(19)
(11)EP 3 459 526 B1

(12)EUROPEAN PATENT SPECIFICATION

(45)Mention of the grant of the patent:
24.02.2021 Bulletin 2021/08

(21)Application number: 17193240.3

(22)Date of filing:  26.09.2017
(51)International Patent Classification (IPC): 
A61K 9/14(2006.01)
A61K 33/00(2006.01)
A61K 9/51(2006.01)

(54)

BIOAVAILABLE DITHIOCARBAMATE-METAL COMPLEX NANOPARTICLES, METHOD OF PREPARATION AND USE THEREOF

BIOVERFÜGBARE DITHIOCARBAMAT-METALL-KOMPLEX-NANOPARTIKEL, VERFAHREN ZUR HERSTELLUNG UND VERWENDUNG DAVON

NANOPARTICULES COMPLEXES DE DITHIOCARBAMATE MÉTALLIQUE BIODISPONIBLES,LEURS PROCÉDÉ DE PRÉPARATION ET D'UTILISATION


(84)Designated Contracting States:
AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

(43)Date of publication of application:
27.03.2019 Bulletin 2019/13

(73)Proprietor: Palacký University In Olomouc
771 47 Olomouc (CZ)

(72)Inventors:
  • Skrott, Zdenek
    75625 Ruzdka (CZ)
  • Mistrik, Martin
    78301 Olomouc (CZ)
  • Hajduch, Marian
    79305 Moravsky Beroun (CZ)
  • Bartek, Jiri
    2670 Greve (DK)
  • Zboril, Radek
    77900 Olomouc (CZ)
  • Dzubak, Petr
    75103 Brodek u Prerova (CZ)

(74)Representative: Hartvichova, Katerina et al
HARBER IP s.r.o. Dukelskych hrdinu 567/52
170 00 Praha 7
170 00 Praha 7 (CZ)


(56)References cited: : 
EP-A1- 1 214 063
US-A1- 2006 040 980
US-A1- 2003 229 064
  
     
    Remarks:
    The file contains technical information submitted after the application was filed and not included in this specification
     
    Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


    Description

    Field of the Invention



    [0001] The present invention relates to nanoparticles , as disclosed in the claims, of R1,R2-dithiocarbamate-metal complex and at least one blood protein, usable for obtaining antitumor compositions suitable for medical applications, including cancer treatment and diagnostics, as well as a process for producing such nanoparticles.

    Background Art



    [0002] R1,R2-dithiocarbamates (DTC) are strong metal ion chelators known in literature. Once complex between DTC and metal is formed some of these complexes display anticancer activity in cellular systems employing various cancer cell models. However, the complexes with proposed antitumor activity are not water soluble, which makes it particularly difficult to administer such complex to patients; also the complexes show low preferential toxicity towards cancer cells and thus nearly narrow therapeutic index. Both limitations can be overcome by present invention.

    [0003] Dithiocarbamates (DTC), particularly in complex with various bivalent metals, exhibit promising anticancer activity in various preclinical models. Metal chelating properties of DTC's are known for a long time, as well as their antitumor activity. Several patent documents have covered the use of dithiocarbamate complexes with heavy metals, especially with copper, zinc, gold or silver, as a treatment strategy for various malignancies (see e.g. US20030229064, US20050096304). However, none of these patent documents has been translated to practical use in humans so far. Apparently, the main obstacle for the use of dithiocarbamate-metal complexes in the clinical routine are unfavourable pharmacologic properties, namely stability and water based formulation capabilities. For instance, for the most promising anticancer complex which is bis(diethyldithiocarbamate)copper(II) (or copper bis(diethyldithiocarbamate)), the solubility constant in water is only in the range of nanograms per litre, which is insufficient to deliver therapeutic doses in patients.

    [0004] US 20030229064 and EP 1214063 relate to treatment of cancer by administering a dithiocarbamate, optionally either in the form of dithiocarbamate-heavy metal ion complex, or in the form of a dithiocarbamate or dithiocarbamate anion with ceruloplasmin.

    [0005] US 20060040980 discloses a method of treatment of cancer, wherein ionophore which is dithiocarbamate or clioquinol, and transition metal or metal-albumin complex are administered in a sequence, wherein the ionophore is administered prior to or preferably after the administration of metal or albumin-metal complex.

    [0006] CN 103222961 discloses nanoparticles which are produced from thiocarbamate-copper salt and protein, wherein the thiocarbamate-copper salt is added in organic solvent. This results in particles containing organic solvent which are not suitable for medical treatment.

    Summary of the Invention



    [0007] The main object of the present invention is a novel nanoparticulate form of dithiocarbamate-metal complex and at least one blood protein according to claim 1, and process for its preparation according to claim 6, the nanoparticulate form being useful as anticancer drug or as a diagnostic agent.

    [0008] According to the present invention, the nanoparticulate form (herein also referred to as nanoparticles) is prepared by combining a first component selected from dithiocarbamate and metal salt with at least one blood protein in an aqueous solvent, and subsequently adding a second component selected from dithiocarbamate and metal salt, whereas if the first component is dithiocarbamate, then the second component is metal salt; and if the first component is metal salt, then the second component is dithiocarbamate.

    [0009] Within the framework of the present invention, it was discovered that when the sequential addition of the reagents is carried out as described in claim 6, then after the addition of the second component, the blood proteins have a considerable capacity to bind to the dithiocarbamate-metal complex that is rapidly formed in the solution and spontaneously assemble into nanoparticles wherein at least 50 % of the nanoparticles have the size within the range of 2-200 nm, thus forming an injectable bioavailable dispersion of nanoparticles. In this nanoparticulate form the molecules of dithiocabamate-copper complex are uniformly distributed, maintain their original chemical properties and exhibit a substantially improved biological activity, both in vitro and in vivo, thus enabling an unexpected pharmaceutical use of otherwise water-insoluble compound, including the use in cancer treatment and tumour visualization.

    [0010] The process of nanoparticle preparation can be performed in a very short time (below 1 minute), even at the patient's bed, in a single reaction vessel without the need of non-polar solvents and which allows either immediate or sustained parenteral administration.

    Detailed Description of the Invention



    [0011] In the present invention, "metal" means a metal selected from copper, zinc, silver and gold.

    [0012] The metal may be in the form of a single isotope or an isotopic mixture. The isotopes may be radioactive isotopes or non-radioactive isotopes. For copper, non-radioactive isotopes are 63Cu and 65Cu, and radioactive isotopes are preferably 6Cu or 67Cu. 64Cu is a positron emitting isotope of copper, with applications for molecular radiotherapy and positron emission tomography.

    [0013] "Metal salt" means a salt of the metal in the form of a cation with an anion. With regard to the intended pharmaceutical use of the nanoparticle solution, the skilled person would understand that the anion should be a pharmaceutically acceptable anion and preferably water-soluble. The anion may be selected, e.g. from inorganic acid anions such as halogenides (in particular chlorides, bromides, iodides), sulfates, sulfites, sulfides, phosphates, nitrates, carbonates; carboxylic acid anions, dicarboxylic acid anions, tricarboxylic acid anions, sulfonic acid anions, amino acid anions, such as formates, acetates, propionates, oxalates, succinates, maleinates, fumarates, maleates, citrates, triflates, gluconates, bis-glycinates.

    [0014] "Dithiocarbamate" means a moiety having the formula (R1)(R2)N-CS2- (also referred to in this text as R1,R2-dithiocarbamate), wherein R1 and R2 are the same or different and are independently selected from C1-C8 alkyl, C2-C8 alkenyl, C3-C10 cycloalkyl, C6-C14 aryl, C4-C14 heteroaryl containing at least one heteroatom selected from O, S, N, C3-C10 heterocyclyl containing at least one heteroatom selected from O, S, N; or R1 and R2 together with the nitrogen atom on which they are bound form a heterocycle, wherein -R1-R2- is a C2-C6 alkylene or a C2-C6 alkenylene, wherein optionally 1-2 carbon atoms may be replaced by heteroatoms selected from O, S, NH. The moieties forming R1 and R2 may be unsubstituted or further substituted by at least one substituent selected from C1-C4 alkyl, hydroxy, mercapto, C1-C4 alkoxy, C1-C4 alkylthio, halogen, phenyl, benzyl, keto group, carboxyl group, C1-C4 alkyloxycarbonyl.

    [0015] More preferably, R1 and R2 are independently selected from C1-C6 (or C1-C4) alkyl, C2-C6 (or C2-C4) alkenyl, C3-C6 cycloalkyl, phenyl; or R1 and R2 together with the nitrogen atom on which they are bound form a heterocycle, wherein -R1-R2- is a C2-C6 alkylene or a C2-C6 alkenylene.

    [0016] Dithiocarbamate can be present in the form of the negatively charged anion, typically in the dithiocarbamate-metal complex. As a starting compound in the process of the present invention, it may be used in the form of a neutral compound (R1)(R2)N-CH2(S)SH or, preferably, in the form of a salt [(R1)(R2)N-CH2S2]m-Catm+, such as alkali metal salt (Cat+ is an alkali metal cation, m=1), ammonium salt (Cat+ is an ammonium cation, m=1) or alkaline earth metal salt (Cat+ is an alkaline earth metal cation, m=2). The skilled person understands which form is meant or which form is necessary, depending on the context in which the term "dithiocarbamate" is used.

    [0017] "Dithiocarbamate-metal complex" is a complex comprising at least one dithiocarbamate moiety and at least one metal, preferably one metal. For example, the dithiocarbamate-metal complex may correspond to formula (I)

    wherein

    M is a metal, preferably copper,

    An is a pharmaceutically acceptable anion, preferably as defined herein above,

    n is the valence of the d-metal, typically, n is 1, 2, or 3,

    and R1 and R2 are as defined herein above.



    [0018] The ratio of the metal to dithiocarbamate may be for example in the range of from 1:5 to 5:1, or in the range of 1:2 to 5:1. The ratio of the metal to dithiocarbamate may optimally correspond to their stoichiometric ratio in the complex, or to their stoichiometric ratio ± 20 %, or to their stechiometric ratio ± 50 %. For example, for copper the stoichiometric ratio is 2:1.

    [0019] "Blood protein" means a blood serum proteins selected from the group comprising albumin, transferrin, immunoglobulin, and mixtures comprising these proteins, including whole blood, blood plasma and blood serum. Preferably, the present invention uses human blood proteins, but also blood proteins of other animals, preferably mammals, can be used, such as dog, cat, horse, mouse, rat, rabbit, goat, sheep, cattle blood proteins. The blood proteins may be isolated from blood or blood serum, respectively, or may be of recombinant origin.

    [0020] Most, preferably, the blood serum protein is albumin.

    [0021] "Aqueous solvent" is water or a water-based buffer, such as phosphate, citrate, acetate, Tris, HEPES, saline or other common buffers. Preferably, the aqueous solvent is sterile.

    [0022] The size of the self-assembled nanoparticles (i.e., molecular assemblies) is such that at least 50 %, or at least 70 %, of the nanoparticles have the size within the range of 2-200 nm, or within the range of 10-100 nm, or within the range of 10-200 nm. The particle sizes and their distributions were measured by Dynamic Light Scattering method.

    [0023] The percentages, unless indicated otherwise, are w/v %.

    [0024] The present invention describes the process of in-situ self-assembly of dithiocarbamate-copper complex and at least one blood protein into a nanoparticle. This is attained by a process comprising the steps of:
    1. (a) solubilising at least one blood protein in an aqueous solvent (e.g. water or water-based buffer) to a concentration in the range from 0.01% (w/v) to saturated solution (preferably from 0.1% to 10% (w/v)), or providing whole blood or blood plasma or blood serum of a patient;
    2. (b) adding at least one dithiocarbamate dissolved in an aqueous solvent (e.g. water or water-based buffer) to a concentration in the range from 1 uM to 100 mM, preferably 1 to 10 mM;
    3. (c) adding a metal salt solution in an aqueous solvent (e.g. water or water-based buffer), having the metal salt concentration in the range from 1 uM to 100 M, preferably 1 to 10 mM,
    wherein the steps are carried out in the sequence (a), (b), (c) or in the sequence (a), (c), (b). Preferably, at least 10 second-delay accompanied by shaking or vortexing is made between individual steps.

    [0025] This single-tube reaction leads to rapid spontaneous self-assembly of protein-dithiocarbamate-copper nanoparticulate form forming a dispersion of nanoparticles.

    [0026] Step (a) is preferably performed at a temperature in the range from 10 to 45 °C, preferably 20 to 38 °C, and/or at pH in the range from 5 to 8, most preferably 6.8.

    [0027] In step (b), the dithiocarbamate is preferably in the form of a neutral compound or a salt.

    [0028] In a preferred embodiment, the molar ratio of metal ions:dithiocarbamate ions is 1:2.

    [0029] The nanoparticles of the present invention form a bioavailable dispersion of nanoparticles, and can be administered to a patient in need of such treatment. The dispersion of nanoparticles can be used in therapy, in particular cancer therapy, both chemotherapy and radiotherapy, such as therapy of solid tumors including melanoma, non-small cell lung cancer, small cell lung cancer, renal cancer, colorectal cancer, breast cancer, pancreatic cancer, gastric cancer, bladder cancer, ovarian cancer, uterine cancer, lymphoma, prostate cancer, adenocarcinoma of the colon and nodal or hepatic metastases. The dispersion of nanoparticles can also be used in diagnostics, such as tumor visualisation, e.g., by positron emission tomography.

    [0030] It is important to note that in case the reaction is performed according to the present invention the resulting dispersion of protein-dithiocarbamate-copper nanoparticles allows direct parenteral applications to the treated subject (human or animal) without the need of additional chemical or physical processing such as extractions, separations, product cleaning, concentration enhancement etc.

    [0031] Moreover, the reaction of the present invention can be performed directly at the bed of the patient or in the hospital pharmacy using a combination of pharmaceutically acceptable ingredients. For example human serum albumin solution, diethyldithiocarbamate and CuCl2 are commonly commercially available in pharmaceutical grades. Such procedure does not require costly chemical reactors, processing in additional devices and may simplify regulatory approval. In addition, it significantly limits some of the logistic problems related to the storage of the dispersion of nanoparticles - a fresh drug can be prepared with high reproducibility and immediately applied.

    [0032] The present invention also enables simple modification of the size of assembled nanoparticles. By changing the ratio between dithiocarbamate-metal (in particular copper) complex and proteins, the formed nanoparticles are of different size following a rule that a higher protein concentration leads to smaller particles. Optimum reaction conditions can be determined to produce nanoparticles with the optimum pharmacologic properties, as the size of nanoparticles is important determinant of its behaviour in-vivo, in particular for biodistribution (e.g. blood-brain barrier penetration) and kinetics. The size of manufactured nanoparticles also depends on the substituents R1, R2 of the dithiocarbamate entering into the reaction. For example, dimethylditihiocarbamate with albumin forms larger particles compared to pyrrolidinedithiocarbamate with albumin despite all the other conditions of the two reactions being identical, suggesting smaller particles formation with longer R1, R2 groups.

    [0033] The prepared dispersion of nanoparticles is relatively stable and can be stored for several weeks at 4 °C without significant degradation or precipitation. The formed nanoparticles of protein-dithiocarbamate-metal can be further processed by drying or lyophilisation to further improve stability, storage and logistics. Dried nanoparticles can be repeatedly dissolved in sterile water-based buffers and used for therapy. This important aspect of the protein-dithiocarbamate-metal nanoparticles properties is particularly valuable for both large- or small-scale industrial production, storage and logistics.

    [0034] As immunoglobulin or immunoglobulin-containing mixtures can be used in the production of the nanoparticles, the resulting nanoparticles may harbour antibodies (or antibody fragments) against specific tumour antigens. This approach enables specific targeting of the nanoparticle into the tumour, yielding a better anticancer effect and a reduced systemic toxicity.

    [0035] To prove general applicability of the nanoparticles, the following examples show preparation and characterization of dithiocarbamate-metal complex nanoparticles with blood proteins including albumin, transferrin and immunogblobulins alone or in combinations to demonstrate generic formulation and cancer targeting and tumour visualization capabilities of the resulting dispersion of nanoparticles. The examples should not be construed as limiting the scope of the claimed invention which is determined by the claims.

    Brief Description of Drawings



    [0036] 

    Figure 1 shows dynamic light scattering (DLS) spectra of the nanoparticles prepared in Example 1. The nanoparticles form a polydispersed system: ca 20 - 100 nm; largest fraction: 30-40 nm.

    Figure 2 shows representative atomic force microscopy (AFM) image showing the particles with vertical dimensions ranging from approx. 20 to 40 nm prepared according to Example 1.

    Figure 3 shows representative transmission electron microscopy (TEM) images of the nanoparticles (approx.. 30 nm); no indication of crystal planes, no electron diffraction, overall character of a molecular assembly (nanoparticles) prepared according to Example 1.

    Figure 4 shows HAADF/EDS images of cca 60 nm molecular assembly; key elements (Cu, N, S) are clearly embedded in the nano-assembly prepared according to Example 1.

    Figure 5 shows X-band electron paramagnetic resonance (EPR) spectra of bis(diethyldithiocarbamate) copper complex (CuET) (a-b are the resonances for the neat CuET powder obtained from TOKYO CHEMICAL INDUSTRY CO.,LTD.; c-d, are the resonances for the CuET-albumin nanoparticles prepared according to Example 1). The estimated gavg-value of 2.044 and the overall resonance profile clearly indicate that upon nanoparticle formation no modification of the copper oxidation-state, and no substantial alteration of the Cu octahedral-field occur.

    Figure 6 shows DLS spectra (upper image) and representative TEM images (lower two images) of polydispersed system of nanoparticles formed during chemical reaction in the presence of 0.01% albumin (Example 2) or 5% albumin (Example 3). The analysis shows a prominent shift towards smaller particles in case higher albumin concentration is entering the chemical reaction.

    Figure 7 shows DLS size distribution (upper image) and representative TEM images (lower two images) of the formed nanoparticles depicting different size of formed nanoparticles (Examples 4, 5) depending on the used type of dithiocarbamate (dimethylditihiocarbamate - DMC vs pyrrolidinedithiocarbamate - PDT)

    Figure 8 shows dynamic light scattering (DLS) spectra of the nanoparticles prepared in Example 7. The nanoparticles form a polydispersed system: ca 10 - 40 nm; largest fraction: 15-30 nm.

    Figure 9 shows dynamic light scattering (DLS) spectra of the nanoparticles prepared in Example 8. The nanoparticles form a polydispersed system: ca 15 - 100 nm; largest fraction: 20-40 nm.

    Figure 10 shows dynamic light scattering (DLS) spectra of the nanoparticles prepared in Example 9. The nanoparticles form a polydispersed system: ca 50 - 800 nm; largest fraction: 60-200 nm.

    Figure 11 shows 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide (XTT) - based cell viability analysis of CuET-albumin (CuET-HSA) nanoparticle toxicity prepared according to Example 1 after 24 hours treatment of various concentrations (from 125 nM to 10 µM). Cancer cell lines (DU-145, MDA-MB-231) show higher sensitivity compared to normal cells (RPE-1, RWPE-1). CuET-HSA nanoparticle has similar potency as neat CuET powder dissolved in dimethyl sulfoxide (DMSO) in cancer cell lines, but it is more tolerated by normal cells.

    Figure 12 shows high pressure liquid chromatography coupled to time-of-flight mass spectrometry (HPLC-TOF/MS) analysis of CuET concentrations in mice serum after single i.p. application of 5 mg/kg CuET formulated as albumin nanoparticle according to Example 1 or as the neat CuET powder (obtained from TOKYO CHEMICAL INDUSTRY CO., LTD.) dissolved in olive oil which is a standard formulation for non-polar compounds applied intraperitoneally in the mice experiments. Measured concentrations show significantly higher circulating concentrations of CuET in case of the nanoparticle formulation.

    Figure 13 shows the presence of the Cu64 radioisotope in the tumours (marked by arrows) visualised by combined positron emission tomography combined with computer tomography (PET/CT) imaging in severe combined immunodeficiency (SCID) mouse. The PET signal was collected 22 hours after the retro-orbital injection of dispersion of nanoparticles of MSA-diethyl-dithiocarbamate-copper64 nanoparticles (left) and dispersion of nanoparticles of human transferrin-diethyldithiocarbamate-copper64 nanoparticles (right) prepared according to examples 10 and 11 respectively. PET signal shows that the Cu64 embedded in complexes is accumulated within the MDA-MB-231 subcutaneous tumour mass. GIT positivity indicates that the compounds are excreted by intestinal tract.

    Figure 14 shows the growth of MDA-MB-231 breast cancer tumours subcutaneously xenografted into the SCID mice after treatment with CuET (1 mg/ml) formulated as either albumin nanoparticle according to Example 1 or as the neat CuET powder (obtained from TOKYO CHEMICAL INDUSTRY CO., LTD.) dissolved in olive oil which is a standard formulation for non-polar compounds applied intraperitoneally in the mice experiments. Only the CuET-albumin nanoparticle shows significant effect on the tumour growth.

    Figure 15 shows the growth of AMO-1 tumours subcutaneously xenografted into the SCID mice treated with CuET (1 mg/ml) formulated as albumin nanoparticle according to Example 1. The nanoparticles treatment significantly reduces the tumour growth (upper image, callipers) and the treated animals show prolonged survival (lower image, Kaplan-Meyer survival plots) compared to control mice treated only by albumin. The animals were treated by intraperitoneal application once a day in a regime 5 days' treatment + 2 days off. These experiments points also at a very good tolerability of the CuET-albumin nanoparticles as the animals were injected more than 100 times.


    Examples


    Materials and Methods



    [0037] Dynamic Light Scattering (DLS) analyses allowing to determine the average size and size distribution of the prepared nanoparticles were performed by the Zetasizer Nano ZS instrument (Malvern, U.K.), with following parameters setting: V = 400 uL, T = 25 °C, Number of runs: 10, Run duration: 1s, Number of measurements: 3, Measurement angle: 173° Backscatter (NIBS default), Cell type: ZEN0040.
    Transmission Electron Microcopy (TEM) images were obtained with TEM JEOL 2010 instrument with LaB6 type emission gun, operating at 100 kV. The analysis of crystal structure/amorphicity of nanoparticles is allowed with Selected Area Electron Diffraction (SAED) analysis. STEM/HAADF (Scanning Transmission Electron Microscopy/High-Angle Annular Dark-Field) analyses for EDS (Energy Dispersive X-ray Spectroscopy) mapping of elemental composition of nanoparticles were performed with a FEI Titan G2 HRTEM microscope operating at 80 kV. For STEM measurements, Super-X system with four silicon drift detectors (Bruker) was used. For all these microscopis analyses, a droplet of dispersion containing nanoparticles was deposited on a carbon-coated copper grid and slowly dried at room temperature.
    Atomic Force Microscopy (AFM) images were taken by scanning tunneling microscope Ntegra (Nt-MDT company) in the semi - contact AFM mode. The sample was scanned with Ha-NC tip. For the sample preparation, one drop of the dispersion containing nanoparticles was placed on the mica support and dried at room temperature.

    [0038] Electron Paramagnetic Resonance (EPR) spectra of the frozen dispersion of nanoparticles were recorded at 140 K on JEOL JES-X-320 instrument operating at X-band frequency (9.17 GHz), equipped with a variable temperature control ES 13060DVT5 apparatus. High purity quartz tubes were employed (Suprasil, Wilmad, ≤ 0.5 OD). Other experimental parameters: 100 KHz modulation frequency, 0.1 mW microwave power, 0.03 s time constant, 8 Gauss modulation width.

    Cell lines



    [0039] Cell lines were cultured in appropriate medium supplemented with 10% fetal bovine serum and penicillin/streptomycin; and maintained at humidified, 5% CO2 atmosphere at 37°C. Lines cultured in DMEM medium were: HCT116 (obtained from American Type Culture Collection, ATCC), DU145 (obtained from European Collection of Authenticated Cell Cultures, ECACC), MDA-MB-231 (ATCC), U-2-OS (ECACC), HeLa (ATCC), CAPAN-1 (ATCC), A253 (ATCC), FaDu (ATCC), h-TERT-RPE1 (ATCC). Cell lines cultured in RPMI1640 medium were: AMO-1 (ATCC), MM-1S (ATCC), OVCAR-3 (NCI60), CCRF-CEM (ATCC), K562 (ATCC), 786-0 (NCI60). Cell lines cultured in EMEM medium were: U87-MG (ATCC), SiHA (ATCC). Cell line A549 (ATCC) was cultured in F12K medium, HT29 (ATCC) in McCoy's medium. RWPE-1 (ATCC) cells were cultured in a keratinocyte serum-free medium supplemented with the bovine pituitary extract and human recombinant epidermal growth factor (Thermo Scientific).

    Cell viability test



    [0040] Cell viability was measured by XTT test. 10000 cells were seeded to 96-well plate. Next day the cells were treated as indicated. After 24 hours XTT assay was performed according manufacturer instructions (Applichem). XTT solution was added to media and incubated for 30-60 minutes, and then dye intensity was measured at 475 nm wavelength using spectrometer (TECAN, Infinite M200PRO). Results are shown as mean values and standard deviations from 3 independent experiments, each performed in 3 replicates. For LD50 (lethal dose) analysis across panel of cell lines listed in Extended data 2d, cell lines were treated with various doses (at least 5 doses) for 48 hours. LD50s are calculated using Graphpad Prism software based on survival curves from at least two independent experiments.

    HPLC-MS detection of copper-diethyldithiocarbamate complex in mice



    [0041] The copper-diethyldithiocarbamate analysis was performed on HPLC-ESI-QTOF system consisting of HPLC chromatograph Thermo UltiMate 3000 with AB Sciex TripleTOF 5600+ mass spectrometer, using the DuoSpray ESI. Data were acquired in Product ion mode with two parent masses 358.9 and 360.9 for analysis of CuET. Chromatographic separation was done by PTFE column especially designed for analysis of strong metal chelators filled by C18 sorbent (IntellMed, cat.no.IM_301). Analysis was performed at room temperature and flow rate 1500 µL/min with isocratic chromatography. Mobile phase consist of HPLC grade acetone (Lachner) 99.9%, HPLC water (Merck Millipore) 0.1% and 0.03% HPLC formic acid (Sigma). Acquired mass spectra were evaluated in software PeakView 1.2, where extracted ion chromatograms of transitions 88.0 and 116.0 (common for both parent masses) with 0.1 mass tolerance was Gaussian smoothened with width of 2 points.

    PET imaging



    [0042] The presence of the Cu64 radioisotope in the tumours was visualised by combined positron emission tomography combined with computer tomography (PET/CT) imaging in severe combined immunodeficiency (SCID) mouse bearing MDA-MB-231 xenografts. The PET signal was collected 22 hours after the retro-orbital injection of dispersion of nanoparticles of MSA-diethyldithiocarbamate-copper64 nanoparticles and dispersion of nanoparticles of human transferrin-diethyldithiocarbamate-copper64 nanoparticles corresponding to activity 33 MBq and 29 MBq, respectively. Signal was recorded using Albira PET/SPECT/CT imaging system (Bruker Biospin Corporation, Woodbridge, CT, USA).

    Mice in-vivo experiments



    [0043] To test directly effect of CuET-albumin nanoparticles we used MDA-MB-231 and AMO1 models. MDA-MB-231 was injected (5*106 cells were transplanted s.c.) to grow tumours in SCID mice (ENVIGO, NL). AMO-1 xenografts were expanded in SCID mice. Each group consisted of 10 animals, each bearing two tumours. CuET was formulated as nanoparticulate system in mouse serum albumin according to Example 12 or as the neat CuET powder (obtained from TOKYO CHEMICAL INDUSTRY CO., LTD.) dissolved in olive oil which is a standard formulation for non-polar compounds applied intraperitoneally in the mice experiments to final concentration 1 mg/ml. Olive oil alone and mouse serum albumin solutions were used as the treatment controls. All solutions were applied intraperitoneally in a schedule 5 day ON and 2 day OFF, the CuET was applied in the final dose 1mg per kg of body weight. All aspects of the animal studies met the accepted criteria for the care and experimental use of laboratory animals, and protocols were approved by the Animal Research Committee of the 1st Faculty of Medicine Charles University in Prague and Ethical Committee of Faculty of Medicine and Dentistry, Palacky University in Olomouc.

    Protein identification using HPLC-MS



    [0044] Acetone precipitated proteins were dissolved in the digestion buffer (8 M urea, 0.5 ammonium bicarbonate, pH = 8) to a protein concentration of 1 - 5 µg/µl). Proteins in the sample were treated with dithiothreitol for reduction for 30 min at 56 °C and followed by iodoacetamide for alkylation for 30 min at room temperature in the dark. Sample was diluted to 0.8 M urea with 50 mM ammonium bicarbonate buffer and proteins were digested with trypsin (1/60) at 37 °C overnight. Digestion was stopped by adding TFA (pH = 2). The peptides were purified by using C18 column. The sample was measured using LC-MS consisting of a Dionex UltiMate 3000 RSLCnano system (Thermo Fisher Scientific) coupled via an EASY-spray ion source (Thermo Fisher Scientific) to an Orbitrap Elite mass spectrometer (Thermo Fisher Scientific). Purified peptides were separated on 50 cm EASY-Spray column (75 µm ID, PepMap C18, 2 µm particles, 100 Å pore size; Thermo Fisher Scientific). For each LC-MS/MS analysis, about 1 µg peptides were used for 165 min runs. First 5 min, peptides were loaded onto 2 cm trap column (Acclaim PepMap 100, 100 µm ID, C18, 5 µm particles, 100 Å pore size; Thermo Fisher Scientific) in loading buffer (98.9%/1%/0.1%, v/v/v, water/acetonitrile/formic acid) at a flow rate of 6 µl/min. Thereafter has been switched valve and peptides were loaded in buffer A (99.9%/0.1%, v/v, water/formic acid) and eluted from EASY-Spray column with a linear 120 min gradient of 2%-35% of buffer B (99.9%/0.1%, v/v, acetonitrile/formic acid), followed by a 5 min 90% B wash at a flow rate 300 nl/min. EASY-Spray column temperature was kept at 35°C. Mass spectrometry data were acquired with a Top12 data-dependent MS/MS scan method. Target values for the full scan MS spectra were 1 x 106 charges in the 300-1700 m/z range, with a maximum injection time of 35 ms and resolution of 120,000 at m/z 400. The 2 m/z isolation window was used for MS/MS scans. Fragmentation of precursor ions was performed by CID dissociation with a normalized collision energy of 35. MS/MS scans were performed in ion trap with ion target value of 1 x 104 and maximum injection time of 100 ms. Dynamic exclusion was set to 70 s to avoid repeated sequencing of identical peptides.

    Protein Data Analysis



    [0045] Mass spectrometry raw files were analysed using the MaxQuant software environment (version 1.5.6.5), and its built-in Andromeda search engine. Proteins were identified by searching MS and MS/MS data against the human proteome from UniProtKB (UP000005640, January 2017) and common contaminants database. Carbamidomethylation of cysteines was set as a fixed modification. N-terminal acetylation and oxidation of methionines were set as variable modification. Trypsin was set as protease, and a maximum of two missed cleavages were allowed in the database search. Peptide identification was performed with an allowed initial precursor mass deviation up to 7 ppm (Orbitrap) and an allowed fragment mass deviation of 0.5 Da (collision-induced dissociation, ion trap). The "matching between runs" option was enabled to match identifications across samples within a time window of 20 s of the aligned retention times. The false discovery rate was set to 0.01 for both proteins and peptides with a minimum length of seven amino. LFQ was performed with a minimum ratio count of 2. Protein abundances were calculated on the basis of summed peptide intensities of unique and "razor" peptide. Protein matching to the reverse database and proteins identified only with modified peptides were filtered out.

    EXAMPLE 1



    [0046] Preparation of dispersion of nanoparticles from 0.2% human serum albumin (HSA) and diethyldithiocarbamate and copper chloride salt.

    Procedure:



    [0047] An injectable aqueous 20% (w/v) HSA solution in accordance with FDA specifications (pH=6.9±0.5) is diluted to 0.2% (w/v) with sterile water for injections. Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to 0.2% HSA to final concentration 5.6 mM, followed by brief stirring. To the 0.2% HSA solution containing 5.6 mM DTC is added sterile copper (ii) chloride (1 M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results:



    [0048] The resulting solution contains 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper-HSA nanoparticles. The nanoparticles resulting from this reaction were further analysed by various advanced analytical tools including DLS, AFM, TEM, EDS and EPR depicting their basic physical and chemical properties (see figures 1-5). The analyses show that the nanoparticles form a polydispersed system. The size of the particles spanned the range ca 20 - 100 nm with largest fraction of 20-40 nm as proved by DLS analysis (figure 1). AFM image (figure 2) and TEM image (figure 3) confirm the results of DLS size distribution. Moreover, transmission electron microscopy images of the nanoparticles show no indications of crystal planes (and no electron diffraction with amorphous-like pattern) - the facts confirming that nanoparticles have a character of a molecular assembly. EDS chemical mapping of single nanoparticle (figure 4) shows that the key elements (Cu, N, S) of copper bis(diethyldithiocarbamate) structure are homogeneously distributed within the molecular assembly. Comparative analysis by X-band EPR spectroscopy (figure 5) shows nearly the same spectra of the CuET embedded in the nanoparticles as the neat bis(diethyldithiocarbamate) copper complex (CuET). The estimated gavg-values and the overall resonance profiles clearly indicate that upon encapsulation of the drug no modification of the copper oxidation and spin states (Cull, S=1/2), and no substantial alteration of the Cu octahedral-field occur. Resulting particles show also low zeta potential (ζ) -35,6 mV measured by electrochemical impedance spectroscopy pointing at good stability in solution.

    [0049] The dispersion of nanoparticles was also tested in biological experiments involving cytotoxicity tests on a panel of human cell lines derived from cancers including plasmacytoma (AMO-1; LD50 = 63 nM), pancreatic adenocarcinoma (Capan-1; LD50 = 64 nM), multiple myeloma (MM1s; LD50 = 69 nM), acute lymphoblastic leukemia (CCRF-CEM; LD50 = 70 nM), ovarian adenocarcinoma (OVCAR3; LD50 = 83 nM), lung carcinoma (A549; LD50 = 91 nM), colorectal carcinoma (HCT116; LD50 = 117 nM), salivary gland carcinoma (A253; LD50 = 214 nM), renal cell adenocarcinoma (786-O; LD50 = 235 nM), osteosarcoma (U2OS; LD50 = 271 nM), squamous cell carcinoma (FADU; LD50 = 330 nM, SiHA; LD50 = 777 nM), chronic myelogenous leukemia (K562; LD50 = 318 nM), breast adenocarcinoma (MDA-MB-231; LD50 = 517 nM), glioblastoma (U87-MG; LD50 = 696 nM), and primary (normal) cells including normal retinal epithelial cells (hTERT-PRE1; LD50 = >1000 nM) and normal prostate epithelial cells (RWPE; LD50 >2000nM). Toxicity of the nanoparticles was tested in the XTT - based cell viability assay. Cancer cell lines (DU-145, MDA-MB-231) show higher sensitivity compared to normal cells (RPE-1, RWPE-1). Importantly, CuET-HSA nanoparticle has similar potency as neat CuET powder dissolved in dimethyl sulfoxide (DMSO) in cancer cell lines, but it is more tolerated by normal cells suggesting general preferential toxicity to tumour cells (see figure 11).

    [0050] The dispersion of nanoparticles was also examined in a long-term storage stability test at 4°C. As readout the activity in cell culture was chosen. Nanoparticles stored for 4 weeks at 4°C showed similar potency in terms of cellular toxicity as freshly prepared nanoparticles in the XTT cell viability test using osteosarcoma line U2OS (LD50 280 nM compared to 271 nM).

    [0051] The dispersion of nanoparticles was also tested for the possibility of drying and follow-up re-solubilisation. The nanoparticles were dried under vacuum for 16 hours at room temperature. Dried powder was stored at 4°C for one week and then mixed with sterile water and keep on shaker for 24 hours to achieve complete re-solubilisation. Resulting re-solubilized nanoparticles were analysed by DLS and electrochemical impedance spectroscopy. Measured most probable average particle size of 82 nm and zeta potential (ζ) - 40 mV indicates minimal changes in the chemical/physical properties of nanoparticles after drying and re-solubilisation process.

    EXAMPLE 2



    [0052] Preparation of dispersion of nanoparticles from 0.01% human serum albumin (HSA) and diethyldithiocarbamate and copper chloride salt.

    Procedure:



    [0053] An injectable aqueous 20% (w/v) HSA solution in accordance with FDA specifications (pH=6.9±0.5) is diluted to 0.01% (w/v) with sterile water for injections. Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to 0.01% HSA to final concentration 5.6 mM, followed by brief stirring. To the 0.01% HSA solution containing 5.6 mM DTC is added sterile copper (ii) chloride (1M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0054] Resulting solution contains 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper-HSA nanoparticles. Then nanoparticles were further examined by DLS and TEM showing that the major fraction of nanoparticles is approx. 50-60 nm with a significant fraction of larger particles, but the sizes did not exceed 500 nm (see figure 6).

    EXAMPLE 3



    [0055] Preparation of dispersion of nanoparticles from 5% human serum albumin (HSA) and diethyldithiocarbamate and copper chloride salt.

    Procedure:



    [0056] An injectable aqueous 20% (w/v) HSA solution in accordance with FDA specifications (pH=6.9±0.5) is diluted to 5% (w/v) with sterile water for injections. Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to 5% HSA to final concentration 5.6 mM, followed by brief stirring. To the 5% HSA solution containing 5.6 mM DTC is added sterile copper (ii) chloride (1 M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0057] Resulting solution contains 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper-HSA nanoparticles. This reaction results into nanoparticles which were examined by DLS and TEM showing the major fraction of particles approx. 20-30 nm; generally the size distribution is significantly shifted towards smaller particles with increased HSA concentration (See figure 6).

    EXAMPLE 4



    [0058] Preparation of dispersion of nanoparticles from 1% human serum albumin (HSA) and dimethyldithiocarbamate and copper chloride salt.

    Procedure:



    [0059] An injectable aqueous 20% (w/v) HSA solution in accordance with FDA specifications (pH=6.9±0.5) is diluted to 1% (w/v) with sterile water for injections. Sterile solution of dimethyldithiocarbamate sodium salt (DMC) solubilised in water of a concentration 280 mM is added to 1% HSA to final concentration 5.6 mM, followed by brief stirring. To the 1% HSA solution containing 5.6 mM DMC is added sterile copper (ii) chloride (1 M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0060] Resulting solution contains 2.8 mM (1 mg/ml) of dimethyldithiocarbamate-copper-HSA nanoparticles. This reaction results into nanoparticles which were examined by DLS and TEM showing a major fraction of particles in the range of 60-70 nm (see figure 7.).

    EXAMPLE 5



    [0061] Preparation of dispersion of nanoparticles from 1% human serum albumin (HSA) and pyrrolidinedithiocarbamate and copper chloride salt.

    Procedure:



    [0062] An injectable aqueous 20% (w/v) HSA solution in accordance with FDA specifications (pH=6.9±0.5) is diluted to 1% (w/v) with sterile water for injections. Sterile solution of pyrrolidinedithiocarbamate amonium salt (PDC) solubilised in water of a concentration 280 mM is added to 1% HSA to final concentration 5.6 mM, followed by brief stirring. To the 1% HSA solution containing 5.6 mM PDC is added sterile copper (ii) chloride (1 M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0063] Resulting solution contains 2.8 mM (1 mg/ml) of pyrrolidinedithiocarbamate-copper-HSA nanoparticles. This reaction results into nanoparticles which were examined by DLS and TEM showing a major fraction of particles about 15 nm (see figure 7). Evidently, dimethylditihiocarbamate with albumin forms larger particles compared to pyrrolidinedithiocarbamate (PDT) with albumin despite all the other conditions of the two reactions being identical. This fact reflects that smaller particles are formed with longer R1, R2 groups.

    EXAMPLE 6



    [0064] Preparation of dispersion of nanoparticles from 1% human serum transferrin and diethyldithiocarbamate and copper chloride salt.

    Procedure:



    [0065] An injectable aqueous 1% (w/v) human serum transferrin solution is prepared in sterile water for injections. Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to 1% transferrin to final concentration 5.6 mM, followed by brief stirring. To the 1% transferrin solution containing 5.6 mM DTC is added sterile copper (ii) chloride (1 M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0066] Resulting solution contain 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper-transferrin nanoparticles. This reaction results into nanoparticles which were examined by DLS showing most probable average size of approx. 60nm. Resulting particles show low zeta potential (ζ) -21,5 mV measured by electrochemical impedance spectroscopy pointing at good stability in solution.

    EXAMPLE 7



    [0067] Preparation of dispersion of nanoparticles from 1% human serum immunoglobulin (IgG) and diethyldithiocarbamate and copper chloride salt.

    Procedure:



    [0068] An injectable aqueous 1% (w/v) immunoglobulin solution (human gamma-globulin fraction) is prepared in sterile water for injections. Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to 1% transferrin to final concentration 5.6 mM, followed by brief stirring. To the 1% immunoglobulin solution containing 5.6 mM DTC is added sterile copper (ii) chloride (1 M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0069] Resulting solution contain nearly 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper-immunoglobulin nanoparticles. This reaction results into nanoparticles which were examined by DLS showing most probable average size of approx.20nm (see Figure 8). Resulting particles show zeta potential (ζ) -1,39 mV as measured by electrochemical impedance spectroscopy pointing at relatively poor stability in solution.

    EXAMPLE 8



    [0070] Preparation of dispersion of nanoparticles from a mixture of 1% human serum transferrin and 1% human serum albumin and diethyldithiocarbamate and copper chloride salt.

    Procedure:



    [0071] An injectable aqueous 1% (w/v) human serum transferrin solution is mixed with an injectable aqueous 1% (w/v) HSA solution. Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to the mixture of proteins to final concentration 5.6 mM, followed by brief stirring. To the protein solution containing 5.6 mM DTC is added sterile copper (ii) chloride (1 M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0072] Resulting solution contain 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper-transferrin/HSA nanoparticles. This reaction results into nanoparticles which were examined by DLS showing most probable average size of approx.30nm (see Figure 9).

    EXAMPLE 9



    [0073] Preparation of dispersion of nanoparticles from a mixture of 1% human serum albumin and 1% (w/v) immunoglobulin solution and diethyldithiocarbamate and copper chloride salt.

    Experimental setup



    [0074] An injectable aqueous 1% (w/v) human serum albumin solution is mixed with an injectable aqueous 1% (w/v) immunoglobulin solution. Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to the mixture of proteins to final concentration 5.6 mM, followed by brief stirring. To the protein solution containing 5.6 mM DTC is added sterile copper (ii) chloride (1 M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0075] Resulting solution contains 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper-albumin/immunoglobulin nanoparticles. This reaction results into nanoparticles which were examined by DLS showing most probable average size of approx.120nm (see Figure 10).

    EXAMPLE 10



    [0076] Preparation of dispersion of nanoparticles from 0.2% mouse serum albumin (MSA) and diethyldithiocarbamate and radioactive copper (Cu64) acetate salt for tumour imaging and biodistribution.

    Experimental setup



    [0077] Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to 1% MSA to final concentration 5.6 mM, followed by brief stirring. To the 1% MSA solution containing 5.6 mM DTC is added a mixture of sterile copper (Cu64) (ii) acetate and non-active copper chloride (100 mM concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0078] Resulting solution contains 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper (Cu64)-MSA nanoparticles. The nanoparticles resulting from this reaction were further analysed in-vivo by retro-orbital injection (dose 1 mg/kg corresponding to activity 33 MBq) into mice bearing subcutaneous MDA-MB-231 tumour xenograft followed by the PET analysis in multiple time points after the application using Albira PET/SPECT/CT imaging system (Bruker Biospin Corporation, Woodbridge, CT, USA). Measured PET signal shows accumulation within the tumour mass (Figure 13).

    EXAMPLE 11



    [0079] Preparation of dispersion of nanoparticles from 0.2% human serum transferin (HST) and diethyldithiocarbamate and radioactive copper (Cu64) acetate salt for tumour imaging and biodistribution.

    Experimental setup



    [0080] Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to 1% HST to final concentration 5.6 mM, followed by brief stirring. To the 1% HST solution containing 5.6 mM DTC is added a mixture of sterile copper (Cu64) (ii) acetate and non-active copper chloride (100 mM concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0081] Resulting solution contains 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper (Cu64)-HST nanoparticles. The nanoparticles resulting from this reaction were further analysed in-vivo by retro-orbital injection (dose 1 mg/kg corresponding to activity 29 MBq) into mice bearing subcutaneous MDA-MB-231 tumour xenograft followed by the PET analysis in multiple time points after the application using Albira PET/SPECT/CT imaging system (Bruker Biospin Corporation, Woodbridge, CT, USA). PET signal shows accumulation within the tumour mass (Figure 13).

    EXAMPLE 12



    [0082] Preparation of dispersion of nanoparticles from 0.2% mouse serum albumin (MSA) and diethyldithiocarbamate and copper chloride salt.

    Experimental setup



    [0083] An injectable aqueous 20% (w/v) MSA solution (pH=6.9±0.5) is diluted to 0.2% (w/v) with sterile water for injections. Sterile solution of diethyldithiocarbamate sodium salt (DTC) solubilised in water of a concentration 280 mM is added to 0.2% MSA to final concentration 5.6 mM, followed by brief stirring. To the 0.2% MSA solution containing 5.6 mM DTC is added sterile copper (ii) chloride (1 M concentration in water) to final concentration 2.8 mM, followed by brief stirring.

    Results



    [0084] Resulting solution contains 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper-MSA nanoparticles. The nanoparticles resulting from this reaction were further analysed by various mouse in-vivo experiments including pharmacokinetics of CuET by HPLC/MS (see figure 12) and antitumor activity (see figures 14-15).

    EXAMPLE 13



    [0085] Preparation of dispersion of nanoparticles from 0.2% human serum albumin (HSA) and copper chloride salt and diethyldithiocarbamate.

    Experimental setup



    [0086] An injectable aqueous 20% (w/v) MSA solution (pH=6.9±0.5) is diluted to 0.2% (w/v) with sterile water for injections. Sterile solution of sterile copper (ii) chloride solubilised in water of a concentration 1 M is added to 0.2% HSA to final concentration 2.8 mM, followed by brief stirring. To the 0.2% HSA solution containing 2.8 mM copper (ii) chloride is added sterile diethyldithiocarbamate sodium salt (280 mM concentration in water) to final concentration 5.6 mM, followed by brief stirring.

    Results



    [0087] Resulting solution contains 2.8 mM (1 mg/ml) of diethyldithiocarbamate-copper-HSA nanoparticles.


    Claims

    1. A nanoparticulate form of dithiocarbamate-metal complex and at least one blood protein, wherein at least 50 % of the nanoparticles have the size within the range of 2-200 nm,
    wherein the said nanoparticulate form of dithiocarbamate-metal complex and at least one blood protein is prepared by combining a first component selected from dithiocarbamate and metal salt with at least one blood protein in an aqueous solvent, and subsequently adding a second component selected from dithiocarbamate and metal salt, wherein

    if the first component is dithiocarbamate, then the second component is metal salt; and

    if the first component is metal salt, then the second component is dithiocarbamate;

    wherein the metal is selected from copper, zinc, silver and gold, and
    wherein the blood proteins are blood serum proteins selected from the group comprising albumin, transferrin, immunoglobulin, and mixtures comprising these proteins, including whole blood, blood plasma and blood serum.
     
    2. The nanoparticulate form according to claim 1, wherein the metal is copper.
     
    3. The nanoparticulate form according to claim 1, wherein the metal is selected from 63Cu, 65Cu, 64Cu and mixtures thereof.
     
    4. The nanoparticulate form according to claim 1, wherein the dithiocarbamate has a formula (R1)(R2)N-CS2, wherein R1 and R2 are the same or different and are independently selected from C1-C8 alkyl, C2-C8 alkenyl, C3-C10 cycloalkyl, C6-C14 aryl, C4-C14 heteroaryl containing at least one heteroatom selected from O, S, N, C3-C10 heterocyclyl containing at least one heteroatom selected from O, S, N; or R1 and R2 together with the nitrogen atom on which they are bound form a heterocycle, wherein -R1-R2- is a C2-C6 alkylene or a C2-C6 alkenylene, wherein optionally 1-2 carbon atoms may be replaced by heteroatoms selected from O, S, NH; and
    the moieties forming R1 and R2 may be unsubstituted or further substituted by at least one substituent selected from C1-C4 alkyl, hydroxy, mercapto, C1-C4 alkoxy, C1-C4 alkylthio, halogen, phenyl, benzyl, keto group, carboxyl group, C1-C4 alkyloxycarbonyl.
     
    5. The nanoparticulate form according to claim 4, wherein R1 and R2 are independently selected from C1-C6 (or C1-C4) alkyl, C2-C6 (or C1-C4) alkenyl, C3-C6 cycloalkyl, phenyl; or R1 and R2 together with the nitrogen atom on which they are bound form a heterocycle, wherein -R1-R2- is a C2-C6 alkylene or a C2-C6 alkenylene.
     
    6. A process for preparation of the nanoparticulate form according to claim 1, wherein the nanoparticulate form is prepared by combining a first component selected from dithiocarbamate and metal salt with at least one blood protein in an aqueous solvent, and subsequently adding a second component selected from dithiocarbamate and metal salt, whereas if the first component is dithiocarbamate, then the second component is metal salt; and if the first component is metal salt, then the second component is dithiocarbamate.
     
    7. The process according to claim 6, comprising the steps of:

    (a) solubilising at least one blood protein in an aqueous solvent to a concentration in the range from 0.01% (w/v) to saturated solution, or providing whole blood or blood plasma or blood serum of a patient;

    (b) adding at least one dithiocarbamate dissolved in an aqueous solvent in the dithiocarbamate in the range from 1 uM to 100 mM, preferably 1 to 10 mM;

    (c) adding a metal salt solution in an aqueous solvent, having the metal salt concentration in the range from 1 uM to 100 M, preferably 1 to 10 mM,

    wherein the steps are carried out in the sequence (a), (b), (c) or in the sequence (a), (c), (b).
     
    8. The process according to claim 6 or 7, wherein molar ratio of metal ions:dithiocarbamate ions is 1:5 to 5:1.
     
    9. The process according to any one of claims 6 to 8, wherein the aqueous solvent is water or water-based buffer, such as phosphate, citrate, acetate, Tris, HEPES, saline; preferably, the aqueous solvent is sterile.
     
    10. The nanoparticulate form according to any one of claims 1 to 5 for use as therapeutic and/or diagnostic agent.
     
    11. The nanoparticulate form according to any one of claims 1 to 5 for use in therapy selected from chemotherapy of cancer, radiotherapy of cancer and/or in visualization of tumors.
     
    12. The nanoparticulate form for use according to claim 10 or 11, wherein the nanoparticulate form is administered parenterally.
     
    13. The nanoparticulate form according to any one of claims 1 to 5, optionally for use according to claim 10, wherein the nanoparticulate form comprises immunoglobulin or immunoglobulin-containing blood protein mixtures as blood proteins, and further harbours antibodies or antibody fragments against tumour antigens.
     


    Ansprüche

    1. Eine nanopartikuläre Form eines Dithiocarbamat-Metall-Komplexes und mindestens eines Blutproteins, wobei mindestens 50% der Nanopartikel eine Größe im Bereich von 2 bis 200 nm haben; wobei die nanopartikuläre Form des Dithiocarbamat-Metall-Komplexes und mindestens eines Blutproteins erhältlich ist durch das Verfahren, indem eine erste Komponente, ausgewählt aus Dithiocarbamat und Metallsalz, mit mindestens einem Blutprotein in einem wässrigen Lösungsmittel kombiniert wird und anschließend wird eine zweite Komponente ausgewählt aus Dithiocarbamat und Metallsalz hinzugefügt, worin
    wenn die erste Komponente Dithiocarbamat ist, ist die zweite Komponente Metallsalz, und
    wenn die erste Komponente Metallsalz ist, ist die zweite Komponente Dithiocarbamat,
    wobei das Metall ausgewählt ist aus Kupfer, Zink, Silber und Gold und
    wobei die Blutproteine Blutserumproteine sind, ausgewählt aus der Gruppe, die umfasst Albumin, Transferrin, Immunglobulin und Gemische umfassend diese Proteine, einschließlich Vollblut, Blutplasma und Blutserum.
     
    2. Nanopartikuläre Form nach Anspruch 1, wobei das Metall Kupfer ist.
     
    3. Nanopartikuläre Form nach Anspruch 1, wobei das Metall ausgewählt ist aus 63Cu, 65Cu, 64Cu und Gemischen davon.
     
    4. Nanopartikuläre Form nach Anspruch 1, wobei das Dithiocarbamat eine Formel (R1)(R2)N-CS2- aufweist, wobei R1 und R2 gleich oder verschieden sind und unabhängig voneinander ausgewählt sind aus C1-C8-Alkyl, C2-C8-Alkenyl, C3-C10-Cycloalkyl, C6-C14-Aryl, C4-C14-Heteroaryl, das mindestens ein aus O, S, N ausgewähltes Heteroatom enthält, C3-C10-Heterocyclyl, das mindestens ein aus O, S, N ausgewähltes Heteroatom enthält; oder R1 und R2 bilden zusammen mit dem Stickstoffatom, an das sie gebunden sind, einen Heterocyclus, wobei -R1-R2- ein C2-C6-Alkylen oder ein C2-C6-Alkenylen ist, wobei gegebenenfalls 1-2 Kohlenstoffatome durch aus O, S, NH ausgewählte Heteroatome ersetzt werden können; und
    die Einheiten, die R1 und R2 bilden, können unsubstituiert oder weiter substituiert sein durch mindestens einen Substituenten, ausgewählt aus C1-C4-Alkyl, Hydroxy, Mercapto, C1-C4-Alkoxy, C1-C4-Alkylthio, Halogen, Phenyl, Benzyl, Ketogruppe, Carboxylgruppe, C1-C4-Alkyloxycarbonyl.
     
    5. Nanopartikuläre Form nach Anspruch 4, wobei R1 und R2 unabhängig voneinander ausgewählt sind aus C1-C6 (oder C1-C4)-Alkyl, C2-C6 (oder C2-C4)-Alkenyl, C3-C6-Cycloalkyl, Phenyl; oder R1 und R2 bilden zusammen mit dem Stickstoffatom, an das sie gebunden sind, einen Heterocyclus, wobei - R1-R2- ein C2-C6-Alkylen oder ein C2-C6-Alkenylen ist.
     
    6. Verfahren zur Herstellung der nanopartikulären Form nach Anspruch 1, wobei die nanopartikuläre Form hergestellt wird so dass eine erste aus Dithiocarbamat und Metallsalz ausgewählte Komponente mit mindestens einem Blutprotein in einem wässrigen Lösungsmittel kombiniert ist und anschließend eine zweite aus Dithiocarbamat und Metallsalz ausgewählte Komponente zugegeben wird, wobei wenn die erste Komponente Dithiocarbamat ist, die zweite Komponente Metallsalz ist; und wenn die erste Komponente Metallsalz ist, dann ist die zweite Komponente Dithiocarbamat.
     
    7. Verfahren nach Anspruch 6, umfassend die Schritte:

    (a) Lösen mindestens eines Blutproteins in einem wässrigen Lösungsmittel auf eine Konzentration im Bereich von 0,01% (Gew./Vol.) bis zu gesättigter Lösung, oder Bereitstellen von Vollblut oder Blutplasma oder Blutserum eines Patienten;

    (b) Zugabe von mindestens einem Dithiocarbamat, gelöst in einem wässrigen Lösungsmittel, mit einer Dithiocarbamatkonzentration im Bereich von 1 uM bis 100 mM, vorzugsweise 1 bis 10 mM;

    (c) Hinzufügen einer Metallsalzlösung in einem wässrigen Lösungsmittel mit einer Metallsalzkonzentration im Bereich von 1 uM bis 100 M, vorzugsweise 1 bis 10 mM,

    wobei die Schritte in der Sequenz (a), (b), (c) oder in der Sequenz (a), (c), (b) ausgeführt werden.
     
    8. Verfahren nach Anspruch 6 oder 7, wobei das Molarverhältnis von Metallionen Dithiocarbamationen 1: 5 bis 5: 1 beträgt.
     
    9. Verfahren nach einem der Ansprüche 6 bis 8, wobei das wässrige Lösungsmittel Wasser oder Puffer auf Wasserbasis ist, wie Phosphat-, Citrat-, Acetat-, Tris-Puffer, HEPES, Salzlösung; vorzugsweise ist das wässrige Lösungsmittel steril.
     
    10. Nanopartikuläre Form nach einem der Ansprüche 1 bis 5 zur Verwendung als therapeutisches und/oder diagnostisches Mittel.
     
    11. Nanopartikuläre Form nach einem der Ansprüche 1 bis 5 zur Verwendung in einer Therapie, ausgewählt aus einer Chemotherapie von Krebs, einer Strahlentherapie von Krebs und/oder zur Visualisierung von Tumoren.
     
    12. Nanopartikuläre Form zur Verwendung nach Anspruch 10 oder 11, wobei die nanopartikuläre Form parenteral verabreicht wird.
     
    13. Nanopartikelform nach einem der Ansprüche 1 bis 5, gegebenenfalls zur Verwendung nach Anspruch 10, wobei die nanopartikuläre Form Immunglobulin oder Immunglobulin enthaltende Blutproteinmischungen als Blutproteine umfasst und ferner Antikörper oder Antikörperfragmente gegen Tumorantigene enthält.
     


    Revendications

    1. Forme nanoparticulaire d'un complexe dithiocarbamate-métal et d'au moins une protéine sanguine, dans laquelle au moins 50% des nanoparticules ont une taille comprise entre 2 et 200 nm,
    où ladite forme nanoparticulaire de complexe dithiocarbamate-métal et au moins une protéine sanguine est préparée en combinant un premier composant choisi parmi un dithiocarbamate et un sel métallique avec au moins une protéine sanguine dans un solvant aqueux, puis en ajoutant un second composant choisi parmi un dithiocarbamate et un sel métallique, où
    si le premier composant est le dithiocarbamate, alors le second composant est le sel métallique; et
    si le premier composant est le sel métallique, alors le second composant est le dithiocarbamate;
    où le métal est choisi parmi le cuivre, le zinc, l'argent et l'or, et
    où les protéines sanguines sont des protéines de sérum sanguin choisies dans le groupe comprenant l'albumine, la transferrine, l'immunoglobuline et des mélanges comprenant ces protéines, y compris le sang total, le plasma sanguin et le sérum sanguin.
     
    2. Forme nanoparticulaire selon la revendication 1, dans laquelle le métal est le cuivre.
     
    3. Forme nanoparticulaire selon la revendication 1, dans laquelle le métal est choisi parmi 63Cu, 65Cu, 64Cu et leurs mélanges.
     
    4. Forme nanoparticulaire selon la revendication 1, dans laquelle le dithiocarbamate a une formule (R1)(R2)N-CS2-, dans laquelle R1 et R2 sont identiques ou différents et sont indépendamment choisis parmi alkyle en C1-C8, alcényle en C2-C8, cycloalkyle en C3-C10, aryle en C6-C14, hétéroaryle en C4-C14 contenant au moins un hétéroatome choisi parmi O, S, N, hétérocyclyle en C3-C10 contenant au moins un hétéroatome choisi parmi O, S, N; ou R1 et R2 conjointement avec l'atome d'azote sur lequel ils sont liés forment un hétérocycle, dans lequel -R1-R2- est un alkylène en C2-C6 ou un alcénylène en C2-C6, où éventuellement 1-2 atomes de carbone peuvent être remplacés par des hétéroatomes choisis de O, S, NH; et
    les groupements formant R1 et R2 peuvent être non substitués ou substitués par au moins un substituant choisi parmi alkyle en C1-C4, hydroxy, mercapto, alcoxy en C1-C4, alkylthio en C1-C4, halogène, phényle, benzyle, groupe céto, groupe carboxyle, alkyloxycarbonyle en C1-C4.
     
    5. Forme nanoparticulaire selon la revendication 4, dans laquelle R1 et R2 sont choisis indépendamment parmi un alkyle en C1-C6 (ou en C1-C4), un alcényle en C2-C6 (ou en C2-C4), un cycloalkyle en C3-C6, un phényle; ou R1 et R2 conjointement avec l'atome d'azote sur lequel ils sont liés forment un hétérocycle, dans lequel -R1-R2- est un alkylène en C2-C6 ou un alcénylène en C2-C6.
     
    6. Procédé de préparation de la forme nanoparticulaire selon la revendication 1, dans lequel la forme nanoparticulaire est préparée en combinant un premier composant choisi parmi un dithiocarbamate et un sel métallique avec au moins une protéine sanguine dans un solvant aqueux, puis en ajoutant un deuxième composant choisi parmi un dithiocarbamate et un sel métallique, tandis que si le premier composant est le dithiocarbamate, alors le deuxième composant est le sel métallique; et si le premier composant est le sel métallique, alors le second composant est le dithiocarbamate.
     
    7. Procédé selon la revendication 6, comprenant les étapes de:

    (a) solubiliser au moins une protéine sanguine dans un solvant aqueux à une concentration dans la gamme de 0,01% (p / v) à une solution saturée, ou fournir du sang total ou du plasma sanguin ou du sérum sanguin d'un patient;

    (b) ajouter au moins un dithiocarbamate dissous dans un solvant aqueux, avec le dithiocarbamate dans la plage de 1 uM à 100 mM, de préférence de 1 à 10 mM;

    (c) ajouter une solution de sel métallique dans un solvant aqueux, ayant la concentration de sel métallique dans la gamme de 1 uM à 100 M, de préférence de 1 à 10 mM,

    où les étapes sont effectuées dans la séquence (a), (b), (c) ou dans la séquence (a), (c), (b).
     
    8. Procédé selon la revendication 6 ou 7, dans lequel le rapport molaire des ions métalliques: ions dithiocarbamate est de 1: 5 à 5: 1.
     
    9. Procédé selon l'une quelconque des revendications 6 à 8, dans lequel le solvant aqueux est l'eau ou un tampon à base d'eau, tel que un tampon phosphate, citrate, acétate, Tris, HEPES, solution saline; de préférence, le solvant aqueux est stérile.
     
    10. Forme nanoparticulaire selon l'une quelconque des revendications 1 à 5 pour utilisation comme agent thérapeutique et/ou diagnostique.
     
    11. Forme nanoparticulaire selon l'une quelconque des revendications 1 à 5 pour utilisation thérapeutique choisie parmi la chimiothérapie du cancer, la radiothérapie du cancer et/ou pour utilisation dans la visualisation de tumeurs.
     
    12. Forme nanoparticulaire pour utilisation selon la revendication 10 ou 11, où la forme nanoparticulaire est administrée par voie parentérale.
     
    13. Forme nanoparticulaire selon l'une quelconque des revendications 1 à 5, éventuellement destinée pour utilisation selon la revendication 10, où la forme nanoparticulaire comprend des immunoglobulines ou des mélanges de protéines sanguines contenant des immunoglobulines en tant que protéines sanguines, et contient en outre des anticorps ou des fragments d'anticorps contre des antigènes tumoraux.
     




    Drawing



































    Cited references

    REFERENCES CITED IN THE DESCRIPTION



    This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

    Patent documents cited in the description