1
|
Li A, Huang K, Pan W, Wu Y, Liang Y, Zhang Z, Wu D, Ma L, Gou Y. Thiosemicarbazone Mixed-Valence Cu(I/II) Complex against Lung Adenocarcinoma Cells through Multiple Pathways Involving Cuproptosis. J Med Chem 2024; 67:9091-9103. [PMID: 38778566 DOI: 10.1021/acs.jmedchem.4c00257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Induction of cuproptosis and targeting of multiple signaling pathways show promising applications in tumor therapy. In this study, we synthesized two thiosemicarbazone-copper complexes ([CuII(L)Cl] 1 and [CuII2CuI(L)2Cl3] 2, where HL is the (E)-N-methyl-2-(phenyl(pyridin-2-yl)methylene ligand), to assess their antilung cancer activities. Both copper complexes showed better anticancer activity than cisplatin and exhibited hemolysis comparable to that of cisplatin. In vivo experiments showed that complex 2 retarded the A549 cell growth in a mouse xenograft model with low systemic toxicity. Primarily, complex 2 kills lung cancer cells in vitro and in vivo by triggering multiple pathways, including cuproptosis. Complex 2 is the first mixed-valent Cu(I/II) complex to induce cellular events consistent with cuproptosis in cancer cells, which may stimulate the development of mixed-valent copper complexes and provide effective cancer therapy.
Collapse
Affiliation(s)
- Aili Li
- Laboratory of Respiratory Diseases, The Affiliated Hospital of Guilin Medical University, Guilin 541001, P. R. China
- Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin 541001, P. R. China
| | - Kai Huang
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin 541001, P. R. China
- Department of Scientific Research, The Affiliated Hospital of Guilin Medical University, Guilin 541001, P. R. China
| | - Weiping Pan
- Laboratory of Respiratory Diseases, The Affiliated Hospital of Guilin Medical University, Guilin 541001, P. R. China
| | - Youru Wu
- Laboratory of Respiratory Diseases, The Affiliated Hospital of Guilin Medical University, Guilin 541001, P. R. China
| | - Yuwei Liang
- Department of Scientific Research, The Affiliated Hospital of Guilin Medical University, Guilin 541001, P. R. China
| | - ZhenLei Zhang
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, P. R. China
| | - Daqi Wu
- Laboratory of Respiratory Diseases, The Affiliated Hospital of Guilin Medical University, Guilin 541001, P. R. China
| | - Libing Ma
- Laboratory of Respiratory Diseases, The Affiliated Hospital of Guilin Medical University, Guilin 541001, P. R. China
- Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin 541001, P. R. China
| | - Yi Gou
- Laboratory of Respiratory Diseases, The Affiliated Hospital of Guilin Medical University, Guilin 541001, P. R. China
- Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin 541001, P. R. China
| |
Collapse
|
2
|
Antiproliferative Activity and DNA Interaction Studies of a Series of N4,N4-Dimethylated Thiosemicarbazone Derivatives. Molecules 2023; 28:molecules28062778. [PMID: 36985750 PMCID: PMC10058200 DOI: 10.3390/molecules28062778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
The exploitation of bioactive natural sources to obtain new anticancer agents with novel modes of action may represent an innovative and successful strategy in the field of medicinal chemistry. Many natural products and their chemical analogues have been proposed as starting molecules to synthesise compounds with increased biological potential. In this work, the design, synthesis, and characterisation of a new series of N4,N4-dimethylated thiosemicarbazone Cu(II), Ni(II), and Pt(II) complexes are reported and investigated for their in vitro toxicological profile against a leukaemia cell line (U937). The antiproliferative activity was studied by MTS assay to determine the GI50 value for each compound after 24 h of treatment, while the genotoxic potential was investigated to determine if the complexes could cause DNA damage. In addition, the interaction between the synthesised molecules and DNA was explored by means of spectroscopic techniques, showing that for Pt and Ni derivatives a single mode of action can be postulated, while the Cu analogue behaves differently.
Collapse
|
3
|
Ganorkar K, Samanta A, Mukherjee S, Joshi R, Gupta S, Sarkar A, Ghosh SK. Switching of the Polarity-Sensitive Aggregation Pattern of a Thiosemicarbazone-Based Anticancer Luminophore and Its Involvement in Cellular Apoptosis of the Human Lung Cancer Cell Line. J Phys Chem B 2023; 127:104-120. [PMID: 36594702 DOI: 10.1021/acs.jpcb.2c06410] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Elucidation of the photophysical and biochemical properties of small molecules can facilitate their applications as prospective therapeutic imaging (theragnostic) agents. Herein, we demonstrate the luminescence behavior of a strategically designed potential therapeutic thiosemicarbazone derivative, (E)-1-(4-(diethylamino)-2-hydroxybenzylidene)-4,4-dimethylthiosemicarbazide (DAHTS), accompanied by the illustration of its solvation and solvation dynamics using spectroscopic techniques and exploring its promising antitumor activities by adopting the necessary biochemical assays. Solvent-dependent photophysical properties, namely UV-vis absorption, fluorescence emission, and excitation profiles, concentration-dependent studies, and time-resolved fluorescence decays, serve as footprints to explain the existence of DAHTS monomers, its excited-state intramolecular proton transfer (ESIPT) product, and dimeric and aggregated forms. The emission intensity progressively intensifies with increasing polarity and proticity of the solvents up to MeOH, but in water, a sudden dip is seen. Solvent polarity and H-bonding modulate the fluorescence behavior of the primary emission peak and significantly influence the formation of the dimer and DAHTS aggregates. The designed luminophore (DAHTS) exhibits significant antiproliferative activity against the human lung cancer (A549) cell lines with inhibitory concentrations (IC50) of 16.88 and 11.92 μM for 24 and 48 h, respectively. DAHTS effectively reduces the cell viability and induces cytotoxicity with extensive morphological changes in A549 cells in the form of spikes when compared to the normal HEK cell lines. More importantly, it increases the p53 expression at the mRNA level that consolidates its potential therapeutic activity. The effect of DAHTS on apoptotic pathways against the A549 cell line has been investigated to determine its probable mechanism of cell death. Thus, the all-inclusive understanding of the photophysical properties and the necessary biochemical assays put forward important steps toward tailoring the thiosemicarbazone core structure for favorable cancer theragnostic applications in academic and pharmaceutical research.
Collapse
Affiliation(s)
- Kapil Ganorkar
- Department of Chemistry, Visvesvaraya National Institute of Technology, Nagpur, Maharashtra440010, India
| | - Angela Samanta
- CMBL, Department of Biological Sciences, BITS-Pilani, K.K. Birla Goa Campus, Zuarinagar, Goa403726, India
| | - Soham Mukherjee
- Department of Chemistry, Visvesvaraya National Institute of Technology, Nagpur, Maharashtra440010, India.,Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York14850, United States
| | - Ritika Joshi
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna, Maharashtra431 203, India
| | - Smruti Gupta
- Department of Chemistry, Visvesvaraya National Institute of Technology, Nagpur, Maharashtra440010, India
| | - Angshuman Sarkar
- CMBL, Department of Biological Sciences, BITS-Pilani, K.K. Birla Goa Campus, Zuarinagar, Goa403726, India
| | - Sujit Kumar Ghosh
- Department of Chemistry, Visvesvaraya National Institute of Technology, Nagpur, Maharashtra440010, India
| |
Collapse
|
4
|
Babak MV, Ahn D. Modulation of Intracellular Copper Levels as the Mechanism of Action of Anticancer Copper Complexes: Clinical Relevance. Biomedicines 2021; 9:biomedicines9080852. [PMID: 34440056 PMCID: PMC8389626 DOI: 10.3390/biomedicines9080852] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/29/2022] Open
Abstract
Copper (Cu) is a vital element required for cellular growth and development; however, even slight changes in its homeostasis might lead to severe toxicity and deleterious medical conditions. Cancer patients are typically associated with higher Cu content in serum and tumor tissues, indicating increased demand of cancer cells for this micronutrient. Cu is known to readily cycle between the +1 and +2 oxidation state in biological systems. The mechanism of action of Cu complexes is typically based on their redox activity and induction of reactive oxygen species (ROS), leading to deadly oxidative stress. However, there are a number of other biomolecular mechanisms beyond ROS generation that contribute to the activity of anticancer Cu drug candidates. In this review, we discuss how interfering with intracellular Cu balance via either diet modification or addition of inorganic Cu supplements or Cu-modulating compounds affects tumor development, progression, and sensitivity to treatment modalities. We aim to provide the rationale for the use of Cu-depleting and Cu-overloading conditions to generate the best possible patient outcome with minimal toxicity. We also discuss the advantages of the use of pre-formed Cu complexes, such as Cu-(bis)thiosemicarbazones or Cu-N-heterocyclic thiosemicarbazones, in comparison with the in situ formed Cu complexes with metal-binding ligands. In this review, we summarize available clinical and mechanistic data on clinically relevant anticancer drug candidates, including Cu supplements, Cu chelators, Cu ionophores, and Cu complexes.
Collapse
|
5
|
Saghatforoush L, Hosseinpour S, Moeini K, Mardani Z, Bezpalko MW, Scott Kassel W. INVESTIGATION OF THE BINDING ABILITY
OF A NEW THIOSEMICARBAZONE-BASED LIGAND
AND ITS Zn(II) COMPLEX TOWARD PROTEINS AND DNA: SPECTRAL, STRUCTURAL, THEORETICAL, AND DOCKING STUDIES. J STRUCT CHEM+ 2021. [DOI: 10.1134/s0022476621050115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
6
|
Abstract
Cancer cells accumulate iron to supplement their aberrant growth and metabolism. Depleting cells of iron by iron chelators has been shown to be selectively cytotoxic to cancer cells in vitro and in vivo. Iron chelators are effective at combating a range of cancers including those which are difficult to treat such as androgen insensitive prostate cancer and cancer stem cells. This review will evaluate the impact of iron chelation on cancer cell survival and the underlying mechanisms of action. A plethora of studies have shown iron chelators can reverse some of the major hallmarks and enabling characteristics of cancer. Iron chelators inhibit signalling pathways that drive proliferation, migration and metastasis as well as return tumour suppressive signalling. In addition to this, iron chelators stimulate apoptotic and ER stress signalling pathways inducing cell death even in cells lacking a functional p53 gene. Iron chelators can sensitise cancer cells to PARP inhibitors through mimicking BRCAness; a feature of cancers trademark genomic instability. Iron chelators target cancer cell metabolism, attenuating oxidative phosphorylation and glycolysis. Moreover, iron chelators may reverse the major characteristics of oncogenic transformation. Iron chelation therefore represent a promising selective mode of cancer therapy.
Collapse
|
7
|
Shakya B, Yadav PN. Thiosemicarbazones as Potent Anticancer Agents and their Modes of Action. Mini Rev Med Chem 2020; 20:638-661. [DOI: 10.2174/1389557519666191029130310] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/17/2019] [Accepted: 10/03/2019] [Indexed: 11/22/2022]
Abstract
:Thiosemicarbazones (TSCs) are a class of Schiff bases usually obtained by the condensation of thiosemicarbazide with a suitable aldehyde or ketone. TSCs have been the focus of chemists and biologists due to their wide range of pharmacological effects. One of the promising areas in which these excellent metal chelators are being developed is their use against cancer. TSCs have a wide clinical antitumor spectrum with efficacy in various tumor types such as leukemia, pancreatic cancer, breast cancer, non-small cell lung cancer, cervical cancer, prostate cancer and bladder cancer. To obtain better activity, different series of TSCs have been developed by modifying the heteroaromatic system in their molecules. These compounds possessed significant antineoplastic activity when the carbonyl attachment of the side chain was located at a position α to the ring nitrogen atom, whereas attachment of the side chain β or γ to the heterocyclic N atom resulted in inactive antitumor agents. In addition, replacement of the heterocyclic ring N with C also resulted in a biologically inactive compound suggesting that a conjugated N,N,S-tridentate donor set is essential for the biological activities of thiosemicarbazones. Several possible mechanisms have been implemented for the anticancer activity of thiosemicarbazones.
Collapse
Affiliation(s)
- Bhushan Shakya
- Amrit Campus, Tribhuvan University, Thamel, Kathmandu, Nepal
| | - Paras Nath Yadav
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| |
Collapse
|
8
|
A novel 8-nitro quinoline-thiosemicarbazone analogues induces G1/S & G2/M phase cell cycle arrest and apoptosis through ROS mediated mitochondrial pathway. Bioorg Chem 2020; 97:103709. [DOI: 10.1016/j.bioorg.2020.103709] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 01/19/2023]
|
9
|
Rudd SG, Tsesmetzis N, Sanjiv K, Paulin CBJ, Sandhow L, Kutzner J, Hed Myrberg I, Bunten SS, Axelsson H, Zhang SM, Rasti A, Mäkelä P, Coggins SA, Tao S, Suman S, Branca RM, Mermelekas G, Wiita E, Lee S, Walfridsson J, Schinazi RF, Kim B, Lehtiö J, Rassidakis GZ, Pokrovskaja Tamm K, Warpman‐Berglund U, Heyman M, Grandér D, Lehmann S, Lundbäck T, Qian H, Henter J, Schaller T, Helleday T, Herold N. Ribonucleotide reductase inhibitors suppress SAMHD1 ara-CTPase activity enhancing cytarabine efficacy. EMBO Mol Med 2020; 12:e10419. [PMID: 31950591 PMCID: PMC7059017 DOI: 10.15252/emmm.201910419] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 12/15/2019] [Accepted: 12/17/2019] [Indexed: 01/23/2023] Open
Abstract
The deoxycytidine analogue cytarabine (ara-C) remains the backbone treatment of acute myeloid leukaemia (AML) as well as other haematological and lymphoid malignancies, but must be combined with other chemotherapeutics to achieve cure. Yet, the underlying mechanism dictating synergistic efficacy of combination chemotherapy remains largely unknown. The dNTPase SAMHD1, which regulates dNTP homoeostasis antagonistically to ribonucleotide reductase (RNR), limits ara-C efficacy by hydrolysing the active triphosphate metabolite ara-CTP. Here, we report that clinically used inhibitors of RNR, such as gemcitabine and hydroxyurea, overcome the SAMHD1-mediated barrier to ara-C efficacy in primary blasts and mouse models of AML, displaying SAMHD1-dependent synergy with ara-C. We present evidence that this is mediated by dNTP pool imbalances leading to allosteric reduction of SAMHD1 ara-CTPase activity. Thus, SAMHD1 constitutes a novel biomarker for combination therapies of ara-C and RNR inhibitors with immediate consequences for clinical practice to improve treatment of AML.
Collapse
|
10
|
Polynuclear zinc(II) complexes of thiosemicarbazone: Synthesis, X-ray structure and biological evaluation. J Inorg Biochem 2019; 203:110908. [PMID: 31683125 DOI: 10.1016/j.jinorgbio.2019.110908] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/21/2019] [Accepted: 10/27/2019] [Indexed: 01/23/2023]
Abstract
Two new dimeric Zn(II) ([{ZnL1(DMSO2)}2]·DMSO (1), [{ZnL2Cl}2] (2)) and a novel tetrameric Zn(II) complex ([(Zn2L3)2(μ-OAc)2(μ3-O)2] (3)), where H2L1 = 4-(p-methoxyphenyl) thiosemicarbazone of o-hydroxynapthaldehyde, HL2 = 4-(p-methoxyphenyl)thiosemicarbazone of benzoyl pyridine and H2L3 = 4-(p-chlorophenyl)thiosemicarbazone of o-vanillin are reported. Ligands and their complexes were characterized by spectroscopic and single crystal X-ray diffraction techniques. In addition, the complexes exhibited good binding affinity towards HSA (1012 M-1), which is supported by their ability to quench the tryptophan fluorescence emission spectra of HSA. The complexes were also screened for their DNA binding propensity through UV-vis absorption titration, circular dichroism and fluorescence spectral studies. Results show that they effectively interact with CT-DNA through an intercalative mode of binding, with binding constants ranging from 103 to 104 M-1. Among the three complexes 1 has the highest binding affinity towards CT-DNA. Further, the phosphatase activity was evaluated using bis(2,4-dinitrophenyl)phosphate (BDNPP) as substrate, however, the complexes did not yield any measurable catalytic activity. Nevertheless the complexes showed significant cytotoxic potential against HeLa and HT-29 cancer cell lines that was assessed through MTT assay and DAPI staining. Remarkably, complex 1 showed better activity than cisplatin against HT-29 cell line.
Collapse
|
11
|
Wang F, Lv H, Zhao B, Zhou L, Wang S, Luo J, Liu J, Shang P. Iron and leukemia: new insights for future treatments. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:406. [PMID: 31519186 PMCID: PMC6743129 DOI: 10.1186/s13046-019-1397-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/27/2019] [Indexed: 01/19/2023]
Abstract
Iron, an indispensable element for life, is involved in all kinds of important physiological activities. Iron promotes cell growth and proliferation, but it also causes oxidative stress damage. The body has a strict regulation mechanism of iron metabolism due to its potential toxicity. As a cancer of the bone marrow and blood cells, leukemia threatens human health seriously. Current studies suggest that dysregulation of iron metabolism and subsequent accumulation of excess iron are closely associated with the occurrence and progress of leukemia. Specifically, excess iron promotes the development of leukemia due to the pro-oxidative nature of iron and its damaging effects on DNA. On the other hand, leukemia cells acquire large amounts of iron to maintain rapid growth and proliferation. Therefore, targeting iron metabolism may provide new insights for approaches to the treatment of leukemia. This review summarizes physiologic iron metabolism, alternations of iron metabolism in leukemia and therapeutic opportunities of targeting the altered iron metabolism in leukemia, with a focus on acute leukemia.
Collapse
Affiliation(s)
- Fang Wang
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Huanhuan Lv
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Bin Zhao
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Liangfu Zhou
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Shenghang Wang
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jie Luo
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Junyu Liu
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China. .,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
12
|
Süleymanoğlu M, Kaya B, Erdem-Kuruca S, Ülküseven B. Iron(III) and nickel(II) complexes of tetradentate thiosemicarbazones: Synthesis, structure, cytotoxicity, and lipophilicity. J Biochem Mol Toxicol 2019; 33:e22383. [PMID: 31392809 DOI: 10.1002/jbt.22383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 07/19/2019] [Accepted: 07/26/2019] [Indexed: 11/07/2022]
Abstract
Eighteen of the iron(III) and nickel(II) complexes with tetradentate thiosemicarbazidato ligands were synthesized and described, by analytical and spectroscopic methods. Two complexes as an example to the iron and nickel centered ones were crystallographically analyzed to confirm the molecular structures. Cytotoxic effects of the complexes on K562 chronic myeloid leukemia cells were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. For comparison, human umbilical vein endothelial cells (HUVECs) was used as a noncancerous cell line. While four of the iron(III) complexes exhibited the antileukemic effect with 50% inhibition of cell growth (IC50 ) values in the 3.4 to 6.9 μg/mL range on K562 cell line, the nickel(II) complexes showed no significant effect on both cell lines. The complexes Fe4, Fe5, and Fe6, bearing 4-methoxy substituent exhibited relatively high antiproliferative activity on both cell lines. Complex Fe3 with 3-methoxy and S-allyl groups exhibited a selectivity between K562 and HUVEC cells by IC50 values of 6.9 and >10 μg/mL, respectively. Lipophilicity, a key parameter for bioavailability and oral administration, was found in the range of -0.3 and +1.3 that desired for drug active ingredients. The results were discussed in the context of a structure-activity relationship.
Collapse
Affiliation(s)
- Mediha Süleymanoğlu
- Department of Medical Biology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Büşra Kaya
- Department of Chemistry, Engineering Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Serap Erdem-Kuruca
- Department of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Bahri Ülküseven
- Department of Chemistry, Engineering Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
13
|
Abstract
Since its discovery, polycythemia vera (PV) has challenged clinicians responsible for its diagnosis and management and scientists investigating its pathogenesis. As a clonal hematopoietic stem cell (HSC) disorder, PV is a neoplasm but its driver mutations result in overproduction of morphologically and functionally normal blood cells. PV arises in an HSC but it can present initially as isolated erythrocytosis, leukocytosis, thrombocytosis, or any combination of these together with splenomegaly or myelofibrosis, and it can take years for a true panmyelopathy to appear. PV shares the same JAK2 mutation as essential thrombocytosis and primary myelofibrosis, but erythrocytosis only occurs in PV. However, unlike secondary causes of erythrocytosis, in PV, the plasma volume is frequently expanded, masking the erythrocytosis and making diagnosis difficult if this essential fact is ignored. PV is not a monolithic disorder: female patients deregulate fewer genes and clinically behave differently than their male counterparts, while some PV patients are genetically predisposed to an aggressive clinical course. Nevertheless, based on what we have learned over the past century, most PV patients can lead long and productive lives. In this review, using clinical examples, I describe how I diagnose and manage PV in an evidence-based manner without relying on chemotherapy.
Collapse
|
14
|
de Siqueira LRP, de Moraes Gomes PAT, de Lima Ferreira LP, de Melo Rêgo MJB, Leite ACL. Multi-target compounds acting in cancer progression: Focus on thiosemicarbazone, thiazole and thiazolidinone analogues. Eur J Med Chem 2019; 170:237-260. [DOI: 10.1016/j.ejmech.2019.03.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/11/2019] [Accepted: 03/10/2019] [Indexed: 02/08/2023]
|
15
|
Novel palladium (II) complexes with tetradentate thiosemicarbazones. Synthesis, characterization, in vitro cytotoxicity and xanthine oxidase inhibition. Invest New Drugs 2019; 37:1187-1197. [PMID: 30874940 DOI: 10.1007/s10637-019-00751-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/25/2019] [Indexed: 10/27/2022]
Abstract
In vitro cytotoxicity and xanthine oxidase inhibition capabilities were investigated for five palladium (II) chelate complexes. The palladium complexes were synthesized by starting from S-alkyl-thiosemicarbazones where the alkyl component is methyl, ethyl, propyl or butyl. The solid complexes are characterized by elemental analysis and spectroscopic techniques (UV-visible, IR and 1H NMR). In order to be able to verify the N2O2-type thiosemicarbazidato ligand (L2-) structure in the square planar geometry, complex 1 has been studied as a representative by using single crystal X-ray crystallography. The in vitro cytotoxic activity measurements were carried out in HepG2 and Hep3B hepatocellular carcinomas, HCT116 colorectal carcinoma, and 3 T3 mouse fibroblast cell lines. The palladium complexes exhibited notable cytotoxic activities in all cell lines at lower μM concentrations compared to the standard chemicals, cisplatin and allopurinol. IC50 values were determined between 0.42 ± 0.01 and 12.01 ± 0.37 μg/ml in examining the antixanthine oxidase abilities of the complexes. Two complexes with S-methyl group exhibited a high inhibition activity on the xanthine oxidase. The results indicated that these complexes could be used as active pharmaceutical ingredients.
Collapse
|
16
|
Heffeter P, Pape VFS, Enyedy ÉA, Keppler BK, Szakacs G, Kowol CR. Anticancer Thiosemicarbazones: Chemical Properties, Interaction with Iron Metabolism, and Resistance Development. Antioxid Redox Signal 2019; 30:1062-1082. [PMID: 29334758 DOI: 10.1089/ars.2017.7487] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE During the past decades, thiosemicarbazones were clinically developed for a variety of diseases, including tuberculosis, viral infections, malaria, and cancer. With regard to malignant diseases, the class of α-N-heterocyclic thiosemicarbazones, and here especially 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (Triapine), was intensively developed in multiple clinical phase I/II trials. Recent Advances: Very recently, two new derivatives, namely COTI-2 and di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) have entered phase I evaluation. Based on the strong metal-chelating/metal-interacting properties of thiosemicarbazones, interference with the cellular iron (and copper) homeostasis is assumed to play an important role in their biological activity. CRITICAL ISSUES In this review, we summarize and analyze the data on the interaction of (α-N-heterocyclic) thiosemicarbazones with iron, with the special aim of bridging the current knowledge on their mode of action from chemistry to (cell) biology. In addition, we highlight the difference to classical iron(III) chelators such as desferrioxamine (DFO), which are used for the treatment of iron overload. FUTURE DIRECTIONS We want to emphasize that thiosemicarbazones are not solely removing iron from the cells/organism. In contrast, they should be considered as iron-interacting drugs influencing diverse biological pathways in a complex and multi-faceted mode of action. Consequently, in addition to the discussion of physicochemical properties (e.g., complex stability, redox activity), this review contains an overview on the diversity of cellular thiosemicarbazone targets and drug resistance mechanisms.
Collapse
Affiliation(s)
- Petra Heffeter
- 1 Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna , Vienna, Austria .,2 Research Cluster "Translational Cancer Therapy Research," Vienna, Austria
| | - Veronika F S Pape
- 3 Institute of Enzymology , Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary .,4 Department of Physiology, Faculty of Medicine, Semmelweis University , Budapest, Hungary
| | - Éva A Enyedy
- 5 Department of Inorganic and Analytical Chemistry, University of Szeged , Szeged, Hungary
| | - Bernhard K Keppler
- 2 Research Cluster "Translational Cancer Therapy Research," Vienna, Austria .,6 Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna , Vienna, Austria
| | - Gergely Szakacs
- 1 Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna , Vienna, Austria .,3 Institute of Enzymology , Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Christian R Kowol
- 2 Research Cluster "Translational Cancer Therapy Research," Vienna, Austria .,6 Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna , Vienna, Austria
| |
Collapse
|
17
|
Kunos CA, Capala J, Ivy SP. Radiopharmaceuticals for Relapsed or Refractory Leukemias. Front Oncol 2019; 9:97. [PMID: 30859091 PMCID: PMC6397856 DOI: 10.3389/fonc.2019.00097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 02/04/2019] [Indexed: 12/21/2022] Open
Abstract
Radiopharmaceuticals, meaning drugs that hold a radionuclide intended for use in cancer patients for treatment of their disease or for palliation of their disease-related symptoms, have gained new interest for clinical development in adult patients with relapsed or refractory leukemia. About one-third of adult patients outlive their leukemia, with the remainder unable to attain complete remission status following the first phase of treatment due to refractory bone marrow or blood residual microscopic disease. The National Cancer Institute (NCI) Cancer Therapy Evaluation Program conducted 49 phase 1-1b trials in adult patients with leukemia between 1986 and 2017 in an effort to discover tolerated and effective therapeutic drug combinations intended to improve remission and mortality rates. None of these trials involved radiopharmaceuticals. In this article, the NCI perspective on the challenges encountered in and on the future potential of radiopharmaceuticals alone or in combination for adult patients with relapsed or refractory leukemia is discussed. An effort is underway already to build-up the NCI's clinical trial enterprise infrastructure for radiopharmaceutical clinical development.
Collapse
Affiliation(s)
- Charles A Kunos
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, United States
| | - Jacek Capala
- Radiation Research Program, National Cancer Institute, Bethesda, MD, United States
| | - Susan Percy Ivy
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
18
|
Gu S, Yu P, Hu J, Liu Y, Li Z, Qian Y, Wang Y, Gou Y, Yang F. Mitochondria-localizing N-heterocyclic thiosemicarbazone copper complexes with good cytotoxicity and high antimetastatic activity. Eur J Med Chem 2019; 164:654-664. [DOI: 10.1016/j.ejmech.2019.01.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/26/2018] [Accepted: 01/06/2019] [Indexed: 01/16/2023]
|
19
|
Fordham SE, Blair HJ, Elstob CJ, Plummer R, Drew Y, Curtin NJ, Heidenreich O, Pal D, Jamieson D, Park C, Pollard J, Fields S, Milne P, Jackson GH, Marr HJ, Menne T, Jones GL, Allan JM. Inhibition of ATR acutely sensitizes acute myeloid leukemia cells to nucleoside analogs that target ribonucleotide reductase. Blood Adv 2018; 2:1157-1169. [PMID: 29789314 PMCID: PMC5965047 DOI: 10.1182/bloodadvances.2017015214] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/09/2018] [Indexed: 12/20/2022] Open
Abstract
The ataxia telangiectasia and Rad3-related (ATR) protein kinase promotes cancer cell survival by signaling stalled replication forks generated by replication stress, a common feature of many cancers including acute myeloid leukemia (AML). Here we show that the antileukemic activity of the chemotherapeutic nucleoside analogs hydroxyurea and gemcitabine was significantly potentiated by ATR inhibition via a mechanism involving ribonucleotide reductase (RNR) abrogation and inhibition of replication fork progression. When administered in combination with gemcitabine, an inhibitor of the M1 RNR subunit, the ATR inhibitor VX-970, eradicated disseminated leukemia in an orthotopic mouse model, eliciting long-term survival and effective cure. These data identify a synergistic interaction between ATR inhibition and RNR loss that will inform the deployment of small molecule inhibitors for the treatment of AML and other hematologic malignancies.
Collapse
Affiliation(s)
- Sarah E Fordham
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Helen J Blair
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Claire J Elstob
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ruth Plummer
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Yvette Drew
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nicola J Curtin
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Olaf Heidenreich
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Deepali Pal
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David Jamieson
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Catherine Park
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John Pollard
- Vertex Pharmaceuticals (Europe) Ltd, Abingdon, Oxfordshire, United Kingdom
| | - Scott Fields
- Vertex Pharmaceuticals (Europe) Ltd, Abingdon, Oxfordshire, United Kingdom
| | - Paul Milne
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; and
| | - Graham H Jackson
- Department of Haematology, Freeman Hospital, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Helen J Marr
- Department of Haematology, Freeman Hospital, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Tobias Menne
- Department of Haematology, Freeman Hospital, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Gail L Jones
- Department of Haematology, Freeman Hospital, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - James M Allan
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
20
|
Rejmund M, Mrozek-Wilczkiewicz A, Malarz K, Pyrkosz-Bulska M, Gajcy K, Sajewicz M, Musiol R, Polanski J. Piperazinyl fragment improves anticancer activity of Triapine. PLoS One 2018; 13:e0188767. [PMID: 29652894 PMCID: PMC5898707 DOI: 10.1371/journal.pone.0188767] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/13/2017] [Indexed: 12/17/2022] Open
Abstract
A new class of TSCs containing piperazine (piperazinylogs) of Triapine, was designed to fulfill the di-substitution pattern at the TSCs N4 position, which is a crucial prerequisite for the high activity of the previously obtained TSC compounds–DpC and Dp44mT. We tested the important physicochemical characteristics of the novel compounds L1-L12. The studied ligands are neutral at physiological pH, which allows them to permeate cell membranes and bind cellular Fe pools more readily than less lipid-soluble ligands, e.g. DFO. The selectivity and anti-cancer activity of the novel TSCs were examined in a variety of cancer cell types. In general, the novel compounds demonstrated the greatest promise as anti-cancer agents with both a potent and selective anti-proliferative activity. We investigated the mechanism of action more deeply, and revealed that studied compounds inhibit the cell cycle (G1/S phase). Additionally we detected apoptosis, which is dependent on cell line’s specific genetic profile. Accordingly, structure-activity relationship studies suggest that the combination of the piperazine ring with Triapine allows potent and selective anticancer chelators that warrant further in vivo examination to be identified. Significantly, this study proved the importance of the di-substitution pattern of the amine N4 function.
Collapse
Affiliation(s)
- Marta Rejmund
- Institute of Chemistry, University of Silesia, Katowice, Poland
| | - Anna Mrozek-Wilczkiewicz
- A. Chełkowski Institute of Physics, University of Silesia, Katowice, Poland
- Silesian Center for Education and Interdisciplinary Research, University of Silesia, Chorzów, Poland
| | - Katarzyna Malarz
- Institute of Chemistry, University of Silesia, Katowice, Poland
- Silesian Center for Education and Interdisciplinary Research, University of Silesia, Chorzów, Poland
| | | | - Kamila Gajcy
- Institute of Chemistry, University of Silesia, Katowice, Poland
| | | | - Robert Musiol
- Institute of Chemistry, University of Silesia, Katowice, Poland
| | - Jaroslaw Polanski
- Institute of Chemistry, University of Silesia, Katowice, Poland
- * E-mail:
| |
Collapse
|
21
|
Bonaccorso C, Grasso G, Musso N, Barresi V, Condorelli DF, La Mendola D, Rizzarelli E. Water soluble glucose derivative of thiocarbohydrazone acts as ionophore with cytotoxic effects on tumor cells. J Inorg Biochem 2018; 182:92-102. [PMID: 29452884 DOI: 10.1016/j.jinorgbio.2018.01.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/28/2018] [Accepted: 01/30/2018] [Indexed: 12/26/2022]
Abstract
A novel water-soluble ionophore based on the thiocarbohydrazone moiety conjugated with glucose (GluTch) was synthesized through a simple two-step procedure. Structural elucidation was carried out in water solution by means of various spectroscopic techniques (NMR, UV-Vis, and CD), electrospray ionization mass spectrometry and density functional theory calculations. The flexible nature of the thiocarbohydrazone moiety of the new glycoderivative compound induced both different coordination motifs and stoichiometry towards copper and zinc. Cytotoxicity assays of the ligands on the human normal keratinocyte NCTC-2544, MDA-MB-231 breast cancer and PC-3 human prostate adenocarcinoma cell lines demonstrated that i) higher activity on cancer cells growth inhibition compared to a normal cell line; ii) the introduction of the glucose unit does not alter the cytotoxic activity of the underivatized ionophore ligand and iii) the presence of copper ion improves the activity of the thiocarbohydrazones.
Collapse
Affiliation(s)
- Carmela Bonaccorso
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy; Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici, via Celso Ulpiani, 27, 70125 Bari, Italy
| | - Giulia Grasso
- Consiglio Nazionale delle Ricerche, Istituto di Biostrutture e Bioimmagini (IBB-CNR), Via P. Gaifami 18, 95126 Catania, Italy
| | - Nicolò Musso
- Dipartimento Scienze Biomediche e Biotecnologiche, Sez. Biochimica Medica, via S. Sofia 64, I-95125, Catania, Italy
| | - Vincenza Barresi
- Dipartimento Scienze Biomediche e Biotecnologiche, Sez. Biochimica Medica, via S. Sofia 64, I-95125, Catania, Italy
| | - Daniele F Condorelli
- Dipartimento Scienze Biomediche e Biotecnologiche, Sez. Biochimica Medica, via S. Sofia 64, I-95125, Catania, Italy
| | - Diego La Mendola
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici, via Celso Ulpiani, 27, 70125 Bari, Italy.
| | - Enrico Rizzarelli
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125 Catania, Italy; Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici, via Celso Ulpiani, 27, 70125 Bari, Italy
| |
Collapse
|
22
|
Mannargudi MB, Deb S. Clinical pharmacology and clinical trials of ribonucleotide reductase inhibitors: is it a viable cancer therapy? J Cancer Res Clin Oncol 2017. [PMID: 28624910 DOI: 10.1007/s00432-017-2457-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Ribonucleotide reductase (RR) enzymes (RR1 and RR2) play an important role in the reduction of ribonucleotides to deoxyribonucleotides which is involved in DNA replication and repair. Augmented RR activity has been ascribed to uncontrolled cell growth and tumorigenic transformation. METHODS This review mainly focuses on several biological and chemical RR inhibitors (e.g., siRNA, GTI-2040, GTI-2501, triapine, gemcitabine, and clofarabine) that have been evaluated in clinical trials with promising anticancer activity from 1960's till 2016. A summary on whether their monotherapy or combination is still effective for further use is discussed. RESULTS Among the RR2 inhibitors evaluated, GTI-2040, siRNA, gallium nitrate and didox were more efficacious as a monotherapy, whereas triapine was found to be more efficacious as combination agent. Hydroxyurea is currently used more in combination therapy, even though it is efficacious as a monotherapy. Gallium nitrate showed mixed results in combination therapy, while the combination activity of didox is yet to be evaluated. RR1 inhibitors that have long been used in chemotherapy such as gemcitabine, cladribine, fludarabine and clofarabine are currently used mostly as a combination therapy, but are equally efficacious as a monotherapy, except tezacitabine which did not progress beyond phase I trials. CONCLUSIONS Based on the results of clinical trials, we conclude that RR inhibitors are viable treatment options, either as a monotherapy or as a combination in cancer chemotherapy. With the recent advances made in cancer biology, further development of RR inhibitors with improved efficacy and reduced toxicity is possible for treatment of variety of cancers.
Collapse
Affiliation(s)
- Mukundan Baskar Mannargudi
- Clinical Pharmacology Program, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Subrata Deb
- Department of Biopharmaceutical Sciences, Roosevelt University College of Pharmacy, 1400 N. Roosevelt Blvd., Schaumburg, IL, 60173, USA.
| |
Collapse
|
23
|
Zaltariov MF, Hammerstad M, Arabshahi HJ, Jovanović K, Richter KW, Cazacu M, Shova S, Balan M, Andersen NH, Radulović S, Reynisson J, Andersson KK, Arion VB. New Iminodiacetate-Thiosemicarbazone Hybrids and Their Copper(II) Complexes Are Potential Ribonucleotide Reductase R2 Inhibitors with High Antiproliferative Activity. Inorg Chem 2017; 56:3532-3549. [PMID: 28252952 DOI: 10.1021/acs.inorgchem.6b03178] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
As ribonucleotide reductase (RNR) plays a crucial role in nucleic acid metabolism, it is an important target for anticancer therapy. The thiosemicarbazone Triapine is an efficient R2 inhibitor, which has entered ∼20 clinical trials. Thiosemicarbazones are supposed to exert their biological effects through effectively binding transition-metal ions. In this study, six iminodiacetate-thiosemicarbazones able to form transition-metal complexes, as well as six dicopper(II) complexes, were synthesized and fully characterized by analytical, spectroscopic techniques (IR, UV-vis; 1H and 13C NMR), electrospray ionization mass spectrometry, and X-ray diffraction. The antiproliferative effects were examined in several human cancer and one noncancerous cell lines. Several of the compounds showed high cytotoxicity and marked selectivity for cancer cells. On the basis of this, and on molecular docking calculations one lead dicopper(II) complex and one thiosemicarbazone were chosen for in vitro analysis as potential R2 inhibitors. Their interaction with R2 and effect on the Fe(III)2-Y· cofactor were characterized by microscale thermophoresis, and two spectroscopic techniques, namely, electron paramagnetic resonance and UV-vis spectroscopy. Our findings suggest that several of the synthesized proligands and copper(II) complexes are effective antiproliferative agents in several cancer cell lines, targeting RNR, which deserve further investigation as potential anticancer drugs.
Collapse
Affiliation(s)
- Mirela F Zaltariov
- Institute of Inorganic Chemistry, University of Vienna , Währinger Strasse 42, 1090 Vienna, Austria.,Inorganic Polymers Department, Petru Poni Institute of Macromolecular Chemistry, Romanian Academy , Aleea G. Ghica Voda 41A, 700487 Iasi, Romania
| | - Marta Hammerstad
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of Oslo , P.O. Box 1066, Blindern, NO-0316 Oslo, Norway
| | | | - Katarina Jovanović
- Institute for Oncology and Radiology of Serbia , Pasterova 14, 11000 Belgrade, Serbia
| | - Klaus W Richter
- Institute of Inorganic Chemistry-Functional Materials, University of Vienna , Althanstrasse 14, 1090 Vienna, Austria
| | - Maria Cazacu
- Inorganic Polymers Department, Petru Poni Institute of Macromolecular Chemistry, Romanian Academy , Aleea G. Ghica Voda 41A, 700487 Iasi, Romania
| | - Sergiu Shova
- Inorganic Polymers Department, Petru Poni Institute of Macromolecular Chemistry, Romanian Academy , Aleea G. Ghica Voda 41A, 700487 Iasi, Romania
| | - Mihaela Balan
- Inorganic Polymers Department, Petru Poni Institute of Macromolecular Chemistry, Romanian Academy , Aleea G. Ghica Voda 41A, 700487 Iasi, Romania
| | - Niels H Andersen
- Department of Chemistry, University of Oslo , P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
| | - Siniša Radulović
- Institute for Oncology and Radiology of Serbia , Pasterova 14, 11000 Belgrade, Serbia
| | - Jóhannes Reynisson
- School of Chemical Sciences, University of Auckland , Auckland, New Zealand
| | - K Kristoffer Andersson
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of Oslo , P.O. Box 1066, Blindern, NO-0316 Oslo, Norway
| | - Vladimir B Arion
- Institute of Inorganic Chemistry, University of Vienna , Währinger Strasse 42, 1090 Vienna, Austria
| |
Collapse
|
24
|
Stacy AE, Palanimuthu D, Bernhardt PV, Kalinowski DS, Jansson PJ, Richardson DR. Zinc(II)-Thiosemicarbazone Complexes Are Localized to the Lysosomal Compartment Where They Transmetallate with Copper Ions to Induce Cytotoxicity. J Med Chem 2016; 59:4965-84. [PMID: 27023111 DOI: 10.1021/acs.jmedchem.6b00238] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
As the di-2-pyridylketone thiosemicarbazone (DpT) and 2-acetylpyridine thiosemicarbazone (ApT) series show potent antitumor activity in vitro and in vivo, we synthesized their fluorescent zinc(II) complexes to assess their intracellular distribution. The Zn(II) complexes generally showed significantly greater cytotoxicity than the thiosemicarbazones alone in several tumor cell-types. Notably, specific structure-activity relationships demonstrated the importance of the di-2-pyridyl pharmacophore in their activity. Confocal fluorescence imaging and live cell microscopy showed that the Zn(II) complex of our lead compound, di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), which is scheduled to enter clinical trials, was localized to lysosomes. Under lysosomal conditions, the Zn(II) complexes were shown to transmetallate with copper ions, leading to redox-active copper complexes that induced lysosomal membrane permeabilization (LMP) and cytotoxicity. This is the first study to demonstrate direct lysosomal targeting of our novel Zn(II)-thiosemicarbazone complexes that mediate their activity via transmetalation with copper ions and LMP.
Collapse
Affiliation(s)
- Alexandra E Stacy
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Blackburn Building (D06), Level 5, Sydney, New South Wales 2006, Australia
| | - Duraippandi Palanimuthu
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Blackburn Building (D06), Level 5, Sydney, New South Wales 2006, Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland , Brisbane, Queensland 4072, Australia
| | - Danuta S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Blackburn Building (D06), Level 5, Sydney, New South Wales 2006, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Blackburn Building (D06), Level 5, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Blackburn Building (D06), Level 5, Sydney, New South Wales 2006, Australia
| |
Collapse
|
25
|
Redox cycling metals: Pedaling their roles in metabolism and their use in the development of novel therapeutics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:727-48. [PMID: 26844773 DOI: 10.1016/j.bbamcr.2016.01.026] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022]
Abstract
Essential metals, such as iron and copper, play a critical role in a plethora of cellular processes including cell growth and proliferation. However, concomitantly, excess of these metal ions in the body can have deleterious effects due to their ability to generate cytotoxic reactive oxygen species (ROS). Thus, the human body has evolved a very well-orchestrated metabolic system that keeps tight control on the levels of these metal ions. Considering their very high proliferation rate, cancer cells require a high abundance of these metals compared to their normal counterparts. Interestingly, new anti-cancer agents that take advantage of the sensitivity of cancer cells to metal sequestration and their susceptibility to ROS have been developed. These ligands can avidly bind metal ions to form redox active metal complexes, which lead to generation of cytotoxic ROS. Furthermore, these agents also act as potent metastasis suppressors due to their ability to up-regulate the metastasis suppressor gene, N-myc downstream regulated gene 1. This review discusses the importance of iron and copper in the metabolism and progression of cancer, how they can be exploited to target tumors and the clinical translation of novel anti-cancer chemotherapeutics.
Collapse
|
26
|
Al-Eisawi Z, Stefani C, Jansson PJ, Arvind A, Sharpe PC, Basha MT, Iskander GM, Kumar N, Kovacevic Z, Lane DJR, Sahni S, Bernhardt PV, Richardson DR, Kalinowski DS. Novel Mechanism of Cytotoxicity for the Selective Selenosemicarbazone, 2-Acetylpyridine 4,4-Dimethyl-3-selenosemicarbazone (Ap44mSe): Lysosomal Membrane Permeabilization. J Med Chem 2016; 59:294-312. [PMID: 26645570 DOI: 10.1021/acs.jmedchem.5b01399] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Selenosemicarbazones show marked antitumor activity. However, their mechanism of action remains unknown. We examined the medicinal chemistry of the selenosemicarbazone, 2-acetylpyridine 4,4-dimethyl-3-selenosemicarbazone (Ap44mSe), and its iron and copper complexes to elucidate its mechanisms of action. Ap44mSe demonstrated a pronounced improvement in selectivity toward neoplastic relative to normal cells compared to its parent thiosemicarbazone. It also effectively depleted cellular Fe, resulting in transferrin receptor-1 up-regulation, ferritin down-regulation, and increased expression of the potent metastasis suppressor, N-myc downstream regulated gene-1. Significantly, Ap44mSe limited deleterious methemoglobin formation, highlighting its usefulness in overcoming toxicities of clinically relevant thiosemicarbazones. Furthermore, Cu-Ap44mSe mediated intracellular reactive oxygen species generation, which was attenuated by the antioxidant, N-acetyl-L-cysteine, or Cu sequestration. Notably, Ap44mSe forms redox active Cu complexes that target the lysosome to induce lysosomal membrane permeabilization. This investigation highlights novel structure-activity relationships for future chemotherapeutic design and underlines the potential of Ap44mSe as a selective anticancer/antimetastatic agent.
Collapse
Affiliation(s)
- Zaynab Al-Eisawi
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Sydney, Level 5, Blackburn Building (D06)New South Wales 2006, Australia
| | - Christian Stefani
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Sydney, Level 5, Blackburn Building (D06)New South Wales 2006, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Sydney, Level 5, Blackburn Building (D06)New South Wales 2006, Australia
| | - Akanksha Arvind
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Sydney, Level 5, Blackburn Building (D06)New South Wales 2006, Australia
| | - Philip C Sharpe
- School of Chemistry and Molecular Biosciences, University of Queensland , Brisbane, Queensland 4072, Australia
| | - Maram T Basha
- School of Chemistry and Molecular Biosciences, University of Queensland , Brisbane, Queensland 4072, Australia
| | - George M Iskander
- School of Chemistry, University of New South Wales , Sydney, New South Wales 2052, Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales , Sydney, New South Wales 2052, Australia
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Sydney, Level 5, Blackburn Building (D06)New South Wales 2006, Australia
| | - Darius J R Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Sydney, Level 5, Blackburn Building (D06)New South Wales 2006, Australia
| | - Sumit Sahni
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Sydney, Level 5, Blackburn Building (D06)New South Wales 2006, Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland , Brisbane, Queensland 4072, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Sydney, Level 5, Blackburn Building (D06)New South Wales 2006, Australia
| | - Danuta S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney , Sydney, Level 5, Blackburn Building (D06)New South Wales 2006, Australia
| |
Collapse
|
27
|
Serda M, Kalinowski DS, Rasko N, Potůčková E, Mrozek-Wilczkiewicz A, Musiol R, Małecki JG, Sajewicz M, Ratuszna A, Muchowicz A, Gołąb J, Šimůnek T, Richardson DR, Polanski J. Exploring the anti-cancer activity of novel thiosemicarbazones generated through the combination of retro-fragments: dissection of critical structure-activity relationships. PLoS One 2014; 9:e110291. [PMID: 25329549 PMCID: PMC4199632 DOI: 10.1371/journal.pone.0110291] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/10/2014] [Indexed: 01/01/2023] Open
Abstract
Thiosemicarbazones (TSCs) are an interesting class of ligands that show a diverse range of biological activity, including anti-fungal, anti-viral and anti-cancer effects. Our previous studies have demonstrated the potent in vivo anti-tumor activity of novel TSCs and their ability to overcome resistance to clinically used chemotherapeutics. In the current study, 35 novel TSCs of 6 different classes were designed using a combination of retro-fragments that appear in other TSCs. Additionally, di-substitution at the terminal N4 atom, which was previously identified to be critical for potent anti-cancer activity, was preserved through the incorporation of an N4-based piperazine or morpholine ring. The anti-proliferative activity of the novel TSCs were examined in a variety of cancer and normal cell-types. In particular, compounds 1d and 3c demonstrated the greatest promise as anti-cancer agents with potent and selective anti-proliferative activity. Structure-activity relationship studies revealed that the chelators that utilized “soft” donor atoms, such as nitrogen and sulfur, resulted in potent anti-cancer activity. Indeed, the N,N,S donor atom set was crucial for the formation of redox active iron complexes that were able to mediate the oxidation of ascorbate. This further highlights the important role of reactive oxygen species generation in mediating potent anti-cancer activity. Significantly, this study identified the potent and selective anti-cancer activity of 1d and 3c that warrants further examination.
Collapse
Affiliation(s)
- Maciej Serda
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
| | - Danuta S. Kalinowski
- Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Nathalie Rasko
- Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Eliška Potůčková
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic
| | - Anna Mrozek-Wilczkiewicz
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
- A. Chełkowski Institute of Physics and Silesian Interdisciplinary Centre for Education and Research, University of Silesia, Katowice, Silesia, Poland
| | - Robert Musiol
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
| | - Jan G. Małecki
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
| | | | - Alicja Ratuszna
- A. Chełkowski Institute of Physics and Silesian Interdisciplinary Centre for Education and Research, University of Silesia, Katowice, Silesia, Poland
| | - Angelika Muchowicz
- Department of Immunology, Medical University of Warsaw, Warsaw, Mazovia, Poland
| | - Jakub Gołąb
- Department of Immunology, Medical University of Warsaw, Warsaw, Mazovia, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Mazovia, Poland
| | - Tomáš Šimůnek
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic
| | - Des R. Richardson
- Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
- * E-mail: (JP); (DRR)
| | - Jaroslaw Polanski
- Institute of Chemistry, University of Silesia, Katowice, Silesia, Poland
- * E-mail: (JP); (DRR)
| |
Collapse
|
28
|
Minden MD, Hogge DE, Weir SJ, Kasper J, Webster DA, Patton L, Jitkova Y, Hurren R, Gronda M, Goard CA, Rajewski LG, Haslam JL, Heppert KE, Schorno K, Chang H, Brandwein JM, Gupta V, Schuh AC, Trudel S, Yee KWL, Reed GA, Schimmer AD. Oral ciclopirox olamine displays biological activity in a phase I study in patients with advanced hematologic malignancies. Am J Hematol 2014; 89:363-8. [PMID: 24273151 DOI: 10.1002/ajh.23640] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 11/19/2013] [Accepted: 11/20/2013] [Indexed: 12/12/2022]
Abstract
The antimycotic ciclopirox olamine is an intracellular iron chelator that has anticancer activity in vitro and in vivo. We developed an oral formulation of ciclopirox olamine and conducted the first-in-human phase I study of this drug in patients with relapsed or refractory hematologic malignancies (Trial registration ID: NCT00990587). Patients were treated with 5-80 mg/m² oral ciclopirox olamine once daily for five days in 21-day treatment cycles. Pharmacokinetic and pharmacodynamic companion studies were performed in a subset of patients. Following definition of the half-life of ciclopirox olamine, an additional cohort was enrolled and treated with 80 mg/m² ciclopirox olamine four times daily. Adverse events and clinical response were monitored throughout the trial. Twenty-three patients received study treatment. Ciclopirox was rapidly absorbed and cleared with a short half-life. Plasma concentrations of an inactive ciclopirox glucuronide metabolite were greater than those of ciclopirox. Repression of survivin expression was observed in peripheral blood cells isolated from patients treated once daily with ciclopirox olamine at doses greater than 10 mg/m², demonstrating biological activity of the drug. Dose-limiting gastrointestinal toxicities were observed in patients receiving 80 mg/m² four times daily, and no dose limiting toxicity was observed at 40 mg/m² once daily. Hematologic improvement was observed in two patients. Once-daily dosing of oral ciclopirox olamine was well tolerated in patients with relapsed or refractory hematologic malignancies, and further optimization of dosing regimens is warranted in this patient population.
Collapse
Affiliation(s)
- Mark D. Minden
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Donna E. Hogge
- Division of Hematology and Leukemia/BMT; University of British Columbia; Vancouver British Columbia Canada
| | - Scott J. Weir
- University of Kansas Cancer Center; Kansas City Kansas
| | - Jim Kasper
- The Leukemia & Lymphoma Society; White Plains New York
| | | | | | - Yulia Jitkova
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Rose Hurren
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Carolyn A. Goard
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Lian G. Rajewski
- Biotechnology Innovation and Optimization (BIO) Center; University of Kansas; Lawrence Kansas
| | - John L. Haslam
- Biotechnology Innovation and Optimization (BIO) Center; University of Kansas; Lawrence Kansas
| | - Kathleen E. Heppert
- Biotechnology Innovation and Optimization (BIO) Center; University of Kansas; Lawrence Kansas
| | - Kevin Schorno
- University of Kansas Cancer Center; Kansas City Kansas
| | - Hong Chang
- Department of Laboratory Hematology; University Health Network; Toronto Ontario Canada
| | - Joseph M. Brandwein
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Vikas Gupta
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Andre C. Schuh
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Suzanne Trudel
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Karen W. L. Yee
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | | | - Aaron D. Schimmer
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| |
Collapse
|
29
|
Basha MT, Rodríguez C, Richardson DR, Martínez M, Bernhardt PV. Kinetic studies on the oxidation of oxyhemoglobin by biologically active iron thiosemicarbazone complexes: relevance to iron-chelator-induced methemoglobinemia. J Biol Inorg Chem 2013; 19:349-57. [PMID: 24317633 DOI: 10.1007/s00775-013-1070-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/18/2013] [Indexed: 12/29/2022]
Abstract
The oxidation of oxyhemoglobin to methemoglobin has been found to be facilitated by low molecular weight iron(III) thiosemicarbazone complexes. This deleterious reaction, which produces hemoglobin protein units unable to bind dioxygen and occurs during the administration of iron chelators such as the well-known 3-aminopyridine-2-pyridinecarbaldehyde thiosemicarbazone (3-AP; Triapine), has been observed in the reaction with Fe(III) complexes of some members of the 3-AP structurally-related thiosemicarbazone ligands derived from di-2-pyridyl ketone (HDpxxT series). We have studied the kinetics of this oxidation reaction in vitro using human hemoglobin and found that the reaction proceeds with two distinct time-resolved steps. These have been associated with sequential oxidation of the two different oxyheme cofactors in the α and β protein chains. Unexpected steric and hydrogen-bonding effects on the Fe(III) complexes appear to be the responsible for the observed differences in the reaction rate across the series of HDpxxT ligand complexes used in this study.
Collapse
Affiliation(s)
- Maram T Basha
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | | | | | | | | |
Collapse
|
30
|
Saravanan RR, Seshadri S, Gunasekaran S, Mendoza-Meroño R, Garcia-Granda S. Crystallographic, experimental (FT-IR and FT-RS) and theoretical (DFT) investigation, UV-Vis, MEP, HOMO-LUMO and NBO/NLMO of (E)-1-[1-(4-Chlorophenyl)ethylidene]thiosemicarbazide. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2013; 121:268-275. [PMID: 24252291 DOI: 10.1016/j.saa.2013.10.081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 10/13/2013] [Accepted: 10/17/2013] [Indexed: 06/02/2023]
Abstract
Crystallographic, experimental (FT-IR and FT-RS) and theoretical density function theory (DFT) and UV-Vis spectra of (E)-1-[1-(4-Chlorophenyl)ethylidene]thiosemicarbazide) (ECET) are investigated. The optimized geometry of the compound was calculated from the DFT-B3LYP gradient calculations employing 6-31G (d,p) basis set and calculated vibrational frequencies are evaluated via comparison with experimental values. Molecular stability has been analyzed using Natural Bond Orbital (NBO) and Natural Localized Molecular Orbital (NLMO) analysis and the limits of the molecular electrostatic potential observed. The calculated HOMO and LUMO energies show the charge transfer occurs within the molecule.
Collapse
Affiliation(s)
- R R Saravanan
- Department of Physics, AMET University, Kanathur, Chennai 603 112, India.
| | - S Seshadri
- Department of Physics, L.N. Govt. Arts College, Ponneri, Thiruvallur 601 204, India
| | - S Gunasekaran
- Research & Development, St. Peter's University, Avadi, Chennai 600 054, India
| | - R Mendoza-Meroño
- Faculty of Chemistry, Department of Physical and Analytical Chemistry, University Oviedo-CINN, C/Julian Claveria, 8, 33006 Oviedo (Asturias), Spain
| | - S Garcia-Granda
- Faculty of Chemistry, Department of Physical and Analytical Chemistry, University Oviedo-CINN, C/Julian Claveria, 8, 33006 Oviedo (Asturias), Spain
| |
Collapse
|
31
|
Mendoza-Meroño R, Menéndez-Taboada L, García-Granda S. 4-(Diphenyl-amino)-benzaldehyde 4-phenyl-thio-semicarbazone. Acta Crystallogr Sect E Struct Rep Online 2012; 68:o2402-3. [PMID: 22904859 PMCID: PMC3414326 DOI: 10.1107/s160053681203053x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 07/04/2012] [Indexed: 11/11/2022]
Abstract
The title molecule, C26H22N4S, is composed of three main parts, viz. a triphenylamine group is connected to a phenyl ring by a thiosemicarbazone moiety. The C= N double bond has an E conformation. The crystal packing is dominated by strong hydrogen bonds through the thiosemicarbazone moiety, with pairs of N—H⋯S hydrogen bonds linking the molecules to form inversion dimers with an R22(8) ring motif. An intramolecular N—H⋯N hydrogen bond is also present, generating an S(5) ring motif. Although the structure contains four phenyl rings, π–π stacking interactions are not formed between them, probably due to the conformation adopted by the triphenylamine group. However, a weak π–π stacking interaction is observed between the phenyl ring and the delocalized thiosemicarbazone moiety.
Collapse
Affiliation(s)
- Rafael Mendoza-Meroño
- Departamento de Química Física y Analítica, Facultad de Química, Universidad de Oviedo - CINN, C/ Julián Clavería, 8, 33006 Oviedo, Spain
| | | | | |
Collapse
|
32
|
Mendoza-Meroño R, García-Granda S. (E)-1-[1-(4-Chloro-phen-yl)ethyl-idene]thio-semicarbazide. Acta Crystallogr Sect E Struct Rep Online 2012; 68:o2309. [PMID: 22904786 PMCID: PMC3414179 DOI: 10.1107/s1600536812029133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 06/26/2012] [Indexed: 11/10/2022]
Abstract
In the crystal structure of the title compound, C(9)H(10)ClN(3)S, the mol-ecules form chains parallel to [001] through N-H⋯S hydrogen bonds. In addition, weak inter-molecular N-H⋯Cl hydrogen bonds connect the chains, forming a two-dimensional network parallel to (001).
Collapse
Affiliation(s)
- Rafael Mendoza-Meroño
- Departamento de Química Física y Analítica, Facultad de Química, Universidad de Oviedo - CINN, C/ Julián Clavería, 8, 33006 Oviedo, Spain
| | | |
Collapse
|
33
|
Tan KW, Seng HL, Lim FS, Cheah SC, Ng CH, Koo KS, Mustafa MR, Ng SW, Maah MJ. Towards a selective cytotoxic agent for prostate cancer: Interaction of zinc complexes of polyhydroxybenzaldehyde thiosemicarbazones with topoisomerase I. Polyhedron 2012. [DOI: 10.1016/j.poly.2012.03.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
34
|
Quach P, Gutierrez E, Basha MT, Kalinowski DS, Sharpe PC, Lovejoy DB, Bernhardt PV, Jansson PJ, Richardson DR. Methemoglobin formation by triapine, di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT), and other anticancer thiosemicarbazones: identification of novel thiosemicarbazones and therapeutics that prevent this effect. Mol Pharmacol 2012; 82:105-14. [PMID: 22508546 DOI: 10.1124/mol.112.078964] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Thiosemicarbazones are a group of compounds that have received comprehensive investigation as anticancer agents. The antitumor activity of the thiosemicarbazone, 3-amino-2-pyridinecarboxaldehyde thiosemicarbazone (3-AP; triapine), has been extensively assessed in more than 20 phase I and II clinical trials. These studies have demonstrated that 3-AP induces methemoglobin (metHb) formation and hypoxia in patients, limiting its usefulness. Considering this problem, we assessed the mechanism of metHb formation by 3-AP compared with that of more recently developed thiosemicarbazones, including di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT). This was investigated using intact red blood cells (RBCs), RBC lysates, purified oxyhemoglobin, and a mouse model. The chelation of cellular labile iron with the formation of a redox-active thiosemicarbazone-iron complex was found to be crucial for oxyhemoglobin oxidation. This observation was substantiated using a thiosemicarbazone that cannot ligate iron and also by using the chelator, desferrioxamine, that forms a redox-inactive iron complex. Of significance, cellular copper chelation was not important for metHb generation in contrast to its role in preventing tumor cell proliferation. Administration of Dp44mT to mice catalyzed metHb and cardiac metmyoglobin formation. However, ascorbic acid administered together with the drug in vivo significantly decreased metHb levels, providing a potential therapeutic intervention. Moreover, we demonstrated that the structure of the thiosemicarbazone is of importance in terms of metHb generation, because the DpT analog, di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), does not induce metHb generation in vivo. Hence, DpC represents a next-generation thiosemicarbazone that possesses markedly superior properties. This investigation is important for developing more effective thiosemicarbazone treatment regimens.
Collapse
Affiliation(s)
- Patricia Quach
- Discipline of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The anticancer effects of thiosemicarbazones were once solely attributed to the inhibition of ribonucleotide reductase, an enzyme involved in the rate-limiting step of DNA synthesis. However, the mechanism behind this inhibition was initially not described. The ability of thiosemicarbazones to chelate metal ions has now been recognized as a major factor in their antiproliferative effects. This mini-review discusses current advances of an emerging 'new wave' of thiosemicarbazones as potent anticancer agents, describing recent insights into their mechanism of action. The redox activity of Fe-thiosemicarbazone complexes is critical in their anticancer activity, resulting in oxidative damage and the inhibition of ribonucleotide reductase. In vivo analysis indicates that some thiosemicarbazones show potential as chemotherapeutic agents.
Collapse
|
36
|
Weir SJ, Patton L, Castle K, Rajewski L, Kasper J, Schimmer AD. The repositioning of the anti-fungal agent ciclopirox olamine as a novel therapeutic agent for the treatment of haematologic malignancy. J Clin Pharm Ther 2010; 36:128-34. [PMID: 21366640 DOI: 10.1111/j.1365-2710.2010.01172.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE 6-Cyclohexyl-1-hydroxy-4-methyl-2(1H)-pyridone (ciclopirox) and specifically its olamine salt 6-cyclohexyl-1-hydroxy-4-methyl-2(1H)-pyridone 2-aminoethanol salt (ciclopirox olamine) are anti-fungal agents currently used for the treatment of mild to moderate cutaneous fungal infection. Our objective is to comment on the opportunity to rapidly reposition ciclopirox and its olamine for the treatment of haematologic malignancy by leveraging its prior published toxicology and pharmacology data. COMMENT Ciclopirox olamine chelates intracellular iron and displays preclinical efficacy in the treatment of haematologic malignancy. Currently, an ongoing study is evaluating topical ciclopirox olamine for the treatment of cervical cancer. Doses of ciclopirox olaine required for a systemic anti-cancer effect appear pharmacologically achievable. However, caution is required as at the highest doses tested in animal toxicology studies, irreversible cardiac degeneration was observed. WHAT IS NEW AND CONCLUSION The existing pharmacology and toxicology data suggest that systemic ciclopirox olamine could be repositioned as a new investigational anti-cancer agent. The available pharmacology and toxicology data should aid in the design of phase I clinical trials of this agent in patients with refractory haematologic malignancies.
Collapse
Affiliation(s)
- S J Weir
- Institute for Advancing Medical Innovation, The University of Kansas Cancer Center, Kansas City, Kansas, USA
| | | | | | | | | | | |
Collapse
|
37
|
Yu Y, Kalinowski DS, Kovacevic Z, Siafakas AR, Jansson PJ, Stefani C, Lovejoy DB, Sharpe PC, Bernhardt PV, Richardson DR. Thiosemicarbazones from the old to new: iron chelators that are more than just ribonucleotide reductase inhibitors. J Med Chem 2009; 52:5271-94. [PMID: 19601577 DOI: 10.1021/jm900552r] [Citation(s) in RCA: 306] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yu Yu
- Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kowol CR, Trondl R, Heffeter P, Arion VB, Jakupec MA, Roller A, Galanski M, Berger W, Keppler BK. Impact of Metal Coordination on Cytotoxicity of 3-Aminopyridine-2-carboxaldehyde Thiosemicarbazone (Triapine) and Novel Insights into Terminal Dimethylation. J Med Chem 2009; 52:5032-43. [DOI: 10.1021/jm900528d] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Christian R. Kowol
- University of Vienna, Institute of Inorganic Chemistry, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Robert Trondl
- University of Vienna, Institute of Inorganic Chemistry, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Vladimir B. Arion
- University of Vienna, Institute of Inorganic Chemistry, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Michael A. Jakupec
- University of Vienna, Institute of Inorganic Chemistry, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Alexander Roller
- University of Vienna, Institute of Inorganic Chemistry, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Markus Galanski
- University of Vienna, Institute of Inorganic Chemistry, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- University of Vienna, Institute of Inorganic Chemistry, Währinger Strasse 42, A-1090 Vienna, Austria
| |
Collapse
|
39
|
Atasever B, Ulküseven B, Bal-Demirci T, Erdem-Kuruca S, Solakoğlu Z. Cytotoxic activities of new iron(III) and nickel(II) chelates of some S-methyl-thiosemicarbazones on K562 and ECV304 cells. Invest New Drugs 2009; 28:421-32. [PMID: 19495562 DOI: 10.1007/s10637-009-9272-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 05/22/2009] [Indexed: 10/20/2022]
Abstract
The S-methyl-thiosemicarbazones of the 2-hydroxy-R-benzaldehyde (R = H, 3-OH 3-OCH(3) or 4-OCH(3)) reacted with the corresponding aldehydes in the presence of FeCl(3) and NiCl(2). New ONNO chelates of iron(III) and nickel(II) with hydroxy- or methoxy-substituted N(1),N(4)-diarylidene-S-methyl-thiosemicarbazones were characterized by means of elemental analysis, conductivity and magnetic measurements, UV-Vis, IR and (1)H-NMR spectroscopies. Cytotoxic activities of the compounds were determined using K562 chronic myeloid leukemia and ECV304 human endothelial cell lines by MTT assay. It was determined that monochloro N(1)-4-methoxysalicylidene-N(4)-4-methoxysalicylidene-S-methyl-thiosemicarbazidato-iron(III) complex showed selective anti-leukemic effects in K562 cells while has no effect in ECV304 cells in the 0.53 microg/ml (IC(50)) concentrations. Also, some methoxy-substituted nickel(II) chelates exhibit high cytotoxic activity against both of these cell lines in low concentrations. Cytotoxicity data were evaluated depending on cell lines origin and position of the substituents on aromatic rings.
Collapse
Affiliation(s)
- Belkis Atasever
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Türkiye
| | | | | | | | | |
Collapse
|
40
|
Noulsri E, Richardson DR, Lerdwana S, Fucharoen S, Yamagishi T, Kalinowski DS, Pattanapanyasat K. Antitumor activity and mechanism of action of the iron chelator, Dp44mT, against leukemic cells. Am J Hematol 2009; 84:170-6. [PMID: 19140186 DOI: 10.1002/ajh.21350] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Iron chelators have been reported to induce apoptosis and cell cycle arrest in cancer cells. Recent studies suggest broad and selective antitumor activity of the new iron chelator, di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT; Whitnall et al., Proc Natl Acad Sci USA 2006;103:14901-14906). However, little is known concerning its effects on hematological malignancies. Using acute leukemia cells, the effect of Dp44mT on apoptosis, cell cycle, caspase-3 activation, and mitochondrial trans-membrane potential has been examined by flow cytometry. Dp44mT acted to induce a G(1)/S arrest in NB4 promyelocytic leukemia cells at low concentrations (0.5-2.5 microM), being far more effective than the clinically used chelator, desferrioxamine (DFO). Moreover, Dp44mT induced apoptosis of NB4 cells in a dose- and time-dependent manner with markedly less effect on nonproliferating cells. The apoptosis-inducing activity of Dp44mT was significantly more effective than DFO. Furthermore, this study also showed that Dp44mT had broad activity, inducing apoptosis in several types of acute leukemia and also multiple myeloma cell lines. Additional studies examining the cytotoxic mechanisms of Dp44mT showed that a reduction in the mitochondrial trans-membrane potential and caspase-3 activation could be involved in the mechanism of apoptosis. Our results suggest that Dp44mT possesses potential as an effective cytotoxic agent for the chemotherapeutic treatment of acute leukemia.
Collapse
Affiliation(s)
- Egarit Noulsri
- Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | | | | | | | | | | |
Collapse
|
41
|
Bernhardt PV, Sharpe PC, Islam M, Lovejoy DB, Kalinowski DS, Richardson DR. Iron chelators of the dipyridylketone thiosemicarbazone class: precomplexation and transmetalation effects on anticancer activity. J Med Chem 2009; 52:407-15. [PMID: 19090766 DOI: 10.1021/jm801012z] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We previously reported a series of di-2-pyridylketone thiosemicarbazone (HDpT) chelators that showed marked and selective antitumor activity (Whitnall, M.; et al. Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 14901-14906). To further understand their biological efficacy, we report the characterization and activity of their Mn(II), Co(III), Ni(II), Cu(II), and Zn(II) complexes. The X-ray crystal structures of four divalent (Mn, Ni, Cu, and Zn) and one trivalent (Fe) complexes are reported. Electrochemistry shows the Fe(III/II) and Cu(II/I) potentials of the complexes may be redox-active within cells. Stability constants were also determined for the Mn(II), Ni(II), Cu(II), and Zn(II) complexes. All divalent complexes underwent transmetalation upon encountering Fe(II), to form low spin ferrous complexes. Importantly, the divalent Mn(II), Ni(II), Cu(II), and Zn(II) complexes of the HDpT analogues are equally active in preventing proliferation as their ligands, suggesting the complexes act as lipophilic vehicles facilitating intracellular delivery of the free ligand upon metal dissociation.
Collapse
Affiliation(s)
- Paul V Bernhardt
- Centre for Metals in Biology, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Qld 4072, Australia.
| | | | | | | | | | | |
Collapse
|
42
|
Penel N, Delord JP, Bonneterre ME, Bachelot T, Ray-Coquard I, Blay JY, Pascal LB, Borel C, Filleron T, Adenis A, Bonneterre J. Development and validation of a model that predicts early death among cancer patients participating in phase I clinical trials investigating cytotoxics. Invest New Drugs 2009; 28:76-82. [PMID: 19205623 DOI: 10.1007/s10637-009-9224-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2008] [Accepted: 01/27/2009] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Selecting patients for phase 1 studies remains challenging. Given the lack of clear and reliable guidance for the estimation of life expectancy, we retrospectively assessed predictive factors of early death (within 90 days following inclusion) among these patients. METHODS Two hundred fifty-seven consecutive cancer patients enrolled in phase I studies investigating cytotoxics at Oscar Lambret Cancer Center and Institut Claudius Regaud were included in the development database. Univariate and multivariate analyses (logistic regression model) were undertaken to determine the prognostic factors. A probability tree described the rate of early death in the different prognostic subgroups. This prognostic model was then evaluated on a second independent cohort of 128 patients treated at Léon Bérard Cancer Center. RESULTS The median overall survival was 8.4 months in the dataset population, and the rate of early death was 15%. In multivariate analysis, the two prognostic factors for early death were albumin <38 g/l (OR = 5.21) and lymphocytes <700/mm(3) (OR = 3.88). According to these two parameters, three prognostic subgroups were defined with early death rates of, respectively, 8/121 (6%), 19/119 (16%) and 13/17 (76%). In the validation dataset, the rates of early death according to three prognostic groups were 13/68 (19%), 20/57 (35%) and 3/3 (100%), respectively. CONCLUSION We do not recommend the enrolment of patients with albumin level below 38g/l and lymphocytes count below 700/mm(3), in phase 1 trial investigating cytotoxics. Our model is helpful to discriminate "patients with reasonable life expectancy" as defined in most phase 1 protocols.
Collapse
Affiliation(s)
- Nicolas Penel
- Département de Cancérologie Générale, Centre Oscar Lambret, 3, Rue F Combemale, Lille, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|