AU2002231463A1 - Metal complexes and therapeutic uses thereof - Google Patents

Metal complexes and therapeutic uses thereof

Info

Publication number
AU2002231463A1
AU2002231463A1 AU2002231463A AU2002231463A AU2002231463A1 AU 2002231463 A1 AU2002231463 A1 AU 2002231463A1 AU 2002231463 A AU2002231463 A AU 2002231463A AU 2002231463 A AU2002231463 A AU 2002231463A AU 2002231463 A1 AU2002231463 A1 AU 2002231463A1
Authority
AU
Australia
Prior art keywords
compound
phenanthroline
platinum
cancer
formula
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.)
Granted
Application number
AU2002231463A
Other versions
AU2002231463B2 (en
Inventor
Janice Aldrich-Wright
Ronald Ralph Fenton
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.)
University of Western Sydney
Original Assignee
University of Western Sydney
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 AUPR3302A external-priority patent/AUPR330201A0/en
Application filed by University of Western Sydney filed Critical University of Western Sydney
Priority to AU2002231463A priority Critical patent/AU2002231463B2/en
Publication of AU2002231463A1 publication Critical patent/AU2002231463A1/en
Assigned to THE UNIVERSITY OF SYDNEY, THE UNIVERSITY OF WESTERN SYDNEY reassignment THE UNIVERSITY OF SYDNEY Request for Assignment Assignors: THE UNIVERSITY OF SYDNEY
Application granted granted Critical
Publication of AU2002231463B2 publication Critical patent/AU2002231463B2/en
Assigned to THE UNIVERSITY OF WESTERN SYDNEY reassignment THE UNIVERSITY OF WESTERN SYDNEY Request for Assignment Assignors: THE UNIVERSITY OF SYDNEY, THE UNIVERSITY OF WESTERN SYDNEY
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

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.
Background The platinum coordination complex cisplatin (cts-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 (cw-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.
Cisplatin 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 ofthe chloride ions ofthe 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 ofthe DNA prevents effective repair.
Another covalent binder, oxaliplatin ([Pt(LT)-oxalato (1R),(2R)- diaminocyclohexane] complex) is prescribed for treating the same type of cancers, more particularly cancers ofthe ovaries, as well as cancers ofthe colon, ofthe upper respiratory tracts and epidermoid cancers. Oxaliplatin belongs to the class of platinum(II)-t7W?5- 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
[ML,L2] + I where M is selected from the group consisting of platinum(II), palladium (II) and copper(II);
Li 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, L 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-l, 10-phenanthroline, 5, 6-dimethyl-l, 10- phenanthroline, 4,7-dimethyl-l, 10-phenanthroline, 3, 8-dimethyl-l, 10-phenanthroline, 3,4,7,8-tetramethyl-l, 10-phenanthroline and 4,7-diamino-l, 10-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'- pyridyOquinoxaline, dipyrido[3,2-d;2'3'-f]quinoxaline (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 Ila or lib and variants thereof.
Ila
lib
wherein in Formula Ila, the group -(CnH2n)- may be acyclic or cyclic, n is 0 to 4 inclusive, R , R , R , R , R5 and R6, which may be the same or different, are independently selected from hydrogen or substituted or unsubstituted alkyl, aryl; phenyl or cycloalkyl; or R1, R2, R5 and R6 are as described above and R3 and R4 are joined to form, with the -(CnH n)- group, a cycloalkane having 6 to 8 ring carbons, and wherein the compound of formula Ila has at least one chiral centre, and wherein in Formula lib, A is an aromatic ring of 6 to 8 carbons, R1, R2, R4 and R , are as described above; and wherein the compound of formula lib may or may not have a chiral centre. An example of a compound of formula lib that does not require a chiral centre is 1, 2-diaminobenzene and derivatives thereof.
In a particularly preferred form ofthe invention, the compound of formula I is a metal complex exemplified by formula III:
[PtLιL2]2+ III where Li and L are as defined above.
Where the compound ofthe 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 ofthe 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 ofthe compounds ofthe 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
[MLιL2]2+
I where M, Li, and L2 are as defined above.
Preferably the compound used in the method ofthe second aspect is a compound of formula III described above. The method of treatment ofthe second aspect may be suitable for treatment ofthe same range of tumours against which cisplatin, carboplatin and oxaliplatin have been described as being active. Moreover, the compounds ofthe 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 ofthe invention may be used include cancers ofthe esophagus, breast, ovary, lung (eg small cell carcinoma), bladder, testicles, endometrium, head and neck, thyroid, cervix, neoblastoma, leukemia, and osteogenic sarcoma. The method ofthe 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 ofthe illness being treated. Accordingly the optimum dosage may be determined by the practitioner who is treating any particular patient. The antitumour agent ofthe 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 ofthe compounds ofthe 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 ofthe simultaneous, sequential or separate administration ofthe 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 ofthe present invention is expected to be improved by conjoint treatment with radiotherapy and/or with an antiangiogenic agent.
The method ofthe 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 ofthe 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-ρaclitaxel, C- 4methylcarbonatepaclitaxel, epothilone A, epothilone B, epothilone C, epothilone D, desoxyepothilone A, desoxyepothilone B, S[1S[1R,3R(E), 7R, 10S, 11R, 12R, 165]]- 7, 1 l-dihydroxy-8,8, 10, 12, 16-pentamethyl-3-[l-methyl-2(2-methyl-4-thiazolyl)- ethenyl]-4-aza-17-oxabicyclo[14,l,0]heptadecane-5,9-dione,[lS-[li?,3R(E),7R,10S, 11R, 12R, 16S,]]-3-[2-[2-(aminomethyl)-4-thiazolyl]-l-methylethenyl]-7,l 1-dihydroxy- 8,8,10,12,16-pentamethyl-4,17-dioxabicyclo-[14,l,0]heptadecane-5,9-dione, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloro- methotrexate, mitomycin C, porflromycin, 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 ofthe ovary, the compound ofthe 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 ofthe present invention may be administered in unit dosage form.
The pharmaceutical composition ofthe 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 ofthe 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 ofthe 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 he 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 ofthe present invention differ in both structure and method of action from cisplatin and carhoplatin (and their analogues). In particular, they differ from cisplatin and carhoplatin in that they are intercalators, that is, they are thought to intercalate or insert into DNA, changing the shape and/or structure ofthe DNA, whereas cisplatin and carhoplatin are covalent binders. Moreover, in one embodiment, the new compound ofthe present invention incorporates a chiral diamine.
Intercalator compounds have been described in Stereochemical Requirements 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 (1974J Activity of Platinum Intercalating Agents Against. Murine Leukemia L1210, W. D. McFayden, L. P. G. Wakelin, I. A. G. Roos and V. A Leopold, J. 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 ofthe 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 ofthe present invention differ from such compounds in that they have a bidentate intercalator on the other side ofthe coordination sphere. In preferred compounds ofthe 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 ofthe present invention, the compounds, of formula I have an overall positive charge (ie. are cationic) on the molecule compared to the neutral cisplatin type compounds, which become charged in vivo through loss chloride ion(s).
The compounds ofthe 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)(l,10-phenanthroline)platinum(π) cation
(2R,3R-butanediamine)(l, 10-phenanthroline)platinum(II) cation
(lS,2S-cyclohexanediamine)(l,10-phenanthroline)platinum(II) cation
( lR,2R-cyclohexanediamine)( 1 , 10-phenanthroline)platinum(LI) cation.
(N,N-dimethyl- 1 S,2S-cyclohexanediamine)( 1 , 10-phenanthroline)platinum(II) cation
(N,N-dimethyl- lR,2R-cyclohexanediamine)( 1 , 10-phenanthroline)platinum(II) cation
SYNTHESIS OF THE PLATINUMOT) 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 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: 1HNMR, solvent DMSO-flfe (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'-DimethyI-R,R- or N,N'-T)imethy\-S,S-l,2- diaminocyclohexane)
The platinum complex, dichloro-l,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 ofthe 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 μm Sartorius™ Minisart® 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 μm Sartorius™ - Minisart filter and put aside for crystal formation. The quantities ofthe 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- e (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(Me -dach)(phen)]2+ complexes were also characterized using: 1H NMR, solvent OMSO-d6 (Acros Organics), ppm: 1.25 (m, 2H); 1.50 (m, IH); 1.70 (m, 4H); 1.95 (m, IH); 2.90 (d, 3H); 3.05 (d, 3H); 3.15 (br s, 2H); 7.30 (m, IH); 7.80 (m, IH); 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)](ClO4)2
(where L = R,R- or S,S-l,2-diaminocyclohexane; Λr >iV-dimeth l-R,R-or VN-dimeth l- ', ,-l,2-diaιninocycIohexane)
ICso Results
The specific cell lines and the IC50 results for the various compounds are shown in the Table 2.
Table 2.
* The concentrations are in μM (standard deviation)
Shaded areas indicate that no tests have been performed at this stage using these 5 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 2S, 3S-diaminobutane (2S3S-bn) and 2R, 3R-diaminobutane (2R,3R-bn) then the concentration inhibiting the exponential 10 growth of cells to 50% is further reduction from 27.0 to 1.11 and 0.32 μM respectively.
On changing the chirality ofthe 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 μM. The IC50 value of 2S S-diaminocyclohexane (S,S-dach) is lower that cisplatin at 0.43 15 μM.
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,E-dach)](ClO4)2 were 55.6 and 23.6 μM 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 ofthe 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%.
Comments:
L1210 cells are mouse leukaemia cells. L1210/DDP are cisplatin-resistant.
Most recent cisplatin controls: L1210 0.5 μM; L1210/DDP 6.9 μM
40 μM 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 μM, C13: 10 μM, SKOV-3: 3 μM
Ligands: dach = 1,2-diaminocyclohexane; Me2-dach = N,N-dimethyl-l,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 ofthe compounds, [Pt(S,S-dach)(phen)](ClO ) 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 ofthe ancillary ligand and the chirality ofthe 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 ofthe complexes. For example, the complex [Pt(Me2-S,S-dach)(phen)](ClO )2 (methyl substituent on each ofthe 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 ofthe common general knowledge ofthe 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 ofthe invention as broadly described. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.

Claims (1)

  1. CLAIMS:
    1. A compound of formula I or a salt thereof
    [ML_ ] 2+ I where M is selected from the group consisting of platinum(II), palladium (II) and copper(II);
    Li 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, L is a chiral bidentate ligand.
    2. A compound according to claim 1, wherein M is platinum(II).
    3. A compound according to claim 1 or claim 2, wherein L is a chiral bidentate ligand.
    4. A compound according to any one ofthe preceding claims, wherein L2 is a compound according to formula Ila or lib:
    R1R2N-CHR3-(C„H2n)-CHR4-NR5R6 Ila
    G CNR.,R2
    - ^CNR4R5 lib
    wherein in Formula Ila, the group -(C„H2n)- may be acyclic or cyclic, n is 0 to 4 inclusive, R1, R2, R3, R4, R5 and R6, which may be the same or different, are independently selected from hydrogen or substituted or unsubstituted alkyl, aryl; phenyl or cycloalkyl; or R1, R2, R5 and R6 are as defined 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 Ila has at least one chiral centre, and wherein in Formula lib, A is an aromatic ring of 6 to 8 carbons, R1, R , R and R5, are as defined in claim 1; and wherein the compound of formula optionally includes a chiral centre.
    5. A compound according to any one ofthe preceding claims, wherein the intercalator moiety forms a square-planar or pseudo-planar complex.
    6. A compound according to any one ofthe preceding claims, wherein Li is selected from 1, 10-phenanthroline or a substituted derivative thereof
    7. A compound according to claim 6, wherein i is selected from the group consisting of 4-methyl-l, 10-phenanthroline, 5, 6-dimethyl-l, 10-phenanthroline, 4,7- dimethyl-l, 10-phenanthroline, 3,8-dimethyl-l, 10-phenanthroline, 3,4,7,8-tetramethyl- 1, 10-phenanthroline and 4,7-diamino-l, 10-phenanthroline.
    8. A compound according to any one of claims 1 to 5, wherein ~L\ is selected from the group consisting of 2-(2'-pyridyl)quinoxaline, dipyrido[3,2-d;2'3'-f]quinoxaline (l,10-tetra-aza-2,3-dihydrotriphenylene), diaminophenathrene and their substituted analogues.
    9. A compounds according to any one ofthe preceding claims which is in the form of a salt.
    10. A compound according to claim 9, wherein the counter ion ofthe salt is selected from the group consisting of chloride, perchlorate, hexafluorophosphate, sulfate and nitrate.
    11. A compound according to claim 10, wherein the counter ion is chloride ion.
    12. A compound according any one ofthe preceding claims, which is in the form of a racemate.
    13. A compound according to any one of claims 1 to 11, which is in the form of a diastereomer.
    14. A compound according to any one of claims 1 to 11, which is in the form of a pure enantiomer.
    15. A compound according to claim 1 selected from the group consisting of (2S,3R- butanediamine)(l,10-phenanthroline)platinum(II) cation, (2S,3S-butanediamine)(l,10- phenanthroline)platinum(II) cation , (2R,3R-butanediamine)(l,10- phenanthroline)platinum(II) cation, (lS,2S-cyclohexanediamine)(l, 10- phenanthroline)platinum(II) cation, (lR,2R-cyclohexanediamine)(l, 10- phenanthroline)platinum(II) cation, (N,N-dimethyl-lS,2S-cyclohexanediamine)(l, 10- phenanthroline)platinum(II) cation and (N,N-dimethyl-lR,2R- cyclohexanediamine)( 1 , 10-phenanthroline)platinum(II) cation.
    16. Use of a compound according to any one ofthe preceding claims in the treatment of an antiproliferative disease.
    17. A use according to claim 16, wherein the disease is a cancer.
    18. Use of a compound according to any one of claims 1 to 16 as an antimicrobial agent.
    19. 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
    [ML!L2]2+ I where M is selected from the group consisting of platinum(II), palladium (II) and copper(II);
    Li 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.
    20. A method according to claim 19, wherein the cancer is selected from the group consisting of cancer ofthe esophagus, breast cancer, cancer ofthe ovary, lung cancer, bladder cancer, testicular cancer, cancer ofthe endometrium, head and neck cancer, thyroid cancer, cervical cancer, neuroblastoma, leukemia and osteogenic sarcoma.
    21. A method according to any one of claims 19 to 20, wherein the compound of formula I is used in simultaneously, sequentially or separately with another anticancer therapy selected from surgery, radiotherapy and/or chemotherapy.
    22. A method according to claim 21, wherein the compound of formula I is administered with an anticancer compound, a cytotoxic agent and/or a secondary anticancer agent.
    23. A method according to claim 22, wherein the compound of formula I is administered with another anticancer compound or cytotoxic agent.
    24. A method according to any one of claims 19 to 23, wherein the compound of formula I is a compounds according to any one of claims 2 to 15.
    25. A pharmaceutical composition comprising a compound in accordance with formula I
    I where M is selected from the group consisting of platinum(LI), palladium (IL) and copper(II);
    Li 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, 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.
    26. A pharmaceutical composition according to claim 25 in the form suitable for oral, parenteral or rectal administration.
    27. A composition according to claim 26 in an injectable form.
    28. A composition according to claim 26 in a dosage form" selected 4om the group consisting of tablets, capsules and powders.
    30. A composition according to any one of claims 25 to 29, wherein the compound of formula I is a compound according to any one of claims 2 to 15.
AU2002231463A 2001-02-23 2002-02-22 Metal complexes and therapeutic uses thereof Ceased AU2002231463B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002231463A AU2002231463B2 (en) 2001-02-23 2002-02-22 Metal complexes and therapeutic uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
AUPR3302A AUPR330201A0 (en) 2001-02-23 2001-02-23 Metal complexes and therapeutic uses thereof
AUPR3302 2001-02-23
PCT/AU2002/000167 WO2002066435A1 (en) 2001-02-23 2002-02-22 Metal complexes and therapeutic uses thereof
AU2002231463A AU2002231463B2 (en) 2001-02-23 2002-02-22 Metal complexes and therapeutic uses thereof

Publications (2)

Publication Number Publication Date
AU2002231463A1 true AU2002231463A1 (en) 2003-02-27
AU2002231463B2 AU2002231463B2 (en) 2008-08-21

Family

ID=3827319

Family Applications (2)

Application Number Title Priority Date Filing Date
AUPR3302A Abandoned AUPR330201A0 (en) 2001-02-23 2001-02-23 Metal complexes and therapeutic uses thereof
AU2002231463A Ceased AU2002231463B2 (en) 2001-02-23 2002-02-22 Metal complexes and therapeutic uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AUPR3302A Abandoned AUPR330201A0 (en) 2001-02-23 2001-02-23 Metal complexes and therapeutic uses thereof

Country Status (9)

Country Link
US (1) US7410960B2 (en)
EP (1) EP1377552B1 (en)
JP (1) JP4294957B2 (en)
AT (1) ATE326450T1 (en)
AU (2) AUPR330201A0 (en)
CA (1) CA2448251C (en)
DE (1) DE60211500T2 (en)
NZ (1) NZ528415A (en)
WO (1) WO2002066435A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002364756A1 (en) * 2002-11-12 2004-06-03 Zakrytoe Aktsionernoe Obschestvo "Asgl-Issledovatelskie Laboratorii Palladium complexes with heterocyclic ligands
CA2530279A1 (en) * 2003-06-27 2005-01-06 Akira Odani Bisphosphonate complexes
US20060264414A1 (en) * 2005-04-08 2006-11-23 The Board of Regents, University and Community College System of Nevada on behalf of the Fluorous 2,2'-bipyridines and fluorinated biphasic sysems for ligand design
EA010431B1 (en) * 2006-08-17 2008-08-29 Общество С Ограниченной Ответственностью "Радифенс" Palladium-based coordination compound and method for preparing thereof
EP2398484A4 (en) 2009-02-23 2013-07-10 Inter K Pty Ltd Inhibition of multiple cell activation pathways
TWI380988B (en) * 2009-12-31 2013-01-01 Univ Nat Cheng Kung Platinum complex and methods for making platinum complex and platinum catalyst therethrough
SG185722A1 (en) 2010-06-04 2013-01-30 Univ Ohio Phosphaplatins and their use for treatment of cancers
GR1009550B (en) * 2018-02-20 2019-07-01 Χριστινα Αννα Δημητριου Μητσοπουλου Bivalent copper compounds with natural products of pyridyl quinoxaline and nitrates exhibiting anti-fugal and anti-cancer action - multi-target medications
CN110746466B (en) * 2019-10-22 2023-08-15 合肥工业大学 Preparation and application of chiral platinum complex
CN111991419A (en) * 2020-09-10 2020-11-27 中国医学科学院基础医学研究所 Use of platinum-containing particles for the preparation of a medicament and/or a medical product for inducing the differentiation of leukemia cells

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2002746A (en) * 1977-06-17 1979-02-28 Gist Brocades Nv Copper complexes
JPH0232086A (en) 1988-07-20 1990-02-01 Toray Ind Inc Novel platinum-containing compound and malignant tumor remedy
MX172248B (en) 1989-12-20 1993-12-09 Univ Mexico PROCEDURE FOR THE OBTAINING OF NEW MIXED COPPER AMINO ACID COMPLEXES BASED ON FENYLATED PHENANTROLINS AS ANTI-CANCER AGENTS
MX172967B (en) 1989-12-20 1994-01-26 Univ Mexico PROCEDURE FOR THE OBTAINING OF NEW MIXED COPPER AMINO ACID COMPLEXES BASED ON FENANTROLINS AND THEIR DERIVATIVES RENTED AS ANTI-CANCER AGENTS
JPH06287021A (en) * 1992-04-22 1994-10-11 Tanaka Kikinzoku Kogyo Kk Optical resolution of optically active platinum complex compound

Similar Documents

Publication Publication Date Title
US8980875B2 (en) Platinum-N-heterocyclic carbene derivatives, preparation thereof and therapeutic use thereof
EP1377552B1 (en) Metal complexes and therapeutic uses thereof
AU2002231463A1 (en) Metal complexes and therapeutic uses thereof
CA2560059A1 (en) Platinum carboxylate anticancer compounds
ITMI940610A1 (en) PLATINUM TRINUCLEAR CATIONIC COMPLEXES FOR ANTI-CANCER ACTIVITIES AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US6313333B1 (en) Multinuclear cationic platinum complexes with antitumor activity
US9220705B2 (en) Method of treating colorectal cancer
US7928097B2 (en) Photoreactive compounds and compositions
EP0793667B1 (en) Trinuclear cationic platinum complexes having antitumour activity and pharmaceutical compositions containing them
US10973849B2 (en) Rhenium complexes and methods of use for treating cancer
US6001872A (en) Water soluble transplatinum complexes with anti-cancer activity and method of using same
US6921824B1 (en) Ruthenium dimeric complexes suitable as antimetastatic and antineoplastic agents
JPH02212497A (en) Novel platinum complex and production thereof
KR20120101798A (en) Novel hexanuclear arene-ruthenium nano prismatic cage compound, preparation method thereof and pharmaceutical composition for preventing and treating cancer as active ingredient
EP2404925A1 (en) New all trans platinum (IV) complexes, their preparation and their use as antitumor agents
ITMI20081660A1 (en) PLATINUM COMPLEX WITH ANTITUMOR ACTIVITY
JPH049393A (en) New platinum complex and tumor remedy comprising the same complex as active ingredient