Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METAL COMPLEXES AND THERAPEUTIC USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2002/066435
Kind Code:
A1
Abstract:
A compound of formula (I) or a salt thereof (I) where M is selected from the group consisting of platinum (II), palladium (II), and copper(II); L1 is an intercalator moiety; and L2 is a bidentate ligand, provided that when L2 is other than a bidentate ligand containing an aryl or phenyl group, L2 is a chiral bidentate ligand.

Inventors:
FENTON RONALD RALPH (AU)
ALDRICH-WRIGHT JANICE (AU)
Application Number:
PCT/AU2002/000167
Publication Date:
August 29, 2002
Filing Date:
February 22, 2002
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV SYDNEY (AU)
FENTON RONALD RALPH (AU)
ALDRICH-WRIGHT JANICE (AU)
International Classes:
A61K31/4745; A61P35/00; C07D471/04; C07F1/00; C07F15/00; (IPC1-7): C07D215/06; C07D241/40; A61K31/555; A61P35/00
Other References:
CUSUMANO M. ET AL: "Noncovalent......DNA", INORG. CHEM., vol. 37, no. 3, 1998, pages 563 - 568, XP002971221
CUSUMANO M. ET AL: "The interaction......DNA", J. INORG. BIOCHEM., vol. 65, no. 2, 1997, pages 137 - 144, XP002971224
ODANI A. ET AL: "Thermodynamic....stacking", INORG. CHEM., vol. 30, no. 24, 1991, pages 4484 - 4486, XP002971222
ODANI A. ET AL: "Platinum......interactions", INORG. CHEM., vol. 30, no. 9, 1991, pages 2133 - 2138, XP002971223
DESHPANDE S.: "Preparation and......complexes", INORG. CHIM. ACTA, vol. 78, no. 2, 1983, pages 75 - 80, XP002971225
MATILLA A.: "The reactions of......", J. INORG. BIOCHEM., vol. 55, no. 4, 1994, pages 235 - 247, XP009018613
See also references of EP 1377552A4
Attorney, Agent or Firm:
F B RICE & CO. (Carlton, Victoria 3053, AU)
Download PDF:
Claims:
AMENDED CLAIMS
1. [(received by the International Bureau on 24 June 2002 (24. 06. 02); original claim 16 replaced by amended claim 16; remaining claims unchanged (1 page) ] 14. A compound according to any one of claims 1 to 11, which is in the ibrm o : fa pure enantiomer.
2. 15 A compound according to claim 1 selected from the group consisting of of 3R butanediamine) (1, 10phenanthroline)platinum(II) cation, (2S,3Sbutanediamine)(1,10 phenanthroline)platinum(II) cation, (2R,3Rbutanediamine)(1,10 phenanthroline)platinum(II) cation, (1S,2Scyclohexanediamine)1,10 phenanthroline)platinum(II) cation, (1S,2Rcyclohexanediamine)1,10 phenanthroline)platinum(II) cation, (N,N'dimethyl1S,2Scyclohexanediamine)(1, 10 phenanthroline)platinum(II) cation and (N,N'dimethyl1R,2R cyclohexanediamine)(1,10phenanthroline)platinum(II) cation.
3. Use of a compound according to any one of the preceding claims in the treatment ofaproliierative disease.
4. A use according to claim 16, wherein the disease is a cancer.
5. Use of a compound according to any one of claims 1 to 16 as an antimicrobial agent.
6. A method of treatment of a cancer in a subject comprising administering to the subject a therapeutically effective amount of a compound of formula I or a salt thereof where M is selected from the group consisting of platinum(II), palladium (II) and copper(II); IJl is an intercalator moiety ; and L2 is a bidentate ligand, provided that when L2 is other than a bidentate ligand containing an aryl or phenyl group, L2 is a chiral bidentate ligand.
7. A method according to claim 19, wherein the cancer is selected from the group consisting of cancer of the esophagus, breast cancer, cancer of the ovary, lung cancer, bladder cancer, testicular cancer, cancer of the endometrium, head and neck cancer, thyroid cancer, cervical cancer, neuroblastoma, leukemia and osteogenic sarcoma.
Description:
Metal Complexes and therapeutic uses thereof Field of the Invention The present invention relates to metal complexes and therapeutic uses thereof.

The invention is particularly concerned with metallointercalator compounds, pharmaceutical compositions containing them and to their use for the therapeutic treatment of various cancers.

Back round The platinum coordination complex cisplatin (cis-diamminedichloroplatinum (II)) is widely prescribed for the treatment of a variety of tumours (eg advanced testicular cancer, ovarian cancer, breast cancer and cancers of the bladder, head, neck, oesophagus and lung). Carboplatin (cis-diammine (l, l- cyclobutanedicarboxylato) platinum (II)) has similar antineoplastic activity and may also be used in bone marrow therapy or peripheral stem cell rescue. US Patent No.

4,177,263, the entire disclosure of which is incorporated herein by reference, describes methods of treating cancer with cisplatin and cisplatin analogues.

Cisplatiti is classified as an alkylating agent. It is believed to kill cancer cells by covalently binding to DNA and interfering with its repair mechanism, eventually leading to cell death. After the cisplatin molecule enters the cell membrane, the first step is for a molecule of water to replace one of the chloride ions of the cisplatin molecule. The resulting complex can then bind to a nitrogen on a DNA nucleotide. The second chloride ion is then replaced by another water molecule and the platinum binds to a second nucleotide. Cisplatin has a preference for nitrogen 7 on two adjacent guanines on the same strand. It also binds to adenine and to a lesser extent across strands. The resulting distortion in shape of the DNA prevents effective repair.

Another covalent binder, oxaliplatin ([Pt (II)-oxalato (1R), (2R)- diaminocyclohexane] complex) is prescribed for treating the same type of cancers, more particularly cancers of the ovaries, as well as cancers of the colon, of the upper respiratory tracts and epidermoid cancers. Oxaliplatin belongs to the class of platinum (II)-t. rans-1, 2-diaminocyclohexane complexes.

Summary of Invention We have identified what we believe to be a new group of compounds that may have utility as therapeutic agents in the treatment of cancers.

Accordingly, in a first aspect, the present invention provides a compound of formula I or a salt thereof 1L212+ I where M is selected from the group consisting of platinum (II), palladium (II) and copper (II) ; Ll is an intercalator moiety; and L2 is a bidentate ligand, provided that when L2 is other than a bidentate ligand containing an aryl or phenyl group, L2 is a chiral bidentate ligand.

By the term"intercalator moiety"we mean any moiety that is capable of non- covalent insertion between pairs of bases in the nucleic acid double helix.

Preferably M is platinum (II).

Preferably L2 is a chiral bidentate ligand.

The intercalator moiety preferably forms a square-planar or pseudo-planar complex. The intercalator moiety may be a planar heterocyclic residue. Preferably, the intercalator is a bidentate ligand. A strongly binding bidentate intercalator is particularly preferred.

The intercalator Li may be 1, 10-phenanthroline or a substituted derivative thereof. The 1,10-phenanthroline may be substituted with one or more alkyl groups, preferably methyl groups. Examples of, but not restricted to, substituted derivatives of 1, 10-phenanthroline include 4-methyl-1, 10-phenanthroline, 5,6-dimethyl-1,10- phenanthroline, 4,7-dimethyl-1,10-phenanthroline, 3,8-dimethyl-1,10-phenanthroline, 3,4,7,8-tetramethyl-1, 1 0-phenanthroline and 4,7-diamino-1, 1 0-phenanthroline.

The substituent positions for 1, 10- phenanthroline are shown below: Substituent positions for 1,10-phenanthroline

The present invention also extends to compounds in which the intercalator compound Li is other than 1, 10-phenanthroline, for example, 2- (2'- pyridyl) quinoxaline, dipyrido [3,2-d; 2'3'-fAquinoxaline (also know as 1, 10-tetra-aza- 2,3-dihydrotriphenylene), diaminophenathrene and their substituted analogues. Whilst compounds based on these other intercalator compounds may be less active as antitumour agents than those based on 1, 10-phenanthroline, our experiments in cancer cell lines suggest that they do have antitumour activity.

By the term"chiral bidentate ligand"we mean a bidentate ligand having at least one chiral centre.

Preferably the bidentate ligand is a chiral diamine. Where the diamine is other than a ligand having an aryl or phenyl moiety, the diamine is a chiral diamine. On the other hand, if the diamine contains an aryl or phenyl moiety (eg 1,2-diaminobenzene and substituted analogues thereof), the diamine need not be a chiral diamine. The diamine may be of formula IIa or IIb and variants thereof.

RtR2N-CHR3-(CnH2n)-CHR4-NR5R6 IIa lib wherein in Formula IIa, the group- (C,, H2,,)- may be acyclic or cyclic, n is 0 to 4 inclusive, Ru, R2, R3, R4, R'and R, which may be the same or different, are independently selected from hydrogen or substituted or unsubstituted alkyl, aryl; phenyl or cycloalkyl ; or Ru, R2, R5 and R6 are as described above and R3 and R4 are joined to form, with the -(CnH2n)- group, a cycloalkane having 6 to 8 ring carbons, and wherein the compound of formula IIa has at least one chiral centre, and wherein in Formula lib, A is an aromatic ring of 6 to 8 carbons, Ru, R, R4 and R5, are as described above; and wherein the compound of formula IIb may or may not have a chiral centre.

An example of a compound of formula IIb that does not require a chiral centre is 1, 2-diaminobenzene and derivatives thereof.

In a particularly preferred form of the invention, the compound of formula I is a metal complex exemplified by formula III: [PtL8L2] 2+ III where Li and L2 are as defined above.

Where the compound of the present invention is in the form of a salt, the anionic counter ion may be any suitable anion. The counter ion may be chosen such that it imparts desirable or special properties, such as increased solubility, on the complex.

Preferably the counter ion is pharmaceutically acceptable. Non-limiting examples of anionic counter ions are chloride, perchlorate, hexafluorophosphate, sulfate and nitrate.

Chloride ion is the preferred counter ion for biological purposes.

Where compounds of the present invention have a chiral centre, they may be in the form of a substantially pure enantiomer, diasteriomer or a racemate.

We have produced compounds in accordance with formula I and found these compounds to have high biological activity with cancer cell lines. These results are indicative of the compounds of the present invention having biological activity as antitumour agents.

Accordingly, in a second aspect, the present invention provides a method of treatment of a cancer in a subject, comprising administering to the subject a therapeutically effective amount of a compound of formula I or salt thereof [ML1L2]2+ I where M, Ll, and L2 are as defined above.

Preferably the compound used in the method of the second aspect is a compound of formula III described above.

The method of treatment of the second aspect may be suitable for treatment of the same range of tumours against which cisplatin, carboplatiri and oxalplaíin have been described as being active. Moreover, the compounds of the present invention may be used to treat tumours which have an acquired or intrinsic resistance to cisplatin.

Examples of tumours for which the treatment method of the invention may be used include cancers of the esophagus, breast, ovary, lung (eg small cell carcinoma), bladder, testicles, endometrium, head and neck, thyroid, cervix, neoblastoma, leukemia,

and osteogenic sarcoma. The method of the invention may also have application in the treatment of neoplasms of childhood.

The compound unit dose may vary depending upon the host treated, the particular route of administration, and the severity of the illness being treated.

Accordingly the optimum dosage may be determined by the practitioner who is treating any particular patient. The antitumour agent of the invention may be administered to a warm-blooded animal at a unit dose of approximately 1-200 mg/kg. The daily dose may be in the range of approximately 40-50 mg/kg, however, as already indicated, the appropriate dosage may be readily determined by the practitioner.

The antitumour effect of the compounds of the present invention may be applied as a sole therapy or may involve, in addition, one or more other substances and/or treatments. Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate administration of the individual components of the treatment. In the field of medical oncology it is normal practice to use a combination of different forms of treatment to treat each patient with cancer such as a combination of surgery, radiotherapy and/or chemotherapy. In particular, it is known that irradiation or treatment with antiangiogenic and/or vascular permeability reducing agents can enhance the amount of hypoxic tissue within a tumour. Therefore the effectiveness of the compounds of the present invention is expected to be improved by conjoint treatment with radiotherapy and/or with an antiangiogenic agent.

The method of the invention may be used with another anti-cancer agent, for example, adriamycin, radiation, surgery, ultrasound, photoreactive compounds, anthracyclines, nitrogen mustards, ethyleneamines, methylmelamines, alkyl-sulfonates, nitrosoureas, triazenes, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodophyllotoxins, antibiotics, tumour-associated proteins and antigens, biological response modifiers, alpha-interferon, platinum coordination complexes, anthracenedione, substituted ureas, methylhydrazine derivatives, adrenocortical suppressants, adreno-corticosteroid steroids, progestins, estrogens, anti-estrogens, androgens, anti-androgens, and solvents that destroy cancer cells. Further examples of cytotoxic agents that may be used in conjunction with the compound of the invention are paclitaxel, docetaxel, 7-O-methylthiomethyl-paclitaxel, 4-desacetyl-4-methylcarbonatepaclitaxel, 3'-tert-butyl-3'-N-tert-butyloxycarbonyl-4- deacetyl-3'-dephenyl-3'-N-debenzoyl-4-O-methoxycarbonyl-pacl itaxel, C- 4methylcarbonatepaclitaxel, epothilone A, epothilone B, epothilone C, epothilone D, desoxyepothilone A, desoxyepothilone B, S [1 S [1R, 3R (E), 7R, 105, 11R, 12R, 16S]]- 7,11-dihydroxy-8, 8, 1 0,12,16-pentamethyl-3- [ 1-methyl-2 (2-methyl-4-thiazolyl)-

ethenyl]-4-aza-17-oxabicyclo [14, l, 0] heptadecane-5,9-dione, [ 1 S- [IR, 3R (E), 7R, 1 OS, <BR> <BR> <BR> 1 1R, 1 2R, 1 6S,]]-3-[2-[2-(aminomethyl)-4-thiazolyl]-1-methylethenyl]-7, 1 1-dihydroxy- 8,8,10,12,16-pentamethyl-4, 17-dioxabicyclo- [ 14, 1,0] heptadecane-5, 9-dione, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloro- methotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine, arabinoside, podophyllotoxin, etoposide, etoposide phosphate, teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine, leurosine, estramustine, cyclophosphamide, bleomycin, ifosamide, melphalan, hexamethylmelamine, thiotepa, cytarabin, idatrexate, trimetrexate, dacarbazine, L- asparaginase, camptothecin, CPT11, topotecan, ara-C, bicalutamide, flutamide, leuprolide, a pyridobenzoindole, an interferon and/or an interleukin.

Particular, although not restricted to, examples of combination therapy include the use of a compound in accordance with the present invention with cisplatin, bleomycin, etoposide, vinblastine in the treatment of testicular cancer. In the case of treatment of carcinoma of the ovary, the compound of the invention may be used with, for example, paclitaxel, cyclophosphamide or doxorubicin.

In a third aspect, the present invention provides a pharmaceutical composition comprising a compound in accordance with formula I in an amount sufficient to have an antitumour effect in an animal or human together with at least one pharmaceutically acceptable excipient, diluent and/or carrier.

The compounds of the present invention may be administered in unit dosage form.

The pharmaceutical composition of the invention may be formulated in any suitable form of administration, for example, oral, parenteral (eg intravenous, subcutaneous, intramuscular or intramedullary injection) or rectal administration.

Preferably, the composition is formulated for parenteral administration.

The compounds may be administered in conjunction with hydration therapy.

This is the"standard"administration method for cisplatin. The hydration therapy minimises/reduces the effect on the kidneys, nephrotoxicity, which is the principal dose limiting factor for treatment.

For oral administration, the pharmaceutical compositions may be in the form of tablets, gelatine capsules, powders, granules or any other form which may be administered orally.

The oral formulation may include components selected from one or more of excipients, carriers, diluents, binders, lubricants, fluidising agents and adhesion inhibitors. The pharmaceutical compositions may further contain pharmaceutically

acceptable vehicles that are compatible with the compounds of the invention. In the case of capsules, a conventional excipient such as starch, lactose, talc, magnesium stearate and so on can be used. Also, in the case of tablet, any conventional excipient can be used. Examples of suitable carriers are starch, crystal cellulose, hydroxypropylmethylcellulose, polyethyleneglycol, lactose, polyvinylpyrrolydone or glyceryl or combinations of two or more thereof. Examples of diluents are glucose, dried lactose, Fast-flolactose, dehydrated lactose, sucrose, starch, starch 1500, calcium hydrogen phosphate, emcompress or avicel. Examples of binders are gum arabic, tragacanth, gelatin solution, starch paste solution, glucose syrup, sucrose syrup, povidone or cellulose derivatives. Examples of lubricants include polyethyleneglycol 4000,6000,8000, lauryl sodium sulfate, lauryl magnesium sulfate, sodium benzoate, polyethylene monostearate, glyceryl triacetate, magnesium stearate, zinc stearate, calcium stearate, stearic acid, talc, hardened vegetable oil, liquid paraffin, paraffin derivatives or wax. The formulation may include a fluidising agent, for example, starch, talc, silicon dioxide, silicate, magnesium carbonate or magnesium oxide. An adhesion inhibitor, for example starch or talc; may be incorporated into the formulation.

The formulation may be formulated for controlled release. Examples of controlled release additives include hydroxypropylmethylcellulose, hydroxypropylcellulose, hydroxyethylcellulose, ethylcellulose, methylcellulose, carboxymethylcellulose, polyacrylic acid, acrylic acid, acrylate derivatives, poly vinylpyrrolydone or polyethyleneglycol.

Injectable formulations may comprise water-soluble solvents such as physiological saline solution, sterilized water, Ringer's solution, an alcohol (eg ethanol, benzylalcohol, propyleneglycol and glycerine), higher fatty acid ester. These injectable formulation may include a diluent, for example, phosphate buffer saline (PBS), 0.9% NaCl (saline) and the like. The formulation may include a preservative, for example, sodium benzoate, methylparaben or propylparaben.

Other additives that may be included in an injectable formulation include an isotonication agent, analgesic, a stabilizing agent, suspending agent, buffering agent, emulsifying agent, all of which are well known to those skilled in the art.

The compounds of the present invention may have antimicrobial properties.

Accordingly, the present invention extends to antimicrobial composition comprising a compound in accordance with the present invention as well as to the use of these compounds as antimicrobial agents.

Moreover, the compounds of the present invention may have application in the treatment of cell proliferation diseases other than cancers (eg psoriasis). Accordingly, in yet another aspect, the present invention provides antiproliferative compositions comprising a compound in accordance with the invention as well as the use of these compounds as antiproliferative agents.

DETAILED DESCRIPTION OF THE INVENTION The compounds of the present invention differ in both structure and method of action from cisplatin and carboplatin (and their analogues). In particular, they differ from cisplatin and carboplatira in that they are intercalators, that is, they are thought to intercalate or insert into DNA, changing the shape and/or structure of the DNA, whereas cisplatirz and carboplatin are covalent binders. Moreover, in one embodiment, the new compound of the present invention incorporates a chiral diamine.

Intercalator compounds have been described in Stereochemical Requrements for Intercalation of Platinum Complexes into Double-stranded DNA,s, S. J. Lippard, P. J.

Bond, K. C. Wu and W. R. Bauer. Science, 194, 726 (1974) Activity of Platinum IntercalatingAgentsAgainstMurineLeukemiaL1210, W. D. McFayden, L. P. G.

Wakelin, 1. A. G. Roos and V. X Leopold, X Med Chem. 28, 1131 (1985), the entire disclosure of which are incorporated by reference. Both of these publications relate to non-chiral molecules, as the coordinated diamine. In contrast, the compounds of the present invention either involve the use of chiral alkyl bidentate ligands or non-chiral aryl or phenyl bidentate ligands (eg 1,2-diaminobenzenes) that need not have a chiral centre.

Some platinum complexes currently used contain chiral diamines in their structure, Oxaliplatin being one example. However, the compounds of the present invention differ from such compounds in that they have a bidentate intercalator on the other side of the coordination sphere. In preferred compounds of the invention, this bidentate intercalator is a strongly binding intercalator and as such would be expected to remain coordinated to the divalent platinum molecule under biological conditions.

Furthermore, in a preferred form of the present invention, the compounds, of formula I have an overall positive charge (ie. are cationic) on the molecule compared to the neutral cisplcctirz type compounds, which become charged in vivo through loss chloride ion (s).

The compounds of the present invention may have other advantages such as solubility and mode of action, which may prove to be better than that for drugs currently in clinical use. Changes in the diamine result in changes in properties such as

solubility, stereochemistry and activity, adding to the flexibility of use in clinical applications.

In order that the present invention may be more readily understood, we provide that following non-limiting embodiments.

Examples of specific molecular structure of intercalator compounds in accordance with the invention are shown below:

(2S, 3R-butanediamine) (1, 10-phenanthroline) platinum (II) cation (2S, 3S-butanediamine) (1, 1 0-phenanthroline) platinum (II) cation (2R, 3R-butanediamine) (1, 1 0-phenanthroline) platinum (II) cation

(1S, 2S-cyclohexanediamine)(1,10-phenanthroline) platinum (II) cation (1R, 2R-cyclohexanediamine) (1, 10-phenanthroline) platinum (II) cation.

(N,-dimethyl-lS, 2S-cyclohexanediamine) (1, 10-phenanthroline) platinum (II) cation (N, N-dimethyl-lR, 2R-cyclohexanediamine) (1, 10-phenanthroline) platinum (II) cation SYNTHESIS OF THE PLATINUM(II) COMPLEXES General synthetic procedure

The following is illustrative of a general synthetic procedure that may be used to make, with appropriate modification, compounds in accordance with the present invention.

Potassium tetrachloroplatinate (II) (0.423 g, 1.02 mmol, Aldrich) was dissolved in 400 mL of water in a large evaporating dish. Sodium chloride (0.529 g, 0.01 mol, Ajax) was added to the solution with gentle stirring until dissolved. The diimine, 1, 10- phenanthroline monohydrate (0.202 g, 1.00 mmol, Aldrich), was dissolved in hydrochloric acid (10 M, 0.8 mL) with a minimal amount of water, and added to the initial solution. The reaction was left to slowly evaporate on a steam bath for-4 hr.

The fluffy yellow product was collected via suction filtration and washed with ice-cold water (4 x 5 mL) and dried in air. The filtrate was preserved and continually reduced by heating, in order to obtain additional crops. The product was collected as described above. Yield: 0.409 g, 90 %. The compound was characterized by : 1H NE, solvent DMSO-d6 (Acros Organics), ppm: 8.19 (dd, 2H) ; 8.29 (s, 2H); 9.06 (d, 2H) ; 9.79 (d, 2H).

The Synthesis of [Pt (L) (phen)] 2+ (where L = R, R-or S, S-1, 2- Diaminocyclohexane ; N, N'-Dimethyl-R, R-or N, N'-Dimethyl-S, S-1, 2- diaminocyclohexane) The platinum complex, dichloro-1, 10-phenanthrolineplatinum (II) (-0. 50 mmol), was dissolved in water (-100 mL) and gently refluxed with stirring for 1 hr.

The diamine, (L), (-0. 50 mmol) was dissolved in minimum water and slowly added to the solution via a syringe. The conical flask was covered in aluminium foil and the mixture was left to gently reflux overnight (-16 hr). The next day, a second equivalent of the diamine (L) in minimum water was added to the mixture to ensure completion of the reaction. The solution was gently refluxed for an additional 8 hr. The solution was cooled to room temperature with stirring, and then filtered through a 0.45 Mm Sartorius Minisarts filter. The solution was reduced to-50 mL on the rotary evaporator at 40°C.

A saturated solution of lithium perchlorate was added to the-50 mL solution to precipitate the diamine-platinum complex as a perchlorate salt. The mixture was briefly heated inside the steam bath and then cooled to room temperature. The product was collected via suction filtration using a micro sintered-glass filter. The microcrystalline solid was washed with ice-water (2 x 5 mL), a minimum volume of ethanol, diethyl ether and dried in air. The product was placed in a vacuum desiccator overnight to dry completely. The filtrate was filtered through a 0.45, um Sartorius

Minisart filter and put aside for crystal formation. The quantities of the reagents used and the yield of each diamine are presented in Table 1.

The [Pt (dach) (phen)] 2+ complexes were characterized by : 1H NMR, solvent DMSO-d6 (Acros Organics), ppm: 1.25 (m, 2H); 1.47 (br m, 2H) ; 1.65 (m, 2H) ; 2.11 (d, 2H); 2.59 (m, 2H); 6.55 (m, 2H); 7.15 (d, 2H) ; 8. 28 (dd, 2H); 8.37 (s, 2H) ; 9.15 (m, 2H); 9.20 (s, 2H).

The [Pt (Me2-dach) (phen)] 2+ complexes were also characterized using : 1H NW, solvent DMSO-d6 (Acros Organics), ppm: 1.25 (m, 2H); 1.50 (m, 1H) ; 1.70 (m, 4H); 1.95 (m, 1H); 2.90 (d, 3H); 3.05 (d, 3H); 3.15 (br s, 2H) ; 7.30 (m, 1H) ; 7.80 (m, 1H) ; 8.30 (m, 2H); 8. 40 (s, 2H) ; 9.15 (d, 2H); 9.30 (m, 2H).

Table 1-The synthetic results of [Pt (L) (phen)] (CI04) 2 (where L = R,R- or S,S-1,2-diaminocyclohexane ; N,N'-dimethyl-R,R- or N,N'-dimethyl-S,S-1,2-diaminocyclohexane) Complex [Pt (R, R-dach) [Pt (S, S-dach) [Pt (Me2-R, R-- [Pt (Me2-S, S- (phen)] (CIO4) 2 (phen)] (CI04) 2 dach) dach) (phen)](ClO4)2) (phen)](ClO4)2 [PtCI2 (phen)] 0. 223 g, 0.220 g, 0.221 g, 0. 224 g, 0.500 mmol 0.493 mmol 0.500 mmol 0.501 mmol (L) R, R-dach S, S-dach Me2-R, R-dach Me2-S, S-dach Ist equiv. 0.062 g, 0.061 g, 0.071 g, 0.073 g, 0.540 mmol 0.535 mmol 0. 571 mmol 0.515 mmol 2nd equiv. 0.062 g, 0.064 g, 0.071 g, 0.073 g, 0.546 mmol 0.559 mmol 0.638 mmol 0.513 mmol Yield 0.270 g, 78 % 0. 290 g, 85 % 0. 260 g, 73 % 0. 260 g, 72 % Colour Pale yellow Pale yellow Medium yellow Medium yellow

IC50 Results The specific cell lines and the IC50 results for the various compounds are shown in the Table 2.

Table 2. Cell lines KYSE 520 A-427 (lung) LCLC-103H 5637 (bladder) 5637 (bladder) Compounds (esophagus) (lung) Repeat [Pt (en) (pheen)] Cl249. 1 (14.9) * 24. 1 (8.0) 41.2 (11.6) 27.0 (3.5) 23.7 (1. 30) [Pt (en) _,,..,.. : [Pt (2R, 3R-bn) (phen)] CI2 0. 67 (0. 03) 0. 54 (0. 12) 0. 78 (0. 19) 0. 32 (0. 18) 0. 43 (0. 22) [Pt en d Cl., 19. 4 3. 1 16. 4 6. 6 23. 6 7. 5 16. 7 3. 1.. : :.... ; ;, : [Pt (meso-2, 3-bn) (phen)] Cl21. 09 (2) 0. 92 (0. 13) 1. 07 (0. 11) 0. 61 (0. 25) 0. 72 (0. 25) [Pt (rac-2, 3-bn) (phen)] C1041. 03 (0. 33) 0. 67 (0. 19) 1. 06 (0. 32) 0. 40 (0. 14) 0. 95 (0. 70) [Pt (pnOH) (phen)] Cl : 86. 8 (4. 9) 41. 7 (12. 8) 97. 9 (2) 66. 1 (8. 9) 65. 4 (12. 47) ".... :... : r. : <a .. :. ; :,, ;,. [Pt (phen) (S, S-dach)] (ClO4) 2 m X w, 0. 15 (0. 06) [Pt (2R, 3R-bn) (phen)] Cl 0. 67 (0. 03) 0. 54 (0. 12) 0. 78 (0. 19) 0. 32 (0. 18) 0. 43 (0,. 22) [Pt (phen) (Me2-R, R-dach)] (Cl04) 2 W g 23. 6 (8. 95) CDDP 6. 63 (1. 23) 4. 46 (0.53) 1.44 (0.45) 0.88 (0.88) 0.43 (0.25) [Pt (rac-2,3-bn) (phen)] C104 1.03 (0.33) 0.67 (0.19) 1.06 (0.32) 0.40 (0.14) 0.95 (0.70) [Pt (pnOH) (phen)] CI : 86. 8 (4. 9) 41. 7 (12. 8) 97. 9 (2) 66. 1 (8. 9) 65. 4 (12. 47) k. : k : .. i.. t--t .- :, ; : ^k : k. : t".--.--2-- k 2 ;--k...... _,. .. : 2. xi .-k.... i : ; ss : : : tL : : G : 22'-.' ; : . 3. : .. 5... ". >.. r ; £ z,.,",.,""". >., 4 0 44 : li.. k: 2Ii2i22kk222Ni'. t%? ftlr, £IISiMi' v£I2IFi>. 222Ok:. IY. YY. '. e. 2Y<£Siiiiii£; LSS2i M. kk2ai2::. i2Ik.: ti \I: i 212: = . =. iAi£::: iii: i:::::: J k: t: =: 2ktkD7..:: CDDP 6.63 (1.23) 4.46 (0.53) 1.44 (0.45) 0.88 (0.88) 0.43 (0.13) * The concentrations are in jjM (standard deviation) Shaded areas indicate that no tests have been performed at this stage using these

particular cell lines.

When we compare the effect of changing the diamine from the non-chiral 1,2- diaminoethane (en) to the chiral ligands and ZS, 3S-diaminobutane (2S, 3S-bn) and 2R, 3R-diaminobutane (2R, 3R-bn) then the concentration inhibiting the exponential growth of cells to 50% is further reduction from 27.0 to 1.11 and 0.32 uM respectively.

On changing the chirality of the diamine such as 2R, 3R-diaminocyclohexane (R, R-dach) to 2S, 3S-diaminocyclohexane (S, S-dach) then the concentration inhibiting the exponential growth of cells to 50% is even further reduced from 0.54 to 0.15 ! The ICso value of 2S, 3S-diaminocyclohexane (S, S-dach) is lower that cisplatin at 0.43 uM.

From the last two examples it is quite clear that chirality plays a crucial role in the effectiveness of these compounds. Even methylation on the nitrogen atoms (which

on coordination to the metal ion produce additional chiral centres) influences the IC50 values [Pt (phen) (Me2-S,S-dach)](ClO4) 2 and [Pt (phen) (Me2-R, R-dach)] (C104) 2 were 55.6 and 23.6 u. M resp ectively.

These results indicate that a modest variation to either the intercalator or the diamine has significant effects on the resulting IC50 values. Systematic variation of each of the components M, L1, L3 in turn to may be used to determine the optimum combination of metal, intercalator and diamine.

Further experiments were carried out and the results of these are shown in Table 3.

Table 3

Results Of Anti-Tumour Drug Screening Unless indicated otherwise, these are the results for growth inhibition studies of compounds using the sulphorhodamine B (SRB) assay. IC50 is the concentration required to inhibit cell growth by 50%. Compound Test Cell line Results-ICso (pM) [Pt (Me2-S, S-dach) (phen)] (C104) 2CCL121040 [Pt (Me2-S, S-dach) (phen)] (C104) 2CC L1210/DDP40 [Pt (S, S-dach) (phen)] (ClO4)2 CC L1210 0.13~0. 00 [Pt (S, S-dach) (phen)] (ClO4) 2 CC L1210/DDP 0.28 ~ 0. 09 [Pt (Me2-R, R-dach) (phen)](ClO4)2 CC L1210 >40 [Pt (Me2-R, R-dach) (phen)] (ClO4) 2 CC L1210/DDP >40 [Pt (R, R-dach) (phen)] (ClO4) 2 CC L1210 1. 4,1.6 [Pt (R, R-dach) (phen)] (Cl04) 2 CC L1210/DDP 4. 3,4.5 [Pt (Me2-S, S-dach) (phen)] (C104) 2 CC L1210 12, 12 [Pt (Me2-S, S-dach) (phen)] (ClO4)2 CC L1210/DDP 18, 16 [Pt (Me2-R, R-dach) (phen)] (C104) 2CCL121026, > 40 [Pt (Me2-R, R-dach) (phen)] (ClO4) 2 CC L1210/DDP 34, > 40 [Pt (R, R-bn) (phen)] Cl2 CC L1210 1. 5,1.5 [Pt (R, R-bn) (phen)] Cl2 CC L1210/DDP 2. 5,1.9 [Pt (S, S-bn) (phen)] C12 CC L1210 3. 8, 3.0 [Pt (S, S-bn) (phen)] Cl2CC L1210/DDP5. 3, 8.0 [Pt (S, S-dach) (phen)] (C104) 2 CC L1210 0. 13 i0. 00 [Pt (S, S-dach) (phen)] (ClO4)2 CC L1210/DDP 0.28 0. 09 [Pt (R, R-bn) (phen)] Cl2 SRB 2008 1. 7,4.5 [Pt (R, R-bn) (phen)] Cl2SRBC135. 2,9.0,7.4 [Pt (R, R-bn) (phen)] ]Cl2 SRB SKOV3 2. 0,4.5,3.0 [Pt (S, S-bn) (phen)] Cl2 SRB 2008 7. 2,11 [Pt (S, S-bn) (phen)] Cl2 SRB Cl3 26, 22,28 [Pt (S, S-bn) (phen)] C12 SRB SKOV-3 9. 8, 9.2,12 [Pt (S, S-dach) (phen)] (ClO4) 2 SRB 2008 0. 37,0.45 [Pt (S, S-dach) (phen)] (C104) 2 SRB C13 0. 56,0.88 [Pt (S, S-dach) (phen)](ClO4)2 SRB SKOV-3 0. 31,0.42 [Pt (R, R-dach) (phen)] (ClO4) 2 CC L1210 1. 4,1.6 [Pt (R, R-dach) (phen)] (C104) 2 CC L1210/DDP 4. 3,4.5 [Pt (R, R-dach) (phen)] (C104) 2 SRB 2008 3. 3,3.1 [Pt (R, R-dach) (phen)] (C104) 2 SRB C13 5. 1,9.2 [Pt (R, R-dach) (phen)] (C104) 2 SRB SKOV3 3. 3,3.6

Comments: L1210 cells are mouse leukaemia cells. L1210/DDP are cisplatin-resistant.

Most recent cisplatin controls: L1210 0.5 uM ; L1210/DDP 6.9 uM 40µM is highest dose tested in CC growth inhibition assay 2008 cells are human ovarian carcinoma cells. C13*5 are cisplatin-resistant. SKOV-3 are intrinsically resistant to cisplatin Reference IC50's for cisplatin: 2008: 0.6 uM, C13 : 10 pLM, SKOV-3 : 3 pLM Ligands : dach = 1, 2-diaminocyclohexane; Me2-dach = N,N'-dimethyl-1, 2- diaminocyclohexane ; bn = 2,3-butanediamine The data presented in Table3 is the results collected from the growth inhibition studies of selected compounds with the following cell lines: L1210 (mouse leukaemia cells), 2008 (human ovarian carcinoma cells), L1210/DDP and C13*5 (acquired

cisplatin resistance) and SKOV-3 (intrinsic cisplatin resistance). The data shows that one of the compounds, [Pt (S, S-dach) (phen)] (104) 2 has far better activity against all the cell lines tested than the current anti-cancer drug, cisplatin. The data also shows that the bulky cyclohexane ring of the ancillary ligand and the chirality of the substituents on this ring (amine groups) plays an important part in the activity of this particular group of compounds. The complex with the absolute chiralities of S, S exhibits more activity than the R, R enantiomer. Moreover, the data shows that small changes of substitution on the ancillary ligand produces large changes on the activity of the complexes. For example, the complex [Pt (Me2-S, S-dach) (phen)] (C104) 2 (methyl substituent on each of the amine groups) is very much less active than the unsubstituted complex with the same absolute chirality.

Throughout this specification the word"comprise", or variations such as "comprises"or"comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.

Any description of prior art documents herein is not an admission that the documents form part of the common general knowledge of the relevant art in Australia.

It will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the invention as shown in the specific embodiments without departing from the spirit or scope of the invention as broadly described. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.