AU695936B2 - Novel dna-targeted alkylating agents - Google Patents

Novel dna-targeted alkylating agents

Info

Publication number
AU695936B2
AU695936B2 AU32475/95A AU3247595A AU695936B2 AU 695936 B2 AU695936 B2 AU 695936B2 AU 32475/95 A AU32475/95 A AU 32475/95A AU 3247595 A AU3247595 A AU 3247595A AU 695936 B2 AU695936 B2 AU 695936B2
Authority
AU
Australia
Prior art keywords
chloroethyl
methyl
piperazinyl
benzimidazole
amino
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.)
Ceased
Application number
AU32475/95A
Other versions
AU3247595A (en
Inventor
William Alexander Denny
Jeffrey Bruce Smaill
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.)
Circadian Pharmaceuticals (Australia) Pty Ltd
Cancer Society of New Zealand Auckland Dvision
Original Assignee
Circadian Pharmaceuticals (Australia) Pty Ltd
Cancer Society of New Zealand Auckland Dvision
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 AUPM7665A external-priority patent/AUPM766594A0/en
Priority claimed from AUPN0242A external-priority patent/AUPN024294A0/en
Application filed by Circadian Pharmaceuticals (Australia) Pty Ltd, Cancer Society of New Zealand Auckland Dvision filed Critical Circadian Pharmaceuticals (Australia) Pty Ltd
Priority to AU32475/95A priority Critical patent/AU695936B2/en
Priority claimed from PCT/AU1995/000520 external-priority patent/WO1996006831A1/en
Publication of AU3247595A publication Critical patent/AU3247595A/en
Application granted granted Critical
Publication of AU695936B2 publication Critical patent/AU695936B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

NOVEL DNA-TARGETED ALKYLATING AGENTS
This invention relates to a novel class of DNA- targeted alkylating agents, to methods of preparing the novel compounds, and to the use of these compounds in the treatment of neoplastic disease. In particular, the invention relates to novel bisbenzimidazole compounds which in addition to having the ability to bind to the minor groove of deoxyribonucleic acid (DNA) have the ability to alkylate DNA. Background Of The Invention
Alkylating agents are an important class of anticancer drugs, which express their cytotoxic and
antitumour effects by forming adducts with cellular DNA. The bisbenzimidazole moiety itself has been reported in the literature as an efficient DNA minor groove binding agent, and a number of compounds of this general structure have been reported to have DNA binding and cytotoxic properties. In particular, the compound pibenzimol (Hoechst 33258), which binds to regions of the minor groove rich in adenine and thymine bases, and its analogues, have been intensively investigated. See for example:
Loewe, V. H. and Urbanietz, I.
"Basisch substituierte 2,6-Bis-benzimidazolederivate, eine neue chemotherapeutisch aktive Korperklass"
Arz.-Forschung, 1974 24 1927-1933.
Bathini, Y. and Lown, J.W.
"Convenient routes to substituted benzimidazoles and imdazolo[4,5-b]pyridines using nitrobenzene as oxidant" Synth. Comm., 1990 20 955-963.
Beerman, T.A., McHugh, M.M., Sigxαund, R., Lown, J.W.,
Rao, K.E. and Bathini, Y.
"Effects of analogs of the DNA minor groove binder Hoechst 33258 on topoisomerase II and I mediated activities"
Biochim. Biophys. Acta., 1992 1131 53-61. Kelly, D.P., Bateman, S.A., Martin, R.F., Reum, M.E., Rose, M. and Whittaker, A.R.D.
"DNA binding compounds. V. Synthesis and characterisation of boron-containing bisbenzimidazoles related to the DNA minor groove binder Hoechst 33258"
Aus. J. Chem., 1994 47 247-262.
Wang, H., Gupta, R. and Lown, J.W.
"Synthesis, DNA binding, sequence preference and biological evaluation of minor groove selective N1-alkoxyalkyl
bisbenzimidazoles"
Anti-Cancer Drug Design, 1994 9 153-180.
A recent publication [Lee, M., Walker, C.D., Eckert, J.M., Bowers, S.K., Montague, D., McAdams, S. and Hartley, J.A., "DNA sequence selective alkylation and cytotoxicity of monoheterocyclic analogues of Hoechst
33258", Med. Chem. Res., 1993 3 79-86] describes three monobenzimidazole mustard compounds. Subsequent to the priority date of this application, a further publication [Gupta, R., Wang, H., Huang, L. And Lown, J.W. "Design, synthesis, DNA sequence preferential alkylation and
biological evaluation of N-mustard derivatives of Hoechst 33258 analogues", Anti-Cancer Drug Design, 1995 10 25-41] has described four mixed benzimidazole/benzoxazole mustard analogues of Hoechst 33258.
We have now developed a novel class of bisbenzimidazole compounds which have not only the ability to bind to DNA in the minor groove, but also to alkylate DNA. These compounds show good anti-tumour activity both in vi tro and in vivo . Summary Of The Invention
According to a first aspect, the invention provides compounds of General Formula I
wherein A represents CN, NHR or any one of the formulae Ila-IId; B represents CH, N, O or S; D represents NH, NR, O or S; X represents (CH2)n, (CH2)nO, (CH2)nS, NHCO, NHCO(CH2)., CONH, CONH(CH2)n or a direct link from the nuclear
2 -position to E; E represents any one of the formulae
Illa-IIIc; m is 1 or 2, n is from 0 to 6, and R is a lower alkyl group optionally substituted with amine and/or hydroxyl functions,
together with N-oxides or pharmaceutically acceptable addition salts thereof,
with the proviso that when A is Ila (where R = Me), B is CH, D is NH, X is a direct link and E is IlIa, then Y in IlIa is not 4-N(CH2CH2Cl)2.
In formula Ila, R is a lower alkyl group optionally substituted with amine and/or hydroxyl
functions.
In formula IlIa, Y is up to two of N(CH2CH2Q)2, N(Me)CH2CH2Q, N(Et)CH2CH2Q, NO2, Cl, Br, F, OMe, Me or CONH- at positions 2 to 6, and Q is Cl, Br, I, OH or OSO2Me.
In formula Illb, G is up to two of COOR,
CH2OCONHR, CH2Q, where R is a lower alkyl group optionally substituted with amine and/or hydroxyl functions, and Q is Cl, Br, I, OH or OSO2Me, and Z is =N- or -CH=.
In formula IIIc, G is up to two of COOR, CH2OCONHR, or CH2Q, where R is a lower alkyl group
optionally substituted with amine and/or hydroxyl functions, and Q is Cl, Br, I, OH or OSO2Me, and the K groups are separately H, Me, or together form a carbocyclic ring -(CH2)3- . Examples of this type of structure have been reported [Anderson, W.K. and Corey, P.F., "Synthesis and antileukemic activity of 5-substituted 2,3-dihydro-6,7- bis(hydroxymethyl)-1H-pyrrolizine diesters", J. Med. Chem., 1977 20 812-818.; Anderson, W.K. and Hala, M.J.,
"Antileukemic activity of derivatives of 1,2-dimethyl-3,4- bis(hydroxymethyl)-5-phenylpyrrole bis(N-methylcarbamate)", J. Med. Chem., 1979 23 997-980]
For the purposes of this specification, the term "lower alkyl" is to be understood to mean an alkyl group having 1 to 4 carbon atoms.
While only some of the ring structures and substituents within the scope of General Formula I are exemplified herein, both the types of ring structure and the types of substituents are known in the art, and the person skilled in the art will readily be able to
synthesise compounds within the scope of the invention other than those specifically described herein. Similarly, such a person will readily be able to synthesize compounds of the invention in which n is 0 to 6.
The compounds of General Formula I form pharmaceutically-acceptable addition salts with both organic and inorganic acids, and these addition salts also form part of the present invention. Examples of suitable acids for salt formation are hydrochloric, sulphuric, phosphoric, acetic, citric, oxalic, malonic, salicylic, malic, fumaric, succinic, ascorbic, maleic,
methanesulfonic, isethionic and the like. Certain members of the class of compounds of formula I, containing tertiary alkyl amines, also form N-oxides of these amines, and these N-oxides also form part of the present invention.
The compounds of General Formula I have cytotoxic and anticancer activity, and are useful as antitumour agents. Therefore according to a second aspect, the invention provides a pharmaceutical composition comprising a compound of General Formula I, together with a
pharmaceutically acceptable carrier.
According to a third aspect, the invention provides a method of treatment of neoplastic disease, comprising the step of administering an effective dose of a compound of General Formula I to a mammal in need of such treatment.
Preferably the dose administered will be in the range of 1 to 200 mg/kg/body weight. This may be
administered as a single dose, or in divided doses. The compound of General Formula I may be administered by any suitable route, for example by intravenous, intramuscular, subcutaneous or intra-tumour injection, or may be
administered orally or topically.
It will be appreciated that the amount of a compound of the invention required for use in treatment will vary not only with the particular compounds selected but also with the route of administration, the nature of the condition being treated, and the age and condition of the individual being treated, and will ultimately be at the discretion of the attendant physician or veterinarian.
According to a fourth aspect, the invention provides methods of synthesis of compounds of the
invention, in accordance with the reaction schemes set out herein. It will be appreciated that the most suitable reaction scheme for synthesis of a given compound of the invention will depend upon the nature of the individual compound. A skilled person will be able to select the most suitable scheme for the desired purpose.
Thus the invention provides a method of synthesis of a compound of General Formula I, said method comprising steps selected from the group consisting of
a) converting a nitrile to a corresponding imine ether hydrochloride, and reacting said imine ether hydrochloride with a freshly prepared phenylene diamine; b) treating an acid with borane-dimethyl sulphide to yield an alcohol; converting the alcohol to an aldehyde by oxidation under Swern conditions; reacting the aldehyde with a phenylenediamine to give a
monobenzimidazole compound of formula I; converting the monobenzimidazole compound to a corresponding imine ether and coupling with a diaminobenzene derivative by cupric ion-promoted oxidation to yield a bisbenzimidazole compound of formula I;
c) converting a monomustard acid to an
aldehyde, coupling the aldehyde with a phenylenediamine to yield a monobenzimidazole compound of formula I together with the corresponding alcohol; converting the
monobenzimidazole to the corresponding imine ether, coupling the imine ether with a diaminobenzene derivative and optionally halogenating to yield a monomustard
bisbenzimidazole compound of formula I;
d) coupling a carboxylic acid derivative of an aniline mustard or aniline half-mustard with a freshly- prepared benzimidazole phenylenediamine using PPE, to yield a mustard or half-mustard bisbenzimidazole of formula I;
e) converting an imidazole dicarboxylic acid to the corresponding diethyl ester, subjecting said ester to N-alkylation with an alkyl bromide to yield a t-butyl ester derivative, selectively hydrolysing the t-butyl ester derivative to yield a carboxylic acid, converting the carboxylic acid to the acid chloride, coupling said acid chloride to a nitroaniline to yield an amide, subjecting said amide to reduction and acid-catalysed ring closure to yield a benzimidazole, coverting said benzimidazole to the corresponding diol by hydride reduction, and bis-carbamate protecting said diol; and
f) directly coupling a carboxylic acid and a freshly prepared phenylenediamine using PPE to yield a bisbenzimidazole, subjecting said benzimidazole to hydride reduction to yield a diol and biscarbamate protecting said diol. Detailed Description of the Invention
The invention will now be described in detail by way of reference only to the following non-limiting examples, and to Figure 1, which shows the results of agarose gel electrophoresis of pSV2gpt DNA treated with representative compounds of the invention or with known alkylating agents. The lanes are as follows, reading from left to right: (1) Untreated DNA, no drug; (2) Denatured DNA, no drug; (3) blank; (4-5) Denatured DNA in the presence of chloroambucil half mustard; 20, 50 μM
respectively; (6-7) Denatured DNA in the presence of chlorambucil; 20, 50 μM respectively; (8) blank; (9-11) Denatured DNA in the presence of compound 3; 0.5, 1, 2 μM respectively; (12) blank; (13-15) Denatured DNA in the presence of compound 5; 0.5, 1, 2 μM respectively; (16) blank; (17-19) Denatured DNA in the presence of compound 6; 0.5, 1, 2 μM respectively.
Abbreviations used herein are as follows:
HRMS High Resolution Mass Spectrometry
PPE polyphosphate ester
Ms methanesulphonyl
DMF dimethylformamide
THF tetrahydrofuran
The compounds of formula I and the acid addition salts and N-oxides thereof may be prepared by the processes outlined in Schemes 1-6.
Scheme 1
i H2/Rh-Al2O3/Pt-C/EtOH/glacial AcOH.
ii EtOH/HCl(g)/(0°C to 20°C)/2 days.
iii EtOH/glacial AcOH/3 h.
In Scheme 1, conversion of nitriles (IV) to the corresponding imine ether hydrochlorides (V), followed by reaction of these with freshly prepared phenylenediamine (VII), gave moderate yields (ca. 20 %) of the desired compounds of formula I (compounds 1 to 4 of Table 1).
Scheme 2
i BH3.DMS/THF/0°C.
ii DMSO/(COCl)2/THF/-78°C; Et3N/-78°C to 20°C.
iii Cu(OAc)2/MeOH(aq)/_/20 min.
iv Na2S (aq).
v Pt-C/Rh-Al2O3/60 psi H2/AcOH.
vi HCl(g)/EtOH/0°C to 20°C/2 days,
vii AcOH/100°C/2 h.
Scheme 2 outlines an alternative method of synthesis of benzimidazole derivatives representative of formula I. Treatment of acids (VIII) with borane-dimethyl sulphide gave the corresponding crude alcohols, which were in turn oxidised under Swern conditions to the aldehydes (IX). Reaction of these aldehydes with the
phenylenediamine (X) gave the desired monobenzimidazole compounds of formula I (compounds 7 to 10 of Table I).
Conversion of these compounds to the corresponding imine ethers, followed by Cu2+-promoted oxidative coupling of these with the diaminobenzene derivative (XIV) gave good yields of the desired bisbenzimidazole compounds of formula I (compounds 2, 4 to 6 of Table 1).
Scheme 3
i BH3.DMS/THF/0°C.
ii DMSO/(COCl)2/THF/-78°C; Et3N/-78°C to 20°C.
iii Cu(OAc)2 (n=3); CuCl2 (n=2 , 6) /MeOH(aq)/Δ/20 min. iv Na2S(aq).
v HCl(g)/EtOH/0°C to 20°C/2 days,
vi AcOH/100°C/3-8 h.
vii MsCl/pyridine/CH2Cl2/0°C.
viii LiCl/DMF/140°C. In Scheme 3, the monomustard acids (XV) were similarly converted to the aldehydes (XVI), which were similarly coupled to (X) to give monobenzimidazole
compounds of formula I (compounds 11 to 13 of Table 1), together with the corresponding alcohols (XIX). The use of CuCl2 as a coupling catalyst minimised hydrolysis to the alcohols. Further coupling of the imine ethers of the monobenzimidazoles with the diaminobenzene derivative
(XIV), followed by halogenation with MsCl.LiCl where necessary, gave the desired monomustard bisbenzimidazole compounds of formula I (compounds 14 to 16 of Table 1).
Scheme 4
i Pt-C/Rh-Al2O3/60 psi H2/EtOAc/MeOH
ii PPE (polyphosphate ester) /100 °C/2 h Scheme 4 outlines the preferred method of
synthesis of bisbenzimidazole mustard and bisbenzimidazole half mustard derivatives representative of formula I.
Coupling of the respective acids (XV), (XXII-XXV) with freshly prepared phenylenediamine (VII) using PPE gave the desired half mustard and mustard bisbenzimidazole compounds of formula I (compounds 17 to 21 of Table 1).
Scheme 5
i EtOH/SOCl2/reflux.
ii K2CO3/DMF/Δ.
iii HCOOH/25°C to 50°C/20 h.
iv SOCl2/DMF; XIII/THF/2 h.
v Pt-C/Rh-Al2O3/60 psi H2/EtOH; HCl(g)/0°C to
70°C/24 h.
vi LiA1H4/THF/0°C/2 h.
vii MeNCO/Bu2Sn(OAc)2/CH2Cl2.
In Scheme 5, the imidazole dicarboxylic acid (XXVI) is converted to its diethyl ester (XXVII) which is then N-alkylated with the alkyl bromide (XXVIII).
Selective hydrolysis of the resulting t-butyl ester
derivative (XXIX) gave the carboxylic acid (XXX), which was then converted to the acid chloride and coupled to the nitroaniline (XIII) to give the amide (XXXI). Reduction and acid catalysed ring closure of amide (XXXI) gave the benzimidazole (XXXII) which upon hydride reduction, and biscarbamate protection of the resulting diol, gave the desired benzimidazole compounds of formula I (compounds 22 and 23 of Table 1). Scheme 5 is a modification of Scheme 4 as described in the priority documents in respect of this application, Australian Patent Applications No. PM 7665 and PN 0242, the entire disclosures of which are incorporated herein by this reference. While Scheme 4 as previously described was successful in enabling production of the diol derivative of General Formula I, 2-[2-[N-(4,5-bishydroxymethyl)imidazolyl]ethyl]-5-(1-methyl-4- piperazinyl)benzimidazole, the yield was very low. Scheme 5 as presently described can be used to make the diol derivative.
Scheme 6
i Pt-C/Rh-Al2O3/60 psi H2/EtOAc/MeOH .
ii PPE (polyphosphate ester)/100°C/2 h.
iii LiAlH1/THF/0°C/2 h.
iv MeNCO/Bu2Sn(OAc)2/CH2Cl2.
In Scheme 6, direct coupling of the carboxylic acid (XXX) and freshly prepared phenylenediamine (VII) using PPE gave the bisbenzimidazole (XXXIII) which upon hydride reduction, and biscarbamate protection of the resulting diol, gave the desired bisbenzimidazole compounds of formula I (compounds 24 and 25 of Table 1).
Table 1 gives physicochemical data for 25
compounds within the General Formula I, representative of it, and which have been prepared by the processes of the invention.
Table 1
Structures and physicochemical properties of representative compounds of formula I
>
The following Examples illustrate the preparation of compounds representative of the general formula I.
Elemental analyses were carried out in the
Microchemical Laboratory, University of Otago. Melting points were determined on an Electrothermal apparatus using the supplied, stem-corrected thermometer, and are as read. NMR spectra were obtained on either Bruker AM-400 or CW-60 spectrometers (Me4Si). Some 13C resonances, particularly quaternary carbons of benzimidazole ring systems, proved difficult to observe and remain unassigned. Mass spectra were obtained on an AEI MS-30 spectrometer at nominal 5000 resolution. Fluorescence spectra were obtained on a Hitachi F-2000 spectrofluorimeter. The names of the compounds referred to herein which were disclosed in the priority documents in respect of this specification, Australian Provisional Patent
Applications No. PM 7665 and No. PN 0242, have been revised in order to accord with IUPAC recommendations. However, these compounds are the same as those which were disclosed in the priority documents.
Example 1 Preparation of 2-[2-[4-(N, N-bis (2 - chloroethyl)amino]phenyl]-5- benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 2 of Table 1) via the method of Scheme 1 - Example of General Procedure
Dry HCl gas was slowly bubbled into a suspension of 4-[N,N-bis(2-chloroethyl)amino]-benzonitrile (IV: n=0) [Degutis, J.; Sukelienne, D. Synthesis and some reactions of nitriles of N,N-bis(2-chloroethyl)aminophenylalkanoic acids. Zh. Obshch. Khim., 1961 31 3326-3330; Chem. Abstr., 1962, 57 3354e] (0.73 g, 3 mmol) in dry EtOH (30 mL) maintained at 0-5°C, and addition of HCl was continued until the solution became saturated. The mixture was brought to room temperature and stirred for a further 48 h, and the resulting suspension was then concentrated under reduced pressure to give the crude imine ether
hydrochloride (V, n=0) which was used without purification.
A solution of 2-(3-nitro-4-aminophenyl)-6-(1- methyl-4-piperazinyl)benzimidazole (VI) [Loewe, H.;
Urbanietz, J., "Basisch substituerte 2,6-bisbenzimidazol- derivat, eine neue chemotherapeutisch aktive korperclasse", Arzneim-Forsch. (Drug Design), 1974 24 1927-1933] (1.06 g, 3 mmol) in EtOH/glacial AcOH (1:1, 60 mL) was hydrogenated over Rh-Al2O3/Pt-C at 60 psi H2 for 12h. After this time the catalysts were removed by filtration and the solution of crude diamine (VII) was added immediately to the imine ether hydrochloride (V, n=0) prepared above. The slurry was refluxed under N2 for 3 h and concentrated under reduced pressure, and the crude residue was basified with concentrated ammonia and partitioned between EtOAc and water. The aqueous phase was repeatedly extracted with portions of EtOAc, and the combined organic phases were dried (Na2SO4), filtered and concentrated, and the residue was chromatographed on Al2O3 (grade III). Elution with
EtOAc/MeOH (99:1) afforded compound 2 of Table 1 (0.30 g, 17% yield) as a pale yellow foam. Trihydrochloride, mp (MeOH/EtOAc) >300°C. 1H NMR (CD3OD) δ 8.52 (d, J4,6 = 1.4 Hz, 1 H, H-4), 8.25 (dd, J6,7 = 8.6 Hz and J4,6 = 1.4 Hz, 1 H, H-
6), 8.11 (d, J2",3" = 9.1 Hz, 2 H, H-2",H-6"), 8.03 (d, J6,7 = 8.6 Hz, 1 H, H-7), 7.77 (d, J6',7, = 9.1 Hz, 1 H, H-7'), 7 .44 (dd, J6' ,7' = 9 . 1 Hz and J4 ' , 6' = 2 . 0 Hz , 1 H, H- 6 ' ) , 7 .37 (d, J4',6' = 2.0 Hz, 1 H, H-4'), 7.10 (d, J2",3" = 9.1 Hz, 2 H, H- 3",H-5"), 4.00 (br d, J = 11.5 Hz, 2 H, piperazinyl
methylene), 3.96 (br t, J = 6.5 Hz, 4 H, NCH2CH2Cl), 3.80 (br t, J = 6.5 Hz, 4 H, NCH2CH2Cl), 3.70 (br d, J = 12.0 Hz, 2 H, piperazinyl methylene), 3.30 (br m, 4 H,
piperazinyl methylene), 3.01 (s, 3 H, NCH3). 13C NMR δ 154.09, 153.42, 151.08, 149.02, 136.20, 134.35, 133.73,
131.44, 127.63, 126.31, 121.36, 119.94, 116.02, 115.72, 114.14, 113.99, 109.92, 100.80, 54.59, 53.85, 48.34, 43.63, 41.46. Fluorescence excitation Vmax (MeOH) 383 nm, emission λmax (MeOH) 510 nm. Anal. (C29H31N7Cl2.3HC L.1.5H2O) C,H,N,Cl.
Further elution with EtOAc/MeOH (19:1) yielded 2-
[2-methyl-5-benzimidazolyl]-6-(1-methyl-4-piperazinyl)- benzimidazole (0.31 g, 27% yield), mp (MeOH/EtOAc) >260°C (dec). 1H NMR ( (CD3) 2SO/D2O) δ 8.62 (s, 1 H, H-4), 8.28 (d, J6,7 m 8.4 Hz, 1 H, H-6), 8.03 (d, J6,7 = 8.4 Hz, 1 H, H-7), 7.74 (d, J6' ,7' = 9.1 Hz, 1 H, H-7'), 7.38 (dd, J6' ,7' = 9.1 Hz and J4 ' , 6' = 1.7 Hz, 1 H, H-6'), 7.28 (d, J4 ' , 6'. = 1.7 Hz, 1 H, H-4'), 3.94 (br d, J = 11.9 Hz, 2 H, piperazinyl
methylene), 3.61 (br d, J = 10.8 Hz, 2 H, piperazinyl methylene), 3.27 (t, J = 11.9 Hz, 2 H, piperazinyl
methylene), 3.20 (t, J = 11.4 Hz, 2 H, piperazinyl
methylene), 2.91 (s, 3 H, CH3), 2.88 (s, 3 H, CH3). 13C NMR δ 154.25, 148.61, 146.95, 133.70, 133.04, 131.44, 126.09, 124.45, 119.95, 117.22, 114.69, 114.33, 113.55, 98.64, 51.94, 46.01, 41.76, 12.60. Anal. (C20H23N6.5H2O) C,H,N.
Example 2 Preparation of 2-[2-[4-(N-methyl-N-(2- chloroethyl)amino)phenyl]-5- benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 1 of Table 1)
A solution of 4-fluorobenzonitrile (5.35 g, 44.2 mmol) in DMSO (30 mL) containing excess methylaminoethanol was heated with stirring to 120°C for 8h. The solution was then cooled and concentrated under reduced pressure. The crude residue was partitioned between H.O/EtOAc and the organic phase then washed with water (x2), dried (Na2SO4), filtered and concentrated and percolated through a pad of SiO2 (CH2Cl2 then EtOAc elution) to give 4-(N-methyl,N-
(hydroxyethyl)amino)benzonitrile (5.6 g, 72% yield) as a colourless liquid, which was used without further
purification. A solution of the above nitrile (5.6 g, 31.8 mmol) and Et3Ν (8.8 mL, 63.6 mmol) in dry CH2Cl2 (50 mL) was treated at 0°C with MsCl (3.0 mL, 38.2 mmol). The mixture was stirred for 20 min and partitioned between aqueous NaHCO3/EtOAc. The organic phase was then dried (Na2SO4), filtered and concentrated under reduced pressure to yield the crude mesylate, which was treated with excess LiCl in DMF (25 mL) at 140°C for 5 min with rapid stirring. Excess DMF was removed under reduced pressure and the residue was partitioned between H2O/EtOAc. Workup of the organic layer and chromatography of the residue on SiO2 and elution with petroleum ether/EtOAc (3:1) afforded pure 4-(N-methyl,N-(2- chloroethyl)amino)benzonitrile (3.6 g, 58% yield), mp
(petroleum ether/CH2Cl2) 104.5-105.5°C. 1H NMR (CDCl3) δ 7.48 (d, J2,3 = 9.1 Hz, 2 H, H-2,H-6), 6.67 (d, J2, 3 = 9.1 Hz, 2 H, H-3, H-5), 3.74 (complex t, J = 7.3 Hz, 2 H,
NCH2CH2Cl), 3.64 (complex t, J = 7.3 Hz, 2 H, NCH2CH2Cl) , 3.09 (s, 3 H, NCH3); 13C NMR δ 150.95, 133.66, 120.30,
111.50, 98.50, 53.77, 40.16, 39.02. Anal. (C10H11ClN2) C,H,N.
Reaction of the crude imine ether hydrochloride prepared from the above nitrile with (VII) as above, followed by chromatography on alumina (grade III) (elution with EtOAc then EtOAc/1-3% MeOH) afforded compound 1 of Table 1 (0.57 g, 13% yield). Trihydrochloride salt, mp (MeOH/EtOAc) >300°C. 1H NMR (CD3OD/D2O) δ 8.16 (d, J4, 6 = 1.6 Hz, 1 H, H-4), 8.00 (dd, J6,7 = 8.6 Hz and J4,6 = 1.6 Hz, 1 H, H- 6 ) , 7 . 87 (d, J6, 7 = 8 . 6 Hz , 1 H, H-7 ) , 7 . 82 (d, J2" , 3 " = 9.0 Hz, 2 H, H-2",H-6"), 7.68 (d, J6',7' = 8.9 Hz, 1 H, H-
7'), 7.32 (dd, J6',7' = 8.9 Hz and J4',6' = 2.1 Hz, 1 H, H-6'), 7.29 (d, J4',6' = 2.1 Hz, 1 H, H-4'), 6.80 (d, J2",3" = 9.0 Hz, 2 H, H-3" ,H 5"), 3.95 (br d, J = 12.1 Hz, 2 H, piperazinyl methylene), 3.72-3.68 (m, 6 H, NCH2CH2Cl and piperazinyl methylene), 3.31 (t, J = 12.4 Hz, 2 H, piperazinyl
methylene), 3.24 (t, J = 11.8 Hz, 2 H, piperazinyl
methylene), 3.04 (s, 3 H, NCH3), 2.99 (s, 3 H, NCH3). 13C NMR δ 154.15, 153.42, 150.44, 148.16, 136.00, 134.03,
133.53, 130.62, 127.41, 125.41, 120.26, 119.31, 115.88, 115.68, 113.17, 112.95, 108.54, 100.50, 54.45, 54.20,
48.06, 43.81, 42.00, 39.30. Fluorescence excitation λmax (MeOH) 397 nm, emission λmax (MeOH) 511 nm. Anal.
(C28H30NCl.3Cl .3H2O) C,H,N.
Example 3 Preparation of 2-[2-[[4-(N,N-bis(2- chloroethyl)amino)phenyl]methyl]-5- benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 3 of Table 1)
A similar reaction between (VII) and the crude imine ether hydrochloride (V, n=1) (prepared from the nitrile (IV, n=1)) [Degutis, J.; Sukelienne, D., "Synthesis and some reactions of nitriles of N,N-bis(2-chloroethyl)- aminophenylalkanoic acids", Zh. Obshch. Khim., 1961 31
3326-3330; Chem. Abstr., 1962 57 3354e] followed by
chromatography on Al2O3 grade III (elution with CH2Cl2 followed by CH2Cl2/MeOH (49:1)) afforded compound 3 of Table 1 (38% yield). Trihydrochloride salt, mp
(MeOH/EtOAc) >300°C. 1H NMR (CD3OD) δ 8.60 (s, 1 H, H-4), 8.28 (dd, J6 ,7= 8.7 Hz and J4 ,6 = 1.5 Hz, 1 H, H-6), 8.07 (d, J6 ,7 = 8.7 Hz, 1 H, H-7), 7.78 (d, J6',7' = 9.1 Hz, 1 H, H-7'), 7.47 (dd, J6',7' = 9.1 Hz and J4',6, = 2.0 Hz, 1 H, H- 6'), 7.39 (d, J4',6, = 2.0 Hz, 1 H, H-4'), 7.33 (d, J2",3" = 8.7 Hz, 2 H, H-2",H-6"), 6.85 (d, J2",3" = 8.7 Hz, 2 H, H- 3",H-5"), 4.53 (s, 2 H, ArCH2Ar), 4.00 (br d, J = 13.7 Hz, 2 H, piperazinyl methylene), 3.80 (br t, J = 6.4 Hz, 4 H, NCH2CH2Cl), 3.70 (br t, J = 6.5 Hz, 6 H, NCH2CH2Cl and piperazinyl methylene), 3.30 (m, 4 H, piperazinyl
methylene), 3.01 (s, 3 H, NCH3). 13C NMR δ 158.70, 151.18, 149.04, 147.78, 135.49, 134.37, 133.13, 131.80, 127.70, 126.56, 122.07, 121.81, 120.13, 116.81, 115.75, 115.33, 114.32, 100.90, 54.60, 54.38, 48.39, 43.61, 41.52, 32.99. Fluorescence excitation λmax (MeOH) 335 nm, emission λmax (MeOH) 462 nm. Anal. (C30H33N7Cl2.3HCl) C,H,N.
Example 4 Preparation of 2-[2-[2-[4-(N,N-bis(2- chloroethyl)amino)phenyl]ethyl]-5- benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 4 of Table 1)
A similar reaction between (VII) and the crude imine ether hydrochloride (V, n=2) (prepared from the nitrile (IV, n=2) ) [Degutis, J.; Jodelyte. A.; Sukelienne, D., "New synthesis of p-[bis(2-chloroethyl)amino]-b- phenylpropionic acid and its derivatives", Lietuvos TSR Aukstuju Mokyklu Mokslo Darbai, Chem. ir Chem. Tech., 1962 2 39-46; Chem. Abstr., 1963 59 8651a] followed by
chromatography on Al2O3 (grade III) (elution with EtOAc followed by EtOAc/MeOH (33:1)) gave compound 4 of Table 1 (19% yield). Trihydrochloride salt, mp (MeOH/EtOAc)
>300°C. 1H NMR (CD3OD) δ 8.60 (d, J4,6 = 1.7 Hz, 1 H, H-4), 8.28 (dd, J6 ,7= 8.7 Hz and J4,6 = 1.7 Hz, 1 H, H-6), 8.08 (d, J6 ,7= 8.7 Hz, 1 H, H-7), 7.79 (d, J6,,7' = 9.1 Hz, 1 H, H-7'), 7.48 (dd, J6',7' = 9.1 Hz and J4',6, = 2.1 Hz, 1 H, H- 6'), 7.39 (d, J4',6, = 2.1 Hz, 1 H, H-4'), 7.12 (d, J2",3" = 8.7 Hz, 2 H, H-2",H-6"), 6.72 (d, J2",3" = 8.7 Hz, 2 H, H- 3",H 5"), 4.01 (br d, J = 13.8 Hz, 2 H, piperazinyl
methylene), 3.73 (br t, J = 7.3 Hz, 4 H,NCH2CH2Cl) , 3.69 (br d, J = 12.6 Hz, 2 H, piperazinyl methylene), 3.62 (br t, J = 7.3 Hz, 4 H, NCH2CH2Cl) , 3.52 [t, J = 7.4 Hz, 2 H, ArCH2CH2C6H4N(CH2CH2Cl)2], 3.31 (broad m, 4H, piperazinyl methylene), 3.21 [t, J = 7.3 Hz, 2 H
ArCH2CH2C6H4N(CH2CH2Cl)2] , 3.01 (s, 3 H, NCH3). 13C NMR δ 158.44, 150.90, 148.98, 146.60, 135.30, 134.30, 133.00, 130.47, 128.36, 127.76, 126.25, 121.78, 120.02, 116.70, 115.78, 114.98, 113.93, 101.00, 54.54, 54.28, 48.33, 43.77, 41.74, 33.06, 30.01. Fluorescence excitation λmax (MeOH) 331 nm, emission λmax (MeOH) 449 nm. Anal.
(C31H35N7Cl2.3HCl.2H2O) C,H,N.
Example 5 Preparation of 2-[2-[4-(N,N-bis(2- chloroethyl)amino)phenyl]-5- benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 2 of Table 1) via the method of Scheme 2 -
Example of General Procedure
A solution of 4- (N,N-bis (2 -chloroethyl) amino) - benzaldehyde (IX, n=0) [Anker, R.M., Cook, A.H.,
"Quinoxaline cyanines. Part IV. Some halogenated styryl derivatives", J. Chem. Soc., 1944 489-492] (1.32 g, 5.4 mmol) in MeOH (25 mL) was added to a stirred solution of 2,3-diaminobenzonitrile (X) [Stephens, F.F., Bower, J.D., "The preparation of benzimindazoles and benzoxazoles from Schiff's bases. Part II.", J. Chem. Soc, 1950 1722-1726] (0.72 g, 5.4 mmol) in aqueous MeOH (25 mL), immediately followed by a 0.05 M aqueous solution of cupric acetate (1.1 mol equiv.). The suspension was brought briefly to the boil and filtered hot. The precipitate was washed with aqueous MeOH and redissolved in cone HCl. This acid solution was treated with an aqueous solution of Na2S
(1.5 mol equiv.) and the precipitated CuS removed by filtration. The filtrate was then basified with cone ammonia, diluted with water and repeatedly extracted with EtOAc. The combined organic extracts were dried (Na2SO4), filtered and concentrated, and the residue was
chromatographed on SiO2. Elution with CH2Cl2/EtOAc (3:1) yielded 2-[4-(N,N-bis(2-chloroethyl)amino)phenyl]-5- cyanobenzimidazole (XI, n=0) [compound 7 of Table 1] (0.80 g, 40% yield), mp (EtOAc/petroleum ether) 80°C (dec). 1H ΝMR ((CD3)2CO) δ 8.15 (d, J2,,3, = 9.0 Hz, 2 H, H-2',H-6,), 7.94 (d, J4 ,6 = 1.5 Hz, 1 H, H-4), 7.69 (d, J6 ,7= 8.5 Hz, 1 H, H-7), 7.50 (dd, J6 ,7 = 8.5 Hz and J4 ,6 = 1.5 Hz, 1 H, H- 6), 6.99 (d, J2,,3, = 9.0 Hz, 2 H, H-3',H-5'), 3.94 (br t, J = 7.5 Hz, 4 H, ΝCH2CH2Cl), 3.83 (br t, J = 7.5 Hz, 4 H, ΝCH2CH2Cl). 13C NMR δ 155.98, 149.75, 143.08, 140.50,
129.62, 126.31, 120.58, 120.13, 118.40, 116.15, 112.93, 105.47, 53.52, 41.43. Anal. (C18H16Cl2N4) C,H,N,Cl.
Dry HCl gas was slowly bubbled into a suspension of the nitrile (XI, n=0) in dry EtOH (10 mL per mmol of nitrile) maintained at 0 to 5°C, with addition of HCl being continued until the solution became saturated. The mixture was brought to room temperature and stirred for 48h, then concentrated under reduced pressure to give the crude imine ether hydrochloride (XII, n=0) which was employed without further purification.
A solution of 4-(1-methyl-4-piperazinyl)-1- nitroaniline (XIII) [Loewe, H.; Urbanietz, J. Basisch substituerte 2,6-bisbenzimidazolderivat, eine neue
chemotherapeutisch aktive korperclasse. Arzneim-Forsch. (Drug Design), 1974 24 1927-1933] (0.40 g, 1.7 mmol) in glacial AcOH (10 mL per mmol of nitroaniline) was
hydrogenated over Rh-Al2O3/Pt-C at 60 psi H2 for 12h. After this time the catalysts were removed by filtration and the resulting crude triamine (XIV) was added immediately to the imine ether hydrochloride (XII, n=0) (0.75 g, 1.7 mmol) prepared above, and the slurry was refluxed under N2 for 3h. The reaction mixture was then concentrated under reduced pressure and the crude residue was basified with cone ammonia and then partitioned between EtOAc/water. The aqueous phase was repeatedly extracted with portions of EtOAc, and the combined organic phases were dried (Na2SO4), filtered and concentrated. Chromatography of the residue as above afforded compound 2 of Table 1 (0.75 g, 74% yield).
Example 6 Preparation of 2-[2-[2-[4-(N,N-bis(2- chloroethyl)amino)phenyl]ethyl]-5- benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 4 of
Table 1)
A solution of borane dimethyl sulphide (23 mmol of a 10 M solution in THF) was added slowly to a stirred solution of 3-[4-(N,N-bis(2-chloroethyl)amino)phenyl]- propanoic acid (VIII, n=2) [Everett, J.L., Roberts, J.J., Ross W.C.J., "Aryl-2-halogenoalkylamines. Part XII. Some carboxylic acid derivatives of Ν,Ν-di-2-chloroethyl- aniline", J. Chem. Soc., 1953 2386-2392] (5.5 g, 19 mmol) in THF (190 mL) maintained at 0°C under a N2 atmosphere. After the initial effervescence the solution was stirred for a further 4h before being quenched with MeOH and concentrated under reduced pressure. The product was partitioned between aqueous NaHCO3 and EtOAc, and the residue from the organic phase was percolated through a pad of SiO2 (EtOAc elution) to give the crude alcohol, which was used without further purification. DMSO (1.15 mol equiv.) was added to a stirred solution of oxalyl chloride (1.1 mol equiv.) in THF (5 mL per mmol of alcohol)
maintained at -78°C under a N2 atmosphere. Stirring was continued for 10 min after which a solution of the above alcohol (1 mol equiv.) in THF (5 mL per mmol of alcohol) was added. Stirring was continued at -78°C for a further 15 min before Et3N (5.0 mol equiv.) was added and the mixture brought slowly to room temperature. The reaction mixture was partitioned between H2O/EtOAc and the residue from the organic layer was chromatographed on SiO2 and eluted with petroleum ether/EtOAc (3:1) to give pure 3-[4- (N,N-bis(2-chloroethyl)amino]phenyl)propanal (IX, n=2) as an unstable oil (3.4 g, 68% overall yield). 1H ΝMR (CDCl3) δ 9.80 (t, J1,2 = 1.6 Hz, 1 H, CHO), 7.08 (d, J2,,3, = 8.8 Hz, 2 H, H-2',H-6'). 6.62 (d, J2,,3, = 8.8 Hz, 2 H, H-3',H-5'),
3.69 (complex t, J = 6.5 Hz, 4 H, ΝCH2CH2Cl), 3.68 (complex t, J = 6.5 Hz, 4 H, NCH2HC2Cl) , 2.87 (t, J2,3 = 7.4 Hz, 2 H, H-3), 2.72 (br dt, J2,3 = 7.4 Hz, and J1,2 = 1.6 Hz, 2 H, H- 2), 2.12 (complex m, 4 H, H-2). 13C NMR δ 201.88, 144.56, 129.53, 129.36, 112.28, 53.54, 45.50, 40.48, 27.04. HRMS (EI, 70 eV): calcd. for C13H17NO3Cl2 273.068720. Found:
273.06881.
The above propanal (IX, n=2) was reacted with (X) as described above, followed by chromatography on silica gel and elution with CH2Cl2/EtOAc (9:1), to give the
nitrile (XI, n=2; compound 8 of Table 1) (1.6 g, 32% yield), mp (EtOAc/petroleum ether) 50°C (dec). 1H NMR ((CD3)2CO) δ 7.95 (d, J4 ,6 = 1.5 Hz, 1 H, H-4), 7.66 (d, J6 ,7 = 8.4 Hz, 1 H, H-7), 7.50 (dd, J6 ,7= 8.4 Hz and J4 ,6 = 1.5 Hz, 1 H, H-6), 7.12 (d, J2,,3, = 8.7 Hz, 2 H, H-2',H-6'),
6.70 (d, J2,,3, = 8.7 Hz, 2 H, H-3',H-5'), 3.73 (m, 8 H, NCH2CH2Cl), 3.22 (br t, J = 7.9 Hz, 2 H, benzylic
methylene), 3.10 (br t, J = 7.9 Hz, 2 H, benzylic
methylene) . 13C NMR δ 158.95, 145.86, 142.30, 140.28,
130.44, 130.26, 125.97, 120.69, 120.57, 116.08, 113.08,
105.27, 53.82, 41.58, 33.42, 31.96. Anal. (C20H20Cl2N4)
C,H,N.
Conversion of (XI, n=2) to the imino ether (XII, n=2), and condensation of this with the triamine (XIV) was carried out as described above to give compound 4 of Table
1 (1.4 g, 65% yield), which had identical properties to the sample prepared by the method of Scheme 1. Example 7 Preparation of 2-[2-[3-[4-(N,N-bis(2- chloroethyl)amino)phenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)- benzimidazole (compound 5 of Table 1)
Similar reaction of 4-[4-(N,N-bis(2-chloroethyl)- amino]phenyl)butanoic acid (VIII, n=3) [Everett, J.L., Roberts, J.J., Ross W.C.J., "Aryl-2-halogenoalkylamines. Part XII. Some carboxylic acid derivatives of N,N-di-2- chloroethylaniline", J. Chem. Soc., 1953 2386-2392]
(4.75 g, 15.6 mmol) according to the above protocol gave 4-
[4-(N,N-bis(2-chloroethyl)amino)phenyl]butanal (IX, n=3) as a pale yellow oil (3.3 g, 73% overall yield). 1H ΝMR
(CDCl3) δ 9.75 (t, J1,2 = 1.6 Hz, 1 H, CHO), 7.06 (d, J2,,3, = 8.6 H z, 2 H, H-2',H-6'), 6.63 (d, J2,,3, = 8.6 Hz, 2 H, H- 3',H-5'), 3.71 (t, J = 7.4 Hz, 4 H, ΝCH2CH2Cl), 3.63 (t, J
= 7.4 Hz, 4 H, NCH2CH2Cl) , 2.56 (t, J3 ,4 = 7.4 Hz, 2 H, H-4), 2.45 (dt, J2 ,3 = 7.3 Hz, and J1,2 = 1.6 Hz, 2 H, H-2), 1.90 (quintet, J = 7.4 Hz, 2 H, H-3). 13C NMR δ 202.52, 144.41, 130.28, 129.62, 112.21, 53.51, 43.10, 40.59, 33.82, 23.85. HRMS (EI, 70 eV) calcd for C14H19NO35Cl2 : 287.08437. Found:
287.08427.
The butanal (IX, n=3) and diamine (X) were reacted together as above, followed by chromatography on silica gel and elution with CH2Cl2/EtOAc (5:1), to give the nitrile (XI, n=3; compound 9 of Table 1) as an oil (2.1 g,
46% yield). 1H NMR ((CD3)2CO) δ 7.94 (dd, J = 0.8, 0.6 Hz, 1 H, H-4), 7.66 (dd, J6 ,7= 8.3, 0.6 Hz, 1 H, H-7), 7.49 (dd, J6 ,7 = 8.3 Hz and J4 ,6 = 1.6 Hz, 1 H, H-6), 7.10 (d, J2,,3, = 8.8 Hz, 2 H, H-2',H-6'), 6.71 (d, J2,,3, = 8.8 Hz, 2 H, H-3',H-5'), 3.79-3.69 (m, 8 H, NCH2CH2Cl) , 2.97 (t, J =
7.4 Hz, 2 H, ArCH2CH2CH2C6H4NR2), 2.64 (t, J = 7.4 Hz, 2 H, ArCH2CH2CH2C6H4NR2), 2.15 (quintet, J = 7.4 Hz, 2 H,
ArCH2CH2CH2C6H4NR2). 13C NMR δ 159.39, 145.63, 142.28,
140.41, 131.12, 130.23, 125.91, 120.63, 120.58, 116.05, 113.11, 105.20, 53.90, 41.63, 34.77, 30.40, 29.06. Anal.
(C21H22Cl2N4.H2O) C,H,N. Conversion of (XI, n=3) to the imine ether (XII, n=3) and condensation of this with (XIV) was carried out as above. Chromatography of the product on Al2O3 (grade V) and elution with EtOAc followed by EtOAc/MeOH (66:1) gave compound 5 of Table 1 as an orange oil (1.2 g, 41% yield). Trihydrochloride salt, mp (MeOH/EtOAc) >300°C. 1H NMR (CD3OD) δ 8.50 (s, 1 H, H 4), 8.26 (dd, J6,7 = 8.7 Hz and J4 ,6 = 1.7 Hz, 1 H, H-6), 7.97 (d, J6 ,7= 8.7 Hz, 1 H, H-7), 7.83 (d, J6',7' = 9.1 Hz, 1 H, H-7'), 7.48 (dd, J6',7' = 9.1 Hz and J4',6, = 2.2 Hz, 1 H, H-6'), 7.44 (d, J4',6, = 2.2 Hz, 1 H, H-4'), 7.09 (d, J2",3" = 8.6 Hz, 2 H, H-2",H-6"), 6.52 (d, J2" ,3" = 8 . 6 Hz , 2 H, H-3 " , H- 5 " ) , 4 . 02 (br d, J = 12 . 9 Hz , 2 H, piperazinyl methylene), 3.72 (br d, J = 11.4 Hz, 2 H, piperazinyl methylene), 3.54 (t, J = 6.0 Hz, 4 H,
NCH2CH2Cl), 3.46 (t, J = 6.0 Hz, 4 H, NCH2CH2Cl), 3.40-3.27 (m, 6 H, piperazinyl methylenes and ArCH2CH2CH2C6H4NR2), 3.03 (s, 3 H, NCH3), 2.79 (t, J = 6.7 Hz, 2 H,
ArCH2CH2CH2C6H4NR2), 2.41 (quintet, J = 6.3 Hz, 2 H,
ArCH2CH2CH2 C6H4NR2). 13C NMR δ 159.12, 150.93, 148.70,
145.18, 135.06, 134.19, 132.70, 130.87, 130.67, 127.59, 126.06, 121.48, 120.06, 116.46, 114.52, 113.80, 100.88, 54.59, 54.51, 48.27, 43.77, 41.73, 34.97, 28.91, 27.53.
Fluorescence excitation λmax (MeOH) 332 nm,
emission λmax (MeOH) 453 nm. Anal. (C32H37N7Cl2.3HCl.H2O) C,H,N.
Example 8 Preparation of 2-[2-[6-[4-(N,N-bis(2- chloroethyl)amino)phenyl]hexyl]-5- benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 6 of Table 1)
Similar reaction of 7-[4-(N,N-bis(2- chloroethyl)amino)phenyl]heptanoic acid (VIII, n=6) (1.50 g, 4.34 mmol) according to the above procedure yielded 7- [4-(N,N-bis(2-chloroethyl)amino)-phenyl]heptanal (IX, n=6) as an unstable colourless gum (1.34 g, 96% overall yield). 1H NMR (60 MHz) (CDCl3) δ 9.8 (t, J1,2 = 1.7 Hz, 1 H, CHO), 6.9 (d, J2,,3, = 8.9 Hz, 2 H, H-2',H-6'), 6.4 (d, J2',3 = 8.9
Hz, 2 H, H-3',H-5'), 3.6 (broad s, 8 H, NCH2CH2Cl), 2.7-2.2
(broad m, 4 H, H-2.H-7), 1.8-0.9 (broad m, 8 H, H-3,H-4,H-
5.H-6). HRMS (EI, 70 eV) calcd for C17H25NO35Cl2 : 329.13132. Found 329.13176.
Heptanal (IX, n=6) and diamine (X) were reacted together as above, followed by chromatography on silica gel and elution with CH2Cl2/EtOAc (5:1), to give the nitrile (XI, n=6; compound 10 of Table 1) (0.65 g, 40% yield) as an oil. 1H NMR ((CD3)2CO) δ 7.94 (d, J4 ,6 = 1.5
Hz, 1 H, H-4), 7.67 (d, J6,7 = 8.3 Hz, 1 H, H-7), 7.50 (dd, J6 ,7= 8.3 Hz and J4 ,6 = 1.5 Hz, 1 H, H-6), 7.05 (d, J2,,3, = 8.7 Hz, 2 H, H-2',H-6'), 6.70 (d, J2,,3, = 8.7 Hz, 2 H, H- 3',H-5'), 3.78-3.68 (m, 8 H, NCH2CH2Cl), 2.97 (t, J = 7.5 Hz, 2 H, ArCH2(CH2)5C6H4NR2), 2.49 (t, J = 7.4 Hz, 2 H,
Ar(CH2)5CH2C6H4NR2), 1.88 (quintet, J = 7.5 Hz, 2 H,
ArCH2CH2(CH2)4C6H4NR2), 1.57 (quintet, J = 7.5 Hz, 2 H
Ar(CH2)4CH2CH2C6H4NR2), 1.49-1.34 (m, 4 H,
ArCH2CH2CH2CH2CH2CH2C6H4NR2). 13C NMR δ 159.41, 145.46, 142.12, 140.12, 132.21, 130.24, 126.02, 120.59, 120.54, 116.04,
113.12, 105.35, 53.96, 41.65, 35.33, 32.37, 30.41, 29.76, 29.58, 29.57, 28.44. HRMS (El, 70 eV) calcd. for
C24H28N4 35Cl2 : 442.169103. Found : 442.16764.
Conversion of (XI, n=6) to the imino ether (XII, n=6) and condensation of this with IV was carried out as above. Chromatography of the product on Al2O3 (grade V) and elution with EtOAc followed by EtOAc/MeOH (66:1) gave compound 6 of Table 1 (0.20 g, 23% yield).
Trihydrochloride salt, mp (MeOH/EtOAc) >300°C. 1H NMR (CD3OD) δ 8.48 (d, J4 ,6 = 1.6 Hz, 1 H, H-4), 8.17 (dd, J6 ,7= 8.7 Hz and J4 ,6 = 1.6 Hz, 1 H, H-6), 8.00 (d, J6 ,7 = 8.7 Hz, 1 H, H-7), 7.71 (d, J6',7' = 9.0 Hz, 1 H, H-7'), 7.36 (dd, J6',7' = 9.0 Hz and J4',6' = 1.9 Hz, 1 H, H-6'), 7.32 (d, J4',6, = 1.9 Hz, 1 H, H-4'), 7.03 (d, J2",3" = 8.5 Hz, 2 H, H-2",H- 6"), 6.75 (d, J2",3" = 8.5 Hz, 2 H, H-3",H-5"), 3.89 (d, J = 12.4 Hz, 2 H, piperazinyl methylene), 3.66 (t, J = 6.5 Hz, 4 H, NCH2CH2Cl) , 3.59 (d, J = 11.4 Hz, 2H, piperazinyl methylene), 3.52 (t, J = 6.5 Hz, 4 H, NCH2CH2Cl) , 3.28-3.10 (complex m, 6 H, piperazinyl methylenes and
ArCH2(CH2)5C6H4NR2) , 2.90 (s, 3 H, NCH3) , 2.45 (t, J = 7.5 Hz, 2 H, Ar(CH2)5CH2C6H4NR2), 1.89 (quintet, J = 7.5 Hz, 2 H, ArCH2CH2(CH2)4C6H4NR2), 1.51 (quintet, J = 7.1 Hz, 2 H,
Ar(CH2)4CH2CH2C6H4NR2), 1.39-1.30 (m, 4 H,
ArCH2CH2CH2CH2CH2CH2C6H4NR2). 13C NMR δ 159.16, 150.86,
148.90, 143.13, 136.04, 135.20, 134.18, 132.84, 130.73, 127.63, 126.31, 121.71, 120.02, 116.75, 115.78 (two signals superimposed), 114.99, 100.97, 55.71, 54.51, 48.31, 43.79,
41.05, 35.54, 32.17, 29.71, 29.39, 27.91, 27.70.
Fluorescence excitation λmax (MeOH) 328 nm, emission λmax (MeOH) 467 nm. Anal. (C35H43N7C12.3HCl.4H2O) C,H,N.
Example 9 Preparation of 2-[2-[3-[4-(N-ethyl-N-(2- chloroethyl)amino)phenyl]propyl]-5- benzimidazolyl]-5-(l-methyl-4- piperazinyl)benzimidazole (compound 14 of Table 1) via the method of Scheme 3
4-[4-(N-Ethyl-N-(2-chloroethyl)amino)phenyl]- butanoic acid (XV, n=3) [Gourdie, T.A., Prakash, A.S.,
Wakelin, L.P.G., Woodgate, P.D., Denny, W.A., "Synthesis and evaluation of DΝA-targeted spatially-separated bis- (aniline) mustards as potential alkylating agents with enhanced DΝA cross-linking capability", J. Med. Chem., 1991 34 240-248] was converted according to the methods in the above examples to give 4-[4-(N-ethyl-N-(2-chloroethyl)- amino)-phenyl]butanal (XVI, n=3) as a colourless oil (87% overall yield). 1H ΝMR (CDCl3) δ 9.22 (t, J1,2 = 1.7 Hz, 1 H, CHO) , 7.04 (d, J2 ,3 = 8.7 Hz, 2 H, H-2',6'), 6.63 (d, J2,,3, = 8.7 Hz, 2 H, H-3',5'), 3.59 (s, 4 H, CH2CH2Cl) , 3.39
(q, J = 7.1 Hz, 2 H, CH2CH3) , 2.56 (t, J3 ,4 = 7.4 Hz, 2 H, H- 4), 2.44 (td, J1,2 = 1.7 Hz, J2 ,3 = 7.3 Hz, 2 H, H-2), 1.91 (quintet, J = 7.4 Hz, 2 H, H-3), 1.16 (t, J = 7.1 Hz, 3 H, CH2CH3). 13C ΝMR δ 202.59, 144.50, 129.42, 129.12, 52.48, 45.44, 43.16, 40.53, 33.88, 23.92, 12.49. HRMS (El, 70eV) calcd for C14H20ΝO35Cl : 253.12334. Found: 253.12281. Reaction between the butanal (XVI; n=3) and diamine (X) according to the method above gave a mixture of compounds which were dissolved in MeOH (10 mL) and stirred with KOH (10 drops, 1 M) for 20 min. Usual workup followed by chromatography on silica gel and elution with
CH2Cl2/EtOAc (1:1) gave first the chloroethyl nitrile
(XVII, n=3; compound 11 of Table 1) (91 mg, 7%) as an oil. 1H NMR (CDCl3) δ 7.86 (br s, 1 H, H-4), 7.58 (dd, J6 ,7= 8.3 Hz and J4,7 = 0.7 Hz, 1 H, H-7), 7.48 (dd, J6 ,7 = 8.3 Hz and J4 ,6 = 0.7 Hz, 1 H, H-6), 6.99 (d, J2 ,3 = 8.7 Hz, 2 H, H-
2',6'), 6.56 (d, J2,,3, = 8.7 Hz, 2 H, H-3',5'), 3.56 (s, 4 H, CH2CH2Cl) , 3.36 (q, J = 7.1 Hz, 2 H, CH2CH3) , 2.97 (t, J = 7.4 Hz, 2 H, ArCH2CH2CH2C6H4NR2) , 2.61 (t, J = 7.3 Hz, 2 H, Ar CH2CH2CH2C6H4NR2) , 2.16 (quintet, J = 7.3 Hz, 2 H,
ArCH2CH2CH2C6H4NR2 ),1.13 (t, J = 7.1 Hz, 3 H, CH2CH3). 13C
NMR δ 158.28, 145.36, 141.00, 137.50, 129.36, 128.66,
125.96, 119.89, 119.74, 115.17, 112.07, 105.08, 52.38, 45.39, 40.56, 34.04, 29.49, 28.50, 12.40. HRMS (El, 70 eV) calcd for C21H23N4 35C1 : 366.16112. Found: 366.16103. Later eluates gave the hydroxyethyl nitrile (XIX, n=3) (0.25 g,
20% overall yield) as an oil. 1H NMR (CDCl3) δ 7.82 (br, 1 H, H-4), 7.55 (d, J6 ,7 = 8.3 Hz, 1 H, H-7), 7.43 (dd, J6 ,7 = 8.3 Hz and J4 ,6 = 1.4 Hz, 1 H, H-6), 6.87 (d, J2,,3, = 8.6 Hz, 2 H, H-2',6'), 6.56 (d, J2,,3, = 8.6 Hz, 2 H, H-3',5'), 3.78 (t, J = 5.8 Hz, 2 H, CH2CH2OH) , 3.35 (m, 4 H,
CH2CH2OH,CH2CH3) , 2.85 (t, J = 7.0 Hz, 2 H,
Ar CH2CH2CH2C6H4NR2) , 2.53 (t, J = 7.0 Hz, 2 H,
ArCH2CH2CH2C6H4NR2) , 2.08 (m, 2 H, ArCH2CH2CH2C6H4NR2) , 1.07 (t, J = 7.0 Hz, 3 H, CH2CH3) . 13C NMR δ 158.73, 146.48, 146.07, 137.50, 129.08, 128.75, 125.72, 120.01, 115.15,
113.03, 111.99, 104.65, 59.71, 52.42, 45.51, 33.99, 29.61, 28.36, 11.68.
The hydroxyethyl nitrile (XIX, n=3) was converted to the corresponding imine ether hydrochloride (XX, n=3) as above, and the latter compound was reacted with (XIV) as above to give a mixture of compounds which were dissolved in MeOH (10 mL) and stirred with KOH (1 mL, 1 M) for 20 min. Usual workup, followed by chromatography on alumina (grade III) and elution with EtOAc/MeOH (93:7) gave (XXI, n=3) (30% yield). 1H NMR (CD3OD) δ 8.18 (br s, 1 H, H-4), 7.86 (dd, J6',7'= 8.5 Hz and J4 ,6 = 1.6 Hz, 1 H, H-6), 7.59 (d, J6 ,7= 8.5 Hz, 1 H, H-7), 7.48 (d, J6',7' = 8.8 Hz, 1 H,
H-7'), 7.11 (d, J4',6' = 2.2 Hz, 1 H, H-4'), 7.01 (dd, J6',7' = 8.8 Hz and J4',6, = 2.2 Hz, 1 H, H-6'), 6.98 (d, J2 ,3 = 8.7 Hz, 2 H, H-2',6'), 6.62 (d, J2 ,3 = 8.7 Hz, 2 H, H-3",5"),
3.63 (t, J = 6.6 Hz, 2 H, CH2CH2OH) , 3.32 (m, 4 H, CH2CH2OH, CH2CH3) , 3.19 (br t, J = 4.9 Hz, 4 H, piperazinyl
methylene), 2.88 (t, J = 7.8 Hz, 2 H, ArCH2CH2CH2C6H4NR2),
2.64 (br t, J = 4.9 Hz, 4 H, piperazinyl methylene), 2.57 (t, partially obscured, J = 7.3 Hz, 2 H, ArCH2CH2CH2C6H4NR2), 2.35 (s, 3 H, NCH3) , 2.07 (partially resolved quintet, J = 7.3 Hz, 2 H, ArCH2CH2CH2C6H4NR2), 1.07 (t, J = 7.0 Hz, 3 H, CH2CH3).
A solution of the above alcohol (XXI, n=3)
(92 mg, 0.17 mmol) and pyridine (0.5 mL) in dry CH2Cl2 (3 mL) was treated with excess MsCl (0.3 mL) at 0°C. The mixture was stirred for 20 min, then diluted with water and extracted with EtOAc. The combined organic extracts were washed with water, dried over anhydrous Na2SO4 and
concentrated under reduced pressure to give the crude mesylate, which was treated with excess LiCl in DMF (5 mL) at 140°C for 5min, with rapid stirring. Excess DMF was removed under reduced pressure, and the residue was
partitioned between H2O/Et0Ac Workup of the organic layer and chromatography of the residue on alumina (grade III) and elution with EtOAc/MeOH (93:7) gave compound 14 of Table 1 (45 mg, 48% yield) as a yellow oil.
Trihydrochloride salt, mp (MeOH/EtOAc) >300°C. 1H NMR (free base in CD3OD) δ 8.19 (d, J4 ,6 = 1.6 Hz, 1 H, H-4), 7.92 (dd, J6 ,7 = 8.4 Hz and J4 ,6 = 1.6 Hz, 1 H, H-6), 7.59 (d, J6 ,7= 8.4 Hz, 1 H, H-7), 7.49 (d, J6,,7, = 8.8 Hz, 1 H, H-7'), 7.12 (d, J4',6, = 2.0 Hz, 1 H, H-4'), 7.01 (partially obscured dd, J6,,7' = 8.8 Hz and J4',6, = 2.0 Hz, 1 H, H-6'), 6.99 (d, J2",3" = 8.6 Hz, 2 H, H-2",6"), 6.59 (d, J2",3" = 8.6 Hz, 2 H, H-3",5"), 3.54 (s, 4 H, CH-CH-Cl), 3.32 (partially obscured q, J = 7.0 Hz, 2 H, CH2CH3), 3.20 (br t, J = 5.0 Hz, 4 H, piperazinyl methylene), 2.88 (t, J = 7.8 Hz, 2 H, ArCH 2CH2CH2C6H4NR2) , 2 . 66 (br t, J = 5 . 0 Hz , 4 H, piperazinyl methylene), 2.58 (t, J = 7.3 Hz, 2 H, ArCH2CH2CH2C6H4NR2) ,
2.36 (s, 3 H, NCH3), 2.10 (br quintet, J = 7.4 Hz,
ArCH2CH2CH2C6H4NR2) , 1.08 (t, J = 7.0 Hz, 3 H, CH2CH3). Anal. (C32H38N7Cl.4HC l.4H2O) C,H,N.
Example 10 Preparation of 2-[2-[2-[4-(N-Ethyl-N-(2- chloroethyl)amino)phenyl]ethyl]-5- benzimidazoyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 15 of Table 1)
Similar reaction of 3-[4-(N-ethyl-N-(2- chloroethyl) amino) phenyl] propanoic acid (XV, n=2) (7.00 g,
27.4 mmol) according to the above protocol gave 3-[4-(N- ethyl-N- (2-chloroethyl) amino)phenyl] propanal (XVI, n=2) as a colourless oil (5.21 g, 79% overall yield). 1H ΝMR
(CDCl3) d 9.81 (t, J1,2 = 1.5 Hz, 1 H, CHO) , 7.05 (d, J2,,3, = 8.6 Hz, 2 H, H-2',6'), 6.63 (d, J2,,3, = 8.6 Hz, 2 H, H-
3', 5'), 3.59 (s, 4 H, CH2CH2Cl) , 3.39 (q, J = 7.0 Hz, 2 H, CH2CH3) , 2.86 (br t, J2 ,3 = 7.6 Hz, 2 H, H-3), 2.72 (m, 2 H, H-2), 1.15 (t, J = 7.0 Hz, 3 H, CH2CH3). 13C ΝMR δ 202.10, 145.45, 129.25 (2), 128.05, 112.05 (2), 52.40, 45.56, 45.39, 40.48, 27.03, 12.43. HRMS (El, 70 eV) calcd for
C13H18ΝO35Cl : 239.10769. Found: 239.10773.
Reaction of (XVI, n=2) with (X) as above, followed by chromatography on silica gel and elution with EtOAc/hexane (1:1), gave the chloroethyl nitrile (XVII, n=2; compound 12 of Table 1) (42% yield) as an oil. 1H NMR
(CDCl3) δ 7.86 (br s, 1 H, H-4), 7.56 (br d, J6 ,7= 8.3 Hz, 1 H, H-7), 7.48 (dd, J6 ,7= 8.3 Hz and J4,6 = 1.4 Hz, 1 H, H- 6), 7.01 (d, J2,,3, = 8.7 Hz, 2 H, H-2',6'), 6.59 (d, J2,,3, = 8.7 Hz, 2 H, H-3',5'), 3.57 (s, 4 H, CH2CH2Cl) , 3.38 (q, J= 7.0 Hz, 2 H, CH2CH3) , 3.24 (br t, J = 7.6 Hz, 2 H,
ArCH2CH2 C6H4NR2),3.08 (br t, J = 7.6 Hz, 2 H, Ar CH2CH2 C6H4NR2), 1.14 (t, J = 7.0 Hz, 3 H, CH2CH3). 13C NMR δ 157.73, 145.79, 129.26 (2), 127.45, 125.98, 119.93, 112.12 (2), 105.11, 52.31, 45.34, 40.50, 32.88, 31.41, 12.40. HRMS (EI, 70 eV) calcd for C20H21N4 37Cl : 354.14252. Found:
354.14332.
The nitrile (XVII, n=2) was converted to the corresponding crude imine ether hydrochloride (XVIII, n=2) and this was reacted with diamine (XIV) as above, and the resulting mixture of compounds was dissolved in MeOH (60 mL) and stirred with KOH (4 mL, 1 M) for 1 h. Usual workup followed by chromatography on alumina (grade III) and elution with EtOAc/MeOH (93:7) gave initially gave compound 15 of Table 1 (23% yield) as a gum. Trihydrochloride salt, mp (MeOH/EtOAc) >300°C. 1H NMR (free base in CD3OD) δ 8.19 (br s, 1 H, H-4), 7.91 (dd, J6 ,7= 8.4 Hz and J4,6 = 1.2 Hz, 1 H, H-6), 7.60 (d, J6 ,7= 8.4 Hz, 1 H, H-7), 7.49 (d, J6',7' = 8.8 Hz, 1 H, H-7'), 7.12 (br s, 1 H, H-4'), 7.01
(partially obscured dd, J4',6, = 2.2 Hz and J6',7' = 8.8 Hz, 1 H, H-6'), 6.99 (d, J2",3" = 8.8 Hz, 2 H, H-2",6"), 6.59 (d, J2",3" = 8.8 Hz, 2 H, H-3",5"), 3.55 (br s, 4 H, CH2CH2Cl),
3.34 (q, J = 7.0 Hz, 2 H, CH2CH3), 3.19 ( br t, J = 4.7 Hz, 4 H, piperazinyl methylene), 3.11 (m, 2 H, CH2CH2C6H4NR2), 3.01 (m, 2 H, CH2CH2C6H4NR2), 2.64 (br t, J = 4.7 Hz, 4 H, piperazinyl methylene), 2.35 (s, 3 H, NCH3), 1.08 (t, J = 7.0 Hz, 3 H, CH2CH3) . 13C NMR δ 158.48, 153.89, 149.60,
147.15, 130.28 (3), 129.61, 125.41, 122.10, 116.39, 113.59 (2), 56.19 (2), 53.64, 51.83 (2), 46.42, 46.12, 41.76, 34.48, 32.44, 12.76. Anal. (C31H36ClN7.4HCl .H2O) C,H,N,Cl.
Example 11 Preparation of 2-[2-[6-[4-(N-ethyl-N-(2- chloroethyl)amino)phenyl]hexyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)- benzimidazole (compound 16 of Table 1) Similar reaction of 7-[4-(N-ethyl-N-(2- chloroethyl)amino)phenyl]heptanoic acid (XV, n=6), (1.97 g, 6.67 mmol) gave 7-[4-(N-ethyl-N-(2-chloroethyl)amino)- phenyl]heptanal (XVI, n=6) as a colourless oil (1.40 g, 75% overall yield). 1H NMR (CDCl3) δ 9.76 (t, J1,2 = 1.6 Hz, 1 H, CHO) , 7.04 (d, J2,,3, = 8.7 Hz, 2 H, H-2',6'), 6.63 (d, J2,,3, = 8.7 Hz, 2 H, H-3',5'), 3.59 (s, 4 H, CH2CH2Cl), 3.39 (q, J = 7.0 Hz, 2 H, CH2CH3), 2.46 (br m, 4 H, H-2,7), 1.60 (br m, 4 H, H-3,6), 1.35 (br m, 4 H, H-4,H-5), 1.16 (t, J = 7.0 Hz, 3 H, CH2CH3).; 13C NMR δ 202.83, 139.40, 130.41, 129.29, 112.05, 52.57, 45.50, 43.84, 40.57, 34.66, 31.49, 29.00, 28.90, 22.00, 12.50. HRMS (El, 70 eV) calcd for C17H26NO35Cl : 295.17029. Found: 295.17100.
A solution of the above heptanal (XVI, n=6)
(100 mg, 0.36 mmol) in aqueous MeOH (20 mL) was added to a stirred solution of (X) (48 mg, 0.36 mmol) in MeOH (50 mL), immediately followed by an aqueous solution of copper (II) chloride (79 mg, 0.47 mmol) in water (10 mL). The
resulting suspension was stirred and brought to reflux for 10 min, before cooling and acidifying with cone HCl. The resulting solution was treated with a solution of Na2S (129 mg, 0.54 mmol) in water (10 mL), giving an immediate brown suspension which was filtered through a pad of celite. The filtrate was then basified with cone ammonia, diluted, and extracted with EtOAc (3x). The organic layer was washed with water, dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was chromatographed on silica gel, elution with EtOAc/hexane (2:3) giving the chloroethyl nitrile (XVII, n=6; compound 13 of Table 1) (91 mg, 62% yield) as an oil. 1H NMR (CDCl3) δ 7.91 (br s, 1 H, H-4), 7.62 (d, J6 ,7= 8.3 Hz, 1 H, H-7), 7.49 (dd, J6 ,7 = 8.3 Hz and J4 ,6 = 1.4 Hz, 1 H, H-6), 6.97 (d, J2,,3, = 8.6 Hz, 2 H, H-2',6'), 6.62 (d, J2,,3, = 8.6 Hz, 2 H, H-3',5'), 3.58 (s, 4 H, CH2CH2Cl) , 3.38 (q, J = 7.0 Hz, 2 H, CH2CH3) , 2.99 (t, J = 7.8 Hz, 2 H, ArCH2(CH2)5CsH4NR2) , 2.45 (t, J = 7.7 Hz, 2 H, Ar(CH2)5CH2C6H4NR2) , 1.89 (quintet, J = 7.8 Hz, 2 H, ArCH2CH2(CH2)4C6H4NR2) , 1.45 (br m, 6 H,
ArCH2CH2(CH2)3CH2C6H4NR2), 1.15 (t, J = 7.0 Hz, 3 H, CH2CH3) . 13C NMR δ 158.38, 144.89, 140.37, 137.80, 129.28, 126.17, 124.26, 119.75, 115.20, 112.20, 105.39, 54.63, 52.58,
45.59, 40.54, 34.60, 31.40, 29.10, 28.77, 27.86, 12.45. HRMS (EI, 70 eV) calcd for C24H29N4 37Cl 410.20512. Found : 410.20644.
The above nitrile (XVII, n=6) was converted to the corresponding crude imine ether hydrochloride (XVIII, n=6), and this was reacted with (XIV) as above to give a mixture of compounds which were dissolved in MeOH (lOmL) and stirred with KOH (1 mL, 1 M) for 20 min. Usual workup followed by chromatography on alumina (grade III) and elution with EtOAc/MeOH (93:7) gave initially the
chloroethyl bisbenzimidazole (compound 16 of Table 1) (36 mg, 5% yield) as a gum. Trihydrochloride salt, mp
(MeOH/EtOAc) >300°C. 1H NMR (free base in CD3OD) δ 8.21 (d, J4 ,6 = 1.6 Hz, 1 H, H-4), 7.93 (dd, J6 ,7= 8.5 Hz and J4 ,6 = 1.6 Hz, 1 H, H-6), 7.60 (d, J6 ,7= 8.5 Hz, 1 H, H-7), 7.48 (d, J6',7' = 8.8 Hz, 1 H, H-7'), 7.10 (d, J4',6, = 2.0 Hz, 1 H,
H-4'), 6.98 (dd, J4',6, = 2.0 Hz and J6',7' = 8.8 Hz, 1 H, H- 6'), 6.89 (d, J2 ,3 = 8.7 Hz, 2 H, H-2",6"), 6.54 (d, J2 ,3 = 8.7 Hz, 2 H, H-3",5"), 3.52 (s, 4 H, CH2CH2Cl), 3.33
(partially obscured q, J = 7.0 Hz, 2 H, CH2CH3) , 3.17 ( br t, J = 4.6 Hz, 4 H, piperazinyl methylene) 2.84 (t, J = 7.3
Hz, 2 H, ArCH2(CH2)5C6H4NR2) , 2.64 (br t, J = 4.6 Hz, 4 H, piperazinyl methylene), 2.39 (partially obscured t, J = 7.2 Hz, Ar(CH2)5CH2C6H4NR2) , 2.35 (s, 3 H, NCH3) , 1.79 (quintet, J = 7.3 Hz, 2 H, ArCH2CH2(CH2)4C6H4NR2) , 1.40 (br m, 6 H, ArCH2CH2(CH2)3CH2C6H4NR2), 1.05 (t, J = 7.0 Hz, 3 H, CH2CH3) .
Anal. (C35H44ClN7.4Cl .10H2O) C,H,N,Cl. Later eluates gave the hydroxyethyl bisbenzimidazole (XXI, n=6) (178 mg, 28% yield). 1H NMR(CD3OD) δ 8.21 (d, J4 ,6 = 1.5 Hz, 1 H, H-4), 7.94 (dd, J6 ,7 = 8.4 Hz, 1 H, H-6), 7.62 (d, J6 ,7= 8.4 Hz, 1 H, H-7), 7.50 (d, J6',7' = 8.8 Hz, 1 H, H-7'), 7.13 (d, J4',6,
= 1.9 Hz, 1 H, H-4'), 7.02 (dd, J6',7' = 8.8 Hz and J4',6,= 1.9 Hz, 1 H, H-6'), 6.91 (d, J2 ,3 = 8.6 Hz, 2 H, H-2",6"), 6.60 (d, J2",3"= 8.6 Hz, 2 H, H-3",5"), 3.64 (t, J = 6.3 Hz, 2 H, CH2CH2OH) , 3.33 (m, 4 H, CH2CH2OH, CH2CH3) , 3.21 (br t, J = 5.1 Hz, 4 H, piperazinyl methylene, 2.88 (t, J = 7.6
Hz, 2 H, ArCH2(CH2)5C6H4NR2) , 2.66 (br t, J = 5.1 Hz, 4 H, piperazinyl methylene), 2.43 (partially obscured t, J = 7.1 Hz, 2 H, Ar(CH2)5CH2C6H4NR2), 2.36 (s, 3 H, NCH3) , 1.83 (br quintet, J = 7.6 Hz, 2 H, ArCH2CH2(CH2)4C6H4NR2), 1.45 (br m, 6 H, ArCH2CH2(CH2)3CH2C6H4NR2), 1.07 (t, J = 7.0 Hz, 3 H,
CH2CH3).
The hydroxyethyl compound (XXI, n=6) was converted back to the chloroethyl bisbenzimidazole
(compound 16 of Table 1) as described above.
Example 12 Preparation of 2-[2-[[4-(N-ethyl-N-(2- chloroethyl)amino)phenyl]methyl]-5- benzimidazoyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 17 of Table 1) via the method of Scheme 4 - Example of General Procedure
The phenylenediamine (VII) (1.34 g, 4.14 mmol) was prepared by catalytic hydrogenation (60 psi, 12 h) of the corresponding nitroaniline (VI) in MeOH/EtOAc (1:2) over Pt-C/Rh-Al2O3. The filtered hydrogenation solution was concentrated in vacuo and then 2-[4-(N-ethyl-N-(2- chloroethyl)amino)phenyl]acetic acid (XV, n=1) (1.00 g, 4.14 mmol) was added, followed by a solution of PPE, as prepared in CHCl3/Et2O [Fieser and Fieser, Reagents for
Organic Synthesis, Vol.l, p, 892, John Wiley and Sons Inc., 1967] . Solvents were removed under reduced pressure, and the resulting syrup was heated at 100 °C for 2 h on an oil bath. The cooled mixture was dissolved in MeOH, diluted with water and basified with cone ammonia, then extracted with EtOAc (3X). The combined organic extracts were washed with water, dried over anhydrous Νa2SO4 and concentrated under redcued pressue, and the residue was chromatographed on alumina (grade III). Elution with EtOAc/MeOH (95:5) gave compound 17 of Table 1 (0.82 g, 38%) as a yellow oil.
Trihydrochloride salt, mp (MeOH/EtOAc) >300°C. 1H NMR
(free base in CD3OD) δ 8.18 (br s, 1 H, H-4), 7.91 (dd, J6 ,7 = 8.5 Hz and J4 ,6 = 1.6 Hz, 1 H, H-6), 7.58 (d, J6 ,7= 8.5 Hz, 1 H, H-7), 7.46 (d, J6',7' = 8.8 Hz, 1 H, H-7'), 7.12 (d, J2",3" = 8.8 Hz, 2 H, H-2",6"), 7.08 (d, J4',6,= 2.3 Hz, 1 H, H-4'), 6.98 (dd, J6',7' = 8.8 Hz and J4',6, = 2.3 Hz, 1 H, H- 6'), 6.61 (d, J2",3" = 8.8 Hz, 2 H, H-3",5"), 4.08 (s, 2 H, ArCH2Ar), 3.53 (s, 4 H, CH2CH2Cl), 3.32 (q, J = 6.9 Hz, 2 H, CH2CH3), 3.16 (br t, J = 4.6 Hz, 4 H, piperazinyl
methylene), 2.61 (br t, J = 4.6 Hz, 4 H, piperazinyl methylene), 2.32 (s, 3 H, NCH3), 1.05 (t, J = 7.0 Hz, 3 H, CH2CH3) . 13C NMR δ 158.28, 153.81, 149.53, 147.58, 140.10, 136.00, 130.86 (2), 125.43, 125.36, 122.16, 116.60, 116.33, 113.53 (2), 102.28, 56.12 (2), 53.47, 51.73 (2), 46.33, 46.07, 41.69, 35.29, 12.71. Anal. (C30H34N7Cl .3HCl .3H2O)
C,H,N.
Example 13 Preparation of 2-[2-[3-[3-(N,N-bis(2- chloroethyl)]amino)phenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole (compound 18 of
Table 1)
To a mixture of m-chlorambucil (XXII) (1.00 g, 3.29 mmol) [Palmer, B.P., Wilson, W.R., Denny, W.A., "Νitro analogues of chlorambucil as potential hypoxia-selective anti-tumour drugs", Anti-Cancer Drug Design, 1990 5, 337-
349] and freshly prepared diamine (VII) (4.00 mmol, 1.30 g) was added PPE solution (40 mL). The method described above was followed to give compound 18 of Table 1 (0.73 g, 38% yield) as a white powder, mp ca 220°C (decomp.). 1H ΝMR (CD3OD) δ 8.19 (br s, 1 H, H-4), 7.92 (dd, J6 ,7= 8.4 Hz and J4,6 = 1.1 Hz, 1 H, H-6), 7.59 (d, J6,7 = 8.2 Hz, 1 H, H-7), 7.48 (d, J6',7' = 8.8 Hz, 1 H, H-7'), 7.11 (partially
obscured s, 1 H, H-4'), 7.10 (t, J = 7.8 Hz, 1H, H-5"), 7.00 (dd, J6',7' = 8.8 Hz and J4',6, = 2.3 Hz, 1 H, H-6'), 6.57-6.50 (m, 3H, H-2", 4", 6"), 3.69-3.58 (m, 8H,
Ν(CH2CH2Cl)2), 3.18 (br t, J = 4.9 Hz, 4H, piperazinyl methylene), 2.91 (t, J = 7.8 Hz, 2H, ArCH2CH2CH2C6H4NR2), 2.64 (partially obscured t, J = 7.6 Hz, 2H,
ArCH2CH2CH2C6H4NR2), 2.62 (br t, J = 5.2 Hz, 4H, piperazinyl methylene), 2.33 (s, 3H, NCH3), 2.15 (quintet, J = 7.6 Hz,
2H, ArCH2CH2CH2C6H4NR2) . 13C NMR δ 158.77, 153.84, 149.57, 147.86, 144.06, 130.65, 125.39, 122.08, 118.78, 116.37, 113.45, 111.11, 56.16 (x2), 54.40 (x2), 51.80 (x2), 46.12, 41.71 (x2), 38.85, 30.84, 29.46. Anal. (C32H37N7Cl2.1.5H2O) C,H,N,Cl. Example 14 Preparation of 2-[2-[3-[3-(N-ethyl-N-(2- chloroethyl)amino)phenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)- benzimidazole (compound 19 of Table 1) To a mixture of m-chlorambucil half mustard
(XXIII) (0.75 g, 2.78 mmol) and freshly prepared diamine
(VII) (3.01 mmol, 0.97 g) was added PPE solution (40 mL). The method described above was followed to give compound 19 of Table 1 (1.05 g, 73% yield) as a white powder, mp ca 220°C (decomp.). 1H ΝMR (CD3OD) δ 8.19 (br s, 1 H, H-4), 7.92 (dd, J6 ,7= 8.4 Hz and J4 ,6 = 1.4 Hz, 1 H, H-6), 7.59
(br d, J6 ,7= 8.2 Hz, 1 H, H-7), 7.47 (d, J6',7' = 8.8 Hz, 1 H, H-7'), 7.11 (br s, 1 H, H-4'), 7.07 (t, J = 8.4 Hz, 1H, H-5"), 6.99 (dd, J6',7' = 8.8 Hz and J4',6, = 2.1 Hz, 1 H, H- 6'), 6.52-6.48 (m, 3H, H-2", 4", 6"), 3.57 (s, 4H,
CH2CH2Cl) , 3.37 (q, J = 7.0 Hz, 2H, CH2CH3) , 3.18 (br t, J =
4.9 Hz, 4H, piperazinyl methylene), 2.91 (t, J = 7.8 Hz, 2H, ArCH2CH2CH2C6H4ΝR2) , 2.63 (m, 6H, ArCH2CH2CH2C6H4NR2, piperazinyl methylene), 2.33 (s, 3H, NCH3) , 2.15 (quintet, J = 7.7 Hz, 2H, ArCH2CH2CH2C6H4NR2) , 1.09 (t, J = 7.0 Hz, 3H, CH2CH3). 13C NMR δ 158.82, 153.86, 149.59, 148.68, 143.74,
130.44, 125.40, 122.25, 117.92, 116.38, 113.45, 111.14, 56.17 (x2), 53.53, 51.81 (x2), 46.33, 46.12, 41.78, 36.93, 30.93, 29.50, 12.81. Anal. (C32H38N7Cl ) C,H,N,Cl.
Example 15 Preparation of 2-[2-[3-[2-(N,N-bis(2- chloroethyl)aminophenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)- benzimidazole (compound 20 of Table 1) To a mixture of o-chlorambucil (XXIV) (1.00 g, 3.29 mmol) and freshly prepared diamine (VII) (4.00 mmol, 1.30 g) was added PPE solution (40 mL). The method described above was followed to give compound 20 of Table 1 (1.27 g, 65% yield) as a white powder, mp ca 230°C
(decomp.). 1H NMR (CD3OD) δ 8.22 (br s, 1 H, H-4), 7.94 (dd, J6 ,7 = 8.5 Hz and J4 ,6 = 1.3 Hz, 1 H, H-6), 7.63 (d, J6 ,7 = 8.3 Hz, 1 H, H-7), 7.49 (d, J6',7' = 8.8 Hz, 1 H, H-7'),
7.29-7.07 (m, 5H, H-4', 3", 4", 5", 6"), 7.01 (dd, J4',6, = 2.2 Hz and J6',7' = 8.8 Hz, 1H, H-6'), 3.38 (br t, J = 6.5 Hz, 4H, CH2CH2Cl) , 3.25 (br t, J = 6.8 Hz, 4H, CH2CH2Cl) , 3.19 (br t, J = 4.9 Hz, 4H, piperazinyl methylene), 2.97 (t, J = 7.5 Hz, 2H, ArCH2CH2CH2C6H4NR2) , 2.84 (br t, J = 7.8
Hz, 2H, ArCH2CH2CH2C6H4NR2) , 2.63 (br t, J = 4.7 Hz, 4H, piperazinyl methylene), 2.34 (s, 3H, NCH3), 2.15 (quintet, J = 7.6 Hz, 2H, ArCH2CH2CH2C6H4NR2). 13C NMR δ 158.80, 153.89, 149.58, 149.12, 140.72, 130.99, 128.13, 126.66, 125.48, 125.42, 122.15, 116.38, 58.11 (x2), 56.17 (x2), 51.80 (x2),
46.11, 42.56 (x2), 30.77, 30.29, 29.99. Anal. (C32H37N7Cl2) C,H,N,Cl.
Example 16 Preparation of 2-[2-[3-[2-(N-ethyl-N-(2- chloroethyl)amino)phenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)- benzimidazole (compound 21 of Table 1)
To a mixture of o-chlorambucil half mustard (XXV) (0.75 g, 2.78 mmol) and freshly prepared diamine (VII)
(3.01 mmol, 0.97 g) was added PPE solution (40 mL). The method described above was followed to give compound 21 of Table 1 (0.74 g, 48% yield) as a white powder, mp ca 220°C (decomp.). 1H ΝMR (CD3OD) δ 8.22 (br s, 1 H, H-4), 7.94 (dd, J6 ,7= 8.5 Hz and J4,6 = 1.4 Hz, 1 H, H-6), 7.62 (d, J6 ,7 = 8.3 Hz, 1 H, H-7), 7.49 (d, J6',7' = 8.8 Hz, 1 H, H-7'), 7.25-7.00 (m, 6H, H-4', 6', 3", 4", 5", 6"), 3.37 (t, J =
6.8 Hz, 2H, ΝCH2CH2Cl), 3.18 (m, 6H, ArCH2CH2CH2C6H4NR2, piperazinyl methylene ), 2.95 (br t, J = 7.4 Hz, 2H,
NCH2CH2Cl), 2.90 (q, J = 7.1 Hz, 2H, CH2CH3) , 2.82 (br t, J = 8.1 Hz, 2H, ArCH2CH2CH2C6H4NR2) , 2.64 (br t, J = 4.5 Hz, 4H, , piperazinyl methylene), 2.34 (s, 3H, NCH3) , 2.14
(quintet, J = 7.7 Hz, 2H, ArCH2CH2CH2C6H4NR2) , 0.89 (t, J = 7.0 Hz, 3H, CH2CH3) . 13C NMR δ 158.89, 153.89, 150.20,
149.59, 140.57, 130.89, 127.88, 126.00, 125.41, 124.53, 122.09, 116.38, 57.13, 56.18 (x2), 51.81 (x2), 50.60,
46.11, 42.59, 31.07, 30.34, 29.98, 13.05. Anal. (C32H38N7Cl) C,H,N,Cl.
Example 17 Preparation of compounds 22 and 23 of
Table 1 via the method of Scheme 5
To a solution of SOCl2 (60 mL) in dry EtOH (300 mL) was added 4,5-imidazoledicarboxylic acid (XXVI)
(12.6 g, 80.7 mmol). The resulting suspension was heated under reflux for 18 h, by which time it had cleared, it was then cooled and concentrated in vacuo to give a crude white solid. The solid was dissolved in EtOAc, washed with dil. ammonia, water and brine and then dried over anhydrous Na2S04 and concentrated under reduced pressure to give
(XXVII) (12.3 g, 72% yield), mp 153-159°C. [Bauer, L., Namburg, C.N.V., Dhawan, D., J. Heterocyc. Chem., 1964 1 275-278 report mp 151- 152 °C] . 1H NMR (CDCl3) δ 7 . 91 (s , 1 H, H-2), 4.39 (q, J = 7.1 Hz, 4 H, CH2CH3), 1.36 (t, J = 7.1 Hz, 6 H, CH2CH3).
To a solution of (XXVII) (6.00 g, 28.3 mmol) in dry DMF (50 mL) was added anhydrous K2CO3 (1.1 mol eq, 4.30 g) and tert butyl-4-bromobutyrate (1.3 mol eq, 8.20 g) (XXVIII). The resulting mixture was heated at 100°C with vigorous stirring under N2 for 1 h, after which time the
DMF was evaporated under reduced pressure. The residue was then partitioned between water and EtOAc, and the residue from usual workup of the organic layer was chromatographed on silica gel. Elution with EtOAc/hexane (7:3) gave the N- alkylated imidazole (XXIX) (7.90 g, 79% yield) as a
colourless oil. 1H NMR (CDCl3, 200 MHz) δ 7.52 (s, 1 H, H- 2), 4.36 (q, J = 7.1 Hz, 4 H, CH2CH3) , 4.27 (t, J = 7.0 Hz, 2 H, NCH2), 2.21 (t, J=6.7 Hz, 2H, CH2COOtBu), 2.03 (m, 2 H, CH2CH2CH2), 1.44 (s, 9H, C(CH3)3), 1.38 (t, J = 7.1 Hz, 3 H, CH2CH3), 1.36 (t, J = 7.1 Hz, 3 H, CH2CH3). A solution of the ester (XXIX) (3.60 g,
10.2 mmol) in formic acid (80 mL) was stirred overnight with a drying tube, then warmed to 50°C and stirred for 4 h until reaction was complete by tic. The solvent was removed under reduced pressure and the crude product was
partitioned between EtOAc and water, the organic layer was washed with water and brine then dried over anhydrous
Na2SO4 and concentrated in vacuo to give a white solid which was recrystalised from toluene/hexane to give the carboxylic acid (XXX) (2.77 g, 91% yield) as white
crystals, mp 78-80°C. 1H NMR (CDCl3) δ 10.49 (br s, 1 H, COOH), 7.76 (s, 1 H, H-2), 4.39 (q, J= 7.0 Hz, 2 H,
CN2CH3) , 4.37 (q, J = 7.0 Hz, 2 H, CH2CH3) , 4.33 (t, J = 7.1 Hz, 2 H, NCH2) , 2.38 (t, J = 6.9 Hz, 2 H, CH2COOH) , 2.12 (quintet, J=7.0 Hz, 2H, CH2CH2CH2), 1.38 (t, J = 7.0 Hz, 3 H, CH2CH3) , 1.37 (t, J = 7.0 Hz, 3 H, CH2CH 3) ; 13C NMR δ 176.00, 162.05, 159.75, 139.80, 136.59, 124.51, 61.83, 61.32, 46.21, 30.34, 25.89, 14.05, 13.84. Anal. (C13H18N2O6) C,H,N.
A solution of the carboxylic acid (1.50 g, 4.62 mmol) in neat SOC12 (10 mL) with one drop of DMF added, was stirred at r.t. for 1 h, after which time the excess SOCl2 was distilled off under reduced pressure and dry benzene was added and then removed in vacuo to ensure all traces of SOCl2 had been removed. The crude acid chloride was then dissolved in dry THF (20 mL) and added dropwise via cannula to a solution of the nitroaniline (XIII) (2.0 mol eq,
9.25 mmol, 2.20 g) in dry THF (30 mL) under N2. The
addition is accompanied by formation of a yellow
precipitate which is presumably the HCl salts of the nitroaniline and product amide. The reaction was stirred for 2 h and then diluted with sat. NaHCO3 and extracted with EtOAc. The combined organic extracts were washed with water and brine, then dried over anhydrous Na2SO4 and concentrated in vacuo. Column chromatography on alumina (grade III) and elution with EtOAc gave recovered
nitroaniline (XIII) (0.80 g), further elution with EtOAc/MeOH (97:3) gave the amide (XXXI) (1.88 g, 79% yield) as a yellow oil which crystalised on standing, mp 66-68°C. 1H NMR (CDCl3) δ 11.05 (s, 1H, NHCO), 8.28 (d, J4 ,6 = 2.9 Hz, 1H, H-4), 8.14 (d, J6 ,7= 9.7 Hz, 1H, H-7), 7.62 (s, 1H, H- 2'), 6.55 (dd, J4,6 = 2.9 Hz and J6,7 = 9.7 Hz, 1H, H-6), 4.38 (m, 6H, CH2CH3 x2, CH2N), 3.51 (t, J = 3.5 Hz, 4H,
piperazinyl methylene), 2.54 (m, 6H, piperazinyl methylene, NHCOCH2), 2.35 (s, 3H, NCH3) , 2.23 (quintet, J = 7.2 Hz, 2H, CH2CH2CH2), 1.39 (t, J = 7.1 Hz, 3H, CH2CH3), 1.38 (t, J= 7.1 Hz, 3H, CH2CH3). 13C NMR δ 170.77, 162.59, 160.01, 155.46, 139.59, 137.82, 137.49, 128.53, 125.99, 124.23, 107.86, 102.70, 61.73, 61.26, 54.41 (x2), 46.53 (x2),
46.01, 45.92, 34.56, 26.19, 14.16, 13.91. Anal. (C24H32N6O7) C,H,N.
A solution of the amide (XXXI) (1.28 g,
2.48 mmol) in dry EtOH (50 mL) was hydrogenated (60 psi) over a mixed catalyst system comprising of Pt-C and Rh- Al2O3 until the solution was colourless (ca. 8 h). The solution was then filtered through celite and cooled to 0°C. HCl (g) was bubbled directly into the solution for 5 min and then the solution was warmed to 70°C for 24 h, after which time the solvent was removed at reduced
pressure, the residue was diluted with water, basified by the addition of dil. ammonia and EtOAc extracted several times. The combined orgsmic extracts were washed with water and brine, then dried over anhydrous Na2SO4 and
concentrated in vacuo. Column chromatography on alumina (grade III) and gradient elution with EtOAc to MeOH/EtOAc (9:1) gave the benzimidazole (XXXII) (0.99 g, 85% yield) as a white powder, mp 122-124°C. 1H NMR (CDCl3) δ 10.96 (br s, 1H, NH), 7.53 (s, 1H, H-2'), 7.44 (br s, 1H, H-7) , 7.02 (br s, 1H, H-4), 6.94 (dd, J4,6 = 2.2 Hz and J6,7 = 8.8 Hz, 1H, H-6), 4.32 (m, 6H, CH2CH3 x2, CH2N) , 3.17 (t, J = 5.0 Hz, 4H, piperazinyl methylene), 2.83 (t, J = 7.0 Hz, 2H,
CH2CH2CH2N) , 2.61 (t, J = 4.9 Hz, 4H, piperazinyl
methylene), 2.36 (s, 3H, NCH3), 2.35 (partially obscured quintet, J = 7.1 Hz, 2H, CH2CH2CH2), 1.33 (t, J = 7.1 Hz, 3H, CH2CH3) , 1 .32 ( t, J = 7 .1 Hz , 3H, CH2CH3) . 13C NMR δ 162.69, 159.99, 152.17, 148.01, 139.69, 137.24, 124.67, 114.43, 61.90, 61.37, 55.19 (x2), 50.94, 46.11 (x2), 46.01, 28.39, 25.36, 14.08, 13.87. Anal. (C24H32N6O4) C,H,N.
To a slurry of LiAlH4 (4 mol eq, 6.06 mmol, 0.23 g) in dry THF (5 mL) at 0°C under N2 was added dropwise a solution of the diester (XXXII) (0.71 g, 1.52 mmol) in dry THF (20 mL). After addition was complete the reaction was stirred for 2 h and then water (~ 2 mL) and 5M NaOH (~ 2 mL) were alternately added dropwise until the excess LiA1H4 had been quenched. The resulting suspension was filtered through a pad of celite which was washed with hot THF. The celite and lithium salts were then soxhlet extracted with THF for 20 h and then the combined filtrate and soxhlet extraction were concentrated at reduced pressure to give a crude white solid which was recrystalised from MeOH/EtOAc to give compound 22 of Table 1 (0.35 g, 60% yield), as a white powder, mp 197-200°C. 1H NMR (CD3OD) δ 7.64 (s, 1H, H-2'), 7.38 (d, J6,7 = 8.8 Hz, 1H, H-7), 7.04 (br s, 1H, H- 4), 6.97 (dd, J4 ,6 = 2.1 Hz and J6 ,7 = 8.8 Hz, 1H, H-6),
4.64 (s, 2H, CH2OH), 4.52 (s, 2H, CH2OH), 4.14 (t, J = 7.2 Hz, 2H, CH2N), 3.16 (t, J = 4.8 Hz, 4H, piperazinyl methylene), 2.88 (t, J = 7.4 Hz, 2H, CH2CH2CH2N) , 2.64 (t, J = 4.8 Hz, 4H, piperazinyl methylene), 2.35 (s, 3H, NCH3), 2.34 (partially obscured quintet, J = 7.3 Hz, 2H,
CH2CH2CH2). 13C NMR δ 154.94, 149.30, 140.03, 139.50, 138.36, 135.35, 129.91, 116.00, 115.98, 102.42, 57.50, 56.17 (x2), 52.77, 51.93 (x2), 46.10, 45.43, 30.12, 26.69. Anal.
(C20H28N6O2.H2O) C,H,N.
To a suspension of the diol (compound 22 of Table
1) (155 mg, 0.403 mmol) in dry CH2Cl2 (8 mL) was added methyl isocyanate (2.5 mol eq, 1.01 mmol, 42 mL) and dibutyl tin diacetate (2 drops). The suspension was stirred for 12 h and then a further two 80 mL portions of methyl isocyanate were added over 8 h. The reaction mixture cleared and appeared to be complete by tic, after which it was put directly onto an alumina (grade III) column and chromatographed with gradient elution from EtOAc to
MeOH/EtOAc (9:1) to give compound 23 of Table 1 (115 mg, 57% yield) as a white powder, mp 168-169°C. 1H NMR (CDCl3) δ 7.42 (partially obscured d, J6 ,7= 8.8 Hz, 1H, H-7), 7.41 (s, 1H, H-2'), 7.01 (br s, 1H, H-4), 6.93 (dd, J4 ,6 = 2.2 Hz and J6,7 = 8.8 Hz, 1H, H-6), 5.78 (q, J = 4.5 Hz, 1H,
CH3NHCO), 5.13 (s, 2H, CH2OCONHCH3), 5.06 (s, 2H,
CH2OCONHCH3), 4.98 (q, J = 4.6 Hz, 1H, CH-NHCO), 4.02 (t, J = 6.8 Hz, 2H, CH2N), 3.16 (t, J = 4.7 Hz, 4H, piperazinyl methylene), 2.78 (t, J = 7.4 Hz, 2H, CH2CH2CH2N), 2.73 (d, J = 4.7 Hz, 3H, CH3NHCO), 2.69 (d, J = 4.7 Hz, 3H, CH3NHCO), 2.61 (t, J = 4.9 Hz, 4H, piperazinyl methylene), 2.36 (s, 3H, NCH3), 2.32 (quintet, J = 7.2 Hz, 2H, CH2CH2CH2). 13C NMR δ 157.07 (x2), 156.64, 152.70, 148.10, 137.84, 137.33, 125.37, 114.49, 101.30, 59.40, 55.22 (x2), 54.23, 50.94 (x2), 46.03, 44.52, 28.60, 27.48 (x2), 25.67. Anal.
(C24H34N8O4) C,H,N.
Example 18 Preparation of compounds 24 and 25 of
Table 1 via the method of Scheme 6
To a mixture of carboxylic acid (XXX) (2.68 g,
8.26 mmol) and freshly prepared diamine (VII) (0.98 mol eq, 8.12 mmol, 2.62 g) was added PPE solution (90 mL). The method above (Example 12) was followed to give the
bisbenzimidazole (XXXIII) (1.94 g, 41% yield) as an orange powder, mp 143-149°C. 1H NMR (CD3OD) δ 8.19 (d, J4 ,6 = 1.6
Hz, 1 H, H-4), 7.92 (dd, J6 ,7 = 8.4 Hz and J4 ,6 = 1.6 Hz, 1 H, H-6), 7.91 (s, 1H, H-2"), 7.61 (d, J6,7 = 8.4 Hz, 1 H, H- 7), 7.48 (d, J6,,7, = 8.8 Hz, 1 H, H-7'), 7.12 (d, J4',6, = 2.2 Hz, 1 H, H-4'), 7.02 (dd, J6',7' = 8.8 Hz and J4',6, = 2.2 Hz, 1 H, H-6'), 4.37 (t, J = 7.0 Hz, 2H, CH2N), 4.30 (q, J =
7.1 Hz, 2H, CH2CH3), 4.29 (q, J = 7.1 Hz, 2H, CH2CH3), 3.23 (t, J = 4.7 Hz, 4H, piperazinyl methylene), 2.94 (t, J = 7.5 Hz, 2H, CH2CH2CH2N), 2.78 (t, J = 4.7 Hz, 4H,
piperazinyl methylene), 2.45 (s, 3H, NCH3), 2.36 (quintet, J = 7.0 Hz, 2H, CH2CH2CH2), 1.32 (t, J = 7.1 Hz, 3H, CH2CH3),
1.29 (t, J = 7.1 Hz, 3H, CH2CH3) . 13C NMR δ 163.97, 161.17, 157.17, 153.84, 149.38, 141.64, 140.35, 137.62, 136.11, 126.27, 125.52, 122.26, 116.65, 116.45, 102.46, 63.06, 62.44, 56.00 (x2), 51.46 (x2), 47.45, 45.72, 30.08, 26.75, 14.52, 14.33. Anal. (C31H36N8O4.½H20) C,H,N.
To a slurry of LiAlH4 (4 mol eq, 12.3 mmol, 0.47 g) in dry THF (20 mL) at 0°C under N2 was added dropwise a solution of the diester (XXXIII) (1.80 g, 3.08 mmol) in dry THF (100 mL). After addition was complete the reaction was stirred for 2 h and then water (~5 mL) and 5M NaOH (~5 mL) were alternately added dropwise until the excess LiAlH4 had been quenched. The resulting suspension was filtered through a pad of celite which was washed with hot THF. The celite and lithium salts were then soxhlet extracted with THF/MeOH (99:1) for 5 days, with the solvent being changed every 24 h. The combined filtrate and soxhlet extractions were concentrated at reduced pressure and the residue chromatographed on alumina (grade III) with gradient elution from MeOH/EtOAc (9:1) to MeOH/EtOAc (7:3) to give compound 24 of Table 1 (0.80 g, 52% yield) as a white solid. A small amount was recrystalised from EtOAc/MeOH to provide an analytically pure sample, mp 238-240°C. 1H NMR
(CD3OD) δ 8.20 (br s, 1 H, H-4), 7.93 (br d, J6 ,7= 8.4 Hz,
1 H, H-6), 7.67 (s, 1H, H-2"), 7.62 (br d, J6 ,7= 8.4 Hz, 1
H, H-7), 7.49 (d, J6',7' = 8.8 Hz, 1 H, H-7'), 7.12 (br s, 1 H, H-4'), 7.02 (dd, J6',7' = 8.8 Hz and J4',6, = 2.3 Hz, 1 H,
H-6'), 4.66 (s, 2H, CH2OH), 4.52 (s, 2H, CH2OH), 4.18 (t, J = 7.1 Hz, 2H, CH2N), 3.21 (br t, J = 4.5 Hz, 4H,
piperazinyl methylene), 2.96 (t, J = 7.4 Hz, 2H,
CH2CH2CH2N), 2.66 (br t, J = 4.5 Hz, 4H, piperazinyl
methylene), 2.39 (quintet, J = 7.1 Hz, 2H, CH2CH2CH2), 2.36
(s, 3H, NCH3) . 13C NMR δ 157.52, 153.79, 149.61, 140.04, 138.41, 129.89, 125.58, 122.26, 116.45, 57.49, 56.16 (x2), 52.79, 51.83 (x2), 46.10, 45.44, 29.96, 26.80. Anal.
(C27H32N8O2) C,H,N.
To a suspension of the diol (compound 24 of
Table 1) (102 mg, 0.204 mmol) in dry CH2Cl2 (10 mL) was added methyl isocyanate (2.5 mol eq, 0.509 mmol, 21 mL) and dibutyl tin diacetate (2 drops). The suspension was stirre for 6 h and then further 50 mL portions of methyl
isocyanate were added every 8 h, until the reaction mixtur cleared (~250 mL), after which it was immediately put onto an alumina (grade III) column and chromatographed with gradient elution from EtOAc to MeOH/EtOAc (6:94) to give compound 25 of Table 1 (72 mg, 57% yield) as an off white powder, 176-177ºC. 1H NMR (CD3OD) δ 8.22 (br s, 1 H, H-4), 7.94 (br d, J6 ,7= 8.2 Hz, 1 H, H-6), 7.75 (s, 1H, H-2"), 7.63 (br s, 1 H, H-7), 7.49 (d, J6',7' = 8.7 Hz, 1 H, H-7'), 7.14 (br s, 1 H, H-4'), 7.04 (dd, J6',7' = 8.8 Hz and J4',6, = 2.3 Hz, 1 H, H-6'), 5.22 (s, 2H, CH2OCONHCH3) , 5.03 (s, 2H, CH2OCONHCH3) , 4.19 (t, J = 7.2 Hz, 2H, CH2N), 3.23 (br t, = 5.0 Hz, 4H, piperazinyl methylene), 2.97 (t, J = 7.6 Hz, 2H, CH2CH2CH2N) , 2.68 (partially obscured t, J = 4.9 Hz, 4H, piperazinyl methylene), 2.66 (s, 3H, CH3NHCO) , 2.64 (s, 3H, CH3NHCO), 2.38 (s, 3H, NCH3) , 2.37 (partially obscured quintet, J ~7.3 Hz, 2H, CH2CH2CH2). HRMS (FAB, 70 eV)
Calculated for C31H39N10O4 : 615.3156. Found: 615.3130. Example 19 Interaction of Representative Compounds of the Invention with DNA
The interaction of compounds 2 to 6 with DNA was examined.
Reversible DNA binding constants and binding sit sizes for calf thymus DNA at 20°C in 0.01 M HEPES buffer, pH 7.00 were determined spectrophotometrically by Scatchar analysis, using analysis times sufficiently short that covalent alkylation reactions did not interfere. The reversible binding of these compounds is 3 to 4 fold stronger than that of pibenzimol itself, and decreases slightly across the homologous series. The binding site size remains essentially constant at 2.6±0.4 nucleotides per binding site, suggesting similar binding contacts across the series. These results are summarized in Table 2. The mustards (compounds 1 to 6) all bind 3 to 4 fold more tightly to calf thymus DNA at 0.01 ionic strength than does pibenzimol itself. Although the binding
constants decrease by a factor of nearly 2 as the linker chain is increased, the binding site size n ' remains essentially constant at 2.6 ± 0.4 nucleotides per binding site, suggesting that the mode of binding and the drug/DNA contacts which comprise it are similar, and dictated by the bisbenzimidazole chromophore.
Example 20 DNA crosslinking assay
The ability of representative compounds of the invention to bind to and cross-link DNA was examined. This was carried out by agarose gel electrophoresis of plasmid DNA using linearized pSV2gpt DNA (Tindall, K.R. and
Stankowski, L.F.; Mutation Research, 1989 220 241-245) and the method reported by Valu et al, 1990, op.cit. Briefly, linearized pSV2gpt DNA (1 μL of a 1 mg/mL solution) was incubated with drug in TE-80 buffer (10 mM Tris.HCl, 1 mM EDTA at pH 8), shielded from ambient light, for 2 h at 30°C, then denatured by the addition of 2 μL of 1% sodium dodecyl sulfate and 10 μL of 50 mM methylmercury hydroxide and incubated for a further 30 min at 20°C in the dark. Renaturation of drug- treated DNA samples was carried out by incubation with 2.5 μL of 2-mercaptoethanol for 1 h.
Samples were prepared for electrophoresis by the addition of 0.1 mg/mL bromophenol blue, 0.5 μg/mL ethidium bromide and 5 μL of 40% sucrose. Electrophoresis was carried out at 120V in 89 mM tris/borate buffer at pH 8 containing
2 mM EDTA and 0.5 μg/mL ethidium bromide. DNA was
visualized using 302 nm trans-illumination, and was
photographed using Polaroid type 55 film and a Wratten 3A filter.
An illustration of the gels is provided in
Figure 1, and a summary of the data is given in Table 3.
After drug treatment, DNA is denatured to the single stranded form, which runs as a lower band in the gels (see lanes 1 and 2 in Figure 1). Cross-linking by the drugs is evidenced by the appearance of increasing amounts of renatured, double stranded DNA, as shown by the upper band in the gels, corresponding to the band for untreated DNA seen in lane 1. The conditions used were similar to those employed in the yeast assays (see Example 17 below), and only the compound with the longest alkyl chain
(compound 6) crosslinked DNA under these conditions. As shown in Table 3, the compounds all bind strongly to DNA. The gel electrophoresis studies show that the analogues 2 to 6 do not unwind closed circular
supercoiled DNA, suggesting strongly that these compounds bind in the minor groove, as does pibenzimol. The kinetic data indicate different rates of alkylation of DNA. While the slower rates for compounds 2 and 3 are largely
explained by the lower reactivities of these mustards, the increase from compound 4 to compound 6 is probably related to their decreasing DNA binding allowing more
conformational freedom for the drug, a phenomenon observed previously with DNA-targeted alkylators. However, cross- linking was only detected for the compound of longest chain length, compound 6. It is of considerable interest that this compound was not the most effective antitumour drug, and it appears that cross-linking is not important for antitumour activity in these drugs, as shown previously for other minor groove- targeted mono-alkylating agents
(Warpehoski, M.H. et al, J. Med. Chem., 1988 31 590-603).
The rate constants for alkylation of calf thymus
DNA, determined by measuring the rate of increase in the DNA-bound fluorescence, are also given in Table 3, and show that the compounds alkylate DNA more rapidly as the chain length increases. Example 21 Anti-Tumour Activity In Vi tro
Representative examples of compounds of the invention were tested for biological activity against tumors. Compounds 1 to 6 and 14 to 21, prepared as
described above, were tested for their ability to inhibit growth in vi tro of cells of the wild-type murine leukaemia cell line P388, and the cell line AA8 which is derived from Chinese hamster ovary cells. For each compound, the IC50, ie. the concentration of compound which inhibited cell growth in treated cultures to 50% of that of controls, was determined, using means of three determinations. In addition, a hypersensitivity factor was determined by comparing activity of the compounds against AA8 cells to activity against UV4. The latter cell line is deficient in aspects of the excision repair pathway for removing DNA adducts, and is hypersensitive to DNA-alkylating agents, especially interstrand crosslinking agents. Previous work has shown that the ratio of IC50s determined in the wild- type AA8 and mutant UV4 lines (the hypersensitivity
factor HF) is a determinant of the mode of cell killing by alkylating agents (Palmer, B.D. et al, J. Med. Chem., 1990 33 112-121; Wilson, W.R. et al, J. Med. Chem., 1989 32
31-38). Compounds with HFs of ca. 1 are unlikely to kill cells via DNA alkylation events. Compounds which show high HF values are likely to form bulky adducts or cross-links, with the latter being especially likely for compounds showing the highest HF values (> ca. 25) (Wilson et al, op. ci t . ) .
Cell lines were maintained in exponential growth phase by subculturing in RPMI 1640 (P338) or Alpha MEM (AA8, UV4) containing 10% fetal calf serum, as previously described (Palmer et al, op. cit . ; Baguley, B.C. and
Wilson, W.R., Eur. J. Cancer Clin. Oncol., 1987 23
607-613). IC50 values were determined using log-phase cultures in 96-well microculture plates, and were
calculated as the nominal drug concentration required to reduce the cell density to 50% of control values, using
8 control cultures on each microplate. For P388 cultures, drug was present throughout the growth period (72 h), and final cell densities were determined using a minor
modification of the MTT method of Mossman (Mossman, T., J. Immunol. Methods, 1983 65 55-63). For AA8 and UV4
cultures, drug exposure was terminated after 18 h by washing three times with fresh medium. Cultures were then grown for a further 72 h before determining cell density by staining with methylene blue (Finlay, G.J. et al, Anal. Biochem., 1984 139 272-277). The results are summarized in Table 4.
In the present series, both of the non-alkylating compounds (pibenzimol itself and the N(Et) 2 derivative) were moderately cytotoxic against wild-type P388 leukaemia, with IC50 values of ca. 1 μM. The compounds were much less active against the AA8 cell line (although a shorter exposure time was used as described above). In other studies, pibenzimol has shown a similar IC50 value against L1210 leukaemia, but very low cytotoxicity in solid tumour lines (Finlay, G.J. and Baguley, B.C., Eur. J. Cancer, 1990 26 585-589). Both pibenzimol and the Ν(Et)2 compound had AA8/UV4 IC50 ratios (HFs) of about unity, as expected for compounds which do not act by DNA alkylation. Some
Phase I-II clinical studies with pibenzimol have been reported, but the mode of action is not known (Patel, S.K. et al, Invest. New Drugs, 1991 9 53-57; Chen, A.Y. et al, Cancer Res., 1993 53 1332-1337). The half-mustard analogue (1) showed slightly increased cytotoxicity against the wild-type P388 and
AA8 lines, but was much more effective against UV4,
resulting in an HF of 14. This is strong evidence for the mode of cytotoxicity being via the formation of DNA
adducts. The corresponding full mustard (2) had roughly similar cytotoxicities against the wild-type lines, but had an increased HF of 24. This compound has the capability of forming DNA cross-links, and the increased HF is consistent with this possibility. Compounds 3 to 6 form a series where the aniline mustard moiety is tethered to the end of an increasingly long polymethylene chain. While IC50 values in both wild-type cell lines differ little between
compounds 2 and 3, there is a large increase in
cytotoxicity (80-fold in P388, 5-fold in AA8) for the
(CH2)2-linked compound 4. This is probably not due simply to increasing lipophilicity, since the longer chain
compounds 5 and 6 are less cytotoxic.
The relative cytotoxicities of compounds 2 to 6 do suggest a positive role for the DNA-targeting
chromophore. The increased HF of compounds 3 to 5 is consistent with them forming a higher proportion of crosslinks than compound 2. The lower HF and absolute
cytotoxicity of compound 6 suggest that there may be an optimal chain length, beyond which entropic factors become important. We have noted this phenomenon previously in a series of acridine-targeted mustards (Valu, K.K. et al, J. Med. Chem., 1990 33 3014-3019).
Example 22 Anti-Tumour Activity In Vivo
In addition, the ability of the compounds to increase the percentage life span of mice bearing P338 murine leukaemia was assessed. The compound was
administered intraperitoneally, as a single dose of a solution in point 1 mL of 30% v/v ethanol/water, 24 hrs after intraperitoneal inoculation of 106 tumour cells.
There were six animals in each group. The percentage increase in life span of treated animals was determined by comparing with control animals which had been injected with an equivalent volume of normal saline. An increase in life-span of more than 20% is considered to be
statistically significant. The results are summarized in Table 5.
Figures in parenthesis represent the average number of animals in a group of six which were long-term survivors. Compound 5 was inactive in vivo at all doses up to the maximum tolerable dose.
Pibenzimol is reported to have moderate in vivo activity against L1210 leukemia on a repeated dose schedule (Chen et al , op. ci t . ) , but was not active against P388 using the single-dose protocol. However, the N(Et)2 derivative showed low but significant activity (ILS 50%). The monoalkylating half-mustard (1) was also active, but the highest activity was shown by the corresponding bis- mustard (2), with a single dose of 30 milligrams/kilogram resulting in some long-term survivors. The antitumour activity of the higher homologues decreased as their toxicity increased, but the CH2 and (CH2)2 compounds 3 and 4 retained significant in vivo activity.
It is clear from the data in Examples 21 and 22 that compounds 1 to 6 and 14 to 21, representative examples of the compounds of general formula I, are potent cytotoxic agents in vitro , and that compounds 1 to 4 also possess in vivo anticancer activity. While compound 5 was inactive in the particular assay described in Example 22, it is
understood in the art that there can be considerable differences in response between different types of in vivo assay. Other assays are known, for example using different tumour cell lines or using primary tumors.
The present study has shown that mustard analogues of the well-characterised reversible minor groove binding ligand pibenzimol have greatly increased
cytotoxicity and in vivo antitumor activity in
antiproliferative assays, compared with pibenzimol itself.
Example 23 Bacterial mutagenicity
The ability of representative compounds of the invention to induce mutations in bacteria was assayed using a standard plating method. Salmonella strains TA1977, TA1978, TA98 and TA100 were kindly supplied by B.N. Ames (Biochemistry Dept, University of California, Berkeley, California, USA). All are deep rough derivatives of the LT2 subline of S. typhimurium . Characteristics of these strains and the rationale for their selection have been previously described (Ferguson, L.R. et al , Mutation
Research, 1988 265 103-104). Since we have found that the use of aliquots of frozen stock is necessary for
reproducibility of experiments, the bacteria were initially grown to stationary phase in nutrient media and frozen (with 10% DMSO) in 1 ml aliquots at -80°C. Strains were routinely characterised for spontaneous reversion
characteristics and reversion rates in response to the following diagnostic mutagens: TA 98; daunomycin (Sigma) and 4-nitro-o-phenylene diamine (Sigma): TA100; sodium azide (BDH) : TA102; bleomycin: TA1978pKM101; mitomycin C: TA1537; 9-aminoacridine (Sigma).
For each experiment, a 1 mL vial of bacteria was removed from the -80°C freezer, inoculated into 20 mL of fresh bacterial complete medium and grown for 4 hours.
Optical density was checked at that time and at intervals thereafter until a one-in-ten dilution into fresh bacterial complete medium gave a reading of between 0.10 and 0.12 at 654 nm, to ensure that all cultures were at the same stage of growth when used. The S. typhimurittm plate
incorporation assay was carried out as described
(Maron, D.M. and Ames, B.N., Mutation Research, 1983 113 173-215). Each drug was tested over a range of
concentrations by adding varying amounts to 2 mL of soft agar containing 5 mM histidine-biotin maintained at 42°C in a temperature block. Bacterial suspension (100 μL) was added, the tube was mixed and quickly poured over the surface of agar plates containing 20 mL of minimal medium (Vogel, H.J. and Bonner, D.M., J. Biol. Chem., 1956 218
97-106). Plates were allowed to harden and then incubated at 37°C for 3 days before scoring colonies for reversion to histidine independence. All assays were performed in triplicate, and repeated at least once. Data presented are an average. Reversion characteristics of each strain were routinely tested in each experiment. Care was taken to exclude light from both the chemical and the assay plates. Colony counts were determined on an Artek Model 880
automatic counter, calibrated with plates counted manually (Maron and Ames, op.cit.).
The compounds were tested in a number of strains, and were found to be completely negative in TA98,
TA1978pKM101 and TA1537. Some compounds showed activity in strains TA100 and TA102, and dose-response curves for these are summarised in Table 6.
Results are mean values (2 experiments, plated in triplicate) ± standard deviation.
Square brackets denote that toxicity was becoming apparent on visual inspection of the plates.
The parent compound and the less reactive mustard derivatives 1-3 showed no statistically significant mutagenic response in TA100 when the data were analysed by a standard method (Mahon, G.A.T. et al, 1989, D.J. Kirkland (Ed.), Statistical evaluation of mutagenicity test data. Cambridge University, Cambridge). However, if the more toxic dose levels were not included in the analysis, compounds 4 and 5 could be considered to show a mutagenic effect in TA100. In TA102, the parent compound showed a weak but significant mutagenic effect. The more reactive mustard analogues 3-5 were also active in TA102, the last two more so than the parent compound.
None of the compounds showed any evidence of frameshift mutagenesis in GCGC regions, as shown by
inactivity in the strains TA98 and TA1978pKM101. They were also inactive against TA1978+, which was included as an example of a uvrB+ strain, a genotype which we have
previously found necessary to detect frameshift mutagenesis by cross-linking drugs such as mitomycin C (Ferguson, L.R. et al, Antitumour Drug Design, 1988 3 67-76). The
compounds were also non-mutagenic in strain TA1537
(McCoy, E.C. et al, Mutation Res., 1981 90 21-30;
Ferguson L.R. and von Borstel, R.C., Mutation Res., 1992 265 103-148 ), and thus appeared to be quite different to the series of acridine-targeted aniline mustards studied earlier (Ferguson, L.R. et al, Anticancer Drug Design, 1989b 4 209-216 and Ferguson, L.R. and Turner, P.M., Eur. J. Cancer Clin. Oncol., 1989c 24 591-596) and also to the parent aniline mustards (Ferguson, L.R. et al, Mutation Res., 1989a 224 95-104). The mutagenic ability of
compounds 4 and 5 in TA100 is consistent with DNA
monoalkylation by these compounds, as is their activity in causing weak mitotic crossing-over and some aberrant colonies in yeast. The lower bacterial mutagenicity and increased mitotic crossing-over activity of compound 6 are consistent with its DNA cross-linking capability.
Both the parent compound and several mustard analogues did show activity in strain TA102. This strain carries the hisG428 ochre (TAA) mutatton (Levin et al, 1982), and detects as mutagens a variety of oxidants and other compounds not detected by the other tester strains. The hisG428 mutation has A:T base pairs at the mutated site, while the other standard tester strains have G:C base pairs at the critical site. Both the parent compound (non- alkylating) and the mustard analogues of the mutation show similar effects, which may be due more to their high binding selectivity for AT sequences than to any alkylation event. Overall, these results support the view that the pibenzimol chromophore may be redirecting mustard
alkylation to unusual sites, presumably in the minor groove, as has been shown for other targeted mustards
(Prakash, A.S. et al, Anti-Cancer Drug Design, 1991 6
195-206). Example 24 Mitotic crossing-over and petite
mutagenesis in yeast
Many anti-cancer agents are themselves mutagenic, and therefore may give rise to other cancers as a side- effect of treatment. This may result from mutagenesis of mitochondrial DNA. The effect of representative compounds of the invention on mitochondrical mutagenesis in yeast was therefore examined.
The Saccharomyces cerevisiae diploid strain D5 (Zimmermann, F.K., Mutation Res., 1973 21 263-269) was kindly provided by Dr B.S. Cox (Botany School, University of Oxford). A single colony isolate was inoculated into liquid yeast complete medium (YC Cox, B.S. and
Bevan, E.A., New Phytol, 1962 61 342-351) and grown to stationary phase for 24 h. Dimethyl sulphoxide was added to 10%. Aliquots (1 mL) were frozen to -70°C, and stored at this temperature before use. For all experiments, the 1 mL sample was thawed, added to 10 mL of fresh medium and grown for exactly 2 hours before use.
This assay has been described in detail (Ferguson, L.R., Mutation Res., 1984 136 223-234).
Briefly, a log-phase culture was washed, then diluted into 0.87% saline. A 96-well microtitre tray (A/S Nunc,
Denmark) was inoculated with (usually) 100 μL aliquots of the diluted yeast culture, and compounds added at various dilutions to the wells, to a maximum of 1000 μg/mL.
Compounds were dissolved in dimethyl sulphoxide, and dilutions made so that there was no more than 1% DMSO in each well. Trays were incubated for either 2 hours or 20 hours at 30 °C, an appropriate dilution made from each well into saline, and 100 μL plated onto each of
10 YC plates. Cell numbers were calculated so that the dilutions at this point were at least 1/104, thereby effectively washing compounds from cells by dilution
(Ferguson, 1984, op.cit.). Plates were incubated at 30°C for 5 days, and scored for ordinary, coloured or sectored colonies by visual inspection (Zimmermann, 1973, op.cit.). They were then overlayed with tetrazolium in order to score petite colonies (Nagai, S., Science, 1959 130 1188-1189). All experiments were performed at least twice, and the data compared for reproducibility. The data presented have been pooled.
Dose response data for each of the compounds are summarised in Table 7. Compounds 1 to 3 caused no
significant activity in either of these assays. However, the more reactive mustard compounds 4 to 6 caused mitotic crossing-over, with the effect increasing with chain length; compound 6 is a moderately effective inducer of mitotic crossing-over. This was the only compound which also showed activity, albeit weak in the petite mutagenesis assays. a Survival is expressed as a percentage. The actual numbers of colonies examined are given in brackets b The indication of petite colonies at the given dose, expressed as a percentage of total surviving cells. c Mitotic crossing over, calculated as the
frequency of red/pink or red/pink/white colonies as a percentage of total surviving cells.
d The total frequency of aberrant colonies,
calculated as the number of coloured colonies as a
percentage of toal surviving cells.
Previous studies have shown that many minor groove binding drugs are very effective "petite" or
mitochondrial mutagens in yeast (Ferguson and Baguley, 1983), an activity which is also seen for both simple nitrogen mustards and those targeted to DNA by an
intercalating (acridine) carrier (Ferguson 1988, op.cit.; 1989, op.cit.; 1991, op.cit.). It is therefore of
considerable interest that both the parent compound and most of the analogues are not active in this system.
Although our results do not rule out some mitochondrial effects for these compounds - for example, some types of mitochondrial enzyme inhibition will not be detected in the petite mutagenesis assay - none of the present series appears to act similarly to the bisquaternary ammonium heterocycle class of minor groove binders (Ferguson and Baguley, 1983, op.cit.). Thus the most effective
antitumour agent, the monoalkylator compound 2, was
inactive as a bacterial mutagen and as a mitochondrial mutagen in yeast.
The weak mutagenic activity of the
bisbenzimidazole-derived mustards of the invention suggests that, unlike many alkylating agents, they may be less likely to be potential carcinogens (Quinto, I. and
Radman, M., Mutation Res., 1987 181 235-242; Schmal, D., IRAC Sci. Publ., 1986 78 143-146; Pederson-Bjergaard, J. et al, Cancer Res., 1989 48 1812-1817).
It will be apparent to the person skilled in the art that while the invention has been described in some detail for the purposes of clarity and understanding, various modifications and alterations to the embodiments and methods described herein may be made without departing from the scope of the inventive concept disclosed in this specification.

Claims (14)

CLAIMS :
1. A compound of general formula I
wherein A represents CN, NHR or any one of the formulae Ila-IId; B represents CH, N, O or S; D represents NH, NR, O or S; X represents (CH2)n, (CH2)nO, (CH2)nS, NHCO, NHCO(CH2)n, CONH, CONH(CH2)n or a direct link from the nuclear
2-position to E; E represents any one of the formulae
Illa-IIIc; m is 1 or 2, n is from 0 to 6, and R is a lower alkyl group optionally substituted with amine and/or hydroxyl functions,
together with N-oxides or pharmaceutically acceptable addition salts thereof,
with the proviso that when A is Ila (where R = Me), B is CH, D is NH, X is a direct link and E is IlIa, then Y in IlIa is not 4-N(CH2CH2Cl)2.
2. A compound according to Claim 1, wherein A represents CN, NHR, or any one of the formulae (Ila-IId).
3. A compound according to Claim 2, wherein A is CN or formula Ila.
4. A compound according to any one of Claims 1 to 3, wherein X represents (CH2)n and n is from 0 to 6.
5. A compound according to any one of Claims 1 to 4, wherein E is formula IlIa or Illb.
6. A compound according to any one of Claims 1 to 5, wherein G in formula IIIc is up to 2 of CH2OCONHR or CH2Q.
7. A compound according to Claim 1 selected from the group consisting of 2-[2-[4-(N-methyl-N-(2- chloroethyl)amino)phenyl]-5-benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole, 2-[2-[4-(N, N-bis (2 - chloroethyl)amino)phenyl]-5-benzimidazolyl]-5-(1-methyl-4- piperazinyl)benzimidazole, 2-[2-[[4-(N,N-bis(2- chloroethyl)amino)phenyl]methyl]-5-benzimidazolyl]-5-(1- methyl-4-piperazinyl)benzimidazole, 2-[2-[2-[4-(N,N-bis(2-chloroethyl)amino)phenyl]ethyl]-5-benzimidazolyl]-5-(1- methyl-4-piperazinyl)benzimidazole, 2-[2-[3-[4-(N,N-bis (2 - chloroethyl)amino)phenyl]propyl]-5-benzimidazolyl]-5-(1- methyl-4-piperazinyl)benzimidazole, 2-[2-[6-[4-(N,N-bis (2 - chloroethyl)amino)phenyl]hexyl]-5-benzimidazolyl]-5-(1- methyl-4-piperazinyl)benzimidazole, 2-[4-(N,N-bis(2- chloroethyl)amino)phenyl]-5-cyanobenzimidazole, 2-[2-[4- (N,N-bis(2-chloroethyl)amino))phenyl]ethyl)-5- cyanobenzimidazole, 2-[3-[4-(N,N-bis (2 - chloroethyl)amino)phenyl]propyl)-5-cyanobenzimidazole, 2- [6-[4-(N,N-bis(2-chloroethyl)amino))phenyl]hexyl)-5- cyanobenzimidazole, 2-[3-[4-(N-ethyl-N-(2- chloroethyl)amino))phenyl]propyl)-5-cyanobenzimidazole, 2- [2-[4-(N-ethyl-N-(2-chloroethyl)amino))pehnyl]ethyl)-5- cyanobenzimidazole, 2-[6-[4-(N-ethyl-N-(2- chloroethyl)amino))phenyl]hexyl)-5-cyanobenzimidazole, 2- [2-[3-[4-(N-ethyl-N-(2-chloroethyl)amino)phenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)benzimidazole, 2- [2-[2-[4-(N-Ethyl-N-(2-chloroethyl)amino)phenyl]ethyl]-5- benzimidazoyl]-5-(1-methyl-4-piperazinyl)benzimidazole, 2- [2-[6-[4-(N-ethyl-N-(2-chloroethyl)amino)phenyl]hexyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)benzimidazole, 2- [2-[[4--N-ethyl-N-(2-chloroethyl)amino)phenyl]methyl]-5- benzimidazoyl]-5-(1-methyl-4-piperazinyl)benzimidazole, 2-
[2-[3-[3-(N,N-bis(2-chloroethyl)amino)phenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)benzimidazole, 2-
[2-[3-[3-(N-ethyl-N-(2-chloroethyl)amino)phenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)benzimidazole, 2-
[2-[3-[2-(N,N-bis(2-chloroethyl)aminophenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)benzimidazole, 2-
[2-[3-[2-(N-ethyl-N-(2-chloroethyl)amino)phenyl]propyl]-5- benzimidazolyl]-5-(1-methyl-4-piperazinyl)benzimidazole, 2- [3-[N-(4,5-bishydroxymethyl)imidazolyl]propyl]-5-(1-methyl- 4-piperazinyl)benzimidzole, 2-[3-[N(4 , 5- bis(methylcarbamoyl)methyl)imidazoly]propyl]-5-(1-methyl-4- piperazinyl)benzimidazole, 2-[2-[3-[Ν-(4,5- bishydroxymethyl)imidazolyl]propyl]-5-(benzimidazolyl]-5- (1-methyl-4-piperazinyl)benzimidazole, 2-[2-[3-[Ν-(4,5- bis(methylcarbamoyl)methyl)imidazolyl]propyl]-5-
(benzimidazolyl]-5-(1-methyl-4-piperazinyl)benzimidazole.
8. An anti-tumour agent comprising as active
ingredient a compound according to any one of Claims 1 to 7.
9. A pharmaceutical composition comprising a
compound according to any one of Claims 1 to 7, together with a pharmaceutically acceptable carrier.
10. Use of a compound according to any one of
Claims 1 to 7 in medicine.
11. Use of a compound according to any one of
Claims 1 to 7 for the manufacture of a medicament for the treatment of neoplastic disease.
12. A method of treatment of neoplastic disease, comprising the step of administering an effective dose of a compound as defined in any one of Claims 1 to 7 to a mammal in need of such treatment.
13. A method according to Claim 12, wherein the dose of the compound is between 1 and 200 mg/kg body weight.
14. A method of synthesis of compound of general formula I according to any one of Claims 1 to 6, comprising steps selected for the group consisting of a) converting a nitrile to a corresponding imine ether hydrochloride, and reacting said imine ether hydrochloride with a freshly prepared phenylene diamine;
b) treating an acid with borane-dimethyl sulphide to yield an alcohol; converting the alcohol to an aldehyde by oxidation under Swern conditions; reacting the aldehyde with a phenylenediamine to give a
monobenzimidazole compound of formula I; converting the monobenzimidazole compound to a corresponding imine ether and coupling with a diaminobenzene derivative by cupric ion-promoted oxidation to yield a bisbenzimidazole compound of formula I;
c) converting a monomustard acid to an
aldehyde, coupling the aldehyde with a phenylenediamine to yield a monobenzimidazole compound of formula I together with the corresponding alcohol; converting the
monobenzimidazole to the corresponding imine ether, coupling the imine ether with a diaminobenzene derivative and optionally halogenating to yield a monomustard
bisbenzimidazole compound of formula I;
d) coupling a carboxylic acid derivative of an aniline mustard or aniline half-mustard with a freshly- prepared benzimidazole phenylenediamine using PPE, to yield a mustard or half-mustard bisbenzimidazole of formula I;
e) converting an imidazole dicarboxylic acid to the corresponding diethyl ester, subjecting said ester to N-alkylation with an alkyl bromide to yield a t-butyl ester derivative, selectively hydrolysing the t-butyl ester derivative to yield a carboxylic acid, converting the carboxylic acid to the acid chloride, coupling said acid chloride to a nitroaniline to yield an amide, subjecting said amide to reduction and acid-catalysed ring closure to yield a benzimidazole, coverting said benzimidazole to the corresponding diol by hydride reduction, and bis-carbamate protecting said diol; and
f) directly coupling a carboxylic acid and a freshly prepared phenylenediamine using PPE to yield a bisbenzimidazole, subjecting said benzimidazole to hydride reduction to yield a diol and biscarbamate protecting said diol.
AU32475/95A 1994-08-26 1995-08-23 Novel dna-targeted alkylating agents Ceased AU695936B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU32475/95A AU695936B2 (en) 1994-08-26 1995-08-23 Novel dna-targeted alkylating agents

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
AUPM7665A AUPM766594A0 (en) 1994-08-26 1994-08-26 Novel dna-targeted alkylating agents
AUPM7665 1994-08-26
AUPN0242 1994-12-21
AUPN0242A AUPN024294A0 (en) 1994-12-21 1994-12-21 Novel DNA-targeted alkylating agents
AU32475/95A AU695936B2 (en) 1994-08-26 1995-08-23 Novel dna-targeted alkylating agents
PCT/AU1995/000520 WO1996006831A1 (en) 1994-08-26 1995-08-23 Novel dna-targeted alkylating agents

Publications (2)

Publication Number Publication Date
AU3247595A AU3247595A (en) 1996-03-22
AU695936B2 true AU695936B2 (en) 1998-08-27

Family

ID=27153459

Family Applications (1)

Application Number Title Priority Date Filing Date
AU32475/95A Ceased AU695936B2 (en) 1994-08-26 1995-08-23 Novel dna-targeted alkylating agents

Country Status (1)

Country Link
AU (1) AU695936B2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE696499A (en) * 1966-04-01 1967-10-03

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE696499A (en) * 1966-04-01 1967-10-03
FR1519964A (en) * 1966-04-01 1968-04-05 Hoechst Ag Bis-benzimidazoles carrying basic substituents and their preparation

Also Published As

Publication number Publication date
AU3247595A (en) 1996-03-22

Similar Documents

Publication Publication Date Title
EP1131301B1 (en) 2-phenylbenzimidazoles and 2-phenylindoles, and production and use thereof
Zhao et al. Synthesis and cytotoxic evaluation of certain 4-anilino-2-phenylquinoline derivatives
CA2736097C (en) Carbazole compounds for inhibition of nf-kb activity
EP0777656B1 (en) Novel dna-targeted alkylating agents
US20070072912A1 (en) Alicyclic-amine-substituted 4-carboxamido-benzimidazoles as parp-inhibitors and antioxidants
US4444776A (en) Dipyrido (4,3-B) (3,4-F) indoles, process for obtaining them, therapeutical use and pharmaceutical compositions
US8222297B2 (en) Aniline or phenol mustards linked to DNA-affinic molecules or water-soluble aromatic rings and their use as cancer therapeutic agents
Atwell et al. Synthesis, DNA interactions and biological activity of DNA minor groove targeted polybenzamide-linked nitrogen mustards
Gravatt et al. DNA-directed alkylating agents. 4. 4-anilinoquinoline-based minor groove directed aniline mustards
US4767769A (en) Antiulcer benzimidazole derivatives
AU695936B2 (en) Novel dna-targeted alkylating agents
US7989451B2 (en) Tricyclic 1,2,4-triazine oxides and compositions for therapeutic use in cancer treatments
EP0799224B1 (en) Quinoxalines and drugs prepared therefrom
KACZMAREK et al. SYNTHESIS OF 6-SUBSTITUTED 6H-INDOLO [2, 3-b] QUINOLINES
DE19718181A1 (en) New aryl-substituted bi:cyclic heterocyclic compounds
Bajwa et al. Antimalarials. 1. Heterocyclic analogs of N-substituted naphthalenebisoxazines
US9193687B2 (en) Phenyl N-mustard linked to DNA-affinic molecules or water-soluble aryl rings, method and their use as cancer therapeutic agents
EP4215521A1 (en) Fused ring diimide derivative, preparation method therefor and use thereof
US20070191372A1 (en) Dna-targeted benzotriazine 1,4-dioxides and their use in cancer therapy
EP1590332A1 (en) A process for the synthesis of bisbenzimidazoles and its derivations
US6750223B2 (en) 4-Anilino[2,3-b]quinoline derivatives, their preparation processes and pharmaceutical compositions comprising the same
CA1241335A (en) 1,4-aminoalkylamino-9h-thioxanthen-9-ones
WO1994000115A1 (en) Bis-(substituted-phenyl) derivatives with cytotoxic and anticancer activity
US4880817A (en) O-functionalized derivatives of substituted isoquinolin-3-ols having cardiotonic and/or phosphodiesterase fraction III inhibiting properties and/or renal vasodilating properties
Chackal et al. Design, synthesis and pharmacological evaluation of new anticancer fused pentacycles