WO2010015590A1 - Organotin(iv) complexes with n-acetylcysteine possessing antitumoral activity, process for their production and their use - Google Patents

Organotin(iv) complexes with n-acetylcysteine possessing antitumoral activity, process for their production and their use Download PDF

Info

Publication number
WO2010015590A1
WO2010015590A1 PCT/EP2009/060006 EP2009060006W WO2010015590A1 WO 2010015590 A1 WO2010015590 A1 WO 2010015590A1 EP 2009060006 W EP2009060006 W EP 2009060006W WO 2010015590 A1 WO2010015590 A1 WO 2010015590A1
Authority
WO
WIPO (PCT)
Prior art keywords
acetylcysteine
compounds
organotin
carbon atoms
cells
Prior art date
Application number
PCT/EP2009/060006
Other languages
French (fr)
Inventor
Claudia Pellerito
Cristina Prinzivalli
Ornella Pellerito
Michela Giuliano
Original Assignee
Universita' Degli Studi Di Palermo
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universita' Degli Studi Di Palermo filed Critical Universita' Degli Studi Di Palermo
Publication of WO2010015590A1 publication Critical patent/WO2010015590A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/32Tin compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/22Tin compounds
    • C07F7/226Compounds with one or more Sn-S linkages

Definitions

  • the present invention relates to novel organotin(IV) derivatives showing antitumour activity, a process for their production and their use in medicine.
  • the invention relates to the synthesis and the study of organotin(IV) derivatives with N-acetylcysteine (NAC) which exhibit excellent selective antitumoral in vitro activities against several cencer cell lines and particularly human hepatocarcinoma HepG2, thus their are suggested for producing drugs for tumors treatment, both alone and mixed with other chemotherapeutic agents.
  • NAC N-acetylcysteine
  • organotin(IV) complexes wherein the metal acceptor is coordinated by a biological organic ligand, containing in particular sulphur ligands, carboxylates, or amine groups.
  • Organotin(IV) compounds which are generally very toxic, even at low concentration, starting from the '50s years of the XX century they were used for industrial applications at first as catalyst in PVC then for agricultural applications as fungicides and miticides.
  • organotin(IV) compounds One of the major and widespread application of organotin(IV) compounds is the use as active ingredients in antifouling marine paint, for preventing biological growths on submerged surfaces in fresh and sea water.
  • organotin(IV) derivatives display antitumoral activity.
  • Ph 3 Sn acetate was evaluated for its inhibitory effects in tumour growth.
  • the biological activity is essentially determined by the number and nature of the organic group bound to the central Sn atom and that di-organotin(IV) derivatives show the highest antitumor activity combined with the lowest mammalian toxicity, while tri- organotin(IV) derivatives are generally very toxic.
  • organotin derivatives suggested as antitumoral drug are disclosed in the patent publications EP 0484596 A1 and EP 0677528 A1 (Gielen et al., both with Pharmachemie B. V. as applicant), and EP 0538517 A1 (Boualam et al., Pharmachemie B. V.).
  • R 2 SnX 2 L 2 derivatives show a biological activity whose mode of action differs from that of the similar cis-diammineplatinum(ll) family drugs from which they have been designed.
  • the c/s-diamminedichloroplatinum(ll) characterized by a Cl-Pt-Cl- bond angle less then 95°, is active in vivo only after that the chloride ion is displaced; that allows the platinum to react with DNA nucleobases.
  • This mechanism of action is theoretically also possible for diorganotin(IV) derivatives, and in fact it is known that organotin(IV) form adducts with purines and pyrimidines.
  • crystallographic data for both active or inactive R 2 SnX 2 L 2 derivatives revealed an average Cl-Sn-Cl bond angle less then 103° which may implicate an activation mechanism similar to that of cis-diammineplatinum(ll).
  • apoptosis is a physiological process of cell death, which is also called “programmed cell death” or PCD because responds to a genetically determined program that is carried out actively by the cells and that requires energy, gene transcription and protein synthesis.
  • Apoptosis, with proliferation, differentiation and cell migration, is a molecular program which is involved in the regulation of the size of tissues and in the maintenance of cell number and cellular homeostasis.
  • the apoptotic mechanism is also activated as a defense mechanism to protect the integrity of organisms when the cells are subjected to different damages. Thus prevents the propagation of cell population with abnormal genetic and irreversible damages.
  • Apoptosis has biochemical and morphological aspects different to necrosis, another type of cell death. Necrosis does not occur through a specific genetic program, unlike apoptosis which is also called "clean death"; it is not a private event that affects only the cell where it turns on, but necrosis extends to many cells in a diffuse inflammatory event that culminates with the swelling of cellular organelles and cell lysis.
  • apoptosis a sequence of events which are common to different cell types occurs and remain constant over the course of phylogenesis. This process begins with a rapid morphological cell transformation, consisting in the reduction of cell volume, cytoskeleton modification, loss of contact between the cells and their separation from original tissue. In the nucleus, the nuclear lamina is degraded and the chromatin is condensed and fragmented and then move to the perinuclear region. Thus the formation of vesicles containing chromatin occurs, which move towards the plasma membrane and are surrounded by the evagination of a plasma membrane. The apoptotic cell takes a look as bubbles, commonly defined blebbing.
  • drugs which are currently used in anticancer therapies act by activating cytotoxic death by apoptosis.
  • the goal of anticancer research is the design of drugs capable of inducing preferentially apoptosis in tumoral cytotypes without affect non-tumoral cells, and to identify the mechanism of activation of apoptosis and the factors associated with it.
  • the caspases are a family of cytosolyc cysteine proteases which are synthesized in proenzimatic form and then are activated by proteolytic cleavage at a specific aspartate residue within a cascade through which functional enzymes are generated.
  • the caspases can be separated into two functionally different groups: the early caspases, as the caspases 2, 8, 9 and 10, and the effector caspases, as the caspases 3, 6 and 7.
  • the caspase activation may occur not only by proteolytic cleavage but also through association with regulatory proteins, such as the pro-caspase 9.
  • the effector caspases act during the final phase of apoptosis on the level of nuclear lamina, ICAD (inhibitor of caspase activated DNase) and PARP (poly-ADP-ribose polymerase).
  • extrinsic pathway is consequent to the linkage of several proteins such as FasL or TNF to death receptors of TNF family (for example Apo/Fas/CD95, TNF- ⁇ , DR4 and DR5) which are localized on cell membrane and mediate various physiological responses such as inflammation, proliferation, antiviral activity and cellular death.
  • the receptor-ligand binding induces the receptor activation, that form trimers and contact, specific cytoplasmatic proteins (for example FADD) through the intracellular portion and this culminates with the activation of caspases cascade.
  • the intrinsic pathway involves the mitochondria and is activated by intracellular events (for example DNA damage, oxidative stress) and also by extracellular events (cytotoxic compound administration, ionizing radiation). These signals converge on mitochondria and cause the dissipation of mitochondrial membrane potential, release in cytosol of cytochrome-c which activates caspase-9 into apoptosome and consequently the executive caspases. It is known that the dissipation of mitochondrial membrane potential may occur as a result of cytoplasmatic ROS (reactive oxygen species) accumulation, after strong oxidative stress, often associated with the mechanism of action of several compounds. During apoptotic activation of mitochondria, several factors of Bcl-2 family are commonly involved.
  • cytoplasmatic ROS reactive oxygen species
  • pro-apoptotic Bak, Bok, Bad, Bim, BcIXs, Bid, Noxa, Puma
  • anti-apoptotic Bcl-2, BclX L , Mcl-1
  • the anti-apoptotic factors moves from the cytosol, where they are inactive, to the mitochondria and then they are activated through different mechanisms.
  • the pro-apoptotic members facilitate the release of cytochrome-c through the formation of channels and compete with the anti-apoptotic factors, which are opposed to these events.
  • This invention relates to anti-tumor organometallic complexes, and specifically to organotin(IV) compounds with ligands of biological origin, said compounds inducing apoptosis in tumoral cells, promising as selective drug for cancer therapy.
  • the above compounds are particular interesting for use in the treatment of hepatocellular carcinoma, colorectal cancer, leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney.
  • Hepatocellular carcinoma is a tumour that develops slowly both in cirrhotic and healthy liver and it is one of the most frequent tumours in the world with unfavourable prognosis: it is estimated that the mortality rate associated with HCC is more then 90%.
  • liver resection represent the only radical treatment of HCC.
  • Liver transplantation needs a patient selection: small and liver limited disease without secondary tumours or metastasis are the fundamental requirements.
  • Other palliative and controversial treatments are radiotherapy, that seems to be palliative and controversial, and systemic chemotherapy, using the drugs combination of fluorouracil (5-FU), adriamycin and doxorubicin.
  • Hepatic arterial (HA) infusional chemotherapy possesses a number of constraints not found in systemic chemotherapy and it is complementary to i.v. drug administration.
  • the drug used should have activity in a dose-responsive way without significant hepatic toxicity.
  • Selective drug administration in the hepatic artery allows the delivery of active agents at a higher local concentration while it reduces the peripheral toxic effects of the cytotoxic drug.
  • the concept of arterial injected metabolic radiotherapy is based on the liver's double vascularization (hepatic artery, portal vein) and on the fact that HCCs are hypervascularized tumours which are mainly fed by the hepatic artery while 80% of the liver unaffected by the tumour is vascularized by the portal vein.
  • HCCs are hypervascularized tumours which are mainly fed by the hepatic artery while 80% of the liver unaffected by the tumour is vascularized by the portal vein.
  • a high "tumour/unaffected liver” ratio which enables us to administer a considerable tumour targeted dose, preserving the healthy part of the liver as long as possible.
  • the recent advances in imaging technology allow a more selective approaches, producing selective ischemia within the tumour while inducing decreased collateral damage to the tumour free liver parenchyma.
  • That interventistic radiological procedures includes chemoembolization and alcoholization, suitable for cirrhotic patients or for patients with unreasectable HCC with large or multiple lesions.
  • Colorectal cancer is the third most common form of cancer and the second leading cause of cancer-related death in the Western world and most commonly it spreads to the liver.
  • one regimen involves the combination of infusional 5- fluorouracil (5-FU) and leucovorin (LV), methotrexate (toxic for retina), epirubicine, paclitaxel (Taxol, Patene), daunorubicin (cardiotoxic), irinotecan or oxaliplatin.
  • the latter is the complex (R,R)-1 ,2-diaminocyclohexane(ethanedioate-0,0) platinum; its cytotoxicity results from inhibition of DNA synthesis by causing crosslinking of
  • the 5-FU is an antimetabolite that causes leucopenia, platelets decreasing, hepatotoxicity and pulmonary toxicity, cardiotoxicity, and neurotoxicity.
  • oxaliplatin is toxic for SNC, hematopoietic system, gastrointestinal tract and allergic reactions can occur.
  • This invention provides novel organotin(IV) complexes with the organic ligand N- acetylcysteine, a N-acetyl derivative of the natural occurring sulphur-containing amino acid L-cysteine, of formula:
  • the compounds of the invention surprisingly show a strong anti-tumour activity, resulting from in vitro cytotoxicity studies on several cancer cellular lines.
  • the compound of the invention showed excellent antitumoral activity in vitro towards hepatoblastoma cells (HepG2), leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney cell lines.
  • HepG2 hepatoblastoma cells
  • the in vitro cytotoxicity was also corroborated by a cytofluorimetric analysis of the cell-cycle-dependent DNA content distribution to probe several aspects of drug-induced cytotoxicity, like apoptosis and by other biological studies described later.
  • the organic ligand used in organotin(IV) complexes is a low molecular weight cysteine derivative showing antioxidant activity both in vitro and in vivo, acting as free radicals or hydrogen peroxide scavenger by reduction.
  • the N-acetylcysteine (NAC) is a drug commonly used as a mucolytic agent in the management of acute or chronic bronchopulmonary diseases.
  • HCC Hepatocellular carcinoma
  • hepatocarcinoma HCC In Caucasian populations in industrialized countries and in Japan the hepatocarcinoma HCC is mainly associated with C or B virus cirrhosis (70-80% of patients); usually post hepatic cirrhosis, less frequently alcoholic cirrhosis or secondary to hemocromatosis.
  • Human hepatoma HepG2 cell line is characterized (from a biomolecular point of view) by an alteration of ⁇ catenin gene, caused by a point mutation or a deletion of exons 3 and 4 (aa 25-140). This deletion removes the potential site of regulation by glycogen synthase kinase 3 ⁇ (GSK-3 ⁇ ) responsible for its phosphorylation.
  • ⁇ -catenin accumulates in the cytoplasm and moves into the nucleus where, after the association with the transcription factor TCF/LEF, activates genes involved in cell cycle progression and protection from apoptosis.
  • ⁇ catenin alteration can contribute to the deregulated proliferation of hepatoma cells, but also to the acquisition of resistance to apoptosis.
  • HepG2 cells represent, therefore, a good hepatoma model to study the biochemical mechanisms which are involved in the apoptotic mechanism.
  • the hepatic Chang liver cells employed as non tumoral cell model, are immortalized cells in active proliferation, as beyond the genetic program of senescence.
  • organotin(IV) derivatives of N- acethyl-L-cysteine that could be synthesized with high yields and low costs by simple reaction of commercial reagents, wherein there are two or three organic groups linked to the tin, show excellent anti-tumor activity inducing apoptosis; at the same time compounds exhibit higher selectivity and lower toxicity then those of the corresponding organotin(IV) parents.
  • this invention provides diorganotin(IV) and triorganotin(IV) complexes with N-acethylcysteine which exhibit high toxicity against human hepatocarcinoma cell lines, but they are less toxic, being some of them not toxic, towards normal human hepatocyte cell line. They are all also less toxics then the corresponding organotin(IV) parents used as reagents for their synthesis.
  • the toxicity is time and concentration dependent: in detail it seems that biological ligand, constituting the complexes, modulates the toxic effects of the organometallic moiety.
  • the cell toxicity occurs by apoptosis induction, as it has been showed and confirmed by biochemical study on caspases expression.
  • organotin(IV) derivatives disclosed in this invention could represent a targeted specific anticancer treatment with high selectivity, and affecting poorly the physiology of normal cells.
  • the compound of the invention showed excellent antitumoral activity in vitro also towards leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney cell lines.
  • organotin(IV) complexes with N- acethylcysteine disclosed in this invention were previously described from a chemical point of view (i.e. G. Domazetis et al., in Inorg.Chim.Acta, (1979), Vol.32,L48-L50; G. Domazetis et al., Tri-n-butyltin(IV) derivatives of L-cysteine ethyl ester, N-acetyl-L-cysteine and ⁇ -glutamylcysteinyl glicine (Glutathione reduced), J.
  • R 15 R 2 and R 3 are each independently selected from the group consisting of: H, an alkyl, alkoxyl, alkenyl or alkynyl group with up to 12 carbon atoms, an isocyclic or heterocyclic, aromatic or aralkyl group with up to 12 carbon atoms, for use as medicinal active ingredients.
  • R- ⁇ ,R 2 and R 3 are each independently selected from the group consisting of: H, an alkyl group with up to 8 carbon atoms, an alkoxyl group with up to 8 carbon atoms, a phenyl group.
  • diorganotin(IV) complexes with N-acetylcysteine either among triorganotin(IV) complexes with N-acetylcysteine disclosed for medical use according to this invention and having respectively the general formula (1 ) and (2)
  • preferred compounds are those wherein R 1 , R 2 and R 3 (when present) are each independently selected in the group consist in methyl, ethyl, propyl, butyl and phenyl groups.
  • R 1 and R 2 are each independently selected from the group consisting of: H, an alkyl, alkoxyl, alkenyl or alkynyl group with up to 12 carbon atoms, an isocyclic or heterocyclic, aromatic or aralkyl group with up to 12 carbon atoms, that are not described in literature for their putative use as antitumoral agents selective for hepatic and colorectal cancer, particularly preferred is dibutyltin(IV) acetylcisteinate complex, of formula (1 a):
  • organotin(IV) complexes with N-acetylcysteine of general formula (1 ) and (2) can be synthesized by reaction of N-acetylcysteine and the corresponding organotin(IV) precursors.
  • Further object of the present invention is a process for producing organotin complexes with N-acetylcysteine with general formula (1 ) or (2), as above described, said process including reacting an organotin(IV) derivative wherein R 1 , R 2 and R 3 (when present) have the meaning above already said, with N- acetylcysteine in methanol, chloroform or water, in a temperature range comprised between 10 °C and 90 °C, preferably between 25 °C and 75 °C.
  • the reaction mixture is quenched by cooling, and the resulting precipitate containing the desired compound of formula (1 ) or (2) is collected by filtration.
  • organotin(IV)complexes with N-acetylcysteine with general formula (1 ) or (2) that includes preferably the reaction between organotin(IV)derivative with N- acetylcysteine in refluxing methanol.
  • the reaction mixture is quenched by cooling, and the resulting precipitate containing the desired compound of formula (1 ) or (2) is collected by filtration.
  • Further aspect of the invention is the use of one ore more compounds having general formula (1 ) or (2), as above described, for the production of a pharmaceutical preparation, in detail an anticancer preparation, and preferably as active ingredient in an anticancer preparation for the treatment of hepatic and colorectal tumours as well as for leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney.
  • the compounds above described can be advantageously used in cancer therapy, administered in a pharmaceutical preparation as toxic agents by themselves or in combination with other chemotherapeutic agent for enhancing the therapeutic effects, and for reducing pharmacoresistences.
  • This invention relates also to a pharmaceutical preparation, including as active component at least one of the organometallic complexes with N-acetylcysteine with general formula (1 ) or (2) together with one ore more pharmaceutically acceptable adjuvants and/or vehicles.
  • the preparation contains as active component dibutyltin(IV) -N-acetylcisteinate complex, of formula (1 a).
  • the pharmaceutical preparations containing organotin(IV) derivatives with N-acetylcysteine suitables for the therapeutical administration can be designed using conventional methods already known by expertises, by using adjuvants and vehicles pharmaceutically acceptable, for obtaining preparations suitable for parenteral injection (intravenous, intramuscular or subcutaneous), for oral administration of solid or liquid formulations, for transdermal, rectal, intravaginal or topical administration, for instance in oronasal mucosa and similar.
  • the percentage of the active compound in the preparation and the method of tumour treatment could be changed for reaching the optimal dosage.
  • the dosage to administer depends on: route of administration, length of treatment, patient conditions and weight, drug effectiveness and patient reaction.
  • the formulation and the choice of adjuvants and vehicles pharmacologically suitable for each need will be made according to the current knowledge in this field.
  • the specific characteristics of the invention, as well as the advantages either for the methods for synthesizing the suggested compounds, either for their biological and pharmacological activity, will be remarked afterwards or in the detailed illustrative description and experimental results related to this invention. Brief description of the figures
  • the figures 1A and 1 B show the effects induced by the compound Bu 2 SnNAC, according to the invention, in comparison with Bu 2 SnCI 2 and NAC, on hepatic HepG2 and Chang liver cell viability, respectively after 8 h (A) and 16 h (B) of treatment;
  • the figure 2 shows the influence of NAC on the cytotoxic effect induced by Bu 2 SnCI 2 in HepG2 cells according to the invention;
  • the figure 3 shows the involvement of caspases in apoptosis induced by Bu 2 SnNAC complex, according to the invention, compared with Bu 2 SnCI 2 , in HepG2 cells;
  • the figure 4 shows the effects of the Bu 2 SnNAC complex, according to the invention, and the others used for a comparison, on the levels of PAR4 factor and the survival protein pAKT.
  • the figures 5A and 5B show the effect of the compound NAC2 against 60 cancer cell lines at a single dose of 10 ⁇ M.
  • the 119 Sn M ⁇ ssbauer spectra were recorded by maintaining the source at room temperature and by cooling the sample, as fine powder pressed between two aluminium foil, at the temperature of 77.3 ⁇ 0.1 K, using a liquid nitrogen cryostat NRD-1258-MB (Cryo Industries of America inc., Atkinson, NH, USA) and using an ITC 502 Oxford Instrument temperature controller to obtain the temperature control.
  • N-acetylcysteine (NAC) (1 mmol,163.18 mg) was dissolved in 30 ml of methanol or water, then added slowly to a dimethyltin(IV) oxide (Me 2 SnO) solution (1 mmol, 164.78 mg), under stirring. After 24 hours at range 25-75 °C, the white solid derivative was recovered after filtration under vacuum and vacuum drying on
  • N-acetylcysteine (NAC) (163.18 mg, 1 mmol) was dissolved in 30 ml of methanol or water, then added slowly to a dibutyltin(IV) oxide (Bu 2 SnO) solution (1 mmol
  • the Ph 2 SnNAC complex was obtained as white solid, mixing a diphenyltin(IV) oxide (Ph 2 SnO) aqueous suspension and a N-acetylcysteine aqueous solution, in molar ratio1 :1 .
  • This reaction allowed the complex formation, without unreacted reagents or secondary products.
  • N-acetylcysteine NAC (163.18 mg, 1 mmol) was dissolved in 30 ml of distillate water, then added slowly to a diphenyltin(IV) oxide (Ph 2 SnO) aqueous suspension
  • (Me 3 Sn) ⁇ NAC complex synthesis The (Me 3 Sn) 2 NAC complex was obtained as white solid, mixing a trimethyltin(IV) hydroxide (Me 3 SnOH) and a N-acetylcysteine methanolic solutions, in molar ratio
  • N-acetylcysteine NAC (163.18 mg, 1 mmol) was dissolved in 30 ml of methanol and then added slowly to a trimethyltin(IV) hydroxide (Me 3 SnOH) methanolic solution (2 mmol, 180.82 mg), under stirring. After 24 hours at range 25-75 °C, the solution volume was reduced to 10ml by a rotating evaporator.
  • the (Bu 3 Sn) 2 N-acetylcysteinate complex was obtained as gel mixing a tributhyltin(IV) hydroxide ((Bu 3 Sn) 2 O) and a N-acetylcysteine methanolic solutions, in molar ratio 2:1.
  • This reaction allowed the complex formation, without unreacted reagents or secondary products.
  • the (Ph 3 Sn) 2 N-acetylcisteinate complex was obtained as white solid, mixing a triphenyltin(IV) hydroxide (Ph 3 SnOH) and a N-acetylcysteine chloroformic or methanolic solutions, in molar ratio2:1.
  • N-acetylcysteine (NAC) (163.18 mg, 1 mmol) was dissolved in 30 ml of chloroform (chloroform or methanol) , then added slowly to a Ph 3 SnOH chloroformic solution (2 mmol; 367.01 mg), under stirring. After 24 hours at range 25-75 °C, the solution volume was reduced to 10ml by a rotating evaporator.
  • reaction mixture was quenched at fridge temperature, and after 24 hours the resulting white desired precipitate was collected by filtration, washed with chloroform and then vacuum dried on P 4 O 10 .
  • N-acetyl-L-cysteine N-acetyl-L-cysteine
  • R 2 Sn(IV)NAC diorganotin
  • NAC N-acetyl-L-cysteine
  • the isomer shift values ⁇ , mms "1 , reflect the meaning of the parameter, increasing their value according to the inductive effects of the organic groups, so they decrease on going from dialkyl to diphenyl derivatives.
  • ⁇ R ⁇ is the p.q.s. of the alkylic and phenylic group in an idealised octahedral configuration (-1.03 mms “1 and -0.95 mms “1 , respectively) and ⁇ is the measured quadrupole splitting:
  • the table contains Mossbauer parameters of the organometallic compounds in object, as isomer shift, ⁇ ,in mm s ⁇ together with the and experimental nuclear quadrupole splitting [
  • organotin(IV) with N-Acetylcysteine complexes was evaluated at the Department of Biochemical Sciences of University of Palermo, Policlinico "Paolo Giaccone", by using MTT assay to evaluate their cytotoxicity, cytofluorimetric analysis to identify cell cycle distribution for apoptosis quantification and to assess ROS production, Western blotting analysis to evaluate the involvement of caspases, pAKT and Par4 in apoptotic mechanism induced by the compounds.
  • MTT assay to evaluate their cytotoxicity
  • cytofluorimetric analysis to identify cell cycle distribution for apoptosis quantification and to assess ROS production
  • Western blotting analysis to evaluate the involvement of caspases, pAKT and Par4 in apoptotic mechanism induced by the compounds.
  • Chang liver and HepG2 hepatoma cells were cultured in polystyrene 75 cm 2 flasks and grown in RPMI 1640 medium supplemented with 10% (v/v) heat- inactivated foetal calf serum, 2.0 mM L-glutamine, 1.0 mM sodium pyruvate and antibiotic antimycotic solution (100 U/ml penicillin, 100 Dg/ml streptomycin and 250 ng/ml amphotericin B) and incubated at 37 °C in a humidified, 5% CO 2 atmosphere, 95% air.
  • RPMI 1640 medium supplemented with 10% (v/v) heat- inactivated foetal calf serum, 2.0 mM L-glutamine, 1.0 mM sodium pyruvate and antibiotic antimycotic solution (100 U/ml penicillin, 100 Dg/ml streptomycin and 250 ng/ml amphotericin B) and incubated at 37 °C in a humidified
  • Cell viability was determined after detachment of cells from flask by Trypsin - EDTA solution and seeding of 6.5x10 3 Chang liver cells and 10 4 HepG2 cells (on 200 ⁇ l of medium) in 96-well cell culture plates (0.3 cm diameter).
  • MTT Cell viability assay is a quantitative colorimetric assay that employs the 3-[4,5- dimethylthiazolyl-2] 2,5-diphenyl-tetrazolium bromide (MTT) (T Mosmann, J.Immunol. Methods, 1983, 65, 55-63). Tetrazolium salt MTT is cleaved to blue formazan by mitochondrial dehydrogenases of vital cells , which belongs to the mitochondrial respiratory chain. Since this enzymatic system is active only in viable (metabolically active) cells, this assay exclusively detects viable cells. The intensity of staining formed is directly proportional to the number of living cells and is measured by means of spectrophotometer.
  • MTT 3-[4,5- dimethylthiazolyl-2] 2,5-diphenyl-tetrazolium bromide
  • MTT stock solution was added to each well (final concentration 1 mg/ml) for 2 hours at 37 °C.
  • the medium was then removed and 0.2 ml of lysis buffer (obtained by dissolving 20% (p/v) SDS (sodium dodecilsulphate) in 50% N,N-dimethylformamide, pH 4.7) was added to dissolve the formazan product.
  • lysis buffer obtained by dissolving 20% (p/v) SDS (sodium dodecilsulphate) in 50% N,N-dimethylformamide, pH 4.7
  • the absorbance at 570 nm (test wavelength) and at 630 nm (reference wavelength) was measured using an ELISA microplate reader (OPSYS MR, Dynex Technologies). Lysis; buffer was employed as the reference test to reset the instrument.
  • the toxicity induced by 10 ⁇ M Bu 2 SnCI 2 added to the cells after the pre-incubation with NAC is significantly greater than that observed in the presence of Bu 2 Sn N-Acetylcysteinate complex.
  • the binding with N-Acetylcysteine significantly modulates the cytotoxic effect of the organometallic portion, probably without that any degradation occurs within the cell.
  • Pl propidium iodide
  • This method allows to evaluate the cell distribution along the phases of proliferative cycle. The presence of a peak of lower fluorescence intensity indicates the presence of cells with fragmented DNA.
  • ROS reactive oxygen species
  • Equal amounts of protein samples (60 ⁇ g/lane) were diluted in 1 X Laemmli buffer (constituted by 0.0625 M Tris, pH 6.8, 2% SDS, 5% ⁇ -mercaptoethanol, 10% glycerol, 0.02% bromophenol blue) (U.K. Laemmli, 1970, Nature, 227, 680- 685), denatured at 90 °C for 5 minm, and subjected to SDS-PAGE in running buffer 1 x (25 mM Tris, 0.1 % SDS, 1.44% glycine, pH 9.0) for about 2 hours at 150 V.
  • 1 X Laemmli buffer constituted by 0.0625 M Tris, pH 6.8, 2% SDS, 5% ⁇ -mercaptoethanol, 10% glycerol, 0.02% bromophenol blue
  • Bu 2 SnCI 2 induced the decrease of the inactive form of protein (57 KDa), but the appearance of the active form of enzyme, indicated as a band at lower molecular weight, was not observed.
  • the decrease of pro-caspase-8 (which was considered as expression of activation in caspase-8) is observed after 8 hours of incubation with the compound and it is more visible after 16 hours. Similar effects were induced also in executioner caspase-9 and caspase-3 proteins.
  • PAR-4 protein is a product of pro-apoptotic gene which induces apoptosis in several tumor cytotypes. This function is induced after phosphorylation in Thr163 by PKA with the consequent migration to the nucleus, where it is able to induce the expression of factors that promote apoptosis (for example through FasL-Fas- FADD-caspase axis) and inhibit the expression of factors which are opposed to it (for example Bcl2 and NF-kB proteins).
  • PAR-4 The inhibition of pro-apoptotic action of PAR-4 occurs as a consequence of the phosphorylation on the residue Ser 249 by pAKT protein, which sequesters it in the cytoplasm.
  • PAR-4 also appears to modulate pAKT through PTEN activation, a fosfatydilinositol-3-phosphate phosphatase which has a strong inhibitory effect on PI-3 kinase by reverting the phosphorylation status and determine accordingly the lack of activation of survival factor p A KT Effect of compounds on the levels of pAKT factor.
  • NAC2 activity was assayed against 60 different human tumor cell lines, representative for leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney. The aim was to evaluate a selective growth inhibition or cell killing of particular tumor cell lines induced by NAC2.
  • the screening is a two-stage process, beginning with the evaluation of the compound against the 60 cell lines at a single dose of 10 uM. The output from the single dose screen is reported as a mean graph and is available for analysis by the COMPARE program. We report the results of this one dose test. The compound exhibited significant growth inhibition.
  • the human tumor cell lines of the cancer screening panel were grown in RPMI 1640 medium containing 5% foetal bovine serum and 2 mM L-glutamine. Cells were inoculated into 96 well microtiter plates in 100 ⁇ l_ at plating densities ranging from 5,000 to 40,000 cells/well depending on the doubling time of individual cell lines. After cell inoculation, the microtiter plates were incubated at 37° C, 5 % CO2, 95 % air and 100 % relative humidity for 24 h prior to addition of experimental drugs. After 24 h, two plates of each cell line were fixed in situ with TCA, to represent a measurement of the cell population for each cell line at the time of drug addition (Tz).
  • TCA time of drug addition
  • the plates were incubated for an additional 48 h at 37°C, 5 % CO2, 95 % air, and 100 % relative humidity.
  • the assay is terminated by the addition of cold TCA.
  • Cells were fixed in situ by the gentle addition of 50 ⁇ l of cold 50 % (w/v) TCA (final concentration, 10 % TCA) and incubated for 60 minutes at 4°C. The supernatant was discarded, and the plates were washed five times with tap water and air dried.
  • Sulforhodamine B (SRB) solution 100 ⁇ l
  • 0.4 % (w/v) in 1 % acetic acid was added to each well, and plates were incubated for 10 minutes at room temperature.
  • One-dose data are reported as a mean graph of the percent growth of treated cells .
  • the number reported for the One- dose assay is growth relative to the no-drug control, and relative to the time zero number of cells. This allows detection of both growth inhibition (values between 0 and 100) and lethality (values less than 0). For example, a value of 100 means no growth inhibition. A value of 40 would mean 60% growth inhibition. A value of 0 means no net growth over the course of the experiment. A value of -40 would mean 40% lethality. A value of -100 means all cells are dead. Information from the
  • the figure 6 shows the results on 60 different human tumor cell lines.
  • the compound NAC2 showed excellent antitumoral activity in vitro towards leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney.

Landscapes

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

Abstract

Organotin(IV) compounds with N-acetylcysteine having the general formula (1 ) o (2): wherein: R1, R2 and R3 are each independently selected from the group consisting of : H, an alkyl, alkoxyl, alkenyl or alkynyl group with up to 12 carbon atoms, an isocyclic or heterocyclic aromatic or aralkyl group with up to 12 carbon atoms, and they are preferably chosen from methyl, ethyl, butyl or phenyl, are synthetized by simple procedures from commercial products, and are active as anticancer agents. In particular, the Bu2Sn-N-acetylcysteinate derivative was found to have a potent and selective anticancer activity in vitro against several cancer cell lines.

Description

ORGANOTIN(IV) COMPLEXES WITH N-ACETYLCYSTEINE POSSESSING ANTITUMORAL ACTIVITY, PROCESS FOR THEIR PRODUCTION AND THEIR USE
Field of the invention The present invention relates to novel organotin(IV) derivatives showing antitumour activity, a process for their production and their use in medicine. In particularl, the invention relates to the synthesis and the study of organotin(IV) derivatives with N-acetylcysteine (NAC) which exhibit excellent selective antitumoral in vitro activities against several cencer cell lines and particularly human hepatocarcinoma HepG2, thus their are suggested for producing drugs for tumors treatment, both alone and mixed with other chemotherapeutic agents. State of the art
Several inorganic salts and metal complexes are used in medicine for many therapeutic applications, but only two metal based complexes have been really successful in clinical practice as antitumoral agents: the cis- diamminedichloroplatinum(ll) (cis-platin) and cis-diammine-(1 ,1 - cyclobutanedicarboxylato)platinum(ll) (carboplatin). The first one was synthesyzed for the first time by Michele Peyrone and pubblished in 1844 and its tumor growth inhibition properties were discovered only in 1965. The second one, a second generation cis-platin analogous (patent US 4140707, published in 1979 to the Research Corp.), obtained Food and Drug Administration approval in 1989 for second-line treatment of ovarian carcinoma.
Some tumors developed immediately resistance to the mentioned compounds that, in addition, show severe side effects, as nefrotoxicity and hematotoxicity, as well as ototoxicity. Particularly in the last period, the research in metal-based antitumorals derivatives was focused on treatment of tumors resistent to treatment with platinum derivatives. In this concern, very interesting examples are anticancer titanium compounds acting against tumors in the gastrointestinal tract, whereas platinum antitumour derivatives are ineffective towards the same tumors.
In the last years, one of the major developments in the field of metal-based antitumour complexes, that have been synthesized and studied, is the focus on organotin(IV) complexes, wherein the metal acceptor is coordinated by a biological organic ligand, containing in particular sulphur ligands, carboxylates, or amine groups.
Organotin(IV) compounds, which are generally very toxic, even at low concentration, starting from the '50s years of the XX century they were used for industrial applications at first as catalyst in PVC then for agricultural applications as fungicides and miticides. One of the major and widespread application of organotin(IV) compounds is the use as active ingredients in antifouling marine paint, for preventing biological growths on submerged surfaces in fresh and sea water.
It is well known that some organotin(IV) derivatives display antitumoral activity. First results date back to the 1972, when Ph3Sn acetate was evaluated for its inhibitory effects in tumour growth. It is also known that the biological activity is essentially determined by the number and nature of the organic group bound to the central Sn atom and that di-organotin(IV) derivatives show the highest antitumor activity combined with the lowest mammalian toxicity, while tri- organotin(IV) derivatives are generally very toxic.
Many organotin(IV) compounds were assayed against several cancer cell lines and there are several publications in this topic. Probably adducts of the type R2SnX2L2 are the most studied organotin compounds (X= halogen, pseudohalogen; L= ligand containing O, N donors, R= ) but also fluorinated derivatives, bis(stannyl) methanes, compounds containing R2SnO group, derivatives with carboxylic acids, aminoacids, purines, pyrimidines, and peptides. Some examples of organotin derivatives suggested as antitumoral drug are disclosed in the patent publications EP 0484596 A1 and EP 0677528 A1 (Gielen et al., both with Pharmachemie B. V. as applicant), and EP 0538517 A1 (Boualam et al., Pharmachemie B. V.).
The above-mentioned R2SnX2L2 derivatives show a biological activity whose mode of action differs from that of the similar cis-diammineplatinum(ll) family drugs from which they have been designed.
The c/s-diamminedichloroplatinum(ll), characterized by a Cl-Pt-Cl- bond angle less then 95°, is active in vivo only after that the chloride ion is displaced; that allows the platinum to react with DNA nucleobases. This mechanism of action is theoretically also possible for diorganotin(IV) derivatives, and in fact it is known that organotin(IV) form adducts with purines and pyrimidines. Actually crystallographic data for both active or inactive R2SnX2L2 derivatives revealed an average Cl-Sn-Cl bond angle less then 103° which may implicate an activation mechanism similar to that of cis-diammineplatinum(ll).
It has been revealed that R2SnX2L2 containing N-donor ligands and showing an average Sn-N bond lengths > 239 pm are active complexes, whereas complexes with stronger Sn-N bond lengths < 239 pm are inactive. This implies that the predissociation of N donors groups is more critical then the loss of halogen ions. Recent work demonstrated that the occurrence of relatively stable ligand— Sn bonds, and a low hydrolytic decomposition of Sn-N bond, allow the ligand to interact with the active site of the target before the R2Sn2+ toxin is released. With regard to the data published on new antitumoral agents, in the last years great attention is focused to the study of apoptosis as process that may represent a therapeutic strategy if opportunely activated.
As it is well known, apoptosis is a physiological process of cell death, which is also called "programmed cell death" or PCD because responds to a genetically determined program that is carried out actively by the cells and that requires energy, gene transcription and protein synthesis. Apoptosis, with proliferation, differentiation and cell migration, is a molecular program which is involved in the regulation of the size of tissues and in the maintenance of cell number and cellular homeostasis.
The apoptotic mechanism is also activated as a defense mechanism to protect the integrity of organisms when the cells are subjected to different damages. Thus prevents the propagation of cell population with abnormal genetic and irreversible damages. Apoptosis has biochemical and morphological aspects different to necrosis, another type of cell death. Necrosis does not occur through a specific genetic program, unlike apoptosis which is also called "clean death"; it is not a private event that affects only the cell where it turns on, but necrosis extends to many cells in a diffuse inflammatory event that culminates with the swelling of cellular organelles and cell lysis. During apoptosis, a sequence of events which are common to different cell types occurs and remain constant over the course of phylogenesis. This process begins with a rapid morphological cell transformation, consisting in the reduction of cell volume, cytoskeleton modification, loss of contact between the cells and their separation from original tissue. In the nucleus, the nuclear lamina is degraded and the chromatin is condensed and fragmented and then move to the perinuclear region. Thus the formation of vesicles containing chromatin occurs, which move towards the plasma membrane and are surrounded by the evagination of a plasma membrane. The apoptotic cell takes a look as bubbles, commonly defined blebbing. Exposure of phosphatidylserine on the outer leaflet of the cell membrane close to the cells indicates that it is ready to engulf. In the next step, the cells form spheroidal subunits surrounded by membranes known as "apoptotic bodies" which, released into the environment, disappear through the process of phagocytosis; thus there is not release of cytosolyc content in the environment and therefore does not rise to any secondary flogistic process.
Many drugs which are currently used in anticancer therapies act by activating cytotoxic death by apoptosis. The goal of anticancer research is the design of drugs capable of inducing preferentially apoptosis in tumoral cytotypes without affect non-tumoral cells, and to identify the mechanism of activation of apoptosis and the factors associated with it.
The study of molecular mechanisms of apoptosis has identified two different pathways through which the programmed cell death is achieved: the extrinsic pathway and the intrinsic pathway. Both pathways require the intervention of several proteins, some with pro-apoptotic function that will be activated during the process and others with anti-apoptotic function that will be inhibited. Many of these proteins can be grouped into two broad families: caspase proteins and Bcl-2 family.
The caspases are a family of cytosolyc cysteine proteases which are synthesized in proenzimatic form and then are activated by proteolytic cleavage at a specific aspartate residue within a cascade through which functional enzymes are generated. The caspases can be separated into two functionally different groups: the early caspases, as the caspases 2, 8, 9 and 10, and the effector caspases, as the caspases 3, 6 and 7. The caspase activation may occur not only by proteolytic cleavage but also through association with regulatory proteins, such as the pro-caspase 9. The effector caspases act during the final phase of apoptosis on the level of nuclear lamina, ICAD (inhibitor of caspase activated DNase) and PARP (poly-ADP-ribose polymerase).
The activation of extrinsic pathway is consequent to the linkage of several proteins such as FasL or TNF to death receptors of TNF family (for example Apo/Fas/CD95, TNF-α, DR4 and DR5) which are localized on cell membrane and mediate various physiological responses such as inflammation, proliferation, antiviral activity and cellular death. The receptor-ligand binding induces the receptor activation, that form trimers and contact, specific cytoplasmatic proteins (for example FADD) through the intracellular portion and this culminates with the activation of caspases cascade.
The intrinsic pathway involves the mitochondria and is activated by intracellular events (for example DNA damage, oxidative stress) and also by extracellular events (cytotoxic compound administration, ionizing radiation). These signals converge on mitochondria and cause the dissipation of mitochondrial membrane potential, release in cytosol of cytochrome-c which activates caspase-9 into apoptosome and consequently the executive caspases. It is known that the dissipation of mitochondrial membrane potential may occur as a result of cytoplasmatic ROS (reactive oxygen species) accumulation, after strong oxidative stress, often associated with the mechanism of action of several compounds. During apoptotic activation of mitochondria, several factors of Bcl-2 family are commonly involved. These factors are divided into pro-apoptotic (Bax, Bak, Bok, Bad, Bim, BcIXs, Bid, Noxa, Puma) and anti-apoptotic (Bcl-2, BclXL, Mcl-1 ) members. The anti-apoptotic factors moves from the cytosol, where they are inactive, to the mitochondria and then they are activated through different mechanisms. In the outer mitochondrial membrane, the pro-apoptotic members facilitate the release of cytochrome-c through the formation of channels and compete with the anti-apoptotic factors, which are opposed to these events.
This invention relates to anti-tumor organometallic complexes, and specifically to organotin(IV) compounds with ligands of biological origin, said compounds inducing apoptosis in tumoral cells, promising as selective drug for cancer therapy. The above compounds are particular interesting for use in the treatment of hepatocellular carcinoma, colorectal cancer, leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney. Hepatocellular carcinoma (HCC) is a tumour that develops slowly both in cirrhotic and healthy liver and it is one of the most frequent tumours in the world with unfavourable prognosis: it is estimated that the mortality rate associated with HCC is more then 90%. At this stage, liver resection represent the only radical treatment of HCC. Liver transplantation needs a patient selection: small and liver limited disease without secondary tumours or metastasis are the fundamental requirements. Other palliative and controversial treatments are radiotherapy, that seems to be palliative and controversial, and systemic chemotherapy, using the drugs combination of fluorouracil (5-FU), adriamycin and doxorubicin. Hepatic arterial (HA) infusional chemotherapy possesses a number of constraints not found in systemic chemotherapy and it is complementary to i.v. drug administration. The drug used should have activity in a dose-responsive way without significant hepatic toxicity. Selective drug administration in the hepatic artery allows the delivery of active agents at a higher local concentration while it reduces the peripheral toxic effects of the cytotoxic drug.
The concept of arterial injected metabolic radiotherapy is based on the liver's double vascularization (hepatic artery, portal vein) and on the fact that HCCs are hypervascularized tumours which are mainly fed by the hepatic artery while 80% of the liver unaffected by the tumour is vascularized by the portal vein. Hence by injecting these therapeutic agents intra-arterially we can obtain a high "tumour/unaffected liver" ratio which enables us to administer a considerable tumour targeted dose, preserving the healthy part of the liver as long as possible. The recent advances in imaging technology allow a more selective approaches, producing selective ischemia within the tumour while inducing decreased collateral damage to the tumour free liver parenchyma. That interventistic radiological procedures includes chemoembolization and alcoholization, suitable for cirrhotic patients or for patients with unreasectable HCC with large or multiple lesions.
Those regional therapies inhibit tumor growth, and cause the destruction of small tumor nodules but there are not radical cures so there are still palliative approaches. Recently another regional treatment have been proposed: the radioembolization with arterially injected Lipiodol 1-131 (radioactive iodine) instead of cytotoxic drugs as in chemoembolization.
Colorectal cancer is the third most common form of cancer and the second leading cause of cancer-related death in the Western world and most commonly it spreads to the liver.
There are many therapeutic protocols for later stage colorectal cancer treatment and for hepatocarcinoma: one regimen involves the combination of infusional 5- fluorouracil (5-FU) and leucovorin (LV), methotrexate (toxic for retina), epirubicine, paclitaxel (Taxol, Patene), daunorubicin (cardiotoxic), irinotecan or oxaliplatin. The latter is the complex (R,R)-1 ,2-diaminocyclohexane(ethanedioate-0,0) platinum; its cytotoxicity results from inhibition of DNA synthesis by causing crosslinking of
DNA. When oxaliplatin is administered mixed with 5-FU/LV, usually the 5-FU toxicity increases, with following symptoms diarrhoea, myelotoxicity, mucosity.
It is clear that current drugs used in chemotherapy are toxic but not selective. For example, the 5-FU is an antimetabolite that causes leucopenia, platelets decreasing, hepatotoxicity and pulmonary toxicity, cardiotoxicity, and neurotoxicity.
Also oxaliplatin is toxic for SNC, hematopoietic system, gastrointestinal tract and allergic reactions can occur.
Against progress in over the past decade has led to a major improvement in survival rate in patients with colorectal cancer with hepatic metastasis, however it is still necessary to better evaluate the efficacy of intra-arterial infusion, the landmark of second-line treatments and post-operative adjuvant treatments. It is also necessary to design new selective antitumoral agents.
Summary of the invention This invention provides novel organotin(IV) complexes with the organic ligand N- acetylcysteine, a N-acetyl derivative of the natural occurring sulphur-containing amino acid L-cysteine, of formula:
Figure imgf000010_0001
The compounds of the invention surprisingly show a strong anti-tumour activity, resulting from in vitro cytotoxicity studies on several cancer cellular lines. The compound of the invention showed excellent antitumoral activity in vitro towards hepatoblastoma cells (HepG2), leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney cell lines. In case of hepatoblastoma cells the in vitro cytotoxicitywas also corroborated by a cytofluorimetric analysis of the cell-cycle-dependent DNA content distribution to probe several aspects of drug-induced cytotoxicity, like apoptosis and by other biological studies described later.
The organic ligand used in organotin(IV) complexes, provided by this invention, is a low molecular weight cysteine derivative showing antioxidant activity both in vitro and in vivo, acting as free radicals or hydrogen peroxide scavenger by reduction. The N-acetylcysteine (NAC) is a drug commonly used as a mucolytic agent in the management of acute or chronic bronchopulmonary diseases.
Studies concerning this invention showed that new synthesized organotin(IV) complexes with N-acetylcysteine induce apoptosis in HCC HepG2 cells, while they are inactive towards non tumoural cells used as control (Chang liver cells). Hepatocellular carcinoma (HCC) is one of the most frequent tumours in the world, being the 90% of all malignant liver cancer and furthermore it has been proved that the incidence of HCC is increasing remarkably in many countries in the last decades. It is estimated that the incidence of HCC reaches 3-6-% of solid tumors in USA and Europe, and up to 20-40 % in Africa and South-East Asia. In Caucasian populations in industrialized countries and in Japan the hepatocarcinoma HCC is mainly associated with C or B virus cirrhosis (70-80% of patients); usually post hepatic cirrhosis, less frequently alcoholic cirrhosis or secondary to hemocromatosis. Human hepatoma HepG2 cell line is characterized (from a biomolecular point of view) by an alteration of β catenin gene, caused by a point mutation or a deletion of exons 3 and 4 (aa 25-140). This deletion removes the potential site of regulation by glycogen synthase kinase 3 β (GSK-3β) responsible for its phosphorylation. The lack of phosphorylation prevents the ubiquitination and its degradation by the proteasome. Accordingly, β-catenin accumulates in the cytoplasm and moves into the nucleus where, after the association with the transcription factor TCF/LEF, activates genes involved in cell cycle progression and protection from apoptosis. β catenin alteration can contribute to the deregulated proliferation of hepatoma cells, but also to the acquisition of resistance to apoptosis. HepG2 cells represent, therefore, a good hepatoma model to study the biochemical mechanisms which are involved in the apoptotic mechanism. The hepatic Chang liver cells, employed as non tumoral cell model, are immortalized cells in active proliferation, as beyond the genetic program of senescence. This feature allows the maintenance in culture. According to this invention, a specific class of organotin(IV) derivatives of N- acethyl-L-cysteine, that could be synthesized with high yields and low costs by simple reaction of commercial reagents, wherein there are two or three organic groups linked to the tin, show excellent anti-tumor activity inducing apoptosis; at the same time compounds exhibit higher selectivity and lower toxicity then those of the corresponding organotin(IV) parents. In detail, this invention provides diorganotin(IV) and triorganotin(IV) complexes with N-acethylcysteine which exhibit high toxicity against human hepatocarcinoma cell lines, but they are less toxic, being some of them not toxic, towards normal human hepatocyte cell line. They are all also less toxics then the corresponding organotin(IV) parents used as reagents for their synthesis. The toxicity is time and concentration dependent: in detail it seems that biological ligand, constituting the complexes, modulates the toxic effects of the organometallic moiety. The cell toxicity occurs by apoptosis induction, as it has been showed and confirmed by biochemical study on caspases expression. As it will be described later in detail, the activity of the compounds concerning this invention has been demonstrated in vitro by MTT cytotoxicity test on HepG2 hepatoblastoma cell line, and it has been confirmed subsequently by a cytofluori metric analysis of the cell-cycle-dependent DNA content distribution to quantify apoptosis, by Western blotting for probing caspases involvement in apoptosis induced by organotin(IV) compounds, by studying the level expression of PAR4 and pAkt factors, as well as by ROS production evaluation. The use of organotin(IV) derivatives disclosed in this invention could represent a targeted specific anticancer treatment with high selectivity, and affecting poorly the physiology of normal cells.
The compound of the invention showed excellent antitumoral activity in vitro also towards leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney cell lines.
It should be pointed out that although some organotin(IV) complexes with N- acethylcysteine disclosed in this invention were previously described from a chemical point of view (i.e. G. Domazetis et al., in Inorg.Chim.Acta, (1979), Vol.32,L48-L50; G. Domazetis et al., Tri-n-butyltin(IV) derivatives of L-cysteine ethyl ester, N-acetyl-L-cysteine and α-glutamylcysteinyl glicine (Glutathione reduced), J. Organometal.Chem., 173 (1979) 357-376), there have not been ever reported so far biological or pharmacological activities or properties of these compound, and least of all their use as selective antitumoral agents for hepatic and colorectal diseases. Detailed description of the invention
Thus the invention relates to organotin(IV) complexes with N-acetylcysteine of the general formula (1 ) or (2) :
Figure imgf000012_0001
Figure imgf000013_0001
wherein:
R15R2 and R3 are each independently selected from the group consisting of: H, an alkyl, alkoxyl, alkenyl or alkynyl group with up to 12 carbon atoms, an isocyclic or heterocyclic, aromatic or aralkyl group with up to 12 carbon atoms, for use as medicinal active ingredients.
In details, as remarked, these active agents are antitumoral agents, and more in detail they are suitable for hepatic and colorectal cancer. According a preferred embodiment of the invention, R-ι,R2 and R3 (when present) are each independently selected from the group consisting of: H, an alkyl group with up to 8 carbon atoms, an alkoxyl group with up to 8 carbon atoms, a phenyl group.
Either among diorganotin(IV) complexes with N-acetylcysteine either among triorganotin(IV) complexes with N-acetylcysteine disclosed for medical use according to this invention and having respectively the general formula (1 ) and (2), preferred compounds are those wherein R1, R2 and R3 (when present) are each independently selected in the group consist in methyl, ethyl, propyl, butyl and phenyl groups. Among diorganotin(IV) derivatives of general formula (1 )
Figure imgf000014_0001
wherein R1 and R2 are each independently selected from the group consisting of: H, an alkyl, alkoxyl, alkenyl or alkynyl group with up to 12 carbon atoms, an isocyclic or heterocyclic, aromatic or aralkyl group with up to 12 carbon atoms, that are not described in literature for their putative use as antitumoral agents selective for hepatic and colorectal cancer, particularly preferred is dibutyltin(IV) acetylcisteinate complex, of formula (1 a):
Figure imgf000014_0002
Compound of formula 1 a showed the highest antitumoral activity in vitro among all compounds tested within the contest of this invention.
According to the synthetic procedure provided for producing compounds of the present invention, organotin(IV) complexes with N-acetylcysteine of general formula (1 ) and (2) can be synthesized by reaction of N-acetylcysteine and the corresponding organotin(IV) precursors. Further object of the present invention is a process for producing organotin complexes with N-acetylcysteine with general formula (1 ) or (2), as above described, said process including reacting an organotin(IV) derivative wherein R1, R2 and R3 (when present) have the meaning above already said, with N- acetylcysteine in methanol, chloroform or water, in a temperature range comprised between 10 °C and 90 °C, preferably between 25 °C and 75 °C. Preferably, at the end of said the reaction, the reaction mixture is quenched by cooling, and the resulting precipitate containing the desired compound of formula (1 ) or (2) is collected by filtration.
According to this inventions when the aim of the process is the production of organotin(IV)complexes with N-acetylcysteine with general formula (1 ) or (2), that includes preferably the reaction between organotin(IV)derivative with N- acetylcysteine in refluxing methanol. Preferably, at the end of the reaction described, the reaction mixture is quenched by cooling, and the resulting precipitate containing the desired compound of formula (1 ) or (2) is collected by filtration.
Further aspect of the invention is the use of one ore more compounds having general formula (1 ) or (2), as above described, for the production of a pharmaceutical preparation, in detail an anticancer preparation, and preferably as active ingredient in an anticancer preparation for the treatment of hepatic and colorectal tumours as well as for leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney. According to the invention, the compounds above described can be advantageously used in cancer therapy, administered in a pharmaceutical preparation as toxic agents by themselves or in combination with other chemotherapeutic agent for enhancing the therapeutic effects, and for reducing pharmacoresistences. This invention relates also to a pharmaceutical preparation, including as active component at least one of the organometallic complexes with N-acetylcysteine with general formula (1 ) or (2) together with one ore more pharmaceutically acceptable adjuvants and/or vehicles. In an even more preferred embodiment, the preparation contains as active component dibutyltin(IV) -N-acetylcisteinate complex, of formula (1 a). According to this invention, the pharmaceutical preparations containing organotin(IV) derivatives with N-acetylcysteine suitables for the therapeutical administration can be designed using conventional methods already known by expertises, by using adjuvants and vehicles pharmaceutically acceptable, for obtaining preparations suitable for parenteral injection (intravenous, intramuscular or subcutaneous), for oral administration of solid or liquid formulations, for transdermal, rectal, intravaginal or topical administration, for instance in oronasal mucosa and similar.
The percentage of the active compound in the preparation and the method of tumour treatment could be changed for reaching the optimal dosage. The dosage to administer depends on: route of administration, length of treatment, patient conditions and weight, drug effectiveness and patient reaction. As soon as the right dosage of active compound has been determined, the formulation and the choice of adjuvants and vehicles pharmacologically suitable for each need will be made according to the current knowledge in this field. The specific characteristics of the invention, as well as the advantages either for the methods for synthesizing the suggested compounds, either for their biological and pharmacological activity, will be remarked afterwards or in the detailed illustrative description and experimental results related to this invention. Brief description of the figures
Some experimental results are illustrated in the enclosed pictures, where: the figures 1A and 1 B show the effects induced by the compound Bu2SnNAC, according to the invention, in comparison with Bu2SnCI2 and NAC, on hepatic HepG2 and Chang liver cell viability, respectively after 8 h (A) and 16 h (B) of treatment; the figure 2 shows the influence of NAC on the cytotoxic effect induced by Bu2SnCI2 in HepG2 cells according to the invention; the figure 3 shows the involvement of caspases in apoptosis induced by Bu2SnNAC complex, according to the invention, compared with Bu2SnCI2, in HepG2 cells; and the figure 4 shows the effects of the Bu2SnNAC complex, according to the invention, and the others used for a comparison, on the levels of PAR4 factor and the survival protein pAKT. the figures 5A and 5B show the effect of the compound NAC2 against 60 cancer cell lines at a single dose of 10μM. Experimental section
As already remarked, in what is concerning the synthesis, all the organotin(IV) derivatives N-acetylcysteine described later have been obtained starting from reactants and solvents reagent grade and commercially available. In the experimental chemical session after described, elemental analysis have been carried out at the Department of Inorganic and Analytical Chemistry "S.Cannizzaro" of the University of Palermo, by a Vario EL III (Elementar GmbH) analyzer instrument using the "purge and trap" method, performing double analysis and evaluating the results as average of the two analysis. The FT-IR spectra of the free ligand and of its organotin(IV) complexes were registered, both Nujol mulls and hexachlorobutadiene, on a Mod. Spectrum ONE Perkin-Elmer FT-IR with the appropriated equipped software , between CsI windows, performing 8 scan with a 4 cm"1 resolution.
The 119Sn Mόssbauer spectra were recorded by maintaining the source at room temperature and by cooling the sample, as fine powder pressed between two aluminium foil, at the temperature of 77.3±0.1 K, using a liquid nitrogen cryostat NRD-1258-MB (Cryo Industries of America inc., Atkinson, NH, USA) and using an ITC 502 Oxford Instrument temperature controller to obtain the temperature control. The multichannel calibration was performed with an enriched iron foil (57Fe = 95.2%, thickness 0.06 mm, Ritverc GmbH, St. Petersburg, Russian Federation) at room temperature by using a 57Co (10 mCi) source (New England Nuclear, Amersham) while the zero point of the Doppler velocity scale was determined at RT through the absorption spectrum of natural CaSnO3 ([119Sn] = 0.50 mg cm-2 ) as source.
The 119Sn Mόssbauer spectra were recorded in duplicate with a model 209 TAKES (Ponteranica, Bergamo, Italy) multichannel analyzer (4096 channels) and the following MWE (Munchen Wissenschaftliche Elektronik, Gmbh, Starnberg, Germany) apparatus: MR250 driving unit, FG2 digital function generator (moving at linear velocity and constant acceleration in a triangular waveform. )and MA250 velocity transducer, In each point of spectrum velocity , a sufficient number of counts were collected to obtain a well defined spectrum. Data so collected have been elaborated by a personal computer, using appropriate calculations software.
For the solution phase, the complexes have been investigated at 296 K by 1H,
13C(1HJ and 119Sn(1HJ NMR using a Bruker AVANCE DRX300 NMR spectrometer, with "probe broad-band" and "inverse detection" respectively, at 300.13, 75.5 and
118.9 MHz. The signals of the solvents were used as internal standard for 1H and
13C(1HJ spectra, and as external standard the 13C signal of TMS (13C d= 0.00). For the 119Sn(1HJ spectra a neat liquid solution of tetramethylstannane (Me4Sn) was used as external reference (119 Sn d=0.00). Molecular geometries were optimized at the density functional level of theory using the software packages Gaussian '03 (G03) 112 and Turbomole. DFT calculations were performed by the hybrid B3LYP method and using the double-zeta valence plus polarization (DZVP) all electron basis set 6-31 G** (H, C, O, S). The calculations were performed using density functional theory (DFT). The calculation of chemical shift and coupling constants values were fully optimised at DFT level using the mPW1 PW91 functional with the same basis set.
EXAMPLE 1
MepSnNAC complex synthesis
N-acetylcysteine (NAC) (1 mmol,163.18 mg) was dissolved in 30 ml of methanol or water, then added slowly to a dimethyltin(IV) oxide (Me2SnO) solution (1 mmol, 164.78 mg), under stirring. After 24 hours at range 25-75 °C, the white solid derivative was recovered after filtration under vacuum and vacuum drying on
P4O10.
White solid obtained was study by FT-IR analysis. The IR spectrum showed that the reaction proceeded with total conversion of the starting material, as well as all compounds presented as further examples in Table 2.
Analytical data of the obtained compound are reported later in Table 1.
EXAMPLE 2
B UgSn N AC complex synthesis N-acetylcysteine (NAC) (163.18 mg, 1 mmol) was dissolved in 30 ml of methanol or water, then added slowly to a dibutyltin(IV) oxide (Bu2SnO) solution (1 mmol
,248.93 mg), under stirring. After 24 hours at range 25-75 °C, the white solid derivative was recovered after filtration under vacuum, ricristalyzed from chloroform and vacuum dried on P4O10).
White solid obtained was study by FT-IR analysis. The IR spectrum showed that the reaction proceeded with total conversion of the starting material, as well as all compounds presented as further examples in Table 2. Analytical data of the obtained compound are reported later in Table 1.
EXAMPLE 3
PhgSnNAC complex synthesis
The Ph2SnNAC complex was obtained as white solid, mixing a diphenyltin(IV) oxide (Ph2SnO) aqueous suspension and a N-acetylcysteine aqueous solution, in molar ratio1 :1 .
This reaction allowed the complex formation, without unreacted reagents or secondary products.
N-acetylcysteine NAC (163.18 mg, 1 mmol) was dissolved in 30 ml of distillate water, then added slowly to a diphenyltin(IV) oxide (Ph2SnO) aqueous suspension
(1 mmol ,288.92 mg), under stirring. After 24 hours at range 25-75 °C, the white solid derivative was recovered after filtration under vacuum , and vacuum dried on
P4O10 P2O5).
White solid obtained was study by FT-IR analysis. The IR spectrum showed that the reaction proceeded with total conversion of the starting material, as well as all compounds presented as further examples in Table 2. Analytical data of the obtained compound are reported later in Table 1.
EXAMPLE 4
(Me3Sn)^NAC complex synthesis The (Me3Sn)2NAC complex was obtained as white solid, mixing a trimethyltin(IV) hydroxide (Me3SnOH) and a N-acetylcysteine methanolic solutions, in molar ratio
2:1 .
This reaction allowed the complex formation, without unreacted reagents or secondary products. N-acetylcysteine NAC (163.18 mg, 1 mmol) was dissolved in 30 ml of methanol and then added slowly to a trimethyltin(IV) hydroxide (Me3SnOH) methanolic solution (2 mmol, 180.82 mg), under stirring. After 24 hours at range 25-75 °C, the solution volume was reduced to 10ml by a rotating evaporator.
The reaction mixture was quenched at fridge temperature, and after 24 hours the resulting white desired precipitate was collected by filtration, washed with chloroform and then vacuum dried on P4Oi0 . White solid obtained was study by FT-IR analysis. The IR spectrum showed that the reaction proceeded with total conversion of the starting material, as well as all compounds presented as further examples in Table 3. Analytical data of the obtained compound are reported later in Table 1.
EXAMPLE 5 (BUc1Sn)9NAC complex synthesis
The (Bu3Sn)2N-acetylcysteinate complex was obtained as gel mixing a tributhyltin(IV) hydroxide ((Bu3Sn)2O) and a N-acetylcysteine methanolic solutions, in molar ratio 2:1.
This reaction allowed the complex formation, without unreacted reagents or secondary products.
N-acetylcysteine NAC (163.18 mg, 1 mmol) was dissolved in 30 ml of methanol, then added slowly to a tributhyltin(IV) hydroxide ((Bu3Sn)2O methanolic solution (1 mmol; d=1.171 g/ml; 0.51 ml), under stirring. After 24 hours at range 25-75 °C, the solution volume was reduced to 10ml by a rotating evaporator. The reaction mixture was quenched at fridge temperature, but after 24 hours no solid was formed. The solution was dried by a rotating evaporator. The resulting gel desired was collected and dried on P4O10
FT-IR analysis of the obtained material correspond to that of the desired compound, as well as for all compounds presented as further examples in Table 3. Analytical data of the obtained compound are reported later in Table 1.
EXAMPLE 6
(Ph3Sn)9NAC complex synthesis
The (Ph3Sn)2 N-acetylcisteinate complex was obtained as white solid, mixing a triphenyltin(IV) hydroxide (Ph3SnOH) and a N-acetylcysteine chloroformic or methanolic solutions, in molar ratio2:1.
This reaction allowed the complex formation, without unreacted reagents or secondary products. N-acetylcysteine (NAC) (163.18 mg, 1 mmol) was dissolved in 30 ml of chloroform (chloroform or methanol) , then added slowly to a Ph3SnOH chloroformic solution (2 mmol; 367.01 mg), under stirring. After 24 hours at range 25-75 °C, the solution volume was reduced to 10ml by a rotating evaporator.
The reaction mixture was quenched at fridge temperature, and after 24 hours the resulting white desired precipitate was collected by filtration, washed with chloroform and then vacuum dried on P4O10 .
FT-IR analysis of the obtained material correspond to that of the desired compound, as well as for all compounds presented as further examples in Table 3.
Analytical data of the obtained compound are reported later in Table 1.
Table 1
Elemental analysis analytical data [experimental (calculated), (%)] for R2Sn(IV)NAC (R = Me, n-Bu, Ph) and (R3Sn(IV))2NAC derivatives (R = Me, n-Bu, Ph); NAC2"= [N-acethlcysteinate]2"
Figure imgf000021_0001
Table 2
Assignment of more relevant absorption bands of N-acetyl-L-cysteine (NAC), of its R2Sn(IV)NAC (diorganotin) derivatives (AIk = Me, n-Bu, Ph), in the range 4000- 250 cm"1
Figure imgf000022_0001
* Signals are hidden by those of Ammide II
Table 3
Assignment of more relevant absorption bands of N-acetyl-L-cysteine (NAC), of its (R3Sn(IV))2NAC(triorganotin(IV) ) derivatives (R = Me, n-Bu, Ph), in the range 4000-250 cm"1.
Figure imgf000023_0001
* Signals are hidden by those of Ammide II
119Sn Mόssbauer characterization
By the Mόssbauer spectroscopy, and in detail by the isomer shift parameter, δ, we obtained useful data on the valence state of tin in complexes and on the structure and the bonding in the tin environment. The isomer shift values δ, mms"1, reflect the meaning of the parameter, increasing their value according to the inductive effects of the organic groups, so they decrease on going from dialkyl to diphenyl derivatives.
In order to verify the correctness of the interpretation, the experimental nuclear quadrupole splitting parameters have been rationalized according to the point charge model formalism. Theorical data of nuclear quadrupole splitting |ΔΘxp| were calculated by the point-charge model formalism and compared with experimental data.
For the diorganotin (IV) derivatives of formula (1 ) the C-Sn-C angles (θ, deg) were calculated via Eq. (1 ),
Figure imgf000024_0001
where {R} is the p.q.s. of the alkylic and phenylic group in an idealised octahedral configuration (-1.03 mms"1 and -0.95 mms"1, respectively) and Δ is the measured quadrupole splitting:
For all compounds, even if the other groups bonded to the tin have not been taken in account, the C-Sn-C angles values were calculated with an error of ± 13°, however the values were useful for evaluating the distorted configuration around the tin atom. The values of the experimental isomer shift δ and of the calculated and experimental nuclear quadrupole splitting [|ΔΘxp| (mm s"1) and |Δcaic| (mm s"1) ], are summarized in Table 4. In details, the table contains Mossbauer parameters of the organometallic compounds in object, as isomer shift, δ,in mm s \ together with the and experimental nuclear quadrupole splitting [|ΔΘxp| (mm s"1)], and the C-Sn-C angles values measured bat the temperature of the liquid nitrogen, and the calculated nuclear quadrupole splitting [|Δca|C| (mm s"1) according to the point charge model formalism for idealised tetraedral and trigonal bipyramidal configuration. Furthermore, in table the suggested geometry for synthesized compounds has also been reported: Mossbauer data strongly suggested a trigonal bipyramidal configuration for R2SnNAC ( R= Me, Bu, Ph) derivatives, where alkyl and phenyl groups lie in equatorial positions.
For derivatives triorganotin(IV) NAC with general formula (2), Mossbauer data indicated more than one environment for the tin atom in these. The obtained |ΔΘxp| values were in the range of a trigonal bipyramidal and tetrahedral configuration around the tin atom, within the experimental error of ±0.4 mm/s. Table 4
Experimental Mossbauer parameters3, isomer shift δ and nuclear quadrupole splitting |ΔΘxp| (mm s"1) measured at liquid N2 temperature, calculated nuclear quadrupole splittings Δcaicd, according to the point charge formalism, and C-Sn-C angles(of synthesized compounds)
Figure imgf000025_0001
a Sample thickness ranged between 0.50 and 0.60 mg Sn cm" ; isomer shift, δ ± 0.03, mms" with respect to room temperature BaSnOs; Fl and F2 values are the full width at half height of the resonant peaks, respectively, at greater and lower velocity respect to the centroid of the Mossbauer spectra; nuclear quadrupole splittings, |zfexp| ± 0.02 mms 1. b NAC2' = N-acethylcysteinate2'.
Computational studies for R2Sn complexes with N-acetylcysteine(R = Me, Bu) with general formula (1)
The computational studies are consistent with the experimental data. The geometries of the Me2SnNAC and Bu2SnNAC complexes, previously synthetized, were optimized and tvibrational frequencies for the two species were also computed. For Bu2SnNAC complex,two structural isomers ((A) and (B)) have been found, whose energy values showed that the most stable isomer is the one presenting the sulfur atom in the equatorial position (A), in agreement with the Mossbauer data. The bond angles and lengths have been compared with literature data of the captopril, a ligand that coordinates dimethyltin(IV) unit similarly to the NAC
The vibrational frequencies have been compared with IR data. The good agreement between the frequencies and the calculated and experimental bond angles and length confirmed the structural hypothesis previously suggested. Study of anticancer activity of organotin (IV) with N-acetylcysteine complexes
The activity of organotin(IV) with N-Acetylcysteine complexes according to the invention was evaluated at the Department of Biochemical Sciences of University of Palermo, Policlinico "Paolo Giaccone", by using MTT assay to evaluate their cytotoxicity, cytofluorimetric analysis to identify cell cycle distribution for apoptosis quantification and to assess ROS production, Western blotting analysis to evaluate the involvement of caspases, pAKT and Par4 in apoptotic mechanism induced by the compounds. Cell cultures
Chang liver and HepG2 hepatoma cells were cultured in polystyrene 75 cm2 flasks and grown in RPMI 1640 medium supplemented with 10% (v/v) heat- inactivated foetal calf serum, 2.0 mM L-glutamine, 1.0 mM sodium pyruvate and antibiotic antimycotic solution (100 U/ml penicillin, 100 Dg/ml streptomycin and 250 ng/ml amphotericin B) and incubated at 37 °C in a humidified, 5% CO2 atmosphere, 95% air.
Cell viability was determined after detachment of cells from flask by Trypsin - EDTA solution and seeding of 6.5x103 Chang liver cells and 104 HepG2 cells (on 200 μl of medium) in 96-well cell culture plates (0.3 cm diameter).
Cytofluorometric studies were carried out seeding 1.5x105 Chang liver cells and 2.0x105 HepG2 cells in 6-well cell culture plates (3 cm diameter), while Western blotting analysis was performed seeding 1 O6 CeIIs in Petri dishes (10 cm diameter). Stock solutions of compounds were prepared in dimethyl sulfoxide (DMSO) obtaining a 1 mM solution which was opportunely diluted in culture medium to the desired final maximum test concentration. The final concentration of DMSO never exceeded 0.04%, a concentration beyond which there may be phenomena of toxicity. Control cells were cultured in the presence of vehicle alone. MTT Cell viability assay MTT assay is a quantitative colorimetric assay that employs the 3-[4,5- dimethylthiazolyl-2] 2,5-diphenyl-tetrazolium bromide (MTT) (T Mosmann, J.Immunol. Methods, 1983, 65, 55-63). Tetrazolium salt MTT is cleaved to blue formazan by mitochondrial dehydrogenases of vital cells , which belongs to the mitochondrial respiratory chain. Since this enzymatic system is active only in viable (metabolically active) cells, this assay exclusively detects viable cells. The intensity of staining formed is directly proportional to the number of living cells and is measured by means of spectrophotometer. After the treatments, MTT stock solution was added to each well (final concentration 1 mg/ml) for 2 hours at 37 °C. The medium was then removed and 0.2 ml of lysis buffer (obtained by dissolving 20% (p/v) SDS (sodium dodecilsulphate) in 50% N,N-dimethylformamide, pH 4.7) was added to dissolve the formazan product. Finally, after 16 hours of incubation at 37 °C, the absorbance at 570 nm (test wavelength) and at 630 nm (reference wavelength) was measured using an ELISA microplate reader (OPSYS MR, Dynex Technologies). Lysis; buffer was employed as the reference test to reset the instrument.
Data were the mean ± S. E. of three independent experiments involving triplicate assays. The percentage of viable cells is calculated as percentage of viability compared to untreated control cells by assumpting the value of 100% for untreated cells.
In initial experiments, we evaluated the sensitivity of HepG2 and Chang liver cells to all the complex and organotin derivatives. The results showed that organotin parents were highly toxic to both the two cell lines, while the organotin (IV) complexes, in particular the dibutyltin(IV) N-Acetylcysteinate resulted the most active. Then, to evaluate the sensitivity of hepatic HepG2 and Chang liver cells to dibutyltin(IV)dich\or\de (Bu2SnCI2) and d/&ufy/f/n(IV)-N-acetylcysteinate (Bu2Sn- N-Acetylcysteinate), we carried out time- and dose-dependent experiments. The results were reported in Figure 1A and 1 B. MTT colorimetric assay was employed as previously reported.
Cells were incubated in 96-well cell culture plates in presence of the NAC ligand (control) or of the Bu2SnCI2 and Bu2SnNAC at the mentioned concentrations. After the treatment cell viability was evaluated by MTT colorimetric assay. Data were reported as the percentage of viable cells, calculated as percentage of viability compared to untreated control cells, mean D S. E. of three independent experiments involving triplicate assays. The results indicated that the compounds exert different effects in Chang liver and HepG2 cells
In particular, 10 μM Bu2SnCI2 induced after 8 hours of treatment a reduction of HepG2cell viability equal to 53%, against a value of 28% observed in the presence of 10 μM Bu2Sn-N-Acetylcysteinate. The cytotoxicity increased with the time reaching the maximum after 48 hours of treatment (-90%) with the two compounds employed at the same dose (10 μ M).
Interestingly, the studies showed that both the compounds, Bu2SnCI2 and Bu2Sn N-Acetylcysteinate, had toxic effects on hepatoma HepG2cell line. However, after 8 hours of incubation 10 μM Bu2SnCI2 was much more toxic than Bu2SnN Acetylcysteinate employed at the same dose. This can be explained with the ability of N-Acetylcysteine, conjugated in the complex, to modulate, during the initial phase of treatment (8 hours), the high toxic activity of Bu2SnCI2.
In order to exclude a protective effect induced by the presence of the free ligand N-Acetylcysteine in the solution, as hydrolysis product of the complex, and to support the hypothesis of the modulating effect of N-Acetylcysteine, cytotoxicity studies on HepG2 cells pre-treated with 10 μM N-Acetylcysteine for 1 hour were performed and then cells were incubated in the presence of 10 μM Bu2SnCI2 for 8 hours. The results are shown in Figure 2. Cell viability was tested using a procedure based on the MTT assay. Values are expressed as percentage of viable cells compared to control.
As indicated in the graph, the toxicity induced by 10 μM Bu2SnCI2 added to the cells after the pre-incubation with NAC is significantly greater than that observed in the presence of Bu2Sn N-Acetylcysteinate complex. Thus, it is possible to conclude that the binding with N-Acetylcysteine significantly modulates the cytotoxic effect of the organometallic portion, probably without that any degradation occurs within the cell.
In immortalized Chang liver cells, after 8 hours of treatment with the compounds, were not observed changes in cell viability and even after longer times of incubation (16 and 24 hours) the death came on the very low values (19%) after treatment with 10 μM Bu2SnCI2 and 17% after treatment with 10 μM Bu2Sn N-Acetylcysteinate. Characterization of apoptosis
Cytofluorometric study of cell distribution along the phases of proliferative cycle: quantification of apoptosis.
Quantification of apoptosis was carried out by staining the cells with propidium iodide (Pl) , a fluorescent dye that intercalates to DNA, according to the protocol of Carollo et al. (M. Carollo, L. Parente, N. D'Alessandro, Oncol. Res., 1998, 10, 245-54). This method allows to evaluate the cell distribution along the phases of proliferative cycle. The presence of a peak of lower fluorescence intensity indicates the presence of cells with fragmented DNA. After treatment with the compounds, cells were resuspended in hypotonic solution of fluorochrome consists in 50 μg/ml propidium iodide, 0.1 % sodium citrate, 0.1 % Nonidet P-40 and 100 μg/ml RNAse, and incubated overnight in the dark at a temperature of 4 °C. Cell distribution along the phases of proliferative cycle was evaluated using Beckman Coulter Epics XL cytofluorimeter and Expo32 software for data processing. Fluorescence emitted from the cells was analyzed as frequency of histograms of individual parameters and the percentage of cellular population located in sub-diploid region (subG0/G1 ) was considered as an indicator of programmed cell death. Data obtained were comparable to those observed with MTT assay. As shown in Table 5 (see below) the reduction of HepG2 cell viability was accompanied with a peak of 59.5% (control 1.4%) of cells in subG0/G1 phase of the cell cycle, indicating DNA fragmentation after 8 hours of treatment with 10 μM Bu2SnCI2.
After incubation with 10 μM Bu2Sn N-Acetylcysteinate, instead, the peak is equal to 2.7%. Prolonging the incubation time up to 16 hours, it was observed that both the compounds, Bu2SnCI2 and Bu2Sn N-Acetylcysteinate, have a similar effect on HepG2 cells, confirming the results observed with cell viability assay. In fact, treatment with 10 μM or 20 μM of both compounds for 16 h induced a significant fragmentation of DNA with about 80% of cells in subG0/G1 phase of cell cycle. Instead after 8 hours of incubation even doubling the concentrations of the compounds (20 μM) was observed the appearance of subG0/G1 peak equal to 65.4% in the presence of Bu2SnCI2 while after treatment with Bu2Sn N- Acetylcysteinate the amount of peak was equal to 27.5%. The compounds showed very modest effects on Chang liver cell distribution along the proliferative phases of the cycle, both prolonging the time of incubation that using increasing concentrations of the compounds; in fact the percentage of cells in subG0/G1 phase did not exceed ever the 10-16%. Table 5
Cytofluorimetric analysis of HepG2 (A) and (B) and Chang liver (C) cells distribution along the phases of the proliferative cycle.
Figure imgf000030_0001
Assessment of ROS production
The production of reactive oxygen species (ROS) was measured using 2', 7'-dichlorofluorescein diacetate (H2DCFDA), a ROS probe, which is converted to fluorescent compound after the removal of acetate groups by the intracellular esterases and after oxidation dependent on the presence of substantial amounts of ROS. After incubation with compounds, cells were washed gently with warm buffer (37 °C) HBSS/Ca/Mg and incubated at 37 °C for 30 min with 5 μM H2DCFDA dye. Then cells were detached with trypsin solution, resuspended in buffer and analyzed by flow cytometry using the excitation wavelength of 488 nm and the emission wavelength of 525 nm. Data were analysed using Expo32 software. The percentage of cells showing a higher fluorescence, which reflects ROS production, was determined by comparison with untreated controls.
The study of ROS production showed that 10 μM Bu2SnCI2 is able to induce, after 8 hours of treatment, ROS accumulation in HepG2 cells, as demonstrated by the appearance of a peak of fluorescence equal to 84% (vs 8.5% in control cells). Interestingly, in cells incubated with 10 μM Bu2Sn N- Acetylcysteinate, ROS production is smaller with a peak to fluorescence equal to 47.5%.
Studies on the involvement of caspases in apoptotic mechanism induced by the compounds in HepG2 cells: western blotting analysis Cell lysates preparation
After treatment with the compounds, cells (seeded in 10 cm culture plates) were trypsinized and centrifuged for 20 min at 800 rpm. Then, cells were incubated for 30 min on ice in denaturating RIPA buffer, constituted by 1 % NP-40, 0.5% Na-deoxycholate and 0.1 % SDS, containing a protease inhibitor cocktail (25 μg/ml aprotinine, 1 mM PMSF, 10 mM sodium orthovanadate, 10 mM Sodium- fluoride, 25 μg /ml leupeptine and 0.2 mM Sodium-pyrophosphate). Cells were sonicated three times for 10 sec and protein concentration was evaluated by means of Lowry method (O. H. Lowry, N.J. Rosebrough, A.L. Farr, R.J. Randali, J.Biol. Chem. 1951 , 193, 265-275). Protein electrophoresis on polyacrylamide gel and blotting on nitrocellulose membrane
Equal amounts of protein samples (60 μg/lane) were diluted in 1 X Laemmli buffer (constituted by 0.0625 M Tris, pH 6.8, 2% SDS, 5% β-mercaptoethanol, 10% glycerol, 0.02% bromophenol blue) (U.K. Laemmli, 1970, Nature, 227, 680- 685), denatured at 90 °C for 5 minm, and subjected to SDS-PAGE in running buffer 1 x (25 mM Tris, 0.1 % SDS, 1.44% glycine, pH 9.0) for about 2 hours at 150 V.
Then, cells were electrotransferred to a nitrocellulose membrane for the detection with specific antibodies. Bands were quantified by densitometric analysis using SMX Image software.
Western blotting analysis indicated that treatment of HepG2 cells with Bu2SnCI2 decreased the levels of inactive pro-enzymatic isoforms of caspase-8, caspase-9 and caspase-3. These three proteins mediate, at different levels, signals that converge in the final mechanism of apoptosis. Caspase-8 and caspase-9 intervene in the initial caspase cascade activation and therefore are referred as "initiators", instead caspase-3 acts in the final stage of apoptosis causing the degradation of the nuclear lamina, of ICAD and PARP proteins. On the contrary Bu2Sn-N-Acetylcysteinate induced the recovery in the levels of caspase- 8, caspase-9 and caspase-3, as shown in Figure 3. This figure illustrates the involvement of caspases in apoptosis induced by Bu2SnNAC and Bu2SnCI2 in HepG2 cells used in the study. Cells were incubated for 8 and 16 h at the indicated compounds concentrations. After treatment, cell lysates were prepared and western blotting was conduced using specific antibody to caspase-8, caspase-9 and caspase-3. The normalization of protein amounts was performed, for each experiment, through actin immunoblotting. The bands were quantified by densitometric analysis using SMX Image software. Experiments performed for the evaluation of caspase-8 indicated that
Bu2SnCI2 induced the decrease of the inactive form of protein (57 KDa), but the appearance of the active form of enzyme, indicated as a band at lower molecular weight, was not observed. The decrease of pro-caspase-8 (which was considered as expression of activation in caspase-8) is observed after 8 hours of incubation with the compound and it is more visible after 16 hours. Similar effects were induced also in executioner caspase-9 and caspase-3 proteins.
Treatment with Bu2Sn-N-Acetylcysteinate, in same experimental conditions, induced the recovery of pro-caspase form levels, compared with Bu2SnCI2 parent, supporting the ability of the complex to modulate the apoptotic effect of parent compound.
Mechanism of cellular survival
Recently, it has been demonstrated the existence of an elaborate mechanism of reciprocal regulation between pAKT, one of the most important survival factor, and PAR-4 (Prostate-apoptosis-response-gene-4) proteins. PAR-4 protein is a product of pro-apoptotic gene which induces apoptosis in several tumor cytotypes. This function is induced after phosphorylation in Thr163 by PKA with the consequent migration to the nucleus, where it is able to induce the expression of factors that promote apoptosis (for example through FasL-Fas- FADD-caspase axis) and inhibit the expression of factors which are opposed to it (for example Bcl2 and NF-kB proteins). The inhibition of pro-apoptotic action of PAR-4 occurs as a consequence of the phosphorylation on the residue Ser 249 by pAKT protein, which sequesters it in the cytoplasm. PAR-4 also appears to modulate pAKT through PTEN activation, a fosfatydilinositol-3-phosphate phosphatase which has a strong inhibitory effect on PI-3 kinase by reverting the phosphorylation status and determine accordingly the lack of activation of survival factor p A KT Effect of compounds on the levels of pAKT factor.
To evaluate the involvement of pAKT, one of the most important survival factor and PAR4 in apoptotic pathway induced by the compounds, first were performed experiments to assess the effect of the compounds on pAKT levels.
In particular, we wanted to assess whether the different effect of the two compounds observed after 8 hours of treatment was due to different mechanisms which are induced within the cell in an early phase of treatment in attempt to escape from apoptotic mechanism.
The results of the experiments are shown in Figure 4. The cells were incubated with the indicated concentrations of compounds for 8 and 16 hours. Then western blotting analysis on cell lysates using specific antibodies for pAKT, phospho-PAR4 (Thr163) and PAR4 were performed. Normalization of protein was performed by immunoblotting for actin. The bands were quantified by densitometry analysis using SMX Image software. The results shown in the figure indicate that after 8 hours of treatment with Bu2SnCI2 the content of active survival factor pAKT decrease in dose-dependent manner, whereas the levels of pAKT recover after incubation with Bu2SnNAC.
Recent data indicate that the phosphorylation on Thr163 of PAR4 determines its activation as a pro-apoptotic factor and subtracts it to the phosphorylation on Ser 249 by pAKT, an event that would render PAR4 inactive (J.W. Lee, K. F. Lee, H. Y. Hsu, L.P. Hsu, W.L. Shin, YC. Chu, WT. Hsiao, P.F. Liu, Cancer Lett, 2007, 257(2), 252-62). This mechanism probably explains why, when the cells were incubated for 8 hours with Bu2SnNAC, pAKT increased (compared to HepG2 cells incubated with Bu2SnCI2 in the same experimental conditions) and simultaneously phospho-PAR4 (Thr163) levels decreased. Then it is expected that, after treatment with Bu2Sn-N- Acetylcysteinate, anti-apoptotic phospho-PAR4 (Ser249) levels increase following pAKT activity, and this would induce a delay, at least during early hours of treatment, in the activation of caspases, and consequently in apoptosis. Instead in cells incubated with Bu2SnCI2, after 8 hours of incubation, the decrease in pAKT levels and the increase in pro-apoptotic phospho-PAR4 (ThM 63) were responsible for the greater sensitivity observed in these cells to apoptotic effects induced by Bu2SnCI2.
Study on antitumoral activity of NAC2 complex (dibuthyltin N.acetylcysteinate) performed at the National Cancer Institute. NAC2 activity was assayed against 60 different human tumor cell lines, representative for leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney. The aim was to evaluate a selective growth inhibition or cell killing of particular tumor cell lines induced by NAC2. The screening is a two-stage process, beginning with the evaluation of the compound against the 60 cell lines at a single dose of 10 uM. The output from the single dose screen is reported as a mean graph and is available for analysis by the COMPARE program. We report the results of this one dose test. The compound exhibited significant growth inhibition. Methodology of The In Vitro Cancer Screen
The human tumor cell lines of the cancer screening panel were grown in RPMI 1640 medium containing 5% foetal bovine serum and 2 mM L-glutamine. Cells were inoculated into 96 well microtiter plates in 100 μl_ at plating densities ranging from 5,000 to 40,000 cells/well depending on the doubling time of individual cell lines. After cell inoculation, the microtiter plates were incubated at 37° C, 5 % CO2, 95 % air and 100 % relative humidity for 24 h prior to addition of experimental drugs. After 24 h, two plates of each cell line were fixed in situ with TCA, to represent a measurement of the cell population for each cell line at the time of drug addition (Tz). Experimental drug were solubilized in dimethyl sulfoxide at 400-fold the desired final maximum test concentration and stored frozen prior to use. At the time of drug addition, an aliquot of frozen concentrate was thawed and diluted to twice the desired final maximum test concentration with complete medium containing 50 μg/ml gentamicin. Aliquots of 100 μl of this drug dilution was added to the appropriate microtiter wells already containing 100 μl of medium, resulting in the required final drug concentration.
Following drug addition, the plates were incubated for an additional 48 h at 37°C, 5 % CO2, 95 % air, and 100 % relative humidity. For adherent cells, the assay is terminated by the addition of cold TCA. Cells were fixed in situ by the gentle addition of 50 μl of cold 50 % (w/v) TCA (final concentration, 10 % TCA) and incubated for 60 minutes at 4°C. The supernatant was discarded, and the plates were washed five times with tap water and air dried. Sulforhodamine B (SRB) solution (100 μl) at 0.4 % (w/v) in 1 % acetic acid was added to each well, and plates were incubated for 10 minutes at room temperature. After staining, unbound dye was removed by washing five times with 1 % acetic acid and the plates were air dried. Bound stain was subsequently solubilized with 10 mM trizma base, and the absorbance was read on an automated plate reader at a wavelength of 515 nm. For suspension cells, the methodology was the same except that the assay was terminated by fixing settled cells at the bottom of the wells by gently adding 50 μl of 80 % TCA (final concentration, 16 % TCA). Using the three absorbance measurements [time zero, (Tz), control growth, (C), and test growth in the presence of drug at the 10-5 M concentration level (Ti)], the percentage growth was calculated as: [(Ti-Tz)/(C-Tz)] x 100 for concentrations for which Ti>/=Tz [(Ti-Tz)/Tz] x 100 for concentrations for which TkTz.
Interpretation of One-Dose Data: The One-dose data are reported as a mean graph of the percent growth of treated cells . The number reported for the One- dose assay is growth relative to the no-drug control, and relative to the time zero number of cells. This allows detection of both growth inhibition (values between 0 and 100) and lethality (values less than 0). For example, a value of 100 means no growth inhibition. A value of 40 would mean 60% growth inhibition. A value of 0 means no net growth over the course of the experiment. A value of -40 would mean 40% lethality. A value of -100 means all cells are dead. Information from the
One-dose mean graph is available for COMPARE analysis. http://dtp.nci.nih.gov/branches/btb/ivclsp.html
The figure 6 shows the results on 60 different human tumor cell lines. The compound NAC2 showed excellent antitumoral activity in vitro towards leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney.
This invention has been described with reference to some forms of specific realizations, but it is intended that variations and changes can be made to it by specialists alone to escape of relative scope of protection.

Claims

1. Organotin(IV) compounds with N-acetylcysteine having the general formula (1 ) o (2):
Figure imgf000037_0001
wherein:
R-I, R2 and R3 are each independently selected from the group consisting of:
H, an alkyl, alkoxyl, alkenyl or alkynyl group with up to 12 carbon atoms, an isocyclic or heterocyclic aromatic or aralkyl group with up to 12 carbon atoms, for use as medicinal active ingredients.
2. Organotin(IV) compounds with N-acetylcysteine according to claim 1 , wherein the said medicinal active ingredients are anticancer agents.
3. Organotin(IV) compounds with N-acetylcysteine according to claim 2, wherein the said anticancer agents are agents for the treatment of hepatic and colorectal tumours, leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney.
4. Organotin(IV) compounds with N-acetylcysteine according to any one of claims 1 -3, wherein R1, R2 and R3 are each independently selected from:
H, an alkyl group with up to 8 carbon atoms, or an alkoxyl group with up to 8 carbon atoms or a phenyl group.
5. Diorganotin(IV) compounds with N-acetylcysteine according to claim 4 having the general formula (1 ), wherein R-i and R2 are each independently selected from methyl, ethyl, propyl, butyl and phenyl.
6. Triorganotin(IV) compounds with N-acetylcysteine according to claim 4 having the general formula (2), wherein R-, , R2 and R3 are each independently selected from methyl, ethyl, propyl, butyl and phenyl.
7. A diorganotin(IV) compound with N-acetylcysteine having the general formula (1 )
Figure imgf000038_0001
wherein:
R-i, R2 are each independently selected from: H, an alkyl, alkoxyl, alkenyl or alkynyl group with up to 12 carbon atoms, an isocyclic or heterocyclic aromatic or aralkyl group with up to 12 carbon atoms, for use as a medicinal active ingredient.
8. A compound according to claim 7, wherein R-i and R2 are each independently selected from methyl, ethyl, propyl, butyl and phenyl.
9. A compound according to claim 8 wherein R-, and R2 represent butyl, having the formula (1 a):
Figure imgf000039_0001
10. A triorganotin(IV) compound with N-acetylcysteine having the general formula (2)
Figure imgf000039_0002
wherein: R1, R2 R3 are each independently selected from:
H, an alkyl, alkoxyl, alkenyl or alkynyl group with up to 12 carbon atoms, an isocyclic or heterocyclic aromatic or aralkyl group with up to 12 carbon atoms, for use as a medicinal active ingredient.
11. A compound according to claim 10, wherein R-, and R2 are each independently selected from methyl, ethyl, propyl, butyl and phenyl.
12. A process for producing the organotin compounds with N-acetylcysteine having the formula (1 ) or (2) as defined in claims 1 , 4, 5 or 6, comprising the step of reacting an organotin derivative wherein R-i, R2 and R3 have the meaning recited in claim 1 with N-acetylcysteine in methanol, or in chloroform, or in water, in a temperature range comprised between 10°C and 90 °C.
13. A process according to claim 12, wherein at the end of the said reaction in methanol or in water or in chloroform the reaction mixture is quenched, and the resulting precipitate containing the desired compound of formula (1 ) or (2) is collected by filtration.
14. A process for producing the compound dibutyltin(IV)-N acetylcysteinate of the general formula (1 a), as defined in claim 9, comprising the step of reacting
N-acetylcysteine with dibutyltin oxide in methanol, or in water or in chloroform, in a temperature range comprised between 10°C and 90 °C.
15. A process according to claim 14, wherein at the end of the said reaction in methanol or in water or in chloroform the reaction mixture is quenched, and the resulting precipitate containing the desired compound of formula (1 a) is collected by filtration.
16. Use of one or more organotin(IV) compounds with N-acetylcysteine of the general formula (1 ) or (2), as defined in claims 1 , 4, 5 o 6, for the production of a pharmaceutical preparation.
17. Use according to claim 16, wherein the said pharmaceutical preparation is an anticancer preparation.
18. Use according to claim 17, wherein the said pharmaceutical preparation is an anticancer preparation for the treatment of hepatic and colorectal tumours, leukemia, melanoma and cancers of the lung, colon, brain, ovary, breast, prostate, and kidney.
19. A pharmaceutical preparation comprising, as active ingredient, at least one of the organotin(IV) compounds with N-acetylcysteine of the general formula (1 ) or (2), as defined in claims 1 , 4, 5 or 6, together with one or more pharmacologically acceptable adjuvants and/or vehicles.
20. A pharmaceutical preparation according to claim 19, comprising as an active ingredient the compound dibutyltin(IV)-N-acetylcysteinate of the formula (1 a), as defined in claim 9.
PCT/EP2009/060006 2008-08-04 2009-08-03 Organotin(iv) complexes with n-acetylcysteine possessing antitumoral activity, process for their production and their use WO2010015590A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ITRM2008A000426 2008-08-04
ITRM2008A000426A IT1392832B1 (en) 2008-08-04 2008-08-04 ORGANOSTAGNO (IV) COMPLEXES WITH N-ACETYLCISTEIN FOR ANTITUMOR ACTIVITY, PROCEDURE FOR THEIR PRODUCTION AND THEIR USE.

Publications (1)

Publication Number Publication Date
WO2010015590A1 true WO2010015590A1 (en) 2010-02-11

Family

ID=40800735

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2009/060006 WO2010015590A1 (en) 2008-08-04 2009-08-03 Organotin(iv) complexes with n-acetylcysteine possessing antitumoral activity, process for their production and their use

Country Status (2)

Country Link
IT (1) IT1392832B1 (en)
WO (1) WO2010015590A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011110838A3 (en) * 2010-03-08 2012-05-18 Procure Therapeutics Limited Therapeutic treatments and screening methods using p63

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CHASAPIS CHRISTOS T ET AL: "Organotin(IV) Derivatives of L-Cysteine and their in vitro Anti-Tumor Properties.", BIOINORGANIC CHEMISTRY AND APPLICATIONS 2004, 2004, pages 43 - 54, XP002536164, ISSN: 1565-3633 *
DOMAZETIS G, MAGEE R, JAMES B: "Reactions of triorganotin (IV) compounds with L-cysteine, L-cysteine ethyl ester, N-acetyl L-cysteine and glutathione reduced", INORGANICA CHIMICA ACTA, vol. 32, 1979, pages L48 - L50, XP002536163 *
DOMAZETIS G, MAGEE R, JAMES B: "Tri-n-butyltin (IV) derivatives of L-cysteine ethyl ester, N-acetyl-L-cysteine and alpha-glutamyl cysteinyl glycine (glutathione reduced)", JOURNAL OF ORGANOMETALLIC CHEMISTRY, vol. 173, 1979, pages 357 - 376, XP002536162 *
NATH M, GOYAL S, GOYAL S, ENG G, OGWURU N: "Synthesis, spectral and thermal studies of organotin (IV) complexes of N-acetylamino acids", SYNTHESIS AND REACTIVITY IN INORGANIC, METAL-ORGANIC NANO-METAL CHEMISTRY, vol. 28, no. 10, 1998, pages 1619 - 1641, XP002536161 *
SILVESTRI A, DUCA D, HUBER F: "A study of dimethyltin(IV)-L-cysteinate in aqueous solution", APPLIED ORGANOMETALLIC CHEMISTRY, vol. 2, 1988, pages 417 - 425, XP002536165 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011110838A3 (en) * 2010-03-08 2012-05-18 Procure Therapeutics Limited Therapeutic treatments and screening methods using p63

Also Published As

Publication number Publication date
ITRM20080426A1 (en) 2010-02-05
IT1392832B1 (en) 2012-03-23

Similar Documents

Publication Publication Date Title
Abdalbari et al. The gold complex auranofin: new perspectives for cancer therapy
Cheng et al. Current progresses in metal-based anticancer complexes as mammalian TrxR inhibitors
Schmidt et al. Are Pt (IV) prodrugs that release combretastatin A4 true multi-action prodrugs?
DK2575802T3 (en) Phosphaplatiner and use thereof for the treatment of cancer
CN102905705A (en) Novel iridium/rhodium anti-cancer compounds
McKeon et al. Platinum (IV) oxaliplatin–peptide conjugates targeting memHsp70+ phenotype in colorectal cancer cells
Shpakovsky et al. The antioxidant 2, 6-di-tert-butylphenol moiety attenuates the pro-oxidant properties of the auranofin analogue
Pettinari et al. Novel osmium (II)–cymene complexes containing curcumin and bisdemethoxycurcumin ligands
ES2683038T3 (en) Calmangafodipir, a new chemical entity, and other mixed metal complexes, preparation methods, compositions and treatment methods
Liu et al. Multi-action platinum (IV) prodrugs conjugated with COX-inhibiting NSAIDs
Li et al. A Pt (IV)-based mononitro-naphthalimide conjugate with minimized side-effects targeting DNA damage response via a dual-DNA-damage approach to overcome cisplatin resistance
Althobaiti et al. A comparative study of novel ruthenium (III) and iron (III) complexes containing uracil; docking and biological studies
JP2012520833A (en) Gold (III) complexes with oligopeptides functionalized with sulfur donors and their use as antitumor agents
Liao et al. Cyclometalated iridium (III) complexes induce immunogenic cell death in HepG2 cells via paraptosis
Ferreira-Silva et al. [Ru (pipe)(dppb)(bipy)] PF6: A novel ruthenium complex that effectively inhibits ERK activation and cyclin D1 expression in A549 cells
Huang et al. A cytotoxic nitrido-osmium (VI) complex induces caspase-mediated apoptosis in HepG2 cancer cells
He et al. Antitumor activity of iridium/ruthenium complexes containing nitro-substituted quinoline ligands in vivo and in vitro
WO2010015590A1 (en) Organotin(iv) complexes with n-acetylcysteine possessing antitumoral activity, process for their production and their use
Kamoon et al. Dithiocarbamates derivatives as anticancer agents: A Review
Zheng et al. Comparative evaluation of the structure and antitumor mechanism of mononuclear and trinucleated thiosemicarbazone Cu (II) complexes
WO2011125911A1 (en) Metal complexes and anticancer agents comprising same as active ingredient
US9220705B2 (en) Method of treating colorectal cancer
US11192902B1 (en) Heterocyclic diazenyl pyridinone copper(II) complexes as pharmacological antitumor agents
Stojanović et al. Platinum (II) complexes with malonic acids: Synthesis, characterization, in vitro and in vivo antitumor activity and interactions with biomolecules
US11459343B2 (en) Gold(I) N-heterocyclic carbene thiourea complex and therapeutic compositions thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09781400

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09781400

Country of ref document: EP

Kind code of ref document: A1