Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Molecules ; 29(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474543

ABSTRACT

Copper-organic compounds have gained momentum as potent antitumor drug candidates largely due to their ability to generate an oxidative burst upon the transition of Cu2+ to Cu1+ triggered by the exogenous-reducing agents. We have reported the differential potencies of a series of Cu(II)-organic complexes that produce reactive oxygen species (ROS) and cell death after incubation with N-acetylcysteine (NAC). To get insight into the structural prerequisites for optimization of the organic ligands, we herein investigated the electrochemical properties and the cytotoxicity of Cu(II) complexes with pyridylmethylenethiohydantoins, pyridylbenzothiazole, pyridylbenzimidazole, thiosemicarbazones and porphyrins. We demonstrate that the ability of the complexes to kill cells in combination with NAC is determined by the potential of the Cu+2 → Cu+1 redox transition rather than by the spatial structure of the organic ligand. For cell sensitization to the copper-organic complex, the electrochemical potential of the metal reduction should be lower than the oxidation potential of the reducing agent. Generally, the structural optimization of copper-organic complexes for combinations with the reducing agents should include uncharged organic ligands that carry hard electronegative inorganic moieties.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Copper/chemistry , Reducing Agents , Antineoplastic Agents/chemistry , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Coordination Complexes/chemistry , Ligands
2.
Anal Chem ; 96(1): 127-136, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38126724

ABSTRACT

In vitro/in vivo detection of copper ions is a challenging task but one which is important in the development of new approaches to the diagnosis and treatment of cancer and hereditary diseases such as Alzheimer's, Wilson's, etc. In this paper, we present a nanopipette sensor capable of measuring Cu2+ ions with a linear range from 0.1 to 10 µM in vitro and in vivo. Using the gold-modified nanopipette sensor with a copper chelating ligand, we evaluated the accumulation ability of the liposomal form of an anticancer Cu-containing complex at three levels of biological organization. First, we detected Cu2+ ions in a single cell model of human breast adenocarcinoma MCF-7 and in murine melanoma B16 cells. The insertion of the nanoelectrode did not result in leakage of the cell membrane. We then evaluated the distribution of the Cu-complex in MCF-7 tumor spheroids and found that the diffusion-limited accumulation was a function of the depth, typical for 3D culture. Finally, we demonstrated the use of the sensor for Cu2+ ion detection in the brain of an APP/PS1 transgenic mouse model of Alzheimer's disease and tumor-bearing mice in response to injection (2 mg kg-1) of the liposomal form of the anticancer Cu-containing complex. Enhanced stability and selectivity, as well as distinct copper oxidation peaks, confirmed that the developed sensor is a promising tool for testing various types of biological systems. In summary, this research has demonstrated a minimally invasive electrochemical technique with high temporal resolution that can be used for the study of metabolism of copper or copper-based drugs in vitro and in vivo.


Subject(s)
Alzheimer Disease , Neoplasms , Mice , Humans , Animals , Copper , Alzheimer Disease/diagnosis , Ions , Electrochemical Techniques
3.
Nanomedicine (Lond) ; 18(28): 2105-2123, 2023 12.
Article in English | MEDLINE | ID: mdl-38127591

ABSTRACT

Aim: To develop an optimized approach for encapsulating a 2-alkylthioimidazolone-based copper coordination compound within liposomes, which could offer treatment of cancer and bacterial infections by reactive oxygen species generation toxicity mechanisms. Materials & methods: For drug-loaded liposome preparation, lipids and drug mixture in organic solvents was injected into copper salt solution, forming a coordination compound simultaneously embedded in the lipid bilayer. In vitro tests were performed on MCF7 and MDA-MB-231 breast cancer cells. Results: Liposomes had a loading capacity of up to 1.75% (molar drug-to-lipid ratio). In vitro tests showed increased viability and accumulation of the liposomal formulation compared with free drug as well as lack of cytotoxicity in hepatocytes. Conclusion: This optimized technique for encapsulating large copper complexes in liposomes could be used to improve their delivery and better treat cancer and bacterial infections.


This work introduces a new technique for copper-containing drugs encapsulation in a drug-delivery system. The drug, a promising copper compound, is embedded in lipid nanovesicles ­ tiny fat particles ­ for intravenous injection. In addition to chemical characterization of the obtained drug form, tests on cancer cells showed a noticeable effect, whereas healthy cell types were not harmed. Copper possesses not only anticancer effects but also antimicrobial properties, which are also shown by the drug form, and a test of combined suppression of cancer cell lines and bacteria was successful. Hence, the obtained drug form has the potential for dual treatment of cancer and bacterial infections.


Subject(s)
Bacterial Infections , Breast Neoplasms , Humans , Female , Liposomes , Copper/therapeutic use , Cell Line, Tumor , Breast Neoplasms/drug therapy
4.
Int J Mol Sci ; 24(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298101

ABSTRACT

Copper-64 (T1/2 = 12.7 h) is a positron and beta-emitting isotope, with decay characteristics suitable for both positron emission tomography (PET) imaging and radiotherapy of cancer. Copper-67 (T1/2 = 61.8 h) is a beta and gamma emitter, appropriate for radiotherapy ß-energy and with a half-life suitable for single-photon emission computed tomography (SPECT) imaging. The chemical identities of 64Cu and 67Cu isotopes allow for convenient use of the same chelating molecules for sequential PET imaging and radiotherapy. A recent breakthrough in 67Cu production opened previously unavailable opportunities for a reliable source of 67Cu with high specific activity and purity. These new opportunities have reignited interest in the use of copper-containing radiopharmaceuticals for the therapy, diagnosis, and theranostics of various diseases. Herein, we summarize recent (2018-2023) advances in the use of copper-based radiopharmaceuticals for PET, SPECT imaging, radiotherapy, and radioimmunotherapy.


Subject(s)
Neoplasms , Radiopharmaceuticals , Humans , Radiopharmaceuticals/chemistry , Copper , Positron-Emission Tomography/methods , Tomography, Emission-Computed, Single-Photon , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy
5.
ACS Appl Mater Interfaces ; 15(10): 12882-12894, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36854172

ABSTRACT

Controlled photoreduction of Pt(IV) prodrugs is a challenging task due to the possibility of targeted light-controlled activation of anticancer agents without affecting healthy tissues. Also, a conjugation of photosensitizers and clinically used platinum drugs into one Pt(IV) prodrug allows combining photodynamic therapy and chemotherapy approaches into one molecule. Herein, we designed the cisplatin-based Pt(IV) prodrug Riboplatin with tetraacetylriboflavin in the axial position. A novel Pt(IV) prodrug is able to act both as a photodynamic therapy (PDT) agent through the conversion of ground-state 3O2 to excited-state 1O2 and as an agent of photoactivated chemotherapy (PACT) through releasing of cisplatin under gentle blue light irradiation, without the requirement of a reducing agent. The light-induced behavior of Riboplatin was investigated using an electrochemical sensor in MCF-7 tumor spheroids. Photocontrolled cisplatin release and ROS generation were detected electrochemically in real time. This appears to be the first confirmation of simultaneous photoactivated release of anticancer drug cisplatin and ROS from a dual-action Pt(IV) prodrug observed from the inside of living tumor spheroids.


Subject(s)
Antineoplastic Agents , Prodrugs , Cisplatin/pharmacology , Cisplatin/chemistry , Prodrugs/pharmacology , Prodrugs/chemistry , Reactive Oxygen Species , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Platinum/chemistry , Cell Line, Tumor
6.
Inorg Chem ; 61(37): 14705-14717, 2022 Sep 19.
Article in English | MEDLINE | ID: mdl-36047922

ABSTRACT

We report herein a Pt(IV) prodrug with metronidazole in axial positions Pt-Mnz. The nitroaromatic axial ligand was conjugated with a cisplatin scaffold to irreversibly reduce under hypoxic conditions, thereby retaining the Pt(IV) prodrug in the area of hypoxia. X-ray near-edge adsorption spectroscopy (XANES) on dried drug-preincubated tumor cell samples revealed a gradual release of cisplatin from the Pt-Mnz prodrug instead of rapid intracellular degradation. The ability of the prodrug to penetrate into three-dimensional (3D) spheroid cellular cultures was evaluated by a novel electrochemical assay via a platinum-coated carbon nanoelectrode, capable of single-cell measurements. Using a unique technique of electrochemical measurements in single tumor spheroids, we were able to both detect the real-time response of the axial ligand to hypoxia and establish the depth of penetration of the drug into the tumor model.


Subject(s)
Antineoplastic Agents , Prodrugs , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbon , Cell Line, Tumor , Cisplatin/chemistry , Humans , Hypoxia , Ligands , Metronidazole/pharmacology , Platinum/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology
7.
J Med Chem ; 65(12): 8227-8244, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35675651

ABSTRACT

We report herein the design, synthesis, and biological investigation of a series of novel Pt(IV) prodrugs with non-steroidal anti-inflammatory drugs naproxen, diclofenac, and flurbiprofen, as well as these with stearic acid in the axial position. Six Pt(IV) prodrugs 5-10 were designed, which showed superior antiproliferative activity compared to cisplatin as well as an ability to overcome tumor cell line resistance to cisplatin. By tuning the drug lipophilicity via variation of the axial ligands, the most potent Pt(IV) prodrug 7 was obtained, with an enhanced cellular accumulation of up to 153-fold that of cisplatin and nanomolar cytotoxicity both in 2D and 3D cell cultures. Pt2+ species were detected at different depths of MCF-7 spheroids after incubation with Pt(IV) prodrugs using a Pt-coated carbon nanoelectrode. Cisplatin accumulation in vivo in the murine mammary EMT6 tumor tissue of BALB/c mice after Pt(IV) prodrug injection was proved electrochemically as well. The drug tolerance study on BALB/c mice showed good tolerance of 7 in doses up to 8 mg/kg.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Antineoplastic Agents , Platinum Compounds , Prodrugs , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Drug Design , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Platinum Compounds/pharmacology , Prodrugs/pharmacology
8.
Anal Chem ; 94(12): 4901-4905, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35285614

ABSTRACT

The biodistribution of chemotherapy compounds within tumor tissue is one of the main challenges in the development of antineoplastic drugs, and techniques for simple, inexpensive, sensitive, and selective detection of various analytes in tumors are of great importance. In this paper we propose the use of platinized carbon nanoelectrodes (PtNEs) for the electrochemical detection of platinum-based drugs in various biological models, including single cells and tumor spheroids in vitro and inside solid tumors in vivo. We have demonstrated the quantitative direct detection of Pt(II) in breast adenocarcinoma MCF-7 cells treated with cisplatin and a cisplatin-based DNP prodrug. To realize the potential of this technique in advanced tumor models, we measured Pt(II) in 3D tumor spheroids in vitro and in tumor-bearing mice in vivo. The concentration gradient of Pt(II) species correlated with the distance from the sample surface in MCF-7 tumor spheroids. We then performed the detection of Pt(II) species in tumor-bearing mice treated intravenously with cisplatin and DNP. We found that there was deeper penetration of DNP in comparison to cisplatin. This research demonstrates a minimally invasive, real-time electrochemical technique for the study of platinum-based drugs.


Subject(s)
Antineoplastic Agents , Prodrugs , Animals , Cisplatin/chemistry , Cisplatin/pharmacology , Humans , MCF-7 Cells , Mice , Prodrugs/chemistry , Tissue Distribution
9.
Int J Mol Sci ; 22(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34681725

ABSTRACT

Copper-containing agents are promising antitumor pharmaceuticals due to the ability of the metal ion to react with biomolecules. In the current study, we demonstrate that inorganic Cu2+ in the form of oxide nanoparticles (NPs) or salts, as well as Cu ions in the context of organic complexes (oxidation states +1, +1.5 and +2), acquire significant cytotoxic potency (2-3 orders of magnitude determined by IC50 values) in combinations with N-acetylcysteine (NAC), cysteine, or ascorbate. In contrast, other divalent cations (Zn, Fe, Mo, and Co) evoked no cytotoxicity with these combinations. CuO NPs (0.1-1 µg/mL) together with 1 mM NAC triggered the formation of reactive oxygen species (ROS) within 2-6 h concomitantly with perturbation of the plasma membrane and caspase-independent cell death. Furthermore, NAC potently sensitized HCT116 colon carcinoma cells to Cu-organic complexes in which the metal ion coordinated with 5-(2-pyridylmethylene)-2-methylthio-imidazol-4-one or was present in the coordination sphere of the porphyrin macrocycle. The sensitization effect was detectable in a panel of mammalian tumor cell lines including the sublines with the determinants of chemotherapeutic drug resistance. The components of the combination were non-toxic if added separately. Electrochemical studies revealed that Cu cations underwent a stepwise reduction in the presence of NAC or ascorbate. This mechanism explains differential efficacy of individual Cu-organic compounds in cell sensitization depending on the availability of Cu ions for reduction. In the presence of oxygen, Cu+1 complexes can generate a superoxide anion in a Fenton-like reaction Cu+1L + O2 → O2-. + Cu+2L, where L is the organic ligand. Studies on artificial lipid membranes showed that NAC interacted with negatively charged phospholipids, an effect that can facilitate the penetration of CuO NPs across the membranes. Thus, electrochemical modification of Cu ions and subsequent ROS generation, as well as direct interaction with membranes, represent the mechanisms of irreversible membrane damage and cell death in response to metal reduction in inorganic and organic Cu-containing compounds.


Subject(s)
Apoptosis/drug effects , Coordination Complexes/pharmacology , Copper/chemistry , Metal Nanoparticles/toxicity , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Humans , Liposomes/chemistry , Liposomes/metabolism , Membrane Potential, Mitochondrial/drug effects , Metal Nanoparticles/chemistry , Oxidation-Reduction , Superoxides/metabolism
10.
J Med Chem ; 64(8): 4532-4552, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33822606

ABSTRACT

Prostate-specific membrane antigen (PSMA), also known as glutamate carboxypeptidase II (GCPII), is a suitable target for specific delivery of antitumor drugs and diagnostic agents due to its overexpression in prostate cancer cells. In the current work, we describe the design, synthesis, and biological evaluation of novel low-molecular PSMA ligands and conjugates with fluorescent dyes FAM-5, SulfoCy5, and SulfoCy7. In vitro evaluation of synthesized PSMA ligands on the activity of PSMA shows that the addition of aromatic amino acids into a linker structure leads to a significant increase in inhibition. The conjugates of the most potent ligand with FAM-5 as well as SulfoCy5 demonstrated high affinities to PSMA-expressing tumor cells in vitro. In vivo biodistribution in 22Rv1 xenografts in Balb/c nude mice of PSMA-SulfoCy5 and PSMA-SulfoCy7 conjugates with a novel PSMA ligand demonstrated good visualization of PSMA-expressing tumors. Also, the conjugate PSMA-SulfoCy7 demonstrated the absence of any explicit toxicity up to 87.9 mg/kg.


Subject(s)
Antigens, Surface/metabolism , Antineoplastic Agents/metabolism , Fluorescent Dyes/chemistry , Glutamate Carboxypeptidase II/metabolism , Ligands , Animals , Antigens, Surface/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Glutamate Carboxypeptidase II/chemistry , Humans , Male , Mice , Mice, Nude , Optical Imaging , Prostatic Neoplasms/drug therapy , Structure-Activity Relationship , Tissue Distribution , Transplantation, Heterologous
11.
J Med Chem ; 63(21): 13031-13063, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32985193

ABSTRACT

A series of 73 ligands and 73 of their Cu+2 and Cu+1 copper complexes with different geometries, oxidation states of the metal, and redox activities were synthesized and characterized. The aim of the study was to establish the structure-activity relationship within a series of analogues with different substituents at the N(3) position, which govern the redox potentials of the Cu+2/Cu+1 redox couples, ROS generation ability, and intracellular accumulation. Possible cytotoxicity mechanisms, such as DNA damage, DNA intercalation, telomerase inhibition, and apoptosis induction, have been investigated. ROS formation in MCF-7 cells and three-dimensional (3D) spheroids was proven using the Pt-nanoelectrode. Drug accumulation and ROS formation at 40-60 µm spheroid depths were found to be the key factors for the drug efficacy in the 3D tumor model, governed by the Cu+2/Cu+1 redox potential. A nontoxic in vivo single-dose evaluation for two binuclear mixed-valence Cu+1/Cu+2 redox-active coordination compounds, 72k and 61k, was conducted.


Subject(s)
Coordination Complexes/chemistry , Copper/chemistry , Imidazoles/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Coordination Complexes/metabolism , Coordination Complexes/pharmacology , Crystallography, X-Ray , DNA Damage/drug effects , Humans , Ligands , MCF-7 Cells , Models, Biological , Molecular Conformation , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Spheroids, Cellular/drug effects , Structure-Activity Relationship , Telomerase/antagonists & inhibitors , Telomerase/metabolism
12.
Bioconjug Chem ; 30(3): 741-750, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30726065

ABSTRACT

A novel approach to the synthesis of pH-sensitive prodrugs has been proposed: thiourea drug modification. Resulting prodrugs can release the cytotoxic agent and the biologically active 2-thiohydantoin in the acidic environment of tumor cells. The concept of acid-catalyzed cyclization of thioureas to 2-thiohydantoins has been proven using a FRET model. Dual prodrugs of model azidothymidine, cytotoxic doxorubicin, and 2-thiohydantoin albutoin were obtained, which release the corresponding drugs in the acidic environment. The resulting doxorubicin prodrug was tested on prostate cancer cells and showed that the thiourea-modified prodrug is less cytotoxic (average IC50 ranging from 0.5584 to 0.9885 µM) than doxorubicin (IC50 ranging from 0.01258 to 0.02559 µM) in neutral pH 7.6 and has similar toxicity (average IC50 ranging from 0.4970 to 0.7994 µM) to doxorubicin (IC50 ranging from 0.2303 to 0.8110 µM) under mildly acidic conditions of cancer cells. Cellular and nuclear accumulation in PC3 tumor cells of Dox prodrug is much higher than accumulation of free doxorubicin.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Hydrogen-Ion Concentration , Prodrugs/pharmacology , Thiourea/chemistry , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Doxorubicin/chemistry , Fluorescein/chemistry , Fluorescence Resonance Energy Transfer , Humans , Male , Naphthalenes/chemistry , Prodrugs/chemistry , Prostatic Neoplasms/pathology
13.
J Med Chem ; 57(14): 6252-8, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24950478

ABSTRACT

We have synthesized and characterized a panel of new binuclear mixed valence Cu(I,II) complexes containing substituted 2-alkylthio-5-arylmethylene-4H-imidazolin-4-ones with unusual structure. These complexes are shown to be cytotoxic for various cell lines. We have found that these compounds did not intercalate DNA, inhibited number of polymerases (telomerase predominantly), accumulated in the cell nucleus, and caused DNA degradation. Preliminary studies revealed that lead compound inhibited human breast adenocarcinoma growth in mice model.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/chemistry , Neoplasms, Experimental/drug therapy , Organometallic Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , DNA Damage , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HEK293 Cells , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Neoplasms, Experimental/pathology , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...