CA2350941A1 - Benzamide analogues as parp dna repair enzyme inhibitors - Google Patents

Benzamide analogues as parp dna repair enzyme inhibitors Download PDF

Info

Publication number
CA2350941A1
CA2350941A1 CA002350941A CA2350941A CA2350941A1 CA 2350941 A1 CA2350941 A1 CA 2350941A1 CA 002350941 A CA002350941 A CA 002350941A CA 2350941 A CA2350941 A CA 2350941A CA 2350941 A1 CA2350941 A1 CA 2350941A1
Authority
CA
Canada
Prior art keywords
compound
carboxamide
alkyl
parp
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002350941A
Other languages
French (fr)
Inventor
Roger John Griffin
Alan Hilary Calvert
Nicola Jane Curtin
David Richard Newell
Bernard Thomas Golding
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Newcastle University Ventures Ltd
Original Assignee
Newcastle University Ventures Limited
Roger John Griffin
Alan Hilary Calvert
Nicola Jane Curtin
David Richard Newell
Bernard Thomas Golding
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB9404485A external-priority patent/GB9404485D0/en
Application filed by Newcastle University Ventures Limited, Roger John Griffin, Alan Hilary Calvert, Nicola Jane Curtin, David Richard Newell, Bernard Thomas Golding filed Critical Newcastle University Ventures Limited
Publication of CA2350941A1 publication Critical patent/CA2350941A1/en
Abandoned legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)

Abstract

A range of benzamide analogues consisting of 3-oxybenzamide derivatives are disclosed which can act as potent inhibitors of the DNA repair enzyme poly(ADP-ribose) polymerase or PARP enzyme (EC 2.4.2.30), and which thereby can provide useful therapeutic compounds for use in conjunction with DNA-damaging cytotoxic drugs or radiotherapy to potentiate the effects of the latter.
The compounds disclosed include certain benzoxazole-4-carboxamide compounds and in general they comprise compounds having the general structural formula I

(see formula I) or pharmaceutically acceptable salt thereof, characterised in that Y and X together form a bridge -Y-X- that represents the grouping (see formula II) or (see formula III) or (see formula IV) where R5 is H, alkyl or an optionally substituted aralkyl or aryl group.

Description

BENZAMIDE ANALOGUES
This application is a division of PCT International Application No. PCT/GB95/00513 bearing Canadian Application Serial No. 2,184,747 with the International S Filing Date of March 9, 1995.
The present invention relates to benzamide analogues, especially certain 3-substituted benzamide derivatives that are of interest as being at least potentially useful chemotherapeutic agents by virtue of an ability to inhibit the activity of the enzyme poly ADP-ribosyltransferase (EC 2.4.2.30), also known as poly(ADP-ribose) polymerase, commonly referred to as ADPRT or PARP. In general, the latter abbreviation, PARP, will be used throughout the present specification.
BACKGROUND
At least in higher organisms, the enzyme poly ADP-ribosyltransferase is known to catalyse a transfer of the ADP-ribose moiety from the oxidized form NAD+ of nicotinamide adenine dinucleotide to nuclear acceptor proteins so as to form homo ADP-ribose polymers, and this process has been implicated in a number of cellular events such as, for example, repair of DNA damage, development of cellular differentiation, transformation of cells by oncogenes, and gene expression. A common feature in a number of these processes is the formation and repair of DNA strand breaks and the stage which involves the PARP enzyme appears to be that of DNA ligase II-mediated strand rejoining. In the majority of cases a role for poly ADP-ribosylation has been implicated by the use of inhibitors of the PARP enzyme, and this has led to suggestions that such inhibitors, by interfering with the intracellular DNA repair mechanism, may have a useful chemotherapeutic role insofar as they should be able to modify treatment resistance characteristics and potentiate or enhance the effectiveness of cytotoxic drugs in chemotherapy or of radiation in radiotherapy where a primary effect of the treatment is that of causing DNA damage in target cells, as for example in many forms of antit-umour therapy.
In this connection, several classes of PARP
inhibitors are already known, including benzamide itself and various nicotinamide and benzamide analogues, especially 3-substituted benzamides with small substituent groups such as 3-amino, 3-hydroxy and 3-methoxy. PARP inhibitory activity of certain N-substituted benzamides has also been reported in EP-A-0305008 wherein it has also been proposed to use these compounds in medicine for increasing the cytotoxicity of radiation or of chemotherapeutic drugs.
Regarding this use of benzamides as chemotherapeutic agents, a number of studies on such compounds that are known to exhibit PARP inhibitory activity have confirmed that they can potentiate the cytoxicity of a range of antitumour agents in vitro, for example, bleomycin and methylating drugs. More limited data has further indicated that such benzamides can also potentiate the activity of cytotoxic drugs in vivo, although the dose requirements have appeared to be rather high (e.g. in the region of 0.5g kg-1 per dose for 3-aminobenzamide) and there may be associated problems in preparing satisfactory pharmaceutical formulations and in avoiding toxicity limitations. Furthermore, a number of the known benzamides have also been shown clearly to have potential as radiosensitizers, increasing for example ionising radiation-induced tumour cell kill both in vitro and in vivo, and it is believed that in many cases this effect is related to these compounds acting as PARP inhibitors and interfering with DNA repair.
However, notwithstanding the existing data from in vitro and in vivo studies suggesting that PARP inhibitors have considerable potential as useful chemotherapeutic 35- agents which merit further clinical evaluation, for instance in connection with cancer therapy, currently available known PARP inhibitors are not considered as yet to be entirely suitable to represent candidate drugs.
Accordingly, there is a need to find and develop a greater range of compounds having potentially useful PARP
inhibitory properties.
DISCLOSURE OF THE INVENTION
The present invention identifies a new range or ranges of compounds of interest as PARP inhibitors that can be useful in medicine, especially when administered in conjunction with at least certain cytotoxic drugs or with radiotherapy for increasing the cytotoxic effectiveness thereof. In general, the compounds of this invention as hereinbelow defined comprise benzamide compounds, or analogues, which include 3-position substituents linked in a ring structure with substituents in the 2-position.
More specifically, from one aspect, the invention resides in the use of a compound as herein defined for the manufacture of a medical or veterinary preparation for use in therapy for inhibiting activity of the enzyme poly(ADP-ribose)polymerase or PARP (also known as ADP-ribosyl transferase or ADPRT), such enzyme inhibition constituting an element of a therapeutic treatment, said compound providing an active PARP enzyme inhibiting agent and being a 3-substituted oxybenzamide compound having the general structural formula I

I
,Y
-X
or a pharmaceutically acceptable salt thereof, and characterised in that 30. Y and X together form a bridge -Y-X- that represents the grouping -N=i- or -0-iH- or -S-iH-Rs Rs Rs where Rs is H, alkyl or an optionally substituted aralkyl or aryl group.
The invention also provides for use in therapy as active pharmaceutical substances 3-substituted oxybenzamide compounds having the general structural formula I

\ ~ I
,Y
-X
or a pharmaceutically acceptable salt thereof, characterised in that Y and X together form a bridge -Y-X- that represents the grouping -N=C-Rs where Rs is H, alkyl or an optionally substituted aralkyl or aryl group.
The invention further provides novel 3-substituted oxybenzamide compounds having the general structural formula I

I
\ Y
X
or a pharmaceutically acceptable salt thereof, characterised in that Y and X together form a bridge -Y-X- that represents the grouping -N=C-Rs where Rs is H, alkyl or' an optionally substituted aralkyl or aryl group.

Alkyl groups when present as such or as a moiety in other groups such as alkoxy will generally be composed of 1-8 carbon atoms, preferably 1-6 carbon atoms, and more usually 1-4 carbon atoms.
5 One very important group of compounds of special interest from the point of view of PARP-inhibitory activity comprises benzoxazole-4-carboxamide compounds, i.e, compounds represented by the formula IV

IV
N
~5 where R5, if not H, is preferably alkyl, phenyl or another aryl group such as naphthyl or pyridyl. When RS is an alkyl group this will generally be C1-6 alkyl, such as for example methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl or cyclohexyl. However, it may in some cases be larger, such as in adamantyl for instance . When RS is a phenyl group this may be substituted, especially in the 4 (para) position but alternatively perhaps in the 2-position or 3-position, by substituents such as alkoxy for example.
Within this group of benzoxazole compounds preferred members which are of particular interest include 2-methylbenzoxazole-4-carboxamide, 2-t-butylbenzoxazole-4-carboxamide, 2-phenylbenzoxazole-4-carboxamide, 2-(4-methoxyphenyl)benzoxazole-4-carboxamide.
In the above-mentioned compounds of formula IV, wherein there is an electron-rich aromatic ring, it is believed that the carboxamide group is constrained in a fixed conformation, particularly favourable for presenting the compound as an alternative substrate to NAD+ for the, PARP enzyme, by an intramolecular hydrogen bond between the ring nitrogen atom and one of the hydrogen atoms of the carboxamide group. A similar, although probably somewhat weaker effect may also occur in other compounds of formula I where the X and Y
substituents form a bridge, as defined above, containing an oxygen or sulphur atom, i . a . where the ring N atom of the benzoxazoles of formula IV is replaced by an 0 or S
atom.
The invention also embraces or extends to methods of preparing compounds as hereinbefore defined (including intermediates in some cases) and to the therapeutic use of such compounds. This includes their use for making medical or veterinary preparations or pharmaceutical formulations containing an effective PARP inhibitory amount of the active compound for administration to a patient in conjunction with a cytotoxic drug or radiotherapy in order to increase the cytotoxic effectiveness of the latter. Such preparations or formulations may be made up in accordance with any of the methods well known in the art of pharmacy for administration in any suitable manner, for example orally, parenterally (including subcutaneously, intramuscularly or intravenously), or topically, the mode of administration, type of preparations or formulation and the dosage being generally determined by the details of the associated cytotoxic drug chemotherapy or radiotherapy that is to be enhanced.
As indicated, the compounds according to this invention have at least potential as PARP inhibitors, and in vitro tests hereinafter described have demonstrated positive pharmacological activity which it is believed reflects the activity to be found in vivo in the course of therapeutic clinical use.
It will be understood that where reference is made in this specification to compounds of formula I (or - formula IV) such reference should be construed as extending also to their pharmaceutically acceptable salts where relevant. Also, where any of the compounds referred to can exist in more than one enantiomeric form, all such forms, mixtures thereof, and their preparation and uses are within the scope of the invention.
DESCRIPTION OF EXAMPLES OF PREFERRED EMBODIMENTS
The following examples and descriptions of stages in synthetic routes of preparation of various preferred compounds of interest serve to further illustrate the present invention, but should not be construed in any way as a limitation thereof.

2-Methylbenzoxazole-4-carboxamide (Compound NU1056) 1st Stage - Preparing Methyl (3-Hydroxy-2-nitro)benzoate 3-hydroxy-2-nitrobenzoic acid (5g; 27.32mM) was dissolved in anhydrous methanol (200m1). Anhydrous hydrogen chloride gas was bubbled through the solution until saturated. The mixture was then refluxed for 20 hours (reaction followed by T.L.C. . l0o methanol/900 dichloromethane). Next, the solvent was removed under vacuum to yield a brown solid. The solid was dissolved in water (100m1) and sodium bicarbonate was added until effervescence stopped. Sodium chloride (15g) was added to the aqueous solution, and the product was extracted into ethyl acetate (3x50m1). The pooled aliquots were dried over magnesium sulphate and the solvent removed under vacuum to yield a malty brown solid.
T.L.C. (as before): r.f.: 0.53 NMR: 200MHz: d6 DMSO: b=3. 9 (s; 3H;OCH3) ; 7. 1 (m; 1H;H4) ;
7 . 35 (m; 1H; H6) ; 7 . 8 (t; 1H; HS) Yield 92s 2nd Stage - Preparation of Methyl (2-Amino-3-hydroxy)-Y, o ,., -n ., ~ + o Under a nitrogen atmosphere a palladium/carbon catalyst was suspended in anhydrous methanol (150m1). To this suspension was added methyl (3-hydroxy-2-nitro) benzoate from Stage 1 (4g; 20.3mM). The mixture was left under a hydrogen atmosphere for 4'~ hours. The catalyst was removed by filtration through a "celite" pad, and the solvent removed from the filtrate to yield an orange/brown product.
T.L.C.: 40o Ethyl acetate/60% petroleum ether 60/80.
r.f. 0.33 NMR 200mHz: doDMSO . b: 3.81(S;3H;OCH3);
6. 2 (broad; 2H; NHZ ) ; 6. 5 (t; 1H; HS) ; 6 . 9 (m; 1H; H6) 7. 3 (m; 1H; H4) 9. 8 (s; 1H; OH) Yield 83%
3rd Staae - Preparation of Methyl 2-methylbenzoxazole-4-carboxvlate To a solution of the methyl (2-amino-3-hydroxy)-benzoate (3g; l8.OlmM) in m-xylene (150m1) was added acetyl chloride (1.518m1; 21.6mM). A precipitate was formed; this was left to stir for 30 minutes. On the addition of triethylamine (2.97m1; 21.6mM) the solution became translucent. Pyridinium-p-toluene sulphonic acid (1.2g; 21.6mM) was added, and the mixture refluxed for 34 hours. The solvent was removed by distillation (vacuum) to yield a brown solid which was column chromatographed (50o ethyl acetate/50o petrol 60/80) to give the desired product as a yellow solid.
T.L.C. As above.
1HNMR :200MHz; CDC13; b: 2.69(3H;s;2-CH3) 3.99(3H;s;OMe); 7.33(3H;t;H6); 7.63(lH;dd;H7) 7 . 94 ( 1H; dd; HS ) 4th Stage - Preparation of 2-methylbenzoxazole-4-carboxylic acid Methyl 2-methylbenzoxazole-4-carboxylate (0.1 g, 0.523 mmol) was dissolved in methanol (3 ml), and to this was added aqueous sodium hydroxide solution (0.2 M, 3 ml). The mixture was stirred at 40°C for 4 hours and acidified with hydrochloric acid (6 M) until pH - 1Ø
The mixture was extracted with ethyl acetate (3 x 20 ml), the combined organic layers were washed with water (2 x 20 ml), dried (MgS04) and the solvent was removed under reduced pressure to afford the carboxylic acid (0.068 g, 730) .
5th Staqe - Preparation of 2-methylbenzoxazole-4-carboxamide A solution of 2-methylbenzoxazole-4-carboxylic acid (0.1 g, 0.28 mmol) in anhydrous THF (10 ml) was stirred under nitrogen, and thionyl chloride (0.022 ml, 0.31 mmol), and DMF (O. lm) were added, whereupon the mixture was stirred for a further 5 hours at room temperature.
Aqueous ammonia (0.5 ml) was added and the mixture was stirred for a further 30 minutes. The solvent was removed under reduced pressure, the residual solid was dissolved in water (20 ml), and the solution was extracted with ethyl acetate (3 x 20 ml). The organic layers were pooled, washed with water (2 x 20 ml) and dried (MaSOa). The solvent was removed under reduced pressure to furnish the carboxamide (0.083 g, 84%).
1H (200 MHz) CDC13 b = 6.0 (brs, 1H, NH) , 7. 4 (t, 1H, H6) , 7. 6 (dd, 1H, H7) , 8. 15 (dd, 1H, H5) , 8. 8 (brs, 1H, NH) .

2-Methylbenzoxazole-4-carboxamide (Compound NU1056) In a modification of the procedure described under Example 1 above, the product of the 3rd stage, methyl 2-methylbenzoxazole-4-carboxylate, was prepared directly from the product of the 1st stage, as described below.
Methyl 3-hydroxy-2-nitrobenzoate (O.lg; 0.59mmo1) from the 1st stage was dissolved in anhydrous ethanol (20m1) and to this was added ethyl acetimidate hydro-chloride (0.0678; 0.59mmo1). The reaction mixture was then heated 'under reflux for 24 hours. The ethanol was removed under reduced pressure yielding a brown crystalline solid. This was dissolved in ethyl acetate (3 x 20m1) to produce a precipitate of excess ethyl acetimidate hydrochloride. The excess imidate was S filtered off and the solution washed with sodium hydroxide solution (O.1N; 3 x 20m1), and water (3 x 50m1). The solvent was dried over magnesium sulphate and removed under reduced pressure, leaving a orange crystalline solid (0.1265g; 85%).

2-t-butylbenzoxazole-4-carboxamide (Compound NU1040) (a) lst Stage - Preparation of Methyl 2-t-butyl-benzoxazole-4-carboxylate 2-Amino-3-hydroxybenzoate (O.lg; 0.598mmo1) was dissolved in m-xylene and warmed to 70°C. To this was added pivaloyl chloride (0.117m1; 0.958mmo1), whereupon a brown precipitate was observed to develop. The mixture was stirred for 30 minutes before the addition of triethylamine (0.099m1; 0.958mmo1) and pyridinium-4-toluenesulphonate (0.048; 0.958mmo1). The mixture was then heated under reflux for 26 hours. The m-xylene was removed under reduced pressure to yield a sticky brown solid. The solid was dissolved in water (50m1) and the organics extracted into ethyl acetate (3 x 30m1), pooled, dried over magnesium sulphate, filtered and the solvent removed under reduced pressure.
The title compound was purified via silica column chromatography, with 1:1 ethyl acetate:petrol as the eluant to yield a yellow solid (69%).
IR cm-1: 3040 (3 x CH3) , 1709 (C=0) . M/Z; 233 (63%; M+) ;
218 (50%; -CH3) 202 (42%; -OCH3) ; 186 (100%) ; 173 (12%;
CH3); 160 (33%); 146 (62%); 117 (43%). 1H: d6 DMSO: b 1. 17 (9H; s; (CCH3) 3) ; 3. 99 (3H; s, OCH3) ; 7.31 (1H; t; H6 J=8Hz), 7.66 (1H; dd: H7; J=7.8, 1Hz), 7.95 (1H; dd; HS
J=7, 1Hz).

(b) 2nd Stage - Preparation of 2-t-Butylbenzoxazole-4-carboxamide Methyl 2-t-butyl-4-benzoxazole carboxylate (O.lg;
0.46mmo1) was dissolved in methanol (5ml), and to this S was added aqueous ammonia ( 5m1 ) . The mixture was warmed to 40°C and left to stir for 6 hours at ambient temperature. Once the reaction was complete the solvent was removed under reduced pressure and the product was recrystallised from boiling ethyl acetate and petrol (73a) .
IR cm-1: 3395; 3304 (amide NH); 3163 (3 x CH3), M/Z; 218 (96a; M+); 202 (43%; -NH2) 186 (850; -CONHZ); 175 (770);
160 (35%) 146 (790) ; 133 (230) ; 41 (1000) .
1H: CDC13: b - 1. 44 (9H; s; (CCH3) 3) ; 5. 95 (1H; br s; NH) ;
7.34 (1H; t; H6 J=8Hz), 7.6 (1H; dd: H7; J=7,& 2Hz) 8.07 (1H; dd; H5 J=6.7, 2Hz); 8.88 (1H; br s; NH) 13C 28. 416 (3xCH3) , 34 . 372 (CCH3) , 113. 905 (Ar) , 123.277 (Ar), 124.338 (Ar), 124.338 (Ar), 125.440 (Ar), 139.371 (0-Ar), 150.804 (N-Ar), 166.457 (Ar), 174.471 (C=C
amide).
CHN: Found: C 65.9150; H 6.390; N 12.480, Required: C
66.038%; H 6.465%; N 12.8350.
wTrrtnT ~ n 2-Phenylbenzoxazole-4-carboxamide (Compound NU1051) (a) 1st Stage - Preparation of Ethyl benzimidate Hydrochloride Benzonitrile (0.514m1; 5mmo1) was added to anhydrous ethanol (0.69g). Anhydrous hydrogen chloride gas was bubbled through the solution until saturated. The mixture was left to stir for 20 hours. The white crystalline solid was collected and dried.
mpt: 125-130°C
IR . cm-1, 28,56, 1631. M/Z; 148 (M+, 30%) , 105 (1000) ;

1H: d6 DMSO; b - 1.5 (3H; t, J=7Hz; CHZCH3), 4.75 (2H; q, J=6.9Hz; CH~CH3), 8.0 (5H; m; aromatics).
Elemental Analysis:
ExpectedC: 58.25; H: 6.51; N: 7.54;
FoundC: 58.13; H: 6.43; N: 7.36.
b) 2nd Staqe - Preparation of Methyl 2-phenylbenzoxazole -4-carboxylate To Methyl 2-amino-3-hydroxybenzoate (O.lOg;
0.59mmo1) was added ethyl benzimidate hydrochloride (0.1678; 0.998mmo1) in anhydrous ethanol (20m1). This was refluxed for 20 hours, and the reaction followed by TLC (1:4 Ethyl acetate: petrol).
The ethanol was removed under reduced pressure and the solid dissolved in water (20m1), the organics were extracted into ethyl acetate (3 x 20m1), pooled, and washed with sodium hydroxide solution (2 x lOml; 0.2M), water (2 x lOml) and then dried over magnesium sulfate.
The solvent was removed under reduced pressure to yield a yellow solid. The title compound was isolated by flash chromatography (eluant as for TLC), yielding an off-white crystalline solid. (85%).
IR . cm-1, 1714 . 1H: d6 DMSO: b - 4. 1 (3H; s; OCH3) , 7. 39 (1H; t; J=6Hz; H6), 7.5 (3H; m; H3'; H4'; HS'), 7.78 (1H;
dd J=7.0, 1.0 Hz; H7;), 8.0 (1H; dd; J= 6.67 & 1.14Hz;
HS' ) , 8 . 3 (2H; m; Hz' ; H6' ) .
(c) 3rd Stage - Preparation of 2-phenylbenzoxazole-4-carboxamide To a solution of methyl 2-phenylbenzoxazole-4-carboxylate (0.028; 0.0905mmo1) in methanol (3m1) was added aqueous ammonia (3m1). This was warmed to 40°C and left to stir, whilst the reaction was monitored by TLC
(1:4 EtOAc:petrol). Once all the starting material had reacted a white precipitate was formed. The solvent was removed to yield a white solid, which was recrystallised from ethyl acetate and petrol (70%).
MPt: 199-201°C.
1H: CDC13: b = 6.02 (1H; br s: NH); 7.43 (1H; t; H6;
J=8Hz ) , 7 . 57 ( 3H; m: H3' ; H4' ; HS' ) , 7 . 74 ( 1H; dd;
H7; J=lHz,& 7Hz); 8.23 (1H; dd; H5; J=1Hz & 7.3Hz);
8.27 (2H; m; HZ' ; H6' ) , 8. 97 (1H; br s; NH) .
13C ; M/Z; (EI ) ; 238 (m+; 100 % ) ; 222 (-NHZ 68 % ) ;
195 (-CONH2; 98 % ) . IR cm-1 3383; 3165.
Elemental Analysis:
ExpectedC:70.58, H:4.23, N:11.76.
Found C:70.41, H:4.24, N:11.77.
L'VTMDT L' 2-(4-Nitrophenyl)benzoxazole-4-carboxamide (Compound NU1053) (a) 1st stage - Preparation of Methyl 3-hydroxy-2-(N-4-nitrobenzoyl)aminobenzoate Methyl 2-amino-3-hydroxybenzoate (0.5g; 2.99mmo1) was dissolved in m-xylene (40m1) with warming to 60°C. 4-Nitrobenzoyl chloride (0.556m1; 2.99mmol) was added dropwise, and this was left to stir for 4 hours. The solution was cooled to ambient temperature and the m-xylene removed under reduced pressure. The solid was dissolved in water (100m1) and the organics extracted into ethyl acetate (3 x 50m1). The organic fractions were pooled, dried over magnesium sulphate, filtered and the solvent removed under reduced pressure.
The title product was purified via column chromatography (1:4 ethyl acetate: petrol as eluent) to yield an orange solid (49%).
_ IR . cm-1: 3443 (OH) , 2953, 1697, 1649, 1404. M/Z; 316 (15% M+) .

1H: CDC13: b = 3.95 (3H; s; OCH3) , 7.25 (1H; t; HS
J=8Hz), 7.31 (IH; dd; H4,; J=6,& 2Hz), 7.67 (1H; dd;
H6,); 8.26 (2H; dd; HZ'; H6' J=2.3Hz); 8.30 (2H; dd;
H3' ; H5' J=2 . 2Hz ) ; 9 . 81 ( 1H; s; OH) ; 12 . 30 ( 1H; s;
S NH ) .
(b) 2nd Stage - Preparation of Methyl 2-(4-nitrophenyl)-benzoxazole-4-carboxylate Methyl 3-hydroxy-2-(N-4-nitrobenzoyl)aminobenzoate (O.lg; 0.34 mmol) was dissolved in m-xylene (20m1), and to this was added triethylamine (0.033m1; 0.45mmo1) and pyridinium-4-toluene sulphonate (0.0708; 0.28mmo1). This was refluxed for 32 hours. The m-xylene was removed under reduced pressure and the~remaining solid dissolved in water. The organics were extracted into ethyl acetate (3 x 30m1), dried, filtered and the solvent removed under reduced pressure to yield a brick red solid (74%).
IR . cm-1:, 1726; 1522; 1556, M/Z; 298 (84%, M+) 267 (100%, -OCH3) , 240 (-CO) 1H: CDC13: b = 4. O1 (3H; s; OCH3) , 7.44 (1H; t; H6 J=8.lHz), 7.76 (1H; dd; H7,; J=7.2,& 1Hz), 8.04 (1H; dd; H5, ) , 8. 35 (2H; dd; HZ' ; H6' J=2.2Hz) ; 8.46 (2H; d; H3'; H5' J=2.2Hz).
(c) 3rd Stage - Preparation of 2-(4-nitrophenyl) benzoxazole-4-carboxylic acid Methyl 2-(4-nitrophenyl)benzoxazole-4-carboxylate (0.1 g, 0.335 mmol) was dissolved in methanol (3 ml), and to this was added aqueous sodium hydroxide solution (0.2 M, 3 ml). The mixture was stirred at 40°C for 4 hours and acidified with hydrochloric acid (6 M) until pH -1Ø The mixture was extracted with ethyl acetate (3 x 20 ml), the combined organic layers were washed with water (2 x 20 ml), dried (MgS04) and the solvent was removed under reduced pressure to afford the carboxylic acid (0.084 g, 890).
(d) 4th Stage - Preparation of 2-(4-nitrophenyl) benzoxazole-4-carboxamide A solution of 2-(4-nitrophenyl)benzoxazole-4-carboxylic acid (0:084 g, 0.29 mmol) in anhydrous THF (10 ml) was stirred under nitrogen, and thionyl chloride (0.022 ml, 0.31 mmol), and DMF (O.lml) were added, 5 whereupon the mixture was stirred for a further 5 hours at room temperature . Aqueous ammonia ( 0 . 5 ml ) was added and the mixture was stirred for a further 30 minutes.
The solvent was removed under reduced pressure, the residual solid dissolved in water (20 ml), and the 10 solution was extracted with ethyl acetate (3 x 20 ml).
The organic layers were pooled, washed with water (2 x 20 ml) and dried (MgS04). The solvent was removed under reduced pressure to furnish the carboxamide (0.07 g, 85%).

2-(4-Methoxyphenyl)benzoxazole-4-carboxamide (Compound NU1054) (a) 1st Stage - Preparation of Methyl 3-hydroxy-2-(N-4-methoxvbenzoyl)aminobenzoate Methyl 2-amino-3-hydroxybenzoate (0.58; 2.99mmo1) was dissolved in m-xylene (40m1) with warming to 60°C.
4-Methoxybenzoyl chloride (0.5098; 2.99mmo1) was added dropwise, and this was left to stir for 3 hours. The solution was cooled to ambient temperature and the m-xylene removed under reduced pressure. The solid was dissolved in water (100m1) and the organics extracted into ethyl acetate (3 x 50m1). The organic fractions were pooled, dried over magnesium sulphate, filtered and the solvent removed under reduced pressure.
The title product was purified via chromatography (1:4 ethyl acetate: petrol as eluent) and recrystallised from boiling ethyl acetate/petrol to yield a brick red solid (33%).
IR . cm-1: 3100 (OH) 2571, 1691, 1643, 1606, M/Z; 301 (23 0, M+) , 2~70 (-OCH3) , 135 (100°s, COPhOCH3) 1H: CDC13: b - 3.87 (3H; s; OCH3) ; 3. 93 (3H; s; COOCH3) , 7. 02 (2H; dd; HS' ; H3' ) ; 7.14 (1H; t; HS J=8Hz) , 7.29 (1H;
dd; H4,; J=6.3,& l.7Hz), 7.63 (1H; dd; H6); 8.03 (2H;
dd; HZ' ; H6' ) ; 10. 38 ( 1H; s; OH) ; 11 . 98 ( 1H; s; NH) .
(b) 2nd Stage - Preparation of Methyl 2-(4-methoxyphenyl) benzoxazole-4-carboxylate Methyl 3-hydroxy-2-(N-4-methoxybenzoyl)aminobenzoate (0.058; 0.166mmo1) was dissolved in m-xylene (20m1), and to this was added triethylamine (0.016m1; 0.215mmo1) and pyridinium-4-toluene sulphonate (0.034g; 0.13mmo1). This was refluxed for 58 hours. The m-xylene was removed under reduced pressure and the remaining solid dissolved in water. The organics were extracted into ethyl acetate (3 x 30m1), dried, filtered and the solvent removed under reduced pressure to yield a solid (74%).
IR cm-1: 1718, 1614, 1502. M/Z; 283 (45%, M+) 252 (31%, -OCH3) , 225, 63 (100%) .
1H: CDC13: b = 3. 88 (3H; s; COOCH3) ; 6. 9 (2H; d; H3.; H5. ) ;
7.35 (1H; t; H6), 7.74 (1H; dd; H7;), 7.96 (1H; dd;
HS) ; 8.2 (2H; d; Hz.; H6. ) .
(c) 3rd Stage - Preparation of 2-(4-methoxyphenyl) benzoxazole-4-carboxamide Methyl 2-(4-methoxyphenyl)benzoxazole-4-carboxylate was dissolved in liquid ammonia (30m1) and sealed in an autoclave. The reaction mixture was left at 55°C, 20bar for >20 hours. Once the reaction was complete the ammonia was removed and the resulting solid recrystallised from boiling ethyl acetate and petrol.
ASSAY FOR PARP INHIBITORY ACTIVITY
Compounds of the present invention, particularly those detailed in the preceding Examples, have been tested in vitro for activity as PARP inhibitors using the following methods and materials.

In principle, the PARP assay used relies upon activating endogenous PARP (as hereinafter described) in cells containing exogenous [32P]-NAD+ introduced therein by suspending the cells in a solution of [32P]-NAD+ to which they have been rendered permeable in an initial pre-treatment step. The poly(ADP-ribose) which is then synthesised by the enzyme can be precipitated by tri-chloracetic acid (TCA) and the amount of radio-labelled 3zP incorporated therein measured, e.a. using a scintillation counter, to give a measure of the activity of the PARP under the particular conditions of the experiment. By repeating the experiment following the same procedure, and under the same conditions, in the presence of each compound to be tested the reduction in enzyme activity, represent-ative of the inhibitory effect of the test compound, can then be ascertained from the reduction, if any, of the amount of [32PJ measured in the TCA precipitated poly(ADP-ribose).
The results of this assay may be expressed in terms of percentage inhibition or reduction in activity for one or more different concentrations of each compound tested, or it may be expressed in terms of that concentration of the tested compound which reduces the enzyme activity by 500, i.e. the ICSO value. Thus, with a range of different compounds a set of comparative values for inhibitory activity can be obtained.
In practice, L1210 murine leukaemia cells were used as the source of the PARP enzyme after being rendered permeable to exogenous [3zP]NAD by exposure to hypotonic buffer and cold shock. In the preferred technique developed, which has been found to give exact and reproducible results, a defined amount of a small synthetic oligonucleotide, in particular a single strand oligo-nucleotide having the palindromic sequence _ CGGAATTCCG, is introduced into the cell suspension for activating the PARP enzyme. This oligonucleotide sequence snaps back on itself to form a double-stranded molecule with a single blunt end and provides an effective substrate for activation of PARP. Its behaviour as a potent activator of the enzyme was confirmed in the tests carried out.
The experimental protocol adopted, in which a synthetic oligonucleotide as mentioned above is introduced as a specific activator of PARP, discriminates between PARP and other mono-ADP-ribosyltransferases in the cells. Thus, introduction of such synthetic oligonucleotides causes a 5 to 6 fold stimulation in the radioactive label incorporated and this is attributable solely to PARP activity.
Further details of the assay are given below.
nn -, ~ 1. ,.. ., -, y.
The materials used included the following:
DTT (Dithiothreitol A 100mM (15.4mg/ml) solution (for use as an anti-oxidant) was made up, divided into 500u1 aliquots and stored at -20°C.
Hypotonic buffer:
9mM Hepes (214mg/100m1) 4.5o Dextran (4.5g/100m1) 4.5mM MgCl2 (92mg/100m1) The above ingredients were dissolved in about 80m1 distilled water, pH was adjusted to 7.8 (NaOH/HC1), the solution was then made up to 100m1 with distilled water, and stored in a refrigerator.
DTT was added to 5mM just before use (50u1/ml).
Isotonic buffer:
40mM Hepes (1.9g/200m1) _ 130mM KC1 (1.94g/200m1) 4o Dextran (8g/200m1) 2mM EGTA (152mg/200m1) 2.3mM MgCl2 (94mg/200m1) 225mM Sucrose (15.39g/200m1) The above ingredients were dissolved in about 150m1 distilled water, pH was adjusted to 7.8 (NaOH/HCl), the solution was then made up to 200m1 with distilled water and stored in a refrigerator.
DTT was added to 2.5mM just before use (25u1/ml).
NAD
NAD was stored as a solid in pre-weighed aliquots at -20°C. From these, solutions of a concentration of approximately 6mM (4-4.5mg/ml) were freshly made up shortly before performing an assay, and the molarity was checked by measuring the optical density (O. D.) at 260nm. The stock solution was then diluted with water to give a concentration of 600~M and a small amount of 32P labelled NAD was added (e. g. 2-5u1/ml).
Oliaonucleotide The oligonucleotide having the palindromic sequence CGGAATTCCG, synthesised by conventional means, was vacuum dried and stored as pellets in a freezer. Before use, it was made up to 200ug/ml in lOmM Tris/HC1, pH 7.8, with each pellet being dissolved completely in 50m1 of buffer. The solution was then heated to 60°C in a water bath for 15 minutes, and allowed to cool slowly to ensure correct reannealing. After adding 9.5m1 of buffer, the concentration was checked by measuring the optical density of a diluted sample at 260nm. The main solution was then diluted to a concentration of 200ug/ml and stored in 500u1 aliquots in a freezer, ready for use.
TCA
Solutions of TCA (Trichloroacetic acid) were prepared at two concentrations. loo TCA + 10%

sodium pyrophosphate, and to TCA + 1% sodium pyrophosphate:-Cells The L1210 cells used as the source of the PARP
5 enzyme were maintained as a suspension culture in RPMI medium + 10% foetal bovine serum + glutamine and antibiotics (penicillin and streptomycin).
HEPES and sodium bicarbonate were also added, and the cells were seeded in 100m1 of medium such that 10 there would be a concentration of approximately 8 x 105/ml at the time of carrying out an assay.
Mot-hnrl The compounds being tested were generally made up as a concentrated solution in DMSO (Dimethyl sulphoxide).
15 The solubility of the compound was then checked by adding a quantity of the DMSO solution to a quantity of the isotonic buffer, in the required final proportions that were to be used in carrying out the assay, and after an interval the solution was examined under a microscope for 20 any signs of crystals forming.
A desired quantity of the cells, ascertained by counting with a haemocytometer, was then centrifuged (1500rpm in a "Europa" model 24M centrifuge for 5 minutes), the supernatant removed, and the pellets obtained were resuspended in 20m1 Dul A at 4°C before centrifuging again at 1500rpm and 4°C. After again removing the supernatant, the cells were resuspended at a concentration of 3 x 107 cells/ml in ice cold hypotonic buffer and left for 30 minutes on ice. Nine volumes were then added of ice cold isotonic buffer, and the cells, now rendered permeable to exogenous NAD+, were then used within the next hour for carrying out an assay. The permeablisation of the cells may be checked at this stage by adding duplicate aliquots of cells to an equal volume of trypan blue, leaving for 5 minutes and then counting on a haemocytometer.

The assay was then carried out using for convenience plastic l5ml conical bottomed assay tubes set up in a shaking water bath at 26°C which is the optimum temperature for this enzyme. In a typical assay using the oligonucleotide solution at a concentration of 5ug/ml and the test compound/DMSO solution at a concentration of 2%, and carrying out the assay in quadruplicate, there would then be placed in each assay tube 5ul of the oligonucleotide solution, 50u1 of the 600um NAD + [3zP]-NAD solution, 8ul of the test compound/DMSO solution, and 37u1 of water. Prior to the start of the experiment this "cocktail" would be pre-warmed for 7 minutes at 26°C, as would be also the cell suspension. The reaction would then be started by adding 300u1 of the cell suspension.
The reaction would be stopped by adding 2m1 of the 10%
TCA + 10% sodium pyrophosphate solution.
In addition to the above, six assay tubes would usually be set up as blanks, these containing the same ingredients as above but, before adding the cell suspension, TCA solution is added to prevent any reaction from taking place. This enables corrections to be applied for any non-specific binding of the labelled material to the filter used (see below).
After adding the cell suspension at timed intervals to each of the assay tubes, the 10% TCA + 10% sodium pyrophosphate at 4°C was added to each assay tube exactly 5 minutes after addition of the cell suspension to that tube. Then, after leaving the tubes on ice for a minimum time of one hour, the contents of each individual tube were filtered through an individual filter funnel of a suction filter apparatus using GF/C filter elements (rough side up) wetted with 10% TCA. After filtering the contents of each tube and rinsing the filters several times with 1% TCA + 1% sodium pyrophosphate solution, the 35_ filters were carefully removed and dried before being placed in individual scintillation vials. Four additional 'scintillation vials were also set up as reference standards containing l0ul of the 600uM NAD +
[3zP]-NAD solution; lOml scintillant then being added to each vial. Counting was carried out for 2 minutes on a counter to obtain measures of the 3zP present, and thus the amount of the poly(ADP-ribose) and activity of the PARP enzyme.
RESULTS OF IN VITRO PARP INHIBITION STUDIES
Apart from applying the PARP enzyme assay in accordance with the standard procedure outlined above to a range of compounds which have been made in accordance with the present invention, for comparison purposes it was also applied to certain benzamide compounds, in particular 3-hydroxybenzamide, 3-methoxybenzamide and 3-aminobenzamide, that are already known to exhibit certain PARP inhibitory activity. A full tabulated list of the compounds which have been made and/or studied is hereinafter presented in TABLE I, together with the PARP
inhibition assay results obtained in different experiments for different concentrations of the compounds when tested using the assay hereinabove described.
In reviewing this list, the known PARP inhibitors 3-aminobenzamide, 3-methoxybenzamide and 3-hydroxybenzamide may be regarded as reference compounds. Although there is considerable variation in activity, and in some cases at least the higher concentrations for aqueous solutions of the test compounds could not be achieved because of low solubility, in general the compounds of the present invention which were tested showed a useful degree of activity. Of especial interest were the benzoxazole analogues, particularly those having the reference numbers NU1056, NU1040, NU1051 and NU1054, which showed relatively high inhibitory activity even at low concentrations.
In contrast to the results obtained for the compounds of the present invention, which have in many cases showed PARP enzyme inhibitory properties that are well above average-and at least comparable with, if not considerably better than, those of other known benzamide PARP inhibitors, various analogous nicotinamide compounds studied showed no, or very poor, inhibitory activity when tested in the same manner at similar concentrations.
FURTHER BIOLOGICAL ACTIVITY STUDIES
Again using cultures of the murine leukaemia L1210 cell line, growth inhibition experiments were carried out to assess the cytostatic effects of the compounds and clonogenic survival assays were performed to assess cytotoxicity, especially in 'relation to use of the compounds in conjunction with DNA damaging cytotoxic agents such as cytotoxic antitumour drugs or high energy radiation, e.g. gamma ray irradiation. DNA damage and the effect of the PARP inhibitors on the process of DNA
strand break formation and repair has also been assessed by carrying out DNA strand break assays and monitoring by alkaline elution in accordance with published techniques.
In the growth inhibition assays, typically the L1210 cells would be seeded at 1 x 104/ml in triplicate in 24 well multidishes, and 24 hours later the compounds or drugs being tested would be added in selected combinations and concentrations. At this time one set of replicates would be counted using a Coulter counter (No), and 48 hours later the remaining samples would be counted (N1). The percentage (%) growth inhibition of drug-treated samples could then be estimated. In drug combination experiments, where evidence of synergistic effects on cell growth or clonogenicity was being sought, a single, fixed concentration of a cytotoxic drug sample, e.g. temozolomide, would be taken as the control value.
Overall, it is believed that the studies carried out give clear evidence that the PARP inhibitory character-istics of compounds of this invention reflects an ability of these compounds to potentiate the cytotoxicity of DNA
damaging agents such as certain cytotoxic antitumour drugs and radiation used in radiotherapy. Accordingly, such compounds should be especially useful for administration in conjunction with such cytotoxic drugs or radiotherapy to potentiate their effect in the course of medical treatment as hereinbefore indicated.
Summary Although the present invention should be regarded overall as comprising each and every novel feature or combination of features disclosed herein, the main aspects of the invention comprise, principally but not exclusively, broadly the following:-(i) Novel compounds of formula (I) or (IV) as defined herein;
(ii) Compounds of formula (I) or (IV) with substituents as hereinbefore defined (including salts thereof) for therapy or for use in medicine and in the manufacture of medical preparations, useful for example as PARP
inhibitors to be administered in conjunction with cytotoxic drugs or with radiotherapy to potentiate the effectiveness of the latter in treatment of cancer;
(iii) Processes for the preparation of novel compounds of formula (I) or (IV) as defined herein, including any novel intermediate compounds produced in carrying out such processes;
(iv) Pharmaceutical formulations comprising a compound of formula (I) or (IV) as defined herein together with a pharmaceutically acceptable carrier therein;
(v) Processes for the preparation of a pharmaceutical formulation as defined in (iv) above, e.g. by methods referred to herein.

TABLE I
House Name Structure % Inhibition Number lOltM 301tM 1001.eM
Ref 3-hydroxybenzamide O 35 59 81 C~H7N0z / NHz MW = 137 \
OH
Ref 3-methoxybenzamide O 55 78 89 CgH9N02 . I \ , NHZ
MW = 151 /

Ref 3-aminobcnzarrtide O 36 63 79 C~HgN202 . / NHZ
MW= 136 . \ , , NU 1034 2,3-methylenedioxy ~ 66 benzam idc O 94 / ~ ~2 ICsp = 53~M
CgH~N03 MW = 165 O
O-NU 1040 2-t-butyibenzoxazote- O 87 88 93 4-carboxam ide / N~ ICsp = 8.4~M
C t2H t aN2~
MW = 218 gut TABLE I (contd.) House Name Structure % Inhibition Number tnmM Zn~~A,f tnn..~,~
_ _ _~ _ __ NU 1 OS 1 2-phenylbenzoxazole- O 82 4-carboxam ide C H N O . / I NHz lCso ° 2.1 ~M .

MW=238 N
. /
NU 1054 2-(4-methoxyphenyl) benzoxazote-4- ICsp = 1.1 ~M
carboxam ide C H N O

MW = 252 y ~~.
NU 1056 2-methytbenzoxazote-4- O
carboxamide ~ ICso = 9.S~M
/ ~NH2 C9HgN202 MW = l76 N

Claims (17)

1. Use of a compound for inhibiting activity of the enzyme poly(ADP-ribose)polymerase or PARP (also known as ADP-ribosyl transferase or ADPRT), such enzyme inhibition constituting an element of a therapeutic treatment, said compound providing an active PARP enzyme inhibiting agent and being a 3-substituted oxybenzamide compound having the general structural formula I

or a pharmaceutically acceptable salt thereof, wherein Y and X together form a bridge -Y-X- that represents the grouping where R5 is H, alkyl or an optionally substituted aralkyl or aryl group.
2. Use of a compound as claimed in Claim 1 wherein the or each alkyl group present in the compound, either as such or as a moiety in an alkoxy or other group, within the group R5 contains 1-6 carbon atoms.
3. Use of a compound as claimed in Claim 1 wherein the compound is a benzoxazole-4-carboxamide having a structural formula IV

wherein R5 is H, alkyl or an optionally substituted aralkyl or aryl group as specified in Claim 1.
4. Use of a compound as claimed in Claim 3 wherein R5 is selected from alkyl, phenyl, naphthyl and pyridyl.
5. Use of a compound as claimed in Claim 3 wherein R5 is a C1-6 alkyl group.
6. Use of a compound as claimed in Claim 3 wherein the compound is one of the following:
(a) 2-methylbenzoxazole-4-carboxamide;
(b) 2-t-butylbenzoxazole-4-carboxamide;
(c) 2-phenylbenzoxazole-4-carboxamide;
(d) 2-(4-methoxyphenyl)benzoxazole-4-carboxamide;
or a pharmaceutically acceptable salt of any of the above compounds (a) to (d).
7. For use as an active pharmaceutical substance, a 3-substituted oxybenzamide compound having the general structural formula I

or a pharmaceutically acceptable salt thereof, wherein Y and X together form a bridge -Y-X- that represents the grouping where R5 is H, alkyl or an optionally substituted aralkyl or aryl group.
8. A 3-substituted oxybenzamide compound having the general structural formula I

or a pharmaceutically acceptable salt thereof, wherein Y and X together form a bridge -Y-X- that represents the grouping where R5 is H, alkyl or an optionally substituted aralkyl or aryl group.
9. A compound as claimed in Claim 7 or 8 wherein the or each alkyl group present, either as such or as a moiety in an alkoxy or other group, within the group R5 contains 1-6 carbon atoms.
10. A compound as claimed in Claim 7 or 8 which is a benzoxazole-4-carboxamide compound having a structural formula IV

wherein R5 is H, alkyl or an optionally substituted aralkyl or aryl group.
11. A compound as claimed in Claim 10 wherein R5 is selected from alkyl, phenyl, naphthyl and pyridyl.
12. A compound as claimed in Claim 10 wherein R5 is a C1-6 alkyl group.
13. A compound as claimed in Claim 10 which is one of the following:
(a) 2-methylbenzoxazole-4-carboxamide;
(b) 2-t-butylbenzoxazole-4-carboxamide;
(c) 2-phenylbenzoxazole-4-carboxamide;
(d) 2-(4-methoxyphenyl)benzoxazole-4-carboxamide;
or a pharmaceutically acceptable salt of any of the above compounds (a) to (d).
14. A compound as claimed in any one of Claims 7 to 13 for use as an active PARP-inhibiting substance.
15. A pharmaceutical formulation or composition containing a compound as claimed in Claim 14 in unit dosage form made up for administration to a mammal likely to benefit from treatment with a PARP-inhibiting agent in the course of therapy.
16. A pharmaceutical formulation or composition for medical use comprising an effective PARP-inhibiting amount of a compound as claimed in any one of Claims 7 to 13 together with a pharmaceutically acceptable carrier.
17. A pharmaceutical formulation or composition as claimed in Claim 15 or 16 for use in conjunction with cytotoxic agents in antitumour therapy.
CA002350941A 1994-03-09 1995-03-09 Benzamide analogues as parp dna repair enzyme inhibitors Abandoned CA2350941A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GB9404485.6 1994-03-09
GB9404485A GB9404485D0 (en) 1994-03-09 1994-03-09 Benzamide analogues
CA002184747A CA2184747C (en) 1994-03-09 1995-03-09 Benzamide analogs, useful as parp (adp-ribosyltransferase, adprt) dna repair enzyme inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA002184747A Division CA2184747C (en) 1994-03-09 1995-03-09 Benzamide analogs, useful as parp (adp-ribosyltransferase, adprt) dna repair enzyme inhibitors

Publications (1)

Publication Number Publication Date
CA2350941A1 true CA2350941A1 (en) 1995-09-14

Family

ID=25678653

Family Applications (2)

Application Number Title Priority Date Filing Date
CA002350941A Abandoned CA2350941A1 (en) 1994-03-09 1995-03-09 Benzamide analogues as parp dna repair enzyme inhibitors
CA002352592A Expired - Fee Related CA2352592C (en) 1994-03-09 1995-03-09 Quinazolinone compounds

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA002352592A Expired - Fee Related CA2352592C (en) 1994-03-09 1995-03-09 Quinazolinone compounds

Country Status (1)

Country Link
CA (2) CA2350941A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2709618A4 (en) * 2011-05-10 2014-11-05 UNIVERSITé LAVAL Methods for the treatment and diagnostic of pulmonary arterial hypertension

Also Published As

Publication number Publication date
CA2352592C (en) 2006-06-06
CA2352592A1 (en) 1995-09-14

Similar Documents

Publication Publication Date Title
EP0879820B1 (en) Benzamide analogues, useful as PARP (ADP-Ribosyltransferase, ADPRT) DNA repair enzyme inhibitors
US6100283A (en) Benzimidazole compounds
EP3428161B1 (en) 2,3-dihydro-isoindole-1-one derivative as btk kinase suppressant, and pharmaceutical composition including same
BG107935A (en) Tetrahydropyridine derivatives, their preparation and their use as cell proliferation inhibitors
JP2572115B2 (en) 5-amino or substituted amino-1,2,3-triazoles useful as antiproliferative agents
Chan et al. Synthesis, biological evaluation, and quantitative structure-activity relationship analysis of 2-hydroxy-1H-isoindolediones as new cytostatic agents
CN113264949B (en) Design synthesis and application of spirobenzoxazine piperidine alpha, beta-unsaturated ketone derivatives
EP2051711B1 (en) Thieno[3,2-c]pyridine-7-carboxylic acid derivatives
CA2350941A1 (en) Benzamide analogues as parp dna repair enzyme inhibitors
CN110305125A (en) 5- pyrimidine -6- oxygen-Pyrazolopyridine analog derivative and its preparation method and application
CN112679409B (en) 4-indole-substituted thiosemicarbazide derivative and preparation method and application thereof
Basavarajaiah et al. Synthesis and anti-microbial activity of (Z)-4-(4-substituted-thiazol-2-yl)-1-(2-oxoindolin-3-ylidene) semicarbazide and its derivatives
CN104961648A (en) Novel acylhydrazone derivatives containing anthracene and phenanthroline, and preparation method and application thereof
CN114409603B (en) Pyrimidinone thioether compound and application thereof
CN109053624B (en) Thiazole derivative compound with IDO (indomethacin diphosphate) inhibitory activity, preparation method and application thereof
JP5433576B2 (en) Adrenergic compounds
CN115572247A (en) A retinoid K 3 Derivatives and medical use thereof
JPS63284165A (en) 5-fluorouracil derivative
ITMI970776A1 (en) DERIVATIVES OF 2, 3-BENZODIAZEPIN
CS244698B2 (en) Production method in position 9 substituted derivatives of 4-oxo-4-h-pyrido(1,2-)-pyrimidine
KR20090118025A (en) Crystal, amorphous form and salt of methyl n-[3-(6,7-dimethoxy-2-methylaminoquinazolin-4-yl)phenyl]terephthalamic acid

Legal Events

Date Code Title Description
EEER Examination request
FZDE Dead