WO2003051877A1 - 2-substituted pyrrolo[2.1-a]isoquinolines against cancer - Google Patents

2-substituted pyrrolo[2.1-a]isoquinolines against cancer Download PDF

Info

Publication number
WO2003051877A1
WO2003051877A1 PCT/US2002/040328 US0240328W WO03051877A1 WO 2003051877 A1 WO2003051877 A1 WO 2003051877A1 US 0240328 W US0240328 W US 0240328W WO 03051877 A1 WO03051877 A1 WO 03051877A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally
alkyl
group
alkoxy
substituents selected
Prior art date
Application number
PCT/US2002/040328
Other languages
French (fr)
Inventor
Chengzhi Zhang
Dongping Fan
Yamin Wang
Tindy Li
Stephen J. Boyer
Jennifer Burke
Brian C. Raudenbush
Wai C. Wong
Shihong Ying
Ming Wang
Qian Zhao
Christopher A. Carter
Nils Burkhardt
Josef Pernerstorfer
Maria Niewoehner
Elizabeth CARLSON-SULLIVAN
Ulrich Niewoehner
Original Assignee
Bayer Corporation
Bayer Aktiengesellschaft
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bayer Corporation, Bayer Aktiengesellschaft filed Critical Bayer Corporation
Priority to AU2002366362A priority Critical patent/AU2002366362A1/en
Publication of WO2003051877A1 publication Critical patent/WO2003051877A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to 2-substituted pyrrolo[2.1-a]isoquinoline derivatives which are inhibitors of phosphodiesterase 10a, a process for preparing these compounds and a method of treating cancer by administering these compounds.
  • Cyclic AMP metabolism is regulated by the opposing activities of adenylyl cyclase, which generates cAMP in response to extracellular stimuli (e.g. engagement of G-protein coupled receptors by their cognate ligands), and 3 ',5 '-cyclic nucleotide phosphodiesterases (PDEs), which hydrolyze cAMP to 5 '-AMP.
  • PDEs 3 ',5 '-cyclic nucleotide phosphodiesterases
  • Signal transduction via cAMP is associated with transcriptional events that can result in the inhibition of cellular proliferation (T. J. Shaw et al, Exp. Cell Res. 273, 95 (2002); T.W. Moody et al., Ann. N.Y Acad. Sci. 921, 26 (2000); W.L.
  • the ICAST (Inhibitor of Cyclic AMP Signal Transduction) gene encodes a specific 3',5'- cyclic nucleotide phosphodiesterase. Compared to corresponding normal tissues, ICAST mRNA is overexpressed in breast carcinoma specimens, liver metastases of colorectal carcinoma and non-small cell lung carcinomas.
  • the ICAST cDNA was also recently cloned by other groups and named PDE 10a (K. Fujishige et al., J. Biol. Chem. 274, 18 438 (1999); S.H. Soderling et al., Proc. Natl. Acad. Sci. USA 96, 7071 (1999); K.
  • R"", R"' Me, Et, i-Pr, C ⁇ H ⁇
  • the compounds (B) are described as having anti-tumor activity due to their ability to intercalate into DNA. It is not mentioned that they have any PDE 10a inhibitory activity.
  • R H, OCH 3
  • R" CH 3
  • R'" C 6 H 5
  • R'" OR , CH 3 , NH 2
  • R 1 and R 2 independently from each other denote hydrogen, C ⁇ - -alkyl or trifluoromethyl or
  • R 1 and R 2 together form a C ⁇ - -alkylene bridge
  • R 3 and R 4 independently from each other denote C ⁇ - 6 -alkyl optionally further substituted with halogen up to perhalo;
  • R 5 denotes a radical of the formula
  • R 6 denotes Ci- 6 -alkyl, trifluoromethyl, trifluoromethoxy, halogen, hydrogen, hydroxy or C ⁇ - 6 -alkoxy; X and Y independently from each other denote
  • C 6 -C 10 -aryl optionally having from 1 to 3 substituents selected from the group consisting of C ⁇ - 6 -alkoxy, C ⁇ - 6 -alkyl, C 3 - 8 -cycloalkyl, trifluoromethyl, trifluoromethoxy and halogen, or
  • heteroaryl or heterocyclyl may optionally have from 1 to 3 substituents selected from the group consisting of i) C 3 . 8 -cycloalkyl; ii) C ⁇ - 6 -alkyl, optionally having from 1 to 3 substituents selected from the group consisting of C 3 - 8 -cycloalkyl, hydroxy, C ⁇ _ 6 -alkoxy, halogen, phenylamino, morpholinyl, (C ⁇ - 6 -alkyl)-aminocarbonyl, benzo[2,3]dioxolyl and C 6 -C 10 -aryl, wherein said aryl is optionally substituted with C ⁇ _ 6 -alkyl or C ⁇ - 6 -alkoxy; iii) C 6 -C ⁇ o-aryl, optionally having from 1 to 3 substituents selected from the group consisting of
  • X and Y together with the nitrogen atom to which they are attached form heterocyclyl fused to C 6 -C 10 -aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Ci- 6 - alkyl, C 3 . 8 -cycloalkyl, C ⁇ . 6 -alkoxy and hydroxymethyl;
  • Y denotes -NR 9 R 10 ; wherein R 9 and R 10 independently from each other denote 1) hydrogen,
  • C 6 -C ⁇ o-aryl optionally having from 1 to 3 substituents selected from the group consisting of halogen, C ⁇ - 6 -alkyl, C 3 . 8 -cycloalkyl, C ⁇ - 6 -alkoxy, trifluoromethoxy and trifluoromethyl, 3) heterocyclyl,
  • R 9 and R 10 together with the nitrogen atom to which they are attached form heterocyclyl or heteroaryl, wherein said heteroaryl or heterocyclyl may optionally have from 1 to 3 substituents selected from the group consisting of Ci- 6 -alkyl, C ⁇ - 6 -alkoxy, halogen and methoxymethyl;
  • R and R independently from each other denote hydrogen, C ⁇ - 4 -alkyl or trifluoromethyl;
  • R 3 and R 4 independently from each other denote Ci- 6 -alkyl optionally further substituted with halogen up to perhalo;
  • R 5 denotes a radical of the formula
  • R 6 denotes halogen, hydrogen, hydroxy or Ci- 6 -alkoxy
  • X and Y together with the nitrogen atom to which they are attached form heterocyclyl fused to C 6 -C ⁇ o-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, C ⁇ - 6 - alkyl, C ⁇ - 6 -alkoxy and hydroxymethyl; or
  • X denotes hydrogen and Y denotes -NR 9 R 10 ; wherein R 9 and R 10 independently from each other denote
  • R 9 and R 10 together with the nitrogen atom to which they are attached form heterocyclyl or heteroaryl, wherein said heterocyclyl or heteroaryl optionally have from 1 to 3 substituents selected from the group consisting of C ⁇ _ 6 -alkyl and methoxymethyl;
  • a further alternative embodiment of the present invention relates to a compound of the formula (I), wherein
  • R 1 and R 2 independently from each other denote hydrogen or C ⁇ _ 4 -alkyl;
  • R j denotes C ⁇ - 6 -alkyl or trifluoromethyl;
  • R 4 denotes C ⁇ - -alkyl
  • R 5 denotes a radical of the formula selected from the group consisting of:
  • X and Y together with the nitrogen atom to which they are attached form i) morpholino, optionally further subsituted with 1 to 2 substituents Ci- 6 -alkyl; ii) piperidinyl, optionally having from 1 to 2 substituents selected from the group consisting of hydroxyl, hydroxymethyl and C ⁇ _ 6 -alkyl; iii) pyrrolidinyl, optionally having from 1 to 2 substituents selected from the group consisting of C ⁇ - 6 -dialkylamino, pyridyl, carboxamido, Ci- 6 -alkoxy, phenylaminomethyl, methoxymethyl and methoxyphenyl; or iv) piperazinyl, optionally having from 1 to 2 substituents selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexylmethyl, benzyl, morpholinoe
  • X and Y together with the nitrogen atom to which they are attached fomi dimethoxytetrahydroisoquinolinyl, 2-methyl-6- fluorotetrahydroquinolinyl, indolinyl, isoindolinyl or
  • X denotes hydrogen and Y denotes a) phenylamino, having from 1 to 2 substituents selected from the group consisting of fluoro, chloro, bromo, iodo, trifluoromethyl and Ci-e-alkyl; b) piperidinyl, optionally further substituted with 1 to 2 - ⁇ - alkyl; c) triazolyl; d) pyrrolidinyl, optionally further substituted with 1 to 2 methoxymethyl; e) morpholino; f) imidazoyl; g) C 3 _ 8 -cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl; h) Ci- 6 -dialkylamino; or i) azepanyl;
  • a further alternative embodiment of the present invention relates to a compound of the formula (T): wherein R >3 to R are defined as described above.
  • a further alternative embodiment of the present invention relates to a compound of the formula (I), wherein
  • R 3 denotes a radical of the formula selected from the group consisting of:
  • a further alternative embodiment of the present invention relates to a compound of the formula (I), wherein
  • x and y independently from each other denote zero or 1 and x+y is 1 or 2;
  • R and R independently from each other denote hydrogen, C ⁇ - -alkyl or CF or R 1 and R 2 together form a C ⁇ - 4 -alkylene bridge;
  • R 3 and R 4 independently from each other denote C ⁇ - -alkyl
  • R s denotes a monovalent radical of the formula
  • the compounds of the formula (I) according to the invention can exist in stereoisomeric forms which are either like image and mirror image (enantiomers) or are not like image and mirror image (diastereomers).
  • the invention relates both to the enantiomers or diastereomers and to their respective mixtures.
  • the racemic forms, like the diastereomers, can be separated in a known manner into the stereoisomerically uniform components.
  • Pharmaceutically acceptable salts according to the invention are non-toxic salts which in general are accessible by reaction of the compounds (I) with an inorganic or organic base or acid conventionally used for this purpose.
  • Non-limiting examples of pharmaceutically acceptable salts of compounds (I) include the alkali metal salts, e.g.
  • the alkaline earth metal salts such as the magnesium and calcium salts
  • the quaternary ammonium salts such as, for example, the triethyl ammonium salt, acetates, benzene sulphonates, benzoates, dicarbonates, disulphates, ditartrates, borates, bromides, carbonates, chlorides, citrates, dihydrochlorides, fumarates, gluconates, glutamates, hexyl resorcinates, hydrobromides, hydrochlorides, hydroxynaphthoates, iodides, isothionates, lactates, laurates, malates, maleates, mandelates, mesylates, methylbromides, methylnitrates, methylsulphates, nitrates, oleates, oxalates, palmitates, pantothenates, phosphates, diphosphates, polygalacturonates
  • hydrates are forms of the compounds of the formula (I) above which, in the solid or liquid state, form a molecular compound (solvate) by hydration with water.
  • examples of hydrates are sesquihydrates, monohydrates, dihydrates and trihydrates. Equally suitable are the hydrates of salts of the compounds according to the invention.
  • Halogen represents fluorine, chlorine, bromine and iodine. Preference is given to chlorine and fluorine.
  • alkylcarbonyl represents a straight-chain or branched alkyl radical preferrably having from 1 to 6 carbon atoms. Examples which may be mentioned are: methyl, ethyl, n-propyl, isopropyl, n-butyl, ⁇ o-butyl, tert-butyl, n-pentyl, n-hexyl, n-octyl and «-decyl.
  • alkyl groups having fewer carbon atoms such as, for example, C ⁇ -C -alkyl, are derived analogously from this definition.
  • C j -Cs-Cvcloalkyl represents a mono- or bicyclic alkyl radical having 3 to 8 carbon atoms. Examples which may be mentioned are: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl.
  • the corresponding cycloalkyl groups having fewer carbon atoms, such as, for example, C 3 -C 6 -cycloalkyl, are derived analogously from this definition.
  • Non-limiting examples of radicals include methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxycarbonyl and isobutoxycarbonyl.
  • the corresponding alkoxycarbonyl groups having fewer carbon atoms such as, for example, C 3 -C 6 -alkoxycarbonyl, are derived analogously from this definition.
  • C j ⁇ -Cg-Alkoxy represents a straight-chain or branched alkoxy radical having 1 to 6 carbon atoms. Examples which may be mentioned are: methoxy, ethoxy, «-propoxy, iso-pxopoxy, n-bvAoxy, iso-butoxy, tert-butoxy, w-pentoxy and n-hexoxy.
  • the corresponding alkoxy groups having fewer carbon atoms, such as, for example, C ⁇ -C 4 -alkoxy, are derived analogously from this definition.
  • Ci-C ⁇ -Dialkylamino represents an alkylamino radical having two (independently selected) alkyl substituents, illustratively and preferably representing NN-dimethylamino, N,N- diethylamino, N-ethyl-N-methylamino, N-meti yl-N-n-propylamino, N-isopropyl-N-n- propylamino, N-t-butyl-N-methylamino, N-ethyl-N-n-pentylamino and N-n-hexyl-N- methylamino.
  • Cfi-C ⁇ -Aryl represents an aromatic radical preferably having 6 to 14, more preferably 6 to 10 carbon atoms.
  • Cg-Cui-aryl radicals include phenyl, and naphthyl.
  • Heteroaryl in the context of the invention represents a preferably 5- to 13-membered heteroaryl or a 5- to 13-membered aromatic heterocycle having from 1, up to 4, heteroatoms from the group consisting of ⁇ , O and S, which ring or ring system can be linked via a carbon atom or a nitrogen atom, if such an atom is present.
  • pyridyl examples which may be mentioned are: pyridyl, pyridyl N-oxide, pyrimidyl, pyridazinyl, pyrazinyl, thienyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl or isoxazolyl, indolicenyl, indolyl, benzo[b]thienyl, benzo[b] furyl, benzothiadiazolyl, indazolyl, quinolyl, isoquinolyl, naphthyridinyl, quinazolinyl.
  • Preferred examples include pyrazolyl, pyrazinyl, pyrrolyl, imidazoyl, triazolyl, indolyl, pyridyl, pyridyl-N-oxide, pyrimidyl, pyridazinyl, furyl, imidazolyl and thienyl.
  • Heterocycle in the context of the invention represents a preferably 3- to 13-membered saturated or partially unsaturated heterocycle having from 1, up to 4, heteroatoms from the group consisting of N, O and S, which ring or ring system can be linked via a carbon atom or a nitrogen atom, if such an atom is present.
  • Examples which may be mentioned are: tetrahydropyranyl, aziridyl, azepanyl, tetrahydrofuryl, pyrrolidinyl, pyrrolinyl, piperidinyl, 1,2-dihydropyridinyl, 1,4-dihydropyridinyl, piperazinyl, morpholinyl, thiomorpholinyl, azepinyl, and 1,4-diazepinyl. Preference is given to azepanyl, piperazinyl, piperidinyl, morpholinyl and pyrrolidinyl.
  • C ⁇ -Alkylene or C ⁇ -alkylene bridge represents a linear or branched, bivalent alkylene radical preferably having 1 to 4 carbon atoms, also known as alkandiyl.
  • Non-limiting examples include methylene, ethylene, propylene, ⁇ -methylethylene, ⁇ -methylethylene, ⁇ - ethylethylene, ⁇ -ethylethylene, butylene, ⁇ -methylpropylene, ⁇ -methylpropylene, and ⁇ - methylpropylene.
  • CyCin-Bridged bicycloalkyl represents a bicyclic alkyl radical having 5 to 10 carbon atoms also containing an alkylene bridge.
  • Non-limiting examples include [2.2.1]bicycloheptane, [2.2.2]bicyclooctanetane, [2.1.1]bicyclohexane,
  • a ring system represents a mono-, bi- or tricyclic system of fused rings. These rings share two ring members. These ring members are preferably adjacent ([0]-bridge).
  • Non limiting examples include naphthalene, benzo[2,3]dioxolyl, 2,3-dihydrobenzo[l,4]dioxinyl, and indole.
  • a * symbol next to a bond denotes the point of attachment in the molecule.
  • the compounds according to the invention exhibit an unforeseeable, useful pharmacological and pharmacokinetic activity spectrum. They are therefore suitable for use as medicaments for the treatment and/or prophylaxis of disorders in humans and animals.
  • the compounds of this invention may be formulated as a solution of lyophilized powders for parenteral administration.
  • Powders may be reconstituted by addition of a suitable diluent or other pharmaceutically acceptable carrier prior to use.
  • the liquid formulation is generally a buffered, isotonic, aqueous solution.
  • suitable diluents are normal isotonic saline solution, standard 5% dextrose in water or in buffered sodium or ammonium acetate solution.
  • Such formulation is especially suitable for parenteral administration, but may also be used for oral administration. It may be desirable to add excipients such as polyvinylpyrrolidone, gelatin, hydroxycellulose, acacia, polyethylene glycol, mannitol, sodium chloride, or sodium acetate.
  • the compounds of the present invention may be encapsulated, tableted, or incorporated into an emulsion (oil-in-water or water-in-oil) syrup for oral administration.
  • Pharmaceutically acceptable solids or liquid carriers which are generally known in the pharmaceutical formulary arts, may be added to enhance or stabilize the composition, or to facilitate preparation of the composition.
  • Solid carriers include starch (corn or potato), lactose, calcium sulfate dihydrate, terra alba, croscarmellose sodium, magnesium stearate or stearic acid, talc, pectin, acacia, agar, gelatin, maltodextrins and microcrystalline cellulose, or colloidal silicon dioxide.
  • Liquid carriers include syrup, peanut oil, olive oil, corn oil, sesame oil, saline, and water.
  • the carrier may also include a sustained release material such as glyceryl monostearate or glyceryl distearate, alone or with a wax.
  • the amount of solid carrier varies but, preferably, will be between about 10 mg to about 1 g per dosage unit.
  • the dosage ranges for administration of the heterocyclics disclosed in this invention are those to produce the desired affect.
  • the dosage will generally vary with age, body weight, extent of the disease, and contraindications, if any.
  • the dosage will also be dete ⁇ nined by the existence of any adverse side effects that may accompany the compounds. It is always desirable, whenever possible, to keep adverse side effects to a minimum.
  • One skilled in the art can easily determine the appropriate dosage, scheduling, and method of administration for the exact formulation of the composition being used in order to achieve the desired effective concentration in the individual patient.
  • the dosage can vary from between about 1 mg/kg/day to about 500 mg/kg/day, and preferable from between about 1 mg/kg/day to about 50 mg/kg/day.
  • the present invention also relates to a process for making the compounds.
  • the compounds of the invention may be prepared by use of known chemical reactions and procedures.
  • the compounds (I) can be synthesized according to the reaction sequence shown in Method Al above.
  • the compounds (VIII) can be synthesized by reacting amino compounds (VI) with compounds (VII), wherein L is a leaving group, for example a halogen radical such as Cl, or a radical of the formula -OC(O)R 5 , wherein R 5 is defined as optionally substituted C ⁇ -alkyl or -CH 2 C(O)OR 4 .
  • Compounds (IN) are obtained by reacting compounds (NIII) with a dehydrating agent such as, e.g., phosphorous pentoxide.
  • the compounds (LV) can also be directly converted to the compounds (f) according to the General Method Bl.
  • treatment of compounds (LV) with compounds (X) and (III) gives compounds (I).
  • the compounds (VI) are commercially available or can be synthesized according to methods commonly known to those skilled in the art (H. Mayer et al., Heterocycles 31, 1035 (1990); E. M ⁇ ller (ed.), "Methoden der Organischen Chemie” (Houben-Weyl), 4 th ed., Vol. 11/1 Stickstoff-Neritatien II, Georg Thieme Nerlag, Stuttgart 1957; Shepard et al. in J. Org. Chem. 17, 568 (1952) and in J. Am. Chem. Soc. 72, 4364 (1950)).
  • the compounds (Nil) are commercially available or can be synthesized according to methods commonly known those skilled in the art [e.g. via acylation of acetic acid with an alkyl chloroformate or dialkyl carbonate (March, Advanced Organic Chemistry, 3 rd ed., p. 440-441, Wiley 1985) and converting the resulting monoester of malonic acid into e.g. the corresponding acid chloride or anhydride by methods commonly known to those skilled in the art (see e.g. March, Advanced Organic Chemistry, 3 rd ed., p. 355, 388, Wiley 1985)].
  • Suitable solvents comprise the customary organic solvents which are inert under the reaction conditions.
  • ethers such as diethyl ether, dioxane, tetrahydrofuran, 1,2-dimethoxy ethane
  • hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions
  • halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, dichloroethane, trichloroethylene, chlorobenzene
  • ketones such as acetone
  • esters such as ethyl acetate
  • nitriles such as acetonitrile
  • heteroaromatics such as pyridine
  • polar solvents such as dimethyl formamide and hexamethyl phosphoric acid tris-amide
  • mixtures such as dimethyl formamide and hexamethyl phosphoric acid tris-amide
  • the compounds (Nil) are generally employed in an amount of from 1 to 4 mol per mol of compounds (NI); an equimolar amount or slight excess of compounds (VII) is preferred.
  • the reaction between the compounds (VI) and (VH) is preferably carried out in the presence of a base.
  • Non-limiting examples include alkali metal hydrides and alkali metal alkoxides such as, for example, sodium hydride and potassium tert-butoxide; C ⁇ -C -alkyl amines such as, for example, triethyl amine; cyclic amines such as, for example, piperidine, pyridine, dimethylamino pyridine and -preferably - l,8-diazabicyclo[4.3.0]undec-7-ene (DBU).
  • the base is generally employed in an amount of from 1 to 4 mol per mol of compounds (VI); an equimolar amount or slight excess of the base is preferred.
  • the reaction of the compounds (VI) and (Nil) can generally be carried out within a relatively wide temperature range. In general, the reaction is carried out within a range of from -20 to
  • 200°C preferably from 0 to 70°C, and more preferably at room temperature.
  • dehydrating agents such as, for example, P 2 O 5 or POCl 3 are generally employed in an amount of from 1 to 10 mol, preferably from 3 to 8 mol, per mol of compounds (VHf).
  • the cyclization reaction of the compounds (NIL-) to yield the compounds (IN) is also preferably carried out in a solvent.
  • a solvent ⁇ on-limiting examples comprise the customary organic solvents which are inert under the reaction conditions. They preferably include ethers such as diethyl ether, dioxane, tetrahydrofuran, 1,2-dimethoxy.
  • ethane hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions; halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, dichloroethane, trichloroethylene, chlorobenzene; esters such as ethyl acetate; ketones such as acetone; nitriles such as acetonitrile; heteroaromatics such as pyridine; polar solvents such as dimethyl formamide and hexamethyl phosphoric acid tris-amide; and mixtures thereof.
  • hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions
  • halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, dichloroethane,
  • Toluene is preferred, if the reaction is carried out with P 2 O 5 , and acetonitrile is preferred, if the reaction is carried out with POCl 3 (Benovsky, Stille, Tetrahedron Lett. 38, 8475-8478 (1997)).
  • the temperature for the cyclization reaction of compounds (NLII) is preferably within a range of from 60 to 200°C and more preferably within a range of from 80 to 120°C.
  • the above process steps are generally carried out under atmospheric pressure. However, it is also possible to carry them out under superatmospheric pressure or under reduced pressure (for example, in a range of from 0.5 to 5 bar).
  • the reaction time can generally be varied within a relatively wide range, i general, the reaction is finished after a period of from 2 to
  • the compounds (II) are commercially available or can be synthesized according to methods commonly known to those skilled in the art (LT. Harrison and S. Harrison, Compendium of Organic Synthetic Methods, pp. 132-176, Wiley-Interscience; E. M ⁇ ller
  • the compounds (III) are commercially available.
  • reaction of the compounds (LV) with compounds (II) and (HI), or with compounds (X) and (in), or with compounds (IX), or with compounds (XI), can be carried out as a one-pot synthesis, preferably in a solvent.
  • Suitable solvents comprise the customary organic solvents which are inert under the reaction conditions.
  • Non-limiting examples include ethers such as diethyl ether, dioxane, tetrahydrofuran, 1,2-dimethoxy ethane; hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions; halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachlori.de, dichloroethane, trichloroethylene, chlorobenzene; alcohols such as methanol, ethanol, n- propanol, isopropanol; esters such as ethyl acetate; ketones such as acetone; nitriles such as acetonitrile; heteroaromatics such as pyridine; polar solvents such as dimethyl formamide and hexamethyl phosphoric acid tris-amide; and mixtures of the above-mentioned solvents.
  • the compounds (LU) are generally employed in an amount of from 1 to 3 mol per mol of compounds (LI); an equimolar amount or slight excess of compounds (LU) is preferred.
  • the compounds (LV) are generally employed in an amount of from 0.1 to 1 mol, preferably from 0.3 to 1 mol, per mol of compounds (II).
  • the compounds (LX) or (XI) are generally employed in an amount from 1 to 3 mol per mol of compounds (IV); an equimolar amount or slight excess of compounds (III) is preferred.
  • the reactions of the compounds (IV) with compounds (II) and (IE), or with compounds (X) and (III), or with compounds (DC), or with compounds (XL), are preferably carried out in the presence of a base.
  • a base include alkali metal hydrides and alkali metal alkoxides such as, for example, sodium hydride and potassium tert-butoxide; Cwalkyl amines such as, for example, triethyl amine; cyclic amines such as, for example, pyridine, dimethylamino pyridine, l,8-diazabicyclo[4.3.0]undec-7-ene (DBU) and - preferably - piperidine.
  • the base is generally employed in an amount of from 0,1 to 1 mol, preferably from 0,3 to 1 mol, per mol of compounds (II) or compounds (V), respectively.
  • the reactions of the compounds (LV) with compounds (II) and (III), or with compounds (X) and (LQ), or with compounds (IX), or with compounds (XI), are generally carried out within a relatively wide temperature range, i general, they are carried out in a range of from -20 to 200°C, preferably from 0 to 100°C, and more preferably from 50 to 90°C.
  • the steps of this reaction are generally carried out under atmospheric pressure. However, it is also possible to carry them out under superatmospheric pressure or under reduced pressure (for example, in a range of from 0.5 to 5 bar).
  • the reaction time can generally be varied within a relatively wide range. In general, the reaction is finished after a period of from 2 to 24 hours, preferably from 6 to 12 hours.
  • the reaction between the compounds (N) and H ⁇ XY is preferably carried out in a solvent.
  • Suitable solvents comprise the customary organic solvents which are inert under the reaction conditions.
  • Non-limiting examples include ethers such as diethyl ether, dioxane, tetrahydrofuran, 1,2-dimethoxy ethane; hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions; halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, dichloroethane, trichloroethylene, chlorobenzene; ketones such as acetone; esters such as ethyl acetate; nitriles such as acetonitrile; heteroaromatics such as pyridine; polar solvents such as dimethyl formamide and hexamethyl phosphoric acid tris-amide; and mixtures of the above-mentioned solvents.
  • the compounds HNXY are generally employed in an amount of from 1 to 4 mol per mol of compounds (V); an equimolar amount or slight excess of compounds HNXY is preferred.
  • reaction between the compounds (V) and HNXY is preferably carried out in the presence of a coupling reagent by methods commonly known to those skilled in the art (see e.g. March, Advanced Organic Chemistry, 4 th ed., pp. 419-421 Wiley 1992).
  • Non-limiting examples include dicyclohexylcarbodiimide, l-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride, N,N'-carbonyldiimidazole, pivaloyl chloride, bis(2-oxo-3- oxazolidinyl)phosphinic chloride and benzotriazol-l-yloxytris(pyrrolidino)- phosphonium hexafluorophosphate.
  • the coupling reagents may optionally be bound to a polymeric resin.
  • the coupling reagent is generally employed in an amount of from 1 to 4 mol per mol of compounds (V); an equimolar amount or slight excess of the reagent is preferred.
  • a promoter may be added to the coupling reaction.
  • Non-limiting examples include 1-hydroxybenzotriazole, N,N'-dimethylaminopyridine, and 3-hydroxy-3H-l,2,3- triazolo[4,5-b]-pyridine.
  • the promoter is generally employed in an amount from 0.1 to 1 mol per mol of compounds (V), preferably from 0.3 to 1 mol, per mol of compounds (V).
  • the reaction of the compounds (V) and HNXY can generally be carried out within a relatively wide temperature range, hi general, the reaction is carried out within a range of from -20 to 200°C, preferably from 0 to 70°C, and generally at room temperature.
  • Compounds (LX) and (XI) are commercially available or can be synthesized according to methods commonly known to those skilled in the art (H. Feuer (ed.) "The Chemistry of the Nitro and Nitroso Groups” Interscience Publishers, New York, 1969, pp. 76-117).
  • Compounds (I) wherein R 3 is hydrogen can be synthesized by General Method A or B using compounds (III) or (V) respectively wherein R 3 is hydrogen.
  • the compounds obtained through General Method A or B can be converted into an isomer, a pharmaceutically acceptable salt, a hydrate or a hydrate of a pharmaceutically acceptable salt thereof
  • the compounds of the present invention are inhibitors of phosphodiesterase 10a (PDE 10a).
  • PDE 10a phosphodiesterase 10a
  • the biological tests described below show that the compounds (I) exhibit a pronounced anti-proliferation activity against tumor cells; they are therefore useful for the treatment of cancer.
  • our investigations showed that they are also useful for treatment of conditions of pain and/or for the lowering of the temperature of the body in fever conditions.
  • the compounds according to the invention can be used as active ingredients for the production of medicaments against carcinomatous disorders.
  • they can be converted into the customary formulations such as tablets, coated tablets, aerosols, pills, granules, syrups, emulsions, suspensions and solutions using inert, non-toxic, pharmaceutically suitable excipients or solvents.
  • the compounds according to the invention are used in an amount such that their concentration is approximately 0.5 to approximately 90% by weight, based on the ready-to-use formulations, the concentration being dependent, inter alia, on the indication of the medicament.
  • the formulations can be produced, for example, by extending the active compounds with solvents and/or excipients having the above properties, where, if appropriate, additionally emulsifiers or dispersants and, in the case of water as the solvent, an organic solvent can additionally be added.
  • Administration can be carried out in a customary manner, preferably orally, transdermally or parenterally, for example perlingually, buccally, interperitoneally, intravenously, nasally, rectally or ihhalationally.
  • the compounds according to the invention are also suitable for use in veterinary medicine.
  • the compounds or their non-toxic salts can be administered in a suitable formulation in accordance with general veterinary practice. Depending on the kind of animal to be treated, the veterinary surgeon can determine the nature of use and the dosage.
  • the present invention provides compounds for the use in a medical application, in particular for combating cancer.
  • the invention further provides a method of manufacturing a pharmaceutical composition by combining at least one of the compounds of the invention with at least one pharmacologically acceptable formulating agent.
  • the invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising as an active ingredient an effective amount of at least one of the compounds of the invention and at least one pharmacologically acceptable formulating agent.
  • the invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising as an active ingredient an effective amount of at least one of the compounds of the invention and at least one pharmaceutical active ingredient which is different from the compounds of the invention.
  • the invention further provides a medicament in dosage unit form comprising an effective amount of a compound according to the invention together with an inert pharmaceutical carrier.
  • the invention further provides a method of combating cancer in mammals comprising administering to a mammal in need thereof an antiproliferative effective amount of at least one compound according to the invention either alone or in admixture with a diluent or in the form of a medicament.
  • TMCremophor® non-ionic emulsifier from BASF, Germany DBU l,8-diazabicyclo[5.4.0]undec-7-ene DMEM Dulbecco's Modified Eagle Medium, Life Technologies,
  • HPLC equipment Thermo Separation Products P4000, AS3000, UN3000HR
  • Racemic l-(4-fluorophenyl)-ethylamine (commercially available) was resolved by (+)- tartaric acid following the procedures of A. Ault (Org. Synth., 1973, Coll. Vol. 5, 932) and S. Takenaka et al. (J.C.S. Perkin II, 1978, 95).
  • the organic material was extracted from the dark biphasic mixture using ethyl acetate (3000 mL). The combined organic extracts were washed with brine (3 x 2000 mL) and concentrated to 1/3 volume. The resultant dark oil was placed on a pad of silica gel 60 (400 cc) and eluted using ethyl acetate/hexane (1:1). The desired fractions were concentrated to a yellow oil which was seeded with a small amount of crystals of the title compound and placed in a refrigerator overnight.
  • LTMP lithium tetramethylpiperidine
  • butyllithium 37.14 mmol, 14.86 mL
  • 2,2,6,6-tetramethylpiperidine 42.86 mmol, 7.23 mL
  • tetrahydrofuran 15 mL
  • the mixture was stirred at 0 °C for 1 h.
  • This solution was added slowly to a solution of 1,2-bromofluorobenzene (5.0 g, 28.57 mmol, 3.12 mL) in tetrahydrofuran (90 mL) at - 75 °C.
  • the amines used in this experiment were prepared as 0.5 M solution in dichloromethane by using Bohdan weight station.
  • the amine solutions (270 ⁇ l, 135.0 ⁇ mol) were dispensed into 96-well format FlexChem ® Multiple Synthesis Reactor Block by using Tecan station.
  • Ethyl 2-(3-carboxyphenyl)-8,9-dimethoxy-3-methyl-5,6-dihydro-pyrrolo[2,l- a]isoquinoline-l-carboxylate 60mg/lml DCM, 137.8 ⁇ mol, Intermediate 1.3.
  • PS-DCC resin (n-cyclohexylcarbodiimide, n'- methyl polystyrene HL, 1.9 mmol/g, 140 mg, 266 ⁇ mol) was then loaded to each well of the 96-well format FlexChem ® Multiple Synthesis Reactor Block. The block was then sealed and stirred by rotation motion in a FlexChem ® Rotating Oven at ambient temperature for 12 h. The solution in each well was released to 96 deep well plate. The resin was washed with dichloromethane (3 X 2 ml) and the dichloromethane was released to the other deep well plates.
  • 4-(4-Formyl-3-methoxyphenoxy)-butyryl aminomethyl resin (NovaBiochem; 10.0 g, 0.78 mmol/g) is suspended in dichloromethane (80 ml)/trimethyl orthoformiate (40 ml). Propylamine (2.31 g, 5 equiv.) is added and the reaction mixture is shaken at room temperature overnight. The mixture is filtered and the resin is washed twice with dimethyl- formamide. The resin is re-suspended in dimethylformamide (100 ml), and tetrabutylammonium borohydride (4.01 g, 2 equiv.) is added to the mixture.
  • the propylamine resin from above (3.00 g, 2.34 mmol) is suspended in dichloromethane (30 ml). N,N-diisopropyl ethylamine (1.81 g, 6 equiv.) is added with stirring, followed by 3-formylbenzoic acid chloride (1.18 g, 3 equiv.). The mixture is shaken for 1 h at room temperature. The resin is filtered and repeatedly washed with methanol, dimethylformamide, methanol, dichloromethane and diethyl ether, and dried in vacuo.
  • the fo ⁇ nylbenzoic acid amide resin from above (1.00 g, 0.78 mmol) is suspended in dioxane (6 ml)/isopropanol (3 ml).
  • the respective nitroalkane (5 equiv.) and ammonium acetate 300 mg, 5 equiv.) are added, and the mixture is shaken at 100 °C overnight.
  • the resin is filtered, repeatedly washed (methanol, water, dimethylformamide, methanol, dichloromethane and diethyl ether), and dried in vacuo.
  • test compounds were serially diluted in DMSO using two-fold dilutions to stock concentrations ranging typically from 200 ⁇ M to 1.6 ⁇ M (final concentrations in the assay range from 4 ⁇ M to 0.032 ⁇ M).
  • 96-well assay isoplates (Wallac Inc., Atlanta, GA, U.S.A.) were loaded with 2 ⁇ L of the serially diluted individual test compounds followed by 50 ⁇ L of a dilution of crude recombinant PDE lOa-containing Sf9 cell lysate.
  • the dilution of the lysate was selected such that less than 70%> of the substrate is converted during the later incubation (typical dilution: 1:10000; dilution buffer: 50 mM Tris/hydrochloric acid pH
  • the substrate [5',8- 3 H] adenosine 3',5'- cyclic phosphate (1 ⁇ Ci/ ⁇ L; Amersham Pharmacia Biotech., Piscataway, NJ, U.S.A.), was diluted 1:2000 in assay buffer (assay buffer: 50 mM Tris/hydrochloric acid pH 7.5, 8.3 mM MgCl 2 , 1.7 mM EDTA) to give a final working concentration of 0.0005 ⁇ Ci/ ⁇ L.
  • assay buffer 50 mM Tris/hydrochloric acid pH 7.5, 8.3 mM MgCl 2 , 1.7 mM EDTA
  • MDA-MB-231 human breast carcinoma cells (ATCC # HTB26) were cultured in standard growth medium (DMEM), supplemented with 10%. heat-inactivated FBS, 10 mM HEPES, 2 mM glutamine, 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin) at 37°C in 5% CO 2 (vol/vol) in a humidified incubator. Cells were plated at a density of 3000 cells per well in 100 ⁇ L growth medium in a 96 well culture dish. 24 hours after plating, lactate dehydrogenase (LDH) activity was determined using the Cytotox 96 Non-radioactive
  • Cytotoxicity Kit (Promega, Madison WI, U.S.A.) to yield T 0 LDH values. Briefly, cells were lysed with the addition of 200 ⁇ L of Lysis Buffer (included in the Promega Kit) and lysates were further diluted 1:50 in Lysis Buffer. 50 ⁇ L of diluted cell lysate were transferred to a fresh 96 well culture plate. The assay was initiated with the addition of 50 ⁇ L of substrate per well. Color development was allowed to proceed for 10-15 minutes.
  • the assay was terminated with the addition of 50 ⁇ L of Stop Solution (included in the Promega Kit). Optical densities were determined spectrophotometrically at 490 nm in a 96 well plate reader (SpectraMax 250, Molecular Devices, Sunnyvale, CA, U.S.A.).
  • Test compounds were dissolved in 100%) DMSO to prepare 10 mM stocks. Stocks were further diluted 1 :250 in growth medium to yield working stocks of 40 ⁇ M test compound in 0.4%) DMSO. Test compounds were serially diluted in growth medium containing 0.4%> DMSO to maintain constant DMSO concentrations for all wells. 50 ⁇ L of fresh growth medium and 50 ⁇ L of diluted test compound were added to each culture well to give a final volume of 200 ⁇ L. The cells with and without individual test compounds were incubated for 72 hours at which time LDH activity was measured to yield T 2h values. Optionally, the IC 50 values can be detennined with a least squares analysis program using compound concentration versus percent inhibition.
  • T 72 ctr i LDH activity at 72 hours in the absence of test compound
  • MDA-MB-231 cells are cultured as described above. The cells are harvested by trypsinization, washed, counted, adjusted to 2.5x10 7 cells/mL with ice cold phosphate-buffered saline (PBS), and subsequently stored on ice until transplantation.
  • Test compounds are formulated for oral administration in a vehicle for oral administration composed of polyethylene glycol-400, Cremophor®, ethanol and 0.9% saline (40:5:5:50).
  • Tumor measurements are performed twice per week. Tumor weights are calculated using the formula (a x vX)!2, where a and w refer to the larger and smaller dimensions collected at each measurement.. Animals are sacrificed on day 15 after transplantation and plasma was harvested for pharmacokinetic analyses.
  • An MX-1 breast tumor xenograft model is maintained by serial passage in NCr nu/nu female mice (Taconic Farms, Germantown, NY, USA). Tumors are aseptically harvested from mice when they weigh approximately lg. The envelope and any non- viable areas are dissected and the viable tissue is cut into 3 x 3 x 3 mm cubes. These fragments are implanted in the axilary region of the flank of recipient mice using a trochar.
  • mice Treatment in anti-tumor efficacy studies is intiated when all mice have tumors ranging in size from 75-125 mg. There are typically 10 mice in each experimental group. Each experiment contains an untreated control group to monitor tumor growth kinetics, a vehicle-treated control group, and a positive agent control group to assess the response of the model in each experiment to an agent with an expected degree of anti-tumor efficacy. Lack of confonnance of any of the controls to the historical ranges for the model constitutes a reason to nullify the study. The test compounds were administered starting at different dosages (e.g. 75 and 150 mg/kg) and different schedules (e.g. qld x 10, bid x 10).
  • Test compounds are formulated for oral administration once per day in a vehicle composed of 51%) PEG400/ 12% ethanol/ 12% Cremophor® EL/ 0.1 N hydrochloric acid .
  • Tumor size is recorded in whole mm as measured in two perpendicular dimensions.
  • Animal body weights are recorded in tenths of grams. Both measurements are collected two to three times per week. Animals are sacrificed on day 10 after the last dose and last measurements.
  • Tumor weights are calculated using the equation (/ x w 2 )/2, where / and w refer to the larger and smaller dimensions collected at each measurement. Efficacy is measured as the percent suppression of tumor growth expressed as % ⁇ T/ ⁇ C, where ⁇ T and ⁇ C represent the change in the size of the average tumor in the treated and control groups, respectively, over the treatment period. Significance is evaluated using a Student's t-test with a p ⁇ 0.05.
  • the compounds according to the invention can be converted into pharmaceutical preparations as follows:
  • a single dose of 100 mg of the compound according to the invention is provided by 10 ml of oral suspension.
  • the Rliodigel is suspended in ethanol and the active component is added to the suspension.
  • the water is added with stirring. Stirring is continued for about 6h until the swelling of the Rhodigel is complete.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to 2-substituted pyrrolo[2.1-a]dihydroisoquinoline compounds which are inhibitors of phosphodiesterase 10a and can be used for com-batting cancer.

Description

2-SUBSTITUTED PYRROLO (2 .1-A) ISOQUINO INES AGAINST CANCER
BACKGROUND OF THE INVENTION The present invention relates to 2-substituted pyrrolo[2.1-a]isoquinoline derivatives which are inhibitors of phosphodiesterase 10a, a process for preparing these compounds and a method of treating cancer by administering these compounds.
Cyclic AMP metabolism is regulated by the opposing activities of adenylyl cyclase, which generates cAMP in response to extracellular stimuli (e.g. engagement of G-protein coupled receptors by their cognate ligands), and 3 ',5 '-cyclic nucleotide phosphodiesterases (PDEs), which hydrolyze cAMP to 5 '-AMP. Signal transduction via cAMP is associated with transcriptional events that can result in the inhibition of cellular proliferation (T. J. Shaw et al, Exp. Cell Res. 273, 95 (2002); T.W. Moody et al., Ann. N.Y Acad. Sci. 921, 26 (2000); W.L. Lowe et al., Endocrinology 138, 2219 (1997); D.A. Albert, J. Clin. Invest. 95, 1490 (1995); M.I. Mednieks et al., FEBS Lett. 254. 83 (1989)). Indeed, elevation of intracellular cAMP concentration is growth inhibitory for several human tumor cell lines, including those derived from breast, lung and colorectal carcinomas (B. Wagner et al., Biochem. Pharmacol. 63, 659 (2002); S.B. Jakowlew et al., Peptides 21, 1831 (2000); LS. Fentimen et al., Mol. Biol. Med. 2, 81 (1984); P. Cassoni et al., Int. J. Cancer 72, 340 (1997); S. Shafer et al, Biochem. Pharmacol. 56, 1229 (1998); N.M. Hoosein et al., Regul.
Peptides 24, 15 (1989)). In several human breast carcinoma cell lines, increased cAMP production through stimulation of adenylate cyclase activity and/or reduction in cAMP catabolism through inhibition of phosphodiesterase activity has been shown to result in increased steady state levels of cAMP and growth inhibition (D. Melck et al., FEBS Letters 463, 235 (1999); N. Veber et al., Eur. J. Cancer 30A. 1352 (1994); J.A. Fontana et al, J. Natl. Cancer hist. 78, 1107 (1987); T.A. Slotkin et al., Breast Cancer Res. and Treatment 60, 153 (2000)). In contrast to breast tumor cell lines, normal human mammary epithelial cells are stimulated to proliferate by elevation of intracellular cAMP (LS. Fentimen et al., Mol. Biol. Med. 2, 81 (1984)). These observations suggest that elevation of intracellular cAMP may selectively inhibit breast tumor cell proliferation. Interestingly, it has been reported that neoplastic mammary tissues have higher levels of low-Km phosphodiesterase activity compared to normal breast tissue, suggesting that tumors may gain a growth or survival advantage by keeping intracellular cAMP levels in check (A. Larks Singer et al., Cancer Res. 36, 60 (1976)).
The ICAST (Inhibitor of Cyclic AMP Signal Transduction) gene encodes a specific 3',5'- cyclic nucleotide phosphodiesterase. Compared to corresponding normal tissues, ICAST mRNA is overexpressed in breast carcinoma specimens, liver metastases of colorectal carcinoma and non-small cell lung carcinomas. The ICAST cDNA was also recently cloned by other groups and named PDE 10a (K. Fujishige et al., J. Biol. Chem. 274, 18 438 (1999); S.H. Soderling et al., Proc. Natl. Acad. Sci. USA 96, 7071 (1999); K. Loughney et al., Gene 234, 109 (1999)). Published expression data for ICAST mRNA show a very limited distribution across adult human tissues, with highest levels observed in the testis, caudate nucleus and putamen (K. Fujishige et al., 1999). Increased expression of ICAST mRNA in human tumor specimens indicates that ICAST may play an important role in tumor cell growth and/or survival under conditions of elevated cAMP generation. Selective inhibition of ICAST activity in tumor cells should lead to increased cAMP concentrations and growth inhibition. The expression profile of ICAST and the published reports indicating that breast, lung and colon carcinomas are particularly sensitive to elevation of intracellular cAMP indicate that ICAST may play critical roles specifically in those tumor types. In addition to elevation of cAMP, inhibition of ICAST activity should also decrease the intracellular concentration of 5-AMP, which could limit purine pools and DNA synthesis in rapidly dividing tumor cells.
Pyrrolo[2.1-a]isoquinoline derivatives of formula (A) are described in J. Med. Chem. 27, 1321 (1984) and in J. Med. Chem. 31, 2097 (1988):
Figure imgf000004_0001
R' = H, OMe, Cl
R" = H, Cl
R"' = H, Me
R"", R"'" = Me, Et, i-Pr, CβH^
These compounds are described as having antineoplastic activity, which however is stated to be due to the carbamate moieties being electrophilic centers enabling the compounds (A) to react via an alkyl-oxygen cleavage mechanism. It is not mentioned that these compounds have any PDE 10a inhibitory activity.
Tetracyclic compounds of formula (B) containing a pyrrolo[2.1-a]isoquinoline moiety are described in Arch. Pharm. 321, 81 (1988):
Figure imgf000004_0002
R = H, OMe
The compounds (B) are described as having anti-tumor activity due to their ability to intercalate into DNA. It is not mentioned that they have any PDE 10a inhibitory activity.
The synthesis of pyrrolo[2.1-a]isoquinoline derivatives of formula (C) is described in H.
Meyer, Liebigs Ann. Chem. 9, 1534-1544 (1981):
Figure imgf000005_0001
R = H, OCH3 R" = CH3, CR 6Hπ5 R'" = C6H5
These compounds are not described as having any biological activity, and it is not mentioned that they have any PDE 10a inhibitor activity.
Compounds of the formula (D) are described in GB 1 153 670 A:
Figure imgf000005_0002
R = H, CO2H, CO2R""
Figure imgf000005_0003
These compounds are described as having hypotensive, sympathicolytic and psychotropic properties, but it is not mentioned that they have any PDE 10a inhibitory activity.
The synthesis of compounds of the formula (E) is described in US Patent 4,694,085:
Figure imgf000006_0001
R = H. CH3, OCH3 R" = H, CH3 R = CβH5, CH3, CO2R R"" = H, CH3
It is not mentioned that these compounds have any PDE 10a inhibitory activity.
Derivatives of the formula (F) are described in WO 98/55118:
Figure imgf000006_0002
R = H, Cl, OCH3 R" = CH3
R'" = OR , CH3, NH2
R"" = H, CH3, OR""'
These compounds are described as useful for the treatment of diseases such as psoriasis. However, the compounds disclosed in WO 98/55118 are described as having virtually no cytotoxic activity; it is not mentioned that they have any PDE 10a inhibitor activity.
BRIEF SUMMARY OF THE INVENTION
Surprisingly, it has been found that the 2-substituted pyrrolo[2.1-a]isoquinoline derivatives of the present invention inhibit PDE 10a and exhibit an antiproliferative activity. The present invention relates to a compound of the formula
Figure imgf000007_0001
wherein
x and y independently from each other denote zero or 1 ;
R1 and R2 independently from each other denote hydrogen, Cι- -alkyl or trifluoromethyl or
R1 and R2 together form a Cι- -alkylene bridge;
R3 and R4 independently from each other denote Cι-6-alkyl optionally further substituted with halogen up to perhalo;
R5 denotes a radical of the formula
Figure imgf000007_0002
wherein
R6 denotes Ci-6-alkyl, trifluoromethyl, trifluoromethoxy, halogen, hydrogen, hydroxy or Cι-6-alkoxy; X and Y independently from each other denote
i) hydrogen; ii) Cι_6-alkoxy; iii) C3.8-cycloalkyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι_6-alkyl, hydroxy, benzyloxy, Cι-6- trialkylsilyloxy, halogen and Ci-β-alkoxy; iv) C5.8-cycloalkyl fused to C6-Cι0-aryl, optionally having from 1 to 3 substituents selected from the group consisting of hydoxyl, Cι_6- alkyl, trifluoromethyl, trifluoromethoxy, C3-8-cycloalkyl, halogen and -6-alkoxy; v) C5-Cι0-bridged bicycloalkyl; vi) C6-C10-aryl optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, C3_8-cycloalkyl, Cι-6-alkoxy, (C\.
6-alkyl)-carbonyl, (Cι-6-alkoxy)-carbonyl, trifluoromethyl, trifluoromethoxy and halogen; vii) phenyl fused to a 5- to 7-membered saturated cycloalkyl, optionally containing up to two hetero atoms selected from the group consisting of O, N, and S; viii) heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkoxy, phenyloxy, benzyloxy, C . 6-alkyl, C -8-cycloalkyl, trifluoromethyl, trifluoromethoxy, halogen and benzyl; ix) heteroaryl; or x) Cι_6-alkyl, optionally having from 1 to 3 substituents selected from the group consisting of a) halogen up to perhalo, b) cyano, c) -OR7, d) -NR7R8, e) C6-C10-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, nitro, C^- alkyl, C3.8-cycloalkyl, -OR7, -NR7R8, (Cι-6-alkyl)-carbonyl, (Cι-6-alkoxy)-carbonyl, C^-alkoxy, trifluoromethyl, trifluoromethoxy, -C(O)NR7R8, cyano, -SR7, and C6-Cι0-aryl, f) phenyl fused to a 5- to 7-membered saturated cycloalkyl, optionally containing up to two hetero atoms selected from the group consisting of O, N, and S, g) heteroaryl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, C3.8-cycloalkyl, hydroxy, Cι_6-alkoxy, benzyloxy, halogen and trifluoromethyl, h) heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, C^ό-alkoxy, C3.8-cycloalkyl, halogen and benzyl, and i) C3-g-cycloalkyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, Cι_6-alkoxy and hydroxy;
wherein R7 and R8 independently from each other denote
1) hydrogen,
2) -o-alkyl, optionally further substituted with 1 to 3 substituents selected from the group consisting of hydroxyl and halogen, 3) C3.8-cycloalkyl,
4) benzyl,
5) C6-C10-aryl optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkoxy, Cι-6-alkyl, C3-8-cycloalkyl, trifluoromethyl, trifluoromethoxy and halogen, or
6) heteroaryl; or
X and Y together with the nitrogen atom to which they are attached form heteroaryl or heterocyclyl, wherein said heteroaryl or heterocyclyl may optionally have from 1 to 3 substituents selected from the group consisting of i) C3.8-cycloalkyl; ii) Cι-6-alkyl, optionally having from 1 to 3 substituents selected from the group consisting of C3-8-cycloalkyl, hydroxy, Cι_6-alkoxy, halogen, phenylamino, morpholinyl, (Cι-6-alkyl)-aminocarbonyl, benzo[2,3]dioxolyl and C6-C10-aryl, wherein said aryl is optionally substituted with Cι_6-alkyl or Cι-6-alkoxy; iii) C6-Cιo-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι_6-alkoxy, Cι_6-alkyl, C3-8- cycloalkyl, trifluoromethyl, trifluoromethoxy and cyano, iv) hydroxy; v) Cι-6-alkoxy; vi) Cι-6-dialkylamino; vii) (Ci_6-alkoxy)-carbonyl; viii) aminocarbonyl; and ix) heteroaryl;
or
X and Y together with the nitrogen atom to which they are attached form heterocyclyl fused to C6-C10-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Ci-6- alkyl, C3.8-cycloalkyl, Cι.6-alkoxy and hydroxymethyl;
or X denotes hydrogen and
Y denotes -NR9R10; wherein R9 and R10 independently from each other denote 1) hydrogen,
2) C6-Cιo-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι-6-alkyl, C3. 8-cycloalkyl, Cι-6-alkoxy, trifluoromethoxy and trifluoromethyl, 3) heterocyclyl,
4) C3.8-cycloalkyl, or
5) d-6-alkyl;
or
R9 and R10 together with the nitrogen atom to which they are attached form heterocyclyl or heteroaryl, wherein said heteroaryl or heterocyclyl may optionally have from 1 to 3 substituents selected from the group consisting of Ci-6-alkyl, Cι-6-alkoxy, halogen and methoxymethyl;
and an isomer, a pharmaceutically acceptable salt, a hydrate or a hydrate of a pharmaceutically acceptable salt thereof.
An alternative embodiment of the present invention relates to compounds of the formula
(I), wherein
x and y independently from each other denote zero or 1 ;
R and R independently from each other denote hydrogen, Cι-4-alkyl or trifluoromethyl; R3 and R4 independently from each other denote Ci-6-alkyl optionally further substituted with halogen up to perhalo;
R5 denotes a radical of the formula
Figure imgf000012_0001
wherein
R6 denotes halogen, hydrogen, hydroxy or Ci-6-alkoxy;
X and Y independently from each other denote
i) hydrogen; ii) Cι-6-alkoxy; iii) C3-8-cycloalkyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, hydroxy, benzyloxy and Cι_ 6-trialkylsilyloxy; iv) Cs-s-cycloalkyl fused to C6-Cι0-aryl, optionally substituted with 1 to
3 hydoxyl; v) C5-C10 bridged bicycloalkyl; vi) Ce- o-aryl, optionally having from 1 to 3 substituents selected from the group consisting of Cι_6-alkyl, Cι-6-alkoxy and (Cι_6-alkyl)- carbonyl; vii) phenyl fused to a 5- to 7-membered saturated cycloalkyl, optionally containing up to two hetero atoms selected from the group consisting of O, N, and S; viii) heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of -β-alkoxy, phenyloxy, benzyloxy, and benzyl; ix) heteroaryl; or x) Cι-6-alkyl, optionally having from 1 to 3 substituents selected from the group consisting of a) halogen up to perhalo, b) cyano, c) -OR7, d) -NR7R8, e) C6-Cιo-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, nitro, hydroxy,
Ci-e-alkyl, -NR7R8, Cι_6-alkoxy, trifluoromethyl, trifluoromethoxy and C6-Cιo-aryl, f) phenyl fused to a 5- to 7-membered saturated cycloalkyl, optionally containing up to two hetero atoms selected from the group consisting of O, N, and S, g) heteroaryl, optionally having from 1 to 3 substituents selected from the group consisting of Cι_6-alkyl, hydroxy, Ct-β-alkoxy, benzyloxy and trifluoromethyl, h) heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl and benzyl, and i) C3-8-cycloalkyl, optionally further substituted with 1 to 3 substituents hydroxy;
wherein R7 and R8 independently from each other denote
1) hydrogen,
2) Cι-6-alkyl, optionally further substituted with 1 to 3 substituents hydroxyl,
3) benzyl, 4) C6-Cιo-aryl, optionally further substituted with 1 to 3 substituents Cι_6-alkoxy, or
5) heteroaryl;
or
X and Y together with the nitrogen atom to which they are attached form heteroaryl or heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of i) C3-8-cycloalkyl; ii) Cι-6-alkyl, optionally having from 1 to 3 substituents selected from the group consisting of C3-8-cycloalkyl, hydroxy, Cι-6-alkoxy, phenylamino, morpholinyl,
Figure imgf000014_0001
benzo[2,3]dioxolyl and C6-Cι0-aryl, wherein said aryl is optionally substituted with Ci-6-alkyl or -6-alkoxy; iii) C6-C10-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι-6-alkoxy, trifluoromethyl and cyano; iv) hydroxy; v) Cι-6-alkoxy; vi) Cι-6-dialkylamino; vii) (Cι_6-alkoxy)-carbonyl; viii) aminocarbonyl; and ix) heteroaryl;
or
X and Y together with the nitrogen atom to which they are attached form heterocyclyl fused to C6-Cιo-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι-6- alkyl, Cι-6-alkoxy and hydroxymethyl; or
X denotes hydrogen and Y denotes -NR9R10; wherein R9 and R10 independently from each other denote
1) hydrogen,
2) C6-C10-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι-6-alkyl and trifluoromethyl,
3) heterocyclyl,
4) C3.8-cycloalkyl, or
5) Ci-β-alkyl;
or
R9 and R10 together with the nitrogen atom to which they are attached form heterocyclyl or heteroaryl, wherein said heterocyclyl or heteroaryl optionally have from 1 to 3 substituents selected from the group consisting of Cι_6-alkyl and methoxymethyl;
and an isomer, a pharmaceutically acceptable salt, a hydrate or a hydrate of a pharmaceutically acceptable salt thereof.
A further alternative embodiment of the present invention relates to a compound of the formula (I), wherein
x and y each other denote 1
R1 and R2 independently from each other denote hydrogen or Cι_4-alkyl; Rj denotes Cι-6-alkyl or trifluoromethyl;
R4 denotes Cι- -alkyl;
R5 denotes a radical of the formula selected from the group consisting of:
Figure imgf000016_0001
wherein
X and Y independently from each other denote
i) hydrogen; ii) Cι_6-alkoxy; iii) C3-8-cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl, said C3.8-cycloalkyl optionally having from 1 to 2 substituents selected from the group consisting of Cι-6-alkyl, hydroxy, benzyloxy and tert- butyldimethylsilyloxy; iv) indanyl, 2-hydroxyindanyl, or 1,2,3,4-tefrahydronaphthalenyl; v) [2.2.1]bicycloheptane; vi) naphthyl, 4-methoxyphenyl, 3-(Cι_6-alkoxycarbonyl)phenyl or 2- methoxy-4-methylphenyl; vii) benzo[2,3]dioxolyl; viii) pyiTolidinyl, optionally having from 1 to 2 substituents selected from the group consisting of Cι-6-alkoxy, phenyloxy, benzyloxy and benzyl; ix) thiazolyl, or pyridyl; or x) Cι-6-alkyl, optionally having from 1 to 2 substituents selected from the group consisting of a) halogen up to perhalo, b) cyano, c) hydroxy, Cι_6-alkoxy, benzyloxy, hydroxy-C2.6-alkoxy, or methoxyphenoxy, d) Cι-6-dialkylamino, di-(hydroxy-Cι-6-alkyl)-amino, pyridylamino, or anilino, e) C6-ιo-aryl selected from the group consisting of naphthyl and phenyl, said C6-10-aryl optionally having from 1 to 2 substituents selected from the group consisting of fluoro, chloro, bromo, iodo, nitro, hydroxy, Cι-6-alkyl, -β-alkoxy, trifluoromethyl, trifluoromethoxy, phenyl, amino and Ci-6- dialkylamino, f) benzo[2,3]dioxolyl, or 2,3-dihydrobenzo[ 1 ,4]dioxinyl, g) heterocyclyl selected from the group consisting of pyrazolyl, pyrazinyl, pyrrolyl, furyl, indolyl, thienyl, imidazoyl, and pyridyl, said heterocyclyl optionally having from 1 to 2 substituents selected from the group consisting of Cι.6-alkyl, hydroxy, Cι-6-alkoxy, benzyloxy and trifluoromethyl, h) morpholino, tetrahydrofuranyl, piperidinyl, pyrrolidinyl, optionally further substituted with 1 to 2 substituents Cι-6- alkyl or benzyl, and i) C3-8-cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl, said C3. 8-cycloalkyl optionally being further substituted with 1 to 2 substituents hydroxy;
or
X and Y together with the nitrogen atom to which they are attached form i) morpholino, optionally further subsituted with 1 to 2 substituents Ci- 6-alkyl; ii) piperidinyl, optionally having from 1 to 2 substituents selected from the group consisting of hydroxyl, hydroxymethyl and Cι_6-alkyl; iii) pyrrolidinyl, optionally having from 1 to 2 substituents selected from the group consisting of Cι-6-dialkylamino, pyridyl, carboxamido, Ci- 6-alkoxy, phenylaminomethyl, methoxymethyl and methoxyphenyl; or iv) piperazinyl, optionally having from 1 to 2 substituents selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexylmethyl, benzyl, morpholinoethyl, Ci-e-alkyl, (C1-6-alkoxy)-carbonyl, (Cι-6-alkylaminocarbonyl)methyl, pyridyl, pyrazinyl, pyridylmethyl, benzo[2,3]dioxolyl and phenyl, wherein said phenyl is optionally substituted with 1 to 2 substituents selected from the group consisting of fluoro, chloro, bromo, iodo, trifluoromethyl, cyano and C^-alkoxy;
or
X and Y together with the nitrogen atom to which they are attached fomi dimethoxytetrahydroisoquinolinyl, 2-methyl-6- fluorotetrahydroquinolinyl, indolinyl, isoindolinyl or
2-hydroxymethyltetrahydroisoquinolinyl; or
X denotes hydrogen and Y denotes a) phenylamino, having from 1 to 2 substituents selected from the group consisting of fluoro, chloro, bromo, iodo, trifluoromethyl and Ci-e-alkyl; b) piperidinyl, optionally further substituted with 1 to 2 -β- alkyl; c) triazolyl; d) pyrrolidinyl, optionally further substituted with 1 to 2 methoxymethyl; e) morpholino; f) imidazoyl; g) C3_8-cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl; h) Ci-6-dialkylamino; or i) azepanyl;
and an isomer, a pharmaceutically acceptable salt, a hydrate or a hydrate of a pharmaceutically acceptable salt thereof.
A further alternative embodiment of the present invention relates to the compounds of the
Examples 4, 6, 67, 77, 90, 93, 158, 170, 171, 227, 234, 244, 284, 295, 313, 376, 392 and 381.
A further alternative embodiment of the present invention relates to a compound of the formula (T):
Figure imgf000020_0001
wherein R >3 to R are defined as described above.
A further alternative embodiment of the present invention relates to a compound of the formula (I), wherein
R3 denotes a radical of the formula selected from the group consisting of:
Figure imgf000020_0002
A further alternative embodiment of the present invention relates to a compound of the formula (I), wherein
x and y independently from each other denote zero or 1 and x+y is 1 or 2;
1 0
R and R independently from each other denote hydrogen, Cι- -alkyl or CF or R1 and R2 together form a Cι-4-alkylene bridge;
R3 and R4 independently from each other denote Cι- -alkyl;
Rs denotes a monovalent radical of the formula
Figure imgf000021_0001
wherein
X and Y i ndepende ntly from each other denot< hydrogen, ^-alkyl optionally substituted with furyl which can be further substituted with 1 to 2 substituents methyl, or
C .8-cycloalkyl or X and Y together with the nitrogen atom to which they are attached, form piperazinyl optionally further substituted (i) with 1 to 2 substituents C3.8-cycloalkyl or (ii) with 1 to 2 substituents C^-alkyl optionally further substituted with 1 substituent C3.8-cycloalkyl;
and an isomer, a pharmaceutically acceptable salt, a hydrate or a hydrate of a pharmaceutically acceptable salt thereof.
Compounds (T) wherein the radicals (R O)x and (R2O)y are attached to the phenyl ring in the following positions, are preferred:
Figure imgf000022_0001
Depending on the substitution pattern, the compounds of the formula (I) according to the invention can exist in stereoisomeric forms which are either like image and mirror image (enantiomers) or are not like image and mirror image (diastereomers). The invention relates both to the enantiomers or diastereomers and to their respective mixtures. The racemic forms, like the diastereomers, can be separated in a known manner into the stereoisomerically uniform components.
Furthermore, certain compounds of the formula (I) can be present in tautomeric forms. This is known to the person skilled in the art, and such compounds are likewise included in the scope of the invention.
DETAILED DESCRIPTION OF THE INVENTION
Pharmaceutically acceptable salts according to the invention are non-toxic salts which in general are accessible by reaction of the compounds (I) with an inorganic or organic base or acid conventionally used for this purpose. Non-limiting examples of pharmaceutically acceptable salts of compounds (I) include the alkali metal salts, e.g. lithium, potassium and sodium salts, the alkaline earth metal salts such as the magnesium and calcium salts, the quaternary ammonium salts such as, for example, the triethyl ammonium salt, acetates, benzene sulphonates, benzoates, dicarbonates, disulphates, ditartrates, borates, bromides, carbonates, chlorides, citrates, dihydrochlorides, fumarates, gluconates, glutamates, hexyl resorcinates, hydrobromides, hydrochlorides, hydroxynaphthoates, iodides, isothionates, lactates, laurates, malates, maleates, mandelates, mesylates, methylbromides, methylnitrates, methylsulphates, nitrates, oleates, oxalates, palmitates, pantothenates, phosphates, diphosphates, polygalacturonates, salicylates, stearates, sulphates, succinates, tartrates, tosylates, valerates, and other salts used for medicinal purposes.
According to the invention, "hydrates" are forms of the compounds of the formula (I) above which, in the solid or liquid state, form a molecular compound (solvate) by hydration with water. Examples of hydrates are sesquihydrates, monohydrates, dihydrates and trihydrates. Equally suitable are the hydrates of salts of the compounds according to the invention.
In the context of the present invention, the substituents, if not stated otherwise, in general have the following meanings:
Halogen represents fluorine, chlorine, bromine and iodine. Preference is given to chlorine and fluorine.
QrCfi-Alkyl per se as well as the prefixes "alkyl" and "alk" in the terms "alkylcarbonyl", "alkoxy", and "alkoxycarbonyl" represents a straight-chain or branched alkyl radical preferrably having from 1 to 6 carbon atoms. Examples which may be mentioned are: methyl, ethyl, n-propyl, isopropyl, n-butyl, ώo-butyl, tert-butyl, n-pentyl, n-hexyl, n-octyl and «-decyl. The corresponding alkyl groups having fewer carbon atoms, such as, for example, Cι-C -alkyl, are derived analogously from this definition.
Cj-Cs-Cvcloalkyl represents a mono- or bicyclic alkyl radical having 3 to 8 carbon atoms. Examples which may be mentioned are: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl. The corresponding cycloalkyl groups having fewer carbon atoms, such as, for example, C3-C6-cycloalkyl, are derived analogously from this definition. Non-limiting examples of
Figure imgf000024_0001
radicals include methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxycarbonyl and isobutoxycarbonyl. The corresponding alkoxycarbonyl groups having fewer carbon atoms, such as, for example, C3-C6-alkoxycarbonyl, are derived analogously from this definition.
Cj^-Cg-Alkoxy represents a straight-chain or branched alkoxy radical having 1 to 6 carbon atoms. Examples which may be mentioned are: methoxy, ethoxy, «-propoxy, iso-pxopoxy, n-bvAoxy, iso-butoxy, tert-butoxy, w-pentoxy and n-hexoxy. The corresponding alkoxy groups having fewer carbon atoms, such as, for example, Cι-C4-alkoxy, are derived analogously from this definition.
Ci-Cή-Dialkylamino represents an alkylamino radical having two (independently selected) alkyl substituents, illustratively and preferably representing NN-dimethylamino, N,N- diethylamino, N-ethyl-N-methylamino, N-meti yl-N-n-propylamino, N-isopropyl-N-n- propylamino, N-t-butyl-N-methylamino, N-ethyl-N-n-pentylamino and N-n-hexyl-N- methylamino.
Cfi-Cκι-Aryl represents an aromatic radical preferably having 6 to 14, more preferably 6 to 10 carbon atoms. Νon-limiting examples of Cg-Cui-aryl radicals include phenyl, and naphthyl.
Heteroaryl in the context of the invention represents a preferably 5- to 13-membered heteroaryl or a 5- to 13-membered aromatic heterocycle having from 1, up to 4, heteroatoms from the group consisting of Ν, O and S, which ring or ring system can be linked via a carbon atom or a nitrogen atom, if such an atom is present. Examples which may be mentioned are: pyridyl, pyridyl N-oxide, pyrimidyl, pyridazinyl, pyrazinyl, thienyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl or isoxazolyl, indolicenyl, indolyl, benzo[b]thienyl, benzo[b] furyl, benzothiadiazolyl, indazolyl, quinolyl, isoquinolyl, naphthyridinyl, quinazolinyl. Preferred examples include pyrazolyl, pyrazinyl, pyrrolyl, imidazoyl, triazolyl, indolyl, pyridyl, pyridyl-N-oxide, pyrimidyl, pyridazinyl, furyl, imidazolyl and thienyl.
Heterocycle in the context of the invention represents a preferably 3- to 13-membered saturated or partially unsaturated heterocycle having from 1, up to 4, heteroatoms from the group consisting of N, O and S, which ring or ring system can be linked via a carbon atom or a nitrogen atom, if such an atom is present. Examples which may be mentioned are: tetrahydropyranyl, aziridyl, azepanyl, tetrahydrofuryl, pyrrolidinyl, pyrrolinyl, piperidinyl, 1,2-dihydropyridinyl, 1,4-dihydropyridinyl, piperazinyl, morpholinyl, thiomorpholinyl, azepinyl, and 1,4-diazepinyl. Preference is given to azepanyl, piperazinyl, piperidinyl, morpholinyl and pyrrolidinyl.
C ^-Alkylene or C ^-alkylene bridge represents a linear or branched, bivalent alkylene radical preferably having 1 to 4 carbon atoms, also known as alkandiyl. Non-limiting examples include methylene, ethylene, propylene, α-methylethylene, β-methylethylene, α- ethylethylene, β-ethylethylene, butylene, α-methylpropylene, β-methylpropylene, and γ- methylpropylene.
CyCin-Bridged bicycloalkyl represents a bicyclic alkyl radical having 5 to 10 carbon atoms also containing an alkylene bridge. Non-limiting examples include [2.2.1]bicycloheptane, [2.2.2]bicyclooctanetane, [2.1.1]bicyclohexane,
[3.3.1]bicyclononane, and [3.3.2]bicyclodecane. A ring system represents a mono-, bi- or tricyclic system of fused rings. These rings share two ring members. These ring members are preferably adjacent ([0]-bridge). Non limiting examples include naphthalene, benzo[2,3]dioxolyl, 2,3-dihydrobenzo[l,4]dioxinyl, and indole.
A * symbol next to a bond denotes the point of attachment in the molecule.
The compounds according to the invention exhibit an unforeseeable, useful pharmacological and pharmacokinetic activity spectrum. They are therefore suitable for use as medicaments for the treatment and/or prophylaxis of disorders in humans and animals.
The compounds of this invention may be formulated as a solution of lyophilized powders for parenteral administration. Powders may be reconstituted by addition of a suitable diluent or other pharmaceutically acceptable carrier prior to use. The liquid formulation is generally a buffered, isotonic, aqueous solution. Examples of suitable diluents are normal isotonic saline solution, standard 5% dextrose in water or in buffered sodium or ammonium acetate solution. Such formulation is especially suitable for parenteral administration, but may also be used for oral administration. It may be desirable to add excipients such as polyvinylpyrrolidone, gelatin, hydroxycellulose, acacia, polyethylene glycol, mannitol, sodium chloride, or sodium acetate.
Alternatively, the compounds of the present invention may be encapsulated, tableted, or incorporated into an emulsion (oil-in-water or water-in-oil) syrup for oral administration. Pharmaceutically acceptable solids or liquid carriers, which are generally known in the pharmaceutical formulary arts, may be added to enhance or stabilize the composition, or to facilitate preparation of the composition. Solid carriers include starch (corn or potato), lactose, calcium sulfate dihydrate, terra alba, croscarmellose sodium, magnesium stearate or stearic acid, talc, pectin, acacia, agar, gelatin, maltodextrins and microcrystalline cellulose, or colloidal silicon dioxide. Liquid carriers include syrup, peanut oil, olive oil, corn oil, sesame oil, saline, and water. The carrier may also include a sustained release material such as glyceryl monostearate or glyceryl distearate, alone or with a wax. The amount of solid carrier varies but, preferably, will be between about 10 mg to about 1 g per dosage unit.
The dosage ranges for administration of the heterocyclics disclosed in this invention are those to produce the desired affect. The dosage will generally vary with age, body weight, extent of the disease, and contraindications, if any. The dosage will also be deteπnined by the existence of any adverse side effects that may accompany the compounds. It is always desirable, whenever possible, to keep adverse side effects to a minimum. One skilled in the art can easily determine the appropriate dosage, scheduling, and method of administration for the exact formulation of the composition being used in order to achieve the desired effective concentration in the individual patient. However, the dosage can vary from between about 1 mg/kg/day to about 500 mg/kg/day, and preferable from between about 1 mg/kg/day to about 50 mg/kg/day.
One skilled in the art will recognize that modifications may be made in the present invention without deviating from the spirit or scope of the invention. The invention is illustrated further by the following experimental information and examples, which are not to be construed as limiting the invention in spirit or scope to the specific procedures or compositions described in them.
The present invention also relates to a process for making the compounds. The compounds of the invention may be prepared by use of known chemical reactions and procedures.
Nevertheless, the following general preparative methods are presented for synthesis of the 2-substituted pyrrolo[2.1-a]isoquinoline compounds of the present invention, with more detailed particular examples being presented below in the experimental section describing the working examples. Variables are defined above in the general description. The processes can be illustrated by the following scheme 1 :
General Method A:
General Method Al
Figure imgf000028_0001
The compounds (I) can be synthesized according to the reaction sequence shown in Method Al above. Thus, the compounds (VIII) can be synthesized by reacting amino compounds (VI) with compounds (VII), wherein L is a leaving group, for example a halogen radical such as Cl, or a radical of the formula -OC(O)R5, wherein R5 is defined as optionally substituted C^-alkyl or -CH2C(O)OR4. Compounds (IN) are obtained by reacting compounds (NIII) with a dehydrating agent such as, e.g., phosphorous pentoxide. Reacting compounds (IN) with carboxybenzaldehydes (II) and R3-CH2-ΝO2 (III) provides compounds (V). Reacting the carboxy compounds (V) with the amine HNXY furnishes the amides (I). General Method A2
Figure imgf000029_0001
Alternatively, the transformation of compounds (LV) into compounds (V) in Method Al can be accomplished with the conditions shown in General Method A2. Thus, treatment of compounds (IN) with compounds (LX) provides compounds (N).
General Method B:
General Method Bl
Figure imgf000029_0002
The compounds (LV) can also be directly converted to the compounds (f) according to the General Method Bl. Thus, treatment of compounds (LV) with compounds (X) and (III) gives compounds (I).
General Method B2
Figure imgf000029_0003
Alternatively, the transformation of compounds (LV) into compounds (I) can be accomplished with the conditions shown in General Method B2. Thus, treatment of compounds (IN) with compounds (XI) furnishes compounds (I).
The compounds (VI) are commercially available or can be synthesized according to methods commonly known to those skilled in the art (H. Mayer et al., Heterocycles 31, 1035 (1990); E. Mϋller (ed.), "Methoden der Organischen Chemie" (Houben-Weyl), 4th ed., Vol. 11/1 Stickstoff-Nerbindungen II, Georg Thieme Nerlag, Stuttgart 1957; Shepard et al. in J. Org. Chem. 17, 568 (1952) and in J. Am. Chem. Soc. 72, 4364 (1950)).
The compounds (Nil) are commercially available or can be synthesized according to methods commonly known those skilled in the art [e.g. via acylation of acetic acid with an alkyl chloroformate or dialkyl carbonate (March, Advanced Organic Chemistry, 3rd ed., p. 440-441, Wiley 1985) and converting the resulting monoester of malonic acid into e.g. the corresponding acid chloride or anhydride by methods commonly known to those skilled in the art (see e.g. March, Advanced Organic Chemistry, 3rd ed., p. 355, 388, Wiley 1985)].
The reaction between the compounds (VI) and (Nil) is preferably carried out in a solvent. Suitable solvents comprise the customary organic solvents which are inert under the reaction conditions. Νon-limiting examples include ethers such as diethyl ether, dioxane, tetrahydrofuran, 1,2-dimethoxy ethane; hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions; halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, dichloroethane, trichloroethylene, chlorobenzene; ketones such as acetone; esters such as ethyl acetate; nitriles such as acetonitrile; heteroaromatics such as pyridine; polar solvents such as dimethyl formamide and hexamethyl phosphoric acid tris-amide; and mixtures of the above-mentioned solvents. Dichloromethane is frequently used.
The compounds (Nil) are generally employed in an amount of from 1 to 4 mol per mol of compounds (NI); an equimolar amount or slight excess of compounds (VII) is preferred. The reaction between the compounds (VI) and (VH) is preferably carried out in the presence of a base. Non-limiting examples include alkali metal hydrides and alkali metal alkoxides such as, for example, sodium hydride and potassium tert-butoxide; Cι-C -alkyl amines such as, for example, triethyl amine; cyclic amines such as, for example, piperidine, pyridine, dimethylamino pyridine and -preferably - l,8-diazabicyclo[4.3.0]undec-7-ene (DBU). The base is generally employed in an amount of from 1 to 4 mol per mol of compounds (VI); an equimolar amount or slight excess of the base is preferred.
The reaction of the compounds (VI) and (Nil) can generally be carried out within a relatively wide temperature range. In general, the reaction is carried out within a range of from -20 to
200°C, preferably from 0 to 70°C, and more preferably at room temperature.
For the cyclization of the compounds (Nffl) to yield compounds (IV), dehydrating agents such as, for example, P2O5 or POCl3 are generally employed in an amount of from 1 to 10 mol, preferably from 3 to 8 mol, per mol of compounds (VHf).
The cyclization reaction of the compounds (NIL-) to yield the compounds (IN) is also preferably carried out in a solvent. Νon-limiting examples comprise the customary organic solvents which are inert under the reaction conditions. They preferably include ethers such as diethyl ether, dioxane, tetrahydrofuran, 1,2-dimethoxy. ethane; hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions; halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, dichloroethane, trichloroethylene, chlorobenzene; esters such as ethyl acetate; ketones such as acetone; nitriles such as acetonitrile; heteroaromatics such as pyridine; polar solvents such as dimethyl formamide and hexamethyl phosphoric acid tris-amide; and mixtures thereof.
Toluene is preferred, if the reaction is carried out with P2O5, and acetonitrile is preferred, if the reaction is carried out with POCl3 (Benovsky, Stille, Tetrahedron Lett. 38, 8475-8478 (1997)).
The temperature for the cyclization reaction of compounds (NLII) is preferably within a range of from 60 to 200°C and more preferably within a range of from 80 to 120°C. The above process steps are generally carried out under atmospheric pressure. However, it is also possible to carry them out under superatmospheric pressure or under reduced pressure (for example, in a range of from 0.5 to 5 bar). The reaction time can generally be varied within a relatively wide range, i general, the reaction is finished after a period of from 2 to
24 hours, preferably from 6 to 12 hours.
The compounds (II) are commercially available or can be synthesized according to methods commonly known to those skilled in the art (LT. Harrison and S. Harrison, Compendium of Organic Synthetic Methods, pp. 132-176, Wiley-Interscience; E. Mύller
(ed.), " Methoden der Organischen Chemie" (Houben-Weyl), Vol. VII/1 Sauerstoff- Verbindungen II, Georg Tl ieme Verlag, Stuttgart 1954).
The compounds (III) are commercially available.
The reaction of the compounds (LV) with compounds (II) and (HI), or with compounds (X) and (in), or with compounds (IX), or with compounds (XI), can be carried out as a one-pot synthesis, preferably in a solvent. Suitable solvents comprise the customary organic solvents which are inert under the reaction conditions. Non-limiting examples include ethers such as diethyl ether, dioxane, tetrahydrofuran, 1,2-dimethoxy ethane; hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions; halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachlori.de, dichloroethane, trichloroethylene, chlorobenzene; alcohols such as methanol, ethanol, n- propanol, isopropanol; esters such as ethyl acetate; ketones such as acetone; nitriles such as acetonitrile; heteroaromatics such as pyridine; polar solvents such as dimethyl formamide and hexamethyl phosphoric acid tris-amide; and mixtures of the above-mentioned solvents. Ethanol/isopropanol (preferably in an approximate vol/vol-ratio of 1:1) mixtures are preferred.
The compounds (LU) are generally employed in an amount of from 1 to 3 mol per mol of compounds (LI); an equimolar amount or slight excess of compounds (LU) is preferred. The compounds (LV) are generally employed in an amount of from 0.1 to 1 mol, preferably from 0.3 to 1 mol, per mol of compounds (II). The compounds (LX) or (XI) are generally employed in an amount from 1 to 3 mol per mol of compounds (IV); an equimolar amount or slight excess of compounds (III) is preferred.
The reactions of the compounds (IV) with compounds (II) and (IE), or with compounds (X) and (III), or with compounds (DC), or with compounds (XL), are preferably carried out in the presence of a base. Non-limiting examples include alkali metal hydrides and alkali metal alkoxides such as, for example, sodium hydride and potassium tert-butoxide; Cwalkyl amines such as, for example, triethyl amine; cyclic amines such as, for example, pyridine, dimethylamino pyridine, l,8-diazabicyclo[4.3.0]undec-7-ene (DBU) and - preferably - piperidine. The base is generally employed in an amount of from 0,1 to 1 mol, preferably from 0,3 to 1 mol, per mol of compounds (II) or compounds (V), respectively.
The reactions of the compounds (LV) with compounds (II) and (III), or with compounds (X) and (LQ), or with compounds (IX), or with compounds (XI), are generally carried out within a relatively wide temperature range, i general, they are carried out in a range of from -20 to 200°C, preferably from 0 to 100°C, and more preferably from 50 to 90°C. The steps of this reaction are generally carried out under atmospheric pressure. However, it is also possible to carry them out under superatmospheric pressure or under reduced pressure (for example, in a range of from 0.5 to 5 bar). The reaction time can generally be varied within a relatively wide range. In general, the reaction is finished after a period of from 2 to 24 hours, preferably from 6 to 12 hours.
The compounds HNXY are commercially available or can be synthesized according to methods commonly known those skilled in the E. Mϋller (Ed.), "Methoden der Organischen Chemie" [Methods of Organic Chemistry] (Houben-Weyl), Vol. 11/1 Stickstoff-Verbindungen, Georg Thieme Nerlag, Stuttgart 1957).
The reaction between the compounds (N) and HΝXY is preferably carried out in a solvent.
Suitable solvents comprise the customary organic solvents which are inert under the reaction conditions. Non-limiting examples include ethers such as diethyl ether, dioxane, tetrahydrofuran, 1,2-dimethoxy ethane; hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions; halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, dichloroethane, trichloroethylene, chlorobenzene; ketones such as acetone; esters such as ethyl acetate; nitriles such as acetonitrile; heteroaromatics such as pyridine; polar solvents such as dimethyl formamide and hexamethyl phosphoric acid tris-amide; and mixtures of the above-mentioned solvents. Dichloromethane and tetrahydrofuran are preferred.
The compounds HNXY are generally employed in an amount of from 1 to 4 mol per mol of compounds (V); an equimolar amount or slight excess of compounds HNXY is preferred.
The reaction between the compounds (V) and HNXY is preferably carried out in the presence of a coupling reagent by methods commonly known to those skilled in the art (see e.g. March, Advanced Organic Chemistry, 4th ed., pp. 419-421 Wiley 1992). Non-limiting examples include dicyclohexylcarbodiimide, l-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride, N,N'-carbonyldiimidazole, pivaloyl chloride, bis(2-oxo-3- oxazolidinyl)phosphinic chloride and benzotriazol-l-yloxytris(pyrrolidino)- phosphonium hexafluorophosphate. The coupling reagents may optionally be bound to a polymeric resin. The coupling reagent is generally employed in an amount of from 1 to 4 mol per mol of compounds (V); an equimolar amount or slight excess of the reagent is preferred.
Additionally, a promoter may be added to the coupling reaction. Non-limiting examples include 1-hydroxybenzotriazole, N,N'-dimethylaminopyridine, and 3-hydroxy-3H-l,2,3- triazolo[4,5-b]-pyridine. The promoter is generally employed in an amount from 0.1 to 1 mol per mol of compounds (V), preferably from 0.3 to 1 mol, per mol of compounds (V).
The reaction of the compounds (V) and HNXY can generally be carried out within a relatively wide temperature range, hi general, the reaction is carried out within a range of from -20 to 200°C, preferably from 0 to 70°C, and generally at room temperature. Compounds (LX) and (XI) are commercially available or can be synthesized according to methods commonly known to those skilled in the art (H. Feuer (ed.) "The Chemistry of the Nitro and Nitroso Groups" Interscience Publishers, New York, 1969, pp. 76-117).
Compounds (X) are commercially available or can be synthesized by coupling compounds
HNXY with compounds (II) in the same manner as described above for the coupling of compounds (V) with compounds HNXY.
Compounds (I) wherein R3 is hydrogen can be synthesized by General Method A or B using compounds (III) or (V) respectively wherein R3 is hydrogen.
Optionally, the compounds obtained through General Method A or B can be converted into an isomer, a pharmaceutically acceptable salt, a hydrate or a hydrate of a pharmaceutically acceptable salt thereof
The compounds of the present invention are inhibitors of phosphodiesterase 10a (PDE 10a). As outlined above, the inhibition of PDE 10a is a promising approach for the treatment of cancer. The biological tests described below show that the compounds (I) exhibit a pronounced anti-proliferation activity against tumor cells; they are therefore useful for the treatment of cancer. Furthermore, our investigations showed that they are also useful for treatment of conditions of pain and/or for the lowering of the temperature of the body in fever conditions.
The compounds according to the invention can be used as active ingredients for the production of medicaments against carcinomatous disorders. For this, they can be converted into the customary formulations such as tablets, coated tablets, aerosols, pills, granules, syrups, emulsions, suspensions and solutions using inert, non-toxic, pharmaceutically suitable excipients or solvents. Preferably, the compounds according to the invention are used in an amount such that their concentration is approximately 0.5 to approximately 90% by weight, based on the ready-to-use formulations, the concentration being dependent, inter alia, on the indication of the medicament. The formulations can be produced, for example, by extending the active compounds with solvents and/or excipients having the above properties, where, if appropriate, additionally emulsifiers or dispersants and, in the case of water as the solvent, an organic solvent can additionally be added.
Administration can be carried out in a customary manner, preferably orally, transdermally or parenterally, for example perlingually, buccally, interperitoneally, intravenously, nasally, rectally or ihhalationally.
For human use, in the case of oral administration, it is recommended to administer doses of from 0.001 to 50 mg/kg, preferably from 0.01 to 20 mg kg. In the case of parenteral administration such as, for example, intravenously or via mucous membranes nasally, buccally or inhalationally, it is recommended to use doses of from 0.001 to 0.5 mg/kg.
If appropriate, it may be necessary to depart from the amounts mentioned above, namely depending on the body weight or the type of administration route, on the individual response towards the medicament, the manner of its formulation and the time or interval at which administration takes place. Thus, in some cases it may be sufficient to manage with less than the above mentioned minimum amount, while in other cases the upper limit mentioned must be exceeded. In the case of the administration of relatively large amounts, it may be recommended to divide these into several individual doses over the course of the day.
The compounds according to the invention are also suitable for use in veterinary medicine. For use in veterinary medicine, the compounds or their non-toxic salts can be administered in a suitable formulation in accordance with general veterinary practice. Depending on the kind of animal to be treated, the veterinary surgeon can determine the nature of use and the dosage.
The present invention provides compounds for the use in a medical application, in particular for combating cancer. The invention further provides a method of manufacturing a pharmaceutical composition by combining at least one of the compounds of the invention with at least one pharmacologically acceptable formulating agent.
The invention further provides a pharmaceutical composition comprising as an active ingredient an effective amount of at least one of the compounds of the invention and at least one pharmacologically acceptable formulating agent.
The invention further provides a pharmaceutical composition comprising as an active ingredient an effective amount of at least one of the compounds of the invention and at least one pharmaceutical active ingredient which is different from the compounds of the invention.
The invention further provides a medicament in dosage unit form comprising an effective amount of a compound according to the invention together with an inert pharmaceutical carrier.
The invention further provides a method of combating cancer in mammals comprising administering to a mammal in need thereof an antiproliferative effective amount of at least one compound according to the invention either alone or in admixture with a diluent or in the form of a medicament.
The percentages in the description above, in the following tests and in the Examples are - if not stated otherwise - percentages by weight; parts are parts by weight. Solvent ratios, dilution ratios and concentrations in solutions of liquids in liquids are ratios by volume. Examples
Abbreviations used in this specification
BSA bovine serum albumin
™Cremophor® non-ionic emulsifier from BASF, Germany DBU l,8-diazabicyclo[5.4.0]undec-7-ene DMEM Dulbecco's Modified Eagle Medium, Life Technologies,
Gaithersburg, MD, U.S.A.
DMF N,N-dimethyl formamide
DMSO dimethyl sulphoxide
EDCI l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
EDTA ethylene diamine tetraacetate
FBS fetal bovine serum
HEPES N-(2-hydroxyethyl)-piperazine-N'-(2-ethane sulphonic acid)
HPLC high pressure liquid chromatography
LC-MS liquid chromatography - coupled mass spectroscopy
LC RT liquid chromatography retention time
LDH lactate dehydrogenase
MP melting point
NMR nuclear resonance spectroscopy
PBS phosphate-buffered saline
PyBOP Bromotripyrrolidinophosphonium hexafluorophosphate
TLC thin layer chromatography
Tris/hydrochloric acid tris(hydroxymethyl)-aminomethane hydrochloride
™Triton X-100® tert.-octyl-phenoxypolyethoxyethanol, Rohm& Haas, USA The yield percentages of the following examples refer to the starting component which was used in the lowest molar amount.
LC-MS / HPLC methods:
Method A:
MS equipment: Finnigan LCQ Ion Trap Mass Spectrometer
ionisation mode: ESI
HPLC equipment: HP 1100
UN detection: 254 nm
Column: YMC pro C-18
23 mm x 2 mm 120 A
Supplier: YMC
Gradient: Time A: % B: % Flow
[min.] [mL/min.]
0.50 90.0 10.0 1.0
3.50 5.0 95.0 1.0
4.00 5.0 95.0 1.0
4.01 90.0 10.0 1.0
6.50 90.0 10.0 1.0 A: 0.02 % strength solution of trifluoroacetic acid in 2 % acetonitrile / 98 % water
B: 0.018 % strength solution of trifluoroacetic acid in 98 % acetonitrile / 2 % water
Method B:
MS equipment: Micromass LCZ
ionisation mode: ESI
HPLC equipment: Gilson 215
UN detection: 254 nm
Column: YMC pro C-18
23 mm x 2 mm 120 A
Supplier: YMC
Gradient: Time A: % B: % Flow
[min.] mL/min.]
0.50 90.0 10.0
3.50 5.0 95.0
4.00 5.0 95.0
4.01 90.0 10.0
4.80 90.0 10.0 A: 0.02 % strength solution of trifluoroacetic acid in 2 % acetonitrile / 98 % water
B: 0.02 % strength solution of trifluoroacetic acid in 98 % acetonitrile / 2 % water
Method C: Column: Kromasil RP-18
60 mm x 2.0 mm 3.5 μm
Gradient: Time A: % B: % Flow
[min.] [mL/min.]
0.00 98.0 2.0 0.75
4.50 10.0 90.0 0.75
6.50 10.0 90.0 0.75
0.5% strength aqueous HClO
B: acetonitrile
Method D:
MS equipment: Micromass Platform LCZ
ionisation mode: ESI positive / negative
HPLC equipment: HP 1100
UN detection: 208-400 nm
temperature: 40 °C
Column: Symmetry C 18
50 mm x 2.1 mm 3.5 μm
Supplier: Waters
Gradient: Time A: % B: % Flow
[min.] [mL/min.]
0.00 90.0 10.0 0.50
4.00 10.0 90.0 0.50
6.00 10.0 90.0 0.50
A: 0.05%) strength solution of formic acid in water
B: 0.05% strength formic acid in acetonitrile
Method E:
MS equipment: Finnigan MAT 900S
ionization mode: ESI - positive
HPLC equipment: Thermo Separation Products P4000, AS3000, UN3000HR
UN detection: 210 mn
temperature: 70 °C
Column: ™Symmetry C 18
50 mm x 2.1 mm 3.5 μm
Supplier: Waters
Gradient: Time A: % B: % C: % Flow
[min.] [mL/min.]
0.00 2.0 49.0 49.0 0.9
2.50 95.0 2.5 2.5 1.2
5.00 95.0 2.5 2.5 1.2
5.50 2.0 49.0 49.0 1.2
6.50 2.0 49.0 49.0 1.2
7.00 2.0 49.0 49.0 0.9
acetonitrile
B: 0.01% HC1 in water
C: water
Method F:
MS equipment: Micromass Quattro LCZ
ionisation mode: ESI positive / negative HPLC equipment: HP 1100
UN detection: 208-400 nm
temperature: 40 °C
Column: ™Symmetry C 18
50 mm x 2.1 mm 3.5 μm
Supplier: Waters
Gradient: Time A: % B: % Flow
[min.] [mL/min.]
0.00 90.0 10.0 0.50
4.00 10.0 90.0 0.50
6.00 10.0 90.0 0.50
A: 0.05% strength solution of formic acid in water
B: 0.05%) strength formic acid in acetonitrile
Method G:
HPLC Equipment: Gilson 215
UN Detection: 220 and 254 nM
Temperature: 25 °C
Column: ™YMC-Pack Pro C18
50 mm x 4.6 mm 5μM
Supplier: Waters Gradient: Time A: % B: % Flow
[min.] [mL/min]
0.00 10.0 90.0 4.00
3.50 90.0 10.0 4.00
4.50 90.0 10.0 4.00
4.60 10.0 90.0 4.00
5.00 10.0 90.0 4.00
A: 0.1%) strength solution of TF A in acetonitrile
B : 0.1%) strength aqueous TF A
Starting Materials
For amines not commercially available the specific preparations are exemplified below:
N-(2-Aminoethyl)-N-(2-pyridinyl)amine
Figure imgf000045_0001
A solution of 2-bromopyridine (1.0 g, 6.3 mmol) and ethylenediamine (2.0 mL, 29.8 mmol) was heated to 120 °C for 18 h. The reaction mixture was diluted with dichloromethane and washed with water and brine. The organic layer was dried over sodium sulfate and concentrated in vacuo to give an oil (775 mg, 90%>): MS (HPLC/ES): m/z = 138.1 (M + 1); LCMS RT (method A): 0.75 min. (6-Methoxy-3-pyridinyl)methanamine dihydrochloride
Figure imgf000046_0001
2-Methyoxypyridine-5-carbonitrile (2.0 g, 14.9 mmol) was added to 10%> Pd/C (200 mg,
10%) w/w), ethanol (lOOmL) and cone, hydrochloric acid (3 mL) in a 500 ml bottle under argon and subjected to 58 psi on a Parr Shaker for 40 h. The reaction mixture was filtered through a pad of Celite® and washed with ethanol. The solution was concentrated in vacuo to give a solid: MS (HPLC/ES): m/z = 138.9 (M + 1); LCMS RT (method A): 0.72 min. The amine was carried on to amide coupling conditions without further purification.
2-(lH-Pyrazol-l-yl)ethylamine
Figure imgf000046_0002
To a solution of 1.50 g (22.0 mmol) lH-pyrazole in 50 mL of acetonitrile was added 3.35 g (83.7 mmol) of powdered sodium hydroxide and 299 mg (0.88 mmol) of tetrabutylammoniumsulfate. The reaction was stirred for 30 min and 3.07 g (26.4 mmol) of 2-chloroethylamine hydrochloride was added. This was stirred at room temperature for 3 d, at which time TLC analysis (silica gel 60, 90:10 dichloromethane/methanol, iodine staining) suggested complete reaction. The mixture was filtered and the filtrate was concentrated in vacuo. The residue was cooled to 0 °C and 15 mL of 40 %> strength hydrobromic acid was added slowly. An equal portion of ethanol was then added and the biphasic solution was heated to form only one phase. The resulting solution was cooled to 0 °C and slowly triturated with ether to form 4.21 g (21.9 mmol, 99 %) of 2-(lH-ρyrazol- l-yl)ethylamine as a white solid: 1H-NMR (DMSO- 6) δ 3.24 (sextet, J= 6.0, 2H), 4.40 (t, J= 5.9, 2H), 6.00 (br s, 3H), 6.28 (t, J= 2.1, IH), 7.50 (d, J= 1.7, IH), 7.77 (d, J= 2.3, IH), 7.92 (br s, 2H); MS (HPLC/ES) m/z = 112.1 (M + 1); LCMS RT(method B): 0.79 min.
3-(lH-Pyrazol-l-yl)-propylamine
Figure imgf000047_0001
This compound was prepared in the manner shown for 2-(lH-pyrazol-l-yl)ethylamine above.
1.50 g (7.28 mmol, 99 %) of 2-(lH-ρyrazol-l-yl)propylamine as a white solid: XH-NMR (DMSO-d6) δ 2.02 (m, 2H), 2.73 (m, 2H), 4.19 (t, J= 6.4, 2H), 6.23 (t, J= 2.2, IH), 7.44 (dd, J = 0.7, 1.9, IH), 7.72 (m, 4H); MS (HPLC/ES) m/z = 126.1 (M + 1); LCMS RT
(method B): 0.80 min.
cis-2-{[tert-Butyl-(dimethyl)-silyl]-oxy}cyclohexylamine
Figure imgf000047_0002
To a suspension of cis-2-aminocyclobexanol (1.00 g, 8.68 mmol) in dichloromethane (6 mL) at 0 °C was added 2,6-lutidine (1.86 g, 17.36 mmol) and t-butyldimethylsilyltriflate (3.44 g, 13.0 mmol) dropwise. The mixture was stirred overnight and concentrated in vacuo. The residue was diluted with ether, washed with water, and dried over magnesium sulfate. The solvent was removed in vacuo to give 2.0 g (100%) of a white solid.
Resolution of l-(4-fluorophenyl)ethylamine. (lR)-l-(4-fluorophenyI)-ethylamine and (1 S)-l -(4-fluorophenyl)-ethylamine
Figure imgf000048_0001
Racemic l-(4-fluorophenyl)-ethylamine (commercially available) was resolved by (+)- tartaric acid following the procedures of A. Ault (Org. Synth., 1973, Coll. Vol. 5, 932) and S. Takenaka et al. (J.C.S. Perkin II, 1978, 95).
3-[4-(CyclohexyϊmethyI)-piperazine-l-carbonyl]-benzaldehyde
Figure imgf000048_0002
To a solution of 3-carboxybenzaldehyde (500 mg, 3.33 mmol) in tetrahydrofuran (10 mL) was added PyBOP(2.25 g, 4.33 mmol) and N,N-diisopropylethylamine (750 μL, 4.33 mmol). The reaction mixture was stirred for 20 min, after which 1- cyclohexylmethylpiperizine (789 mg, 4.33 mmol) was added. The reaction mixture was stirred for 18h, at which time TLC analysis (silica gel 60, 50:50 ethyl acetate/hexanes, UN detection) suggested almost complete conversion to the amide. The reaction was concentrated in vacuo, and the resulting crude product was purified by using the Biotage Quad4 (25 M column) eluting with 35:65 ethyl acetate/hexanes. The desired fractions were collected and concenfrated in vacuo to afford the title compound as a yellow oil which turned pink upon standing (563 mg, 1.79 mmol, 54%): 1H-ΝMR (CDC13) δ 10.02 (s, IH), 7.91 (m, 2H), 7.66 (m, IH), 7.26 (m, IH), 3.81 (br. s., 2H), 3.42 (br. s., 2H), 2.50 (br. s., 2H), 2.36 (br. s., 2H), 2.17 (d, J= 6.8 Hz, 2H), 1.73 (m, 6H), 1.51 (m, IH), 1.21 (m, 2H), 0.86 m, 2H).
3-Formyl-N-cyclopropyl-benzanιide
Figure imgf000049_0001
To a solution of 3-carboxybenzaldehyde (500 mg, 3.33 mmol) in tetrahydrofuran (5 mL) was added PyBOP (2.25 g, 4.33 mmol) and N,N-diisopropylethylamine (750 μL, 4.33 mmol). The reaction mixture was stirred for 20 min, after which cyclopropylamine (300 μL, 4.33 mmol) was added. The reaction mixture was stirred for 18h, at which time TLC analysis (silica gel 60, 50:50 ethyl acetate/hexanes, UN detection) suggested complete consumption of the starting material. The reaction mixture was concentrated in vacuo and purified via column chromatography eluting with 2:1 - 3:1 ethyl acetate/hexanes, which afforded the title compound as a white solid (534 mg, 2.82 mmol, 85%): 1H-ΝMR (DMSO, d6) δ 10.03 (s, IH), 8.65 (m, 1H),8.31 (m, IH), 8.10 (m, IH), 8.02 (m, IH), 7.66 (t, J = 7.6 Hz, IH), 2.87 (m, IH), 0.71 (m, 2H), 0.59 (m, 2H); HPLC RT (Method G): 1.20 min. 3-formyl-N-(2,4-difluorobenzyl)-benzamide
Figure imgf000050_0001
To a solution of 3-carboxybenzaldehyde (500 mg, 3.33 mmol) in tetrahydrofuran (10 mL) was added PyBOP (2.25 g, 4.33 mmol) and N,N-diisopropylethylamine (750 μL, 4.33 mmol). The reaction mixture was stirred for 20 min, after which 2,4-difluorobenzylamine (510 μL, 4.33 mmol) was added. The reactoin mixture was stirred for 18h, at which time TLC analysis (silica gel 60, 50:50 ethyl acetate/hexanes, UN detection) suggested complete consumption of the starting material. The reaction mixture was concentrated in vacuo and purified via prep HPLC eluting with 40-90%> acetonitrile/water over 3.5 minutes to afford 733 mg (2.66 mmol, 80%) of RDB 121-1 as a white solid: 1H-ΝMR (DMSO, d6) δ 10.05 (s, IH), 9.25 (t, J= 5.7 Hz, IH), 8.39 (m, IH), 8.17 (m, IH), 8.06 (dt, J= 7.6, 1.3 Hz, IH), 7.70 (t, J= 1.6 Hz, IH), 7.43 (m, IH), 7.23 (m, IH), 7.05 (m, IH), 4.49 (d, J = 5.5 Hz, 2H).
Intermediates
1.1. Ethyl 2-{N-[2-(3,4-dimethoxyphenyl)-ethyl]-carbamoyl} acetate
H
Figure imgf000050_0002
A solution of 50.0 g (275.9 mmol) of 3,4-dimethoxyphenethylamine in 500 mL of dichloromethane was treated with 42.0 g (275.9 mmol) of l,8-diazabicyclo[5.4.0]undec-7- ene, followed by dropwise addition of 35.0 mL (41.6 g, 276.4 mmol) of ethyl malonyl chloride at a rate that kept the internal temperature below 30 °C. The resultant clear yellow solution was stirred at room temperature under an argon atmosphere for 16 h, at which time TLC analysis (silica gel 60, 5:95 methanol/dichloromethane, UN detection) suggested complete reaction. The organics were washed with brine (3 X 1000 mL), dried over sodium sulfate and concentrated in vacuo. The residue was dried under high vacuum at 30 °C for 24 h to provide 80.6 g (272.8 mmol, 99%) of a yellow oil: 1H-ΝMR (DMSO- d_): δ = 1.16, 1.18 (t, mixture of rotamers; J= 7.0, 3H), 2.63 (t, J= 1.1, 2H), 3.18 (s, 2H,),
3.25 (m, 2H), 3.70 (s, 3H), 3.73 (s, 3H), 4.05 (q, J= 7.0, 2H), 6.69 (dd, J= 2.2, 8.4, IH), 6.79 (d, J= 2.2, IH), 6.83 (d, J= 8.4, IH), 8.1 (br t, IH, J= 5.4); MS (HPLC/ES): m/z = 296 (M + 1); TLC (10:90 methanol/dichloromethane, UN detection): R/= 0.70.
1.2. Ethyl (6,7-dimethoxy-3,4-dihydro-l(2H)-isoquinolinylidene)-ethanoate
Figure imgf000051_0001
To a refluxing solution of methanesulfonic anhydride (648.83 g, 3.72 mol) in toluene (4 L) was added Intermediate 1.1, ethyl 2-{Ν-[2-(3,4-dimethoxyphenyl)ethyl]-carbamoyl}- acetate, (1000 g, 3.39 mol) portionwise over 20 minutes. The reaction was stirred at reflux for 30 minutes at which point the heat was removed and the toluene was decanted. The resulting dark oil was then dissolved in water (3000 mL) and treated portionwise with solid potassium carbonate until a pH of about 8 was achieved. The organic material was extracted from the dark biphasic mixture using ethyl acetate (3000 mL). The combined organic extracts were washed with brine (3 x 2000 mL) and concentrated to 1/3 volume. The resultant dark oil was placed on a pad of silica gel 60 (400 cc) and eluted using ethyl acetate/hexane (1:1). The desired fractions were concentrated to a yellow oil which was seeded with a small amount of crystals of the title compound and placed in a refrigerator overnight. The yellow crystalline solid which formed was filtered, washed with ethyl acetate/ hexane (1:1) (2 x 50 ml), and vacuum dried for 12 hours to give 533.26 g of the desired product . The filtrate was concentrated to a dark oil and seeded a second time. After 1 hour, the newly formed yellow solid was filtered, washed with ethyl acetate/ hexane (1:1) (2 x 50 ml), and vacuum dried for 12 hours to provide 106.23 g of a second crop . The two batches of crystals were combined to provide the title compound (639.49 g, 68 %). 1H-NMR (OMSO-d6): δ 1.18 (t, J = 7.0 Hz, 3H); 2.76 (t, J = 6.5 Hz, 2); 3.36
(m, 2H); 3.78 (s, 6H); 4.02 (q, J = 7.0 Hz, 2H); 5.05 (s, IH); 6.87 (s, IH); 7.15 (s, IH); 8.95 (bs, IH). MS (HPLC/ES; method A): m/z = 278 (M + 1). TLC [ethyl acetate/hexane (1:1)]: R/ = 0.63
Instead of methanesulfonic anhydride also phosphorous pentoxide can be used according to this method.
1.3. Ethyl 2-(3-carboxyphenyl)-8,9-dimethoxy-3-methyl-5,6-dihydro-pyrrolo [2,1- a]isoquinoline-l-carboxylate
Figure imgf000052_0001
5.18 mL (72.1 mmol) of nitroethane, 8.56 mL (86.5 mmol) of piperidine, and 10.8 g (72.1 mmol) of 3-carboxy-benzaldehyde were added to a solution of 10 g (36.06 mmol) of ethyl 2-(6,7-dimethoxy-2,3,4-trihydro-isoquinolylidene)-acetate (Intermediate 1.2.) in 300 mL of isopropanol/ ethanol (1:1 v/v). The solution was heated at reflux for 16 h. Subsequently, the mixture was cooled to room temperature, and the volatiles were removed in vacuo. The resultant solid was suspended in 150 mL of 1 N hydrochloric acid, and the aqueous mixture was extracted with trichloromethane (3 X 75 mL). The combined extracts were dried over sodium sulfate, filtered, and concentrated in vacuo to afford the crude product as an orange solid, which was purified by trituration with ether (14 g, 89 %): 1H-NMR (DMSO- 6): δ = 0.81 (t, J= 6.9, 3H), 2.13 (s, 3H), 2.95 (t, J= 6.5, 2H), 3.72 (s, 3H) 3.78 (s, 3H), 3.90-3.96 (m, 4H), 6.93 (s, IH), 7.40-7.49 (m, 2H), 7.72 (d, J= 1.3, 2H), 7.81 ( , IH); MS (HPLC/ES): m/z = 436 (M + 1).
2.1. 5-[l-(ethoxycarbonyl)-8,9-dimethoxy-3-metkyl-5,6-dihydropyrrolo[2,l-a]iso- quinolin-2-yl]-2-hydroxybenzoic acid
Figure imgf000053_0001
A suspension of ethyl 2-(6,7-dimethoxy-2,3,4-trihydro-isoquinolylidene)-acetate (11.67 g, 42.08 mmol, Intermediate 1.2.) and 5-formylsalicylic acid (14.0 g, 84.27 mmol) in dry ethanol (175 mL) and 2-propanol (175 mL) was treated with nitroethane (6.37 g, 84.85 mmol) and piperidine (8.62 g, 101.23 mmol). The contents were heated at reflux under argon for 12 h, at which time TLC analysis (silica gel 60, 10% methanol/dichloromethane, UN detection) suggested complete reaction. The contents were concentrated in vacuo and the dark residue was dissolved in dichloromethane (500 mL). The organics were washed with 1.0 Ν aqueous hydrochloric acid (150 mL) in brine (200 mL) and the layers were separated. Concentration of the organic phase to ~150 mL effected precipitation of a yellow solid. The crude material was filtered, dissolved in hot 10% methanol/ethyl acetate (300 mL), and allowed to cool to room temperature. The contents were further cooled to 3 °C for 2 h, and the resultant precipitate was filtered and dried under high vacuum at 40 °C for 3.5 d to afford the product (10.68 g, 23.66 mmol, 56%) as a pale-yellow solid: 1H- ΝMR (DMSO- ): δ 0.89 (t, J = 7.2, 3H), 2.11 (s, 3H), 2.94 (br t, J = 6.0, 2H), 3.72 (s,
3H), 3.78 (s, 3H), 3.93 (m, 2H), 3.95 (q, J= 7.2, 2H), 6.94 (s, IH), 6.95 (d, J= 8.6, IH), 7.33 (dd, J = 2.0, 8.6, IH), 7.58 (d, J= 2.0, IH), 7.68 (s, IH), absorptions for the phenol and carboxylic acid moieties were not observed; EA: calcd for C 5H25NO : C, 66.51; H, 5.58; N, 3.10. Found: C, 66.25; H, 5.70; N, 3.10; MS (HPLC/ES): m/z = 452 (M + 1); TLC (silica gel 60, 15%o methanol/dichloromethane, UN detection): one spot, R/= 0.56.
3.1. 3-Bromo-2-fluorobenzaldehyde
Figure imgf000054_0001
LTMP (lithium tetramethylpiperidine) was prepared by adding butyllithium (37.14 mmol, 14.86 mL) slowly to a solution of 2,2,6,6-tetramethylpiperidine (42.86 mmol, 7.23 mL) in tetrahydrofuran (15 mL) at 0 °C. The mixture was stirred at 0 °C for 1 h. This solution was added slowly to a solution of 1,2-bromofluorobenzene (5.0 g, 28.57 mmol, 3.12 mL) in tetrahydrofuran (90 mL) at - 75 °C. The mixture was allowed to stir at this temperature for 2 h, then N,N-dimethylformamide (142.86 mmol, 11.05 mL) was added dropwise. After 1 h, the reaction was quenched with water and was concentrated in vacuo. The residue was partitioned between ether and water. The aqueous layer was extracted with ether twice and the combined organic layers were washed with brine, dried over anhydrous magnesium sulfate and concentrated in vacuo. The crude residue was dissolved in dichloromethane and passed through a pad of silica eluting with 100:0 to 95:5 hexanes/ethyl acetate to obtain a colorless oil (4.32 g, 70.8 %): 1H-ΝMR (DMSO- 6): δ =
7.34 (m, IH), 7.82 (m, IH), 8.03 (m, IH), 10.15 (s, IH); TLC (10:90 ethyl acetate/hexanes): R/= 0.70. 3.2. Ethyl 2-(3-bromo-2-fluorophenyl)-8,9-dimetkoxy-3-methyl-5,6- dihydropyrrolo[2,l-a]isoquinoline-l-carboxylate
Figure imgf000055_0001
To a mixture of 3-bromo-2-fluorobenzaldehyde (4.2 g, 20.7 mmol, Intermediate 3.1.) was added ethyl 2-(6,7-dimethoxy-2,3,4-trihydro-isoquinolylidene)-acetate (5.76 g, 20.7 mmol, Intermediate 1.2.), nitroethane (41.5 mmol, 2.98 mL) and piperidine (49.8 mmol, 4.93 mL) in 1:1 ratio of isopropanol/ethanol (100 mL) and was heated to 80 °C overnight. The reaction was concentrated in vacuo to a minimum volume and treated with methanol (40 mL) until precipitation occurred. The resulting residue was filtered, washed with methanol and concentrated in vacuo to give the title compound as a pale pink solid (5.03 g, 49.2 %>): 1H-NMR (CD2C12): δ = 0.94 (t, J= 5.4, 3H), 2.15 (s, 3H), 3.02 (t, J = 5.0, 2H), 3.89 (s, 6H), 3.98 (t, J= 5.0, 2H), 4.03 (m, 2H), 6.77 (s, IH), 7.05 (m, IH), 7.18 (m, IH), 7.43 (m, IH), 8.03 (s, IH).
3.3. 3-[l-(Ethoxycarbonyl)-8,9-dimethoxy-3-methyI-5,6-dihydropyrrolo[2,l- a]isoquinolin-2-yl]-2-fluorobenzoic acid
Figure imgf000055_0002
A solution of ethyl 2-(3-bromo-2-fluorophenyl)-8,9-dimethoxy-3-methyl-5,6- dihydropyrrolo[2,l-a]isoquinoline-l-carboxylate (3.00 g, 6.14 mmol, Intermediate 3.2.) in tetrahydrofuran (60 mL) was cooled to - 78 °C and degassed for 5 min. BuLi (9.23 mmol, 3.69 mL) was added dropwise. The mixture was stirred at - 78 °C for 1 h and CO2 was bubbled into the solution over a period of 2 h. The reaction was quenched with methanol and concentrated in vacuo. The crude residue was purified with 40 M Biotage by eluting with 10:90 methanol/ethyl acetate to obtain desired product as a yellow solid (2.00 g, 71.8 %): 1H-NMR (CD2C12): δ = 0.92 (t, J= 7.0, 3H), 2.14 (s, 3H), 3.03 (t, J= 6.6, 2H), 3.88 (s, 6H), 3.99 (m, 2H), 4.09 (m, 2H), 6.77 (s, IH), 7.27 (t, J= 7.6, IH), 7.49 (t, IH), 7.95 (m, IH), 8.03 (s, IH).
4.1. 2-Fluoro-5-formyl-benzoic acid
Figure imgf000056_0001
A mixture of 2-fluoro-5-formylbenzonitrile (30.0 g, 201.2 mmol) and concentrated hydrochloric acid (500.0 mL) was heated at reflux under argon for 16 h. Upon cooling to room temperature, a white precipitate was formed. The solid was separated and washed with water (2 X 1500 mL) and dissolved in ethyl acetate (2000 mL). The solution was washed with brine, dried over magnesium sulfate and concentrated in vacuo to afford 25.8 g of the title compound as a white solid (153.6 mmol, 76.3 %): 1H-NMR (OMSO-d6): δ = 7.54 (dd, J= 8.5, 10.6, IH), 8.15 (m, IH), 8.41 (dd, J= 2.4, 7.2, IH), 1O.01 (s, IH), 13.63 (br s, IH); EA: calcd for C8H5FO3: C, 57.15; H, 3.00; F, 11.30. Found: C, 57.00; H, 2.76; F, 11.36.
4.2. 5-[l-(Ethoxycarbonyl)-8,9-dimethoxy-3-methyl-5,6-dihydropyrrolo[2,l- a]isoquinolin-2-yl]-2-fluorobenzoic acid
Figure imgf000057_0001
A mixture of 2-fluoro-5-formylbenzoic acid (15.0 g, 89.2 mmol, Intermediate 4.1.), ethyl 2-(6,7-dimethoxy-2,3,4-trihydro-isoquinolylidene)-acetate (12.4 g, 44.6 mmol, Intermediate 1.2.), nitroethane (6.70 g, 89.2 mmol), and piperidine (7.60 g, 89.2 mmol) in a 1 : 1 mixture of ethanol/isopropanol (500mL) was heated at reflux for 5 h at which time
TLC analysis (silica gel 60, 10:90 methanol/dichloromethane, UN detection) suggested complete reaction. The mixture was cooled to 0 °C and washed with concentrated hydrochloric acid (300 mL). The mixture was diluted with water and extracted with dichloromethane (3 X 300 mL). The organic layers were combined, washed with brine, dried over sodium sulfate and concentrated in vacuo. The residue was passed through a pad of silica gel eluting with 2:98 methanol/dichloromethane. Further purification by recrysatUization from ethyl acetate/hexanes provided 8.25 g of the title compound as a light yellow solid (18.2 mmol, 40.2 %): 1H-ΝMR (DMSO- 6): δ = 0.86 (t, J= 5.4, 3H), 2.12 (s, 3H), 2.95 (t, J= 4.5, 2H), 3.72 (s, 3H), 3.78 (s, 3H), 3.96 (m, 4H), 6.93 (s, IH), 7.26 (dd, J = 6.1, 8.0, IH), 7.42 (m, IH), 7.60 (dd, J = 1.8, 5.4, IH), 7.72 (s, IH); MS
(HPLC/ES): m/z = 454.0 (M + 1); LCMS RT (method A): 3.07 min.
5.1. 2-(Acetyloxy)-3-methylbenzoic acid
Figure imgf000057_0002
To a solution of 3-methylsalicylic acid (20.0 g, 131.5 mmol) in anhydrous pyridine (42.5 mL, 525.8 mmol) was added acetic anhydride (49.6 mL, 525.5 mmol). The reaction was stirred at room temperature under an argon atmosphere for 16 h. The solution was poured into ice-water and extracted with dichloromethane (3 X 200 mL). The organic extracts were washed with aqueous IN hydrochloric acid (5 X 80 mL) and brine (2 X 100 mL). The organic layer was dried over magnesium sulfate and concentrated in vacuo to give a white solid (20.67 g, 81 1%): 1H-NMR (DMSO- ): δ 2.16, (s, IH), 2.27 (s, 3H), 7.25 (t, J= 1.6, IH), 7.51 (d, J= 7.3, IH), 7.73 (d, J= 1.1, IH), 12.95 (s, IH).
5.2. 3-Formylsalicylic acid
Figure imgf000058_0001
A suspension of 2-(acetyloxy)-3-methylbenzoic acid (13.35 g, 68.8 mmol, Intermediate 5.1.) in carbon tetrachloride (200 mL) was stirred under an argon atmosphere. A 500-watt tungsten lamp was positioned 2 inches directly in front of the reaction. The solution was gently heated with the lamp while a solution of bromine (7.08 mL, 137.5 mmol) in carbon tetrachloride (100 mL) was added dropwise, so that a red color persisted at all times. After the addition was complete, the reaction was allowed to stir an additional 2 h under the light. The reaction was allowed to cool to room temperature and the mixture was washed several times with a saturated aqueous Na2S2O3 solution (3 X 150 mL). The organic layer was dried over sodium sulfate and concentrated in vacuo. The crude residue was purified by flash chromatography (0:100 to 5:95 methanol/dichloromethane) followed by crystallization (ethyl acetate/hexanes) to give a white solid which was then suspended in 8%o aqueous sodium carbonate and stirred, heating to reflux gradually (60° C for 16 h, 85°
C for 22 h, reflux 24 h), for 3 d. The mixture was cooled to room temperature and made acidic with the addition of IN hydrochloric acid. The resulting solid was collected by filtration and dried under high vacuum for 14 h to provide the title compound (7.21 g, 63%): 1H-NMR (DMSO- ): δ 7.05 (t, J= 7.8, IH), 7.91 (dd, J= 1.8, 7.8, IH), 8.09 (dd, J= 1.8, 7.7, IH), 10.35 (s, IH). 5.3. 3-[l-(Ethoxycarbonyl)-8,9-dimethoxy-3-methyl-5,6-dihydropyrrolo[2,l- a] isoquinolin-2-yl] -2-hydroxybenzoic acid
Figure imgf000059_0001
To a solution of ethyl 2-(6,7-dimethoxy-2,3,4-trihydro-isoquinolylidene)-acetate (14.44 g, 52.1 mmol, Intermediate 1.2.) in ethanol (40 mL) was added 3-formylsalicylic acid (7.21 g, 43.4 mmol, Intermediate 5.2.), nitroethane (3.74 mL, 52.1 mmol), and piperidine (5.15 mL, 52.1 mmol). The solution was heated at reflux under an argon atmosphere for 72 h. The reaction was allowed to cool to room temperature and the volatiles were removed in vacuo. The crude material was purified by silica gel flash chromatography (eluant 0:100 to 10:90 methanol/dichloromethane) and concentrated in vacuo to afford the product as an orange solid (9.7 g, 50 %): 1H-NMR (OMSO-d6): δ 0.77 (t, J= 7.0, 3H), 2.07 (s, 3H), 2.98 - 3.03 (m, 2H), 3.71 (s, 3H), 3.78 (s, 3H), 3.86 (q, J= 7.1, 2H), 3.93 (t, J= 6.2, 2H), 6.73 (t, J= 7.5, IH), 6.92 (s, IH), 7.13 (d, J= 7.5, IH), 7.66 (d, J= 7.5, IH), 7.87 (s, IH),
8.43 (br s, IH).
5.4. Ethyl 8,9-dimethoxy-2-[2-methoxy-3-(methoxycarbonyl)phenyl]-3-methyl-5,6- dihydropyrrolo[2,l-a]isoquinoline-l-carboxylate
Figure imgf000059_0002
To a solution of 3-[l-(ethoxycarbonyl)-8,9-dimethoxy-3-methyl-5,6-dihydropyrrolo[2,l- a]isoquinolin-2-yl]-2-hydroxybenzoic acid (5.00 g, 11.1 mmol, Intermediate 5.3.) in methyl sulfoxide (70 mL) was added potassium carbonate (9.18 g, 66.4 mmol) and iodomethane (2.76 mL, 44.3 mmol). The solution was stirred at room temperature under an argon atmosphere for 16 h. The reaction mixture was poured into water (150 mL) and was extracted with ethyl acetate (4 X 150 mL). The combined extracts were washed with water (3 X 100 mL), dried over magnesium sulfate and concentrated in vacuo to afford the crude product as an yellow solid, which was purified by trituration with methanol (4.1 g, 77 %): 1H-NMR (DMSO-d6) δ 0.74 (t, J = 6.8, 3H), 2.07 (s, 3H), 2.97 (t, J = 6.2, 2H),
3.39 (s, 3H), 3.72 (s, 3H), 3.79 (s, 3H), 3.82 (s, 3H), 3.83 - 3.91 (m, 2H) 3.89 - 4.06 (m, 2H), 6.94 (s, IH), 7.17 (t, J = 7.5, IH), 7.29 (dd, J = 1.9, 7.6, IH), 7.55 (dd, J = 1.8, 7.8, IH), 7.90 (s, IH); MS (HPLC/ES): m/z = 480.3 (M + 1).
6.0. 3-[l-(Ethoxycarbonyl)-8,9-dimethoxy-3-trifluoromethyl-5,6- dihydropyrrolo[2,l-a]isoquinolin-2-yl]-2-benzoic acid methyl ester
Figure imgf000060_0001
To a solution of ethyl 2-(6,7-dimethoxy-2,3,4-trihydroisoquinolylidene)acetate (1.13 g, 4.06 mmol Intermediate 1.2.) in 100 mL ethanol was added methyl-3-formylbenzoate
(1.00 g, 6.09 mmol), trifluoromethylnitromethane (1.00 mL, 8.12 mmol), and piperidine (600 μL, 6.09 mmol). The resulting solution was heated to 80 °C and was stirred for 18 hours, at which time TLC analysis (silica gel 60, 50:50 ethyl acetate/hexanes, UN detection) suggested complete reaction. The crude reaction mixture was concentrated in vacuo, and the residue was purified by flash column chromatography eluting with 1 :2 ethyl acetate/hexanes to afford 700 mg (1.39 mmol, 34%) of the title compound: XH-NMR (CD3CN) δ 8.01 (m, IH), 7.89 (s, IH), 7.65 (s, IH), 7.66 (s, IH), 4.19 (t, J- 6.8 Hz, 2H), 3.98 (q, J= 8.5 Hz, 2H), 3.90 (s, 3H), 3.89 (s, 3H), 3.81 (s, 3H), 3.09 (t, J= 8.5 Hz, 2H), 0.85 (t, J= 6.8 Hz, 3H); MS (HPLC/ES) m/z = 504.2 (M + 1); LC RT (Method G): 3.45 mm.
6.1 3-[l-(Ethoxycarbonyl)-8,9-dimethoxy-3-trifluoromethyl-5,6- dihydropyrrolo[2,l-a]isoquinolin-2-yl]-2-benzoic acid
Figure imgf000061_0001
To a solution of 3-[l-(Ethoxycarbonyl)-8,9-dimethoxy-3-trifluoromethyl-5,6- dihydropyrrolo[2,l-a]isoquinolin-2-yl]-2-benzoic acid methyl ester (660 mg, 1.31 mmol Intermediate 6.0.) in water/tetrahydrofuran (20 mL, 1:1 vol/vol) was added of lithium hydroxide (628 mg, 26.22 mmol). The resulting solution was heated to 50 °C and was stirred for 18 hours, at which time TLC analysis (silica gel 60, 50:50 ethyl acetate/hexanes,
UN detection) suggested complete reaction. The crude reaction mixture was concentrated in vacuo, and the residue was dissolved in aqueous ΝaHCO3 (20 mL). The aqueous layer was washed with EtOAc (2 x 20 mL), was made acidic with aqueous 2N hydrochloric acid . The aquesous layer was extracted with dichloromethane/isopropanol (4 x 40 mL, 10:1 vol/vol), and the combined organics were dried (MgSO4), filtered and concentrated to afford the title compound (344 mg, 0.704 mmol, 54%): 1H-NMR (DMSO, d6) δ 13.01 (br. s., IH), 7.92 (dt, J = 7.0, 1.5 Hz, IH), 7.76 (s, IH), 7.59 (s, IH), 7.49 (m, 2H), 7.02 (s, IH), 4.15 (t, J= 7.1 Hz, 2H), 3.90 (q, J= 7.1 Hz, 2H), 3.81 (s, 3H), 3.72 (s, 3H), 3.07 (t, J = 7.1 Hz, 2H), 0.75 (t, J = 7.1 Hz, 3H); MS (HPLC/ES) m/z = 490.2 (M + 1); LC RT (Method G): 2.92 min. Amine Coupling Procedures
Method A: Parallel Synthesis Method
Figure imgf000062_0001
The amines used in this experiment were prepared as 0.5 M solution in dichloromethane by using Bohdan weight station. The amine solutions (270μl, 135.0 μmol) were dispensed into 96-well format FlexChem® Multiple Synthesis Reactor Block by using Tecan station. Ethyl 2-(3-carboxyphenyl)-8,9-dimethoxy-3-methyl-5,6-dihydro-pyrrolo[2,l- a]isoquinoline-l-carboxylate (60mg/lml DCM, 137.8 μmol, Intermediate 1.3.) was added to each well by Bohdan dispense station. PS-DCC resin (n-cyclohexylcarbodiimide, n'- methyl polystyrene HL, 1.9 mmol/g, 140 mg, 266 μmol) was then loaded to each well of the 96-well format FlexChem® Multiple Synthesis Reactor Block. The block was then sealed and stirred by rotation motion in a FlexChem® Rotating Oven at ambient temperature for 12 h. The solution in each well was released to 96 deep well plate. The resin was washed with dichloromethane (3 X 2 ml) and the dichloromethane was released to the other deep well plates. After concentration to remove solvents, the combined material was diluted with dichloromethane and transferred to 96-well format FlexChem® Multiple Synthesis Reactor Block. PS-Isocynate (80 mg, lOOμmol, 1.7 mmol/g) was loaded to each of well which contains the reaction mixture. The block was then sealed and stirred by rotation motion in a FlexChem® Rotating Oven at ambient temperature for 12 h to scavenge the excess amine. The resin was filtered and washed with dichlormethane (3
X 2ml). The dichloromethane solution was collected into a 96 deep well plate. The solution was then transferred by Tecan to 80 individual pre-weighed 8 ml vials. The solvent was removed in vacuo and the product was weighed with a Bohdan weight station. Products were confirmed by both 1HNMR and LC-MS.
Method B Example 1: Ethyl 2-[4-hydroxy-3-({[2-(2 pyridinylamino)ethyl] amino} carbonyl)- phenyl]-8,9-dimethoxy-3-methyl-5,6-dihydropyrrolo[2,l-a]isoquinoline-l-carboxylate
Figure imgf000063_0001
A mixture of 5-[l-(ethoxycarbonyl)-8,9-dimethoxy-3-methyl-5,6-dihydropyrrolo[2,l- a]isoquinolin-2-yl]-2-hydroxybenzoic acid (100 mg, 0.230 mmol, Intermediate 2.1.), N-(2- aminoethyl)-N-(2-ρyridinyl)-amine (63.0 mg, 0.46 mmol), PS-DCC (N- cyclohexylcarbodiimide, N'-methyl polystyrene HL, 117 mg, 0.344 mmol), and 1- hydroxybenzotriazole (62.0 mg, 0.46 mmol) in tefrahydrofuran (1 mL) and dichloromethane (2 mL) was reacted at room temperature for 18 h. The reaction mixture was filtered and the filtrate was concentrated in vacuo. Purification by silica gel flash chromatography provided ethyl 2-[4-hydroxy-3-({[2-(2-pyridinylamino)ethyl]- amino}carbonyl)phenyl]-8,9-dimethoxy-3-methyl-5,6-dihydropyrrolo[2,l-a]isoquinoline- 1-carboxylate (135 mg, 71%): MS (HPLC/ES): m/z = 571.0 (M + 1; LCMS RT (method A): 2.49 min.
Method C
Example 2: Ethyl 2-[4-hydroxy-3-({[2-(lH-pyrazol-l-yl)-ethyl]-amino}-carbonyl)- phenyl]-8,9-dimethoxy-3-methyl-5,6-dihydropyrrolo[2,l-a]-isoquinoline-l- carboxylate
Figure imgf000064_0001
To a solution of 70 mg (0.16 mmol) of 5-[l-(ethoxycarbonyl)-8,9-dimethoxy-3-methyl- 5,6-dihydropyrrolo[2,l-a]isoquinolin-2-yl]-2-hydroxybenzoic acid (Intermediate 2.1.) in 5 mL of tetrahydrofuran was added 45 mg (0.23 mmol) of EDCI, 31 mg (0.23 mmol) of 1- hydroxybenzotriazole, and 60 μL of triethylamine. After 30 min on the J-Kem block, 45 mg (0.23 mmol) of 2-(lH-pyrazol-l-yl)ethylamine was added. This was allowed to shake for 18 h, at which time TLC analysis (silica gel 60, 90:10 dichloromethane/methanol, UN detection) suggested complete reaction. The reaction mixture was concentrated in vacuo and the crude product was purified by preparative HPLC, which afforded 35 mg (0.064 mmol, 41%) of product as a pink solid: 1H-ΝMR (CD3CN) δ 0.95 (t, J = 7.0, 3H), 2.16 (s, 3H), 3.01 (t, J = 6.4, 2H), 3.77 (m, 2H), 3.82 (s, 3H), 3.86 (s, 3H), 3.98 (m, 2H), 4.03 (q, J = 7.2, 2H), 4.36 (t, J = 5.7, 2H), 6.24 (t, J = 1.9, IH), 6.92 (m, 2H), 7.28 (dd, J = 2.2, 8.6, IH), 7.37 (d, J = 2.3, IH), 7.47 (m, IH), 7.53 (m, IH), 7.63 (br s, IH), 7.79 (s, IH) 12.5 (s, IH); MS (HPLC/ES) m/z = 545.0 (M + 1); LCMS RT (method B) 3.14 min.
Method D
Example 3: Ethyl 8,9-dimethoxy-3-methyl-2-{3-[(4-phenyl-l-piperazinyl)-carbonyl]- phenyl}-5,6-dihydropyrrolo[2,l-a]-isoquinoline-l-carboxylate
Figure imgf000064_0002
To a solution of 500 mg (1.148 mmol) of 3-[l-(ethoxycarbonyl)-8,9-dimethoxy-3-methyl- 5,6-dihydropyrrolo[2,l-a]isoquinolin-2-yl]benzoic acid (Intermediate 1.3.) in 10 mL of dichloromethane was added 279 μL (1.72 mmol) of 1-phenylpiperazine, followed by 160 μL (148 mmol) triethylamine, 242 mg (1.26 mmol) of l-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride, and 14 mg (0.115 mmol) of 4-dimethylaminopyridine. The resultant clear yellow solution was stirred at room temperature under an argon atmosphere for 16 h, at which time TLC analysis (silica gel 60, 2:3 ethyl acetate/hexanes, UN detection) suggested complete reaction. The organics were washed with 1 Ν hydrochloric acid (1 X 25 mL) and saturated aqueous ΝaHCO solution (1 X 25 mL), dried over sodium sulfate and concentrated in vacuo. The residue was recrystallized from ether:hexanes to provide 189.1 mg (0.326 mmol, 28%) of a cystalline solid: XH-NMR (CDC13): δ = 0.95 (t, J= 7.1, 3H), 2.17 (s, 3H), 3.00 (t, J= 6.0, 2H), 3.20 (m, 4H), 3.67 (m, 2H), 3.91 (s, 3H), 3.92 (s, 3H), 4.04 (q, J = 7.0, 2H), 6.73 (s, 3H), 6.92 (m, 3H), 7.30 (m, 5H), 7.93 (s, IH); MS (HPLC/ES): m/z = 580.9 (M + 1); LCMS RT (method A):
3.41 min; TLC (30:70 ethyl acetate/hexanes): = 0.31.
Method E
Example 4: Ethyl 2-(3-{[4-(cyclohexylmethyl)-l-piperazinyl]-carbonyl}-phenyl)-8,9- dimethoxy-3-methyl-5,6-dihydropyrrolo [2,1-a] -isoquinoline-1-carboxylate hydrochloride
Figure imgf000065_0001
To a solution of 3-[l-(ethoxycarbonyl)-8,9-dimethoxy-3-methyl-5,6-dihydropyrrolo[2,l- a]-isoquinolin-2-yl]-benzoic acid (871 mg, 2 mmol, Intermediate 1.3.) in dichloromethane (10 mL) was added 1-hydroxybenzotriazole (270 mg, 2 mmol) and 4-methylmorpholine (0.66 mL, 6 mmol). The reaction mixture was cooled to -10° C, and l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (383 mg, 2.0 mmol) was added. After being stirred for 10 min, the mixture was allowed to warm to room temperature for 20 min, and was subsequently cooled to — 10°C. To the cooled reaction mixture was added 1-cyclohexylmethylpiperazine (438 mg, 2.4 mmol). The solution was allowed to warm to room temperature, and was stirred for 16 h. Subsequently, aqueous IN KHSO solution (5 mL) was added, and the two phases were separated. The organic layer was washed with saturated aqueous NaHCO3 solution (10 mL), dried over sodium sulfate and concentrated in vacuo. The crude product was purified with a Biotage Quad 4 system (25 M column) eluting with 50:50 ethyl acetate/hexanes to afford the free base as a solid. The solid was treated with hydrochloric acid (1M in ether, 2 mL). The resultant precipitate was recovered by filtration and was dried in a vacuum oven to afford the title compound (760 mg, 60%): MS (HPLC/ES): m/z = 600.2 (M + 1); LCMS RT (method A): 2.57 min.
Method F Example 5: Ethyl-2-{3-[(cyclohexylamino)-carbonyl]-4-fluorophenyl}-8,9-dimethoxy-
3-5,6-dihydropyrrolo [2,1-α] isoquinoline-1-carboxylate
Figure imgf000066_0001
To a 0 °C solution of 100 mg (0.22 mmol) of 5-[l-(ethoxycarbonyl)-8,9-dimethoxy-3- methyl-5,6-dihydropyrrolo[2,l-a]isoquinolin-2-yl]-2-fluorobenzoic acid (Intermediate 4.2.) in 4 mL of tetrahydrofuran was added 20 μL (0.26 mmol) of oxalyl chloride dropwise followed by one drop of N,N-dimethylformamide. This was warmed to room temperature and was stirred for 1.5 h. The resulting solution was concentrated in vacuo and the oil was diluted with 4 mL of tetrahydrofuran, followed by the addition of 60 μL (0.44 mmol) of triethylamine and 40 μL (0.33 mmol) of cyclohexylamine. This was stirred for 1 h, at which time TLC analysis (silica gel 60, 50:50 ethyl acetate/hexanes, UN detection) suggested complete reaction. The reaction mixture was diluted with water and ethyl acetate and the layers were separated. The aqueous layer was extracted with ethyl acetate (3 X 10 mL), and the organic layer was dried over magnesium sulfate and concentrated in vacuo. The crude product was purified by preparative TLC eluting with 50:50 ethyl acetate/hexanes which afforded 23.2 mg (0.043 mmol, 20%) of the title compound as an off-white solid: MP 140-142 °C; 1H-ΝMR (CD3CN) δ 0.99 (t, J= 7.5, 3H), 1.39 (m, 2H),
1.67 (br s, IH), 1.79 (br s, 2H), 2.17 (s, 3H), 3.00 (t, J= 7.0, 2H), 3.82 (s, 3H), 3.86 (s, 3H), 3.97 (t, J= 6.3, 2H), 4.07 (m, 2H), 6.83 (br s, IH), 6.90 (s, IH), 7.18 (m, IH), 7.34 (m, IH), 7.60 (dd, J= 2.3, 6.9, IH), 7.73 (s, IH); MS (HPLC/ES) 535.2 m/z = (M + 1); LCMS RT (method B): 3.53 min.
Method G
Example 6: Ethyl-8,9-dimethoxy-2-(3-{[4-(3-methoxyphenyl)-l-piperazinyl]}-phenyl)-
3-methyl-5,6-dihydropyrrolo[2,l-a]isoquinoline-l-carboxylate
Figure imgf000068_0001
To a solution of 150 mg (0.345 mmol) of 3-[l-(ethoxycarbonyl)-8,9-dimethoxy-3-methyl- 5,6-dihydropyrrolo[2,l-a]isoquinolin-2-yl]benzoic acid (Intermediate 1.3.) in 4 mL tetrahydrofuran was added 0.08 mL (0.448 mmol) of NN-diisopropylethyl amine. After 5 min 233 mg (0.448) of PyBop and 86.0 mg (0.448 mmol) of l-(3- methoxyphenyl)piperazine were added. The resulting mixture was stirred for 18 h at which time TLC analysis (silica gel 60, 50:50 ethyl acetate/hexanes, UV detection) suggested complete reaction. The reaction mixture was diluted with water, the layers were separated and the aqueous layer was extracted with ethyl acetate (3 X 10 mL). The organic layer was dried over magnesium sulfate and concentrated in vacuo. The crude product was purified by preparative TLC eluting with 50:50 ethyl acetate/hexanes to afford 140 mg (0.23 mmol, 67%) of the title compound: MP: 97-99 °C; 1H-ΝMR (CDC13) δ 0.94 (t, J = 7.6, 3H), 2.17 (s, 3H), 3.00 (t, J= 6.4, 2H), 3.19 (m, 4H), 3.79 (s, 3H), 3.92 (m, 12H), 4.04 (q, J= 1.6, 2H), 6.46 (m, 2H), 6.54 (d, J= 7.1, IH), 6.72 (s, IH), 7.18 (t, J= 8.5, IH), 7.37
(m, 4H), 7.93 (s, IH); MS (HPLC/ES) m/z = 610.4 (M + 1); LCMS RT (method B) 3.25 min.
Method H Example 7: Ethyl 8,9-dimethoxy-3-methyl-2-{3-[(2-methyl-2-phenylhydrazino)- carbonyl]-phenyl}-5,6-dihydropyrrolo[2,l-a]isoquinoline-l-carboxylate
Figure imgf000069_0001
3-[l-(Ethoxycarbonyl)-8,9-dimethoxy-3-methyl-5, 6-dihydropyrrol[2,l-a] isoquinolin -2- yl] benzoic acid (100 mg, 0.23 mmol, Intermediate 1.3.), 1 -methyl- 1-phenylhydrazine (56.1 mg, 0.46 mmol), PS-DCC (n-cyclohexylcarbodiimide, n'-methyl polystyrene HL,
340 mg, 0.35 mmol) and 1-hydroxybenzotriazole (62.06 mg, 0.46 mmol) were dissolved in dry dichloromethane (1.5 mL) and tetrahydrofuran (1.5 mL) and was stirred overnight. The resin was filtered off and the solvent was removed in vacuo. The residue was purified by Biotage using 50:50 ethyl acetate/hexanes to give 103.9 mg (83.9 %>) of the title compound: MS (HPLC/ES): m/z = 540.2 (M + 1); LCMS RT (method A): 3.27 min;
TLC (50:50 ethyl acetate/hexanes): Rf = 0.22.
General Procedures
Method I
Example 8: Ethyl 2-(3-{[(2-chlorobenzyl)-amino]-carbonyl}-4-methoxyphenyl)-8,9- dimethoxy-3-methyl-5,6-dihy dropyrrolo [2,1-a] isoquinoline-l-carboxylate
Figure imgf000070_0001
To a solution of 99.8 mg (0.17 mmol) of ethyl 2-(3-{[(2-chlorobenzyl)-amino]-carbonyl}- 4-hydroxyphenyl)-8,9-dimethoxy-3 -methyl-5 ,6-dihydropyrrolo[2, 1 -ajisoquinoline- 1 - carboxylate was added 25 mg (1.04 mmol) of potassium carbonate and 40 μL (0.69 mmol) of iodomethane. The reaction was heated to 80 °C for 18 h, at which time TLC analysis
(silica gel 60, 50:50 ethyl acetate/hexanes, UV detection) suggested complete reaction. The reaction contents were diluted with water and ethyl acetate and the layers were separated. The organic layer was washed with water (3 X 5 mL), dried over magnesium sulfate and concentrated in vacuo. The crude product was purified by preparative TLC eluting with 50:50 ethyl acetate/hexanes to afford 32.0 mg (0.054 mmol, 31%.) of the title compound as a yellowish solid: 1H-NMR (CD3CN) δ 0.98 (t, J= 6.8, 3H), 2.19 (s, 3H), 2.99 (t, J= 6.4, 2H), 3.81 (s, 3H), 3.86 (s, 3H), 3.95 (t, J= 6.4, 2H), 4.02 (s, 3H), 4.06 (m, 2H), 4.69 (d, J= 5.5, 2H), 6.89 (s, IH), 7.15 (d, J= 8.3, IH), 7.33 (m, 3H), 7.44 (m, 2H), 7.68 (s, IH), 7.87 (d, J= 2.3, IH), 8.53 (t, J= 4.2, IH); MS (HPLC/ES) m/z = 589.4 (M + 1); LCMS RT (method B) 3.81 min.
Method J
1. Formation of solid phase bound propylamine:
4-(4-Formyl-3-methoxyphenoxy)-butyryl aminomethyl resin (NovaBiochem; 10.0 g, 0.78 mmol/g) is suspended in dichloromethane (80 ml)/trimethyl orthoformiate (40 ml). Propylamine (2.31 g, 5 equiv.) is added and the reaction mixture is shaken at room temperature overnight. The mixture is filtered and the resin is washed twice with dimethyl- formamide. The resin is re-suspended in dimethylformamide (100 ml), and tetrabutylammonium borohydride (4.01 g, 2 equiv.) is added to the mixture. After shaking for 15 min at room temperature, the mixture is cooled to - 40 °C, a solution of acetic acid (44 mi dimethylformamide (20 ml) is added, and the reaction mixture is warmed to room temperature again and shaken for an additional 30 min. The resin is filtered and repeatedly washed with dimethylformamide, dimethylformamide/N,N-diisopropyl ethylamine (9:1), dimethylformamide, methanol, dichloromethane and diethyl ether, and dried in vacuo.
2. Acylation of propylamine on solid phase:
The propylamine resin from above (3.00 g, 2.34 mmol) is suspended in dichloromethane (30 ml). N,N-diisopropyl ethylamine (1.81 g, 6 equiv.) is added with stirring, followed by 3-formylbenzoic acid chloride (1.18 g, 3 equiv.). The mixture is shaken for 1 h at room temperature. The resin is filtered and repeatedly washed with methanol, dimethylformamide, methanol, dichloromethane and diethyl ether, and dried in vacuo.
3. Formation of heterocycle:
The foπnylbenzoic acid amide resin from above (1.00 g, 0.78 mmol) is suspended in dioxane (6 ml)/isopropanol (3 ml). The respective nitroalkane (5 equiv.) and ammonium acetate (300 mg, 5 equiv.) are added, and the mixture is shaken at 100 °C overnight. The resin is filtered, repeatedly washed (methanol, water, dimethylformamide, methanol, dichloromethane and diethyl ether), and dried in vacuo. The resin is re-suspended in dioxane (6 ml)/ethanol (3 ml), Intermediate 1.2 (432 mg, 2 equiv.) is added, and the mixture is shaken at 80 °C overnight. The resin is filtered, repeatedly washed (methanol, dichloromethane, diethyl ether) and dried in vacuo. The crude product is cleaved from solid phase with 50%) trifluoroacetic acid in dichloromethane. Method K
Example: 9: Ethyl 2-(3-{[(2,4-difluorobenzyl)-amino]-carbonyl}-phenyl)-8,9- dimethoxy-3-methyl-5,6-dihydropyrrolo[2,l-a]isoquinoline-l-carboxylate
Figure imgf000072_0001
To a solution of of ethyl 2-(6,7-dimethoxy-2,3,4-trihydroisoquinolylidene)acetate (100 mg, 0.36 mmol, Intermediate 1.2.) in ethanol (5 mL) was 3-formyl-N-(2,4-difluorobenzyl)- benzamide (149 mg, 0.54 mmol), trifluoromethylnitiOmethane (90 μL, 0.72 mmol), and piperidine (50 μL, 0.54 mmol). The resulting solution was heated to 80 °C and was stirred for 18 hours, at which time TLC analysis (silica gel 60, 50:50 ethyl acetate/hexanes, UV detection) suggested complete reaction. The crude reaction mixture was concentrated in vacuo and the resulting crude solid was purified by preparative HPLC eluting with 10-
90% acetonitrile/water over 3.5 minutes to afford the title compound as a white solid (24.8 mg, 0.040 mmol, 11%): 1H-NMR (CD3CN) δ 7.82 (dt, J= 7.4, 1.6 Hz, IH), 7.73 (m, IH),
7.66 (s, IH), 7.56 (m, IH), 7.46 (m, 3H), 6.96 (m, 3H), 4.57 (d, J= 5.8 Hz, 2H), 4.19 (t, J
= 6.2 Hz, 2H), 3.97 (q, J= 7.2 Hz, 2H), 3.88 (s, 3H), 3.81 (m, 3H), 3.09 (t, J= 6.8 Hz, 2H) 0.82 (t, J = 7.2 Hz, 3H); MS (HPLC/ES) m/z = 615.1 (M + 1); HPLC RT (Method G):
3.37 min. Preparation Examples
Table 1
The following amides were prepared from Intermediate 1.3. using the amine coupling procedure indicated.
Figure imgf000073_0001
Figure imgf000073_0002
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Table 2
The following amides were prepared from Intermediate 2.1. using the amine coupling procedure indicated.
Figure imgf000098_0001
Figure imgf000098_0002
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Table3
The following amides were prepared from Intermediate 3.3. using the amine coupling procedure indicated.
Figure imgf000116_0001
Figure imgf000116_0002
Figure imgf000117_0001
Figure imgf000118_0001
Table 4
The following amides were prepared from Intermediate 4.2. using the amine coupling procedure indicated.
Figure imgf000119_0001
Figure imgf000119_0002
table 4 Table 5
The following amides were prepared from Intermediate 5.3. using the amine coupling procedure indicated.
Figure imgf000120_0001
Figure imgf000120_0002
Figure imgf000121_0001
Figure imgf000122_0001
Table 6
The following amide was prepared from hitermediate 2.1. using the amine coupling procedure indicated, followed by methylation of the phenol using general procedure I.
t
Figure imgf000123_0001
Table 7
The following amides were prepared from Intennediate 5.4. using the amine coupling procedure indicated.
Figure imgf000124_0001
Figure imgf000124_0002
Figure imgf000125_0001
table 7 Table 8
The following amides were prepared using general procedure J.
Figure imgf000126_0001
Figure imgf000126_0002
table 8
Table 9
The following amides were prepared from intennediate 6.1 using general the amine coupling procedure indicated.
Figure imgf000127_0001
Figure imgf000127_0002
table 9
Table 10
The following amides were prepared from intennediates indicated, following the same procedure used for the synthesis of Intennediate 6.0.
Figure imgf000128_0001
tab
Figure imgf000128_0002
Biological tests
In vitro Enzyme Inhibition Assay :
Full-length recombinant PDE 10a was expressed in Sf9 insect cells (Invitrogen, Carlsbad,
California, U.S.A.) using the Bac-to-Bac™ Baculovirus Expression System (Life Technologies, Gaithersburg, MD, U.S.A.). 48 hours post infection, cells were harvested and resuspended in 20 mL (per IL culture) Lysis Buffer (50 mM Tris-hydrochloric acid, pH 7.4, 50 mM NaCl, 1 mM MgCl2, 1.5 mM EDTA, 10% glycerol plus 20 μL Protease Inhibitor Cocktail Set III [CalBiochem, La Jolla, CA, U.S.A.]). Cells were sonicated at 4°C for 1 minute and centrifuged at 10,000 RPM for 30 minutes at 4°C. Supernatant was removed and stored at -20°C for activity assays.
The test compounds were serially diluted in DMSO using two-fold dilutions to stock concentrations ranging typically from 200 μM to 1.6 μM (final concentrations in the assay range from 4 μM to 0.032 μM). 96-well assay isoplates (Wallac Inc., Atlanta, GA, U.S.A.) were loaded with 2 μL of the serially diluted individual test compounds followed by 50 μL of a dilution of crude recombinant PDE lOa-containing Sf9 cell lysate. The dilution of the lysate was selected such that less than 70%> of the substrate is converted during the later incubation (typical dilution: 1:10000; dilution buffer: 50 mM Tris/hydrochloric acid pH
7.5, 8.3 mM MgCl2, 1.7 mM EDTA, 0.2% BSA). The substrate, [5',8-3H] adenosine 3',5'- cyclic phosphate (1 μCi/μL; Amersham Pharmacia Biotech., Piscataway, NJ, U.S.A.), was diluted 1:2000 in assay buffer (assay buffer: 50 mM Tris/hydrochloric acid pH 7.5, 8.3 mM MgCl2, 1.7 mM EDTA) to give a final working concentration of 0.0005 μCi/μL. The enzymatic assay was initiated by addition of 50 μL (0.025 μCi) of diluted substrate.
Reactions were incubated at room temperature for 60 minutes and terminated by addition of 25 μL of 18 mg/mL Yttrium Scintillation Proximity Beads (Amersham Pharmacia Biotech., Piscataway, NJ, U.S.A.). Plates were sealed and incubated at room temperature for 60 minutes. Plates were read for 30 seconds/well using a Microbeta counter (Wallac Inc., Atlanta, GA, U.S.A.). The IC50 values were determined by plotting compound concentration versus percent inhibition. Representative results are shown in Table 11 : Table 11
Figure imgf000130_0001
Figure imgf000131_0001
table 11
In vitro Proliferation Inhibition Assay:
MDA-MB-231 human breast carcinoma cells (ATCC # HTB26) were cultured in standard growth medium (DMEM), supplemented with 10%. heat-inactivated FBS, 10 mM HEPES, 2 mM glutamine, 100 U/mL penicillin, and 100 μg/mL streptomycin) at 37°C in 5% CO2 (vol/vol) in a humidified incubator. Cells were plated at a density of 3000 cells per well in 100 μL growth medium in a 96 well culture dish. 24 hours after plating, lactate dehydrogenase (LDH) activity was determined using the Cytotox 96 Non-radioactive
Cytotoxicity Kit (Promega, Madison WI, U.S.A.) to yield T0 LDH values. Briefly, cells were lysed with the addition of 200 μL of Lysis Buffer (included in the Promega Kit) and lysates were further diluted 1:50 in Lysis Buffer. 50 μL of diluted cell lysate were transferred to a fresh 96 well culture plate. The assay was initiated with the addition of 50 μL of substrate per well. Color development was allowed to proceed for 10-15 minutes.
The assay was terminated with the addition of 50 μL of Stop Solution (included in the Promega Kit). Optical densities were determined spectrophotometrically at 490 nm in a 96 well plate reader (SpectraMax 250, Molecular Devices, Sunnyvale, CA, U.S.A.).
Test compounds were dissolved in 100%) DMSO to prepare 10 mM stocks. Stocks were further diluted 1 :250 in growth medium to yield working stocks of 40 μM test compound in 0.4%) DMSO. Test compounds were serially diluted in growth medium containing 0.4%> DMSO to maintain constant DMSO concentrations for all wells. 50 μL of fresh growth medium and 50 μL of diluted test compound were added to each culture well to give a final volume of 200 μL. The cells with and without individual test compounds were incubated for 72 hours at which time LDH activity was measured to yield T 2h values. Optionally, the IC50 values can be detennined with a least squares analysis program using compound concentration versus percent inhibition.
% Inhibition = [l-(T72htest-Toh)/(T72h ctri-T0h)] 100 where
T 2h test - LDH activity at 72 hours in the presence of test compound
T72 ctri = LDH activity at 72 hours in the absence of test compound
Toh = LDH activity at Time Zero
Representative results are shown in Table 12 below:
Table 12
Figure imgf000132_0001
Figure imgf000133_0001
table 12 In vivo Tumor Growth Inhibition Assay: MDA-MB-231 Tumor Xenograft Model
Inhibition of tumor growth in vivo is readily detennined via the following assay:
MDA-MB-231 cells are cultured as described above. The cells are harvested by trypsinization, washed, counted, adjusted to 2.5x107 cells/mL with ice cold phosphate-buffered saline (PBS), and subsequently stored on ice until transplantation. Xenograft experiments are conducted using eight-to-ten week-old female athymic mice with an average body mass of about 20-25 g. Approximately 5 x 106 cells in a total volume of 0.2 mL PBS are injected subcutaneously in the flank region. Thereafter the mice are randomized and divided into several groups that reflect different dosages or schedules, respectively (n = 10 mice/group). The test compounds are administered starting at day 1 at different dosages (e.g. 10, 20 and 40 mg/kg) and different schedules (e.g. QlDxl5, Q2Dx7, Q3Dx5). Test compounds are formulated for oral administration in a vehicle for oral administration composed of polyethylene glycol-400, Cremophor®, ethanol and 0.9% saline (40:5:5:50).
Tumor measurements are performed twice per week. Tumor weights are calculated using the formula (a x vX)!2, where a and w refer to the larger and smaller dimensions collected at each measurement.. Animals are sacrificed on day 15 after transplantation and plasma was harvested for pharmacokinetic analyses.
In vivo Tumor Growth Inhibition Assay: MX-1 Tumor Xenograft Model
An MX-1 breast tumor xenograft model is maintained by serial passage in NCr nu/nu female mice (Taconic Farms, Germantown, NY, USA). Tumors are aseptically harvested from mice when they weigh approximately lg. The envelope and any non- viable areas are dissected and the viable tissue is cut into 3 x 3 x 3 mm cubes. These fragments are implanted in the axilary region of the flank of recipient mice using a trochar.
Treatment in anti-tumor efficacy studies is intiated when all mice have tumors ranging in size from 75-125 mg. There are typically 10 mice in each experimental group. Each experiment contains an untreated control group to monitor tumor growth kinetics, a vehicle-treated control group, and a positive agent control group to assess the response of the model in each experiment to an agent with an expected degree of anti-tumor efficacy. Lack of confonnance of any of the controls to the historical ranges for the model constitutes a reason to nullify the study. The test compounds were administered starting at different dosages (e.g. 75 and 150 mg/kg) and different schedules (e.g. qld x 10, bid x 10). Test compounds are formulated for oral administration once per day in a vehicle composed of 51%) PEG400/ 12% ethanol/ 12% Cremophor® EL/ 0.1 N hydrochloric acid . Tumor size is recorded in whole mm as measured in two perpendicular dimensions. Animal body weights are recorded in tenths of grams. Both measurements are collected two to three times per week. Animals are sacrificed on day 10 after the last dose and last measurements.
Tumor weights are calculated using the equation (/ x w2)/2, where / and w refer to the larger and smaller dimensions collected at each measurement. Efficacy is measured as the percent suppression of tumor growth expressed as %ΔT/ΔC, where ΔT and ΔC represent the change in the size of the average tumor in the treated and control groups, respectively, over the treatment period. Significance is evaluated using a Student's t-test with a p<0.05.
B. Operative examples relating to pharmaceutical compositions
The compounds according to the invention can be converted into pharmaceutical preparations as follows:
Tablet:
Composition:
100 mg of the compound of Example 1, 50 mg of lactose (monohydrate), 50 mg of maize starch (native), 10 mg of polyvinylpyrrolidone (PNP 25) (from BASF, Ludwigshafen,
Germany) and 2 mg of magnesium stearate.
Tablet weight 212 mg, diameter 8 mm, curvature radius 12 mm.
Preparation:
The mixture of active component, lactose and starch is granulated with a 5%> solution
(m/m) of the PNP in water. After drying, the granules are mixed with magnesium stearate for 5 min. This mixture is moulded using a customary tablet press (tablet format, see above). The moulding force applied is typically 15 kΝ.
Orally administrable suspension:
Composition:
1000 mg of the compound of Example 1, 1000 mg of ethanol (96%>), 400 mg of Rhodigel (xanthan gum from FMC, Pennsylvania, USA) and 99 g of water.
A single dose of 100 mg of the compound according to the invention is provided by 10 ml of oral suspension. Preparation:
The Rliodigel is suspended in ethanol and the active component is added to the suspension. The water is added with stirring. Stirring is continued for about 6h until the swelling of the Rhodigel is complete.

Claims

ClaimsWe claim:
1. A compound of the formula
Figure imgf000138_0001
wherein
x and y independently from each other denote zero or 1 ;
R1 and R2 independently from each other denote hydrogen, Cι-4-alkyl or trifluoromethyl or R1 and R2 together form a Cι- -alkylene bridge;
R3 and R4 independently from each other denote Cι-6-alkyl optionally further substituted with halogen up to perhalo;
R5 denotes a radical of the formula
Figure imgf000138_0002
wherein
R6 denotes C^-alky!, trifluoromethyl, trifluoromethoxy, halogen, hydrogen, hydroxy or Cι-6-alkoxy;
X and Y independently from each other denote
i) hydrogen; ii) Cι-6-alkoxy; iii) C -8-cycloalkyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, hydroxy, benzyloxy, Cι-6- trialkylsilyloxy, halogen and C].-6-alkoxy; iv) C5.8-cycloalkyl fused to C6-C10-aryl, optionally having from 1 to 3 substituents selected from the group consisting of hydoxyl, Cι_6- alkyl, trifluoromethyl, trifluoromethoxy, C3-8-cycloalkyl, halogen and Cι_6-alkoxy; v) C5-Cιo-bridged bicycloalkyl; vi) C6-Cιo-aryl optionally having from 1 to 3 substituents selected from the group consisting of C^-a-kyl, C3-8-cycloalkyl, Cι-6-alkoxy, (Cι_ 6-alkyl)-carbonyl, (C1.6-alkoxy)-carbonyl, trifluoromethyl, trifluoromethoxy and halogen; vii) phenyl fused to a 5- to 7-membered saturated cycloalkyl, optionally containing up to two hetero atoms selected from the group consisting of O, N, and S; viii) heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι_6-alkoxy, phenyloxy, benzyloxy, . 6-alkyl, C3.8-cycloalkyl, trifluoromethyl, trifluoromethoxy, halogen and benzyl; ix) heteroaryl; or x) Cι_6-alkyl, optionally having from 1 to 3 substituents selected from the group consisting of a) halogen up to perhalo, b) cyano, c) -OR7, d) -NR7R8, e) C6-C10-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, nitro, Cι_6- alkyl, C3-8-cycloalkyl, -OR7, -NR7R8, (Cι-6-alkyl)-carbonyl, (Cι-6-alkoxy)-carbonyl, Cι-6-alkoxy, trifluoromethyl, trifluoromethoxy, -C(O)NR7R8, cyano, -SR7, and C6-Cι0-aryl, f) phenyl fused to a 5- to 7-membered saturated cycloalkyl, optionally containing up to two hetero atoms selected from the group consisting of O, N, and S, g) heteroaryl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, C3_8-cycloalkyl, hydroxy, Cι_6-alkoxy, benzyloxy, halogen and trifluoromethyl, h) heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of -ό-alkyl, Cι-6-alkoxy, C3-8-cycloalkyl, halogen and benzyl, and i) C3-8-cycloalkyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, Cι-6-alkoxy and hydroxy;
wherein R7 and R8 independently from each other denote 1) hydrogen,
2) Cι-6-alkyl, optionally ftirther substituted with 1 to 3 substituents selected from the group consisting of hydroxyl and halogen,
3) C3-8-cycloalkyl, 4) benzyl, 5) C6-Cιo-aryl optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkoxy, -6-alkyl, C3.8-cycloalkyl, trifluoromethyl, trifluoromethoxy and halogen, or
6) heteroaryl;
or
X and Y together with the nitrogen atom to which they are attached form heteroaryl or heterocyclyl, wherein said heteroaryl or heterocyclyl may optionally have from 1 to 3 substituents selected from the group consisting of i) C .8-cycloalkyl; ii) Ci-6-alkyl, optionally having from 1 to 3 substituents selected from the group consisting of C .8-cycloalkyl, hydroxy, Ct-δ-alkoxy, halogen, phenylamino, morpholinyl, (Cι_6-alkyl)-aminocarbonyl, benzo[2,3]dioxolyl and C6-Cι0-aryl, wherein said aryl is optionally substituted with Cι_6-alkyl or Cι_6-alkoxy; iii) C6-Cιo-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι-6-alkoxy, C^-alky!, C3-8- cycloalkyl, trifluoromethyl, trifluoromethoxy and cyano, iv) hydroxy; v) Cι-6-alkoxy; vi) Cι-6-dialkylamino; vii) (Cι_6-alkoxy)-carbonyl; viii) aminocarbonyl; and ix) heteroaryl;
or X and Y together with the nitrogen atom to which they are attached fonn heterocyclyl fused to C6-Cι0-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Chalky!, C3.8-cycloalkyl, Cι-6-alkoxy and hydroxymethyl;
or
X denotes hydrogen and
Y denotes -NR9R10; wherein R9 and R10 independently from each other denote
1) hydrogen,
2) C6-Cιo-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι-6-alkyl, C3. 8-cycloalkyl, Cι_6-alkoxy, trifluoromethoxy and trifluoromethyl,
3) heterocyclyl,
4) C3.8-cycloalkyl, or
5) Cι-6-alkyl;
or
R9 and R10 together with the nitrogen atom to which they are attached fonn heterocyclyl or heteroaryl, wherein said heteroaryl or heterocyclyl may optionally have from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, Cι_6-alkoxy, halogen and methoxymethyl;
and an isomer, a pharmaceutically acceptable salt, a hydrate or a hydrate of a pharmaceutically acceptable salt thereof.
2. A compound as recited in claim 1, wherein x and y independently from each other denote zero or 1 ;
R1 and R2 independently from each other denote hydrogen, Cι- -alkyl or trifluoromethyl;
R3 and R4 independently from each other denote Cι-6-alkyl optionally further substituted with halogen up to perhalo;
R5 denotes a radical of the formula
Figure imgf000143_0001
wherein
R6 denotes halogen, hydrogen, hydroxy or -ό-alkoxy;
X and Y independently from each other denote
i) hydrogen; ii) Cι-6-alkoxy; iii) C3.8-cycloalkyl, optionally having from 1 to 3 substituents selected from the group consisting of Cι_ -alkyl, hydroxy, benzyloxy and Cι_ 6-trialkylsilyloxy; iv) C5-g-cycloalkyl fused to C6-Cι0-aryl, optionally substituted with 1 to
3 hydoxyl; v) C5-Cιo bridged bicycloalkyl; vi) C6-Cιo-aryl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, Cι.6-alkoxy and (Ci-6-alkyl)- carbonyl; vii) phenyl fused to a 5- to 7-membered saturated cycloalkyl, optionally containing up to two hetero atoms selected from the group consisting of O, N, and S; viii) heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of Ci-β-alkoxy, phenyloxy, benzyloxy, and benzyl; ix) heteroaryl; or x) Cι- -alkyl, optionally having from 1 to 3 substituents selected from the group consisting of a) halogen up to perhalo, b) cyano, c) -OR7, d) -NR7R8, e) C6-Cιo-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, nitro, hydroxy, Cι-6-alkyl, -NR7R8, Cι_6-alkoxy, trifluoromethyl, trifluoromethoxy and C6-Cι0-aryl, f) phenyl fused to a 5- to 7-membered saturated cycloalkyl, optionally containing up to two hetero atoms selected from the group consisting of O, N, and S, g) heteroaryl, optionally having from 1 to 3 substituents selected from the group consisting of Cι-6-alkyl, hydroxy, C^-alkoxy, benzyloxy and trifluoromethyl, h) heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of Ct-ό-alkyl and benzyl, and i) C3-s-cycloalkyl, optionally further substituted with 1 to 3 substituents hydroxy; wherein R and R independently from each other denote
1) hydrogen,
2) Cι_6-alkyl, optionally further substituted with 1 to 3 substituents hydroxyl,
3) benzyl,
4) C6-Cι0-aryl, optionally further substituted with 1 to 3 substituents -ό-alkoxy, or
5) heteroaryl;
or
X and Y together with the nitrogen atom to which they are attached form heteroaryl or heterocyclyl, optionally having from 1 to 3 substituents selected from the group consisting of i) C3.8-cycloalkyl; ii) Cι_6-alkyl, optionally having from 1 to 3 substituents selected from the group consisting of C3.8-cycloalkyl, hydroxy, Cι_6-alkoxy, phenylamino, morpholinyl,
Figure imgf000145_0001
benzo[2,3]dioxolyl and C6-Cιo-aryl, wherein said aryl is optionally substituted with Ci-6-alkyl or Cι-6-alkoxy; iii) C6-CiQ-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι- -alkoxy, trifluoromethyl and cyano; iv) hydroxy; v) Cι-6-alkoxy; vi) Cι-6-dialkylamino; vii) (Cι-6-alkoxy)-carbonyl; viii) aminocarbonyl; and ix) heteroaryl; or
X and Y together with the nitrogen atom to which they are attached fonn heterocyclyl fused to C6-Cι0-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι_6- alkyl, Cι-6-alkoxy and hydroxymethyl;
or
X denotes hydrogen and
Y denotes -NR9R10; wherein R9 and R10 independently from each other denote
1) hydrogen,
2) C6-C10-aryl, optionally having from 1 to 3 substituents selected from the group consisting of halogen, Cι.6-alkyl and trifluoromethyl,
3) heterocyclyl,
4) C3-8-cycloalkyl, or
5) d-e-alkyl;
or
R9 and R10 together with the nitrogen atom to which they are attached form heterocyclyl or heteroaryl, wherein said heterocyclyl or heteroaryl optionally have from 1 to 3 substituents selected from the group consisting of Ci-β-alkyl and methoxymethyl;
and an isomer, a pharmaceutically acceptable salt, a hydrate or a hydrate of a pharmaceutically acceptable salt thereof.
3. A compound as recited in claim 1, wherein x and y each other denote 1
R , ι and R independently from each other denote hydrogen or Cι-4-alkyl;
Rj denotes Cι_6-alkyl or trifluoromethyl;
R4 denotes Ci^-alkyl;
R> denotes a radical of the formula selected from the group consisting of:
Figure imgf000147_0001
wherein
X and Y independently from each other denote
i) hydrogen; ii) Cι-6-alkoxy; iii) C3.8-cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl, said C3-8-cycloalkyl optionally having from 1 to 2 substituents selected from the group consisting of -β-alkyl, hydroxy, benzyloxy and tert- butyldimethylsilyloxy; iv) indanyl, 2-hydroxyindanyl, or 1,2,3,4-tetrahydronaphthalenyl; v) [2.2.1]bicycloheptane; vi) naphthyl, 4-methoxyphenyl, 3-(Cι-6-alkoxycarbonyl)phenyl or 2- methoxy-4-methylphenyl; vii) benzo[2,3]dioxolyl; viii) pynolidinyl, optionally having from 1 to 2 substituents selected from the group consisting of Cι-6-alkoxy, phenyloxy, benzyloxy and benzyl; ix) thiazolyl, or pyridyl; or x) Cι_6-alkyl, optionally having from 1 to 2 substituents selected from the group consisting of a) halogen up to perhalo, b) cyano, c) hydroxy, Cι-6-alkoxy, benzyloxy, hydroxy-C2.6-alkoxy, or methoxyphenoxy, d) -δ-dialkylamino, di- ydroxy- -δ-alky^-amino, pyridylamino, or anilino, e) C6.10-aryl selected from the group consisting of naphthyl and phenyl, said C60-aryl optionally having from 1 to 2 substituents selected from the group consisting of fluoro, chloro, bromo, iodo, nitro, hydroxy, Cι-6-alkyl, Cι_6-alkoxy, trifluoromethyl, trifluoromethoxy, phenyl, amino and
Figure imgf000148_0001
dialkylamino, f) benzo[2,3]dioxolyl, or 2,3-dihydrobenzo[ 1 ,4]dioxinyl, g) heterocyclyl selected from the group consisting of pyrazolyl, pyrazinyl, pyrrolyl, furyl, indolyl, thienyl, imidazoyl, and pyridyl, said heterocyclyl optionally having from 1 to 2 substituents selected from the group consisting of Cι-6-alkyl, hydroxy, Cι-6-alkoxy, benzyloxy and trifluoromethyl, h) morpholino, tetrahydrofuranyl, piperidinyl, pyrrolidinyl, optionally further substituted with 1 to 2 substituents Cι_6- alkyl or benzyl, and i) C3.8-cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl, said C3- 8-cycloalkyl optionally being further substituted with 1 to 2 substituents hydroxy;
or
X and Y together with the nitrogen atom to which they are attached form i) morpholino, optionally further subsituted with 1 to 2 substituents Cι_ 6-alkyl; ii) piperidinyl, optionally having from 1 to 2 substituents selected from the group consisting of hydroxyl, hydroxymethyl and Cι_6-alkyl; iii) pyrrolidinyl, optionally having from 1 to 2 substituents selected from the group consisting of Ci-ό-dialkylamino, pyridyl, carboxamido, - 6-alkoxy, phenylaminomethyl, methoxymethyl and methoxyphenyl; or iv) piperazinyl, optionally having from 1 to 2 substituents selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexylmethyl, benzyl, morpholinoethyl, Cι_6-alkyl, (Cι-6-alkoxy)-carbonyl, (Cι-6-alkylaminocarbonyl)methyl, pyridyl, pyrazinyl, pyridylmethyl, benzo[2,3]dioxolyl and phenyl, wherein said phenyl is optionally substituted with 1 to 2 substituents selected from the group consisting of fluoro, chloro, bromo, iodo, trifluoromethyl, cyano and Cι_6-alkoxy;
or X and Y together with the nitrogen atom to which they are attached fonn dimethoxytetrahydroisoquinolinyl, 2-methyl-6- fluorotetrahydroquinolinyl, indolinyl, isoindolinyl or
2-hydroxymethyltetrahydroisoquinolinyl;
or
X denotes hydrogen and Y denotes a) phenylamino, having from 1 to 2 substituents selected from the group consisting of fluoro, chloro, bromo, iodo, trifluoromethyl and Cι-6-alkyl; b) piperidinyl, optionally further substituted with 1 to 2 Cι-6- alkyl; c) triazolyl; d) pyrrolidinyl, optionally further substituted with 1 to 2 methoxymethyl; e) morpholino; f) imidazoyl; g) C3.8-cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl; h) -ό-dialkylamino; or i) azepanyl;
and an isomer, a pharmaceutically acceptable salt, a hydrate or a hydrate of a pharmaceutically acceptable salt thereof.
4. A compound as recited in claim 1, wherein the compound has the fonnula (V):
Figure imgf000151_0001
wherein R to R are defined as described in claim 1.
5. A compound as recited in claim 1, wherein
R> denotes a radical of the formula selected from the group consisting of:
Figure imgf000151_0002
6. A process for manufacturing a compound of claim 1, comprising the reaction of a compound of the formula
Figure imgf000152_0001
wherein x, y, R , ι1, R and R are as defined in claim 1,
[A] with the compounds of the formulae
Figure imgf000152_0002
(III) (III)
wherein R .3J a. nd RD are as defined in claim 1, or
[B] with a compound of the formula
Figure imgf000152_0003
wherein RJ and R3 are as defined in claim 1, and optionally
[C] conversion of the compound obtained through either process [A] or [B] into an isomer, a phannaceutically acceptable salt, a hydrate, or a hydrate of a phannaceutically acceptable salt thereof.
7. A process for manufacturing a compound of claim 1, comprising the reaction of a compound of the formula
Figure imgf000153_0001
wherein X, Y, R , R and R are as defined in claim 1,
with a compound of formula HNXY.
8. A compound according to claim 1 for the treatment and/or prophylaxis of disorders.
9. A medicament containing at least one compound according claim 1 in combination with at least one pharmaceutically acceptable, phannaceutically safe carrier or excipient.
10. The use of a compound according to claim 1 for manufacturing a medicament for the treatment and/or prophylaxis of cancer.
11. The medicament according to Claim 9 for the treatment and/or prophylaxis of cancer.
12. The process for controlling cancer in humans and animals by administration of an therapeutically effective amount of at least one compound according to claim 1.
PCT/US2002/040328 2001-12-18 2002-12-18 2-substituted pyrrolo[2.1-a]isoquinolines against cancer WO2003051877A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002366362A AU2002366362A1 (en) 2001-12-18 2002-12-18 2-substituted pyrrolo(2.1-a)isoquinolines against cancer

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US34136701P 2001-12-18 2001-12-18
US60/341,367 2001-12-18
US34231001P 2001-12-19 2001-12-19
US60/342,310 2001-12-19

Publications (1)

Publication Number Publication Date
WO2003051877A1 true WO2003051877A1 (en) 2003-06-26

Family

ID=26992477

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/040328 WO2003051877A1 (en) 2001-12-18 2002-12-18 2-substituted pyrrolo[2.1-a]isoquinolines against cancer

Country Status (2)

Country Link
AU (1) AU2002366362A1 (en)
WO (1) WO2003051877A1 (en)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005003130A1 (en) * 2003-06-30 2005-01-13 Altana Pharma Ag Novel pyrrolodihydroisoquinolines useful in the treatment of cancer
WO2005002579A1 (en) * 2003-06-30 2005-01-13 Altana Pharma Ag Pyrrolo-dihydroisoquinoline derivatives as pde10 inhibitors
WO2005028426A1 (en) * 2003-09-19 2005-03-31 Chugai Seiyaku Kabushiki Kaisha Novel 4-phenylamino-benzaldoxime derivative and use thereof as mek inhibitor
WO2005035514A2 (en) * 2003-10-08 2005-04-21 Vertex Pharmaceuticals Incorporated Modulators of atp-binding cassette transporters containing cycloalkyl or pyranyl groups
EP1645556A1 (en) * 2004-10-07 2006-04-12 Boehringer Ingelheim International GmbH Arylpiperazine-benzoylamide derivatives useful as pharmaceutical agents
WO2006089815A1 (en) * 2005-01-12 2006-08-31 Nycomed Gmbh Novel pyrrolodihydroisoquinolines as pde10 inhibitors
JP2007537163A (en) * 2004-05-06 2007-12-20 サイトキネティクス・インコーポレーテッド Compounds, compositions and methods
WO2008044054A2 (en) * 2006-10-12 2008-04-17 Astex Therapeutics Limited Hydroxy-substituted benzoic acid amide compounds for use in therapy
US7700625B2 (en) 2005-04-13 2010-04-20 Astex Therapeutics Ltd. Hydroxybenzamide derivatives and their use as inhibitors of Hsp90
US7745663B2 (en) 2004-07-26 2010-06-29 Chugai Seiyaku Kabushiki Kaisha 5-Substituted-2-phenylamino benzamides as MEK inhibitors
US7754725B2 (en) 2006-03-01 2010-07-13 Astex Therapeutics Ltd. Dihydroxyphenyl isoindolymethanones
WO2011008597A1 (en) * 2009-07-14 2011-01-20 Schering Corporation Dihydroimidazoisoquinoline derivatives useful as pde10 inhibitors
WO2012080220A1 (en) * 2010-12-13 2012-06-21 Katholieke Universiteit Leuven, K.U. Leuven R&D Novel compounds for the treatment of neurodegenerative diseases
US8277807B2 (en) 2006-10-12 2012-10-02 Astex Therapeutics Limited Pharmaceutical combinations
US8383619B2 (en) 2008-04-11 2013-02-26 Astex Therapeutics Limited Pharmaceutical compounds
US8394789B2 (en) 2008-02-08 2013-03-12 Msd Oss B.V. (Dihydro)pyrrolo[2,1-α]isoquinolines
US8653084B2 (en) 2006-10-12 2014-02-18 Astex Therapeutics Ltd. Hydrobenzamide derivatives as inhibitors of Hsp90
US8779132B2 (en) 2006-10-12 2014-07-15 Astex Therapeutics Limited Pharmaceutical compounds
US8883790B2 (en) 2006-10-12 2014-11-11 Astex Therapeutics Limited Pharmaceutical combinations
US8916552B2 (en) 2006-10-12 2014-12-23 Astex Therapeutics Limited Pharmaceutical combinations
RU2575174C1 (en) * 2015-03-12 2016-02-20 федеральное государственное автономное образовательное учреждение высшего образования "Российский университет дружбы народов" (РУДН) METHOD OF OBTAINING DERIVATIVES OF 5,6-DIHYDROPYRROLO[2,1-a]ISOQUINOLINES, WHICH CONTAIN FUNCTIONAL GROUP IN POSITION 2
WO2016023028A3 (en) * 2014-08-08 2016-05-26 The Regents Of The University Of Michigan G protein-coupled receptor kinase 2 inhibitors and methods for use of the same
US10155259B2 (en) 2011-09-11 2018-12-18 Merck Sharp & Dohme B.V. FSH receptor antagonists
WO2023060811A1 (en) * 2021-10-11 2023-04-20 丽珠集团新北江制药股份有限公司 Fluralaner intermediate and method for preparing fluralaner thereby
US11952378B1 (en) 2023-12-14 2024-04-09 King Faisal University Pyrrolo[2,3-c]isoquinoline-1,2-dione compounds as CK2 inhibitors

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002048144A1 (en) * 2000-12-13 2002-06-20 Bayer Aktiengesellschaft Pyrrolo (2.1-a) dihydroisoquinolines and their use as phosphodiesterase 10a inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002048144A1 (en) * 2000-12-13 2002-06-20 Bayer Aktiengesellschaft Pyrrolo (2.1-a) dihydroisoquinolines and their use as phosphodiesterase 10a inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ANDERSON W K ET AL: "SYNTHESIS AND ANTILEUKEMIC ACTIVITY OF BIS(CARBAMOYL)OXYMETHYL-SUBSTI TUTED PYRROLO2,1-AISOQUINOLINES, PYRROLO1,2-AQUINOLINES, PYRROLO2,1-AISOBENZAZEPINES, AND PYRROLO1,2-ABENZAZEPINES", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 31, no. 11, 1988, pages 2097 - 2102, XP001068970, ISSN: 0022-2623 *

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005002579A1 (en) * 2003-06-30 2005-01-13 Altana Pharma Ag Pyrrolo-dihydroisoquinoline derivatives as pde10 inhibitors
WO2005003129A1 (en) * 2003-06-30 2005-01-13 Altana Pharma Ag Pyrrolodihydroisoquinolines as pde10 inhibitors
AU2004253690B2 (en) * 2003-06-30 2010-03-25 Nycomed Gmbh Pyrrolodihydroisoquinolines as PDE10 inhibitors
WO2005003130A1 (en) * 2003-06-30 2005-01-13 Altana Pharma Ag Novel pyrrolodihydroisoquinolines useful in the treatment of cancer
EA012110B1 (en) * 2003-06-30 2009-08-28 Алтана Фарма Аг Pyrrolodihydroisoquinolines as pde10 inhibitors
JP2009513495A (en) * 2003-06-30 2009-04-02 フォーエスシー アクチエンゲゼルシャフト Novel pyrrolodihydroisoquinolines effective in the treatment of cancer
JP2009513494A (en) * 2003-06-30 2009-04-02 ニコメッド ゲゼルシャフト ミット ベシュレンクテル ハフツング Pyrrodihydroisoquinoline as a PDE10 inhibitor
JP4931419B2 (en) * 2003-09-19 2012-05-16 中外製薬株式会社 Novel 4-phenylamino-benzaldoxime derivatives and their use as MEK inhibitors
WO2005028426A1 (en) * 2003-09-19 2005-03-31 Chugai Seiyaku Kabushiki Kaisha Novel 4-phenylamino-benzaldoxime derivative and use thereof as mek inhibitor
US8084645B2 (en) 2003-09-19 2011-12-27 Chugai Seiyaku Kabushiki Kaisha 4-phenylamino-benzaldoxime derivatives and uses thereof as mitogen-activated protein kinase kinase (MEK) inhibitors
JPWO2005028426A1 (en) * 2003-09-19 2007-11-15 中外製薬株式会社 Novel 4-phenylamino-benzaldoxime derivatives and their use as MEK inhibitors
US7598412B2 (en) 2003-10-08 2009-10-06 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
WO2005035514A3 (en) * 2003-10-08 2005-07-14 Vertex Pharma Modulators of atp-binding cassette transporters containing cycloalkyl or pyranyl groups
WO2005035514A2 (en) * 2003-10-08 2005-04-21 Vertex Pharmaceuticals Incorporated Modulators of atp-binding cassette transporters containing cycloalkyl or pyranyl groups
JP2007537163A (en) * 2004-05-06 2007-12-20 サイトキネティクス・インコーポレーテッド Compounds, compositions and methods
JP4805916B2 (en) * 2004-05-06 2011-11-02 サイトキネティクス・インコーポレーテッド Compounds, compositions and methods
US8575391B2 (en) 2004-07-26 2013-11-05 Chugai Seiyaku Kabushiki Kaisha 5-substituted-2-phenylamino benzamides as MEK inhibitors
US7745663B2 (en) 2004-07-26 2010-06-29 Chugai Seiyaku Kabushiki Kaisha 5-Substituted-2-phenylamino benzamides as MEK inhibitors
EP1645556A1 (en) * 2004-10-07 2006-04-12 Boehringer Ingelheim International GmbH Arylpiperazine-benzoylamide derivatives useful as pharmaceutical agents
WO2006089815A1 (en) * 2005-01-12 2006-08-31 Nycomed Gmbh Novel pyrrolodihydroisoquinolines as pde10 inhibitors
US8101648B2 (en) 2005-04-13 2012-01-24 Astex Therapeutics, Ltd. Hydroxybenzamide derivatives and their use as inhibitors of HSP90
US9914719B2 (en) 2005-04-13 2018-03-13 Astex Therapeutics Ltd. Hydroxybenzamide derivatives and their use as inhibitors of HSP90
US7700625B2 (en) 2005-04-13 2010-04-20 Astex Therapeutics Ltd. Hydroxybenzamide derivatives and their use as inhibitors of Hsp90
US8816087B2 (en) 2005-04-13 2014-08-26 Astex Therapeutics Limited Hydroxybenzamide derivatives and their use as inhibitors of Hsp90
US8530469B2 (en) 2005-04-13 2013-09-10 Astex Therapeutics Ltd. Therapeutic combinations of hydroxybenzamide derivatives as inhibitors of HSP90
US7754725B2 (en) 2006-03-01 2010-07-13 Astex Therapeutics Ltd. Dihydroxyphenyl isoindolymethanones
US8106057B2 (en) 2006-03-01 2012-01-31 Astex Therapeutics, Ltd. Dihydroxyphenyl isoindolylmethanones
US9730912B2 (en) 2006-10-12 2017-08-15 Astex Therapeutics Limited Pharmaceutical compounds
US8277807B2 (en) 2006-10-12 2012-10-02 Astex Therapeutics Limited Pharmaceutical combinations
US8916552B2 (en) 2006-10-12 2014-12-23 Astex Therapeutics Limited Pharmaceutical combinations
WO2008044054A3 (en) * 2006-10-12 2008-08-14 Astex Therapeutics Ltd Hydroxy-substituted benzoic acid amide compounds for use in therapy
WO2008044054A2 (en) * 2006-10-12 2008-04-17 Astex Therapeutics Limited Hydroxy-substituted benzoic acid amide compounds for use in therapy
US8653084B2 (en) 2006-10-12 2014-02-18 Astex Therapeutics Ltd. Hydrobenzamide derivatives as inhibitors of Hsp90
US9428439B2 (en) 2006-10-12 2016-08-30 Astex Therapeutics Ltd. Hydrobenzamide derivatives as inhibitors of Hsp90
US8779132B2 (en) 2006-10-12 2014-07-15 Astex Therapeutics Limited Pharmaceutical compounds
JP2010505933A (en) * 2006-10-12 2010-02-25 アステックス、セラピューティックス、リミテッド Pharmaceutical compounds
US8883790B2 (en) 2006-10-12 2014-11-11 Astex Therapeutics Limited Pharmaceutical combinations
US8394789B2 (en) 2008-02-08 2013-03-12 Msd Oss B.V. (Dihydro)pyrrolo[2,1-α]isoquinolines
RU2495037C2 (en) * 2008-02-08 2013-10-10 Мерк Шарп и Доум Б.В. Pyrrolo[2,1-a]isoquinolines
US8664218B2 (en) 2008-04-11 2014-03-04 Astex Therapeutics Ltd. Pharmaceutical compounds
US8383619B2 (en) 2008-04-11 2013-02-26 Astex Therapeutics Limited Pharmaceutical compounds
WO2011008597A1 (en) * 2009-07-14 2011-01-20 Schering Corporation Dihydroimidazoisoquinoline derivatives useful as pde10 inhibitors
US9284271B2 (en) 2010-12-13 2016-03-15 Katholieke Universiteit Leuven, K.U. Leuven R&D Compounds for the treatment of neurodegenerative diseases
WO2012080220A1 (en) * 2010-12-13 2012-06-21 Katholieke Universiteit Leuven, K.U. Leuven R&D Novel compounds for the treatment of neurodegenerative diseases
US10155259B2 (en) 2011-09-11 2018-12-18 Merck Sharp & Dohme B.V. FSH receptor antagonists
WO2016023028A3 (en) * 2014-08-08 2016-05-26 The Regents Of The University Of Michigan G protein-coupled receptor kinase 2 inhibitors and methods for use of the same
US10023564B2 (en) 2014-08-08 2018-07-17 The Regents Of The University Of Michigan G protein-coupled receptor kinase 2 inhibitors and methods for use of the same
RU2575174C1 (en) * 2015-03-12 2016-02-20 федеральное государственное автономное образовательное учреждение высшего образования "Российский университет дружбы народов" (РУДН) METHOD OF OBTAINING DERIVATIVES OF 5,6-DIHYDROPYRROLO[2,1-a]ISOQUINOLINES, WHICH CONTAIN FUNCTIONAL GROUP IN POSITION 2
WO2023060811A1 (en) * 2021-10-11 2023-04-20 丽珠集团新北江制药股份有限公司 Fluralaner intermediate and method for preparing fluralaner thereby
US11952378B1 (en) 2023-12-14 2024-04-09 King Faisal University Pyrrolo[2,3-c]isoquinoline-1,2-dione compounds as CK2 inhibitors

Also Published As

Publication number Publication date
AU2002366362A1 (en) 2003-06-30

Similar Documents

Publication Publication Date Title
WO2003051877A1 (en) 2-substituted pyrrolo[2.1-a]isoquinolines against cancer
US7723347B2 (en) Substituted phenylamino-pyrimidines
US8329716B2 (en) Hetaryloxy-substituted phenylamino pyrimidines as Rho kinase inhibitors
US7737153B2 (en) Heteroaryloxy-substituted phenylaminopyrimidines as rho-kinase inhibitors
US6608056B1 (en) Fused heteroaryl derivatives
US9085575B2 (en) Tri- and tetracyclic pyrazolo[3,4-B]pyridine compounds as antineoplastic agent
US20080249105A1 (en) Pyrrolopyridine-Substituted Benzol Derivatives for Treating Cardiovascular Diseases
US20040138249A1 (en) Pyrrolo (2.1a)dihydroisoquinolines and their use as phosphodiesterase 10a inhibitors
JPH11513398A (en) Bicyclic heteroaromatics as protein tyrosine kinase inhibitors
BR122018075851B1 (en) COMPOUNDS RELATED TO THE TREATMENT OF HYPERPROLIFERATIVE DISEASE AND PROCESSES FOR PREPARING COMPOUNDS
MX2008014618A (en) Compositions and methods for fgf receptor kinases inhibitors.
BR112013004750B1 (en) QUINOLINE AND QUINOXALINE DERIVATIVES AS KINASE INHIBITORS
US20080318946A1 (en) Di-amino-substituted heterocyclic compounds and methods of use
JP4559851B2 (en) Phenylaminopyrimidine and its use as a Rho-kinase inhibitor
JPWO2004043936A1 (en) PLK inhibitor
WO2020231739A2 (en) Compounds and methods for treating cancer
WO2003014117A1 (en) 3-substituted pyrrolo[2.1-a]isoquinoline derivatives
WO2003014116A1 (en) Pyrrolo[2.1-a]isoquinoline derivatives
WO2012121168A1 (en) Kinase inhibitor
AU2007272830B2 (en) Alkyne-substituted pyridone compounds and methods of use
WO2006110447A2 (en) Pyrimidine derivatives and their use in the treatment of cancer
WO2023185073A1 (en) Parp7 inhibitor and use thereof
US20170015664A1 (en) Fused Acridine Derivative and Pharmaceutical Composition, Preparation Method and Use Thereof
CN116262750A (en) Aromatic heterocyclic compound and preparation method and application thereof
KR20160010522A (en) Pyrimidin-4-yl)oxy)-1h-indole-1-carboxamide derivatives and use thereof

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 BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL 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): GH GM KE LS MW MZ 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 IE IT LU MC NL PT 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
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP