloamides (e.g, ⁇ /-bromosuccinimide (NBS), tetraalkylammonium polyhalides).
  • Scheme 4 is used to prepare Compound 24 from Compound 1 , Compound 26 from Compound 2 and Compound 27 from Compound 14.
  • Prodrugs are prepared by routine chemical modifications such as described in Jerry March, supra, including esterification (Scheme 1 ) and alkylation reactions, i.e., use of activated acids or mixed anhydrides (acyl halides, use of coupling reagents, etc), and by the use of alkylating agents (R-X, wherein X is a leaving group, such as diazo, and R is the desired group).
  • Phosphate prodrugs are prepared by phosphorylation, for example, with dialkyl phosphites using procedure such as described in Silverberg et al. (1996), Tet. Lett, Vol. 37, 711-774, U.S.
  • compositions comprising the compounds of the invention
  • the invention provides a pharmaceutical composition comprising a compound of the invention, or a pharmaceutically acceptable salt, solvate or prodrug thereof, in combination with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprising a dibenzodiazepinone analogue is useful for treating diseases and disorders associated with uncontrolled cellular growth and proliferation, such as a neoplastic condition.
  • the pharmaceutical composition is also useful in treating other diseases and disorders, including inflammation, autoimmune diseases, infections, neurodegenerative diseases and stress.
  • the pharmaceutical composition comprising a dibenzodiazepinone analogue may be packaged into a convenient commercial package providing the necessary materials, such as the pharmaceutical composition and written instructions for its use in treating a neoplastic condition, in a suitable container.
  • the compounds of the present invention can be formulated for oral, sublingual, intranasal, intraocular, rectal, transdermal, mucosal, topical or parenteral administration for the therapeutic or prophylactic treatment of neoplastic and proliferative diseases and disorders.
  • Parenteral modes of administration include without limitation, intradermal, subcutaneous (s.c, s.q., sub-Q, Hypo), intramuscular (i.m.), intravenous (i.v.), intraperitoneal (i.p.), intra-arterial, intramedulary, intracardiac, intra-articular (joint), intrasynovial (joint fluid area), intracerebral or intracranial, intraspinal, intracistemal, and intrathecal (spinal fluids). Any known device useful for parenteral injection or infusion of drug formulations can be used to effect such administration.
  • compositions comprising a compound of the present invention will contain from about 0.1 % to about 99.9%, about 1% to about 98%, about 5% to about 95%, about 10% to about 80% or about 15% to about 60% by weight of the active compound.
  • the pharmaceutical preparations disclosed herein are prepared in accordance with standard procedures and are administered at dosages that are selected to reduce, prevent, or eliminate cancer.
  • dosages that are selected to reduce, prevent, or eliminate cancer.
  • unit dosage refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of dibenzodiazepinone analogue calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutically acceptable carriers.
  • the unit dosage contains from 10 to 3000 mg of active ingredient. In another embodiment, the unit dosage contains 20 to 1000 mg of active ingredient.
  • the compositions of the present invention can be delivered using controlled (e.g., capsules) or sustained release delivery systems (e.g., bioerodable matrices). Exemplary delayed release delivery systems for drug delivery that are suitable for administration of the compositions of the invention are described in U.S. Patent Nos 4,452,775 (issued to Kent), 5,039,660 (issued to Leonard), and 3,854,480 (issued to Zaffaroni), incorporated herein by reference in their entirety.
  • compositions of the present invention comprise one or more compounds of the present invention in association with one or more non ⁇ toxic, pharmaceutically-acceptable carriers and/or diluents and/or adjuvants and/or excipients, collectively referred to herein as "carrier” materials, and if desired other active ingredients.
  • Pharmaceutically acceptable carriers include, for example, solvents, vehicles or medium such as saline, buffered saline, dextrose, water, glycerol, ethanol, propylene glycol, polysorbate 80 (Tween-80TM), poly(ethylene) glycol 300 and 400 (PEG 300 and 400), PEGylated castor oil (E.g.
  • Cremophor EL poloxamer 407 and 188
  • hydrophobic carriers include, for example, fat emulsions, lipids, PEGylated phopholids, polymer matrices, biocompatible polymers, lipospheres, vesicles, particles, and liposomes. The term specifically excludes cell culture medium.
  • Excipients or additives included in a formulation have different purposes depending, for example on the nature of the drug, and the mode of administration.
  • Examples of generally used excipients include, without limitation: stabilizing agents, solubilizing agents and surfactants, buffers, antioxidants and preservatives, tonicity agents, bulking agents, lubricating agents, emulsifiers, suspending or viscosity agents, inert diluents, fillers, disintegrating agents, binding agents, wetting agents, lubricating agents, antibacterials, chelating agents, sweetners, perfuming agents, flavouring agents, coloring agents, administration aids, and combinations thereof.
  • compositions may contain common carriers and excipients, such as cornstarch or gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid.
  • the compositions may contain crosarmellose sodium, microcrystalline cellulose, sodium starch glycolate and alginic acid.
  • Formulations for parenteral administration can be in the form of aqueous or non ⁇ aqueous isotonic sterile injection solutions, suspensions or fat emulsions, comprising a compound of this invention, or a pharmaceutically acceptable salt or prodrug thereof.
  • the parenteral form used for injection must be fluid to the extent that easy syringability exists.
  • solutions or suspensions can be prepared from sterile concentrated liquids, powders or granules.
  • the compounds can be dissolved in a carrier such as a solvent or vehicle, for example, polyethylene glycol, propylene glycol, ethanol, corn oil, benzyl alcohol, glycofurol, ⁇ /,/V-dimethylacetamide, /V-methylpyrrolidone, glycerine, saline, dextrose, water, glycerol, hydrophobic carriers, and combinations thereof.
  • a carrier such as a solvent or vehicle, for example, polyethylene glycol, propylene glycol, ethanol, corn oil, benzyl alcohol, glycofurol, ⁇ /,/V-dimethylacetamide, /V-methylpyrrolidone, glycerine, saline, dextrose, water, glycerol, hydrophobic carriers, and combinations thereof.
  • Excipients used in parenteral preparations also include, without limitation,
  • cetrimide sodium docusate., glyceryl monooleate, polyvinylpyrolidone > (PVP) and polyethylene glycol (PEG)) and surfactants (e.g. polysorbates, tocopherol PEG succinate, poloxamer and CremophorTM), buffers (e.g. acetates, citrates, phosphates, tartrates, lactates, succinates, amino acids and the like), antioxidants and preservatives (e.g.
  • BHA, BHT, gentisic acids such as sulfites, bisulfites, metabisulfites, thioglycerols, thioglycolates and the like), tonicity agents (for adjusting physiological compatibility), suspending or viscosity agents, antibacterials (e.g. thimersol, benzethonium chloride, benzalkonium chloride, phenol, cresol and chlorobutanol), chelating agents, and administration aids (e.g. local anesthetics, anti-inflammatory agents, anti-clotting agents, vaso-constrictors for prolongation and agents that increase tissue permeability), and combinations thereof.
  • agents such as sulfites, bisulfites, metabisulfites, thioglycerols, thioglycolates and the like
  • tonicity agents for adjusting physiological compatibility
  • suspending or viscosity agents e.g. thimersol, benzethonium chloride, benzal
  • Parenteral formulations using hydrophobic carriers include, for example, fat emulsions and formulations containing lipids, lipospheres, vesicles, particles and liposomes.
  • Fat emulsions include in addition to the above-mentioned excipients, a lipid and an aqueous phase, and additives such as emulsifiers (e.g. phospholipids, poloxamers, polysorbates, and polyoxyethylene castor oil), and osmotic agents (e.g. sodium chloride, glycerol, sorbitol, xylitol and glucose).
  • emulsifiers e.g. phospholipids, poloxamers, polysorbates, and polyoxyethylene castor oil
  • osmotic agents e.g. sodium chloride, glycerol, sorbitol, xylitol and glucose.
  • Liposomes include natural or derived phospholipids and optionally stabilizing agents such as
  • the parenteral unit dosage form of the compound can be a ready-to-use solution of the compound in a suitable carrier in sterile, hermetically sealed ampoules or in sterile pre-loaded syringes.
  • the suitable carrier optionally comprises any of the above-mentioned excipients.
  • the unit dosage of the compound of the present invention can be in a concentrated liquid, powder or granular form for ex tempore reconstitution in the appropriate pharmaceutically acceptable carrier at the time of delivery.
  • powder forms optionally include bulking agents (e.g. mannitol, glycine, lactose, sucrose, trehalose, dextran, hydroxyethyl starch, ficoll and gelatin), and cryo or lyoprotectants.
  • a sterile formulation of the compound of formula I and optionally one or more additives, including solubilizers or surfactants, can be dissolved or suspended in any of the commonly Used intravenous fluids and administered by infusion.
  • Intravenous fluids include, without limitation, physiological saline, phosphate buffered saline, 5% glucose or Ringer'sTM solution.
  • a sterile formulation of the compound of the present invention or suitable soluble salts or prodrugs forming the compound can be dissolved and administered in a pharmaceutical diluent such as Water-for-lnjection (WFI), physiological saline or 5% glucose.
  • WFI Water-for-lnjection
  • a suitable insoluble form of the compound may be prepared and administered as a suspension in an aqueous base or a pharmaceutically acceptable oil base, e.g. an ester of a long chain fatty acid such as ethyl oleate.
  • solid formulations such as tablets and capsules are particularly useful. Sustained released or enterically coated preparations may also be devised.
  • the pharmaceutical compositions are in the form of, for example, tablets, capsules, suspensions or liquid syrups or elixirs, wafers and the like.
  • excipient or additives include, but are not limited to inert diluents, fillers, disintegrating agents, binding agents, wetting agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives.
  • the oral pharmaceutical composition is preferably made in the form of a unit dosage containing a therapeutically-effective amount of the active ingredient.
  • Examples of such dosage units are tablets and capsules.
  • the tablets and capsules which can contain, in addition to the active ingredient, conventional carriers such as: inert diluents (e.g., sodium and calcium carbonate, sodium and calcium phosphate, and lactose), binding agents (e.g., acacia gum, starch, gelatin, sucrose, polyvinylpyrrolidone (Providone), sorbitol, or tragacanth methylcellulose, sodium carboxymethylcellulose, hydroxypropyl methylcellulose, and ethylcellulose), fillers (e.g., calcium phosphate, glycine, lactose, maize-starch, sorbitol, or sucrose), lubricants or lubricating agents (e.g., magnesium stearate or other metallic stearates, stearic acid, polyethylene glycol, waxes, oils, silica and colloical silica, silicon fluid or talc), disintegrants or dis
  • Oral liquid preparations may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous agents, preservatives, coloring agents and flavoring agents.
  • additives for liquid preparations include acacia, almond oil, ethyl alcohol, fractionated coconut oil, gelatin, glucose syrup, glycerin, hydrogenated edible fats, lecithin, methyl cellulose, methyl or propyl para ⁇ hydroxybenzoate, propylene glycol, sorbitol, or sorbic acid.
  • flavoring agents such as peppermint, oil of wintergreen, cherry, grape, fruit flavoring or the like can also be used. It may also be desirable to add a coloring agent to make the dosage form more aesthetic in appearance or to help identify the product.
  • the compounds of present invention can also be prepared in suitable forms to be applied to the skin, or mucus membranes of the nose and throat, and can take the form of creams, ointments, liquid sprays or inhalants, lozenges, or throat paints.
  • Such topical formulations further can . include chemical compounds such as dimethylsulfoxide (DMSO) to facilitate surface penetration of the active ingredient.
  • DMSO dimethylsulfoxide
  • the compounds of the present invention can be presented in liquid or semi-liquid form formulated in hydrophobic or hydrophilic bases as ointments, creams, lotions, paints or powders.
  • the compounds of the present invention can be administered in the form of suppositories admixed with conventional carriers such as cocoa butter, wax or other glyceride.
  • the invention relates to a method for inhibiting growth and/or proliferation of cancer cells in a mammal.
  • the invention provides a method for treating neoplasms in a mammal.
  • Mammals include ungulates (e.g. sheeps, goats, cows, horses, pigs), and non-ungulates, ' including rodents, felines, canines and primates (i.e. human and non-human primates).
  • the mammal is a human.
  • the dibenzodiazepinone analogues of the present invention may bind to or interact with other cancer-associated proteins and polypeptides, including, without limitation, polypeptides encoded by oncogenes, polypeptides that induce angiogenesis, proteins involved in metastasizing and/or invasive processes, and proteases that regulate apoptosis and the cell cycle. Regardless of the mechanism of action, the dibenzodiazepinone analogues of the invention have been demonstrated to exhibit anti ⁇ cancer activity both in vitro and in vivo. Based on these discoveries, applicants have developed methods for treating neoplasms.
  • neoplasm As used herein, the terms “neoplasm”, “neoplastic disorder”, “neoplasia” “cancer,” “tumor” and “proliferative disorder” refer to cells having the capacity for autonomous growth, i.e., an abnormal state of condition characterized by rapidly proliferating cell growth which generally forms a distinct mass that show partial or total lack of structural organization and functional coordination with normal tissue.
  • the terms are meant to encompass hematopoietic neoplasms (e.g. lymphomas or leukemias) as well as solid neoplasms (e.g.
  • Hematopoietic neoplasms are malignant tumors affecting hematopoietic structures (structures pertaining to the formation of blood cells) and components of the immune system, including leukemias (related to leukocytes (white blood cells) and their precursors in the blood and bone marrow) arising from myeloid, lymphoid or erythroid lineages, and lymphomas (relates to lymphocytes).
  • Solid neoplasms include sarcomas, which are malignant neoplasms that originate from connective tissues such as muscle, cartilage, blood vessels, fibrous tissue, fat or bone.
  • Solid neoplasms also include carcinomas, which are malignant neoplasms arising from epithelial structures (including external epithelia (e.g., skin and linings of the gastrointestinal tract, lungs, and cervix), and internal epithelia that line various glands (e.g., breast, pancreas, thyroid).
  • neoplasms that are particularly susceptible to treatment by the methods of the invention include leukemia, and hepatocellular cancers, sarcoma, vascular endothelial cancers, breast careers, central nervous system cancers (e.g. astrocytoma, gliosarcoma, neuroblastoma, oligodendroglioma and glioblastoma), prostate cancers, lung and bronchus cancers, larynx cancers, esophagus cancers, colon cancers, colorectal cancers, gastro-intestinal cancers, melanomas, ovarian and endometrial cancer, renal and bladder cancer, liver cancer, endocrine cancer (e.g. thyroid), and pancreatic cancer.
  • leukemia and hepatocellular cancers
  • sarcoma vascular endothelial cancers
  • breast careers central nervous system cancers (e.g. astrocytoma, gliosarcoma, neuroblastoma, oli
  • the dibenzodiazepinone analogue is brought into contact with or introduced into a cancerous cell or tissue.
  • the methods of the invention for delivering the compositions of the invention in vivo utilize art-recognized protocols for delivering therapeutic agents with the only substantial procedural modification being the substitution of the dibenzodiazepinone analogue of the present invention for the therapeutic agent in the art-recognized protocols.
  • the route by which the dibenzodiazepinone analogue is administered, as well as the formulation, carrier or vehicle will depend on the location as well as the type of the neoplasm. A wide variety of administration routes can be employed.
  • the dibenzodiazepinone analogue may be administered by intravenous or intraperitoneal infusion or injection.
  • the compound of the invention may be administered by injection directly into the neoplasm.
  • the compound may be administered intravenously or intravascularly.
  • the compound may be administered in a manner such that it can be transported systemically through the body of the mammal and thereby reach the neoplasm and distant metastases for example intrathecal ⁇ , intravenously or intramuscularly or orally.
  • the compound can be administered directly to the tumor.
  • the compound can also be administered subcutaneously, intraperitoneally, topically (for example for melanoma), rectally (for example colorectal neoplasm) vaginally (for example for cervical or vaginal neoplasm), nasally or by inhalation spray (for example for lung neoplasm).
  • the dibenzodiazepinone analogue is administered in an amount that is sufficient to inhibit the growth or proliferation of a neoplastic cell, or to treat a neoplastic disorder.
  • the term "inhibition” refers to suppression, killing, stasis, or destruction of cancer cells.
  • the inhibition of mammalian cancer cell growth according to this method can be monitored in several ways. Cancer cells grown in vitro can be treated with the compound and monitored for growth or death relative to the same cells cultured in the absence of the compound.
  • a cessation of growth or a slowing of the growth rate is indicative of cancer cell inhibition (see Anticancer Drug Development Guide: preclinical screening, clinical trials and approval; B.A. Teicher and P.A. Andrews, ed., 2004, Humana Press, Totowa, NJ).
  • cancer cell inhibition can be monitored by administering the compound to an animal model of the cancer of interest. Examples of experimental non-human animal cancer models are known in the art and described below and in the examples herein.
  • a cessation of tumor growth (i.e., no further increase in size) or a reduction in tumor size (i.e., tumor volume by least a 58%) in animals treated with the compound relative to tumors in control animals not treated with the compound is indicative of significant tumor growth inhibition (see Anticancer Drug Development Guide: preclinical screening, clinical trials and approval; B.A. Teicher and P.A. Andrews, ed., 2004, Humana Press, Totowa, NJ).
  • treatment refers to the application or administration of a dibenzodiazepinone analogue to a mammal, or application or administration of a dibenzodiazepinone analogue to an isolated tissue or cell line from a mammal, who has a neoplastic disorder, a symptom of a neoplastic disorder or a predisposition toward a neoplastic disorder, with the purpose to cure, heal, alleviate, relieve, alter, ameliorate, improve, or control the disorder, the symptoms of disorder, or the predisposition toward disorder.
  • treating is defined as administering, to a mammal, an amount of a dibenzodiazepinone analogue sufficient to result in the prevention, reduction or elimination of neoplastic cells in a mammal ("therapeutically effective amount").
  • the therapeutically effective amount and timing of dosage will be determined on an individual basis and may be based, at least in part, on consideration of the age, body weight, sex, diet and general health of the recipient subject, on the nature and severity of the disease condition, and on previous treatments and other diseases present. Other factors also include the route and frequency of administration, the activity of the administered compound, the metabolic stability, length of action and excretion of the compound, drug combination, the tolerance of the recipient subject to the compound and the type of neoplasm or proliferative disorder.
  • a therapeutically effective amount of the compound is in the range of about 0.01 to about 750 mg/kg of body weight of the mammal. In another embodiment, the therapeutically effective amount is in the range of about 0.01 to about 300 mg/kg body weight per day. In yet another embodiment, the therapeutically effective amount is in the range of 10 to about 50 mg/kg body weight per day.
  • the therapeutically effective doses of the above embodiments may also be expressed in milligrams per square meter (mg/m 2 ) in the case of a human patient. Conversion factors for different mammalian species may be found in:Freireich et al, Quantitative comparison of toxicity of anticancer agents in mouse, rat, dog, monkey and man, . Cancer Chemoth.
  • tumor size and/or tumor morphology is measured before and after initiation of the treatment, and treatment is considered effective if either the tumor size ceases further growth, or if the tumor is reduced in size, e.g., by at least 10% or more (e.g., 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or even 100%, that is, the absence of the tumor).
  • Prolongation of survival, time-to-disease progression, partial response and objective response rate are surrogate measures of clinical activity of the investigational agent.
  • Tumor shrinkage is considered to be one treatment-specific response. This system is limited by the requirement that patients have visceral masses that are amenable to accurate measurement.
  • Methods of determining the size of a tumor in vivo vary with the type of tumor, and include, for example, various imaging techniques well known to those in the medical imaging or oncology fields (MRI, CAT, PET, etc.), as well as histological techniques and flow cytometry.
  • evaluation of serum tumor markers are also used to evaluate response (eg prostate-specific antigen (PSA) for prostate cancer, and carcino-embryonic antigen (CEA), for colon cancer).
  • PSA prostate-specific antigen
  • CEA carcino-embryonic antigen
  • Other methods of monitoring cancer growth include cell counts (e.g. in leukemias) in blood or relief in bone pain (e.g. prostate cancer).
  • the dibenzodiazepinone compound may be administered once daily, or the compound may be administered as two, three, four, or more sub-doses at appropriate intervals throughout the day. In that case, the dibenzodiazepinone compound contained in each sub-dose must be correspondingly smaller in order to achieve the total daily dosage.
  • the dosage unit can also be compounded for delivery over several days, e.g., using a conventional sustained release formulation which provides sustained release of the dibenzodiazepinone compound over a several day period. Sustained release formulations are well known in the art. In this embodiment, the dosage unit contains a corresponding multiple of the daily dose.
  • the effective dose can be administered either as a single administration event (e.g., a bolus injection) or as a slow injection or infusion, e.g. over 30 minutes to about 24 hours.
  • the compound may be administered as a treatment, for up to 30 days.
  • treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or a . series of treatments (e.g., a four-week treatment repeated 3 times, with a 2 months interval between each treatment).
  • Estimates of effective dosages, toxicities and in vivo half-lives for the dibenzodiazepinone compounds encompassed by the invention can be made using conventional methodologies or on the basis of in vivo testing using an appropriate animal model.
  • the dibenzodiazepinone compound may be administered in conjunction with or in addition to known other anticancer treatments such as radiotherapy, or other known anticancer.compounds or chemotherapeutic agents.
  • agents include, but are not limited to, 5-flurouracil, mitomycin C, methotrexate, hydroxyurea, cyclophosphamide, dacarbazine, mitoxantrone, anthracyclines (Epirubicin and Doxurubicin), etopside, pregnasome, platinum compounds such as carboplatin and cisplatin, taxanes such as PaclitaxelTM and DocetaxelTM; hormone therapies such as tamoxifen and anti- estrogens; antibodies to receptors, such as herceptin and Iressa; aromatase inhibitors, progestational agents and LHRH analogues; biological response modifiers such as IL2 and interferons; multidrug reversing agents such as the cyclosporin analogue PSC 8
  • Toxicity and therapeutic efficacy of dibenzodiazepinone compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals. Therapeutic efficacy is determined in animal models as described above and in the examples herein. Toxicity studies are done to determine the lethal dose for 10% of tested animals (LD10). Animals are treated at the maximum tolerated dose (MTD): the highest dose not producing mortality or greater than 20% body weight loss.
  • the effective dose (ED) is related to the MTD in a given tumor model to determine the therapeutic index of the compound.
  • a therapeutic index (MTD/ED) close to 1.0 has been found to be acceptable for some chemotherapeutic drugs, a preferred therapeutic index for classical chemotherapeutic drugs is 1.25 or higher.
  • the data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of compositions of the invention will generally be within a range of circulating concentrations that include the MTD.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range of the compound. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by HPLC.
  • mice Animal models to determine antitumor efficacy of a compound are generally carried out in mice. Either murine tumor cells are inoculated subcutaneously into the hind flank of mice from the same species (syngeneic models) or human tumor cells are inoculated subcutaneously into the hind flank of severe combined immune deficient (SCID) mice or other immune deficient mice (nude mice) (xenograft models).
  • SCID severe combined immune deficient mice
  • nude mice xenograft models
  • MMHCC Mae models of Human Cancer Consortium
  • Cancermodels.nci.nih.gov searchable Cancer Models Database
  • NCI-MMHCC mouse repository NCI-MMHCC mouse repository.
  • Mouse repositories can also be found at: The Jackson Laboratory, Charles River Laboratories, Taconic, Harlan, Mutant Mouse Regional Resource Centers (MMRRC) National Network and at the European Mouse Mutant Archive.
  • Such models may be used for in vivo testing of dibenzodiazepinone compounds, as well as for determining a therapeutically effective dose.
  • pharmaceutically acceptable salts, solvates or prodrugs of said compounds may also be employed in compositions to treat or prevent the above-identified disorders.
  • Micromonospora sp. (deposit accession number IDAC 070303-01 ) was maintained on agar plates of ISP2 agar (Difco Laboratories, Detroit, Ml).
  • An inoculum for the production phase was prepared by transferring the surface growth of the Micromonospora sp. from the agar plates to 125-mL flasks containing 25 mL of sterile medium comprised of glucose 10g, potato dextrin type IV (Sigma) 20 g, yeast extract 5 g, N Z Amine-A 5 g, 1 g CaCO 3 made up to one liter with tap water (pH 7.0).
  • the culture was incubated at about 28 0 C for approximately 70-72 hours on a rotary shaker set at 250 rpm. Following incubation, 10 mL of culture was transferred to a 2L baffled flask containing 600 mL of sterile production medium containing 20 g/L potato dextrin type IV (sigma), 30 g/L glycerol, 2.5 g/L Bacto-peptone, 8.34 g/L yeast extract, 3 g/L CaCO 3 , pH 7.0. Fermentation broth was prepared by incubating the production culture at 28 0 C in a rotary shaker set at 250 rpm for 5 days. b) Alternate procedure:
  • the fermentation was accomplished as a 1 x 10L batch in a 14.5 L fermentor (BioFlo 110TM Fermentor, New Brunswick Scientific, Edison, NJ, USA) using an improved procedure described in CA patent application 2,466,340, filed January 21 , 2004.
  • Micromonospora sp. (deposit accession number IDAC 070303-01) was maintained on agar plates of ISP2 agar (Difco Laboratories, Detroit, Ml). An inoculum for the production phase was prepared by transferring the surface growth of the Micromonospora sp. from the agar plates to 2-L flasks containing 500 mL of sterile medium comprised of 10 g glucose, 20 g potato dextrin, 5 g yeast extract, 5 g NZ-
  • Amine A, and 1 g CaCO 3 made up to one liter with tap water (pH 7.0).
  • the culture was incubated at about 28 0 C for approximately 70 hours on a rotary shaker set at 250 rpm.
  • 300 ml_ of culture was transferred to a 14.5 L fermentor containing 10 L of sterile production medium.
  • Each liter of production medium was composed of 20 g potato dextrin, 30 g glycerol, 2.5 g Bacto-peptone, 8.34 g yeast extract, 0.3 ml_ Silicone defoamer oil (Chem Service), 0.05 ml Proflo oilTM (Traders protein) and 3 g CaCO 3 made to one liter with distilled water and adjusted to pH 7.0.
  • the culture was incubated at 28 0 C, with dissolved oxygen (dO 2 ) controlled at 25% in a cascade loop with agitation varied between 320-600 RPM and aeration set at a fixed rate of 0.5 v/v/m.
  • dO 2 dissolved oxygen
  • Compound 1 by fermentation are provided in Table 1 (QB, MA, KH, RM, JA, FA, CL). Any one of Micromonospora sp. 046-ECO11 or [S01]046 may be used in these exemplified methods.
  • Fractions containing Compound 1 were evaporated to dryness and 100 mg was digested in the 5 mL of the upper phase of a mixture prepared from chloroform, cyclohexane, methanol, and water in the ratios, by volume, of 5:2:10:5.
  • the sample was subjected to centrifugal partition chromatography using a High Speed Countercurrent Chromatography (HSCC) system (Kromaton Technologies, Angers, France) fitted with a 200 mL cartridge and prepacked with the upper phase of this two- phase system.
  • the HSCC was run with the lower phase mobile and Compound 1 was eluted at approximately one-half column volume.
  • Compound 1 was also isolated using the following alternative protocol. At the end of the incubation period, the fermentation broth from the baffled flasks of Example 1 was centrifuged and the supernatant decanted from the pellet containing the bacterial mycelia. 100 mL of 100% MeOH was added to the mycelial pellet and the sample was stirred for 10 minutes and centrifuged for 15 minutes. The methanolic supernatant was decanted and saved. 100 mL of acetone was then added to the mycelial pellet and stirred for 10 minutes then centrifuged for 15 minutes. The acetonic supernatant was decanted and combined with the methanolic supernatant. Finally, 100 mL of 20% MeOH/H 2 O was added to the mycelial pellet, stirred for 10 minutes and centrifuged for 15 minutes. The supernatant was combined with the acetonic and methanolic supernatants.
  • HP-20 resin was washed successively with 2 ⁇ 500mL of 50% MeOH/H 2 O, 2 ⁇ 500mL of 75% MeOH/H 2 O and 2 ⁇ 500mL of MeOH.
  • the column was eluted with acetonitrile/buffer (5 mM of NH 4 HCO 3 ) according to gradient shown in Table 3.
  • the two methanol water layers were combined and treated with 200 ml_ of EtOAc and 400 ml_ of water. The layers were separated and the aqueous layer extracted twice further with 200 mL portions of EtOAc. The EtOAc layers are combined and concentrated. The residue obtained (220 mg) was suitable for final purification, either by HSCC or by HPLC as described above. This extraction process achieved a ten-fold purification when compared with the extraction protocol used in (a) or (b).
  • NMR data were collected dissolved in MeOH-c/4 including proton, carbon and multidimensional pulse sequences gDQCOSY, gHSQC, gHMBC, and NOESY.
  • a number of cross peaks in the 2D spectra of Compound 1 are key in the structural determination.
  • the famesyl chain is placed on the amide nitrogen by a strong cross peak between the proton signal of the terminal methylene of that chain at 4.52 ppm and the amide carbonyl carbon at 170 ppm in the gHMBC experiment.
  • N/A not applicable, group not present in the molecule a.
  • CH in Compound 2 CH 2 in Compound 18 b.
  • Signals for 4', 5', 8' and 9' are very close; assignment was based on Compound 1 b) Synthesis and structural elucidation of Compounds 3, 21, and 22
  • the fraction containing Compound 3 was further purified by HPLC using the same column (20 mL/min, H 2 O/CH 3 CN gradient 80:20-30:70, 0-8 min; 30:70- 0:100, 8-18 min, curve 7), to give substantially pure Compound 3 (13.85 mg, RT: 17.9 min).
  • N/A not applicable, group not present in the molecule ** signal 1.22-1.49 ppm, 10 protons a. CH in Compounds 3 and 21 , CH 2 in Compound 22 b. CH in Compound 3, CH 2 in Compounds 21 and 22
  • N/A not applicable, group not present in the molecule a. CH 2 in Compound 4, CD 3 in Compound 13.
  • Compounds 2 to 4, 6, and 8 to 12 are produced by the procedure described in a) and b), by substituting the alkylating agent used in these procedures respectively by the following alkylating agents: iodomethane, bromoethane, 1-bromopropane, 1- bromopentane, 1-bromoheptane, 1-chlorooctane, trifuoromethyl iodide, heptafluoro-1- iodopropane, and 2-iodo-1 ,1 ,1 -trifluoroethane.
  • alkylating agent used in these procedures respectively by the following alkylating agents: iodomethane, bromoethane, 1-bromopropane, 1- bromopentane, 1-bromoheptane, 1-chlorooctane, trifuoromethyl iodide, heptafluoro-1- iodopropane, and 2-
  • N/A not applicable, group not present in the molecule
  • N/A Not applicable, group not present in the molecule. a. Signals of 4', 5', 8' and 9' are very close; assignment was based on Compound 1.
  • the famesyl olefinic protons of positions 6'-7' and 10'-11 ' on the NMR spectra were replaced by aliphatic proton signals and, together with the aliphatic protons of positions 5', 8' and 9', are observed in the region of about 1.07 to 1.51 ppm, integrating for 12 protons, 2CH, 5CH 2 .
  • the famesyl olefinic proton at position 2'(CH) was shown on the NMR to have a chemical shift of 5.41 ppm.
  • the methylene group at positions 4' was shown at 2.03 ppm.
  • the characteristic 5- ⁇ /-methyl group was also easily assigned to a chemical shift of 2.93 ppm. .
  • aliphatic proton signals at positions 2'-1 V all have very close chemical shifts ranging from about 1 to 1.75 ppm (integrating for 17 protons), methyl protons at positions 12' and 1"-3" are all very close as well (shifts 0.8-0.95 ppm, integrating for 12 protons). These signals are also complex from the fact that 2 diastereomers of positions 3' and T are present in the mixture, and in different proportions. Labile protons were not observed since NMR was done in deuterated methanol.
  • N/A not applicable, group not present in the molecule
  • EXAMPLE 8 AROMATIC SUBSTITUTION REACTION a) Synthesis and structural elucidation of Compound 24 by bromination
  • the semi-purified sample was further purified by HPLC (SymmetryTM C-18 25 x 100 mm column: 20 mL/min, H 2 O/CH 3 CN gradient 70:30-30:70, 0-15 min), to give pure Compound 24 (9.5 mg, RT 13.0 min).
  • In vitro cytotoxic activities (Gl 50 ) of Compounds 1 , 2 to 5, 7, 13, 14, 15, 18, 21 , 22, and 24 were determined using propidium iodide (Pl) as being the concentration of drug resulting in 50% growth inhibition, and by using the following method.
  • Pl propidium iodide
  • Two 96-well plates were seeded in duplicate with each cell line at the appropriate inoculation density (HT29: 3,000; SF268: 3,000; PC-3: 3,000; and MDA-MB-231 : 7,500 cells) and according to the technical data sheet of each cell line (rows A-G, 75 ⁇ l_ of media per well). Row H was filled wih medium only (150 ⁇ l_, negative control-medium).
  • the plates were incubated at appropriate temperature and CO 2 concentration for 24 hrs.
  • Test Compounds were prepared as 15X stock solutions in appropriate medium and corresponding to 450, 45, 0.45, 0.045, and 0.0045 ⁇ M (prepared the day of the experiment). An aliquot of each was diluted 7.5-fold in appropriate test medium to give a set of six 2X concentration solutions (60, 6, 0.6, 0.06, 0.006, and 0.0006 ⁇ M). A 75 ⁇ l_ aliquot of each concentration was added to each corresponding well (rows A to F) of the second plate. Row G was filled with 75 ⁇ l_ of medium/0.6% DMSO (negative control-cells). The second plate was incubated at appropriate temperature and CO2 concentration for 96 hrs.
  • Second plate was processed as the first one, except there were three rounds of freeze/thaw instead of two.
  • First measurement gave the treated dead cells value (TDC), and the second measurement gave the treated total cells value (TTC). Both values were collected for each treated well and control (CTC and CDC).
  • the Gl 50 value emphasizes the correction for the cell count at time zero for cell survival.
  • the T/C values are transposed in a graph to determine GI 50 values, the concentration at with the T/C is 50%.
  • the aim of this study was to test whether Compounds 1 and 2 prevent or delay tumor growth in C6 glioblastoma cell-bearing mice, and to determine an effective dosage regimen.
  • mice A total of 60 six-week-old female mice (Mus musculus nude mice), ranging between 18 to 25 g in weight, were observed for 7 days before treatment. Animal experiments were performed according to ethical guidelines of animal experimentation (Charte du comite d'ethique du CNRSj ' uillet 2003) and the English guidelines for the welfare of animals in experimental neoplasia (WORKMAN, P., TWENTYMAN, P., BALKWILL, F., et al. (1998). United Kingdom Coordinating Committee on Cancer Research (UKCCCR) Guidelines for the welfare of animals in experimental neoplasia (Second Edition, July 1997; British Journal of Cancer, 77:1-10).
  • mice were euthanized upon removal from the cage. Animals were maintained in rooms under controlled conditions of temperature (23 ⁇ 2°C), humidity (45 ⁇ 5%), photoperiodicity (12 hrs light / 12 hrs dark) and air exchange. Animals were housed in polycarbonate cages (5/single cage) that were equipped to provide food and water. Animal bedding consisted of sterile wood shavings that were replaced every other day. Food was provided ad libitum, being placed in the metal lid on the top of the cage. Autoclaved tap water was provided ad libitum. Water bottles were equipped with rubber stoppers and sipper tubes. Water bottles were cleaned, sterilized and replaced once a week. Two different numbers engraved on two earrings identified the animals. Each cage was labeled with a specific code.
  • Tumor Cell Line The C6 cell line was cloned from a rat glial tumor induced by N- nitrosomethyurea (NMU) according to Premont et al. (Premont J, Benda P, Jard S., [3H] norepinephrine binding by rat glial cells in culture. Lack of correlation between binding and adenylate cyclase activation. Biochim Biophys Acta. 1975 Feb 13;381(2):368-76.) after series of alternate culture and animal passages. Cells were grown as adherent monolayers at 37 0 C in a humidified atmosphere (5% CO 2 , 95% air).
  • NMU N- nitrosomethyurea
  • the culture medium was DMEM supplemented with 2 mM L-glutamine and 10% fetal bovine serum.
  • tumor cells were detached from the culture flask by a 10 min treatment with trypsin-versen. The cells were counted in a hemocytometer and their viability assessed by 0.25% trypan blue exclusion.
  • test article For the test article, the following procedure was followed for reconstitution (performed immediately preceding injection).
  • the vehicle consisted of a mixture of benzyl alcohol (1.5%), ethanol (8.5%), propylene glycol (27%), PEG 400 (27%), dimethylacetamide (6%) and water (30%).
  • the vehicle solution was first vortexed in order to obtain a homogeneous liquid.
  • 0.6 ml_ of the vortexed vehicle solution was added to each vial containing the test article (Compound 1 ). Vials were mixed thoroughly by vortexing for 1 minute and inverted and shaken vigorously. Vials were mixed again prior to injection into each animal.
  • mice Experiment started at day 0 (D 0 ). On Do, mice . received a superficial intramuscular injection of C6 tumor cells (5 x 10 5 cells) in 0.1 mL of DMEM complete medium into the upper right posterior leg.
  • treatment started 24 hrs following inoculation of C6 cells. On the day of the treatment, each mouse was slowly injected with 100 ⁇ l_ of test or control articles by the i.p. route. For all groups, treatment was performed until the tumor volume of the saline-treated mice (group 1 ) reached approximately 3 cm 3 (around day 16). Mice of group 1 were treated daily with a saline isosmotic solution for 16 days. Mice of group 2 were treated daily with the vehicle solution for 16 days. Mice of group 3 were treated daily with 10 mg/kg of Compound 1 for 16 days. Mice of group 4 were treated every two days with 30 mg/kg of Compound 1 and received 8 treatments.
  • mice of group 5 were treated every three days with 30 mg/kg of Compound 1 and received 6 treatments. Measurement of tumor volume started as soon as tumors became palpable (>100 mm 3 ; day 11 post-inoculation) and was evaluated every second day until the end of the treatment using callipers. As shown in Table 14 and Figure 1 , the mean value of the tumor volume of all Compound 1 treated groups (6 mice/group) was significantly reduced as demonstrated by the one-way analysis of variance (Anova) test followed by the non-parametric Dunnett's multiple comparison test comparing treated groups to the saline group. An asterisk in the P value column of Table 14 indicates a statistically significant value, while "ns" signifies not significant.
  • Tumor volume was measured every second day until the end of the treatment using callipers. As shown in Table 15 and Figure 2, the mean value of the tumor volume of all Compound 1 treated groups (6 mice/group) was significantly reduced as demonstrated by the one-way analysis of variance (Anova) test followed by the non-parametric Dunnett's multiple comparison test comparing treated groups to the saline group. An asterisk in the P value column of Table 15 indicates a statistically significant value, while "ns" signifies not significant.
  • Compounds 1 and 2 were separately dissolved in ethanol (5%), Polysorbate 80 (15%), PEG 400 (5%) and dextrose (5%) at a final concentration of 6 mg/ml (for all parenteral administration routes).
  • Compound 1 was solubilized in Cremophor® EL/ Ethanol (50%:50%) at a final concentration of 6 mg/ml.
  • animals female CrI: CD1 mice; 6 weeks of age, 22-24g
  • Compound 1 was administered by the intravenous (iv), subcutaneous (sc), intraperitoneal (ip), or oral (po) route to the assigned animals.
  • Compound 2 was administered by the intravenous (iv), or intraperitoneal (ip) route to the assigned animals.
  • the dosing volume of Compounds 1 and 2 was 5 mL per kg body weight. Animals were anesthetized with 5% isoflurane prior to bleeding. Blood was collected into microtainer tubes containing the anticoagulant K 2 EDTA by cardiac puncture from each of 4 animals per bleeding timepoint (2 min, 5 min, 15 min, 30 min, 1 h, 2h, 4h and 8h). Following collection, the samples were centrifuged and the plasma obtained from each sample was recovered and stored frozen (at approximately -8O 0 C) pending analysis.
  • Plasma values of Compounds 1 and 2 falling below the limit of quantitation (LOQ) were set to zero.
  • the following pharmacokinetic parameters were calculated: area under the plasma concentration versus time curve from time zero to the last measurable concentration " time point (AUC 0-t ), area under the plasma concentration versus time curve extrapolated to infinity (AUQn f ), maximum observed plasma concentration (C ma ⁇ ), time of maximum plasma concentration (t ma ⁇ ), apparent first-order terminal elimination rate constant (k e ⁇ ), apparent first-order terminal elimination half-life will be calculated as 0.693/kel (ti /2 ).
  • the systemic clearance (CL) of Compound 1 after intravenous administration was calculated using Dose/AUCinf.
  • Pharmacokinetic parameters were calculated using KineticaTM 4.1.1 (InnaPhase Corporation, Philadelphia, PA).
  • Mean plasma concentrations of Compound 1 following intravenous (iv), intraperitoneal (ip), subcutaneous (sc), and oral (po) administrations at 30 mg/kg are presented in Figure 5.
  • Mean plasma concentrations of Compound 2 following iv and ip administrations at 30 mg/kg, compared with Compound 1 via the same routes of administration, are presented in Figure 6.
  • Compound 2 When administered iv, Compound 2 had an AUC of 92.08 ⁇ M » h and an observed Cm ax of 105 ⁇ g/mL, compared to an AUC of 40.4 ⁇ M » h and an observed C max of 130 ⁇ g/mL for Compound 1.
  • Compound 2 When administered ip, Compound 2 had an AUC of 58.75 ⁇ M*h and an observed C max of 5.8 ⁇ g/mL, compared to an AUC of 9.5 ⁇ M*h and an observed C max of 2.25 ⁇ g/mL for Compound 1.
  • Acute toxicity studies in CD-1 nu/nu mice for Compound 2 using the same formulation, gave an MTD > 50 mg/kg (ip, NOAEL: 30 mg/kg) and ⁇ 100 mg/kg (iv, NOAEL: 75 mg/kg), with weight losses of about 7% for several days post-injection.
  • Compound 1 had an MTD of 150 mg/kg when administered iv.
  • Acute toxicity studies with Compound 46 gave an MTD of 30 mg/kg (ip).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
PCT/CA2005/001467 2004-09-27 2005-09-26 Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals WO2006034574A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US11/663,890 US20090029972A1 (en) 2004-09-27 2005-09-26 Dibenzodiazepinone Analogues, Processes for Their Production and Their Use as Pharmaceuticals
AU2005289317A AU2005289317A1 (en) 2004-09-27 2005-09-26 Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals
EP05791575A EP1809612A4 (de) 2004-09-27 2005-09-26 Dibenzodiazepinonanaloga, verfahren zu deren herstellung und deren verwendung als pharmazeutika
JP2007532739A JP2008514551A (ja) 2004-09-27 2005-09-26 ジベンゾジアゼピノンアナログ、その製造方法及び医薬品としての使用
CA002581658A CA2581658A1 (en) 2004-09-27 2005-09-26 Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals
IL182214A IL182214A0 (en) 2004-09-27 2007-03-26 Dibenzodiaepinone analogues, processes for their production and their use as pharmaceuticals

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US10/951,436 2004-09-27
US10/951,436 US7186713B2 (en) 2003-01-21 2004-09-27 Farnesyl dibenzodiazepinones and methods of treating cancer using same
US62565304P 2004-11-08 2004-11-08
US60/625,653 2004-11-08
US64738105P 2005-01-28 2005-01-28
US60/647,381 2005-01-28
CA2,497,031 2005-02-11
CA 2497031 CA2497031A1 (en) 2004-11-08 2005-02-11 Farnesyl dibenzodiazepinones, processes for their production and their use as pharmaceuticals
US70147205P 2005-07-22 2005-07-22
US60/701,472 2005-07-22

Publications (1)

Publication Number Publication Date
WO2006034574A1 true WO2006034574A1 (en) 2006-04-06

Family

ID=36118533

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2005/001467 WO2006034574A1 (en) 2004-09-27 2005-09-26 Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals

Country Status (4)

Country Link
EP (1) EP1809612A4 (de)
JP (1) JP2008514551A (de)
AU (1) AU2005289317A1 (de)
WO (1) WO2006034574A1 (de)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006128288A1 (en) * 2005-06-02 2006-12-07 Thallion Pharmaceuticals Inc. Formulation comprising farnesyl dibenzodiazepinone and a pharmaceutically acceptable surfactant
JP2009236734A (ja) * 2008-03-27 2009-10-15 Kinjo Gakuin 高速向流クロマトグラフィーによるターメリックパウダー(ウコン粉末)からのクルクミン類の分離精製法
WO2010019775A2 (en) * 2008-08-15 2010-02-18 Thallion Pharmaceuticals Inc. Methods of treating ras driven cancer in a subject

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5541181A (en) * 1994-05-26 1996-07-30 Bristol-Myers Squibb Company Compound produced by a strain of micromonospora
CA2466340A1 (en) * 2003-01-21 2004-08-09 Ecopia Biosciences Inc. Farnesyl dibenzodiazepinone, processes for its production and its use as a pharmaceutical

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2007015171A (es) * 2005-06-02 2008-04-22 Thallion Pharmaceuticals Inc Formulacion que comprende farnesil dibenzodiazepinona y un tensoactivo farmaceuticamente aceptable.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5541181A (en) * 1994-05-26 1996-07-30 Bristol-Myers Squibb Company Compound produced by a strain of micromonospora
CA2466340A1 (en) * 2003-01-21 2004-08-09 Ecopia Biosciences Inc. Farnesyl dibenzodiazepinone, processes for its production and its use as a pharmaceutical

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHARAN R.D. ET AL, J. NAT. PROD., vol. 67, 2004, pages 1431 - 1433, XP008117531 *
See also references of EP1809612A4 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006128288A1 (en) * 2005-06-02 2006-12-07 Thallion Pharmaceuticals Inc. Formulation comprising farnesyl dibenzodiazepinone and a pharmaceutically acceptable surfactant
JP2009236734A (ja) * 2008-03-27 2009-10-15 Kinjo Gakuin 高速向流クロマトグラフィーによるターメリックパウダー(ウコン粉末)からのクルクミン類の分離精製法
WO2010019775A2 (en) * 2008-08-15 2010-02-18 Thallion Pharmaceuticals Inc. Methods of treating ras driven cancer in a subject
WO2010019775A3 (en) * 2008-08-15 2010-05-27 Thallion Pharmaceuticals Inc. Methods of treating ras driven cancer in a subject

Also Published As

Publication number Publication date
AU2005289317A1 (en) 2006-04-06
JP2008514551A (ja) 2008-05-08
EP1809612A4 (de) 2009-05-27
EP1809612A1 (de) 2007-07-25

Similar Documents

Publication Publication Date Title
US7304054B2 (en) Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals
CA2507567A1 (en) Use of compositions comprising farnesyl dibenzodiazepinones for treating neoplastic cells and conditions
US20090170837A1 (en) Methods for treating ras driven cancer in a subject
US20040138196A1 (en) Salinosporamides and methods for use thereof
US7358241B2 (en) Compositions and methods comprising farnesyl dibenzodiazepinones for treating neoplastic cells and conditions
US8076287B2 (en) Cyclic hexadepsipeptides, processes for their production and their use as pharmaceuticals
EP1809612A1 (de) Dibenzodiazepinonanaloga, verfahren zu deren herstellung und deren verwendung als pharmazeutika
US7655646B2 (en) Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals
EP1285928B1 (de) Antibiotische tripropeptine und verfahren ihrer herstellung
CA2511750C (en) Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals
US20090062255A1 (en) Tumor-targeting evaluation methodology and compounds related thereto
US20090029972A1 (en) Dibenzodiazepinone Analogues, Processes for Their Production and Their Use as Pharmaceuticals
CA2581658A1 (en) Dibenzodiazepinone analogues, processes for their production and their use as pharmaceuticals
US7384935B2 (en) Phosphate prodrugs of a farnesyl dibenzodiazepinone, processes for their production and their use as pharmaceuticals
WO2002072617A1 (fr) Composes thiopeptides
US4772595A (en) Synthetic virginiamycin M1 analogs
US4762923A (en) Fermentation analogs of virginiamycin M1
US4859690A (en) Therapeutic virginiamycin M1 analogs
US20060106028A1 (en) Polycyclic aromatics and derivatives thereof and processes for their preparation
US20090186878A1 (en) Crystalline forms of a farnesyl dibenzodiazepinone
US5189050A (en) Fermentation analogs of virginiamycin m1 to treat panic and anxiety disorder
US5006466A (en) Fermentation analogs of virginiamycin M1
DE102004010219B4 (de) HKI10311129, neues Antibiotikum, Verfahren zu dessen Herstellung und Arzneimittel enthaltend HKI10311129
CA2497031A1 (en) Farnesyl dibenzodiazepinones, processes for their production and their use as pharmaceuticals
JPH11124368A (ja) 生理活性物質、その製造法及び剤

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2581658

Country of ref document: CA

Ref document number: 182214

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2007532739

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005289317

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2867/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2005791575

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200580040322.8

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005791575

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11663890

Country of ref document: US