Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biomater Adv ; 140: 213054, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35964389

ABSTRACT

A series of nanostructured SBA-15-based materials functionalized with the tetraorganotin(IV) metallodrugs Ph3Sn(CH2)nOH (n = 3, 4, 6, 8 and 11) are synthesized and structurally characterized by different techniques used in solid-state chemistry. The cytotoxicity of both the organotin(IV) compounds and the tin-functionalized SBA-15 materials are studied against different cancer cell lines observing that the materials have similar cytotoxic activity in comparison with the free organotin compounds in terms of mass. However, considering that the percentage of active metal compound loaded into material is low, the utilization of mesoporous silica as drug vehicle clearly improves the cytotoxic effectiveness of metal-based drugs against cancer cells. One of the most potent between all tested systems is material SBA-15~Cl|Ph3Sn(CH2)8OH. Its cytotoxicity seems to come from additional mechanisms apart from apoptosis provoking cell reprogram in B16 melanoma into more mature and less aggressive phenotype. Moderated production of ROS/RNS is probably in the background of observed phenomenon. Obtained results are further confirmed in syngeneic mouse model of melanoma in C57BL6 mice. The in vivo results show that SBA-15 do not disturb tumor growth, while both Ph3Sn(CH2)8OH and SBA-15~Cl|Ph3Sn(CH2)8OH significantly decreases tumor volume with an enhancement of the antitumor potential of the tetraorganotin(IV) compound upon immobilization in SBA-15.


Subject(s)
Antineoplastic Agents , Melanoma, Experimental , Organotin Compounds , Animals , Antineoplastic Agents/pharmacology , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Organotin Compounds/pharmacology , Silicon Dioxide/chemistry
3.
Cancer Cell ; 37(1): 37-54.e9, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31883968

ABSTRACT

Cyclin-dependent kinase 7 (CDK7) is a central regulator of the cell cycle and gene transcription. However, little is known about its impact on genomic instability and cancer immunity. Using a selective CDK7 inhibitor, YKL-5-124, we demonstrated that CDK7 inhibition predominately disrupts cell-cycle progression and induces DNA replication stress and genome instability in small cell lung cancer (SCLC) while simultaneously triggering immune-response signaling. These tumor-intrinsic events provoke a robust immune surveillance program elicited by T cells, which is further enhanced by the addition of immune-checkpoint blockade. Combining YKL-5-124 with anti-PD-1 offers significant survival benefit in multiple highly aggressive murine models of SCLC, providing a rationale for new combination regimens consisting of CDK7 inhibitors and immunotherapies.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Genomic Instability , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , Animals , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Chemokine CXCL9/metabolism , DNA Damage , Female , Humans , Immune System , Inflammation , Interferon-gamma/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Male , Mice , Micronucleus Tests , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrroles/pharmacology , Signal Transduction , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/immunology , Tumor Necrosis Factor-alpha/metabolism , Cyclin-Dependent Kinase-Activating Kinase
4.
J Biol Inorg Chem ; 24(2): 223-234, 2019 03.
Article in English | MEDLINE | ID: mdl-30759278

ABSTRACT

Extraordinary progress in medicinal inorganic chemistry in the past few years led to the rational design of novel platinum compounds, as well as nonplatinum metal-based antitumor agents, including organotin compounds, whose activity is not based on unrepairable interaction with DNA. To overcome poor solubility and toxicity problems that limited the application of these compounds numerous delivering systems were used (Lila et al. in Biol Pharm Bull 37:206-211, 2014; Yue and Cao in Curr Cancer Drug Targets 16:480-488, 2016; Duan et al. in WIREs Nanomed Nanobiotechnol 8:776-791, 2016). Regarding high drug loading capacity, mesoporous silica nanoparticles like SBA-15 became more important for targeted drug delivery. In this study, cellular uptake and biological activities responsible for organotin(IV) compound Ph3Sn(CH2)6OH (Sn6) grafted into (3-chloropropyl)triethoxysilane functionalized SBA-15 (SBA-15p → SBA-15p|Sn6) were evaluated in human melanoma A375 cell line. Moreover, the influence of SBA-15p grafted with organotin(IV) compound on the stemness of A375 cell was tested. Given the fact that SBA-15p|Sn6 nanoparticles are nonspherical and relatively large, their internalization efficiently started even after 15 min with stable adhesion to the cell membrane. After only 2 h of incubation of A375 cells with SBA-15p|Sn6 passive fluid-phase uptake and macropinocytosis were observed. Inside of the cell, treatment with SBA-15p loaded with Sn6 promoted caspase-dependent apoptosis in parallel with senescence development. The subpopulation of cells expressing Schwann-like phenotype arose upon the treatment, while the signaling pathway responsible for maintenance of pluripotency and invasiveness, Wnt, Notch1, and Oct3/4 were modulated towards less aggressive signature. In summary, SBA-15p enhances the efficacy of free Sn6 compound through efficient uptake and well profiled intracellular response followed with decreased stem characteristics of highly invasive A375 melanoma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Organotin Compounds/pharmacology , Silicon Dioxide/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Melanoma/pathology , Organotin Compounds/chemistry , Particle Size , Phenotype , Silicon Dioxide/chemistry , Surface Properties
5.
Oncogene ; 38(13): 2380-2393, 2019 03.
Article in English | MEDLINE | ID: mdl-30518877

ABSTRACT

The backbone of ovarian cancer treatment is platinum-based chemotherapy and aggressive surgical debulking. New therapeutic approaches using immunotherapy via immune checkpoint blockade, which have demonstrated clinical efficacy in other tumor types, have been less promising in ovarian cancer. To increase their clinical efficacy, checkpoint inhibitors are now being tested in clinical trials in combination with chemotherapy. Here, we evaluated the impact of cisplatin on tumor immunogenicity and its in vivo roles when used alone or in combination with anti-PD-L1, in two novel murine ovarian cancer cell models. The 2F8 and its platinum-resistant derivative 2F8cis model, display distinct inflammatory profiles and chemotherapy sensitivities, and mirror the primary and recurrent human disease, respectively. Acute and chronic exposure to cisplatin enhances tumor immunogenicity by increasing calreticulin, MHC class I, antigen presentation and T-cell infiltration. Cisplatin also upregulates PD-L1 expression in vitro and in vivo, demonstrating a dual, paradoxical immune modulatory effect and supporting the rationale for combination with immune checkpoint blockade. One of the pathways activated by cisplatin treatment is the cGAS/STING pathway. Chronic cisplatin treatment led to upregulation of cGAS and STING proteins in 2F8cis compared to parental 2F8 cells, while acute exposure to cisplatin further increases cGAS and STING levels in both 2F8 and 2F8cis cells. Overexpression of cGAS/STING modifies tumor immunogenicity by upregulating PD-L1, MHC I and calreticulin in tumor cells. Anti-PD-L1 alone in a platinum-sensitive model or with cisplatin in a platinum-resistant model increases survival. These studies have high translational potential in ovarian cancer.


Subject(s)
Carcinoma, Ovarian Epithelial/immunology , Cisplatin/pharmacology , Immune System/drug effects , Immunomodulation/drug effects , Ovarian Neoplasms/immunology , Animals , Antibodies, Monoclonal/administration & dosage , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/therapy , Cell Line, Tumor , Cisplatin/administration & dosage , Combined Modality Therapy , Disease Models, Animal , Female , HEK293 Cells , Humans , Immunotherapy , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Transgenic , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Pathol Oncol Res ; 23(2): 335-343, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27617337

ABSTRACT

Increased circulating alpha-1-antitrypsin (AAT) correlates with cancer stage/aggressiveness, but its role in cancer biology is unclear. We revealed antagonistic effect of AAT to cisplatin-induced cytotoxicity in prostate (PC3) and melanoma (A375) cancer cell lines. Moreover, AAT abrogated cytotoxicity of MEK inhibitor U0126 in PC3 cell line. Weaker antagonistic effect of AAT on cytotoxicity of PI3/Akt and NF-kB inhibitors was also observed. In addition, cisplatin increased AAT gene expression in transfected PC3 cells. However, AAT derived from transfected PC3 cells did not antagonize cisplatin-induced cytotoxicity. In conclusion, these results suggest possible association between high circulating AAT and cisplatin resistance.


Subject(s)
Cisplatin/pharmacology , Melanoma/drug therapy , Melanoma/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , alpha 1-Antitrypsin/metabolism , Cell Line, Tumor , Humans , Male , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
7.
Anticancer Agents Med Chem ; 16(11): 1455-1460, 2016.
Article in English | MEDLINE | ID: mdl-26510901

ABSTRACT

In vitro studies with the ruthenium(II) and analogous iridium(III) complexes [Ru(η6- p-cymene)Cl2{Ph2PCH2CH2CH2S(O)xPh-κP}], [Ru(η6-p-cymene)Cl{Ph2PCH2CH2CH2S(O)xPh- κP,κS}][PF6] (1-4), [Ir(η5-C5Me5)Cl2{Ph2PCH2CH2CH2S(O)xPh-κP}] and [Ir(η5-C5Me5)Cl{Ph2 PCH2CH2CH2S(O)xPh-κP,κS}][PF6] (5-8; x = 0, 1) revealed the high selectivity toward the 8505C, A253, MCF-7, SW480 and 518A2 cancer cell lines. Thus, the cationic ruthenium complex 4 proved to be the most selective one. In case of the neutral and cationic ruthenium complexes 1-4 the caspase-dependent apoptotic cell death was proven as the main cause of the drug's tumoricidal action on 8505C cell line.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Iridium/pharmacology , Ruthenium/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Iridium/chemistry , Molecular Structure , Ruthenium/chemistry , Structure-Activity Relationship
8.
Leuk Res ; 39(10): 1088-95, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26220866

ABSTRACT

Covalent attachment of NO to the first approved HIV protease inhibitor Saquinavir (Saq-NO) expands the therapeutic potential of the original drug. Apart from retained antiviral activity, the modified drug exerts strong antitumor effects and lower toxicity. In the present study, we have evaluated the sensitivity of different hematological malignancies to Saq-NO. Saq-NO efficiently diminished the viability of Jurkat, Raji, HL-60 and K562 cells. While Jurkat and Raji cells (established from pediatric patients) displayed abrogated proliferative potential, HL-60 and K652 cells (originated from adults) exposed to Saq-NO treatment underwent caspase dependent apoptosis. In addition, similar sensitivity to Saq-NO was observed in mononuclear blood cells obtained from pediatric patients with acute lymphoblastic leukemia (ALL) and adult patients with acute myeloid leukemia (AML). Western blot analysis indicated p70S6 kinase as a possible intracellular target of Saq-NO action. Moreover, the addition of a NO moiety to Lopinavir resulted in improved antitumor potential as compared to the parental compound, suggesting that NO-derived HIV protease inhibitors are a potential new source of anticancer drugs with unique mode of action.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Ribosomal Protein S6 Kinases, 70-kDa/drug effects , Saquinavir/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , HIV Protease Inhibitors/pharmacology , HL-60 Cells , Humans , Inhibitory Concentration 50 , Leukemia, Myeloid, Acute/enzymology , Nitric Oxide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology
9.
Eur J Pharmacol ; 760: 136-44, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-25912798

ABSTRACT

(O,O'-Diisobutyl-ethylenediamine-N,N'-di-3-propionate)tetrachloridoplatinum(IV), [PtCl4(iBu2eddp)], shows an improved pharmacological profile in comparison to cisplatin. This is manifested through accelerated dying process led by necrotic cell death, reflected through mitochondrial collapse, strong ATP depletion and reactive oxygen species production. Loss of mitochondrial potential was further followed with intensive apoptosis that finalized with DNA fragmentation. Different dynamic of tumoricidal action could be partly ascribed to less affected repair mechanisms in comparison to cisplatin. Importantly, [PtCl4(iBu2eddp)] did not induce necrosis in primary fibroblasts suggesting different intracellular response of normal vs. tumor cells. This selectivity toward malignant phenotype is further confirmed by retained tumoricidal potential in hypoxic conditions, while cisplatin became completely inefficient.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Platinum Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Apoptosis/physiology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Survival/physiology , Dose-Response Relationship, Drug , Mice , NIH 3T3 Cells , Platinum Compounds/chemistry
10.
Angew Chem Int Ed Engl ; 53(23): 5982-7, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24828572

ABSTRACT

The strong therapeutic potential of an organotin(IV) compound loaded in nanostructured silica (SBA-15pSn) is demonstrated: B16 melanoma tumor growth in syngeneic C57BL/6 mice is almost completely abolished. In contrast to apoptosis as the basic mechanism of the anticancer action of numerous chemotherapeutics, the important advantage of this SBA-15pSn mesoporous material is the induction of cell differentiation, an effect unknown for metal-based drugs and nanomaterials alone. This non-aggressive mode of drug action is highly efficient against cancer cells but is in the concentration range used nontoxic for normal tissue. JNK (Jun-amino-terminal kinase)-independent apoptosis accompanied by the development of the melanocyte-like nonproliferative phenotype of survived cells indicates the extraordinary potential of SBA-15pSn to suppress tumor growth without undesirable compensatory proliferation of malignant cells in response to neighboring cell death.


Subject(s)
Melanoma, Experimental/drug therapy , Neoplasms/therapy , Organotin Compounds/chemistry , Silicon Dioxide/pharmacology , Animals , Apoptosis , Cell Proliferation , Mice , Mice, Inbred C57BL , Nanostructures
11.
ChemMedChem ; 9(7): 1586-93, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24470190

ABSTRACT

Iridium(III) complexes of the type [Ir(η(5) -C5 Me5 )Cl2 {Ph2 PCH2 CH2 CH2 S(O)x Ph-κP}] (x=0-2; 1-3) and [Ir(η(5) -C5 Me5 )Cl{Ph2 PCH2 CH2 CH2 S(O)x Ph-κP,κS}][PF6 ] (x=0-1; 4 and 5) with 3-(diphenylphosphino)propyl phenyl sulfide, sulfoxide, and sulfone ligands Ph2 PCH2 CH2 CH2 S(O)x Ph were designed, synthesized, and characterized fully, including X-ray diffraction analyses for complexes 3 and 4. In vitro studies against human thyroid carcinoma (8505C), submandibular carcinoma (A253), breast adenocarcinoma (MCF-7), colon adenocarcinoma (SW480), and melanoma (518A2) cell lines provided evidence for the high biological potential of the neutral and cationic iridium(III) complexes. Neutral iridium(III) complex 5 proved to be the most active, with IC50 values up to about 0.1 µM, representing activities of up to one order of magnitude higher than cisplatin. Using 8505C cells, apoptosis was shown to be the main mechanism through which complex 5 exerts its tumoricidal action. The described iridium(III) complexes represent potential leads in the search for novel metal-based anticancer agents.


Subject(s)
Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Iridium/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Crystallography, X-Ray , Humans , Ligands , MCF-7 Cells , Molecular Conformation , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
12.
Eur J Med Chem ; 69: 216-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24042044

ABSTRACT

Neutral iridium(III) complexes of the type [Ir(η(5)-C5Me5)Cl2{Ph2PCH2S(O)xPh-κP}] (1-3) with diphenylphosphino-functionalized methyl phenyl sulfides, sulfoxides, and sulfones Ph2PCH2S(O)xPh (x = 0, L1; 1, L2; 2, L3) and the cationic complex [Ir(η(5)-C5Me5)Cl{Ph2PCH2SPh-κP,κS}][PF6] (4) were synthesized and fully characterized analytically and spectroscopically. Furthermore, the structure of 2 was determined by X-ray diffraction analysis. The biological potential of the neutral and cationic iridium(III) complexes was tested in vitro against the cell lines 8505C, A253, MCF-7, SW480 and 518A2. Complex [Ir(η(5)-C5Me5)Cl2{Ph2PCH2S(O)Ph-κP}] (2), with ligand L2 κP coordinated containing a pendent sulfinyl group, is the most active one (IC50 values of about 3 µM), thus, with activities comparable to cisplatin. Complex 2 proved to have an even a higher antiproliferative activity than cisplatin against 8505C and SW480 cell lines, used as a model system of highly anaplastic cancers with low sensitivity to conventional chemotherapeutics such as cisplatin. Additional experiments demonstrated that apoptosis and autophagic cell death contribute to the drug's tumoricidal action.


Subject(s)
Antineoplastic Agents/pharmacology , Iridium/chemistry , Organometallic Compounds/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cations/chemical synthesis , Cations/chemistry , Cations/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/chemistry , Cisplatin/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Ligands , MCF-7 Cells , Models, Molecular , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
13.
J Membr Biol ; 246(4): 307-14, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23417033

ABSTRACT

Soy isoflavones represent hopeful unconventional remedies in the therapy of prostate cancer. The aim of our study was to determine the effects of genistein and daidzein on the parameters that reflect metastatic potential, membrane fluidity, invasiveness and dynamic phenotype in Matrigel of LNCaP and PC-3 prostate cancer cells. Cell viability tests, using a wide range of concentrations of soy isoflavones (6-75 µg/ml for 72 h), were conducted to determine their IC50 concentrations. Electron paramagnetic resonance investigations of prostate cancer cell membrane fluidity were performed at IC50 concentrations of genistein and daidzein (12.5 and 25 µg/ml, respectively, for 10 min). Genistein provoked significant increases in the membrane order parameter (which is reciprocally proportional to membrane fluidity) of 0.722 ± 0.006 (LNCaP), 0.753 ± 0.010 (LNCaP + genistein), 0.723 ± 0.007 (PC-3) and 0.741 ± 0.004 (PC-3 + genistein); however, no such effects were observed for daidzein. While both genistein and daidzein reduced the proliferation of prostate cancer cells at their respective IC50 concentrations, during the 72 h of incubation only genistein provoked effects on the dynamic phenotype and decreased invasiveness. The effect was more evident in PC-3 cells compared to LNCaP cells. Our results imply that (1) invasive activity is at least partially dependent on membrane fluidity, (2) genistein may exert its antimetastatic effects by changing the mechanical properties of prostate cancer cells and (3) daidzein should be applied at higher concentrations than genistein in order to achieve pharmacological effects.


Subject(s)
Genistein/pharmacology , Glycine max/chemistry , Isoflavones/pharmacology , Membrane Fluidity/drug effects , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Inhibitory Concentration 50 , Male
14.
Metallomics ; 4(9): 979-87, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22820831

ABSTRACT

Several new R(2)eddp (R = i-Pr, i-Bu; eddp = ethylenediamine-N,N'-di-3-propionate) esters and corresponding platinum(ii) and platinum(iv) complexes of the general formula [PtCl(n)(R(2)edda-type)] (n = 2, 4) were synthesized and characterized by spectroscopic methods (IR, (1)H and (13)C NMR) and elemental analysis. The crystal structure of platinum(iv) complex [PtCl(4){(c-Pe)(2)eddip}] (3a) was resolved and is given herein. Ligand precursors, platinum(ii), and platinum(iv) complexes were tested against eight tumor cell lines (CT26CL25, HTC116, SW620, PC3, LNCaP, U251, A375, and B16). Selectivity in the action of those compounds between tumor and two normal primary cells (fibroblasts and keratinocytes) are discussed. A structure-activity relationship of these compounds is discussed. Furthermore, cell cycle distribution, induction of necrosis, apoptosis, autophagy, anoikis, caspase activation, ROS, and RNS are presented on the cisplatin-resistant colon carcinoma HCT116 cell line.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Organoplatinum Compounds/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/chemistry , Cisplatin/pharmacology , Cisplatin/toxicity , Humans , Ligands , Mice , Molecular Conformation , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/toxicity , Oxidative Stress/drug effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
15.
J Inorg Biochem ; 109: 40-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22369771

ABSTRACT

This paper focuses on the synthesis, characterization and biological activity of new N,N'-methylene modified cyclohexyl ethylenediamine-N,N'-diacetate (edda)-type ligands and their Pt(IV) complexes. Both the ligands and complexes were characterized by infrared, UV-vis, ESI-MS, 1D ((1)H, (13)C, (195)Pt) and 2D (COSY, HSQC, HMBC) NMR spectroscopy and elemental analysis. The possible correlation between the reduction potentials and the cytotoxicity of the complexes was examined. The potential antitumoral activity of all compounds was tested in vitro on human melanoma A375, human glioblastoma U251, human prostate cancer PC3, human colon cancer HCT116, mouse melanoma B16 and mouse colon cancer CT26CL25 cells, as well as primary fibroblasts and keratinocytes. The results obtained revealed strong antitumor potential of the newly synthesized drugs with preserved efficacy against cisplatin resistant lines and less toxicity towards nonmalignant counterparts. The mechanism found to be responsible for the observed tumoricidal action of each synthesized compound was induction of apoptosis generally accompanied with caspase activation. Taken together, the effective response to the treatment of a wide range of different cell lines, including cisplatin resistant subclones, as well as induction of apoptosis, as the mechanism suggested to be the most desirable way of eliminating malignant cells, represents a great advantage of this novel group of drugs in comparison to other members in this metallo-drug family.


Subject(s)
Antineoplastic Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Cyclohexanes/chemical synthesis , Ethylenediamines/chemical synthesis , Organoplatinum Compounds/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Cyclohexanes/chemistry , Cyclohexanes/pharmacology , Edetic Acid/chemistry , Ethylenediamines/chemistry , Ethylenediamines/pharmacology , Flow Cytometry , Humans , Magnetic Resonance Spectroscopy , Mice , Molecular Structure , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Spectrometry, Mass, Electrospray Ionization , Spectroscopy, Fourier Transform Infrared
SELECTION OF CITATIONS
SEARCH DETAIL
...